1
|
Tecedor L, Chen YH, Leib DE, Ranum PT, Keiser MS, Lewandowski BC, Carrell EM, Lysenko E, Huerta-Ocampo I, Arora S, Cheng C, Liu X, Davidson BL. An AAV variant selected through NHP screens robustly transduces the brain and drives secreted protein expression in NHPs and mice. Sci Transl Med 2025; 17:eadr2531. [PMID: 40367194 DOI: 10.1126/scitranslmed.adr2531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/08/2024] [Accepted: 04/23/2025] [Indexed: 05/16/2025]
Abstract
Recent work has shown that prolonged expression of recombinant proteins after adeno-associated virus (AAV)-mediated delivery of gene therapy to long-lived, ventricle-lining ependymal cells can profoundly affect disease phenotypes in animal models of neurodegenerative diseases. Here, we performed in vivo screens of millions of peptide-modified capsid variants of AAV1, AAV2, and AAV9 parental serotypes in adult nonhuman primates (NHPs) to identify capsids with potent transduction of key brain tissues, including ependyma, after intracerebroventricular injection. Through these screens, we identified an AAV capsid, AAV-Ep+, with markedly increased potency in transducing ependymal cells and cerebral neurons in NHPs. AAV-Ep+'s potency was conserved in three species of NHP, two mouse strains, and human neurons derived from induced pluripotent stem cells. To apply AAV-Ep+ to the treatment of ceroid lipofuscinosis type 2 disease, a lysosomal storage disorder caused by loss-of-function mutations in tripeptidyl-peptidase 1 (TPP1), we used the capsid to package the human TPP1 transgene (AAV-Ep+.hTPP1) and delivered the construct by intracerebroventricular injection into mice lacking TPP1 activity. AAV-Ep+ provided robust and therapeutically relevant TPP1 protein concentrations in these mice, significantly improving tremor and life span. In NHPs, high cerebrospinal fluid (CSF) TPP1 concentrations were achieved after intracerebroventricular delivery of AAV-Ep+.hTPP1 at a total dose of 1 × 1012 viral genomes, which was more than 30× lower than previously reported doses in NHPs. These results suggest that AAV-Ep+ may be a potent vector for gene therapy applications where CSF protein expression is required.
Collapse
Affiliation(s)
- Luis Tecedor
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yong Hong Chen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David E Leib
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Latus Bio, Philadelphia, PA 19104, USA
| | - Paul T Ranum
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Latus Bio, Philadelphia, PA 19104, USA
| | - Megan S Keiser
- Department of Neurological Surgery, NeuroTech Institute, Ohio State University, Columbus, OH 43210 USA
| | - Brian C Lewandowski
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elli M Carrell
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elena Lysenko
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Icnelia Huerta-Ocampo
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sakshi Arora
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Congsheng Cheng
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Xueyuan Liu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
2
|
Rasmussen CLM, Frederiksen SF, Heegaard CW, Thomsen MS, Hede E, Laczek B, Körbelin J, Wüstner D, Thomsen LB, Schwaninger M, Jensen ON, Moos T, Burkhart A. Endothelial and neuronal engagement by AAV-BR1 gene therapy alleviates neurological symptoms and lipid deposition in a mouse model of Niemann-Pick type C2. Fluids Barriers CNS 2025; 22:13. [PMID: 39891227 PMCID: PMC11786545 DOI: 10.1186/s12987-025-00621-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Patients with the genetic disorder Niemann-Pick type C2 disease (NP-C2) suffer from lysosomal accumulation of cholesterol causing both systemic and severe neurological symptoms. In a murine NP-C2 model, otherwise successful intravenous Niemann-Pick C2 protein (NPC2) replacement therapy fails to alleviate progressive neurodegeneration as infused NPC2 cannot cross the blood-brain barrier (BBB). Genetic modification of brain endothelial cells (BECs) is thought to enable secretion of recombinant proteins thereby overcoming the restrictions of the BBB. We hypothesized that an adeno-associated virus (AAV-BR1) encoding the Npc2 gene could cure neurological symptoms in Npc2-/- mice through transduction of BECs, and possibly neurons via viral passage across the BBB. METHODS Six weeks old Npc2-/- mice were intravenously injected with the AAV-BR1-NPC2 vector. Composite phenotype scores and behavioral tests were assessed for the following 6 weeks and visually documented. Post-mortem analyses included gene expression analyses, verification of neurodegeneration in Purkinje cells, determination of NPC2 transduction in the CNS, assessment of gliosis, quantification of gangliosides, and co-detection of cholesterol with NPC2 in degenerating neurons. RESULTS Treatment with the AAV-BR1-NPC2 vector improved motor functions, reduced neocortical inflammation, and preserved Purkinje cells in most of the mice, referred to as high responders. The vector exerted tropism for BECs and neurons resulting in a widespread NPC2 distribution in the brain with a concomitant reduction of cholesterol in adjacent neurons, presumably not transduced by the vector. Mass spectrometry imaging revealed distinct lipid alterations in the brains of Npc2-/- mice, with increased GM2 and GM3 ganglioside accumulation in the cerebellum and hippocampus. AAV-BR1-NPC2 treatment partially normalized these ganglioside distributions in high responders, including restoration of lipid profiles towards those of Npc2+/+ controls. CONCLUSION The data suggests cross-correcting gene therapy to the brain via delivery of NPC2 from BECs and neurons.
Collapse
Affiliation(s)
- Charlotte Laurfelt Munch Rasmussen
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Selma Lagerlöfts Vej 249, 9260, Gistrup, Denmark
| | - Signe Frost Frederiksen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Christian Würtz Heegaard
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000, Aarhus C, Denmark
| | - Maj Schneider Thomsen
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Selma Lagerlöfts Vej 249, 9260, Gistrup, Denmark
| | - Eva Hede
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Selma Lagerlöfts Vej 249, 9260, Gistrup, Denmark
| | - Bartosz Laczek
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Selma Lagerlöfts Vej 249, 9260, Gistrup, Denmark
| | - Jakob Körbelin
- Department of Oncology, Hematology, and Bone Marrow Transplantation, University of Medical Center Hamburg-Eppendorf, Martinisstr. 52, 20246, Hamburg, Germany
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Louiza Bohn Thomsen
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Selma Lagerlöfts Vej 249, 9260, Gistrup, Denmark
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Ole N Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Torben Moos
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Selma Lagerlöfts Vej 249, 9260, Gistrup, Denmark.
| | - Annette Burkhart
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Selma Lagerlöfts Vej 249, 9260, Gistrup, Denmark.
| |
Collapse
|
3
|
Hunter JE, Vite CH, Molony CM, O'Donnell PA, Wolfe JH. Intracisternal vs intraventricular injection of AAV1 result in comparable, widespread transduction of the dog brain. Gene Ther 2024:10.1038/s41434-024-00510-9. [PMID: 39653737 DOI: 10.1038/s41434-024-00510-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/13/2024] [Accepted: 11/28/2024] [Indexed: 03/29/2025]
Abstract
Widespread distribution of transduced brain cells following delivery of AAV vectors into the cerebrospinal fluid (CSF) of the cisterna magna (CM) has been demonstrated in large animal brains. In humans, intraventricular injection is preferred to intracisternal injection for CSF delivery due to the risk of brain stem injury. One study in the dog reported adverse reactions to AAV vectors expressing GFP injected into the lateral ventricle but not when injected into the CM. In contrast, AAV expressing mammalian genes in diseased animals have not triggered adverse responses since many genetic diseases also have compromised immune systems. Differences in circulation of CSF from each site could potentially affect vector spread within the brain, but a direct comparison has not been made using both a mammalian gene and immunologically normal animals. In this study we evaluated the dopamine-2-receptor (D2R) variant D2R80A, which is inactivated for intracellular signaling and has been used as a reporter gene in large animal brains. No adverse reactions to the D2R80A gene were observed from either injection route in normal dogs and both routes resulted in comparable distribution of D2R80A within the brain.
Collapse
Affiliation(s)
- Jacqueline E Hunter
- Research Institute of Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Charles H Vite
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32608, USA
| | - Caitlyn M Molony
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Patricia A O'Donnell
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John H Wolfe
- Research Institute of Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
4
|
Xie S, Li F. Ependymal cells: roles in central nervous system infections and therapeutic application. J Neuroinflammation 2024; 21:255. [PMID: 39385253 PMCID: PMC11465851 DOI: 10.1186/s12974-024-03240-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Ependymal cells are arranged along the inner surfaces of the ventricles and the central canal of the spinal cord, providing anatomical, physiological and immunological barriers that maintain cerebrospinal fluid (CSF) homeostasis. Based on this, studies have found that alterations in gene expression, cell junctions, cytokine secretion and metabolic disturbances can lead to dysfunction of ependymal cells, thereby participating in the onset and progression of central nervous system (CNS) infections. Additionally, ependymal cells can exhibit proliferative and regenerative potential as well as secretory functions during CNS injury, contributing to neuroprotection and post-injury recovery. Currently, studies on ependymal cell primarily focus on the basic investigations of their morphology, function and gene expression; however, there is a notable lack of clinical translational studies examining the molecular mechanisms by which ependymal cells are involved in disease onset and progression. This limits our understanding of ependymal cells in CNS infections and the development of therapeutic applications. Therefore, this review will discuss the molecular mechanism underlying the involvement of ependymal cells in CNS infections, and explore their potential for application in clinical treatment modalities.
Collapse
Affiliation(s)
- Shiqi Xie
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China
| | - Feng Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
- Shanghai Institute of Infectious Disease and Biosecurity, 130 Dong An Road, Xuhui District, Shanghai, China.
- Tuberculosis Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
| |
Collapse
|
5
|
Zhang Y, Chen Z, Wang X, Yan R, Bao H, Chu X, Guo L, Wang X, Li Y, Mu Y, He Q, Zhang L, Zhang C, Zhou D, Ji D. Site-specific tethering nanobodies on recombinant adeno-associated virus vectors for retargeted gene therapy. Acta Biomater 2024; 187:304-315. [PMID: 39025389 DOI: 10.1016/j.actbio.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Recombinant adeno-associated viruses (rAAVs) have been extensively studied for decades as carriers for delivering therapeutic genes. However, designing rAAV vectors with selective tropism for specific cell types and tissues has remained challenging. Here, we introduce a strategy for redirecting rAAV by attaching nanobodies with desired tropism at specific sites, effectively replacing the original tropism. To demonstrate this concept, we initially modified the genetic code of rAAV2 to introduce an azido-containing unnatural amino acid at a precise site within the capsid protein. Following a screening process, we identified a critical site (N587+1) where the introduction of unnatural amino acid eliminated the natural tropism of rAAV2. Subsequently, we successfully redirected rAAV2 by conjugating various nanobodies at the N587+1 site, using click and SpyTag-Spycatcher chemistries to form nanobody-AAV conjugates (NACs). By investigating the relationship between NACs quantity and effect and optimizing the linker between rAAV2 and the nanobody using a cathepsin B-susceptible valine-citrulline (VC) dipeptide, we significantly improved gene delivery efficiency both in vitro and in vivo. This enhancement can be attributed to the facilitated endosomal escape of rAAV2. Our method offers an exciting avenue for the rational modification of rAAV2 as a retargeting vehicle, providing a convenient platform for precisely engineering various rAAV2 vectors for both basic research and therapeutic applications. STATEMENT OF SIGNIFICANCE: AAVs hold great promise in the treatment of genetic diseases, but their clinical use has been limited by off-target transduction and efficiency. Here, we report a strategy to construct NACs by conjugating a nanobody or scFv to an rAAV capsid site, specifically via biorthogonal click chemistry and a spy-spycatcher reaction. We explored the structure-effect and quantity-effect relationships of NACs and then optimized the transduction efficiency by introducing a valine-citrulline peptide linker. This approach provides a biocompatible method for rational modification of rAAV as a retargeting platform without structural disruption of the virus or alteration of the binding capacity of the nanobody, with potential utility across a broad spectrum of applications in targeted imaging and gene delivery.
Collapse
Affiliation(s)
- Yuanjie Zhang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Zhiqian Chen
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Xiaoyang Wang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Rongding Yan
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Han Bao
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Xindang Chu
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Lingfeng Guo
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Xinchen Wang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Yuanhao Li
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Yu Mu
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China.
| | - Qiuchen He
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Lihe Zhang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Chuanling Zhang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Demin Zhou
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Dezhong Ji
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| |
Collapse
|
6
|
Hunter JE, Molony CM, Bagel JH, O’Donnell P, Vite CH, Chawla S, Poptani H, Wolfe JH. Widespread correction of brain pathology in feline alpha-mannosidosis by dose escalation of intracisternal AAV vector injection. Mol Ther Methods Clin Dev 2024; 32:101272. [PMID: 38946937 PMCID: PMC11214173 DOI: 10.1016/j.omtm.2024.101272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/20/2024] [Indexed: 07/02/2024]
Abstract
Alpha-mannosidosis is caused by a genetic deficiency of lysosomal alpha-mannosidase, leading to the widespread presence of storage lesions in the brain and other tissues. Enzyme replacement therapy is available but is not approved for treating the CNS, since the enzyme does not penetrate the blood-brain barrier. However, intellectual disability is a major manifestation of the disease; thus, a complimentary treatment is needed. While enzyme replacement therapy into the brain is technically feasible, it requires ports and frequent administration over time that are difficult to manage medically. Infusion of adeno-associated viral vectors into the cerebrospinal fluid is an attractive route for broadly targeting brain cells. We demonstrate here the widespread post-symptomatic correction of the globally distributed storage lesions by infusion of a high dose of AAV1-feline alpha-mannosidase (fMANB) into the CSF via the cisterna magna in the gyrencephalic alpha-mannosidosis cat brain. Significant improvements in clinical parameters occurred, and widespread global correction was documented pre-mortem by non-invasive magnetic resonance imaging. Postmortem analysis demonstrated high levels of MANB activity and reversal of lysosomal storage lesions throughout the brain. Thus, CSF treatment by adeno-associated viral vector gene therapy appears to be a suitable complement to systemic enzyme replacement therapy to potentially treat the whole patient.
Collapse
Affiliation(s)
- Jacqueline E. Hunter
- Research Institute of Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Caitlyn M. Molony
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jessica H. Bagel
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patricia O’Donnell
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles H. Vite
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - John H. Wolfe
- Research Institute of Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Jackson RJ, Keiser MS, Meltzer JC, Fykstra DP, Dierksmeier SE, Hajizadeh S, Kreuzer J, Morris R, Melloni A, Nakajima T, Tecedor L, Ranum PT, Carrell E, Chen Y, Nishtar MA, Holtzman DM, Haas W, Davidson BL, Hyman BT. APOE2 gene therapy reduces amyloid deposition and improves markers of neuroinflammation and neurodegeneration in a mouse model of Alzheimer disease. Mol Ther 2024; 32:1373-1386. [PMID: 38504517 PMCID: PMC11081918 DOI: 10.1016/j.ymthe.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/05/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do, they have a later age of onset, milder clinical course, and less severe neuropathological findings than people without this allele. The contrast is especially stark when compared with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, a more aggressive clinical course and more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Here, we demonstrate that brain exposure to APOE ε2 via a gene therapy approach, which bathes the entire cortical mantle in the gene product after transduction of the ependyma, reduces Aβ plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE ε4. This result suggests a promising protective effect of exogenous APOE ε2 and reveals a cell nonautonomous effect of the protein on microglial activation, which we show is similar to plaque-associated microglia in the brain of Alzheimer disease patients who inherit APOE ε2. These data increase the potential that an APOE ε2 therapeutic could be effective in Alzheimer disease, even in individuals born with the risky ε4 allele.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Megan S Keiser
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jonah C Meltzer
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Dustin P Fykstra
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Steven E Dierksmeier
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA; Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | - Soroush Hajizadeh
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Johannes Kreuzer
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Robert Morris
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK
| | - Alexandra Melloni
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Tsuneo Nakajima
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Luis Tecedor
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul T Ranum
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ellie Carrell
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - YongHong Chen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Maryam A Nishtar
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Wilhelm Haas
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bradley T Hyman
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
8
|
Carrell EM, Chen YH, Ranum PT, Coffin SL, Singh LN, Tecedor L, Keiser MS, Hudry E, Hyman BT, Davidson BL. VWA3A-derived ependyma promoter drives increased therapeutic protein secretion into the CSF. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:296-304. [PMID: 37547292 PMCID: PMC10400871 DOI: 10.1016/j.omtn.2023.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
Recombinant adeno-associated viral vectors (rAAVs) are a promising strategy to treat neurodegenerative diseases because of their ability to infect non-dividing cells and confer long-term transgene expression. Despite an ever-growing library of capsid variants, widespread delivery of AAVs in the adult central nervous system remains a challenge. We have previously demonstrated successful distribution of secreted proteins by infection of the ependyma, a layer of post-mitotic epithelial cells lining the ventricles of the brain and central column of the spinal cord, and subsequent protein delivery via the cerebrospinal fluid (CSF). Here we define a functional ependyma promoter to enhance expression from this cell type. Using RNA sequencing on human autopsy samples, we identified disease- and age-independent ependyma gene signatures. Associated promoters were cloned and screened as libraries in mouse and rhesus macaque to reveal cross-species function of a human DNA-derived von Willebrand factor domain containing 3A (VWA3A) promoter. When tested in mice, our VWA3A promoter drove strong, ependyma-localized expression of eGFP and increased secreted ApoE protein levels in the CSF by 2-12× over the ubiquitous iCAG promoter.
Collapse
Affiliation(s)
- Ellie M. Carrell
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yong Hong Chen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul T. Ranum
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stephanie L. Coffin
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Larry N. Singh
- Center for Mitochondrial and Epigenomic Medicine, 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
| | - Megan S. Keiser
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eloise Hudry
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Bradley T. Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, 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 & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
9
|
Ellison S, Parker H, Bigger B. Advances in therapies for neurological lysosomal storage disorders. J Inherit Metab Dis 2023; 46:874-905. [PMID: 37078180 DOI: 10.1002/jimd.12615] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
Lysosomal Storage Disorders (LSDs) are a diverse group of inherited, monogenic diseases caused by functional defects in specific lysosomal proteins. The lysosome is a cellular organelle that plays a critical role in catabolism of waste products and recycling of macromolecules in the body. Disruption to the normal function of the lysosome can result in the toxic accumulation of storage products, often leading to irreparable cellular damage and organ dysfunction followed by premature death. The majority of LSDs have no curative treatment, with many clinical subtypes presenting in early infancy and childhood. Over two-thirds of LSDs present with progressive neurodegeneration, often in combination with other debilitating peripheral symptoms. Consequently, there is a pressing unmet clinical need to develop new therapeutic interventions to treat these conditions. The blood-brain barrier is a crucial hurdle that needs to be overcome in order to effectively treat the central nervous system (CNS), adding considerable complexity to therapeutic design and delivery. Enzyme replacement therapy (ERT) treatments aimed at either direct injection into the brain, or using blood-brain barrier constructs are discussed, alongside more conventional substrate reduction and other drug-related therapies. Other promising strategies developed in recent years, include gene therapy technologies specifically tailored for more effectively targeting treatment to the CNS. Here, we discuss the most recent advances in CNS-targeted treatments for neurological LSDs with a particular emphasis on gene therapy-based modalities, such as Adeno-Associated Virus and haematopoietic stem cell gene therapy approaches that encouragingly, at the time of writing are being evaluated in LSD clinical trials in increasing numbers. If safety, efficacy and improved quality of life can be demonstrated, these therapies have the potential to be the new standard of care treatments for LSD patients.
Collapse
Affiliation(s)
- S Ellison
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| | - H Parker
- Division of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - B Bigger
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
10
|
Jackson RJ, Keiser MS, Meltzer JC, Fykstra DP, Dierksmeier SE, Melloni A, Nakajima T, Tecedor L, Ranum PT, Carrell E, Chen Y, Holtzman DM, Davidson BL, Hyman BT. APOE2 gene therapy reduces amyloid deposition, and improves markers of neuroinflammation and neurodegeneration in a mouse model of Alzheimer disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.552850. [PMID: 37645718 PMCID: PMC10461997 DOI: 10.1101/2023.08.14.552850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do they have a later age of onset, milder clinical course, and less severe neuropathological findings than others with Alzheimer disease. The contrast is especially stark in comparison to the phenotype associated with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, as well as a more aggressive clinical course and notably more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Even one APOE ε2 allele improves phenotype, but it is uncertain if that is due to the replacement of a more toxic allele by APOE ε2, or if APOE ε2 has a protective, neuro-modulatory effect. Here, we demonstrate that brain exposure to APOE2 via a gene therapy approach which bathes the entire cortical mantle in the gene product after transduction of the ependyma, rapidly ameliorates established Aβ plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE4. This result suggests a promising protective effect of exogenous APOE2, revealing a cell non-autonomous effect of the protein on microglial activation. We also show that plaque associated microglia in the brain of patients who inherit APOE2 similarly have less microglial reactivity to plaques. These data raise the potential that an APOE2 therapeutic could be effective in Alzheimer disease even in individuals born with the risk ε4 allele. One Sentence Summary Introduction of ApoE2 using an AAV that transduces the ependymal cells of the ventricle causes a reduction in amyloid load and plaque associated synapse loss, and reduces neuroinflammation by modulating microglial responsiveness to plaques.
Collapse
|
11
|
Mitchell NL, Murray SJ, Wellby MP, Barrell GK, Russell KN, Deane AR, Wynyard JR, Palmer MJ, Pulickan A, Prendergast PM, Casy W, Gray SJ, Palmer DN. Long-term safety and dose escalation of intracerebroventricular CLN5 gene therapy in sheep supports clinical translation for CLN5 Batten disease. Front Genet 2023; 14:1212228. [PMID: 37614821 PMCID: PMC10442658 DOI: 10.3389/fgene.2023.1212228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023] Open
Abstract
CLN5 neuronal ceroid lipofuscinosis (NCL, Batten disease) is a rare, inherited fatal neurodegenerative disorder caused by mutations in the CLN5 gene. The disease is characterised by progressive neuronal loss, blindness, and premature death. There is no cure. This study evaluated the efficacy of intracerebroventricular (ICV) delivery of an adeno-associated viral vector encoding ovine CLN5 (scAAV9/oCLN5) in a naturally occurring sheep model of CLN5 disease. CLN5 affected (CLN5-/-) sheep received low, moderate, or high doses of scAAV9/oCLN5 at three disease stages. The treatment delayed disease progression, extended survival and slowed stereotypical brain atrophy in most animals. Of note, one high dose treated animal only developed mild disease symptomology and survived to 60.1 months of age, triple the natural life expectancy of an untreated CLN5-/- sheep. Eyesight was not preserved at any administration age or dosage. Histopathologic examination revealed that greater transduction efficiency was achieved through higher ICV doses, and this resulted in greater amelioration of disease pathology. Together with other pre-clinical data from CLN5-/- sheep, the safety and efficacy data from these investigational new drug (IND)-enabling studies supported the initiation of the first in-human CLN5 gene therapy clinical study using the ICV delivery route for the treatment of CLN5 NCL. Clinical Trial Registration: https://clinicaltrials.gov/, identifier NCT05228145.
Collapse
Affiliation(s)
- Nadia L. Mitchell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
- Department of Radiology, University of Otago, Christchurch, New Zealand
| | - Samantha J. Murray
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Martin P. Wellby
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Graham K. Barrell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Katharina N. Russell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Ashley R. Deane
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - John R. Wynyard
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Madeleine J. Palmer
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Anila Pulickan
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | | | - Widler Casy
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Steven J. Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David N. Palmer
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
- Department of Radiology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
12
|
Swier VJ, White KA, Johnson TB, Wang X, Han J, Pearce DA, Singh R, Drack AV, Pfeifer W, Rogers CS, Brudvig JJ, Weimer JM. A novel porcine model of CLN3 Batten disease recapitulates clinical phenotypes. Dis Model Mech 2023; 16:dmm050038. [PMID: 37305926 PMCID: PMC10434985 DOI: 10.1242/dmm.050038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
Mouse models of CLN3 Batten disease, a rare lysosomal storage disorder with no cure, have improved our understanding of CLN3 biology and therapeutics through their ease of use and a consistent display of cellular pathology. However, the translatability of murine models is limited by disparities in anatomy, body size, life span and inconsistent subtle behavior deficits that can be difficult to detect in CLN3 mutant mouse models, thereby limiting their use in preclinical studies. Here, we present a longitudinal characterization of a novel miniswine model of CLN3 disease that recapitulates the most common human pathogenic variant, an exon 7-8 deletion (CLN3Δex7/8). Progressive pathology and neuron loss is observed in various regions of the CLN3Δex7/8 miniswine brain and retina. Additionally, mutant miniswine present with retinal degeneration and motor abnormalities, similar to deficits seen in humans diagnosed with the disease. Taken together, the CLN3Δex7/8 miniswine model shows consistent and progressive Batten disease pathology, and behavioral impairment mirroring clinical presentation, demonstrating its value in studying the role of CLN3 and safety/efficacy of novel disease-modifying therapeutics.
Collapse
Affiliation(s)
- Vicki J. Swier
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Katherine A. White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Tyler B. Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | | | - Jimin Han
- Department of Ophthalmology, Center for Visual Science, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David A. Pearce
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Ruchira Singh
- Department of Ophthalmology, Center for Visual Science, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Arlene V. Drack
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
- University of Iowa Institute for Vision Research, Iowa City, IA 52242, USA
| | - Wanda Pfeifer
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
| | | | - Jon J. Brudvig
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| | - Jill M. Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
13
|
Sleat DE, Banach-Petrosky W, Larrimore KE, Nemtsova Y, Wiseman JA, Najafi A, Johnson D, Poole TA, Takahashi K, Cooper JD, Lobel P. A mouse mutant deficient in both neuronal ceroid lipofuscinosis-associated proteins CLN3 and TPP1. J Inherit Metab Dis 2023; 46:720-734. [PMID: 37078466 PMCID: PMC10330656 DOI: 10.1002/jimd.12619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
Late-infantile neuronal ceroid lipofuscinosis (LINCL) and juvenile neuronal ceroid lipofuscinosis (JNCL) are inherited neurodegenerative diseases caused by mutations in the genes encoding lysosomal proteins tripeptidyl peptidase 1 (TPP1) and CLN3 protein, respectively. TPP1 is well-understood and, aided by animal models that accurately recapitulate the human disease, enzyme replacement therapy has been approved and other promising therapies are emerging. In contrast, there are no effective treatments for JNCL, partly because the function of the CLN3 protein remains unknown but also because animal models have attenuated disease and lack robust survival phenotypes. Mouse models for LINCL and JNCL, with mutations in Tpp1 and Cln3, respectively, have been thoroughly characterized but the phenotype of a double Cln3/Tpp1 mutant remains unknown. We created this double mutant and find that its phenotype is essentially indistinguishable from the single Tpp1-/- mutant in terms of survival and brain pathology. Analysis of brain proteomic changes in the single Tpp1-/- and double Cln3-/- ;Tpp1-/- mutants indicates largely overlapping sets of altered proteins and reinforces earlier studies that highlight GPNMB, LYZ2, and SERPINA3 as promising biomarker candidates in LINCL while several lysosomal proteins including SMPD1 and NPC1 appear to be altered in the Cln3-/- animals. An unexpected finding was that Tpp1 heterozygosity significantly decreased lifespan of the Cln3-/- mouse. The truncated survival of this mouse model makes it potentially useful in developing therapies for JNCL using survival as an endpoint. In addition, this model may also provide insights into CLN3 protein function and its potential functional interactions with TPP1.
Collapse
Affiliation(s)
- David E. Sleat
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers University, Piscataway, NJ, United States of America
| | - Whitney Banach-Petrosky
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Katherine E. Larrimore
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Yuliya Nemtsova
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Jennifer A. Wiseman
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Allison Najafi
- The Lundquist Institute (formerly Los Angeles Biomedical Research Institute), Harbor-UCLA Medical Center, and David Geffen School of Medicine, University of California, Los Angeles, Torrance, CA United States of America
| | - Dymonn Johnson
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Timothy A. Poole
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Keigo Takahashi
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Jonathan D. Cooper
- The Lundquist Institute (formerly Los Angeles Biomedical Research Institute), Harbor-UCLA Medical Center, and David Geffen School of Medicine, University of California, Los Angeles, Torrance, CA United States of America
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
- Genetics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
- Neurology, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Peter Lobel
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers University, Piscataway, NJ, United States of America
| |
Collapse
|
14
|
Placci M, Giannotti MI, Muro S. Polymer-based drug delivery systems under investigation for enzyme replacement and other therapies of lysosomal storage disorders. Adv Drug Deliv Rev 2023; 197:114683. [PMID: 36657645 PMCID: PMC10629597 DOI: 10.1016/j.addr.2022.114683] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/30/2022] [Accepted: 12/25/2022] [Indexed: 01/18/2023]
Abstract
Lysosomes play a central role in cellular homeostasis and alterations in this compartment associate with many diseases. The most studied example is that of lysosomal storage disorders (LSDs), a group of 60 + maladies due to genetic mutations affecting lysosomal components, mostly enzymes. This leads to aberrant intracellular storage of macromolecules, altering normal cell function and causing multiorgan syndromes, often fatal within the first years of life. Several treatment modalities are available for a dozen LSDs, mostly consisting of enzyme replacement therapy (ERT) strategies. Yet, poor biodistribution to main targets such as the central nervous system, musculoskeletal tissue, and others, as well as generation of blocking antibodies and adverse effects hinder effective LSD treatment. Drug delivery systems are being studied to surmount these obstacles, including polymeric constructs and nanoparticles that constitute the focus of this article. We provide an overview of the formulations being tested, the diseases they aim to treat, and the results observed from respective in vitro and in vivo studies. We also discuss the advantages and disadvantages of these strategies, the remaining gaps of knowledge regarding their performance, and important items to consider for their clinical translation. Overall, polymeric nanoconstructs hold considerable promise to advance treatment for LSDs.
Collapse
Affiliation(s)
- Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Marina I Giannotti
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; CIBER-BBN, ISCIII, Barcelona, Spain; Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona 08028, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; Institute of Catalonia for Research and Advanced Studies (ICREA), Barcelona 08010, Spain; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
15
|
El-Hage N, Haney MJ, Zhao Y, Rodriguez M, Wu Z, Liu M, Swain CJ, Yuan H, Batrakova EV. Extracellular Vesicles Released by Genetically Modified Macrophages Activate Autophagy and Produce Potent Neuroprotection in Mouse Model of Lysosomal Storage Disorder, Batten Disease. Cells 2023; 12:1497. [PMID: 37296618 PMCID: PMC10252192 DOI: 10.3390/cells12111497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/11/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Over the recent decades, the use of extracellular vesicles (EVs) has attracted considerable attention. Herein, we report the development of a novel EV-based drug delivery system for the transport of the lysosomal enzyme tripeptidyl peptidase-1 (TPP1) to treat Batten disease (BD). Endogenous loading of macrophage-derived EVs was achieved through transfection of parent cells with TPP1-encoding pDNA. More than 20% ID/g was detected in the brain following a single intrathecal injection of EVs in a mouse model of BD, ceroid lipofuscinosis neuronal type 2 (CLN2) mice. Furthermore, the cumulative effect of EVs repetitive administrations in the brain was demonstrated. TPP1-loaded EVs (EV-TPP1) produced potent therapeutic effects, resulting in efficient elimination of lipofuscin aggregates in lysosomes, decreased inflammation, and improved neuronal survival in CLN2 mice. In terms of mechanism, EV-TPP1 treatments caused significant activation of the autophagy pathway, including altered expression of the autophagy-related proteins LC3 and P62, in the CLN2 mouse brain. We hypothesized that along with TPP1 delivery to the brain, EV-based formulations can enhance host cellular homeostasis, causing degradation of lipofuscin aggregates through the autophagy-lysosomal pathway. Overall, continued research into new and effective therapies for BD is crucial for improving the lives of those affected by this condition.
Collapse
Affiliation(s)
- Nazira El-Hage
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (N.E.-H.); (M.R.)
| | - Matthew J. Haney
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.J.H.); (Y.Z.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| | - Yuling Zhao
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.J.H.); (Y.Z.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| | - Myosotys Rodriguez
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (N.E.-H.); (M.R.)
| | - Zhanhong Wu
- Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; (Z.W.); (H.Y.)
| | - Mori Liu
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| | - Carson J. Swain
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| | - Hong Yuan
- Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; (Z.W.); (H.Y.)
| | - Elena V. Batrakova
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.J.H.); (Y.Z.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| |
Collapse
|
16
|
Knoernschild K, Johnson HJ, Schroeder KE, Swier VJ, White KA, Sato TS, Rogers CS, Weimer JM, Sieren JC. Magnetic resonance brain volumetry biomarkers of CLN2 Batten disease identified with miniswine model. Sci Rep 2023; 13:5146. [PMID: 36991106 PMCID: PMC10060411 DOI: 10.1038/s41598-023-32071-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Late-infantile neuronal ceroid lipofuscinosis type 2 (CLN2) disease (Batten disease) is a rare pediatric disease, with symptom development leading to clinical diagnosis. Early diagnosis and effective tracking of disease progression are required for treatment. We hypothesize that brain volumetry is valuable in identifying CLN2 disease at an early stage and tracking disease progression in a genetically modified miniswine model. CLN2R208X/R208X miniswine and wild type controls were evaluated at 12- and 17-months of age, correlating to early and late stages of disease progression. Magnetic resonance imaging (MRI) T1- and T2-weighted data were acquired. Total intercranial, gray matter, cerebrospinal fluid, white matter, caudate, putamen, and ventricle volumes were calculated and expressed as proportions of the intracranial volume. The brain regions were compared between timepoints and cohorts using Gardner-Altman plots, mean differences, and confidence intervals. At an early stage of disease, the total intracranial volume (- 9.06 cm3), gray matter (- 4.37% 95 CI - 7.41; - 1.83), caudate (- 0.16%, 95 CI - 0.24; - 0.08) and putamen (- 0.11% 95 CI - 0.23; - 0.02) were all notably smaller in CLN2R208X/R208X miniswines versus WT, while cerebrospinal fluid was larger (+ 3.42%, 95 CI 2.54; 6.18). As the disease progressed to a later stage, the difference between the gray matter (- 8.27%, 95 CI - 10.1; - 5.56) and cerebrospinal fluid (+ 6.88%, 95 CI 4.31; 8.51) continued to become more pronounced, while others remained stable. MRI brain volumetry in this miniswine model of CLN2 disease is sensitive to early disease detection and longitudinal change monitoring, providing a valuable tool for pre-clinical treatment development and evaluation.
Collapse
Affiliation(s)
- Kevin Knoernschild
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Hans J Johnson
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Kimberly E Schroeder
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
| | - Vicki J Swier
- 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
| | - Takashi S Sato
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
| | | | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Jessica C Sieren
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA.
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
17
|
Johnson TB, Brudvig JJ, Likhite S, Pratt MA, White KA, Cain JT, Booth CD, Timm DJ, Davis SS, Meyerink B, Pineda R, Dennys-Rivers C, Kaspar BK, Meyer K, Weimer JM. Early postnatal administration of an AAV9 gene therapy is safe and efficacious in CLN3 disease. Front Genet 2023; 14:1118649. [PMID: 37035740 PMCID: PMC10080320 DOI: 10.3389/fgene.2023.1118649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
CLN3 disease, caused by biallelic mutations in the CLN3 gene, is a rare pediatric neurodegenerative disease that has no cure or disease modifying treatment. The development of effective treatments has been hindered by a lack of etiological knowledge, but gene replacement has emerged as a promising therapeutic platform for such disorders. Here, we utilize a mouse model of CLN3 disease to test the safety and efficacy of a cerebrospinal fluid-delivered AAV9 gene therapy with a study design optimized for translatability. In this model, postnatal day one administration of the gene therapy virus resulted in robust expression of human CLN3 throughout the CNS over the 24-month duration of the study. A range of histopathological and behavioral parameters were assayed, with the therapy consistently and persistently rescuing a number of hallmarks of disease while being safe and well-tolerated. Together, the results show great promise for translation of the therapy into the clinic, prompting the launch of a first-in-human clinical trial (NCT03770572).
Collapse
Affiliation(s)
- Tyler B. Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Amicus Therapeutics, Cranbury, NJ, United States
| | - Jon J. Brudvig
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Amicus Therapeutics, Cranbury, NJ, United States
| | - Shibi Likhite
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Melissa A. Pratt
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Katherine A. White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Jacob T. Cain
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Amicus Therapeutics, Cranbury, NJ, United States
| | - Clarissa D. Booth
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Derek J. Timm
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Samantha S. Davis
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Brandon Meyerink
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Ricardo Pineda
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | | | - Brian K. Kaspar
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Kathrin Meyer
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Jill M. Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Amicus Therapeutics, Cranbury, NJ, United States
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
18
|
Munesue Y, Ageyama N, Kimura N, Takahashi I, Nakayama S, Okabayashi S, Katakai Y, Koie H, Yagami KI, Ishii K, Tamaoka A, Yasutomi Y, Shimozawa N. Cynomolgus macaque model of neuronal ceroid lipofuscinosis type 2 disease. Exp Neurol 2023; 363:114381. [PMID: 36918063 DOI: 10.1016/j.expneurol.2023.114381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are autosomal-recessive fatal neurodegenerative diseases that occur in children and young adults, with symptoms including ataxia, seizures and visual impairment. We report the discovery of cynomolgus macaques carrying the CLN2/TPP1 variant and our analysis of whether the macaques could be a new non-human primate model for NCL type 2 (CLN2) disease. Three cynomolgus macaques presented progressive neuronal clinical symptoms such as limb tremors and gait disturbance after about 2 years of age. Morphological analyses using brain MRI at the endpoint of approximately 3 years of age revealed marked cerebellar and cerebral atrophy of the gray matter, with sulcus dilation, gyrus thinning, and ventricular enlargement. Histopathological analyses of three affected macaques revealed severe neuronal loss and degeneration in the cerebellar and cerebral cortices, accompanied by glial activation and/or changes in axonal morphology. Neurons observed throughout the central nervous system contained autofluorescent cytoplasmic pigments, which were identified as ceroid-lipofuscin based on staining properties, and the cerebral cortex examined by transmission electron microscopy had curvilinear profiles, the typical ultrastructural pattern of CLN2. These findings are commonly observed in all forms of NCL. DNA sequencing analysis identified a homozygous single-base deletion (c.42delC) of the CLN2/TPP1 gene, resulting in a frameshifted premature stop codon. Immunohistochemical analysis showed that tissue from the affected macaques lacked a detectable signal against TPP1, the product of the CLN2/TPP1 gene. Analysis for transmission of the CLN2/TPP1 mutated gene revealed that 47 (49.5%) and 48 (50.5%) of the 95 individuals genotyped in the CLN2-affected macaque family were heterozygous carriers and homozygous wild-type individuals, respectively. Thus, we identified cynomolgus macaques as a non-human primate model of CLN2 disease. The CLN2 macaques reported here could become a useful resource for research and the development of drugs and methods for treating CLN2 disease, which involves severe symptoms in humans.
Collapse
Affiliation(s)
- Yoshiko Munesue
- Division of Clinical Medicine, Department of Neurology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Naohide Ageyama
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Nobuyuki Kimura
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan; Department of Veterinary Associated Science, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari, Ehime 794-8555, Japan
| | - Ichiro Takahashi
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Shunya Nakayama
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan; Laboratory of Veterinary Physiology/Pathophysiology, Nihon University, College of Bioresource Science, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Sachi Okabayashi
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0843, Japan
| | - Yuko Katakai
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0843, Japan
| | - Hiroshi Koie
- Laboratory of Veterinary Physiology/Pathophysiology, Nihon University, College of Bioresource Science, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Ken-Ichi Yagami
- Laboratory Animal Resource Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Kazuhiro Ishii
- Division of Clinical Medicine, Department of Neurology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Akira Tamaoka
- Division of Clinical Medicine, Department of Neurology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan; Department of Molecular and Experimental Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Nobuhiro Shimozawa
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan.
| |
Collapse
|
19
|
Nittari G, Tomassoni D, Roy P, Martinelli I, Tayebati SK, Amenta F. Batten disease through different in vivo and in vitro models: A review. J Neurosci Res 2023; 101:298-315. [PMID: 36434776 DOI: 10.1002/jnr.25147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022]
Abstract
Batten disease consists of a family of primarily autosomal recessive, progressive neuropediatric disorders, also known as neuronal ceroid lipofuscinoses (NCLs). These pathologies are characterized by seizures and visual, cognitive and motor decline, and premature death. The pathophysiology of this rare disease is still unclear despite the years of trials and financial aids. This paper has reviewed advantages and limits of in vivo and in vitro models of Batten disease from murine and larger animal models to primitive unicellular models, until the most recently developed patient-derived induced pluripotent stem cells. For each model advantages, limits and applications were analyzed. The first prototypes investigated were murine models that due to their limits were replaced by larger animals. In vitro models gradually replaced animal models for practical, cost, and ethical reasons. Using induced pluripotent stem cells to study neurodegeneration is a new way of studying the disease, since they can be distinguished into differentiating elements like neurons, which are susceptible to neurodegeneration. In vivo and in vitro models have contributed to clarifying to some extent the pathophysiology of the disease. The collection and sharing of suitable human bio samples likely through biobanks can contribute to a better understanding, prevention, and to identify possible treatment strategies of Batten disease.
Collapse
Affiliation(s)
- Giulio Nittari
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Ilenia Martinelli
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Seyed Khosrow Tayebati
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Francesco Amenta
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| |
Collapse
|
20
|
Kick GR, Whiting REH, Ota-Kuroki J, Castaner LJ, Morgan-Jack B, Sabol JC, Meiman EJ, Ortiz F, Katz ML. Intravitreal gene therapy preserves retinal function in a canine model of CLN2 neuronal ceroid lipofuscinosis. Exp Eye Res 2023; 226:109344. [PMID: 36509165 PMCID: PMC9839638 DOI: 10.1016/j.exer.2022.109344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/02/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
CLN2 neuronal ceroid lipofuscinosis is a rare hereditary neurodegenerative disorder characterized by deleterious sequence variants in TPP1 that result in reduced or abolished function of the lysosomal enzyme tripeptidyl peptidase 1 (TPP1). Children with this disorder experience progressive neurological decline and vision loss starting around 2-4 years of age. Ocular disease is characterized by progressive retinal degeneration and impaired retinal function culminating in total loss of vision. Similar retinal pathology occurs in a canine model of CLN2 disease with a null variant in TPP1. A study using the dog model was performed to evaluate the efficacy of ocular gene therapy to provide a continuous, long-term source of human TPP1 (hTPP1) to the retina, inhibit retinal degeneration and preserve retinal function. TPP1-/- dogs received an intravitreal injection of 1 x 1012 viral genomes of AAV2.CAG.hTPP1 in one eye and AAV2.CAG.GFP in the contralateral eye at 4 months of age. Ophthalmic exams, in vivo ocular imaging and electroretinography were repeated monthly to assess retinal structure and function. Retinal morphology, hTPP1 and GFP expression in the retina, optic nerve and lateral geniculate nucleus, and hTPP1 concentrations in the vitreous were evaluated after the dogs were euthanized at end stage neurological disease at approximately 10 months of age. Intravitreal administration of AAV2.CAG.hTPP1 resulted in stable, widespread expression of hTPP1 throughout the inner retina, prevented disease-related declines in retinal function and inhibited disease-related cell loss and storage body accumulation in the retina for at least 6 months. Uveitis occurred in eyes treated with the hTPP1 vector, but this did not prevent therapeutic efficacy. The severity of the uveitis was ameliorated with anti-inflammatory treatments. These results indicate that a single intravitreal injection of AAV2.CAG.hTPP1 is an effective treatment to inhibit ocular disease progression in canine CLN2 disease.
Collapse
Affiliation(s)
- Grace Robinson Kick
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Rebecca E H Whiting
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Juri Ota-Kuroki
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Leilani J Castaner
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Brandie Morgan-Jack
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Julianna C Sabol
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Elizabeth J Meiman
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Francheska Ortiz
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Martin L Katz
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA.
| |
Collapse
|
21
|
Moura E, Tasqueti UI, Mangrich-Rocha RMV, Filho JRE, de Farias MR, Pimpão CT. Inborn Errors of Metabolism in Dogs: Historical, Metabolic, Genetic, and Clinical Aspects. Top Companion Anim Med 2022; 51:100731. [DOI: 10.1016/j.tcam.2022.100731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/11/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
|
22
|
Swier VJ, White KA, Johnson TB, Sieren JC, Johnson HJ, Knoernschild K, Wang X, Rohret FA, Rogers CS, Pearce DA, Brudvig JJ, Weimer JM. A Novel Porcine Model of CLN2 Batten Disease that Recapitulates Patient Phenotypes. Neurotherapeutics 2022; 19:1905-1919. [PMID: 36100791 PMCID: PMC9723024 DOI: 10.1007/s13311-022-01296-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2022] [Indexed: 12/13/2022] Open
Abstract
CLN2 Batten disease is a lysosomal disorder in which pathogenic variants in CLN2 lead to reduced activity in the enzyme tripeptidyl peptidase 1. The disease typically manifests around 2 to 4 years of age with developmental delay, ataxia, seizures, inability to speak and walk, and fatality between 6 and 12 years of age. Multiple Cln2 mouse models exist to better understand the etiology of the disease; however, these models are unable to adequately recapitulate the disease due to differences in anatomy and physiology, limiting their utility for therapeutic testing. Here, we describe a new CLN2R208X/R208X porcine model of CLN2 disease. We present comprehensive characterization showing behavioral, pathological, and visual phenotypes that recapitulate those seen in CLN2 patients. CLN2R208X/R208X miniswine present with gait abnormalities at 6 months of age, ERG waveform declines at 6-9 months, vision loss at 11 months, cognitive declines at 12 months, seizures by 15 months, and early death at 18 months due to failure to thrive. CLN2R208X/R208X miniswine also showed classic storage material accumulation and glial activation in the brain at 6 months, and cortical atrophy at 12 months. Thus, the CLN2R208X/R208X miniswine model is a valuable resource for biomarker discovery and therapeutic development in CLN2 disease.
Collapse
Affiliation(s)
- Vicki J Swier
- 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
| | - Tyler B Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Jessica C Sieren
- Department of Radiology, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Hans J Johnson
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Kevin Knoernschild
- Department of Radiology, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | | | | | | | - David A Pearce
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Jon J Brudvig
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, 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, University of South Dakota, Sioux Falls, SD, USA.
| |
Collapse
|
23
|
Rybarikova M, Almacellas Barbanoj A, Schorge S, Déglon N. CNS gene therapy: present developments and emerging trends accelerating industry-academia pathways. Hum Gene Ther 2022; 33:913-922. [PMID: 36070435 DOI: 10.1089/hum.2022.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The recent success of first central nervous system gene therapies has reinvigorated the growing community of gene therapy researchers and strengthened the field's market position. We are witnessing an increase of clinical trials with long-term efficiency mainly for neurometabolic, neurodegenerative and neurodevelopmental diseases caused by loss-of-function mutations. The ever-expanding knowledge and accessibility to the most advanced tools allow enrichment of applications to more complex diseases. This gradually contributes towards sealing the gap between top diseases impacting current global health and those towards which gene therapy development is currently aimed. Here, we highlight innovative therapeutic approaches that have reached the clinics and outline the latest improvements of vector design and targeting. Finally, we address the pressing challenges faced by clinical trials and the direction they are heading.
Collapse
Affiliation(s)
- Margareta Rybarikova
- Lausanne University Hospital, Department of Clinical Neurosciences, Lausanne, Vaud, Switzerland.,Lausanne University Hospital, Neuroscience Research Center , Lausanne, Vaud, Switzerland;
| | - Amanda Almacellas Barbanoj
- University College London, Institute of Neurology (IoN), Department of Clinical and Experimental Epilepsy (DCEE), London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Stephanie Schorge
- University College London, Institute of Neurology (IoN), Department of Clinical and Experimental Epilepsy (DCEE), London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Nicole Déglon
- Lausanne University Hospital, Department of Clinical Neurosciences, Lausanne, Vaud, Switzerland.,Lausanne University Hospital, Neuroscience Research Center, Lausanne, Vaud, Switzerland;
| |
Collapse
|
24
|
Hunter JE, Molony CM, Bagel JH, O’Donnell PA, Kaler SG, Wolfe JH. Transduction characteristics of alternative adeno-associated virus serotypes in the cat brain by intracisternal delivery. Mol Ther Methods Clin Dev 2022; 26:384-393. [PMID: 36034772 PMCID: PMC9391516 DOI: 10.1016/j.omtm.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 07/12/2022] [Indexed: 11/18/2022]
Abstract
Multiple studies have examined the transduction characteristics of different AAV serotypes in the mouse brain, where they can exhibit significantly different patterns of transduction. The pattern of transduction also varies with the route of administration. Much less information exists for the transduction characteristics in large-brained animals. Large animal models have brains that are closer in size and organization to the human brain, such as being gyrencephalic compared to the lissencephalic rodent brains, pathway organization, and certain electrophysiologic properties. Large animal models are used as translational intermediates to develop gene therapies to treat human diseases. Various AAV serotypes and routes of delivery have been used to study the correction of pathology in the brain in lysosomal storage diseases. In this study, we evaluated the ability of selected AAV serotypes to transduce cells in the cat brain when delivered into the cerebrospinal fluid via the cisterna magna. We previously showed that AAV1 transduced significantly greater numbers of cells than AAV9 in the cat brain by this route. In the present study, we evaluated serotypes closely related to AAVs 1 and 9 (AAVs 6, AS, hu32) that may mediate more extensive transduction, as well as AAVs 4 and 5, which primarily transduce choroid plexus epithelial (CPE) and ependymal lining cells in the rodent brain. The related serotypes tended to have similar patterns of transduction but were divergent in some specific brain structures.
Collapse
Affiliation(s)
- Jacqueline E. Hunter
- Research Institute of Children’s Hospital of Philadelphia, 502-G Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Caitlyn M. Molony
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jessica H. Bagel
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patricia A. O’Donnell
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen G. Kaler
- Section on Translational Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - John H. Wolfe
- Research Institute of Children’s Hospital of Philadelphia, 502-G Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA,W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Corresponding author John H. Wolfe, Children’s Hospital of Philadelphia, 502-G Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104-4399, USA.
| |
Collapse
|
25
|
Kaminiów K, Kozak S, Paprocka J. Recent Insight into the Genetic Basis, Clinical Features, and Diagnostic Methods for Neuronal Ceroid Lipofuscinosis. Int J Mol Sci 2022; 23:5729. [PMID: 35628533 PMCID: PMC9145894 DOI: 10.3390/ijms23105729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are a group of rare, inherited, neurodegenerative lysosomal storage disorders that affect children and adults. They are traditionally grouped together, based on shared clinical symptoms and pathological ground. To date, 13 autosomal recessive gene variants, as well as one autosomal dominant gene variant, of NCL have been described. These genes encode a variety of proteins, whose functions have not been fully defined; most are lysosomal enzymes, transmembrane proteins of the lysosome, or other organelles. Common symptoms of NCLs include the progressive loss of vision, mental and motor deterioration, epileptic seizures, premature death, and, in rare adult-onset cases, dementia. Depending on the mutation, these symptoms can vary, with respect to the severity and onset of symptoms by age. Currently, all forms of NCL are fatal, and no curative treatments are available. Herein, we provide an overview to summarize the current knowledge regarding the pathophysiology, genetics, and clinical manifestation of these conditions, as well as the approach to diagnosis.
Collapse
Affiliation(s)
- Konrad Kaminiów
- Students’ Scientific Society, Department of Pediatric Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (K.K.); (S.K.)
| | - Sylwia Kozak
- Students’ Scientific Society, Department of Pediatric Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (K.K.); (S.K.)
| | - Justyna Paprocka
- Pediatric Neurology Department, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
| |
Collapse
|
26
|
Bartsch U, Storch S. Experimental Therapeutic Approaches for the Treatment of Retinal Pathology in Neuronal Ceroid Lipofuscinoses. Front Neurol 2022; 13:866983. [PMID: 35509995 PMCID: PMC9058077 DOI: 10.3389/fneur.2022.866983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are a group of childhood-onset neurodegenerative lysosomal storage disorders mainly affecting the brain and the retina. In the NCLs, disease-causing mutations in 13 different ceroid lipofuscinoses genes (CLN) have been identified. The clinical symptoms include seizures, progressive neurological decline, deterioration of motor and language skills, and dementia resulting in premature death. In addition, the deterioration and loss of vision caused by progressive retinal degeneration is another major hallmark of NCLs. To date, there is no curative therapy for the treatment of retinal degeneration and vision loss in patients with NCL. In this review, the key findings of different experimental approaches in NCL animal models aimed at attenuating progressive retinal degeneration and the decline in retinal function are discussed. Different approaches, including experimental enzyme replacement therapy, gene therapy, cell-based therapy, and immunomodulation therapy were evaluated and showed encouraging therapeutic benefits. Recent experimental ocular gene therapies in NCL animal models with soluble lysosomal enzyme deficiencies and transmembrane protein deficiencies have shown the strong potential of gene-based approaches to treat retinal dystrophies in NCLs. In CLN3 and CLN6 mouse models, an adeno-associated virus (AAV) vector-mediated delivery of CLN3 and CLN6 to bipolar cells has been shown to attenuate the retinal dysfunction. Therapeutic benefits of ocular enzyme replacement therapies were evaluated in CLN2 and CLN10 animal models. Since brain-targeted gene or enzyme replacement therapies will most likely not attenuate retinal neurodegeneration, there is an unmet need for treatment options additionally targeting the retina in patients with NCL. The long-term benefits of these therapeutic interventions aimed at attenuating retinal degeneration and vision loss in patients with NCL remain to be investigated in future clinical studies.
Collapse
Affiliation(s)
- Udo Bartsch
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Storch
- University Children's Research@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Stephan Storch
| |
Collapse
|
27
|
Meiman EJ, Kick GR, Jensen CA, Coates JR, Katz ML. Characterization of neurological disease progression in a canine model of CLN5 neuronal ceroid lipofuscinosis. Dev Neurobiol 2022; 82:326-344. [PMID: 35427439 PMCID: PMC9119968 DOI: 10.1002/dneu.22878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/14/2022] [Accepted: 02/25/2022] [Indexed: 11/08/2022]
Abstract
Golden Retriever dogs with a frameshift variant in CLN5 (c.934_935delAG) suffer from a progressive neurodegenerative disorder analogous to the CLN5 form of neuronal ceroid lipofuscinosis (NCL). Five littermate puppies homozygous for the deletion allele were identified prior to the onset of disease signs. Studies were performed to characterize the onset and progression of the disease in these dogs. Neurological signs that included restlessness, unwillingness to cooperate with the handlers, and proprioceptive deficits first became apparent at approximately 12 months of age. The neurological signs progressed over time and by 21 to 23 months of age included general proprioceptive ataxia, menace response deficits, aggressive behaviors, cerebellar ataxia, intention tremors, decreased visual tracking, seizures, cognitive decline, and impaired prehension. Due to the severity of these signs, the dogs were euthanized between 21 and 23 months of age. Magnetic resonance imaging revealed pronounced progressive global brain atrophy with a more than sevenfold increase in the volume of the ventricular system between 9.5 and 22.5 months of age. Accompanying this atrophy were pronounced accumulations of autofluorescent inclusions throughout the brain and spinal cord. Ultrastructurally, the contents of these inclusions were found to consist primarily of membrane‐like aggregates. Inclusions with similar fluorescence properties were present in cardiac muscle. Similar to other forms of NCL, the affected dogs had low plasma carnitine concentrations, suggesting impaired carnitine biosynthesis. These data on disease progression will be useful in future studies using the canine model for therapeutic intervention studies.
Collapse
Affiliation(s)
- Elizabeth J. Meiman
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine University of Missouri Columbia MO 65211 USA
| | - Grace Robinson Kick
- Neurodegenerative Diseases Research Laboratory University of Missouri Columbia MO 65212 USA
| | - Cheryl A. Jensen
- Neurodegenerative Diseases Research Laboratory 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
| | - Martin L. Katz
- Neurodegenerative Diseases Research Laboratory University of Missouri Columbia MO 65212 USA
| |
Collapse
|
28
|
Takahashi K, Nelvagal HR, Lange J, Cooper JD. Glial Dysfunction and Its Contribution to the Pathogenesis of the Neuronal Ceroid Lipofuscinoses. Front Neurol 2022; 13:886567. [PMID: 35444603 PMCID: PMC9013902 DOI: 10.3389/fneur.2022.886567] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/16/2022] [Indexed: 01/05/2023] Open
Abstract
While significant efforts have been made in developing pre-clinical treatments for the neuronal ceroid lipofuscinoses (NCLs), many challenges still remain to bring children with NCLs a cure. Devising effective therapeutic strategies for the NCLs will require a better understanding of pathophysiology, but little is known about the mechanisms by which loss of lysosomal proteins causes such devastating neurodegeneration. Research into glial cells including astrocytes, microglia, and oligodendrocytes have revealed many of their critical functions in brain homeostasis and potential contributions to neurodegenerative diseases. Genetically modified mouse models have served as a useful platform to define the disease progression in the central nervous system across NCL subtypes, revealing a wide range of glial responses to disease. The emerging evidence of glial dysfunction questions the traditional “neuron-centric” view of NCLs, and would suggest that directly targeting glia in addition to neurons could lead to better therapeutic outcomes. This review summarizes the most up-to-date understanding of glial pathologies and their contribution to the pathogenesis of NCLs, and highlights some of the associated challenges that require further research.
Collapse
Affiliation(s)
- Keigo Takahashi
- Pediatric Storage Disorders Laboratory, Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Hemanth R. Nelvagal
- Department of Pharmacology, School of Pharmacy, University College London, London, United Kingdom
| | - Jenny Lange
- Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Jonathan D. Cooper
- Pediatric Storage Disorders Laboratory, Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Genetics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- *Correspondence: Jonathan D. Cooper
| |
Collapse
|
29
|
Murray SJ, Mitchell NL. Ocular therapies for neuronal ceroid lipofuscinoses: more than meets the eye. Neural Regen Res 2022; 17:1755-1756. [PMID: 35017433 PMCID: PMC8820703 DOI: 10.4103/1673-5374.332148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Samantha J Murray
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Nadia L Mitchell
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, New Zealand
| |
Collapse
|
30
|
Taghian T, Batista AR, Kamper S, Caldwell M, Lilley L, Li H, Rodriguez P, Mesa K, Zheng S, King RM, Gounis MJ, Todeasa S, Maguire A, Martin DR, Sena-Esteves M, Meade TJ, Gray-Edwards HL. Real-time MR tracking of AAV gene therapy with βgal-responsive MR probe in a murine model of GM1-gangliosidosis. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:128-134. [PMID: 34703836 PMCID: PMC8517204 DOI: 10.1016/j.omtm.2021.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022]
Abstract
Transformative results of adeno-associated virus (AAV) gene therapy in patients with spinal muscular atrophy and Leber's congenital amaurosis led to approval of the first two AAV products in the United States to treat these diseases. These extraordinary results led to a dramatic increase in the number and type of AAV gene-therapy programs. However, the field lacks non-invasive means to assess levels and duration of therapeutic protein function in patients. Here, we describe a new magnetic resonance imaging (MRI) technology for real-time reporting of gene-therapy products in the living animal in the form of an MRI probe that is activated in the presence of therapeutic protein expression. For the first time, we show reliable tracking of enzyme expression after a now in-human clinical trial AAV gene therapy (ClinicalTrials.gov: NTC03952637) encoding lysosomal acid beta-galactosidase (βgal) using a self-immolative βgal-responsive MRI probe. MRI enhancement in AAV-treated enzyme-deficient mice (GLB-1-/-) correlates with βgal activity in central nervous system and peripheral organs after intracranial or intravenous AAV gene therapy, respectively. With >1,800 gene therapies in phase I/II clinical trials (ClinicalTrials.gov), development of a non-invasive method to track gene expression over time in patients is crucial to the future of the gene-therapy field.
Collapse
Affiliation(s)
- Toloo Taghian
- 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
| | - 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
| | - Sarah Kamper
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, USA
| | - Michael Caldwell
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, USA
| | - Laura Lilley
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, USA
| | - Hao Li
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, USA
| | - Paola Rodriguez
- 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
| | - Katerina Mesa
- 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
| | - Shaokuan Zheng
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Robert M King
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Matthew J Gounis
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Sophia Todeasa
- 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
| | - Anne Maguire
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, 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
| | - Thomas J Meade
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, 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
|
31
|
Seven-year follow-up of durability and safety of AAV CNS gene therapy for a lysosomal storage disorder in a large animal. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:370-389. [PMID: 34761052 PMCID: PMC8550992 DOI: 10.1016/j.omtm.2021.09.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/25/2021] [Accepted: 09/29/2021] [Indexed: 12/15/2022]
Abstract
Delivery of adeno-associated viral vectors (AAVs) to cerebrospinal fluid (CSF) has emerged as a promising approach to achieve widespread transduction of the central nervous system (CNS) and peripheral nervous system (PNS), with direct applicability to the treatment of a wide range of neurological diseases, particularly lysosomal storage diseases. Although studies in small animal models have provided proof of concept and experiments in large animals demonstrated feasibility in bigger brains, there is not much information on long-term safety or durability of the effect. Here, we report a 7-year study in healthy beagle dogs after intra-CSF delivery of a single, clinically relevant dose (2 × 1013 vg/dog) of AAV9 vectors carrying the canine sulfamidase, the enzyme deficient in mucopolysaccharidosis type IIIA. Periodic monitoring of CSF and blood, clinical and neurological evaluations, and magnetic resonance and ultrasound imaging of target organs demonstrated no toxicity related to treatment. AAV9-mediated gene transfer resulted in detection of sulfamidase activity in CSF throughout the study. Analysis at tissue level showed widespread sulfamidase expression and activity in the absence of histological findings in any region of encephalon, spinal cord, or dorsal root ganglia. Altogether, these results provide proof of durability of expression and long-term safety for intra-CSF delivery of AAV-based gene transfer vectors encoding therapeutic proteins to the CNS.
Collapse
|
32
|
Morsy A, Carmona AV, Trippier PC. Patient-Derived Induced Pluripotent Stem Cell Models for Phenotypic Screening in the Neuronal Ceroid Lipofuscinoses. Molecules 2021; 26:molecules26206235. [PMID: 34684815 PMCID: PMC8538546 DOI: 10.3390/molecules26206235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 11/16/2022] Open
Abstract
Batten disease or neuronal ceroid lipofuscinosis (NCL) is a group of rare, fatal, inherited neurodegenerative lysosomal storage disorders. Numerous genes (CLN1–CLN8, CLN10–CLN14) were identified in which mutations can lead to NCL; however, the underlying pathophysiology remains elusive. Despite this, the NCLs share some of the same features and symptoms but vary in respect to severity and onset of symptoms by age. Some common symptoms include the progressive loss of vision, mental and motor deterioration, epileptic seizures, premature death, and in the rare adult-onset, dementia. Currently, all forms of NCL are fatal, and no curative treatments are available. Induced pluripotent stem cells (iPSCs) can differentiate into any cell type of the human body. Cells reprogrammed from a patient have the advantage of acquiring disease pathogenesis along with recapitulation of disease-associated phenotypes. They serve as practical model systems to shed new light on disease mechanisms and provide a phenotypic screening platform to enable drug discovery. Herein, we provide an overview of available iPSC models for a number of different NCLs. More specifically, we highlight findings in these models that may spur target identification and drug development.
Collapse
Affiliation(s)
- Ahmed Morsy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68106, USA; (A.M.); (A.V.C.)
| | - Angelica V. Carmona
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68106, USA; (A.M.); (A.V.C.)
| | - Paul C. Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68106, USA; (A.M.); (A.V.C.)
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68106, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE 68106, USA
- Correspondence:
| |
Collapse
|
33
|
On the cusp of cures: Breakthroughs in Batten disease research. Curr Opin Neurobiol 2021; 72:48-54. [PMID: 34571324 DOI: 10.1016/j.conb.2021.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/27/2022]
Abstract
Batten disease is a family of rare, lysosomal disorders caused by mutations in one of at least 13 genes, which encode a diverse set of lysosomal and extralysosomal proteins. Despite decades of research, the development of effective therapies has remained intractable. But now, the field is experiencing rapid, unprecedented progress on multiple fronts. New tools are providing insights into previously unsolvable problems, with molecular functions now known for nine Batten disease proteins. Protein interactome data are uncovering potential functional overlap between several Batten disease proteins, providing long-sought links between seemingly disparate proteins. Understanding of cellular etiology is elucidating contributions from and interactions between various CNS cell types. Collectively, this explosion in insight is hastening an unparalleled period of therapeutic breakthroughs, with multiple therapies showing great promise in preclinical and clinical studies. The coming years will provide a continuation of this rapid progress, with the promise of effective treatments giving patients hope.
Collapse
|
34
|
Golikova PI, Petukhova DA, Sukhomyasova AL, Nikolaeva TY, Gurinova EE, Ivanova RN, Maksimova NR. [Clinical and genetic description of neuronal ceroid lipofuscinosis 6 type in the yakut family]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:71-76. [PMID: 34481439 DOI: 10.17116/jnevro202112108171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neuronal ceroid lipofuscinosis type 6 (NCL 6) is a rare progressive neurodegenerative disease that belongs to the group of lysosomal storage diseases. A clinical and genetic description of NCL 6 in a Yakut family was carried out. The proband and her sibling showed characteristic clinical signs, including myoclonic epilepsy, ataxia, psychomotor regression, dementia, and visual impairment. The onset of the disease in the age range from 3-4 years. The disease is caused by the frameshift mutation c.396dupT (p.Val133CysfsTer18) in exon 4 of the CLN6 in a homozygous state, which was detected using targeted next generation sequencing. Diagnosis of NCL is difficult due to the pronounced genetic heterogeneity of the disease, as well as the similarity with other hereditary metabolic diseases in clinical manifestations. The method of DNA diagnostics of NCL type 6 using NGS and direct sequencing according to Sanger has been introduced into the practice of medical genetic counseling.
Collapse
Affiliation(s)
- P I Golikova
- Ammosov North-Eastern Federal University, Yakutsk, Russia
| | - D A Petukhova
- Ammosov North-Eastern Federal University, Yakutsk, Russia
| | - A L Sukhomyasova
- Ammosov North-Eastern Federal University, Yakutsk, Russia.,Republic Hospital No. 1 - National Center of Medicine, Yakutsk, Russia
| | - T Ya Nikolaeva
- Ammosov North-Eastern Federal University, Yakutsk, Russia
| | - E E Gurinova
- Republic Hospital No. 1 - National Center of Medicine, Yakutsk, Russia
| | - R N Ivanova
- Ammosov North-Eastern Federal University, Yakutsk, Russia.,Republic Hospital No. 1 - National Center of Medicine, Yakutsk, Russia
| | - N R Maksimova
- Ammosov North-Eastern Federal University, Yakutsk, Russia
| |
Collapse
|
35
|
Kick GR, Meiman EJ, Sabol JC, Whiting REH, Ota-Kuroki J, Castaner LJ, Jensen CA, Katz ML. Visual system pathology in a canine model of CLN5 neuronal ceroid lipofuscinosis. Exp Eye Res 2021; 210:108686. [PMID: 34216614 PMCID: PMC8429270 DOI: 10.1016/j.exer.2021.108686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/17/2021] [Accepted: 06/28/2021] [Indexed: 10/21/2022]
Abstract
CLN5 neuronal ceroid lipofuscinosis is a hereditary neurodegenerative disease characterized by progressive neurological decline, vision loss and seizures. Visual impairment in children with CLN5 disease is attributed to a progressive decline in retinal function accompanied by retinal degeneration as well as impaired central nervous system function associated with global brain atrophy. We studied visual system pathology in five Golden Retriever littermates homozygous for the CLN5 disease allele previously identified in the breed. The dogs exhibited signs of pronounced visual impairment by 21-22 months of age. Electroretinogram recordings showed a progressive decline in retinal function primarily affecting cone neural pathways. Altered visual evoked potential recordings indicated that disease progression affected visual signal processing in the brain. Aside from several small retinal detachment lesions, no gross retinal abnormalities were observed with in vivo ocular imaging and histologically the retinas did not exhibit apparent abnormalities by 23 months of age. However, there was extensive accumulation of autofluorescent membrane-bound lysosomal storage bodies in almost all retinal layers, as well as in the occipital cortex, by 20 months of age. In the retina, storage was particularly pronounced in retinal ganglion cells, the retinal pigment epithelium and in photoreceptor cells just interior to the outer limiting membrane. The visual system pathology of CLN5-affected Golden Retrievers is similar to that seen early in the human disease. It was not possible to follow the dogs to an advanced stage of disease progression due to the severity of behavioral and motor disease signs by 23 months of age. The findings reported here indicate that canine CLN5 disease will be a useful model of visual system disease in CLN5 neuronal ceroid lipofuscinosis. The baseline data obtained in this investigation will be useful in future therapeutic intervention studies. The findings indicate that there is a fairly broad time frame after disease onset within which treatments could be effective in preserving vision.
Collapse
Affiliation(s)
- Grace Robinson Kick
- Neurodegenerative Diseases Research Laboratory, University of Missouri, Columbia, MO, 65212, USA
| | - Elizabeth J Meiman
- Neurodegenerative Diseases Research Laboratory, University of Missouri, Columbia, MO, 65212, USA
| | - Julianna C Sabol
- Neurodegenerative Diseases Research Laboratory, University of Missouri, Columbia, MO, 65212, USA
| | | | - Juri Ota-Kuroki
- Neurodegenerative Diseases Research Laboratory, University of Missouri, Columbia, MO, 65212, USA
| | - Leilani J Castaner
- Neurodegenerative Diseases Research Laboratory, University of Missouri, Columbia, MO, 65212, USA
| | - Cheryl A Jensen
- Neurodegenerative Diseases Research Laboratory, University of Missouri, Columbia, MO, 65212, USA
| | - Martin L Katz
- Neurodegenerative Diseases Research Laboratory, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
36
|
Carvill GL, Matheny T, Hesselberth J, Demarest S. Haploinsufficiency, Dominant Negative, and Gain-of-Function Mechanisms in Epilepsy: Matching Therapeutic Approach to the Pathophysiology. Neurotherapeutics 2021; 18:1500-1514. [PMID: 34648141 PMCID: PMC8608973 DOI: 10.1007/s13311-021-01137-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2021] [Indexed: 02/04/2023] Open
Abstract
This review summarizes the pathogenic mechanisms that underpin the monogenic epilepsies and discusses the potential of novel precision therapeutics to treat these disorders. Pathogenic mechanisms of epilepsy include recessive (null alleles), haploinsufficiency, imprinting, gain-of-function, and dominant negative effects. Understanding which pathogenic mechanism(s) that underlie each genetic epilepsy is pivotal to design precision therapies that are most likely to be beneficial for the patient. Novel therapeutics discussed include gene therapy, gene editing, antisense oligonucleotides, and protein replacement. Discussions are illustrated and reinforced with examples from the literature.
Collapse
Affiliation(s)
- Gemma L Carvill
- Departments of Neurology, Pharmacology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tyler Matheny
- Department Biochemistry and Molecular Genetics, School of Medicine, RNA Bioscience Initiative, University of Colorado, PO Box 6511, Aurora, CO, USA
| | - Jay Hesselberth
- Department Biochemistry and Molecular Genetics, School of Medicine, RNA Bioscience Initiative, University of Colorado, PO Box 6511, Aurora, CO, USA
| | - Scott Demarest
- Departments of Pediatrics and Neurology, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA.
| |
Collapse
|
37
|
Sondhi D, Kaminsky SM, Hackett NR, Pagovich OE, Rosenberg JB, De BP, Chen A, Van de Graaf B, Mezey JG, Mammen GW, Mancenido D, Xu F, Kosofsky B, Yohay K, Worgall S, Kaner RJ, Souwedaine M, Greenwald BM, Kaplitt M, Dyke JP, Ballon DJ, Heier LA, Kiss S, Crystal RG. Slowing late infantile Batten disease by direct brain parenchymal administration of a rh.10 adeno-associated virus expressing CLN2. Sci Transl Med 2021; 12:12/572/eabb5413. [PMID: 33268510 DOI: 10.1126/scitranslmed.abb5413] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022]
Abstract
Late infantile Batten disease (CLN2 disease) is an autosomal recessive, neurodegenerative lysosomal storage disease caused by mutations in the CLN2 gene encoding tripeptidyl peptidase 1 (TPP1). We tested intraparenchymal delivery of AAVrh.10hCLN2, a nonhuman serotype rh.10 adeno-associated virus vector encoding human CLN2, in a nonrandomized trial consisting of two arms assessed over 18 months: AAVrh.10hCLN2-treated cohort of 8 children with mild to moderate disease and an untreated, Weill Cornell natural history cohort consisting of 12 children. The treated cohort was also compared to an untreated European natural history cohort of CLN2 disease. The vector was administered through six burr holes directly to 12 sites in the brain without immunosuppression. In an additional safety assessment under a separate protocol, five children with severe CLN2 disease were treated with AAVrh.10hCLN2. The therapy was associated with a variety of expected adverse events, none causing long-term disability. Induction of systemic anti-AAVrh.10 immunity was mild. After therapy, the treated cohort had a 1.3- to 2.6-fold increase in cerebral spinal fluid TPP1. There was a slower loss of gray matter volume in four of seven children by MRI and a 42.4 and 47.5% reduction in the rate of decline of motor and language function, compared to Weill Cornell natural history cohort (P < 0.04) and European natural history cohort (P < 0.0001), respectively. Intraparenchymal brain administration of AAVrh.10hCLN2 slowed the progression of disease in children with CLN2 disease. However, improvements in vector design and delivery strategies will be necessary to halt disease progression using gene therapy.
Collapse
Affiliation(s)
- Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Neil R Hackett
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Odelya E Pagovich
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jonathan B Rosenberg
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Bishnu P De
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Alvin Chen
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Benjamin Van de Graaf
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jason G Mezey
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA.,Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
| | - Grace W Mammen
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Denesy Mancenido
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Fang Xu
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Barry Kosofsky
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kaleb Yohay
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Stefan Worgall
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA.,Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Robert J Kaner
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mark Souwedaine
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Bruce M Greenwald
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Michael Kaplitt
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jonathan P Dyke
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Douglas J Ballon
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA.,Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Linda A Heier
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Szilard Kiss
- Department of Ophthalmology, Retina Service, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA. .,Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
38
|
Singh RB, Gupta P, Kartik A, Farooqui N, Singhal S, Shergill S, Singh KP, Agarwal A. Ocular Manifestations of Neuronal Ceroid Lipofuscinoses. Semin Ophthalmol 2021; 36:582-595. [PMID: 34106804 DOI: 10.1080/08820538.2021.1936571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are a group of rare neurodegenerative storage disorders associated with devastating visual prognosis, with an incidence of 1/1,000,000 in the United States and comparatively higher incidence in European countries. The pathophysiological mechanisms causing NCLs occur due to enzymatic or transmembrane defects in various sub-cellular organelles including lysosomes, endoplasmic reticulum, and cytoplasmic vesicles. NCLs are categorized into different types depending upon the underlying cause i.e., soluble lysosomal enzyme deficiencies or non-enzymatic deficiencies (functions of identified proteins), which are sub-divided based on an axial classification system. In this review, we have evaluated the current evidence in the literature and reported the incidence rates, underlying mechanisms and currently available management protocols for these rare set of neuroophthalmological disorders. Additionally, we also highlighted the potential therapies under development that can expand the treatment of these rare disorders beyond symptomatic relief.
Collapse
Affiliation(s)
- Rohan Bir Singh
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.,Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Prakash Gupta
- Department of Internal Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Akash Kartik
- Department of Hepatobiliary and Pancreatic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Naba Farooqui
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sachi Singhal
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sukhman Shergill
- Department of Anesthesiology, Yale-New Haven Hospital, New Haven, CT, USA
| | - Kanwar Partap Singh
- Department of Ophthalmology, Dayanand Medical College & Hospital, Ludhiana, India
| | - Aniruddha Agarwal
- Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
39
|
Murray SJ, Russell KN, Melzer TR, Gray SJ, Heap SJ, Palmer DN, Mitchell NL. Intravitreal gene therapy protects against retinal dysfunction and degeneration in sheep with CLN5 Batten disease. Exp Eye Res 2021; 207:108600. [PMID: 33930398 DOI: 10.1016/j.exer.2021.108600] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 01/01/2023]
Abstract
Neuronal ceroid lipofuscinoses (NCL; Batten disease) are a group of inherited neurodegenerative diseases primarily affecting children. A common feature across most NCLs is the progressive loss of vision. We performed intravitreal injections of self-complementary AAV9 vectors packaged with either ovine CLN5 or CLN6 into one eye of 3-month-old CLN5-/- or CLN6-/- animals, respectively. Electroretinography (ERG) was performed every month following treatment, and retinal histology was assessed post-mortem in the treated compared to untreated eye. In CLN5-/- animals, ERG amplitudes were normalised in the treated eye whilst the untreated eye declined in a similar manner to CLN5 affected controls. In CLN6-/- animals, ERG amplitudes in both eyes declined over time although the treated eye showed a slower decline. Post-mortem examination revealed significant attenuation of retinal atrophy and lysosomal storage body accumulation in the treated eye compared with the untreated eye in CLN5-/- animals. This proof-of-concept study provides the first observation of efficacious intravitreal gene therapy in a large animal model of NCL. In particular, the single administration of AAV9-mediated intravitreal gene therapy can successfully ameliorate retinal deficits in CLN5-/- sheep. Combining ocular gene therapy with brain-directed therapy presents a promising treatment strategy to be used in future sheep trials aiming to halt neurological and retinal disease in CLN5 Batten disease.
Collapse
Affiliation(s)
- Samantha J Murray
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand
| | - Katharina N Russell
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand
| | - Tracy R Melzer
- Department of Medicine, University of Otago, Christchurch and the New Zealand Brain Research Institute, Christchurch, 8011, New Zealand
| | - Steven J Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Stephen J Heap
- McMaster & Heap Veterinary Practice, Christchurch, 8025, New Zealand
| | - David N Palmer
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand; Department of Radiology, University of Otago, Christchurch, 8140, New Zealand
| | - Nadia L Mitchell
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand; Department of Radiology, University of Otago, Christchurch, 8140, New Zealand.
| |
Collapse
|
40
|
Massaro G, Geard AF, Liu W, Coombe-Tennant O, Waddington SN, Baruteau J, Gissen P, Rahim AA. Gene Therapy for Lysosomal Storage Disorders: Ongoing Studies and Clinical Development. Biomolecules 2021; 11:611. [PMID: 33924076 PMCID: PMC8074255 DOI: 10.3390/biom11040611] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Rare monogenic disorders such as lysosomal diseases have been at the forefront in the development of novel treatments where therapeutic options are either limited or unavailable. The increasing number of successful pre-clinical and clinical studies in the last decade demonstrates that gene therapy represents a feasible option to address the unmet medical need of these patients. This article provides a comprehensive overview of the current state of the field, reviewing the most used viral gene delivery vectors in the context of lysosomal storage disorders, a selection of relevant pre-clinical studies and ongoing clinical trials within recent years.
Collapse
Affiliation(s)
- Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Amy F. Geard
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
| | - Wenfei Liu
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Oliver Coombe-Tennant
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Simon N. Waddington
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
- Gene Transfer Technology Group, EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK
| | - Julien Baruteau
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK;
- Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, National Institute of Health Research, University College London, London WC1N 1EH, UK;
| | - Paul Gissen
- Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, National Institute of Health Research, University College London, London WC1N 1EH, UK;
| | - Ahad A. Rahim
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| |
Collapse
|
41
|
Chowdhury EA, Meno-Tetang G, Chang HY, Wu S, Huang HW, Jamier T, Chandran J, Shah DK. Current progress and limitations of AAV mediated delivery of protein therapeutic genes and the importance of developing quantitative pharmacokinetic/pharmacodynamic (PK/PD) models. Adv Drug Deliv Rev 2021; 170:214-237. [PMID: 33486008 DOI: 10.1016/j.addr.2021.01.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/17/2022]
Abstract
While protein therapeutics are one of the most successful class of drug molecules, they are expensive and not suited for treating chronic disorders that require long-term dosing. Adeno-associated virus (AAV) mediated in vivo gene therapy represents a viable alternative, which can deliver the genes of protein therapeutics to produce long-term expression of proteins in target tissues. Ongoing clinical trials and recent regulatory approvals demonstrate great interest in these therapeutics, however, there is a lack of understanding regarding their cellular disposition, whole-body disposition, dose-exposure relationship, exposure-response relationship, and how product quality and immunogenicity affects these important properties. In addition, there is a lack of quantitative studies to support the development of pharmacokinetic-pharmacodynamic models, which can support the discovery, development, and clinical translation of this delivery system. In this review, we have provided a state-of-the-art overview of current progress and limitations related to AAV mediated delivery of protein therapeutic genes, along with our perspective on the steps that need to be taken to improve clinical translation of this therapeutic modality.
Collapse
|
42
|
Piguet F, de Saint Denis T, Audouard E, Beccaria K, André A, Wurtz G, Schatz R, Alves S, Sevin C, Zerah M, Cartier N. The Challenge of Gene Therapy for Neurological Diseases: Strategies and Tools to Achieve Efficient Delivery to the Central Nervous System. Hum Gene Ther 2021; 32:349-374. [PMID: 33167739 DOI: 10.1089/hum.2020.105] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
For more than 10 years, gene therapy for neurological diseases has experienced intensive research growth and more recently therapeutic interventions for multiple indications. Beneficial results in several phase 1/2 clinical studies, together with improved vector technology have advanced gene therapy for the central nervous system (CNS) in a new era of development. Although most initial strategies have focused on orphan genetic diseases, such as lysosomal storage diseases, more complex and widespread conditions like Alzheimer's disease, Parkinson's disease, epilepsy, or chronic pain are increasingly targeted for gene therapy. Increasing numbers of applications and patients to be treated will require improvement and simplification of gene therapy protocols to make them accessible to the largest number of affected people. Although vectors and manufacturing are a major field of academic research and industrial development, there is a growing need to improve, standardize, and simplify delivery methods. Delivery is the major issue for CNS therapies in general, and particularly for gene therapy. The blood-brain barrier restricts the passage of vectors; strategies to bypass this obstacle are a central focus of research. In this study, we present the different ways that can be used to deliver gene therapy products to the CNS. We focus on results obtained in large animals that have allowed the transfer of protocols to human patients and have resulted in the generation of clinical data. We discuss the different routes of administration, their advantages, and their limitations. We describe techniques, equipment, and protocols and how they should be selected for safe delivery and improved efficiency for the next generation of gene therapy trials for CNS diseases.
Collapse
Affiliation(s)
- Françoise Piguet
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Timothée de Saint Denis
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Emilie Audouard
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Kevin Beccaria
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Arthur André
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Neurosurgery, Hôpitaux Universitaires La Pitié-Salpêtrière, Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Guillaume Wurtz
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Raphael Schatz
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Sandro Alves
- BrainVectis-Askbio France, iPeps Paris Brain Institute, Paris, France
| | - Caroline Sevin
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,BrainVectis-Askbio France, iPeps Paris Brain Institute, Paris, France.,APHP, Department of Neurology, Hopital le Kremlin Bicetre, Paris, France
| | - Michel Zerah
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Nathalie Cartier
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
43
|
Kose M, Kose E, Ünalp A, Yılmaz Ü, Edizer S, Tekin HG, Karaoğlu P, Özdemir TR, Er E, Onay H, Yildirim ES. Neuronal ceroid lipofuscinosis: genetic and phenotypic spectrum of 14 patients from Turkey. Neurol Sci 2021; 42:1103-1111. [PMID: 33486620 DOI: 10.1007/s10072-021-05067-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION AND PURPOSE Neuronal ceroid lipofuscinoses (NCLs) is a group of congenital metabolic diseases where the neurodegenerative process with the accumulation of ceroid and lipofuscin autofluorescent storage materials is at the forefront. According to the age of presentation, NCLs are classified as congenital, infantile (INCL), late infantile (LINCL), juvenile (JNCL), and adult (ANCL) NCLs. In our study, it was aimed to discuss the clinical and molecular characteristics of our patients diagnosed with NCL. MATERIAL AND METHOD This is a descriptive cross-sectional study which was conducted in 14 patients from 10 unrelated families who were diagnosed with different types of NCL based on clinical presentation, neuroimaging, biochemical measurements, and molecular analyses, at the department of pediatric metabolism between June 2015 and June 2020. RESULTS A total of 14 patients were diagnosed with different types of NCL. Of those, 4 patients were diagnosed with NCL7 (4/14; 30%), 3/14 (23%) with NCL1, 3/14 (23%) with NCL2, 2/14 (14.2%) with NCL13, and 1/14 (7.1%) with NCL10. Eleven pathogenic variants were detected, 5 of which are novel (c.721G>T [p.Gly241Ter] and c.301G>C [p.Ala146Pro] in MFDS8 gene; c.316C>T [p.Gln106Ter] in PPT1 gene; c.341C>T [p.Ala114Val] in TPP1 gene; c.686A>T [p.Glu229Val] in CTSD gene) CONCLUSION: This study is one of the pioneer comprehensive researches from Turkey that provides information about disease-causing variants and clinical presentation of different and rare types of NCLs. The identification of novel variants and phenotypic expansion is important for genetic counselling in Turkey and expected to improve understanding of NCLs.
Collapse
Affiliation(s)
- Melis Kose
- Department of Pediatrics, Division of Metabolism and Nutrition, Izmir Katip Çelebi University Faculty of Medicine, Ismet Kaptan Street, 35100, Izmir, Turkey.
| | - Engin Kose
- Department of Pediatrics, Division of Pediatric Metabolism and Nutrition, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Aycan Ünalp
- Pediatric Neurology Department, University of Health Sciences, Behçet Uz Children Training and Research Hospital, Izmir, Turkey
| | - Ünsal Yılmaz
- Pediatric Neurology Department, University of Health Sciences, Behçet Uz Children Training and Research Hospital, Izmir, Turkey
| | - Selvinaz Edizer
- Pediatric Neurology Department, University of Health Sciences, Behçet Uz Children Training and Research Hospital, Izmir, Turkey
| | - Hande Gazeteci Tekin
- Faculty of Medicine, Çiğli Research and Training Hospital, Department of Pediatrics, Division of Neurology, Bakırçay University, Izmir, Turkey
| | - Pakize Karaoğlu
- Pediatric Neurology Department, University of Health Sciences, Behçet Uz Children Training and Research Hospital, Izmir, Turkey
| | - Taha Reşid Özdemir
- Genetics Department, University of Health Sciences Tepecik Training & Research Hospital, Izmir, Turkey
| | - Esra Er
- Department of Pediatrics, Division of Nutrition and Metabolism, Izmir Katip Çelebi University, Ataturk Training and Research Hospital, Izmir, Turkey
| | - Hüseyin Onay
- Department of Medical Genetics, Ege University Faculty of Medicine, Izmir, Turkey
| | - Eser Sozmen Yildirim
- Clinical Chemistry Department, Ege University Faculty of Medicine, İzmir, Turkey
| |
Collapse
|
44
|
White KA, Nelvagal HR, Poole TA, Lu B, Johnson TB, Davis S, Pratt MA, Brudvig J, Assis AB, Likhite S, Meyer K, Kaspar BK, Cooper JD, Wang S, Weimer JM. Intracranial delivery of AAV9 gene therapy partially prevents retinal degeneration and visual deficits in CLN6-Batten disease mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:497-507. [PMID: 33665223 PMCID: PMC7887332 DOI: 10.1016/j.omtm.2020.12.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 12/31/2020] [Indexed: 02/07/2023]
Abstract
Batten disease is a family of rare, fatal, neuropediatric diseases presenting with memory/learning decline, blindness, and loss of motor function. Recently, we reported the use of an AAV9-mediated gene therapy that prevents disease progression in a mouse model of CLN6-Batten disease (Cln6 nclf ), restoring lifespans in treated animals. Despite the success of our viral-mediated gene therapy, the dosing strategy was optimized for delivery to the brain parenchyma and may limit the therapeutic potential to other disease-relevant tissues, such as the eye. Here, we examine whether cerebrospinal fluid (CSF) delivery of scAAV9.CB.CLN6 is sufficient to ameliorate visual deficits in Cln6 nclf mice. We show that intracerebroventricular (i.c.v.) delivery of scAAV9.CB.CLN6 completely prevents hallmark Batten disease pathology in the visual processing centers of the brain, preserving neurons of the superior colliculus, thalamus, and cerebral cortex. Importantly, i.c.v.-delivered scAAV9.CB.CLN6 also expresses in many cells throughout the central retina, preserving many photoreceptors typically lost in Cln6 nclf mice. Lastly, scAAV9.CB.CLN6 treatment partially preserved visual acuity in Cln6 nclf mice as measured by optokinetic response. Taken together, we report the first instance of CSF-delivered viral gene reaching and rescuing pathology in both the brain parenchyma and retinal neurons, thereby partially slowing visual deterioration.
Collapse
Affiliation(s)
- Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Hemanth R Nelvagal
- Pediatric Storage Disorders Laboratory, Division of Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Pediatrics, The Lundquist Institute at Harbor-UCLA Medical Center and David Geffen School of Medicine, UCLA, Torrance, CA 90502, USA
| | - Timothy A Poole
- Pediatric Storage Disorders Laboratory, Division of Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Lu
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tyler B Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA.,Amicus Therapeutics, Philadelphia, PA 19104, USA
| | - Samantha Davis
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Melissa A Pratt
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Jon Brudvig
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Ana B Assis
- Department of Pediatrics, The Lundquist Institute at Harbor-UCLA Medical Center and David Geffen School of Medicine, UCLA, Torrance, CA 90502, USA
| | - Shibi Likhite
- Nationwide Children's Hospital. He was involved in AAV9 construct development
| | - Kathrin Meyer
- The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA.,Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Brian K Kaspar
- The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA.,Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Jonathan D Cooper
- Pediatric Storage Disorders Laboratory, Division of Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Pediatrics, The Lundquist Institute at Harbor-UCLA Medical Center and David Geffen School of Medicine, UCLA, Torrance, CA 90502, USA
| | - Shaomei Wang
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA.,Amicus Therapeutics, Philadelphia, PA 19104, USA.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57069, USA
| |
Collapse
|
45
|
Aylward SC, Pindrik J, Abreu NJ, Cherny WB, O’Neal M, de Los Reyes E. Cerliponase alfa for CLN2 disease, a promising therapy. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1856654] [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: 10/22/2022]
Affiliation(s)
- Shawn C. Aylward
- Department of Pediatrics and Neurology, Nationwide Children‘s Hospital, Columbus, OH, USA
| | - Jonathan Pindrik
- Division of Pediatric Neurosurgery, Nationwide Children‘s Hospital, Columbus, OH, USA
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Nicolas J. Abreu
- Department of Pediatrics and Neurology, Nationwide Children‘s Hospital, Columbus, OH, USA
| | - W. Bruce Cherny
- Department of Pediatric Neurosurgery, St. Luke‘s Children‘s Hospital, Boise, ID, USA
| | - Matthew O’Neal
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Emily de Los Reyes
- Department of Pediatrics and Neurology, Nationwide Children‘s Hospital, Columbus, OH, USA
| |
Collapse
|
46
|
Abstract
Neuronal ceroid lipofuscinosis (NCLs) is a group of inherited neurodegenerative lysosomal storage diseases that together represent the most common cause of dementia in children. Phenotypically, patients have visual impairment, cognitive and motor decline, epilepsy, and premature death. A primary challenge is to halt and/or reverse these diseases, towards which developments in potential effective therapies are encouraging. Many treatments, including enzyme replacement therapy (for CLN1 and CLN2 diseases), stem-cell therapy (for CLN1, CLN2, and CLN8 diseases), gene therapy vector (for CLN1, CLN2, CLN3, CLN5, CLN6, CLN7, CLN10, and CLN11 diseases), and pharmacological drugs (for CLN1, CLN2, CLN3, and CLN6 diseases) have been evaluated for safety and efficacy in pre-clinical and clinical studies. Currently, cerliponase alpha for CLN2 disease is the only approved therapy for NCL. Lacking is any study of potential treatments for CLN4, CLN9, CLN12, CLN13 or CLN14 diseases. This review provides an overview of genetics for each CLN disease, and we discuss the current understanding from pre-clinical and clinical study of potential therapeutics. Various therapeutic interventions have been studied in many experimental animal models. Combination of treatments may be useful to slow or even halt disease progression; however, few therapies are unlikely to even partially reverse the disease and a complete reversal is currently improbable. Early diagnosis to allow initiation of therapy, when indicated, during asymptomatic stages is more important than ever.
Collapse
|
47
|
Johnson TB, White KA, Brudvig JJ, Cain JT, Langin L, Pratt MA, Booth CD, Timm DJ, Davis SS, Meyerink B, Likhite S, Meyer K, Weimer JM. AAV9 Gene Therapy Increases Lifespan and Treats Pathological and Behavioral Abnormalities in a Mouse Model of CLN8-Batten Disease. Mol Ther 2020; 29:162-175. [PMID: 33010819 DOI: 10.1016/j.ymthe.2020.09.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/24/2020] [Accepted: 09/20/2020] [Indexed: 12/26/2022] Open
Abstract
CLN8 disease is a rare form of neuronal ceroid lipofuscinosis caused by biallelic mutations in the CLN8 gene, which encodes a transmembrane endoplasmic reticulum protein involved in trafficking of lysosomal enzymes. CLN8 disease patients present with myoclonus, tonic-clonic seizures, and progressive declines in cognitive and motor function, with many cases resulting in premature death early in life. There are currently no treatments that can cure the disease or substantially slow disease progression. Using a mouse model of CLN8 disease, we tested the safety and efficacy of an intracerebroventricularly (i.c.v.) delivered self-complementary adeno-associated virus serotype 9 (scAAV9) gene therapy vector driving expression of human CLN8. A single neonatal injection was safe and well tolerated, resulting in robust transgene expression throughout the CNS from 4 to 24 months, reducing histopathological and behavioral hallmarks of the disease and restoring lifespan from 10 months in untreated animals to beyond 24 months of age in treated animals. While it is unclear whether some of these behavioral improvements relate to preserved visual function, improvements in learning/memory, or other central or peripheral benefits, these results demonstrate, by far, the most successful degree of rescue reported in an animal model of CLN8 disease, and they support further development of gene therapy for this disorder.
Collapse
Affiliation(s)
- Tyler B Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA; Amicus Therapeutics, Philadelphia, PA, USA
| | - Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Jon J Brudvig
- 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; Amicus Therapeutics, Philadelphia, PA, USA
| | - Logan Langin
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Melissa A Pratt
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Clarissa D Booth
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Derek J Timm
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Samantha S Davis
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Brandon Meyerink
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Shibi Likhite
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Kathrin Meyer
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA; Amicus Therapeutics, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Nelvagal HR, Lange J, Takahashi K, Tarczyluk-Wells MA, Cooper JD. Pathomechanisms in the neuronal ceroid lipofuscinoses. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165570. [DOI: 10.1016/j.bbadis.2019.165570] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 12/22/2022]
|
49
|
Butz ES, Chandrachud U, Mole SE, Cotman SL. Moving towards a new era of genomics in the neuronal ceroid lipofuscinoses. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165571. [DOI: 10.1016/j.bbadis.2019.165571] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
|
50
|
Samaranch L, Pérez-Cañamás A, Soto-Huelin B, Sudhakar V, Jurado-Arjona J, Hadaczek P, Ávila J, Bringas JR, Casas J, Chen H, He X, Schuchman EH, Cheng SH, Forsayeth J, Bankiewicz KS, Ledesma MD. Adeno-associated viral vector serotype 9-based gene therapy for Niemann-Pick disease type A. Sci Transl Med 2020; 11:11/506/eaat3738. [PMID: 31434754 DOI: 10.1126/scitranslmed.aat3738] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 01/23/2019] [Accepted: 07/29/2019] [Indexed: 11/02/2022]
Abstract
Niemann-Pick disease type A (NPD-A) is a lysosomal storage disorder characterized by neurodegeneration and early death. It is caused by loss-of-function mutations in the gene encoding for acid sphingomyelinase (ASM), which hydrolyzes sphingomyelin into ceramide. Here, we evaluated the safety of cerebellomedullary (CM) cistern injection of adeno-associated viral vector serotype 9 encoding human ASM (AAV9-hASM) in nonhuman primates (NHP). We also evaluated its therapeutic benefit in a mouse model of the disease (ASM-KO mice). We found that CM injection in NHP resulted in widespread transgene expression within brain and spinal cord cells without signs of toxicity. CM injection in the ASM-KO mouse model resulted in hASM expression in cerebrospinal fluid and in different brain areas without triggering an inflammatory response. In contrast, direct cerebellar injection of AAV9-hASM triggered immune response. We also identified a minimally effective therapeutic dose for CM injection of AAV9-hASM in mice. Two months after administration, the treatment prevented motor and memory impairment, sphingomyelin (SM) accumulation, lysosomal enlargement, and neuronal death in ASM-KO mice. ASM activity was also detected in plasma from AAV9-hASM CM-injected ASM-KO mice, along with reduced SM amount and decreased inflammation in the liver. Our results support CM injection for future AAV9-based clinical trials in NPD-A as well as other lysosomal storage brain disorders.
Collapse
Affiliation(s)
- Lluis Samaranch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103, USA
| | | | | | - Vivek Sudhakar
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103, USA
| | | | - Piotr Hadaczek
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103, USA
| | - Jesús Ávila
- Centro Biologia Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
| | - John R Bringas
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103, USA
| | | | | | - Xingxuan He
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Edward H Schuchman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - John Forsayeth
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103, USA
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103, USA.
| | | |
Collapse
|