1
|
Said M, Jing J, Montigon O, Collomb N, Vossier F, Chovelon B, El Amine B, Jeacomine I, Lemasson B, Barbier EL, Detante O, Rome C, Auzély-Velty R. A T1 MRI detectable hyaluronic acid hydrogel for in vivo tracking after intracerebral injection in stroke. J Mater Chem B 2025; 13:4103-4117. [PMID: 40042261 DOI: 10.1039/d4tb02722a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Injectable hydrogels have emerged as a promising strategy for treating stroke and neurodegenerative diseases, but their effectiveness depends on precise injection, defect filling, and long-term retention at the target site. While MRI can help visualize hydrogels, distinguishing them from fluid-filled spaces, like a post-stroke cavity at a chronic stage, is challenging owing to their high water content and similar MR properties. In this study, a T1 MRI detectable hyaluronic acid (HA) hydrogel that is injectable and self-healing was developed for in vivo tracking after intracerebral injection in stroke. This HA hydrogel was functionalized with a thermodynamically stable and kinetically inert gadolinium(III) complex for monitoring its long-term fate in the brain with T1-contrast enhanced MRI. The dynamic covalent cross-links based on boronate ester bonds in the hydrogel network ensured precise injection and instantaneous self-healing. The HA network did not induce adverse tissue response and was biocompatible with therapeutic cells (human adipose stromal/stem cells). Furthermore, this labeling strategy enabled accurate tracking of hydrogel distribution and degradation in stroke condition, allowing a better assessment of efficacy and safety. This MRI-visible hydrogel has significant potential as a scaffold for stem cells, growth factors, and/or drugs, paving the way for more effective treatments for brain disorders.
Collapse
Affiliation(s)
- Moustoifa Said
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
- Univ. Grenoble Alpes, Centre de Recherches sur les Macromolécules Végétales (CERMAV-CNRS), 38041 Grenoble, France.
| | - Jing Jing
- Univ. Grenoble Alpes, Centre de Recherches sur les Macromolécules Végétales (CERMAV-CNRS), 38041 Grenoble, France.
| | - Olivier Montigon
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
- Univ. Grenoble Alpes, IRMaGe, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Nora Collomb
- Univ. Grenoble Alpes, IRMaGe, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Frédérique Vossier
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Benoît Chovelon
- Univ. Grenoble-Alpes, Département de Pharmacochimie Moléculaire UMR 5063, 38400 Grenoble, France
- CHU de Grenoble-Alpes, Institut de Biologie et Pathologie, 38700 La Tronche, France
| | - Bayan El Amine
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Isabelle Jeacomine
- Univ. Grenoble Alpes, Centre de Recherches sur les Macromolécules Végétales (CERMAV-CNRS), 38041 Grenoble, France.
| | - Benjamin Lemasson
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Emmanuel Luc Barbier
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
- Univ. Grenoble Alpes, IRMaGe, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Olivier Detante
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
- CHU Grenoble Alpes, Stroke Unit, Department of Neurology, 38043 Grenoble, France
| | - Claire Rome
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Rachel Auzély-Velty
- Univ. Grenoble Alpes, Centre de Recherches sur les Macromolécules Végétales (CERMAV-CNRS), 38041 Grenoble, France.
| |
Collapse
|
2
|
Eisel MLS, Burns M, Ashizawa T, Byrne B, Corti M, Subramony SH. Emerging therapies in hereditary ataxias. Trends Mol Med 2025; 31:181-194. [PMID: 39153956 DOI: 10.1016/j.molmed.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
Recent investigations have defined the pathophysiological basis of many hereditary ataxias (HAs), including loss-of-function as well as gain-of-function mechanisms at either the RNA or protein level. Preclinical studies have assessed gene editing, gene and protein replacement, gene enhancement, and gene knockdown strategies. Methodologies include viral vector delivery of genes, oligonucleotide therapies, cell-penetrating peptides, synthetic transcription factors, and technologies to deliver therapies to defined targets. In this review, we focus on Friedreich ataxia (FRDA) and the polyglutamine ataxias in which translational research is active. However, much remains to be done to identify safe and effective molecules, create ideal delivery methods, and perform innovative clinical trials to prove the safety and efficacy of treatments for these rare but devastating diseases.
Collapse
Affiliation(s)
- Mallory L S Eisel
- Department of Neurology and the Fixel Institute for Neurological Disorders, University of Florida College of Medicine, Gainesville, FL, USA
| | - Matthew Burns
- Department of Neurology and the Fixel Institute for Neurological Disorders, University of Florida College of Medicine, Gainesville, FL, USA
| | - Tetsuo Ashizawa
- Stanley H. Appel Department of Neurology, Weill Cornell Medicine at Houston Methodist Hospital, Houston, TX, USA
| | - Barry Byrne
- Department of Pediatrics and the Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Manuela Corti
- Department of Pediatrics and the Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Sub H Subramony
- Department of Neurology and the Fixel Institute for Neurological Disorders, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
3
|
Wiseman JP, Scarrott JM, Alves-Cruzeiro J, Saffari A, Böger C, Karyka E, Dawes E, Davies AK, Marchi PM, Graves E, Fernandes F, Yang ZL, Coldicott I, Hirst J, Webster CP, Highley JR, Hackett N, Angyal A, Silva TD, Higginbottom A, Shaw PJ, Ferraiuolo L, Ebrahimi-Fakhari D, Azzouz M. Pre-clinical development of AP4B1 gene replacement therapy for hereditary spastic paraplegia type 47. EMBO Mol Med 2024; 16:2882-2917. [PMID: 39358605 PMCID: PMC11554807 DOI: 10.1038/s44321-024-00148-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Spastic paraplegia 47 (SPG47) is a neurological disorder caused by mutations in the adaptor protein complex 4 β1 subunit (AP4B1) gene leading to AP-4 complex deficiency. SPG47 is characterised by progressive spastic paraplegia, global developmental delay, intellectual disability and epilepsy. Gene therapy aimed at restoring functional AP4B1 protein levels is a rational therapeutic strategy to ameliorate the disease phenotype. Here we report that a single delivery of adeno-associated virus serotype 9 expressing hAP4B1 (AAV9/hAP4B1) into the cisterna magna leads to widespread gene transfer and restoration of various hallmarks of disease, including AP-4 cargo (ATG9A) mislocalisation, calbindin-positive spheroids in the deep cerebellar nuclei, anatomical brain defects and motor dysfunction, in an SPG47 mouse model. Furthermore, AAV9/hAP4B1-based gene therapy demonstrated a restoration of plasma neurofilament light (NfL) levels of treated mice. Encouraged by these preclinical proof-of-concept data, we conducted IND-enabling studies, including immunogenicity and GLP non-human primate (NHP) toxicology studies. Importantly, NHP safety and biodistribution study revealed no significant adverse events associated with the therapeutic intervention. These findings provide evidence of both therapeutic efficacy and safety, establishing a robust basis for the pursuit of an IND application for clinical trials targeting SPG47 patients.
Collapse
Affiliation(s)
- Jessica P Wiseman
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - Joseph M Scarrott
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Gene Therapy Innovation & Manufacturing Centre (GTIMC), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - João Alves-Cruzeiro
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Afshin Saffari
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Child Neurology and Inherited Metabolic Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Cedric Böger
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Evangelia Karyka
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Gene Therapy Innovation & Manufacturing Centre (GTIMC), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Emily Dawes
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Alexandra K Davies
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Paolo M Marchi
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Emily Graves
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Fiona Fernandes
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Zih-Liang Yang
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - Ian Coldicott
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - Jennifer Hirst
- Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Christopher P Webster
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - J Robin Highley
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | | | - Adrienn Angyal
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Thushan de Silva
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
- Sheffield NIHR Biomedical Research Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Glossop Road, Sheffield, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Darius Ebrahimi-Fakhari
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK.
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK.
- Gene Therapy Innovation & Manufacturing Centre (GTIMC), Division of Neuroscience, University of Sheffield, Sheffield, UK.
| |
Collapse
|
4
|
Massaro G, Geard AF, Nelvagal HR, Gore K, Clemo NK, Waddington SN, Rahim AA. Comparison of different promoters to improve AAV vector-mediated gene therapy for neuronopathic Gaucher disease. Hum Mol Genet 2024; 33:1467-1480. [PMID: 38757200 PMCID: PMC11336133 DOI: 10.1093/hmg/ddae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
Gaucher Disease (GD) is an inherited metabolic disorder caused by mutations in the GBA1 gene. It can manifest with severe neurodegeneration and visceral pathology. The most acute neuronopathic form (nGD), for which there are no curative therapeutic options, is characterised by devastating neuropathology and death during infancy. In this study, we investigated the therapeutic benefit of systemically delivered AAV9 vectors expressing the human GBA1 gene at two different doses comparing a neuronal-selective promoter with ubiquitous promoters. Our results highlight the importance of a careful evaluation of the promoter sequence used in gene delivery vectors, suggesting a neuron-targeted therapy leading to high levels of enzymatic activity in the brain but lower GCase expression in the viscera, might be the optimal therapeutic strategy for nGD.
Collapse
Affiliation(s)
- Giulia Massaro
- UCL School of Pharmacy, University College London, 29-38 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Amy F Geard
- UCL School of Pharmacy, University College London, 29-38 Brunswick Square, London, WC1N 1AX, United Kingdom
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand Medical, School, 7 York Road, Parktown 2193, South Africa
| | - Hemanth R Nelvagal
- UCL School of Pharmacy, University College London, 29-38 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Katrina Gore
- Apollo Therapeutics, Stevenage Bioscience Catalyst, 50-60 Station Road, Cambridge, CB1 2JH, United Kingdom
| | - Nadine K Clemo
- Apollo Therapeutics, Stevenage Bioscience Catalyst, 50-60 Station Road, Cambridge, CB1 2JH, United Kingdom
| | - Simon N Waddington
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand Medical, School, 7 York Road, Parktown 2193, South Africa
- UCL EGA Institute for Women's Health, University College London, Medical School Building, 74 Huntley Street, London, WC1E 6AU, United Kingdom
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, 29-38 Brunswick Square, London, WC1N 1AX, United Kingdom
| |
Collapse
|
5
|
Burla GKR, Shrestha D, Bowen M, Horvath JD, Martin BA. Evaluating the effect of injection protocols on intrathecal solute dispersion in non-human primates: an in vitro study using a cynomolgus cerebrospinal fluid system. Fluids Barriers CNS 2024; 21:61. [PMID: 39061067 PMCID: PMC11282645 DOI: 10.1186/s12987-024-00556-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 06/16/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Achieving effective drug delivery to the central nervous system (CNS) remains a challenge for treating neurological disorders. Intrathecal (IT) delivery, which involves direct injection into the cerebrospinal fluid (CSF), presents a promising strategy. Large animal studies are important to assess the safety and efficacy of most drugs and treatments and translate the data to humans. An understanding of the influence of IT injection parameters on solute distribution within the CNS is essential to optimize preclinical research, which would potentially help design human clinical studies. METHODS A three-dimensional (3D) in vitro model of a cynomolgus monkey, based on MRI data, was developed to evaluate the impact of lumbar injection parameters on intrathecal solute dispersion. The parameters evaluated were (a) injection location, (b) bolus volume, (c) flush volume, (d) bolus rate, and (e) flush rate. To simulate the CSF flow within the subarachnoid space (SAS), an idealized CSF flow waveform with both cardiac and respiratory-induced components was input into the model. A solution of fluorescein drug surrogate tracer was administered in the lumbar region of the 3D in vitro model filled with deionized water. After injection of the tracer, the CSF system wide-solute dispersion was imaged using high-resolution cameras every thirty seconds for a duration of three hours. To ensure repeatability each injection protocol was repeated three times. For each protocol, the average spatial-temporal distribution over three hours post-injection, the area under the curve (AUC), and the percent injected dose (%ID) to extra-axial CSF (eaCSF) at three hours were determined. RESULTS The changes to the lumbar injection parameters led to variations in solute distribution along the neuro-axis. Specifically, injection location showed the most impact, enhancing the delivery to the eaCSF up to + 10.5%ID (p = 0.0282) at three hours post-injection. Adding a post-injection flush of 1.5 ml at 1 ml/min increased the solute delivery to the eaCSF by + 6.5%ID (p = 0.0218), while the larger bolus volume resulted in a + 2.3%ID (p = 0.1910) increase. The bolus and flush rates analyzed had minimal, statistically non-significant effects. CONCLUSION These results predict the effects of lumbar injection parameters on solute distribution in the intrathecal space in NHPs. Specifically, the choice of injection location, flush, and bolus volume significantly improved solute delivery to eaCSF. The in vitro NHP CSF model and results offer a system to help predict and optimize IT delivery protocols for pre-clinical NHP studies.
Collapse
Affiliation(s)
- Goutham Kumar Reddy Burla
- Department of Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MC1122, Moscow, ID, 83844, USA
| | - Dev Shrestha
- Department of Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MC1122, Moscow, ID, 83844, USA
| | - Mayumi Bowen
- Genentech, Inc., a member of the Roche Group, South San Francisco, CA, USA
| | - Joshua D Horvath
- Genentech, Inc., a member of the Roche Group, South San Francisco, CA, USA
| | - Bryn A Martin
- Department of Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MC1122, Moscow, ID, 83844, USA.
- Alcyone Therapeutics Inc., Lowell, MA, USA.
- Flux Neuroscience, LLC., Troy, ID, USA.
| |
Collapse
|
6
|
Patel RV, Nanda P, Richardson RM. Neurosurgical gene therapy for central nervous system diseases. Neurotherapeutics 2024; 21:e00434. [PMID: 39191071 PMCID: PMC11445594 DOI: 10.1016/j.neurot.2024.e00434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/16/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Viral vector mediated gene therapies for neurodegenerative and neurodevelopmental conditions that require neurosurgical administration continue to expand. We systematically reviewed the National Institutes of Health (NIH) ClinicalTrials.gov database to identify all clinical trials studying in-vivo viral vector mediated gene therapies targeted to the CNS for neurodegenerative and neurodevelopmental diseases. We isolated studies which delivered therapies using neurosurgical approaches: intracisternal, intraventricular, and/or intraparenchymal. Clinical trials primarily registered in international countries were included if they were referenced by an NIH registered clinical trial. We performed a scoping review to identify the preclinical studies that supported each human clinical trial. Key preclinical and clinical data were aggregated to characterize vector capsid design, delivery methods, gene expression profile, and clinical benefit. A total of 64 clinical trials were identified in active, completed, terminated, and long-term follow-up stages. A range of CNS conditions across pediatric and adult populations are being studied with CNS targeted viral vector gene therapy, including Alzheimer's disease, Parkinson's disease, AADC deficiency, sphingolipidoses, mucopolysaccharidoses, neuronal ceroid lipofuscinoses, spinal muscular atrophy, adrenoleukodystrophy, Canavan disease, frontotemporal dementia, Huntington's disease, Rett syndrome, Dravet syndrome, mesial temporal lobe epilepsy, and glutaric acidemia. Adeno-associated viral vectors (AAVs) were utilized by the majority of tested therapies, with vector serotypes, regulatory elements, delivery methods, and vector monitoring varying based on the disease being studied. Intraparenchymal delivery has evolved significantly, with MRI-guided convection-enhanced delivery established as a gold standard method for pioneering novel gene targets.
Collapse
Affiliation(s)
- Ruchit V Patel
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Pranav Nanda
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - R Mark Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Stavrou M, Georgiou E, Kleopa KA. Lumbar Intrathecal Injection in Adult and Neonatal Mice. Curr Protoc 2024; 4:e1091. [PMID: 38923413 DOI: 10.1002/cpz1.1091] [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] [Indexed: 06/28/2024]
Abstract
This article describes a step-by-step process of lumbar intrathecal injection of Evans blue dye and AAV9-EGFP in adult (2-month-old) and neonatal (postnatal day 10) mice. Intrathecal injection is a clinically translatable technique that has already been extensively applied in humans. In mice, intrathecal injection is considered a challenging procedure that requires a trained and experienced researcher. For both adult and neonatal mice, lumbar intrathecal injection is directed into the L5-L6 intervertebral space. Intrathecally injected material enters the cerebrospinal fluid (CSF) within the intrathecal space from where it can directly access the central nervous system (CNS) parenchyma. Simultaneously, intrathecally injected material exits the CSF with pressure gradient and enters the endoneurial fluid and ultimately the peripheral nerves. While in the CSF, the injectable material also enters the bloodstream and systemic circulation through the arachnoid villi. A successful lumbar intrathecal injection results in adequate biodistribution of the injectable material in the CNS, PNS, and peripheral organs. When correctly applied, this technique is considered as minimally invasive and non-disruptive and can be used for the lumbar delivery of any solute. © 2024 Wiley Periodicals LLC. Basic Protocol 1: C57BL/6 adult and P10 mice lumbar intrathecal injection Basic Protocol 2: Tissue collection and preparation for evaluating Evans blue dye diffusion Basic Protocol 3: Tissue collection and preparation for immunohistochemistry staining Basic Protocol 4: Tissue collection and vector genome copy number analysis.
Collapse
Affiliation(s)
- Marina Stavrou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Elena Georgiou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Center for Neuromuscular Disorders, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
8
|
Zhou XA, Man W, Liu X, Choi S, Jiang Y, Hike D, Cid LG, Lin C, Nedergaard M, Yu X. Mapping glymphatic solute transportation through the perivascular space of hippocampal arterioles with 14 Tesla MRI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.14.557634. [PMID: 38826414 PMCID: PMC11142069 DOI: 10.1101/2023.09.14.557634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The perivascular space (PVS) plays a crucial role in facilitating the clearance of waste products and the exchange of cerebrospinal fluid and interstitial fluid in the central nervous system. While optical imaging methods identify the glymphatic transport of fluorescent tracers through PVS of surface-diving arteries, their limited depth penetration impedes the study of glymphatic dynamics in deep brain regions. In this study, we introduced a novel high-resolution dynamic contrast-enhanced MRI mapping approach based on single-vessel multi-gradient-echo methods. This technique allowed the differentiation of penetrating arterioles and venules from adjacent parenchymal tissue voxels and enabled the detection of Gd-enhanced signals coupled to PVS of penetrating arterioles in the deep cortex and hippocampus. By directly infusing Gd into the lateral ventricle, we eliminated delays in cerebrospinal fluid flow and focused on PVS Gd transport through PVS of hippocampal arterioles. The study revealed significant PVS-specific Gd signal enhancements, shedding light on glymphatic function in deep brain regions. These findings advance our understanding of brain-wide glymphatic dynamics and hold potential implications for neurological conditions characterized by impaired waste clearance, warranting further exploration of their clinical relevance and therapeutic applications.
Collapse
|
9
|
Sevigny J, Uspenskaya O, Heckman LD, Wong LC, Hatch DA, Tewari A, Vandenberghe R, Irwin DJ, Saracino D, Le Ber I, Ahmed R, Rohrer JD, Boxer AL, Boland S, Sheehan P, Brandes A, Burstein SR, Shykind BM, Kamalakaran S, Daniels CW, David Litwack E, Mahoney E, Velaga J, McNamara I, Sondergaard P, Sajjad SA, Kobayashi YM, Abeliovich A, Hefti F. Progranulin AAV gene therapy for frontotemporal dementia: translational studies and phase 1/2 trial interim results. Nat Med 2024; 30:1406-1415. [PMID: 38745011 PMCID: PMC11108785 DOI: 10.1038/s41591-024-02973-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 04/03/2024] [Indexed: 05/16/2024]
Abstract
GRN mutations cause progranulin haploinsufficiency, which eventually leads to frontotemporal dementia (FTD-GRN). PR006 is an investigational gene therapy delivering the granulin gene (GRN) using an adeno-associated virus serotype 9 (AAV9) vector. In non-clinical studies, PR006 transduced neurons derived from induced pluripotent stem cells of patients with FTD-GRN, resulted in progranulin expression and improvement of lipofuscin, lysosomal and neuroinflammation pathologies in Grn-knockout mice, and was well tolerated except for minimal, asymptomatic dorsal root ganglionopathy in non-human primates. We initiated a first-in-human phase 1/2 open-label trial. Here we report results of a pre-specified interim analysis triggered with the last treated patient of the low-dose cohort (n = 6) reaching the 12-month follow-up timepoint. We also include preliminary data from the mid-dose cohort (n = 7). Primary endpoints were safety, immunogenicity and change in progranulin levels in cerebrospinal fluid (CSF) and blood. Secondary endpoints were Clinical Dementia Rating (CDR) plus National Alzheimer's Disease Coordinating Center (NACC) Frontotemporal Lobar Degeneration (FTLD) rating scale and levels of neurofilament light chain (NfL). One-time administration of PR006 into the cisterna magna was generally safe and well tolerated. All patients developed treatment-emergent anti-AAV9 antibodies in the CSF, but none developed anti-progranulin antibodies. CSF pleocytosis was the most common PR006-related adverse event. Twelve serious adverse events occurred, mostly unrelated to PR006. Deep vein thrombosis developed in three patients. There was one death (unrelated) occurring 18 months after treatment. CSF progranulin increased after PR006 treatment in all patients; blood progranulin increased in most patients but only transiently. NfL levels transiently increased after PR006 treatment, likely reflecting dorsal root ganglia toxicity. Progression rates, based on the CDR scale, were within the broad ranges reported for patients with FTD. These data provide preliminary insights into the safety and bioactivity of PR006. Longer follow-up and additional studies are needed to confirm the safety and potential efficacy of PR006. ClinicalTrials.gov identifier: NCT04408625 .
Collapse
Affiliation(s)
- Jeffrey Sevigny
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA.
| | - Olga Uspenskaya
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Laura Dean Heckman
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Li Chin Wong
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Daniel A Hatch
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Ambika Tewari
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Rik Vandenberghe
- Neurology Service, University Hospitals Leuven, Leuven, Belgium and Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - David J Irwin
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Dario Saracino
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, Inserm, CNRS UMR 7225 APHP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, Inserm, CNRS UMR 7225 APHP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Rebekah Ahmed
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Center, UCL Queen Square Institute of Neurology, London, UK
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Sebastian Boland
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Patricia Sheehan
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Alissa Brandes
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Suzanne R Burstein
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Benjamin M Shykind
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Sitharthan Kamalakaran
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Carter W Daniels
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - E David Litwack
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Erin Mahoney
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Jenny Velaga
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Ilan McNamara
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Patricia Sondergaard
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Syed A Sajjad
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Yvonne M Kobayashi
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Asa Abeliovich
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Franz Hefti
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| |
Collapse
|
10
|
Daci R, Flotte TR. Delivery of Adeno-Associated Virus Vectors to the Central Nervous System for Correction of Single Gene Disorders. Int J Mol Sci 2024; 25:1050. [PMID: 38256124 PMCID: PMC10816966 DOI: 10.3390/ijms25021050] [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: 11/30/2023] [Revised: 12/26/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Genetic disorders of the central nervous system (CNS) comprise a significant portion of disability in both children and adults. Several preclinical animal models have shown effective adeno-associated virus (AAV) mediated gene transfer for either treatment or prevention of autosomal recessive genetic disorders. Owing to the intricacy of the human CNS and the blood-brain barrier, it is difficult to deliver genes, particularly since the expression of any given gene may be required in a particular CNS structure or cell type at a specific time during development. In this review, we analyzed delivery methods for AAV-mediated gene therapy in past and current clinical trials. The delivery routes analyzed were direct intraparenchymal (IP), intracerebroventricular (ICV), intra-cisterna magna (CM), lumbar intrathecal (IT), and intravenous (IV). The results demonstrated that the dose used in these routes varies dramatically. The average total doses used were calculated and were 1.03 × 1013 for IP, 5.00 × 1013 for ICV, 1.26 × 1014 for CM, and 3.14 × 1014 for IT delivery. The dose for IV delivery varies by patient weight and is 1.13 × 1015 IV for a 10 kg infant. Ultimately, the choice of intervention must weigh the risk of an invasive surgical procedure to the toxicity and immune response associated with a high dose vector.
Collapse
Affiliation(s)
- Rrita Daci
- Department of Neurosurgery, University of Massachusetts Chan Medical School, 55 N Lake Ave, Worcester, MA 01655, USA;
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Terence R. Flotte
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
- Department of Pediatrics, University of Massachusetts Chan Medical School, 55 N Lake Ave, Worcester, MA 01655, USA
| |
Collapse
|
11
|
Ingusci S, Hall BL, Goins WF, Cohen JB, Glorioso JC. Viral vectors for gene delivery to the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:59-81. [PMID: 39341663 DOI: 10.1016/b978-0-323-90120-8.00001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Brain diseases with a known or suspected genetic basis represent an important frontier for advanced therapeutics. The central nervous system (CNS) is an intricate network in which diverse cell types with multiple functions communicate via complex signaling pathways, making therapeutic intervention in brain-related diseases challenging. Nevertheless, as more information on the molecular genetics of brain-related diseases becomes available, genetic intervention using gene therapeutic strategies should become more feasible. There remain, however, several significant hurdles to overcome that relate to (i) the development of appropriate gene vectors and (ii) methods to achieve local or broad vector delivery. Clearly, gene delivery tools must be engineered for distribution to the correct cell type in a specific brain region and to accomplish therapeutic transgene expression at an appropriate level and duration. They also must avoid all toxicity, including the induction of inflammatory responses. Over the last 40 years, various types of viral vectors have been developed as tools to introduce therapeutic genes into the brain, primarily targeting neurons. This review describes the most prominent vector systems currently approaching clinical application for CNS disorders and highlights both remaining challenges as well as improvements in vector designs that achieve greater safety, defined tropism, and therapeutic gene expression.
Collapse
Affiliation(s)
- Selene Ingusci
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bonnie L Hall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - William F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Justus B Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
12
|
Pacak CA, Suzuki-Hatano S, Khadir F, Daugherty AL, Sriramvenugopal M, Gosiker BJ, Kang PB, Cade WT. One episode of low intensity aerobic exercise prior to systemic AAV9 administration augments transgene delivery to the heart and skeletal muscle. J Transl Med 2023; 21:748. [PMID: 37875924 PMCID: PMC10598899 DOI: 10.1186/s12967-023-04626-1] [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: 08/11/2023] [Accepted: 10/13/2023] [Indexed: 10/26/2023] Open
Abstract
INTRODUCTION The promising potential of adeno-associated virus (AAV) gene delivery strategies to treat genetic disorders continues to grow with an additional three AAV-based therapies recently approved by the Food and Drug Administration and dozens of others currently under evaluation in clinical trials. With these developments, it has become increasingly apparent that the high doses currently needed for efficacy carry risks of toxicity and entail enormous manufacturing costs, especially for clinical grade products. Strategies to increase the therapeutic efficacy of AAV-mediated gene delivery and reduce the minimal effective dose would have a substantial impact on this field. We hypothesized that an exercise-induced redistribution of tissue perfusion in the body to favor specific target organs via acute aerobic exercise prior to systemic intravenous (IV) AAV administration could increase efficacy. BACKGROUND Aerobic exercise triggers an array of downstream physiological effects including increased perfusion of heart and skeletal muscle, which we expected could enhance AAV transduction. Prior preclinical studies have shown promising results for a gene therapy approach to treat Barth syndrome (BTHS), a rare monogenic cardioskeletal myopathy, and clinical studies have shown the benefit of low intensity exercise in these patients, making this a suitable disease in which to test the ability of aerobic exercise to enhance AAV transduction. METHODS Wild-type (WT) and BTHS mice were either systemically administered AAV9 or completed one episode of low intensity treadmill exercise immediately prior to systemic administration of AAV9. RESULTS We demonstrate that a single episode of acute low intensity aerobic exercise immediately prior to IV AAV9 administration improves marker transgene delivery in WT mice as compared to mice injected without the exercise pre-treatment. In BTHS mice, prior exercise improved transgene delivery and additionally increased improvement in mitochondrial gene transcription levels and mitochondrial function in the heart and gastrocnemius muscles as compared to mice treated without exercise. CONCLUSIONS Our findings suggest that one episode of acute low intensity aerobic exercise improves AAV9 transduction of heart and skeletal muscle. This low-risk, cost effective intervention could be implemented in clinical trials of individuals with inherited cardioskeletal disease as a potential means of improving patient safety for human gene therapy.
Collapse
Affiliation(s)
- Christina A Pacak
- Paul and Sheila Wellstone Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, 420 Delaware St SE, Minneapolis, MN, 55455, USA.
| | - Silveli Suzuki-Hatano
- College of Medicine, Department of Pediatrics, University of Florida, Gainesville, USA
| | - Fatemeh Khadir
- Paul and Sheila Wellstone Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, 420 Delaware St SE, Minneapolis, MN, 55455, USA
| | - Audrey L Daugherty
- Paul and Sheila Wellstone Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, 420 Delaware St SE, Minneapolis, MN, 55455, USA
| | | | - Bennett J Gosiker
- College of Medicine, Department of Pediatrics, University of Florida, Gainesville, USA
| | - Peter B Kang
- Paul and Sheila Wellstone Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, 420 Delaware St SE, Minneapolis, MN, 55455, USA
| | - William Todd Cade
- Physical Therapy Division, Department of Orthopaedic Surgery, Duke University School of Medicine, 311 Trent Drive, Durham, NC, 27710, USA.
| |
Collapse
|
13
|
Powers S, Likhite S, Gadalla KK, Miranda CJ, Huffenberger AJ, Dennys C, Foust KD, Morales P, Pierson CR, Rinaldi F, Perry S, Bolon B, Wein N, Cobb S, Kaspar BK, Meyer KC. Novel MECP2 gene therapy is effective in a multicenter study using two mouse models of Rett syndrome and is safe in non-human primates. Mol Ther 2023; 31:2767-2782. [PMID: 37481701 PMCID: PMC10492029 DOI: 10.1016/j.ymthe.2023.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 04/10/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023] Open
Abstract
The AAV9 gene therapy vector presented in this study is safe in mice and non-human primates and highly efficacious without causing overexpression toxicity, a major challenge for clinical translation of Rett syndrome gene therapy vectors to date. Our team designed a new truncated methyl-CpG-binding protein 2 (MECP2) promoter allowing widespread expression of MECP2 in mice and non-human primates after a single injection into the cerebrospinal fluid without causing overexpression symptoms up to 18 months after injection. Additionally, this new vector is highly efficacious at lower doses compared with previous constructs as demonstrated in extensive efficacy studies performed by two independent laboratories in two different Rett syndrome mouse models carrying either a knockout or one of the most frequent human mutations of Mecp2. Overall, data from this multicenter study highlight the efficacy and safety of this gene therapy construct, making it a promising candidate for first-in-human studies to treat Rett syndrome.
Collapse
Affiliation(s)
- Samantha Powers
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| | - Shibi Likhite
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Kamal K Gadalla
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Carlos J Miranda
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Amy J Huffenberger
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Cassandra Dennys
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Kevin D Foust
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Pablo Morales
- The Mannheimer Foundation Inc, Homestead, FL 33034, USA
| | - Christopher R Pierson
- The Department of Pathology & Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH 43205, USA; Department of Pathology and the Department of Biomedical Education & Anatomy, The Ohio State University, Columbus, OH 43210, USA
| | - Federica Rinaldi
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Stephanie Perry
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | | - Nicolas Wein
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA; Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Stuart Cobb
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Brian K Kaspar
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA; Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Kathrin C Meyer
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
14
|
Totten DJ, Booth KTA, Mosier KM, Cumpston EC, Whitted C, Okechuku V, Koontz NA, Nelson RF. Human cochlear diffusion from the cerebrospinal fluid space with gadolinium contrast. Mol Ther 2023; 31:2566-2569. [PMID: 37582360 PMCID: PMC10492018 DOI: 10.1016/j.ymthe.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/01/2023] [Accepted: 08/01/2023] [Indexed: 08/17/2023] Open
Affiliation(s)
- Douglas J Totten
- Department of Otolaryngology, Head & Neck Surgery, Indiana University, Indianapolis. IN 46202, USA
| | - Kevin T A Booth
- Department of Otolaryngology, Head & Neck Surgery, Indiana University, Indianapolis. IN 46202, USA; Department of Medical & Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
| | - Kristine M Mosier
- Department of Otolaryngology, Head & Neck Surgery, Indiana University, Indianapolis. IN 46202, USA; Department of Radiology & Imaging Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Evan C Cumpston
- Department of Otolaryngology, Head & Neck Surgery, Indiana University, Indianapolis. IN 46202, USA
| | - Cody Whitted
- School of Medicine, Indiana University, IN 46202, USA
| | - Vanessa Okechuku
- School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Nicholas A Koontz
- Department of Otolaryngology, Head & Neck Surgery, Indiana University, Indianapolis. IN 46202, USA; Department of Radiology & Imaging Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Rick F Nelson
- Department of Otolaryngology, Head & Neck Surgery, Indiana University, Indianapolis. IN 46202, USA; Department of Neurological Surgery, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
15
|
De BP, Rosenberg JB, Selvan N, Wilson I, Yusufzai N, Greco A, Kaminsky SM, Heier LA, Ricart Arbona RJ, Miranda IC, Monette S, Nair A, Khanna R, Crystal RG, Sondhi D. Assessment of Safety and Biodistribution of AAVrh.10hCLN2 Following Intracisternal Administration in Nonhuman Primates for the Treatment of CLN2 Batten Disease. Hum Gene Ther 2023; 34:905-916. [PMID: 37624739 PMCID: PMC10517331 DOI: 10.1089/hum.2023.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/10/2023] [Indexed: 08/27/2023] Open
Abstract
CLN2 disease is a fatal, childhood autosomal recessive disorder caused by mutations in ceroid lipofuscinosis type 2 (CLN2) gene, encoding tripeptidyl peptidase 1 (TPP-1). Loss of TPP-1 activity leads to accumulation of storage material in lysosomes and resultant neuronal cell death with neurodegeneration. Genotype/phenotype comparisons suggest that the phenotype should be ameliorated with increase of TPP-1 levels to 5-10% of normal with wide central nervous system (CNS) distribution. Our previous clinical study showed that intraparenchymal (IPC) administration of AAVrh.10hCLN2, an adeno-associated vector serotype rh.10 encoding human CLN2, slowed, but did not stop disease progression, suggesting that this may be insufficient to distribute the therapy throughout the CNS (Sondhi 2020). In this study, we assessed whether the less invasive intracisternal delivery route would be safe and provide a wider distribution of TPP-1. A study was conducted in nonhuman primates (NHPs) with intracisternal delivery to cerebrospinal fluid (CSF) of AAVrh.10hCLN2 (5 × 1013 genome copies) or phosphate buffered saline (PBS). No abnormal behavior was noted. CNS magnetic resonance imaging and clinical chemistry data were all unremarkable. Histopathology of major organs had no abnormal finding attributable to the intervention or the vector, except that in one out of two animals treated with AAVrh.10hCLN2, dorsal root ganglia showed mild-to-moderate mononuclear cell infiltrates and neuronal degeneration. In contrast to our previous NHP study (Sondhi 2012) with IPC administration where TPP-1 activity was >2 × above controls in 30% of treated brains, in the two intracisternal treated NHPs, the TPP-1 activity was >2 × above controls in 50% and 41% of treated brains, and 52% and 84% of brain had >1,000 vector genomes/μg DNA, compared to 0% in the two PBS NHP. CSF TPP1 levels in treated animals were 43-62% of normal human levels. Collectively, these data indicate that AAVrh.10hCLN2 delivered by intracisternal route is safe and widely distributes TPP-1 in brain and CSF at levels that are potentially therapeutic. Clinical Trial Registration: NCT02893826, NCT04669535, NCT04273269, NCT03580083, NCT04408625, NCT04127578, and NCT04792944.
Collapse
Affiliation(s)
- Bishnu P. De
- Department of Genetic Medicine, New York, New York, USA
| | | | | | | | | | | | | | - Linda A. Heier
- Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - Rodolfo J. Ricart Arbona
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Ileana C. Miranda
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, New York, USA
| | - Sebastien Monette
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, New York, USA
| | - Anju Nair
- LEXEO Therapeutics, New York, New York, USA
| | | | | | - Dolan Sondhi
- Department of Genetic Medicine, New York, New York, USA
| |
Collapse
|
16
|
Salegio EA, Hancock K, Korszen S. Pre-clinical delivery of gene therapy products to the cerebrospinal fluid: challenges and considerations for clinical translation. Front Mol Neurosci 2023; 16:1248271. [PMID: 37664241 PMCID: PMC10469667 DOI: 10.3389/fnmol.2023.1248271] [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: 06/26/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
While the majority of gene therapy studies in neurological indications have focused on direct gene transfer to the central nervous system (CNS), there is growing interest in the delivery of therapeutics using the cerebrospinal fluid (CSF) as a conduit. Historically, direct CNS routes-of-administration (RoAs) have relied on tissue dynamics, displacement of interstitial fluid, and regional specificity to achieve focal delivery into regions of interest, such as the brain. While intraparenchymal delivery minimizes peripheral organ exposure, one perceived drawback is the relative invasiveness of this approach to drug delivery. In this mini review, we examine the CSF as an alternative RoA to target CNS tissue and discuss considerations associated with the safety of performing such procedures, biodistribution of therapeutics following single administration, and translation of findings given differences between small and large animals. These factors will help delineate key considerations for translating data obtained from animal studies into clinical settings that may be useful in the treatment of neurological conditions.
Collapse
|
17
|
O'Leary C, Forte G, Mitchell NL, Youshani AS, Dyer A, Wellby MP, Russell KN, Murray SJ, Jolinon N, Jones SA, Stacey K, Davis DM, Henckaerts E, Palmer DN, Kamaly-Asl I, Bigger BW. Intraparenchymal convection enhanced delivery of AAV in sheep to treat Mucopolysaccharidosis IIIC. J Transl Med 2023; 21:437. [PMID: 37407981 PMCID: PMC10320977 DOI: 10.1186/s12967-023-04208-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Mucopolysaccharidosis IIIC (MPSIIIC) is one of four Sanfilippo diseases sharing clinical symptoms of severe cognitive decline and shortened lifespan. The missing enzyme, heparan sulfate acetyl-CoA: α-glucosaminide-N-acetyltransferase (HGSNAT), is bound to the lysosomal membrane, therefore cannot cross the blood-brain barrier or diffuse between cells. We previously demonstrated disease correction in MPSIIIC mice using an Adeno-Associated Vector (AAV) delivering HGSNAT via intraparenchymal brain injections using an AAV2 derived AAV-truetype (AAV-TT) serotype with improved distribution over AAV9. METHODS Here, intraparenchymal AAV was delivered in sheep using catheters or Hamilton syringes, placed using Brainlab cranial navigation for convection enhanced delivery, to reduce proximal vector expression and improve spread. RESULTS Hamilton syringes gave improved AAV-GFP distribution, despite lower vector doses and titres. AAV-TT-GFP displayed moderately better transduction compared to AAV9-GFP but both serotypes almost exclusively transduced neurons. Functional HGSNAT enzyme was detected in 24-37% of a 140g gyrencephalic sheep brain using AAV9-HGSNAT with three injections in one hemisphere. CONCLUSIONS Despite variabilities in volume and titre, catheter design may be critical for efficient brain delivery. These data help inform a clinical trial for MPSIIIC.
Collapse
Affiliation(s)
- Claire O'Leary
- Stem Cell & Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
- The Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester Academic Health Science Centre, Northern Care Alliance, Manchester, UK
| | - Gabriella Forte
- Stem Cell & Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Nadia L Mitchell
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand
- Department of Radiology, University of Otago, Christchurch, 8140, New Zealand
| | - Amir Saam Youshani
- Stem Cell & Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
- The Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester Academic Health Science Centre, Northern Care Alliance, Manchester, UK
| | - Adam Dyer
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Martin P Wellby
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand
| | - Katharina N Russell
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand
| | - Samantha J Murray
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand
| | - Nelly Jolinon
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Simon A Jones
- Manchester Centre for Genomic Medicine, Willink Unit, Manchester University NHS Foundation Trust, Manchester, UK
| | - Kevin Stacey
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Daniel M Davis
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington, London, UK
| | - Els Henckaerts
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
- Laboratory of Viral Cell Biology & Therapeutics, Department of Cellular and Molecular Medicine and Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - David N Palmer
- Department of Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand
- Department of Radiology, University of Otago, Christchurch, 8140, New Zealand
| | - Ian Kamaly-Asl
- The Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester Academic Health Science Centre, Northern Care Alliance, Manchester, UK
- Department of Paediatric Neurosurgery, Royal Manchester Children's Hospital, Manchester, UK
| | - Brian W Bigger
- Stem Cell & Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
18
|
Sigurdsson B, Hauglund NL, Lilius TO, Mogensen FLH, Mortensen KN, Beschorner N, Klinger L, Bærentzen SL, Rosenholm MP, Shalgunov V, Herth M, Mori Y, Nedergaard M. A SPECT-based method for dynamic imaging of the glymphatic system in rats. J Cereb Blood Flow Metab 2023; 43:1153-1165. [PMID: 36809165 PMCID: PMC10291457 DOI: 10.1177/0271678x231156982] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/17/2022] [Accepted: 12/07/2022] [Indexed: 02/23/2023]
Abstract
The glymphatic system is a brain-wide waste drainage system that promotes cerebrospinal fluid circulation through the brain to remove waste metabolites. Currently, the most common methods for assessing glymphatic function are ex vivo fluorescence microscopy of brain slices, macroscopic cortical imaging, and MRI. While all these methods have been crucial for expanding our understanding of the glymphatic system, new techniques are required to overcome their specific drawbacks. Here, we evaluate SPECT/CT imaging as a tool to assess glymphatic function in different anesthesia-induced brain states using two radiolabeled tracers, [111In]-DTPA and [99mTc]-NanoScan. Using SPECT, we confirmed the existence of brain state-dependent differences in glymphatic flow and we show brain state-dependent differences of CSF flow kinetics and CSF egress to the lymph nodes. We compare SPECT and MRI for imaging glymphatic flow and find that the two imaging modalities show the same overall pattern of CSF flow, but that SPECT was specific across a greater range of tracer concentrations than MRI. Overall, we find that SPECT imaging is a promising tool for imaging the glymphatic system, and that qualities such as high sensitivity and the variety of available tracers make SPECT imaging a good alternative for glymphatic research.
Collapse
Affiliation(s)
- Björn Sigurdsson
- Center for Translational Neuromedicine, University of Copenhagen, Denmark
| | - Natalie L Hauglund
- Center for Translational Neuromedicine, University of Copenhagen, Denmark
| | - Tuomas O Lilius
- Center for Translational Neuromedicine, University of Copenhagen, Denmark
- INDIVIDRUG Research Program, University of Helsinki, Finland
- Department of Pharmacology, University of Helsinki, Finland
- Department of Emergency Medicine and Services, Helsinki University Hospital and University of Helsinki, Finland
| | - Frida L-H Mogensen
- Center for Translational Neuromedicine, University of Copenhagen, Denmark
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Doctoral School of Science and Technology, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | | - Natalie Beschorner
- Center for Translational Neuromedicine, University of Copenhagen, Denmark
| | - Laura Klinger
- Center for Translational Neuromedicine, University of Copenhagen, Denmark
| | - Simone L Bærentzen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Marko P Rosenholm
- Center for Translational Neuromedicine, University of Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Matthias Herth
- Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
- Department of Clinical Physiology, Copenhagen University Hospital, Denmark
| | - Yuki Mori
- Center for Translational Neuromedicine, University of Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, USA
| |
Collapse
|
19
|
Tian G, Cao C, Li S, Wang W, Zhang Y, Lv Y. rAAV2-Mediated Restoration of GALC in Neural Stem Cells from Krabbe Patient-Derived iPSCs. Pharmaceuticals (Basel) 2023; 16:ph16040624. [PMID: 37111381 PMCID: PMC10143348 DOI: 10.3390/ph16040624] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Krabbe disease is a rare neurodegenerative fatal disease. It is caused by deficiency of the lysosomal enzyme galactocerebrosidase (GALC), which results in progressive accumulation of galactolipid substrates in myelin-forming cells. However, there is still a lack of appropriate neural models and effective approaches for Krabbe disease. We generated induced pluripotent stem cells (iPSCs) from a Krabbe patient previously. Here, Krabbe patient-derived neural stem cells (K-NSCs) were induced from these iPSCs. By using nine kinds of recombinant adeno-associated virus (rAAV) vectors to infect K-NSCs, we found that the rAAV2 vector has high transduction efficiency for K-NSCs. Most importantly, rAAV2-GALC rescued GALC enzymatic activity in K-NSCs. Our findings not only establish a novel patient NSC model for Krabbe disease, but also firstly indicate the potential of rAAV2-mediated gene therapy for this devastating disease.
Collapse
Affiliation(s)
- Guoshuai Tian
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Chunyu Cao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443000, China
| | - Shuyue Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443000, China
| | - Wei Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Ye Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yafeng Lv
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443000, China
| |
Collapse
|
20
|
Simon MJ, Logan T, DeVos SL, Di Paolo G. Lysosomal functions of progranulin and implications for treatment of frontotemporal dementia. Trends Cell Biol 2023; 33:324-339. [PMID: 36244875 DOI: 10.1016/j.tcb.2022.09.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 12/12/2022]
Abstract
Loss-of-function heterozygous mutations in GRN, the gene encoding progranulin (PGRN), were identified in patients with frontotemporal lobar degeneration (FTLD) almost two decades ago and are generally linked to reduced PGRN protein expression levels. Although initial characterization of PGRN function primarily focused on its role in extracellular signaling as a secreted protein, more recent studies revealed critical roles of PGRN in regulating lysosome function, including proteolysis and lipid degradation, consistent with its lysosomal localization. Emerging from these studies is the notion that PGRN regulates glucocerebrosidase activity via direct chaperone activities and via interaction with prosaposin (i.e., a key regulator of lysosomal sphingolipid-metabolizing enzymes), as well as with the anionic phospholipid bis(monoacylglycero)phosphate. This emerging lysosomal biology of PGRN identified novel and promising opportunities in therapeutic discovery as well as biomarker development.
Collapse
Affiliation(s)
| | - Todd Logan
- Denali Therapeutics, South San Francisco, CA, USA
| | | | | |
Collapse
|
21
|
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
|
22
|
AAV vectors applied to the treatment of CNS disorders: Clinical status and challenges. J Control Release 2023; 355:458-473. [PMID: 36736907 DOI: 10.1016/j.jconrel.2023.01.067] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
In recent years, adeno-associated virus (AAV) has become the most important vector for central nervous system (CNS) gene therapy. AAV has already shown promising results in the clinic, for several CNS diseases that cannot be treated with drugs, including neurodegenerative diseases, neuromuscular diseases, and lysosomal storage disorders. Currently, three of the four commercially available AAV-based drugs focus on neurological disorders, including Upstaza for aromatic l-amino acid decarboxylase deficiency, Luxturna for hereditary retinal dystrophy, and Zolgensma for spinal muscular atrophy. All these studies have provided paradigms for AAV-based therapeutic intervention platforms. AAV gene therapy, with its dual promise of targeting disease etiology and enabling 'long-term correction' of disease processes, has the advantages of immune privilege, high delivery efficiency, tissue specificity, and cell tropism in the CNS. Although AAV-based gene therapy has been shown to be effective in most CNS clinical trials, limitations have been observed in its clinical applications, which are often associated with side effects. In this review, we summarized the therapeutic progress, challenges, limitations, and solutions for AAV-based gene therapy in 14 types of CNS diseases. We focused on viral vector technologies, delivery routes, immunosuppression, and other relevant clinical factors. We also attempted to integrate several hurdles faced in clinical and preclinical studies with their solutions, to seek the best path forward for the application of AAV-based gene therapy in the context of CNS diseases. We hope that these thoughtful recommendations will contribute to the efficient translation of preclinical studies and wide application of clinical trials.
Collapse
|
23
|
Horiuchi M, Hinderer CJ, Greig JA, Dyer C, Buza EL, Bell P, Chichester JA, Hayashi PM, Yan H, Goode T, Wilson JM. Intravenous immunoglobulin prevents peripheral liver transduction of intrathecally delivered AAV vectors. Mol Ther Methods Clin Dev 2022; 27:272-280. [PMID: 36320416 PMCID: PMC9593247 DOI: 10.1016/j.omtm.2022.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Gene therapy using neurotropic adeno-associated virus vectors represents an emerging solution for genetic disorders affecting the central nervous system. The first approved central nervous system-targeting adeno-associated virus gene therapy, Zolgensma®, for treating spinal muscular atrophy is administered intravenously at high doses that cause liver-associated adverse events in 20%–30% of patients. Intrathecal routes of vector administration, such as the intra-cisterna magna route, provide efficient gene transduction to central nervous system cells while reducing off-target liver transduction. However, significant levels of liver transduction often occur upon intra-cisterna magna vector delivery in preclinical studies. Using vectors expressing monoclonal antibody transgenes, we examined whether passive transfer of adeno-associated virus-neutralizing antibodies as intravenous immunoglobulin before intrathecal adeno-associated virus delivery improved the safety of viral gene therapy targeting the central nervous system in mice and nonhuman primates. We used intracerebroventricular and intra-cisterna magna routes for vector administration to mice and nonhuman primates, respectively, and evaluated transgene expression and vector genome distribution. Our data indicate that pretreatment with intravenous immunoglobulin significantly reduced gene transduction to the liver and other peripheral organs but not to the central nervous system in both species. With further refinement, this method may improve the safety of adeno-associated virus-based, central nervous system-targeting gene therapies in clinical settings.
Collapse
Affiliation(s)
- Makoto Horiuchi
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christian J. Hinderer
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jenny A. Greig
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cecilia Dyer
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth L. Buza
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jessica A. Chichester
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter M. Hayashi
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hanying Yan
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tamara Goode
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James M. Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Corresponding author James M. Wilson, Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, 125 South 31st Street, Suite 1200, Philadelphia, PA 19104, USA.
| |
Collapse
|
24
|
Marrone L, Marchi PM, Azzouz M. Circumventing the packaging limit of AAV-mediated gene replacement therapy for neurological disorders. Expert Opin Biol Ther 2022; 22:1163-1176. [PMID: 34904932 DOI: 10.1080/14712598.2022.2012148] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Gene therapy provides the exciting opportunity of a curative single treatment for devastating diseases, eradicating the need for chronic medication. Adeno-associated viruses (AAVs) are among the most attractive vector carriers for gene replacement in vivo. Yet, despite the success of recent AAV-based clinical trials, the clinical use of these vectors has been limited. For instance, the AAV packaging capacity is restricted to ~4.7 kb, making it a substantial challenge to deliver large gene products. AREAS COVERED In this review, we explore established and emerging strategies that circumvent the packaging limit of AAVs to make them effective vehicles for gene replacement therapy of monogenic disorders, with a particular focus on diseases affecting the nervous system. We report historical references, design remarks, as well as strengths and weaknesses of these approaches. We additionally discuss examples of neurological disorders for which such strategies have been attempted. EXPERT OPINION The field of AAV-gene therapy has experienced enormous advancements in the last decade. However, there is still ample space for improvement aimed at overcoming existing challenges that are slowing down the progressive trajectory of this field.
Collapse
Affiliation(s)
- Lara Marrone
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Paolo M Marchi
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Mimoun Azzouz
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| |
Collapse
|
25
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
26
|
Abeliovich A, Hefti F, Sevigny J. Gene Therapy for Parkinson's Disease Associated with GBA1 Mutations. JOURNAL OF PARKINSON'S DISEASE 2022; 11:S183-S188. [PMID: 34151863 PMCID: PMC8543272 DOI: 10.3233/jpd-212739] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 11/24/2022]
Abstract
Human genetic studies as well as studies in animal models indicate that lysosomal dysfunction plays a key role in the pathogenesis of Parkinson's disease. Among the lysosomal genes involved, GBA1 has the largest impact on Parkinson's disease risk. Deficiency in the GBA1 encoded enzyme glucocerebrosidase (GCase) leads to the accumulation of the GCase glycolipid substrates glucosylceramide and glucosylsphingosine and ultimately results in toxicity and inflammation and negatively affect many clinical aspects of Parkinson's disease, including disease risk, the severity of presentation, age of onset, and likelihood of progression to dementia. These findings support the view that re-establishing normal levels of GCase enzyme activity may reduce the progression of Parkinson's disease in patients carrying GBA1 mutations. Studies in mouse models indicate that PR001, a AAV9 vector-based gene therapy designed to deliver a functional GBA1 gene to the brain, suggest that this therapeutic approach may slow or stop disease progression. PR001 is currently being evaluated in clinical trials with Parkinson's disease patients carrying GBA1 mutations.
Collapse
Affiliation(s)
- Asa Abeliovich
- Prevail Therapeutics, A Wholly-Owned Subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Franz Hefti
- Prevail Therapeutics, A Wholly-Owned Subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Jeffrey Sevigny
- Prevail Therapeutics, A Wholly-Owned Subsidiary of Eli Lilly and Company, New York, NY, USA
| |
Collapse
|
27
|
Morris JA, Boshoff CH, Schor NF, Wong LM, Gao G, Davidson BL. Next-generation strategies for gene-targeted therapies of central nervous system disorders: A workshop summary. Mol Ther 2021; 29:3332-3344. [PMID: 34547463 DOI: 10.1016/j.ymthe.2021.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/30/2022] Open
Abstract
The National Institute of Neurological Disorders and Stroke (NINDS) held a workshop titled "Next generation strategies for gene-targeted therapies of central nervous system (CNS) disorders" in September 2019 in Bethesda, MD, USA. The meeting brought together a multi-disciplinary group of experts in the field of CNS-directed gene-targeted therapy delivery from academia, industry, advocacy, and the government. The group was charged with identifying the key challenges and gaps in this evolving field, as well as suggesting potential solutions. The workshop was divided into four sessions: (1) control of level and location, (2) improving delivery and distribution, (3) enhancing models and manufacturing, and (4) impacting patients. Prior to the workshop, NINDS established working groups of key opinion leaders (KOLs) for each session. In pre-meeting teleconferences, KOLs were tasked with identifying the research gaps and key obstacles that delay and/or prevent gene-targeted therapies to move into the clinic. This approach allowed for the workshop to begin with problem-solving discussions and strategy development, as the key issues had been established. The overall purpose of the workshop was to consider knowledge gaps and potential strategies to inform the community around CNS gene-targeted therapies, including but not limited to researchers and funders.
Collapse
Affiliation(s)
- Jill A Morris
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Chris H Boshoff
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nina F Schor
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ling M Wong
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guangping Gao
- Horae Gene Therapy Center, UMass Medical School, Worcester, MA 01605, USA
| | - Beverly L Davidson
- Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
28
|
Marchi PM, Marrone L, Azzouz M. Delivery of therapeutic AAV9 vectors via cisterna magna to treat neurological disorders. Trends Mol Med 2021; 28:79-80. [PMID: 34756547 DOI: 10.1016/j.molmed.2021.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 11/18/2022]
Affiliation(s)
- Paolo M Marchi
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Lara Marrone
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK.
| |
Collapse
|
29
|
Fischell JM, Fishman PS. A Multifaceted Approach to Optimizing AAV Delivery to the Brain for the Treatment of Neurodegenerative Diseases. Front Neurosci 2021; 15:747726. [PMID: 34630029 PMCID: PMC8497810 DOI: 10.3389/fnins.2021.747726] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Despite major advancements in gene therapy technologies, there are no approved gene therapies for diseases which predominantly effect the brain. Adeno-associated virus (AAV) vectors have emerged as the most effective delivery vector for gene therapy owing to their simplicity, wide spread transduction and low immunogenicity. Unfortunately, the blood-brain barrier (BBB) makes IV delivery of AAVs, to the brain highly inefficient. At IV doses capable of widespread expression in the brain, there is a significant risk of severe immune-mediated toxicity. Direct intracerebral injection of vectors is being attempted. However, this method is invasive, and only provides localized delivery for diseases known to afflict the brain globally. More advanced methods for AAV delivery will likely be required for safe and effective gene therapy to the brain. Each step in AAV delivery, including delivery route, BBB transduction, cellular tropism and transgene expression provide opportunities for innovative solutions to optimize delivery efficiency. Intra-arterial delivery with mannitol, focused ultrasound, optimized AAV capsid evolution with machine learning algorithms, synthetic promotors are all examples of advanced strategies which have been developed in pre-clinical models, yet none are being investigated in clinical trials. This manuscript seeks to review these technological advancements, and others, to improve AAV delivery to the brain, and to propose novel strategies to build upon this research. Ultimately, it is hoped that the optimization of AAV delivery will allow for the human translation of many gene therapies for neurodegenerative and other neurologic diseases.
Collapse
Affiliation(s)
- Jonathan M Fischell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Paul S Fishman
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
30
|
Pflepsen KR, Peterson CD, Kitto KF, Riedl MS, McIvor RS, Wilcox GL, Vulchanova L, Fairbanks CA. Biodistribution of Adeno-Associated Virus Serotype 5 Viral Vectors Following Intrathecal Injection. Mol Pharm 2021; 18:3741-3749. [PMID: 34460254 DOI: 10.1021/acs.molpharmaceut.1c00252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The pharmacokinetic profile of AAV particles following intrathecal delivery has not yet been clearly defined. The present study evaluated the distribution profile of adeno-associated virus serotype 5 (AAV5) viral vectors following lumbar intrathecal injection in mice. After a single bolus intrathecal injection, viral DNA concentrations in mouse whole blood, spinal cord, and peripheral tissues were determined using quantitative polymerase chain reaction (qPCR). The kinetics of AAV5 vector in whole blood and the concentration over time in spinal and peripheral tissues were analyzed. Distribution of the AAV5 vector to all levels of the spinal cord, dorsal root ganglia, and into systemic circulation occurred rapidly within 30 min following injection. Vector concentration in whole blood reached a maximum 6 h postinjection with a half-life of approximately 12 h. Area under the curve data revealed the highest concentration of vector distributed to dorsal root ganglia tissue. Immunohistochemical analysis revealed AAV5 particle colocalization with the pia mater at the spinal cord and macrophages in the dorsal root ganglia (DRG) 30 min after injection. These results demonstrate the widespread distribution of AAV5 particles through cerebrospinal fluid and preferential targeting of DRG tissue with possible clearance mechanisms via DRG macrophages.
Collapse
Affiliation(s)
- Kelsey R Pflepsen
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Cristina D Peterson
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kelley F Kitto
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Maureen S Riedl
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - R Scott McIvor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - George L Wilcox
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States.,Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, United States.,Department of Dermatology, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carolyn A Fairbanks
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota 55455, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States.,Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
31
|
Adeno-Associated Viral Vectors as Versatile Tools for Parkinson's Research, Both for Disease Modeling Purposes and for Therapeutic Uses. Int J Mol Sci 2021; 22:ijms22126389. [PMID: 34203739 PMCID: PMC8232322 DOI: 10.3390/ijms22126389] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/17/2022] Open
Abstract
It is without any doubt that precision medicine therapeutic strategies targeting neurodegenerative disorders are currently witnessing the spectacular rise of newly designed approaches based on the use of viral vectors as Trojan horses for the controlled release of a given genetic payload. Among the different types of viral vectors, adeno-associated viruses (AAVs) rank as the ones most commonly used for the purposes of either disease modeling or for therapeutic strategies. Here, we reviewed the current literature dealing with the use of AAVs within the field of Parkinson’s disease with the aim to provide neuroscientists with the advice and background required when facing a choice on which AAV might be best suited for addressing a given experimental challenge. Accordingly, here we will be summarizing some insights on different AAV serotypes, and which would be the most appropriate AAV delivery route. Next, the use of AAVs for modeling synucleinopathies is highlighted, providing potential readers with a landscape view of ongoing pre-clinical and clinical initiatives pushing forward AAV-based therapeutic approaches for Parkinson’s disease and related synucleinopathies.
Collapse
|
32
|
Galvan A, Petkau TL, Hill AM, Korecki AJ, Lu G, Choi D, Rahman K, Simpson EM, Leavitt BR, Smith Y. Intracerebroventricular Administration of AAV9-PHP.B SYN1-EmGFP Induces Widespread Transgene Expression in the Mouse and Monkey Central Nervous System. Hum Gene Ther 2021; 32:599-615. [PMID: 33860682 PMCID: PMC8236560 DOI: 10.1089/hum.2020.301] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Viral vectors made from adeno-associated virus (AAV) have emerged as preferred tools in basic and translational neuroscience research to introduce or modify genetic material in cells of interest. The use of viral vectors is particularly attractive in nontransgenic species, such as nonhuman primates. Injection of AAV solutions into the cerebrospinal fluid is an effective method to achieve a broad distribution of a transgene in the central nervous system. In this study, we conducted injections of AAV9-PHP.B, a recently described AAV capsid mutant, in the lateral ventricle of mice and rhesus macaques. To enhance the expression of the transgene (the tag protein emerald green fluorescent protein [EmGFP]), we used a gene promoter that confers high neuron-specific expression of the transgene, the human synapsin 1 (SYN1) promoter. The efficacy of the viral vector was first tested in mice. Our results show that intracerebroventricular injections of AAV9-PHP.B SYN1-EmGFP-woodchuck hepatitis virus posttranscriptional regulatory element resulted in neuronal EmGFP expression throughout the mice and monkey brains. We have provided a thorough characterization of the brain regions expressing EmGFP in both species. EmGFP was observed in neuronal cell bodies over the whole cerebral cortex and in the cerebellum, as well as in some subcortical regions, including the striatum and hippocampus. We also observed densely labeled neuropil in areas known to receive projections from these regions. Double fluorescence studies demonstrated that EmGFP was expressed by several types of neurons throughout the mouse and monkey brain. Our results demonstrate that a single injection in the lateral ventricle is an efficient method to obtain transgene expression in many cortical and subcortical regions, obviating the need of multiple intraparenchymal injections to cover large brain areas. The use of intraventricular injections of AAV9-PHP.B SYN1-EmGFP could provide a powerful approach to transduce widespread areas of the brain and may contribute to further development of methods to genetically target-specific populations of neurons.
Collapse
Affiliation(s)
- Adriana Galvan
- Department of Neurology, Yerkes National Primate Research Center, Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia, USA
| | - Terri L. Petkau
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Austin M. Hill
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrea J. Korecki
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ge Lu
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Diane Choi
- Department of Neurology, Yerkes National Primate Research Center, Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia, USA
- Molecular Systems and Pharmacology Graduate Program, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Kazi Rahman
- Department of Neurology, Yerkes National Primate Research Center, Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia, USA
| | - Elizabeth M. Simpson
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Blair R. Leavitt
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, Vancouver, British Columbia, Canada
- Center for Brain Health, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Yoland Smith
- Department of Neurology, Yerkes National Primate Research Center, Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
33
|
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
|
34
|
Wang J, Zhang L. Retrograde Axonal Transport Property of Adeno-Associated Virus and Its Possible Application in Future. Microbes Infect 2021; 23:104829. [PMID: 33878458 DOI: 10.1016/j.micinf.2021.104829] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 12/19/2022]
Abstract
Gene therapy has become a treatment method for many diseases. Adeno-associated virus (AAV) is one of the most common virus vectors, is also widely used in the gene therapy field. During the past 2 decades, the retrograde axonal transportability of AAV has been discovered and utilized. Many studies have worked on the retrograde axonal transportability of AAV, and more and more people are interested in this field. This review described the current application, influence factors, and mechanism of retrograde axonal transportability of AAV and predicted its potential use in disease treatment in near future.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Gastroenterology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
| | - Liqin Zhang
- Department of Otolaryngology, Peking Union Medical College Hospital, Dongcheng Qu, Beijing, 100730, China.
| |
Collapse
|
35
|
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
|
36
|
Bradbury AM, Bagel JH, Nguyen D, Lykken EA, Pesayco Salvador J, Jiang X, Swain GP, Assenmacher CA, Hendricks IJ, Miyadera K, Hess RS, Ostrager A, ODonnell P, Sands MS, Ory DS, Shelton GD, Bongarzone ER, Gray SJ, Vite CH. Krabbe disease successfully treated via monotherapy of intrathecal gene therapy. J Clin Invest 2021; 130:4906-4920. [PMID: 32773406 DOI: 10.1172/jci133953] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Globoid cell leukodystrophy (GLD; Krabbe disease) is a progressive, incurable neurodegenerative disease caused by deficient activity of the hydrolytic enzyme galactosylceramidase (GALC). The ensuing cytotoxic accumulation of psychosine results in diffuse central and peripheral nervous system (CNS, PNS) demyelination. Presymptomatic hematopoietic stem cell transplantation (HSCT) is the only treatment for infantile-onset GLD; however, clinical outcomes of HSCT recipients often remain poor, and procedure-related morbidity is high. There are no effective therapies for symptomatic patients. Herein, we demonstrate in the naturally occurring canine model of GLD that presymptomatic monotherapy with intrathecal AAV9 encoding canine GALC administered into the cisterna magna increased GALC enzyme activity, normalized psychosine concentration, improved myelination, and attenuated inflammation in both the CNS and PNS. Moreover, AAV-mediated therapy successfully prevented clinical neurological dysfunction, allowing treated dogs to live beyond 2.5 years of age, more than 7 times longer than untreated dogs. Furthermore, we found that a 5-fold lower dose resulted in an attenuated form of disease, indicating that sufficient dosing is critical. Finally, postsymptomatic therapy with high-dose AAV9 also significantly extended lifespan, signifying a treatment option for patients for whom HSCT is not applicable. If translatable to patients, these findings would improve the outcomes of patients treated either pre- or postsymptomatically.
Collapse
Affiliation(s)
- Allison M Bradbury
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica H Bagel
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Duc Nguyen
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Erik A Lykken
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jill Pesayco Salvador
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Xuntian Jiang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gary P Swain
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Charles A Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian J Hendricks
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Keiko Miyadera
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rebecka S Hess
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Arielle Ostrager
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patricia ODonnell
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark S Sands
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel S Ory
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Steven J Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Charles H Vite
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
37
|
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
|
38
|
Ballon DJ, Rosenberg JB, Fung EK, Nikolopoulou A, Kothari P, De BP, He B, Chen A, Heier LA, Sondhi D, Kaminsky SM, Mozley PD, Babich JW, Crystal RG. Quantitative Whole-Body Imaging of I-124-Labeled Adeno-Associated Viral Vector Biodistribution in Nonhuman Primates. Hum Gene Ther 2020; 31:1237-1259. [PMID: 33233962 PMCID: PMC7769048 DOI: 10.1089/hum.2020.116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/03/2020] [Indexed: 12/19/2022] Open
Abstract
A method is presented for quantitative analysis of the biodistribution of adeno-associated virus (AAV) gene transfer vectors following in vivo administration. We used iodine-124 (I-124) radiolabeling of the AAV capsid and positron emission tomography combined with compartmental modeling to quantify whole-body and organ-specific biodistribution of AAV capsids from 1 to 72 h following administration. Using intravenous (IV) and intracisternal (IC) routes of administration of AAVrh.10 and AAV9 vectors to nonhuman primates in the absence or presence of anticapsid immunity, we have identified novel insights into initial capsid biodistribution and organ-specific capsid half-life. Neither I-124-labeled AAVrh.10 nor AAV9 administered intravenously was detected at significant levels in the brain relative to the administered vector dose. Approximately 50% of the intravenously administered labeled capsids were dispersed throughout the body, independent of the liver, heart, and spleen. When administered by the IC route, the labeled capsid had a half-life of ∼10 h in the cerebral spinal fluid (CSF), suggesting that by this route, the CSF serves as a source with slow diffusion into the brain. For both IV and IC administration, there was significant influence of pre-existing anticapsid immunity on I-124-capsid biodistribution. The methodology facilitates quantitative in vivo viral vector dosimetry, which can serve as a technique for evaluation of both on- and off-target organ biodistribution, and potentially accelerate gene therapy development through rapid prototyping of novel vector designs.
Collapse
Affiliation(s)
- Douglas J. Ballon
- Department of Radiology, Citigroup Biomedical Imaging Center
- Department of Genetic Medicine
| | | | - Edward K. Fung
- Department of Radiology, Citigroup Biomedical Imaging Center
| | | | - Paresh Kothari
- Department of Radiology, Citigroup Biomedical Imaging Center
| | | | - Bin He
- Department of Radiology, Citigroup Biomedical Imaging Center
| | | | - Linda A. Heier
- Department of Radiology; Weill Cornell Medical College, New York, New York, USA
| | | | | | | | - John W. Babich
- Department of Radiology, Citigroup Biomedical Imaging Center
| | | |
Collapse
|
39
|
Benveniste H, Lee H, Ozturk B, Chen X, Koundal S, Vaska P, Tannenbaum A, Volkow ND. Glymphatic Cerebrospinal Fluid and Solute Transport Quantified by MRI and PET Imaging. Neuroscience 2020; 474:63-79. [PMID: 33248153 DOI: 10.1016/j.neuroscience.2020.11.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/04/2020] [Accepted: 11/07/2020] [Indexed: 12/13/2022]
Abstract
Over the past decade there has been an enormous progress in our understanding of fluid and solute transport in the central nervous system (CNS). This is due to a number of factors, including important developments in whole brain imaging technology and computational fluid dynamics analysis employed for the elucidation of glymphatic transport function in the live animal and human brain. In this paper, we review the technical aspects of dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) in combination with administration of Gd-based tracers into the cerebrospinal fluid (CSF) for tracking glymphatic solute and fluid transport in the CNS as well as lymphatic drainage. Used in conjunction with advanced computational processing methods including optimal mass transport analysis, one gains new insights into the biophysical forces governing solute transport in the CNS which leads to intriguing new research directions. Considering drainage pathways, we review the novel T1 mapping technique for quantifying glymphatic transport and cervical lymph node drainage concurrently in the same subject. We provide an overview of knowledge gleaned from DCE-MRI studies of glymphatic transport and meningeal lymphatic drainage. Finally, we introduce positron emission tomography (PET) and CSF administration of radiotracers as an alternative method to explore other pharmacokinetic aspects of CSF transport into brain parenchyma as well as efflux pathways.
Collapse
Affiliation(s)
- Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States; Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, United States.
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States
| | - Burhan Ozturk
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States
| | - Xinan Chen
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, Stony Brook, NY, United States
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States
| | - Paul Vaska
- Department of Radiology and Biomedical Engineering, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Allen Tannenbaum
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, Stony Brook, NY, United States
| | - Nora D Volkow
- Laboratory for Neuroimaging, NIAAA, Bethesda, MD, United States
| |
Collapse
|
40
|
Pardridge WM. Treatment of Alzheimer's Disease and Blood-Brain Barrier Drug Delivery. Pharmaceuticals (Basel) 2020; 13:E394. [PMID: 33207605 PMCID: PMC7697739 DOI: 10.3390/ph13110394] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the enormity of the societal and health burdens caused by Alzheimer's disease (AD), there have been no FDA approvals for new therapeutics for AD since 2003. This profound lack of progress in treatment of AD is due to dual problems, both related to the blood-brain barrier (BBB). First, 98% of small molecule drugs do not cross the BBB, and ~100% of biologic drugs do not cross the BBB, so BBB drug delivery technology is needed in AD drug development. Second, the pharmaceutical industry has not developed BBB drug delivery technology, which would enable industry to invent new therapeutics for AD that actually penetrate into brain parenchyma from blood. In 2020, less than 1% of all AD drug development projects use a BBB drug delivery technology. The pathogenesis of AD involves chronic neuro-inflammation, the progressive deposition of insoluble amyloid-beta or tau aggregates, and neural degeneration. New drugs that both attack these multiple sites in AD, and that have been coupled with BBB drug delivery technology, can lead to new and effective treatments of this serious disorder.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, CA 90024, USA
| |
Collapse
|
41
|
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
|
42
|
Hinderer C, Katz N, Dyer C, Goode T, Johansson J, Bell P, Richman L, Buza E, Wilson JM. Translational Feasibility of Lumbar Puncture for Intrathecal AAV Administration. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:969-974. [PMID: 32420410 PMCID: PMC7218226 DOI: 10.1016/j.omtm.2020.04.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/13/2020] [Indexed: 12/30/2022]
Abstract
Preclinical studies have demonstrated that a single injection of an adeno-associated virus (AAV) vector into the cerebrospinal fluid (CSF) can achieve widespread gene transfer throughout the central nervous system. Successfully translating this approach to humans requires identifying factors that influence AAV distribution in the CSF so that optimal parameters can be replicated in the clinic. In the context of developing a motor neuron-targeted gene therapy for spinal muscular atrophy, we conducted studies in nonhuman primates to evaluate the impact of injection volume on spinal cord transduction after AAV delivery via lumbar puncture. Lumbar injection of an AAVhu68 vector targeted motor neurons throughout the spinal cord, but only in juvenile nonhuman primates administered large injection volumes, equivalent to about half of the total CSF volume. Upon repeating this study with clinically relevant injection volumes and larger animals, we found that lumbar puncture failed to achieve significant transduction of the spinal cord. In contrast, vector administered into the cisterna magna distributed reproducibly throughout the spinal cord in both juvenile and adult animals. These findings highlight the challenges of translating AAV delivery via lumbar puncture to humans and suggest that delivery into the cisterna magna may represent a more feasible alternative.
Collapse
Affiliation(s)
- Christian Hinderer
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathan Katz
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cecilia Dyer
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tamara Goode
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julia Johansson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter Bell
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura Richman
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Buza
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Wilson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
43
|
Bailey RM, Rozenberg A, Gray SJ. Comparison of high-dose intracisterna magna and lumbar puncture intrathecal delivery of AAV9 in mice to treat neuropathies. Brain Res 2020; 1739:146832. [PMID: 32289279 DOI: 10.1016/j.brainres.2020.146832] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023]
Abstract
Gene therapy clinical trials for neurological disorders are ongoing using intrathecal injection of adeno-associated virus (AAV) vector directly into the cerebral spinal fluid. Preliminary findings from these trials and results from extensive animal studies provides compelling data supporting the safety and benefit of intrathecal delivery of AAV vectors for inherited neurological disorders. Intrathecal delivery can be achieved by a lumbar puncture (LP) or intracisterna magna (ICM) injection, although ICM is not commonly used in clinical practice due to increased procedural risk. Few studies directly compared these delivery methods and there are limited reports on transduction of the PNS. To further test the utility of ICM or LP delivery for neuropathies, we performed a head to head comparison of AAV serotype 9 (AAV9) vectors expressing GFP injected into the cisterna magna or lumbar subarachnoid space in mice. We report that an intrathecal gene delivery of AAV9 in mice leads to stable transduction of neurons and glia in the brain and spinal cord and has a widespread distribution that includes components of the PNS. Vector expression was notably higher in select brain and PNS regions following ICM injection, while higher amounts of vector was found in the lower spinal cord and peripheral organs following LP injection. These findings support that intrathecal AAV9 delivery is a translationally relevant delivery method for inherited neuropathies.
Collapse
Affiliation(s)
- Rachel M Bailey
- Gene Therapy Center, University of North Carolina at Chapel Hill, United States; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States; Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Alejandra Rozenberg
- Gene Therapy Center, University of North Carolina at Chapel Hill, United States
| | - Steven J Gray
- Gene Therapy Center, University of North Carolina at Chapel Hill, United States; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, NC, United States; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
44
|
Segal DJ. Grand Challenges in Gene and Epigenetic Editing for Neurologic Disease. Front Genome Ed 2020; 1:1. [PMID: 34713209 PMCID: PMC8525068 DOI: 10.3389/fgeed.2019.00001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/23/2019] [Indexed: 12/26/2022] Open
Affiliation(s)
- David J. Segal
- Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States
| |
Collapse
|
45
|
Pardridge WM. Blood-Brain Barrier and Delivery of Protein and Gene Therapeutics to Brain. Front Aging Neurosci 2020; 11:373. [PMID: 31998120 PMCID: PMC6966240 DOI: 10.3389/fnagi.2019.00373] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/19/2019] [Indexed: 01/02/2023] Open
Abstract
Alzheimer’s disease (AD) and treatment of the brain in aging require the development of new biologic drugs, such as recombinant proteins or gene therapies. Biologics are large molecule therapeutics that do not cross the blood-brain barrier (BBB). BBB drug delivery is the limiting factor in the future development of new therapeutics for the brain. The delivery of recombinant protein or gene medicines to the brain is a binary process: either the brain drug developer re-engineers the biologic with BBB drug delivery technology, or goes forward with brain drug development in the absence of a BBB delivery platform. The presence of BBB delivery technology allows for engineering the therapeutic to enable entry into the brain across the BBB from blood. Brain drug development may still take place in the absence of BBB delivery technology, but with a reliance on approaches that have rarely led to FDA approval, e.g., CSF injection, stem cells, small molecules, and others. CSF injection of drug is the most widely practiced approach to brain delivery that bypasses the BBB. However, drug injection into the CSF results in limited drug penetration to the brain parenchyma, owing to the rapid export of CSF from the brain to blood. A CSF injection of a drug is equivalent to a slow intravenous (IV) infusion of the pharmaceutical. Given the profound effect the existence of the BBB has on brain drug development, future drug or gene development for the brain will be accelerated by future advances in BBB delivery technology in parallel with new drug discovery.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
46
|
Vieira S, Strymecka P, Stanaszek L, Silva-Correia J, Drela K, Fiedorowicz M, Malysz-Cymborska I, Rogujski P, Janowski M, Reis RL, Lukomska B, Walczak P, Oliveira JM. Methacrylated gellan gum and hyaluronic acid hydrogel blends for image-guided neurointerventions. J Mater Chem B 2020; 8:5928-5937. [DOI: 10.1039/d0tb00877j] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mn-Based gellan gum hydrogels for cell delivery and real-time tracking on image-guided neuro-procedures.
Collapse
Affiliation(s)
- Sílvia Vieira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| | - Paulina Strymecka
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Luiza Stanaszek
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Joana Silva-Correia
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| | - Katarzyna Drela
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Michał Fiedorowicz
- Small Animal Magnetic Resonance Imaging Laboratory
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Izabela Malysz-Cymborska
- Department of Neurology and Neurosurgery, School of Medicine
- Collegium Medicum
- University of Warmia and Mazury
- Olsztyn
- Poland
| | - Piotr Rogujski
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Miroslaw Janowski
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| | - Barbara Lukomska
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Piotr Walczak
- Department of Neurology and Neurosurgery, School of Medicine
- Collegium Medicum
- University of Warmia and Mazury
- Olsztyn
- Poland
| | - J. Miguel Oliveira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| |
Collapse
|
47
|
Taghian T, Marosfoi MG, Puri AS, Cataltepe OI, King RM, Diffie EB, Maguire AS, Martin DR, Fernau D, Batista AR, Kuchel T, Christou C, Perumal R, Chandra S, Gamlin PD, Bertrand SG, Flotte TR, McKenna-Yasek D, Tai PWL, Aronin N, Gounis MJ, Sena-Esteves M, Gray-Edwards HL. A Safe and Reliable Technique for CNS Delivery of AAV Vectors in the Cisterna Magna. Mol Ther 2019; 28:411-421. [PMID: 31813800 DOI: 10.1016/j.ymthe.2019.11.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 11/29/2022] Open
Abstract
Global gene delivery to the CNS has therapeutic importance for the treatment of neurological disorders that affect the entire CNS. Due to direct contact with the CNS, cerebrospinal fluid (CSF) is an attractive route for CNS gene delivery. A safe and effective route to achieve global gene distribution in the CNS is needed, and administration of genes through the cisterna magna (CM) via a suboccipital puncture results in broad distribution in the brain and spinal cord. However, translation of this technique to clinical practice is challenging due to the risk of serious and potentially fatal complications in patients. Herein, we report development of a gene therapy delivery method to the CM through adaptation of an intravascular microcatheter, which can be safely navigated intrathecally under fluoroscopic guidance. We examined the safety, reproducibility, and distribution/transduction of this method in sheep using a self-complementary adeno-associated virus 9 (scAAV9)-GFP vector. This technique was used to treat two Tay-Sachs disease patients (30 months old and 7 months old) with AAV gene therapy. No adverse effects were observed during infusion or post-treatment. This delivery technique is a safe and minimally invasive alternative to direct infusion into the CM, achieving broad distribution of AAV gene transfer to the CNS.
Collapse
Affiliation(s)
- Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Miklos G Marosfoi
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Ajit S Puri
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Neurological Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Oguz I Cataltepe
- Department of Neurological Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Robert M King
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Elise B Diffie
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA
| | - Anne S Maguire
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA; Department of Anatomy, Physiology and Pharmacology, Auburn University, AL 36849, USA
| | - Deborah Fernau
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ana Rita Batista
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Tim Kuchel
- South Australian Health and Medical Research Institute, Gillies Plains, SA 5086, Australia
| | - Chris Christou
- South Australian Health and Medical Research Institute, Gillies Plains, SA 5086, Australia
| | - Raj Perumal
- South Australian Health and Medical Research Institute, Gillies Plains, SA 5086, Australia
| | | | - Paul D Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stephanie G Bertrand
- Department of Environmental Population Health, Cummings Veterinary School at Tufts University, Grafton, MA 01536, USA
| | - Terence R Flotte
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Diane McKenna-Yasek
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Phillip W L Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Neil Aronin
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Matthew J Gounis
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Miguel Sena-Esteves
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|