51
|
Chen W, Hu Y, Ju D. Gene therapy for neurodegenerative disorders: advances, insights and prospects. Acta Pharm Sin B 2020; 10:1347-1359. [PMID: 32963936 PMCID: PMC7488363 DOI: 10.1016/j.apsb.2020.01.015] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/09/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023] Open
Abstract
Gene therapy is rapidly emerging as a powerful therapeutic strategy for a wide range of neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Some early clinical trials have failed to achieve satisfactory therapeutic effects. Efforts to enhance effectiveness are now concentrating on three major fields: identification of new vectors, novel therapeutic targets, and reliable of delivery routes for transgenes. These approaches are being assessed closely in preclinical and clinical trials, which may ultimately provide powerful treatments for patients. Here, we discuss advances and challenges of gene therapy for neurodegenerative disorders, highlighting promising technologies, targets, and future prospects.
Collapse
Key Words
- AADC, aromatic-l-amino-acid
- AAVs, adeno-associated viruses
- AD, Alzheimer's disease
- ARSA, arylsulfatase A
- ASOs, antisense oligonucleotides
- ASPA, aspartoacylase
- Adeno-associated viruses
- Adv, adenovirus
- BBB, blood–brain barrier
- BCSFB, blood–cerebrospinal fluid barrier
- BRB, blood–retina barrier
- Bip, glucose regulated protein 78
- CHOP, CCAAT/enhancer binding homologous protein
- CLN6, ceroidlipofuscinosis neuronal protein 6
- CNS, central nervous system
- CSF, cerebrospinal fluid
- Central nervous system
- Delivery routes
- ER, endoplasmic reticulum
- FDA, U.S. Food and Drug Administration
- GAA, lysosomal acid α-glucosidase
- GAD, glutamic acid decarboxylase
- GDNF, glial derived neurotrophic factor
- Gene therapy
- HD, Huntington's disease
- HSPGs, heparin sulfate proteoglycans
- HTT, mutant huntingtin
- IDS, iduronate 2-sulfatase
- LVs, retrovirus/lentivirus
- Lamp2a, lysosomal-associated membrane protein 2a
- NGF, nerve growth factor
- Neurodegenerative disorders
- PD, Parkinson's disease
- PGRN, Progranulin
- PINK1, putative kinase 1
- PTEN, phosphatase and tensin homolog
- RGCs, retinal ganglion cells
- RNAi, RNA interference
- RPE, retinal pigmented epithelial
- SGSH, lysosomal heparan-N-sulfamidase gene
- SMN, survival motor neuron
- SOD, superoxide dismutase
- SUMF, sulfatase-modifying factor
- TFEB, transcription factor EB
- TPP1, tripeptidyl peptidase 1
- TREM2, triggering receptor expressed on myeloid cells 2
- UPR, unfolded protein response
- ZFPs, zinc finger proteins
- mTOR, mammalian target of rapamycin
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Wei Chen
- Department of Biological Medicines, Fudan University School of Pharmacy, Shanghai 201203, China
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Dianwen Ju
- Department of Biological Medicines, Fudan University School of Pharmacy, Shanghai 201203, China
| |
Collapse
|
52
|
Mudali D, Jeevanandam J, Danquah MK. Probing the characteristics and biofunctional effects of disease-affected cells and drug response via machine learning applications. Crit Rev Biotechnol 2020; 40:951-977. [PMID: 32633615 DOI: 10.1080/07388551.2020.1789062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Drug-induced transformations in disease characteristics at the cellular and molecular level offers the opportunity to predict and evaluate the efficacy of pharmaceutical ingredients whilst enabling the optimal design of new and improved drugs with enhanced pharmacokinetics and pharmacodynamics. Machine learning is a promising in-silico tool used to simulate cells with specific disease properties and to determine their response toward drug uptake. Differences in the properties of normal and infected cells, including biophysical, biochemical and physiological characteristics, plays a key role in developing fundamental cellular probing platforms for machine learning applications. Cellular features can be extracted periodically from both the drug treated, infected, and normal cells via image segmentations in order to probe dynamic differences in cell behavior. Cellular segmentation can be evaluated to reflect the levels of drug effect on a distinct cell or group of cells via probability scoring. This article provides an account for the use of machine learning methods to probe differences in the biophysical, biochemical and physiological characteristics of infected cells in response to pharmacokinetics uptake of drug ingredients for application in cancer, diabetes and neurodegenerative disease therapies.
Collapse
Affiliation(s)
- Deborah Mudali
- Department of Computer Science, University of Tennessee, Chattanooga, TN, USA
| | - Jaison Jeevanandam
- Department of Chemical Engineering, Faculty of Engineering and Science, Curtin University, Miri, Malaysia
| | - Michael K Danquah
- Chemical Engineering Department, University of Tennessee, Chattanooga, TN, USA
| |
Collapse
|
53
|
Intravitreal enzyme replacement inhibits progression of retinal degeneration in canine CLN2 neuronal ceroid lipofuscinosis. Exp Eye Res 2020; 198:108135. [PMID: 32634395 DOI: 10.1016/j.exer.2020.108135] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 01/13/2023]
Abstract
CLN2 neuronal ceroid lipofuscinosis is a rare recessive hereditary retinal and neurodegenerative disease resulting from deleterious sequence variants in TPP1 that encodes the soluble lysosomal enzyme tripeptidyl peptidase-1 (TPP1). Children with this disorder develop normally, but starting at 2-4 years of age begin to exhibit neurological signs and visual deficits. Vision loss that progresses to blindness is associated with progressive retinal degeneration and impairment of retinal function. Similar progressive loss of retinal function and retinal degeneration occur in a dog CLN2 disease model with a TPP1 null sequence variant. Studies using the dog model were conducted to determine whether intravitreal injection of recombinant human TPP1 (rhTPP1) administered starting after onset of retinal functional impairment could slow or halt the progression of retinal functional decline and degeneration. TPP1-null dogs received intravitreal injections of rhTPP1 in one eye and vehicle in the other eye beginning at 23.5-25 weeks of age followed by second injections at 34-40 weeks in 3 out of 4 dogs. Ophthalmic exams, in vivo ocular imaging, and electroretinography (ERG) were repeated regularly to monitor retinal structure and function. Retinal histology was evaluated in eyes collected from these dogs when they were euthanized at end-stage neurological disease (40-45 weeks of age). Intravitreal rhTPP1 injections were effective in preserving retinal function (as measured with the electroretinogram) and retinal morphology for as long as 4 months after a single treatment. These findings indicate that intravitreal injection of rhTPP1 administered after partial loss of retinal function is an effective treatment for preserving retinal structure and function in canine CLN2 disease.
Collapse
|
54
|
Whiting REH, Pearce JW, Vansteenkiste DP, Bibi K, Lim S, Robinson Kick G, Castaner LJ, Sinclair J, Chandra S, Nguyen A, O'Neill CA, Katz ML. Intravitreal enzyme replacement preserves retinal structure and function in canine CLN2 neuronal ceroid lipofuscinosis. Exp Eye Res 2020; 197:108130. [PMID: 32622066 DOI: 10.1016/j.exer.2020.108130] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 01/01/2023]
Abstract
CLN2 neuronal ceroid lipofuscinosis is a hereditary neurodegenerative disorder characterized by progressive vision loss, neurological decline, and seizures. CLN2 disease results from mutations in TPP1 that encodes the lysosomal enzyme tripeptidyl peptidase-1 (TPP1). Children with CLN2 neuronal ceroid lipofuscinosis experience ocular disease, characterized by progressive retinal degeneration associated with impaired retinal function and gradual vision loss culminating in total blindness. A similar progressive loss of retinal function is also observed in a dog CLN2 model with a TPP1 null mutation. A study was conducted to evaluate the efficacy of periodic intravitreal injections of recombinant human (rh) TPP1 in inhibiting retinal degeneration and preserving retinal function in the canine model. TPP1 null dogs received periodic intravitreal injections of rhTPP1 in one eye and vehicle in the other eye beginning at approximately 12 weeks of age. Ophthalmic exams, in vivo ocular imaging, and electroretinography (ERG) were repeated regularly to monitor retinal structure and function. Retinal histology was evaluated in eyes collected from these dogs when they were euthanized at end-stage neurological disease (43-46 weeks of age). Intravitreal rhTPP1 dosing prevented disease-related declines in ERG amplitudes in the TPP1-treated eyes. At end-stage neurologic disease, TPP1-treated eyes retained normal morphology while the contralateral vehicle-treated eyes exhibited loss of inner retinal neurons and photoreceptor disorganization typical of CLN2 disease. The treatment also prevented the development of disease-related focal retinal detachments observed in the control eyes. Uveitis occurred secondary to the administration of the rhTPP1 but did not hinder the therapeutic benefits. These findings demonstrate that periodic intravitreal injection of rhTPP1 preserves retinal structure and function in canine CLN2 disease.
Collapse
Affiliation(s)
- Rebecca E H Whiting
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, USA
| | - Jacqueline W Pearce
- Veterinary Medicine and Surgery, University of Missouri College of Veterinary Medicine, Columbia, USA
| | - Daniella P Vansteenkiste
- Veterinary Medicine and Surgery, University of Missouri College of Veterinary Medicine, Columbia, USA
| | - Katherine Bibi
- Veterinary Medicine and Surgery, University of Missouri College of Veterinary Medicine, Columbia, USA
| | - Stefanie Lim
- Veterinary Medicine and Surgery, University of Missouri College of Veterinary Medicine, Columbia, USA
| | - Grace Robinson Kick
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, USA
| | - Leilani J Castaner
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, USA
| | | | | | | | | | - Martin L Katz
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, USA.
| |
Collapse
|
55
|
Kwok D, Okada H. T-Cell based therapies for overcoming neuroanatomical and immunosuppressive challenges within the glioma microenvironment. J Neurooncol 2020; 147:281-295. [PMID: 32185647 PMCID: PMC7182069 DOI: 10.1007/s11060-020-03450-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/05/2020] [Indexed: 12/22/2022]
Abstract
Glioblastoma remains as the most common and aggressive primary adult brain tumor to date. Within the last decade, cancer immunotherapy surfaced as a broadly successful therapeutic approach for a variety of cancers. However, due to the neuroanatomical and immunosuppressive nature of malignant gliomas, conventional chemotherapy and radiotherapy treatments garner limited efficacy in patients with these tumors. The intricate structure of the blood brain barrier restricts immune accessibility into the tumor microenvironment, and malignant gliomas can activate various adaptive responses to subvert anticancer immune responses and reinstate an immunosuppressive milieu. Yet, evidence of lymphocyte infiltration within the brain and recent advancements made in cell engineering technologies implicate the vast potential in the future of neuro-oncological immunotherapy. Previous immunotherapy platforms have paved way to improved modalities, which includes but is not limited to personalized vaccines and chimeric antigen receptor T-cell therapy. This review will cover the various neuroanatomical and immunosuppressive features of central nervous system tumors and highlight the innovations made in T-cell based therapies to overcome the challenges presented by the glioblastoma microenvironment.
Collapse
Affiliation(s)
- Darwin Kwok
- Department of Neurological Surgery, University of California, San Francisco, Helen Diller Family Cancer Research Building HD 472 1450 3rd Street, San Francisco, CA, 94158-0520, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, Helen Diller Family Cancer Research Building HD 472 1450 3rd Street, San Francisco, CA, 94158-0520, USA.
- The Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
- Cancer Immunotherapy Program, University of California, San Francisco, CA, USA.
| |
Collapse
|
56
|
Specchio N, Pietrafusa N, Trivisano M. Changing Times for CLN2 Disease: The Era of Enzyme Replacement Therapy. Ther Clin Risk Manag 2020; 16:213-222. [PMID: 32280231 PMCID: PMC7127909 DOI: 10.2147/tcrm.s241048] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/18/2020] [Indexed: 01/23/2023] Open
Abstract
Neuronal ceroid lipofuscinosis type 2 (CLN2 disease) is a progressive neurodegenerative disease that results in early-onset, severe, progressive, neurological disabilities, leading to death in late childhood or early adolescence. Management has relied on symptomatic care, and supportive and palliative strategies, but the approval of the enzyme replacement therapy cerliponase alfa in the USA and Europe in 2017 brought different treatment opportunities. We describe the natural history of CLN2 disease, its diagnosis and management, and the preclinical and clinical development of cerliponase alfa. A PubMed search was undertaken for cerliponase alfa and rhTPP1 to identify preclinical and clinical studies. The hallmark-presenting symptoms of CLN2 disease are unprovoked seizures and a history of language delay, and progression involves motor dysfunction, and cognitive and visual decline. Cerliponase alfa has shown efficacy and tolerability in mouse and canine models of CLN2 disease when delivered intracerebroventricularly. Administration of cerliponase alfa in patients with CLN2 disease has led to significant reductions in the rate of decline of motor and language functions in comparison with a natural history population. The approval of cerliponase alfa has brought a new era for CLN2 disease, highlighting the need to understand different patterns of disease progression and clinical needs in treated patients.
Collapse
Affiliation(s)
- Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Nicola Pietrafusa
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marina Trivisano
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
57
|
Liu W, Kleine-Holthaus SM, Herranz-Martin S, Aristorena M, Mole SE, Smith AJ, Ali RR, Rahim AA. Experimental gene therapies for the NCLs. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165772. [PMID: 32220628 DOI: 10.1016/j.bbadis.2020.165772] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023]
Abstract
The neuronal ceroid lipofuscinoses (NCLs), also known as Batten disease, are a group of rare monogenic neurodegenerative diseases predominantly affecting children. All NCLs are lethal and incurable and only one has an approved treatment available. To date, 13 NCL subtypes (CLN1-8, CLN10-14) have been identified, based on the particular disease-causing defective gene. The exact functions of NCL proteins and the pathological mechanisms underlying the diseases are still unclear. However, gene therapy has emerged as an attractive therapeutic strategy for this group of conditions. Here we provide a short review discussing updates on the current gene therapy studies for the NCLs.
Collapse
Affiliation(s)
- Wenfei Liu
- UCL School of Pharmacy, University College London, UK
| | | | - Saul Herranz-Martin
- UCL School of Pharmacy, University College London, UK; Centro de Biología Molecular Severo Ochoa (UAM-CSIC) and Departamento de Biología Molecular,Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | | | - Sara E Mole
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK
| | | | - Robin R Ali
- UCL Institute of Ophthalmology, University College London, UK; NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust, UK
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, UK.
| |
Collapse
|
58
|
Story BD, Miller ME, Bradbury AM, Million ED, Duan D, Taghian T, Faissler D, Fernau D, Beecy SJ, Gray-Edwards HL. Canine Models of Inherited Musculoskeletal and Neurodegenerative Diseases. Front Vet Sci 2020; 7:80. [PMID: 32219101 PMCID: PMC7078110 DOI: 10.3389/fvets.2020.00080] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Mouse models of human disease remain the bread and butter of modern biology and therapeutic discovery. Nonetheless, more often than not mouse models do not reproduce the pathophysiology of the human conditions they are designed to mimic. Naturally occurring large animal models have predominantly been found in companion animals or livestock because of their emotional or economic value to modern society and, unlike mice, often recapitulate the human disease state. In particular, numerous models have been discovered in dogs and have a fundamental role in bridging proof of concept studies in mice to human clinical trials. The present article is a review that highlights current canine models of human diseases, including Alzheimer's disease, degenerative myelopathy, neuronal ceroid lipofuscinosis, globoid cell leukodystrophy, Duchenne muscular dystrophy, mucopolysaccharidosis, and fucosidosis. The goal of the review is to discuss canine and human neurodegenerative pathophysiologic similarities, introduce the animal models, and shed light on the ability of canine models to facilitate current and future treatment trials.
Collapse
Affiliation(s)
- Brett D. Story
- Auburn University College of Veterinary Medicine, Auburn, AL, United States
- University of Florida College of Veterinary Medicine, Gainesville, FL, United States
| | - Matthew E. Miller
- Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Allison M. Bradbury
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily D. Million
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Dominik Faissler
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, United States
| | - Deborah Fernau
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Sidney J. Beecy
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, United States
| | - Heather L. Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
59
|
Gurda BL, Vite CH. Large animal models contribute to the development of therapies for central and peripheral nervous system dysfunction in patients with lysosomal storage diseases. Hum Mol Genet 2020; 28:R119-R131. [PMID: 31384936 DOI: 10.1093/hmg/ddz127] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/16/2019] [Accepted: 06/07/2019] [Indexed: 12/12/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of 70 monogenic disorders characterized by the lysosomal accumulation of a substrate. As a group, LSDs affect ~1 in 5000 live births; however, each individual storage disease is rare, limiting the ability to perform natural history studies or to perform clinical trials. Perhaps in no other biomedical field have naturally occurring large animal (canine, feline, ovine, caprine, and bovine) models been so essential for understanding the fundamentals of disease pathogenesis and for developing safe and effective therapies. These models were critical for the development of hematopoietic stem cell transplantation in α- and β- mannosidosis, fucosidosis, and the mucopolysaccharidoses; enzyme replacement therapy for fucosidosis, the mucopolysaccharidoses, and neuronal ceroid lipofuscinosis; and small molecule therapy in Niemann-Pick type C disease. However, their most notable contributions to the biomedical field are in the development of gene therapy for LSDs. Adeno-associated viral vectors to treat nervous system disease have been evaluated in the large animal models of α-mannosidosis, globoid cell leukodystrophy, GM1 and GM2 gangliosidosis, the mucopolysaccharidoses, and neuronal ceroid lipofuscinosis. This review article will summarize the large animal models available for study as well as their contributions to the development of central and peripheral nervous system dysfunction in LSDs.
Collapse
Affiliation(s)
- Brittney L Gurda
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles H Vite
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
60
|
Nelvagal HR, Cooper JD. An update on the progress of preclinical models for guiding therapeutic management of neuronal ceroid lipofuscinosis. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1703672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Hemanth Ramesh Nelvagal
- Department of Pediatrics, Division of genetics and genomics, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | - Jonathan D Cooper
- Department of Pediatrics, Division of genetics and genomics, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| |
Collapse
|
61
|
Cardiac pathology in neuronal ceroid lipofuscinoses (NCL): More than a mere co-morbidity. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165643. [PMID: 31863828 DOI: 10.1016/j.bbadis.2019.165643] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are mostly seen as diseases affecting the central nervous system, but there is accumulating evidence that they have co-morbidities outside the brain. One of these co-morbidities is a decline in cardiac function. This is becoming increasingly recognised in teenagers and adolescents with juvenile CLN3, but it may also occur in individuals with other NCLs. The purpose of this review is to summarise the current knowledge of the structural and functional changes found in the hearts of animal models and people diagnosed with NCL. In addition, we present evidence of structural changes that were observed in a systematic comparison of the cardiomyocytes from CLN3Δex7/8 mice.
Collapse
|
62
|
Rosenberg JB, Chen A, Kaminsky SM, Crystal RG, Sondhi D. Advances in the Treatment of Neuronal Ceroid Lipofuscinosis. Expert Opin Orphan Drugs 2019; 7:473-500. [PMID: 33365208 PMCID: PMC7755158 DOI: 10.1080/21678707.2019.1684258] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022]
Abstract
Neuronal ceroid lipofuscinoses (NCL) represent a class of neurodegenerative disorders involving defective lysosomal processing enzymes or receptors, leading to lysosomal storage disorders, typically characterized by observation of cognitive and visual impairments, epileptic seizures, ataxia, and deterioration of motor skills. Recent success of a biologic (Brineura®) for the treatment of neurologic manifestations of the central nervous system (CNS) has led to renewed interest in therapeutics for NCL, with the goal of ablating or reversing the impact of these devastating disorders. Despite complex challenges associated with CNS therapy, many treatment modalities have been evaluated, including enzyme replacement therapy, gene therapy, stem cell therapy, and small molecule pharmacotherapy. Because the clinical endpoints for the evaluation of candidate therapies are complex and often reliant on subjective clinical scales, the development of quantitative biomarkers for NCLs has become an apparent necessity for the validation of potential treatments. We will discuss the latest findings in the search for relevant biomarkers for assessing disease progression. For this review, we will focus primarily on recent pre-clinical and clinical developments for treatments to halt or cure these NCL diseases. Continued development of current therapies and discovery of newer modalities will be essential for successful therapeutics for NCL. AREAS COVERED The reader will be introduced to the NCL subtypes, natural histories, experimental animal models, and biomarkers for NCL progression; challenges and different therapeutic approaches, and the latest pre-clinical and clinical research for therapeutic development for the various NCLs. This review corresponds to the literatures covering the years from 1968 to mid-2019, but primarily addresses pre-clinical and clinical developments for the treatment of NCL disease in the last decade and as a follow-up to our 2013 review of the same topic in this journal. EXPERT OPINION Much progress has been made in the treatment of neurologic diseases, such as the NCLs, including better animal models and improved therapeutics with better survival outcomes. Encouraging results are being reported at symposiums and in the literature, with multiple therapeutics reaching the clinical trial stage for the NCLs. The potential for a cure could be at hand after many years of trial and error in the preclinical studies. The clinical development of enzyme replacement therapy (Brineura® for CLN2), immunosuppression (CellCept® for CLN3), and gene therapy vectors (for CLN1, CLN2, CLN3, and CLN6) are providing encouragement to families that have a child afflicted with NCL. We believe that successful therapies in the future may involve the combination of two or more therapeutic modalities to provide therapeutic benefit especially as the patients grow older.
Collapse
Affiliation(s)
- Jonathan B Rosenberg
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Alvin Chen
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
63
|
Huber RJ, Hughes SM, Liu W, Morgan A, Tuxworth RI, Russell C. The contribution of multicellular model organisms to neuronal ceroid lipofuscinosis research. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165614. [PMID: 31783156 DOI: 10.1016/j.bbadis.2019.165614] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023]
Abstract
The NCLs (neuronal ceroid lipofuscinosis) are forms of neurodegenerative disease that affect people of all ages and ethnicities but are most prevalent in children. Commonly known as Batten disease, this debilitating neurological disorder is comprised of 13 different subtypes that are categorized based on the particular gene that is mutated (CLN1-8, CLN10-14). The pathological mechanisms underlying the NCLs are not well understood due to our poor understanding of the functions of NCL proteins. Only one specific treatment (enzyme replacement therapy) is approved, which is for the treating the brain in CLN2 disease. Hence there remains a desperate need for further research into disease-modifying treatments. In this review, we present and evaluate the genes, proteins and studies performed in the social amoeba, nematode, fruit fly, zebrafish, mouse and large animals pertinent to NCL. In particular, we highlight the use of multicellular model organisms to study NCL protein function, pathology and pathomechanisms. Their use in testing novel therapeutic approaches is also presented. With this information, we highlight how future research in these systems may be able to provide new insight into NCL protein functions in human cells and aid in the development of new therapies.
Collapse
Affiliation(s)
- Robert J Huber
- Department of Biology, Trent University, Peterborough, Ontario K9L 0G2, Canada
| | - Stephanie M Hughes
- Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre and Genetics Otago, University of Otago, Dunedin, New Zealand
| | - Wenfei Liu
- School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Alan Morgan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown St., Liverpool L69 3BX, UK
| | - Richard I Tuxworth
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Claire Russell
- Dept. Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK.
| |
Collapse
|
64
|
Taghian T, Marosfoi MG, Puri AS, Cataltepe OI, King RM, Diffie EB, Maguire AS, Martin DR, Fernau D, Batista AR, Kuchel T, Christou C, Perumal R, Chandra S, Gamlin PD, Bertrand SG, Flotte TR, McKenna-Yasek D, Tai PWL, Aronin N, Gounis MJ, Sena-Esteves M, Gray-Edwards HL. A Safe and Reliable Technique for CNS Delivery of AAV Vectors in the Cisterna Magna. Mol Ther 2019; 28:411-421. [PMID: 31813800 DOI: 10.1016/j.ymthe.2019.11.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 11/29/2022] Open
Abstract
Global gene delivery to the CNS has therapeutic importance for the treatment of neurological disorders that affect the entire CNS. Due to direct contact with the CNS, cerebrospinal fluid (CSF) is an attractive route for CNS gene delivery. A safe and effective route to achieve global gene distribution in the CNS is needed, and administration of genes through the cisterna magna (CM) via a suboccipital puncture results in broad distribution in the brain and spinal cord. However, translation of this technique to clinical practice is challenging due to the risk of serious and potentially fatal complications in patients. Herein, we report development of a gene therapy delivery method to the CM through adaptation of an intravascular microcatheter, which can be safely navigated intrathecally under fluoroscopic guidance. We examined the safety, reproducibility, and distribution/transduction of this method in sheep using a self-complementary adeno-associated virus 9 (scAAV9)-GFP vector. This technique was used to treat two Tay-Sachs disease patients (30 months old and 7 months old) with AAV gene therapy. No adverse effects were observed during infusion or post-treatment. This delivery technique is a safe and minimally invasive alternative to direct infusion into the CM, achieving broad distribution of AAV gene transfer to the CNS.
Collapse
Affiliation(s)
- Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Miklos G Marosfoi
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Ajit S Puri
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Neurological Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Oguz I Cataltepe
- Department of Neurological Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Robert M King
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Elise B Diffie
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA
| | - Anne S Maguire
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA; Department of Anatomy, Physiology and Pharmacology, Auburn University, AL 36849, USA
| | - Deborah Fernau
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ana Rita Batista
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Tim Kuchel
- South Australian Health and Medical Research Institute, Gillies Plains, SA 5086, Australia
| | - Chris Christou
- South Australian Health and Medical Research Institute, Gillies Plains, SA 5086, Australia
| | - Raj Perumal
- South Australian Health and Medical Research Institute, Gillies Plains, SA 5086, Australia
| | | | - Paul D Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stephanie G Bertrand
- Department of Environmental Population Health, Cummings Veterinary School at Tufts University, Grafton, MA 01536, USA
| | - Terence R Flotte
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Diane McKenna-Yasek
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Phillip W L Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Neil Aronin
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Matthew J Gounis
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Miguel Sena-Esteves
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
65
|
Mice deficient in the lysosomal enzyme palmitoyl-protein thioesterase 1 (PPT1) display a complex retinal phenotype. Sci Rep 2019; 9:14185. [PMID: 31578378 PMCID: PMC6775149 DOI: 10.1038/s41598-019-50726-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/13/2019] [Indexed: 01/09/2023] Open
Abstract
Neuronal ceroid lipofuscinosis (NCL) type 1 (CLN1) is a neurodegenerative storage disorder caused by mutations in the gene encoding the lysosomal enzyme palmitoyl-protein thioesterase 1 (PPT1). CLN1 patients suffer from brain atrophy, mental and motor retardation, seizures, and retinal degeneration ultimately resulting in blindness. Here, we performed an in-depth analysis of the retinal phenotype of a PPT1-deficient mouse, an animal model of this condition. Reactive astrogliosis and microgliosis were evident in mutant retinas prior to the onset of retinal cell loss. Progressive accumulation of storage material, a pronounced dysregulation of various lysosomal proteins, and accumulation of sequestosome/p62-positive aggregates in the inner nuclear layer also preceded retinal degeneration. At advanced stages of the disease, the mutant retina was characterized by a significant loss of ganglion cells, rod and cone photoreceptor cells, and rod and cone bipolar cells. Results demonstrate that PPT1 dysfunction results in early-onset pathological alterations in the mutant retina, followed by a progressive degeneration of various retinal cell types at relatively late stages of the disease. Data will serve as a reference for future work aimed at developing therapeutic strategies for the treatment of retinal degeneration in CLN1 disease.
Collapse
|
66
|
Rapid progression of a walking disability in a 5-year-old boy with a CLN6 mutation. Brain Dev 2019; 41:726-730. [PMID: 31029456 DOI: 10.1016/j.braindev.2019.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Neuronal ceroid lipofuscinoses (NCLs; CLN) are mainly autosomal recessive neurodegenerative disorders characterized by the accumulation of autofluorescent lipopigments in neuronal and other cells. Symptoms include visual disabilities, motor decline, and epilepsy. Causative genes are CLN1, CLN2, CLN3, CLN5, CLN6, CLN7, CLN8, CLN10, CLN11, CLN12, CLN13, and CLN14. We present the fourth Japanese case with a CLN6 mutation. CASE PRESENTATION At 3 years of age, our patient became clumsy and fell down easily. He developed focal seizures with impaired consciousness and was started on carbamazepine. He showed ataxic walking and dysarthria with increased deep tendon reflexes. Interictal electroencephalogram revealed slow waves in the left temporal and occipital areas. Brain magnetic resonance imaging showed cerebellar atrophy and ventriculomegaly. In optical coherence tomography (OCT), the inner layer of the retina was thick and highly reflective. Exome sequencing revealed a known homozygous mutation, C.794_976del, p. (Ser265del) in CLN6. DISCUSSION A total of 130 cases of NCL with CLN6 mutations have been reported globally, of which only four were from Japan including the current patient. The deletion of serine at position 265 has been reported in six cases. Ser265 is located in a region of short repeated sequences that is susceptible to mutation. Clinical trials of gene therapy using adeno-associated virus serotype 9 have started for NCL6, making early diagnosis crucial. OCT examination might be helpful in achieving a diagnosis.
Collapse
|
67
|
Li H, Okada H, Suzuki S, Sakai K, Izumi H, Matsushima Y, Ichinohe N, Goto YI, Okada T, Inoue K. Gene suppressing therapy for Pelizaeus-Merzbacher disease using artificial microRNA. JCI Insight 2019; 4:125052. [PMID: 31092737 DOI: 10.1172/jci.insight.125052] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 04/17/2019] [Indexed: 11/17/2022] Open
Abstract
Copy number increase or decrease of certain dosage-sensitive genes may cause genetic diseases with distinct phenotypes, conceptually termed genomic disorders. The most common cause of Pelizaeus-Merzbacher disease (PMD), an X-linked hypomyelinating leukodystrophy, is genomic duplication encompassing the entire proteolipid protein 1 (PLP1) gene. Although the exact molecular and cellular mechanisms underlying PLP1 duplication, which causes severe hypomyelination in the central nervous system, remain largely elusive, PLP1 overexpression is likely the fundamental cause of this devastating disease. Here, we investigated if adeno-associated virus-mediated (AAV-mediated) gene-specific suppression may serve as a potential cure for PMD by correcting quantitative aberrations in gene products. We developed an oligodendrocyte-specific Plp1 gene suppression therapy using artificial microRNA under the control of human CNP promoter in a self-complementary AAV (scAAV) platform. A single direct brain injection achieved widespread oligodendrocyte-specific Plp1 suppression in the white matter of WT mice. AAV treatment in Plp1-transgenic mice, a PLP1 duplication model, ameliorated cytoplasmic accumulation of Plp1, preserved mature oligodendrocytes from degradation, restored myelin structure and gene expression, and improved survival and neurological phenotypes. Together, our results provide evidence that AAV-mediated gene suppression therapy can serve as a potential cure for PMD resulting from PLP1 duplication and possibly for other genomic disorders.
Collapse
Affiliation(s)
- Heng Li
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hironori Okada
- Department of Molecular and Medical Genetics, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Sadafumi Suzuki
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kazuhisa Sakai
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hitomi Izumi
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yukiko Matsushima
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Noritaka Ichinohe
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yu-Ichi Goto
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Okada
- Department of Molecular and Medical Genetics, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Ken Inoue
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
68
|
Ren XT, Wang XH, Ding CH, Shen X, Zhang H, Zhang WH, Li JW, Ren CH, Fang F. Next-Generation Sequencing Analysis Reveals Novel Pathogenic Variants in Four Chinese Siblings With Late-Infantile Neuronal Ceroid Lipofuscinosis. Front Genet 2019; 10:370. [PMID: 31105743 PMCID: PMC6494930 DOI: 10.3389/fgene.2019.00370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/08/2019] [Indexed: 12/24/2022] Open
Abstract
Neuronal Ceroid Lipofuscinoses (NCLs) are progressive degenerative diseases mainly affect brain and retina. They are characterized by accumulation of autofluorescent storage material, mitochondrial ATPase subunit C, or sphingolipid activator proteins A and D in lysosomes of most cells. Heterogenous storage material in NCLs is not completely disease-specific. Most of CLN proteins and their natural substrates are not well-characterized. Studies have suggested variants of Late-Infantile NCLs (LINCLs) include the major type CLN2 and minor types CLN5, CLN6, CLN7, and CLN8. Therefore, combination of clinical and molecular analysis has become a more effective diagnosis method. We studied 4 late-infantile NCL siblings characterized by seizures, ataxia as early symptoms, followed by progressive regression in intelligence and behavior, but mutations are located in different genes. Symptoms and progression of 4 types of LINCLs are compared. Pathology of LINCLs is also discussed. We performed Nest-Generation Sequencing on these phenotypically similar families. Three novel variants c.1551+1insTGAT in TPP1, c.244G>T in CLN6, c.554-5A>G in MFSD8 were identified. Potential outcome of the mutations in structure and function of proteins are studied. In addition, we observed some common and unique clinical features of Chinese LINCL patient as compared with those of Western patients, which greatly improved our understanding of the LINCLs.
Collapse
Affiliation(s)
- Xiao-Tun Ren
- Department of Neurology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xiao-Hui Wang
- Department of Neurology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chang-Hong Ding
- Department of Neurology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | | | | | - Wei-Hua Zhang
- Department of Neurology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Jiu-Wei Li
- Department of Neurology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chang-Hong Ren
- Department of Neurology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Fang Fang
- Department of Neurology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
69
|
Johnson TB, Cain JT, White KA, Ramirez-Montealegre D, Pearce DA, Weimer JM. Therapeutic landscape for Batten disease: current treatments and future prospects. Nat Rev Neurol 2019; 15:161-178. [PMID: 30783219 PMCID: PMC6681450 DOI: 10.1038/s41582-019-0138-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Batten disease (also known as neuronal ceroid lipofuscinoses) constitutes a family of devastating lysosomal storage disorders that collectively represent the most common inherited paediatric neurodegenerative disorders worldwide. Batten disease can result from mutations in 1 of 13 genes. These mutations lead to a group of diseases with loosely overlapping symptoms and pathology. Phenotypically, patients with Batten disease have visual impairment and blindness, cognitive and motor decline, seizures and premature death. Pathologically, Batten disease is characterized by lysosomal accumulation of autofluorescent storage material, glial reactivity and neuronal loss. Substantial progress has been made towards the development of effective therapies and treatments for the multiple forms of Batten disease. In 2017, cerliponase alfa (Brineura), a tripeptidyl peptidase enzyme replacement therapy, became the first globally approved treatment for CLN2 Batten disease. Here, we provide an overview of the promising therapeutic avenues for Batten disease, highlighting current FDA-approved clinical trials and prospective future treatments.
Collapse
Affiliation(s)
- Tyler B Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Jacob T Cain
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | | | - David A Pearce
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA.
- Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Sioux Falls, SD, USA.
| | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA.
- Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Sioux Falls, SD, USA.
| |
Collapse
|
70
|
Amado DA, Rieders JM, Diatta F, Hernandez-Con P, Singer A, Mak JT, Zhang J, Lancaster E, Davidson BL, Chen-Plotkin AS. AAV-Mediated Progranulin Delivery to a Mouse Model of Progranulin Deficiency Causes T Cell-Mediated Toxicity. Mol Ther 2019; 27:465-478. [PMID: 30559071 PMCID: PMC6369714 DOI: 10.1016/j.ymthe.2018.11.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/01/2018] [Accepted: 11/11/2018] [Indexed: 11/16/2022] Open
Abstract
Adeno-associated virus-mediated gene replacement is emerging as a safe and effective means of correcting single-gene mutations affecting the CNS. AAV-mediated progranulin gene (GRN) delivery has been proposed as a treatment for GRN-deficient frontotemporal dementia and neuronal ceroid lipofuscinosis, and recent studies using intraparenchymal AAV-Grn delivery to brain have shown moderate success in histopathologic and behavioral rescue in mouse models. Here, we used AAV9 to deliver GRN to the lateral ventricle to achieve widespread expression in the Grn null mouse brain. We found that, despite a global increase in progranulin, overexpression resulted in dramatic and selective hippocampal toxicity and degeneration affecting neurons and glia. Hippocampal degeneration was preceded by T cell infiltration and perivascular cuffing. GRN delivery with an ependymal-targeting AAV for selective secretion of progranulin into the cerebrospinal fluid similarly resulted in T cell infiltration, as well as ependymal hypertrophy. Interestingly, overexpression of GRN in wild-type animals also provoked T cell infiltration. These results call into question the safety of GRN overexpression in the CNS, with evidence for both a region-selective immune response and cellular proliferative response. Our results highlight the importance of careful consideration of target gene biology and cellular response to overexpression prior to progressing to the clinic.
Collapse
Affiliation(s)
- Defne A Amado
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julianne M Rieders
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, 5060 CTRB, Philadelphia, PA 19104, USA
| | - Fortunay Diatta
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pilar Hernandez-Con
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adina Singer
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jordan T Mak
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junxian Zhang
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric Lancaster
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Beverly L Davidson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, 5060 CTRB, Philadelphia, PA 19104, USA.
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
71
|
Kohlschütter A, Schulz A, Bartsch U, Storch S. Current and Emerging Treatment Strategies for Neuronal Ceroid Lipofuscinoses. CNS Drugs 2019; 33:315-325. [PMID: 30877620 PMCID: PMC6440934 DOI: 10.1007/s40263-019-00620-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The neuronal ceroid lipofuscinoses comprise a group of neurodegenerative lysosomal storage disorders caused by mutations in at least 13 different genes and primarily affect the brain and the retina of children or young adults. The disorders are characterized by progressive neurological deterioration with dementia, epilepsy, loss of vision, motor disturbances, and early death. While various therapeutic strategies are currently being explored as treatment options for these fatal disorders, there is presently only one clinically approved drug that has been shown to effectively attenuate the progression of a specific form of neuronal ceroid lipofuscinosis, CLN2 disease (cerliponase alfa, a lysosomal enzyme infused into the brain ventricles of patients with CLN2 disease). Therapeutic approaches for the treatment of other forms of neuronal ceroid lipofuscinosis include the administration of immunosuppressive agents to antagonize neuroinflammation associated with neurodegeneration, the use of various small molecules, stem cell therapy, and gene therapy. An important aspect of future work aimed at developing therapies for neuronal ceroid lipofuscinoses is the need for treatments that effectively attenuate neurodegeneration in both the brain and the retina.
Collapse
Affiliation(s)
- Alfried Kohlschütter
- Department of Pediatrics, University Medical Center Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Angela Schulz
- 0000 0001 2180 3484grid.13648.38Department of Pediatrics, University Medical Center Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Udo Bartsch
- 0000 0001 2180 3484grid.13648.38Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Stephan Storch
- 0000 0001 2180 3484grid.13648.38Department of Pediatrics, Section Biochemistry, University Medical Center Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
72
|
Hitti FL, Gonzalez-Alegre P, Lucas TH. Gene Therapy for Neurologic Disease: A Neurosurgical Review. World Neurosurg 2019; 121:261-273. [DOI: 10.1016/j.wneu.2018.09.097] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 01/01/2023]
|
73
|
Sheth J, Mistri M, Bhavsar R, Pancholi D, Kamate M, Gupta N, Kabra M, Mehta S, Nampoothiri S, Thakker A, Jain V, Shah R, Sheth F. Batten disease: biochemical and molecular characterization revealing novel PPT1 and TPP1 gene mutations in Indian patients. BMC Neurol 2018; 18:203. [PMID: 30541466 PMCID: PMC6292089 DOI: 10.1186/s12883-018-1206-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
Background Neuronal ceroid lipofuscinoses type I and type II (NCL1 and NCL2) also known as Batten disease are the commonly observed neurodegenerative lysosomal storage disorder caused by mutations in the PPT1 and TPP1 genes respectively. Till date, nearly 76 mutations in PPT1 and approximately 140 mutations, including large deletion/duplications, in TPP1 genes have been reported in the literature. The present study includes 34 unrelated Indian patients (12 females and 22 males) having epilepsy, visual impairment, cerebral atrophy, and cerebellar atrophy. Methods The biochemical investigation involved measuring the palmitoyl protein thioesterase 1 and tripeptidy peptidase l enzyme activity from the leukocytes. Based on the biochemical analysis all patients were screened for variations in either PPT1 gene or TPP1 gene using bidirectional Sanger sequencing. In cases where Sanger sequencing results was uninformative Multiplex Ligation-dependent Probe Amplification technique was employed. The online tools performed the protein homology modeling and orthologous conservation of the novel variants. Results Out of 34 patients analyzed, the biochemical assay confirmed 12 patients with NCL1 and 22 patients with NCL2. Molecular analysis of PPT1 gene in NCL1 patients revealed three known mutations (p.Val181Met, p.Asn110Ser, and p.Trp186Ter) and four novel variants (p.Glu178Asnfs*13, p.Pro238Leu, p.Cys45Arg, and p.Val236Gly). In the case of NCL2 patients, the TPP1 gene analysis identified seven known mutations and eight novel variants. Overall these 15 variants comprised seven missense variants (p.Met345Leu, p.Arg339Trp, p.Arg339Gln, p.Arg206Cys, p.Asn286Ser, p.Arg152Ser, p.Tyr459Ser), four frameshift variants (p.Ser62Argfs*19, p.Ser153Profs*19, p.Phe230Serfs*28, p.Ile484Aspfs*7), three nonsense variants (p.Phe516*, p.Arg208*, p.Tyr157*) and one intronic variant (g.2023_2024insT). No large deletion/duplication was identified in three NCL1 patients where Sanger sequencing study was normal. Conclusion The given study reports 34 patients with Batten disease. In addition, the study contributes four novel variants to the spectrum of PPT1 gene mutations and eight novel variants to the TPP1 gene mutation data. The novel pathogenic variant p.Pro238Leu occurred most commonly in the NCL1 cohort while the occurrence of a known pathogenic mutation p.Arg206Cys dominated in the NCL2 cohort. This study provides an insight into the molecular pathology of NCL1 and NCL2 disease for Indian origin patients. Electronic supplementary material The online version of this article (10.1186/s12883-018-1206-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jayesh Sheth
- FRIGE's Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad, Gujarat, 380015, India.
| | - Mehul Mistri
- FRIGE's Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad, Gujarat, 380015, India
| | - Riddhi Bhavsar
- FRIGE's Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad, Gujarat, 380015, India
| | - Dhairya Pancholi
- FRIGE's Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad, Gujarat, 380015, India
| | - Mahesh Kamate
- Department of Pediatric Neurology, KLES Prabhakar Kore Hospital, Belgaum, Karnataka, 590010, India
| | - Neerja Gupta
- Division of Genetics (Pediatrics), All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Madhulika Kabra
- Division of Genetics (Pediatrics), All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sanjiv Mehta
- Usha-Deep Children Neurology and Epilepsy clinic, Ahmedabad, 380014, India
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Science and Research Centre, Kochi, Kerala, 682041, India
| | - Arpita Thakker
- Department of Neurology, Lokmanya Tilak Medical College, Sion Hospital, Mumbai, Maharashtra, 400022, India
| | - Vivek Jain
- Department of Neurology, Santokba Durlabhji Hospital, Jaipur, 302015, Rajasthan, India
| | - Raju Shah
- Ankur Neonatal Nursery, Ahmedabad, 380009, Gujarat, India
| | - Frenny Sheth
- FRIGE's Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad, Gujarat, 380015, India
| |
Collapse
|
74
|
Abstract
Viral vectors are a promising tool for effective delivery of genetic material into cells. They take advantage of the natural ability of a virus to deliver a genetic payload into cells while being genetically modified such that their ability to replicate is crippled or removed. Here, an updated overview of routinely used viral vectors, including adeno-associated viruses (AAV), retroviruses/lentiviruses, and adenoviruses (Ads), is provided, as well as perspectives on their advantages and disadvantages in research and gene therapy. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Yong Hong Chen
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Megan S Keiser
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Beverly L Davidson
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
75
|
Mitchell NL, Russell KN, Wellby MP, Wicky HE, Schoderboeck L, Barrell GK, Melzer TR, Gray SJ, Hughes SM, Palmer DN. Longitudinal In Vivo Monitoring of the CNS Demonstrates the Efficacy of Gene Therapy in a Sheep Model of CLN5 Batten Disease. Mol Ther 2018; 26:2366-2378. [PMID: 30078766 PMCID: PMC6171082 DOI: 10.1016/j.ymthe.2018.07.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 07/08/2018] [Accepted: 07/12/2018] [Indexed: 02/03/2023] Open
Abstract
Neuronal ceroid lipofuscinoses (NCLs; Batten disease) are neurodegenerative lysosomal storage diseases predominantly affecting children. Single administration of brain-directed lentiviral or recombinant single-stranded adeno-associated virus 9 (ssAAV9) vectors expressing ovine CLN5 into six pre-clinically affected sheep with a naturally occurring CLN5 NCL resulted in long-term disease attenuation. Treatment efficacy was demonstrated by non-invasive longitudinal in vivo monitoring developed to align with assessments used in human medicine. The treated sheep retained neurological and cognitive function, and one ssAAV9-treated animal has been retained and is now 57 months old, almost triple the lifespan of untreated CLN5-affected sheep. The onset of visual deficits was much delayed. Computed tomography and MRI showed that brain structures and volumes remained stable. Because gene therapy in humans is more likely to begin after clinical diagnosis, self-complementary AAV9-CLN5 was injected into the brain ventricles of four 7-month-old affected sheep already showing early clinical signs in a second trial. This also halted disease progression beyond their natural lifespan. These findings demonstrate the efficacy of CLN5 gene therapy, using three different vector platforms, in a large animal model and, thus, the prognosis for human translation.
Collapse
Affiliation(s)
- Nadia L Mitchell
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand; Department of Radiology, University of Otago, Christchurch 8140, New Zealand
| | - Katharina N Russell
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Martin P Wellby
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Hollie E Wicky
- Department of Biochemistry, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand
| | - Lucia Schoderboeck
- Department of Biochemistry, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand
| | - Graham K Barrell
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Tracy R Melzer
- Department of Medicine, University of Otago, Christchurch 8140, New Zealand
| | - Steven J Gray
- Gene Therapy Center and Department of Ophthalmology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Stephanie M Hughes
- Department of Biochemistry, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand
| | - David N Palmer
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand; Department of Radiology, University of Otago, Christchurch 8140, New Zealand.
| |
Collapse
|
76
|
Hudry E, Andres-Mateos E, Lerner EP, Volak A, Cohen O, Hyman BT, Maguire CA, Vandenberghe LH. Efficient Gene Transfer to the Central Nervous System by Single-Stranded Anc80L65. Mol Ther Methods Clin Dev 2018; 10:197-209. [PMID: 30109242 PMCID: PMC6083902 DOI: 10.1016/j.omtm.2018.07.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 07/10/2018] [Indexed: 12/27/2022]
Abstract
Adeno-associated viral vectors (AAVs) have demonstrated potential in applications for neurologic disorders, and the discovery that some AAVs can cross the blood-brain barrier (BBB) after intravenous injection has further expanded these opportunities for non-invasive brain delivery. Anc80L65, a novel AAV capsid designed from in silico reconstruction of the viral evolutionary lineage, has previously demonstrated robust transduction capabilities after local delivery in various tissues such as liver, retina, or cochlea, compared with conventional AAVs. Here, we compared the transduction efficacy of Anc80L65 with conventional AAV9 in the CNS after intravenous, intracerebroventricular (i.c.v.), or intraparenchymal injections. Anc80L65 was more potent at targeting the brain and spinal cord after intravenous injection than AAV9, and mostly transduced astrocytes and a wide range of neuronal subpopulations. Although the efficacy of Anc80L65 and AAV9 is similar after direct intraparenchymal injection in the striatum, Anc80L65's diffusion throughout the CNS was more extensive than AAV9 after i.c.v. infusion, leading to widespread EGFP expression in the cerebellum. These findings demonstrate that Anc80L65 is a highly efficient gene transfer vector for the murine CNS. Systemic injection of Anc80L65 leads to notable expression in the CNS that does not rely on a self-complementary genome. These data warrant further testing in larger animal models.
Collapse
Affiliation(s)
- Eloise Hudry
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Eva Andres-Mateos
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
- Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Eli P. Lerner
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Adrienn Volak
- Department of Neurology, The Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Olivia Cohen
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Bradley T. Hyman
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Casey A. Maguire
- Department of Neurology, The Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Luk H. Vandenberghe
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
- Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
77
|
Deverman BE, Ravina BM, Bankiewicz KS, Paul SM, Sah DWY. Gene therapy for neurological disorders: progress and prospects. Nat Rev Drug Discov 2018; 17:641-659. [DOI: 10.1038/nrd.2018.110] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
78
|
Attenuation of the Niemann-Pick type C2 disease phenotype by intracisternal administration of an AAVrh.10 vector expressing Npc2. Exp Neurol 2018; 306:22-33. [DOI: 10.1016/j.expneurol.2018.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/28/2018] [Accepted: 04/01/2018] [Indexed: 11/18/2022]
|
79
|
Lotfi P, Tse DY, Di Ronza A, Seymour ML, Martano G, Cooper JD, Pereira FA, Passafaro M, Wu SM, Sardiello M. Trehalose reduces retinal degeneration, neuroinflammation and storage burden caused by a lysosomal hydrolase deficiency. Autophagy 2018; 14:1419-1434. [PMID: 29916295 PMCID: PMC6103706 DOI: 10.1080/15548627.2018.1474313] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The accumulation of undegraded molecular material leads to progressive neurodegeneration in a number of lysosomal storage disorders (LSDs) that are caused by functional deficiencies of lysosomal hydrolases. To determine whether inducing macroautophagy/autophagy via small-molecule therapy would be effective for neuropathic LSDs due to enzyme deficiency, we treated a mouse model of mucopolysaccharidosis IIIB (MPS IIIB), a storage disorder caused by deficiency of the enzyme NAGLU (alpha-N-acetylglucosaminidase [Sanfilippo disease IIIB]), with the autophagy-inducing compound trehalose. Treated naglu–/ – mice lived longer, displayed less hyperactivity and anxiety, retained their vision (and retinal photoreceptors), and showed reduced inflammation in the brain and retina. Treated mice also showed improved clearance of autophagic vacuoles in neuronal and glial cells, accompanied by activation of the TFEB transcriptional network that controls lysosomal biogenesis and autophagic flux. Therefore, small-molecule-induced autophagy enhancement can improve the neurological symptoms associated with a lysosomal enzyme deficiency and could provide a viable therapeutic approach to neuropathic LSDs. Abbreviations: ANOVA: analysis of variance; Atg7: autophagy related 7; AV: autophagic vacuoles; CD68: cd68 antigen; ERG: electroretinogram; ERT: enzyme replacement therapy; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFAP: glial fibrillary acidic protein; GNAT2: guanine nucleotide binding protein, alpha transducing 2; HSCT: hematopoietic stem cell transplantation; INL: inner nuclear layer; LC3: microtubule-associated protein 1 light chain 3 alpha; MPS: mucopolysaccharidoses; NAGLU: alpha-N-acetylglucosaminidase (Sanfilippo disease IIIB); ONL: outer nuclear layer; PBS: phosphate-buffered saline; PRKCA/PKCα: protein kinase C, alpha; S1BF: somatosensory cortex; SQSTM1: sequestosome 1; TEM: transmission electron microscopy; TFEB: transcription factor EB; VMP/VPL: ventral posterior nuclei of the thalamus
Collapse
Affiliation(s)
- Parisa Lotfi
- a Department of Molecular and Human Genetics , Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital , Houston , TX , USA
| | - Dennis Y Tse
- b Department of Ophthalmology , Cullen Eye Institute, Baylor College of Medicine , Houston , TX , USA.,c School of Optometry , The Hong Kong Polytechnic University , Kowloon , Hong Kong
| | - Alberto Di Ronza
- a Department of Molecular and Human Genetics , Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital , Houston , TX , USA
| | - Michelle L Seymour
- d Huffington Center on Aging, Department of Molecular and Cellular Biology , Baylor College of Medicine , Houston , TX , USA.,e Department of Otolaryngology-Head & Neck Surgery , Baylor College of Medicine , Houston , TX , USA
| | | | - Jonathan D Cooper
- g Department of Basic and Clinical Neuroscience , Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience , Kings College London , London , UK.,h Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center , David Geffen School of Medicine, UCLA , Torrance , CA , USA
| | - Fred A Pereira
- d Huffington Center on Aging, Department of Molecular and Cellular Biology , Baylor College of Medicine , Houston , TX , USA.,e Department of Otolaryngology-Head & Neck Surgery , Baylor College of Medicine , Houston , TX , USA
| | | | - Samuel M Wu
- b Department of Ophthalmology , Cullen Eye Institute, Baylor College of Medicine , Houston , TX , USA
| | - Marco Sardiello
- a Department of Molecular and Human Genetics , Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital , Houston , TX , USA
| |
Collapse
|
80
|
Donsante A, Boulis NM. Progress in gene and cell therapies for the neuronal ceroid lipofuscinoses. Expert Opin Biol Ther 2018; 18:755-764. [PMID: 29936867 DOI: 10.1080/14712598.2018.1492544] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The neuronal ceroid lipofuscinoses (NCLs) are a subset of lysosomal storage diseases (LSDs) that cause myoclonic epilepsy, loss of cognitive and motor function, degeneration of the retina leading to blindness, and early death. Most are caused by loss-of-function mutations in either lysosomal proteins or transmembrane proteins. Current therapies are supportive in nature. NCLs involving lysosomal enzymes are amenable to therapies that provide an exogenous source of protein, as has been used for other LSDs. Those that involve transmembrane proteins, however, require new approaches. AREAS COVERED This review will discuss potential gene and cell therapy approaches that have been, are, or may be in development for these disorders and those that have entered clinical trials. EXPERT OPINION In animal models, gene therapy approaches have produced remarkable improvements in neurological function and lifespan. However, a complete cure has not been reached for any NCL, and a better understanding of the limits of the current crop of vectors is needed to more fully address these diseases. The prospects for gene therapy, particularly those that can be delivered systemically and treat both the brain and peripheral tissue, are high. The future is beginning to look bright for NCL patients and their families.
Collapse
Affiliation(s)
- Anthony Donsante
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| | - Nicholas M Boulis
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| |
Collapse
|
81
|
Itagaki R, Endo M, Yanagisawa H, Hossain MA, Akiyama K, Yaginuma K, Miyajima T, Wu C, Iwamoto T, Igarashi J, Kobayashi Y, Tohyama J, Iwama K, Matsumoto N, Shintaku H, Eto Y. Characteristics of PPT1 and TPP1 enzymes in neuronal ceroid lipofuscinosis (NCL) 1 and 2 by dried blood spots (DBS) and leukocytes and their application to newborn screening. Mol Genet Metab 2018; 124:64-70. [PMID: 29599076 DOI: 10.1016/j.ymgme.2018.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 03/17/2018] [Indexed: 10/17/2022]
Abstract
We first characterized PPT1 and TPP1 enzymes in dried blood spots (DBS), plasma/serum, and leukocytes/lymphocytes using neuronal ceroid lipofuscinosis (NCL) 1 and 2 patients and control subjects. PPT1 enzyme had only one acid form in control DBS, plasma/serum, and leukocytes/lymphocytes and showed deficient activities in these samples from NCL 1 patients. Conversely, TPP1 enzymes in control DBS and leukocytes/lymphocytes consisted of two forms, an acidic form and a neutral form, whereas serum TPP1 enzyme had only a neutral form. In control subjects, the optimal pH of PPT1 enzyme in DBS, plasma/serum, and leukocytes/lymphocytes was 4.5 to 5.0 in the acidic form, whereas TPP1 enzyme in control DBS and leukocytes/lymphocytes was pH 4.5 and 6.5, respectively. In NCL 1 and 2, both PPT1 and TPP1 enzyme activities in DBS, plasma, and leukocytes/lymphocytes were markedly reduced in acidic pH, whereas heterozygotes of NCL 1 and 2 in the acidic form showed intermediate activities between patients and control subjects. In neutral conditions, pH 6.0, the PPT1 enzyme activities in NCL 1 patients showed rather higher residual activities and intermediate activities in heterozygotes in NCL 1, which was probably caused by mutated proteins in three cases with NCL 1 patients. TPP1 enzyme activities at neutral pH 6.5 to 7.0 in DBS and leukocytes/lymphocytes showed higher enzyme activities in NCL 2 patients and heterozygotes. The reason for the increases of neutral TPP1 enzyme activities at pH 6.5 to 7.0 in NCL 2 DBS and leukocytes/lymphocytes, is obscure, but possibly caused by secondary activation of neutral TPP1 enzyme due to the absence of the acidic form. Interestingly, TPP1 activity in serum only consisted of a neutral form, no acidic form, and was not deficient in any NCL 2 patient. Therefore, we can diagnose NCL 1 patients by plasma/serum enzyme assay of PPT1, but not diagnose NCL 2 by serum TPP1 enzyme assay. A pilot study of newborn screening of NCL 1 and 2 has been established by more than 1000 newborn DBS assays. Using this assay system, we will be able to perform newborn screening of NCL 1 and 2 by DBS.
Collapse
Affiliation(s)
- Rina Itagaki
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan
| | - Masahiro Endo
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan
| | - Hiroko Yanagisawa
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan
| | - Mohammad Arif Hossain
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan
| | - Keiko Akiyama
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan
| | - Keiko Yaginuma
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan
| | - Takashi Miyajima
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan; Institute of Rare disease, AnGes Co., Tokyo, Japan
| | - Chen Wu
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan; Institute of Rare disease, AnGes Co., Tokyo, Japan
| | - Takeo Iwamoto
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan; Core Laboratory, Institute of Medical Science, Tokyo Jikei University School of Medicine, Tokyo, Japan
| | | | - Yu Kobayashi
- Department of Child Neurology, Epilepsy Center, Nishi-Niigata, Chuo National Hospital, Niigata, Japan
| | - Jun Tohyama
- Department of Child Neurology, Epilepsy Center, Nishi-Niigata, Chuo National Hospital, Niigata, Japan
| | - Kazuhiro Iwama
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Institute of Neurological Disorder, Kanagawa, Japan; Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
82
|
Rosenberg JB, Kaplitt MG, De BP, Chen A, Flagiello T, Salami C, Pey E, Zhao L, Ricart Arbona RJ, Monette S, Dyke JP, Ballon DJ, Kaminsky SM, Sondhi D, Petsko GA, Paul SM, Crystal RG. AAVrh.10-Mediated APOE2 Central Nervous System Gene Therapy for APOE4-Associated Alzheimer's Disease. HUM GENE THER CL DEV 2018; 29:24-47. [PMID: 29409358 DOI: 10.1089/humc.2017.231] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive degenerative neurological disorder affecting nearly one in nine elderly people in the United States. Population studies have shown that an inheritance of the apolipoprotein E (APOE) variant APOE4 allele increases the risk of developing AD, whereas APOE2 homozygotes are protected from late-onset AD. It was hypothesized that expression of the "protective" APOE2 variant by genetic modification of the central nervous system (CNS) of APOE4 homozygotes could reverse or prevent progressive neurologic damage. To assess the CNS distribution and safety of APOE2 gene therapy for AD in a large-animal model, intraparenchymal, intracisternal, and intraventricular routes of delivery to the CNS of nonhuman primates of AAVrh.10hAPOE2-HA, an AAVrh.10 serotype coding for an HA-tagged human APOE2 cDNA sequence, were evaluated. To evaluate the route of delivery that achieves the widest extent of APOE2 expression in the CNS, the expression of APOE2 in the CNS was evaluated 2 months following vector administration for APOE2 DNA, mRNA, and protein. Finally, using conventional toxicology assays, the safety of the best route of delivery was assessed. The data demonstrated that while all three routes are capable of mediating ApoE2 expression in AD relevant regions, intracisternal delivery of AAVrh.10hAPOE2-HA safely mediated wide distribution of ApoE2 with the least invasive surgical intervention, thus providing the optimal strategy to deliver vector-mediated human APOE2 to the CNS.
Collapse
Affiliation(s)
- Jonathan B Rosenberg
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Michael G Kaplitt
- 2 Department of Neurosurgery, Weill Cornell Medical College , New York, New York
| | - Bishnu P De
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Alvin Chen
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Thomas Flagiello
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Christiana Salami
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Eduard Pey
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Lingzhi Zhao
- 3 Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College , New York, New York
| | - Rodolfo J Ricart Arbona
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sebastien Monette
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University , Weill Cornell Medical College, New York, New York
| | - Jonathan P Dyke
- 6 Department of Radiology, Weill Cornell Medical College , New York, New York
| | - Douglas J Ballon
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York.,6 Department of Radiology, Weill Cornell Medical College , New York, New York
| | - Stephen M Kaminsky
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Dolan Sondhi
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Gregory A Petsko
- 3 Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College , New York, New York
| | - Steven M Paul
- 7 Voyager Therapeutics, Inc. , Cambridge, Massachusetts
| | - Ronald G Crystal
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| |
Collapse
|
83
|
Chen Y, Zheng S, Tecedor L, Davidson BL. Overcoming Limitations Inherent in Sulfamidase to Improve Mucopolysaccharidosis IIIA Gene Therapy. Mol Ther 2018; 26:1118-1126. [PMID: 29503202 PMCID: PMC6079371 DOI: 10.1016/j.ymthe.2018.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 01/09/2018] [Accepted: 01/12/2018] [Indexed: 11/18/2022] Open
Abstract
Sulfamidase (SGSH) deficiency causes mucopolysaccharidosis type IIIA (MPS IIIA), a lysosomal storage disease (LSD) that affects the CNS. In earlier work in LSD mice and dog models, we exploited the utility of adeno-associated viruses (AAVs) to transduce brain ventricular lining cells (ependyma) for secretion of lysosomal hydrolases into the cerebrospinal fluid (CSF), with subsequent distribution of enzyme throughout the brain resulting in improved cognition and extending lifespan. A critical feature of this approach is efficient secretion of the expressed enzyme from transduced cells, for delivery by CSF to nontransduced cells. Surprisingly, we found that SGSH was poorly secreted from cells, resulting in retention of the expressed product. Using site-directed mutagenesis of native SGSH, we identified an improved secretion variant that also displayed enhanced uptake properties that were mannose-6-phosphate receptor independent. In studies in MPS IIIA-deficient mice, ependymal transduction with AAVs expressing variant SGSH improved spatial learning and reduced memory deficits, substrate accumulation, and astrogliosis. Secondary lysosomal enzyme elevations in the CSF and brain parenchyma were also resolved. In contrast, ependymal transduction with AAVs expressing wild-type SGSH had significantly lower CSF SGSH levels and limited impacts on behavior. These results demonstrate the utility of a previously undescribed SGSH variant for improved MPS IIIA brain gene therapy.
Collapse
Affiliation(s)
- Yonghong Chen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Shujuan Zheng
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Luis Tecedor
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
84
|
Progranulin Gene Therapy Improves Lysosomal Dysfunction and Microglial Pathology Associated with Frontotemporal Dementia and Neuronal Ceroid Lipofuscinosis. J Neurosci 2018; 38:2341-2358. [PMID: 29378861 DOI: 10.1523/jneurosci.3081-17.2018] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/27/2017] [Accepted: 01/20/2018] [Indexed: 01/18/2023] Open
Abstract
Loss-of-function mutations in progranulin, a lysosomal glycoprotein, cause neurodegenerative disease. Progranulin haploinsufficiency causes frontotemporal dementia (FTD) and complete progranulin deficiency causes CLN11 neuronal ceroid lipofuscinosis (NCL). Progranulin replacement is a rational therapeutic strategy for these disorders, but there are critical unresolved mechanistic questions about a progranulin gene therapy approach, including its potential to reverse existing pathology. Here, we address these issues using an AAV vector (AAV-Grn) to deliver progranulin in Grn-/- mice (both male and female), which model aspects of NCL and FTD pathology, developing lysosomal dysfunction, lipofuscinosis, and microgliosis. We first tested whether AAV-Grn could improve preexisting pathology. Even with treatment after onset of pathology, AAV-Grn reduced lipofuscinosis in several brain regions of Grn-/- mice. AAV-Grn also reduced microgliosis in brain regions distant from the injection site. AAV-expressed progranulin was only detected in neurons, not in microglia, indicating that the microglial activation in progranulin deficiency can be improved by targeting neurons and thus may be driven at least in part by neuronal dysfunction. Even areas with sparse transduction and almost undetectable progranulin showed improvement, indicating that low-level replacement may be sufficiently effective. The beneficial effects of AAV-Grn did not require progranulin binding to sortilin. Finally, we tested whether AAV-Grn improved lysosomal function. AAV-derived progranulin was delivered to the lysosome, ameliorated the accumulation of LAMP-1 in Grn-/- mice, and corrected abnormal cathepsin D activity. These data shed light on progranulin biology and support progranulin-boosting therapies for NCL and FTD due to GRN mutations.SIGNIFICANCE STATEMENT Heterozygous loss-of-function progranulin (GRN) mutations cause frontotemporal dementia (FTD) and homozygous mutations cause neuronal ceroid lipofuscinosis (NCL). Here, we address several mechanistic questions about the potential of progranulin gene therapy for these disorders. GRN mutation carriers with NCL or FTD exhibit lipofuscinosis and Grn-/- mouse models develop a similar pathology. AAV-mediated progranulin delivery reduced lipofuscinosis in Grn-/- mice even after the onset of pathology. AAV delivered progranulin only to neurons, not microglia, but improved microgliosis in several brain regions, indicating cross talk between neuronal and microglial pathology. Its beneficial effects were sortilin independent. AAV-derived progranulin was delivered to lysosomes and corrected lysosomal abnormalities. These data provide in vivo support for the efficacy of progranulin-boosting therapies for FTD and NCL.
Collapse
|
85
|
Gene Therapy Approaches to Treat the Neurodegeneration and Visual Failure in Neuronal Ceroid Lipofuscinoses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1074:91-99. [PMID: 29721932 DOI: 10.1007/978-3-319-75402-4_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are a group of fatal, inherited lysosomal storage disorders mostly affecting the central nervous system of children. Symptoms include vision loss, seizures, motor deterioration and cognitive decline ultimately resulting in premature death. Studies in animal models showed that the diseases are amenable to gene supplementation therapies, and over the last decade, major advances have been made in the (pre)clinical development of these therapies. This mini-review summarises and discusses current gene therapy approaches for NCL targeting the brain and the eye.
Collapse
|
86
|
Ricca A, Gritti A. Perspective on innovative therapies for globoid cell leukodystrophy. J Neurosci Res 2017; 94:1304-17. [PMID: 27638612 DOI: 10.1002/jnr.23752] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/25/2016] [Accepted: 03/30/2016] [Indexed: 12/24/2022]
Abstract
Globoid cell leukodystrophy (GLD), or Krabbe's disease, is a lysosomal storage disorder resulting from deficiency of the lysosomal hydrolase galactosylceramidase. The infantile forms are characterized by a unique relentless and aggressive progression with a wide range of neurological symptoms and complications. Here we review and discuss the basic concepts and the novel mechanisms identified as key contributors to the peculiar GLD pathology, highlighting their therapeutic implications. Then, we evaluate evidence from extensive experimental studies on GLD animal models that have highlighted fundamental requirements to obtain substantial therapeutic benefit, including early and timely intervention, high levels of enzymatic reconstitution, and global targeting of affected tissues. Continuous efforts in understanding GLD pathophysiology, the interplay between various therapies, and the mechanisms of disease correction upon intervention may allow advancing research with innovative approaches and prioritizing treatment strategies to develop more efficacious treatments. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alessandra Ricca
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
87
|
Suzuki JI, Dezawa M, Kitada M. Prolonged but non-permanent expression of a transgene in ependymal cells of adult rats using an adenovirus-mediated transposon gene transfer system. Brain Res 2017; 1675:20-27. [DOI: 10.1016/j.brainres.2017.08.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 01/08/2023]
|
88
|
Giugliani R, Vairo F, Kubaski F, Poswar F, Riegel M, Baldo G, Saute JA. Neurological manifestations of lysosomal disorders and emerging therapies targeting the CNS. THE LANCET CHILD & ADOLESCENT HEALTH 2017; 2:56-68. [PMID: 30169196 DOI: 10.1016/s2352-4642(17)30087-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/01/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022]
Abstract
Lysosomal disorders have been an area of interest since intravenous enzyme replacement therapy was successfully introduced for the treatment of Gaucher's disease in the early 1990s. This treatment approach has also been developed for several other lysosomal disorders, including Fabry's disease, Pompe's disease, lysosomal acid lipase deficiency, and five types of mucopolysaccharidosis. Despite the benefits of enzyme replacement therapy, it has limitations-most importantly, its ineffectiveness in treating the neurological components of lysosomal disorders, as only a small proportion of recombinant enzymes can cross the blood-brain barrier. Development of strategies to improve drug delivery to the CNS is now the primary focus in lysosomal disorder research. This Review discusses the neurological manifestations and emerging therapies for the CNS component of these diseases. The therapies in development (which are now in phase 1 or phase 2 clinical trials) might be for specific lysosomal disorders (enzyme replacement therapy via intrathecal or intracerebroventricular routes or with fusion proteins, or gene therapy) or applicable to more than one lysosomal disorder (haemopoietic stem cell transplantation, pharmacological chaperones, substrate reduction therapy, or stop codon readthrough). The combination of early diagnosis with effective therapies should change the outlook for patients with lysosomal disorders with neurological involvement in the next 5-10 years.
Collapse
Affiliation(s)
- Roberto Giugliani
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | | | | | - Fabiano Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mariluce Riegel
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Baldo
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Postgraduate Program in Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jonas Alex Saute
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
89
|
Katz ML, Rustad E, Robinson GO, Whiting REH, Student JT, Coates JR, Narfstrom K. Canine neuronal ceroid lipofuscinoses: Promising models for preclinical testing of therapeutic interventions. Neurobiol Dis 2017; 108:277-287. [PMID: 28860089 DOI: 10.1016/j.nbd.2017.08.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/26/2017] [Indexed: 10/19/2022] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are devastating inherited progressive neurodegenerative diseases, with most forms having a childhood onset of clinical signs. The NCLs are characterized by progressive cognitive and motor decline, vision loss, seizures, respiratory and swallowing impairment, and ultimately premature death. Different forms of NCL result from mutations in at least 13 genes. The clinical signs of some forms overlap significantly, so genetic testing is the only way to definitively determine which form an individual patient suffers from. At present, an effective treatment is available for only one form of NCL. Evidence of NCL has been documented in over 20 canine breeds and in mixed-breed dogs. To date, 12 mutations in 8 different genes orthologous to the human NCL genes have been found to underlie NCL in a variety of dog breeds. A Dachshund model with a null mutation in one of these genes is being utilized to investigate potential therapeutic interventions, including enzyme replacement and gene therapies. Demonstration of the efficacy of enzyme replacement therapy in this model led to successful completion of human clinical trials of this treatment. Further research into the other canine NCLs, with in-depth characterization and understanding of the disease processes, will likely lead to the development of successful therapeutic interventions for additional forms of NCL, for both human patients and animals with these disorders.
Collapse
Affiliation(s)
- Martin L Katz
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| | - Eline Rustad
- Blue Star Animal Hospital, Göteborg 417 07, Sweden
| | - Grace O Robinson
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Rebecca E H Whiting
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Jeffrey T Student
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Joan R Coates
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Kristina Narfstrom
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
90
|
Nelvagal HR, Cooper JD. Translating preclinical models of neuronal ceroid lipofuscinosis: progress and prospects. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1360182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Hemanth R. Nelvagal
- Pediatric Storage Disorders Laboratory, Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, David Geffen School of Medicine, UCLA, Torrance, CA, USA
| | - Jonathan D. Cooper
- Pediatric Storage Disorders Laboratory, Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, David Geffen School of Medicine, UCLA, Torrance, CA, USA
| |
Collapse
|
91
|
Cooper JD, Nelvagal HR. Progress toward Fulfilling the Potential of Immunomodulation in Childhood Neurodegeneration? Mol Ther 2017. [PMID: 28625570 DOI: 10.1016/j.ymthe.2017.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Jonathan D Cooper
- Department of Pediatrics, Los Angeles Biomedical Research Institute, David Geffen School of Medicine, UCLA, 1124 W Carson St., Torrance, CA 90502, USA.
| | - Hemanth R Nelvagal
- Department of Pediatrics, Los Angeles Biomedical Research Institute, David Geffen School of Medicine, UCLA, 1124 W Carson St., Torrance, CA 90502, USA
| |
Collapse
|
92
|
Wegmann S, Bennett RE, Amaral AS, Hyman BT. Studying tau protein propagation and pathology in the mouse brain using adeno-associated viruses. Methods Cell Biol 2017; 141:307-322. [PMID: 28882310 DOI: 10.1016/bs.mcb.2017.06.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The progressive spread of pathological brain lesions containing aggregated tau protein is a hallmark of Alzheimer's disease and other neurodegenerative diseases. In AD, this process follows a distinct pattern along neuronal connections from the entorhinal cortex to hippocampal areas and further on through the limbic system. In other tauopathies, the spread of tau appears less hierarchical throughout the brain, and also nonpathological tau is reported to cross-synaptic connections in the brain. To be able to study the process of cell-to-cell transport of tau and the associated neurotoxicity in the brain in vivo, adeno-associated virus-mediated expression of tau can be used to express different forms of tau in distinct brain areas in rodent models. As an example, we describe how the expression of FTD-mutant human tauP301L in the entorhinal cortex of wild-type mice can be used to study the propagation of tau to connected neurons and to determine pathological consequences such as tau hyperphosphorylation, misfolding, and gliosis. The approach described can easily be translated to study other aggregating and/or propagating proteins in the brain such as synuclein, Abeta, or SOD1.
Collapse
Affiliation(s)
- Susanne Wegmann
- Massachusetts General Hospital, Harvard Medical School, Mass. Institute for Neurodegenerative Diseases (MIND), Boston, MA, United States.
| | - Rachel E Bennett
- Massachusetts General Hospital, Harvard Medical School, Mass. Institute for Neurodegenerative Diseases (MIND), Boston, MA, United States
| | - Ana S Amaral
- Massachusetts General Hospital, Harvard Medical School, Mass. Institute for Neurodegenerative Diseases (MIND), Boston, MA, United States
| | - Bradley T Hyman
- Massachusetts General Hospital, Harvard Medical School, Mass. Institute for Neurodegenerative Diseases (MIND), Boston, MA, United States
| |
Collapse
|
93
|
Synergistic effects of treating the spinal cord and brain in CLN1 disease. Proc Natl Acad Sci U S A 2017; 114:E5920-E5929. [PMID: 28673981 DOI: 10.1073/pnas.1701832114] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Infantile neuronal ceroid lipofuscinosis (INCL, or CLN1 disease) is an inherited neurodegenerative storage disorder caused by a deficiency of the lysosomal enzyme palmitoyl protein thioesterase 1 (PPT1). It was widely believed that the pathology associated with INCL was limited to the brain, but we have now found unexpectedly profound pathology in the human INCL spinal cord. Similar pathological changes also occur at every level of the spinal cord of PPT1-deficient (Ppt1-/- ) mice before the onset of neuropathology in the brain. Various forebrain-directed gene therapy approaches have only had limited success in Ppt1-/- mice. Targeting the spinal cord via intrathecal administration of an adeno-associated virus (AAV) gene transfer vector significantly prevented pathology and produced significant improvements in life span and motor function in Ppt1-/- mice. Surprisingly, forebrain-directed gene therapy resulted in essentially no PPT1 activity in the spinal cord, and vice versa. This leads to a reciprocal pattern of histological correction in the respective tissues when comparing intracranial with intrathecal injections. However, the characteristic pathological features of INCL were almost completely absent in both the brain and spinal cord when intracranial and intrathecal injections of the same AAV vector were combined. Targeting both the brain and spinal cord also produced dramatic and synergistic improvements in motor function with an unprecedented increase in life span. These data show that spinal cord pathology significantly contributes to the clinical progression of INCL and can be effectively targeted therapeutically. This has important implications for the delivery of therapies in INCL, and potentially in other similar disorders.
Collapse
|
94
|
Kim K, Kleinman HK, Lee HJ, Pahan K. Safety and potential efficacy of gemfibrozil as a supportive treatment for children with late infantile neuronal ceroid lipofuscinosis and other lipid storage disorders. Orphanet J Rare Dis 2017. [PMID: 28623936 PMCID: PMC5474050 DOI: 10.1186/s13023-017-0663-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neuronal Ceroid Lipofuscinosis (NCL), also known as Batten disease, is a group of genetically distinct lysosomal disorders that mainly affect the central nervous system, resulting in progressive motor and cognitive decline primarily in children. Multiple distinct genes involved in the metabolism of lipids have been identified to date with various mutations in this family of diseases. There is no cure for these diseases but some new therapeutic approaches have been tested that offer more hope than the standard palliative care. Many of the therapeutic advances require invasive procedures but some progress in slowing the disease has been found and more options can be expected in the future. We also review the literature on children with disease/conditions other than NCL for the non-invasive use, safety, and tolerability of a lipid-lowering drug, gemfibrozil, as a potential treatment for NCLs. Gemfibrozil has shown efficacy in an animal model of NCL known as CLN2 (late infantile classic juvenile) and has been shown to be safe for lowering lipids in children. Among the 200 non-NCL children found in the published literature who were treated with gemfibrozil for NCL-related problems, only 3 experienced adverse events, including 2 with muscle pain and 1 with localized linear IgA bullous dermatitis. We conclude that gemfibrozil is safe for long-term use in children, causes minimal adverse events, is well tolerated, and may delay the progression of NCLs. Gemfibrozil may potentially be an alternative to more invasive therapeutic approaches currently under investigation and has the potential to be used in combination with other therapeutic approaches.
Collapse
Affiliation(s)
- Kyeongsoon Kim
- Department of Pharmaceutical Engineering, Inje University, Gimhae, South Korea
| | - Hynda K Kleinman
- Polaryx Therapeutics Inc., Paramus, NJ, USA. .,The George Washington University Medical Center, Washington, DC, USA.
| | | | | |
Collapse
|
95
|
Groh J, Martini R. Neuroinflammation as modifier of genetically caused neurological disorders of the central nervous system: Understanding pathogenesis and chances for treatment. Glia 2017; 65:1407-1422. [PMID: 28568966 DOI: 10.1002/glia.23162] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/10/2017] [Accepted: 04/18/2017] [Indexed: 12/21/2022]
Abstract
Genetically caused neurological disorders of the central nervous system (CNS) are usually orphan diseases with poor or even fatal clinical outcome and few or no treatments that will improve longevity or at least quality of life. Neuroinflammation is common to many of these disorders, despite the fact that a plethora of distinct mutations and molecular changes underlie the disorders. In this article, data from corresponding animal models are analyzed to define the roles of innate and adaptive inflammation as modifiers and amplifiers of disease. We describe both common and distinct patterns of neuroinflammation in genetically mediated CNS disorders and discuss the contrasting mechanisms that lead to adverse versus neuroprotective effects. Moreover, we identify the juxtaparanode as a neuroanatomical compartment commonly associated with inflammatory cells and ongoing axonopathic changes, in models of diverse diseases. The identification of key immunological effector pathways that amplify neuropathic features should lead to realistic possibilities for translatable therapeutic interventions using existing immunomodulators. Moreover, evidence emerges that neuroinflammation is not only able to modify primary neural damage-related symptoms but also may lead to unexpected clinical outcomes such as neuropsychiatric syndromes.
Collapse
Affiliation(s)
- Janos Groh
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Josef-Schneider-Str. 11, Würzburg, D-97080, Germany
| | - Rudolf Martini
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Josef-Schneider-Str. 11, Würzburg, D-97080, Germany
| |
Collapse
|
96
|
Fingolimod and Teriflunomide Attenuate Neurodegeneration in Mouse Models of Neuronal Ceroid Lipofuscinosis. Mol Ther 2017; 25:1889-1899. [PMID: 28506594 PMCID: PMC5542710 DOI: 10.1016/j.ymthe.2017.04.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 12/17/2022] Open
Abstract
CLN diseases are rare lysosomal storage diseases characterized by progressive axonal degeneration and neuron loss in the CNS, manifesting in disability, blindness, and premature death. We have previously demonstrated that, in animal models of infantile and juvenile forms of CLN disease (CLN1 and CLN3, respectively), secondary neuroinflammation in the CNS substantially amplifies neural damage, opening the possibility that immunomodulatory treatment might improve disease outcome. First, we recapitulated the inflammatory phenotype, originally seen in mice in autopsies of CLN patients. We then treated mouse models of CLN1 and CLN3 disease with the clinically approved immunomodulatory compounds fingolimod (0.5 mg/kg/day) and teriflunomide (10 mg/kg/day) by consistent supply in the drinking water for 5 months. The treatment was well tolerated and reduced T cell numbers and microgliosis in the CNS of both models. Moreover, axonal damage, neuron loss, retinal thinning, and brain atrophy were substantially attenuated in both models, along with reduced frequency of myoclonic jerks in Ppt1−/− mice. Based on these findings, and because side effects were not detected, we suggest that clinically approved immune modulators such as fingolimod and teriflunomide may be suitable to attenuate progression of CLN1 and CLN3 disease and, possibly, other orphan diseases with pathogenically relevant neuroinflammation.
Collapse
|
97
|
Best HL, Neverman NJ, Wicky HE, Mitchell NL, Leitch B, Hughes SM. Characterisation of early changes in ovine CLN5 and CLN6 Batten disease neural cultures for the rapid screening of therapeutics. Neurobiol Dis 2017; 100:62-74. [DOI: 10.1016/j.nbd.2017.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/19/2016] [Accepted: 01/01/2017] [Indexed: 01/12/2023] Open
|
98
|
Extraneuronal pathology in a canine model of CLN2 neuronal ceroid lipofuscinosis after intracerebroventricular gene therapy that delays neurological disease progression. Gene Ther 2017; 24:215-223. [PMID: 28079862 PMCID: PMC5398942 DOI: 10.1038/gt.2017.4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/23/2016] [Accepted: 01/03/2017] [Indexed: 01/04/2023]
Abstract
CLN2 neuronal ceroid lipofuscinosis is a hereditary lysosomal storage disease with primarily neurological signs that results from mutations in TPP1, which encodes the lysosomal enzyme tripeptidyl peptidase-1 (TPP1). Studies using a canine model for this disorder demonstrated that delivery of TPP1 enzyme to the cerebrospinal fluid (CSF) by intracerebroventricular administration of an AAV-TPP1 vector resulted in substantial delays in the onset and progression of neurological signs and prolongation of life span. We hypothesized that the treatment may not deliver therapeutic levels of this protein to tissues outside the central nervous system that also require TPP1 for normal lysosomal function. To test this hypothesis, dogs treated with CSF administration of AAV-TPP1 were evaluated for the development of non-neuronal pathology. Affected treated dogs exhibited progressive cardiac pathology reflected by elevated plasma cardiac troponin-1, impaired cardiac function and development of histopathological myocardial lesions. Progressive increases in the plasma activity levels of alanine aminotransferase and creatine kinase indicated development of pathology in the liver and muscles. The treatment also did not prevent disease-related accumulation of lysosomal storage bodies in the heart or liver. These studies indicate that optimal treatment outcomes for CLN2 disease may require delivery of TPP1 systemically as well as directly to the central nervous system.
Collapse
|
99
|
Golebiowski D, van der Bom IMJ, Kwon CS, Miller AD, Petrosky K, Bradbury AM, Maitland S, Kühn AL, Bishop N, Curran E, Silva N, GuhaSarkar D, Westmoreland SV, Martin DR, Gounis MJ, Asaad WF, Sena-Esteves M. Direct Intracranial Injection of AAVrh8 Encoding Monkey β-N-Acetylhexosaminidase Causes Neurotoxicity in the Primate Brain. Hum Gene Ther 2017; 28:510-522. [PMID: 28132521 DOI: 10.1089/hum.2016.109] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
GM2 gangliosidoses, including Tay-Sachs disease and Sandhoff disease, are lysosomal storage disorders caused by deficiencies in β-N-acetylhexosaminidase (Hex). Patients are afflicted primarily with progressive central nervous system (CNS) dysfunction. Studies in mice, cats, and sheep have indicated safety and widespread distribution of Hex in the CNS after intracranial vector infusion of AAVrh8 vectors encoding species-specific Hex α- or β-subunits at a 1:1 ratio. Here, a safety study was conducted in cynomolgus macaques (cm), modeling previous animal studies, with bilateral infusion in the thalamus as well as in left lateral ventricle of AAVrh8 vectors encoding cm Hex α- and β-subunits. Three doses (3.2 × 1012 vg [n = 3]; 3.2 × 1011 vg [n = 2]; or 1.1 × 1011 vg [n = 2]) were tested, with controls infused with vehicle (n = 1) or transgene empty AAVrh8 vector at the highest dose (n = 2). Most monkeys receiving AAVrh8-cmHexα/β developed dyskinesias, ataxia, and loss of dexterity, with higher dose animals eventually becoming apathetic. Time to onset of symptoms was dose dependent, with the highest-dose cohort producing symptoms within a month of infusion. One monkey in the lowest-dose cohort was behaviorally asymptomatic but had magnetic resonance imaging abnormalities in the thalami. Histopathology was similar in all monkeys injected with AAVrh8-cmHexα/β, showing severe white and gray matter necrosis along the injection track, reactive vasculature, and the presence of neurons with granular eosinophilic material. Lesions were minimal to absent in both control cohorts. Despite cellular loss, a dramatic increase in Hex activity was measured in the thalamus, and none of the animals presented with antibody titers against Hex. The high overexpression of Hex protein is likely to blame for this negative outcome, and this study demonstrates the variations in safety profiles of AAVrh8-Hexα/β intracranial injection among different species, despite encoding for self-proteins.
Collapse
Affiliation(s)
- Diane Golebiowski
- 1 Department of Neurology, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Imramsjah M J van der Bom
- 3 Department of Radiology, University of Massachusetts Medical School , Worcester, Massachusetts.,4 New England Center for Stroke Research, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Churl-Su Kwon
- 5 Department of Neurosurgery, Massachusetts General Hospital , Boston, Massachusetts
| | - Andrew D Miller
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Keiko Petrosky
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Allison M Bradbury
- 7 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University , Alabama.,8 Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University , Alabama
| | - Stacy Maitland
- 1 Department of Neurology, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Anna Luisa Kühn
- 3 Department of Radiology, University of Massachusetts Medical School , Worcester, Massachusetts.,4 New England Center for Stroke Research, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Nina Bishop
- 9 Department of Animal Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Elizabeth Curran
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Nilsa Silva
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Dwijit GuhaSarkar
- 1 Department of Neurology, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Susan V Westmoreland
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Douglas R Martin
- 7 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University , Alabama.,8 Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University , Alabama
| | - Matthew J Gounis
- 3 Department of Radiology, University of Massachusetts Medical School , Worcester, Massachusetts.,4 New England Center for Stroke Research, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Wael F Asaad
- 10 Department of Neurosurgery, Alpert Medical School, Brown University , Providence, Rhode Island.,11 Brown Institute for Brain Science, Brown University , Providence, Rhode Island.,12 Rhode Island Hospital , Providence, Rhode Island
| | - Miguel Sena-Esteves
- 1 Department of Neurology, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
100
|
Kothari P, De BP, He B, Chen A, Chiuchiolo MJ, Kim D, Nikolopoulou A, Amor-Coarasa A, Dyke JP, Voss HU, Kaminsky SM, Foley CP, Vallabhajosula S, Hu B, DiMagno SG, Sondhi D, Crystal RG, Babich JW, Ballon D. Radioiodinated Capsids Facilitate In Vivo Non-Invasive Tracking of Adeno-Associated Gene Transfer Vectors. Sci Rep 2017; 7:39594. [PMID: 28059103 PMCID: PMC5216390 DOI: 10.1038/srep39594] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 11/24/2016] [Indexed: 01/07/2023] Open
Abstract
Viral vector mediated gene therapy has become commonplace in clinical trials for a wide range of inherited disorders. Successful gene transfer depends on a number of factors, of which tissue tropism is among the most important. To date, definitive mapping of the spatial and temporal distribution of viral vectors in vivo has generally required postmortem examination of tissue. Here we present two methods for radiolabeling adeno-associated virus (AAV), one of the most commonly used viral vectors for gene therapy trials, and demonstrate their potential usefulness in the development of surrogate markers for vector delivery during the first week after administration. Specifically, we labeled adeno-associated virus serotype 10 expressing the coding sequences for the CLN2 gene implicated in late infantile neuronal ceroid lipofuscinosis with iodine-124. Using direct (Iodogen) and indirect (modified Bolton-Hunter) methods, we observed the vector in the murine brain for up to one week using positron emission tomography. Capsid radioiodination of viral vectors enables non-invasive, whole body, in vivo evaluation of spatial and temporal vector distribution that should inform methods for efficacious gene therapy over a broad range of applications.
Collapse
Affiliation(s)
- P. Kothari
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - B. P. De
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - B. He
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - A. Chen
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - M. J. Chiuchiolo
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - D. Kim
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - A. Nikolopoulou
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - A. Amor-Coarasa
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - J. P. Dyke
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - H. U. Voss
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - S. M. Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - C. P. Foley
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - S. Vallabhajosula
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - B. Hu
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - S. G. DiMagno
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - D. Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - R. G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - J. W. Babich
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - D. Ballon
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medical College, New York, New York, USA
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|