1
|
Sartorelli J, Ng J, Rahim AA, Waddington SN, Kurian MA. Genetic therapies for movement disorders - current status. J Neurol 2025; 272:220. [PMID: 39985571 PMCID: PMC11846774 DOI: 10.1007/s00415-025-12940-5] [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: 01/02/2025] [Accepted: 01/26/2025] [Indexed: 02/24/2025]
Abstract
Movement disorders are a group of heterogeneous neurological conditions associated with alterations of tone, posture and voluntary movement. They may either occur in isolation or as part of a multisystemic condition. More recently, the advent of next generation sequencing technologies has facilitated better understanding of the underlying causative genes and molecular pathways, thereby identifying targets for genetic therapy. In this review, we summarize the advances in genetic therapy approaches for both hyperkinetic and hypokinetic movement disorders, including Parkinson's Disease, Huntington's Disease and rarer monogenic conditions of childhood onset. While there have been significant advances in the field, multiple challenges remain, related to safety, toxicity, efficacy and brain biodistribution, which will need to be addressed by the next generation of genetic therapies.
Collapse
Affiliation(s)
- J Sartorelli
- Developmental Neurosciences, Zayed Centre for Research Into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, 20 Guilford Street, London, WC1N 1DZ, UK
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - J Ng
- Genetic Therapy Accelerator Centre, UCL Queen Square Institute of Neurology, London, UK
| | - A A Rahim
- UCL School of Pharmacy, University College London, London, UK
| | - S N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, UK
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - M A Kurian
- Developmental Neurosciences, Zayed Centre for Research Into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, 20 Guilford Street, London, WC1N 1DZ, UK.
- Department of Neurology, Great Ormond Street Hospital, London, UK.
| |
Collapse
|
2
|
Elder JB, Lonser RR. Direct Convective Delivery for Nervous System Gene Therapy. Neurosurg Clin N Am 2025; 36:101-111. [PMID: 39542544 DOI: 10.1016/j.nec.2024.09.002] [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: 11/17/2024]
Abstract
Convection-enhanced delivery for central nervous system gene therapy is an emerging treatment strategy to modify the course of previously untreatable or inadequately treated neurologic conditions, including brain tumors, metabolic disorders, epilepsy, and neurodegenerative disorders. Ongoing nervous system gene therapy clinical trials highlight advantages and ongoing challenges to this therapeutic paradigm.
Collapse
Affiliation(s)
- James Bradley Elder
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Russell R Lonser
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
3
|
Kim J, Chang MY. Gene Therapy for Parkinson's Disease Using Midbrain Developmental Genes to Regulate Dopaminergic Neuronal Maintenance. Int J Mol Sci 2024; 25:12369. [PMID: 39596436 PMCID: PMC11594980 DOI: 10.3390/ijms252212369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder. It is characterized by the progressive loss of dopaminergic (DAnergic) neurons in the substantia nigra and decreased dopamine (DA) levels, which lead to both motor and non-motor symptoms. Conventional PD treatments aim to alleviate symptoms, but do not delay disease progression. PD gene therapy offers a promising approach to improving current treatments, with the potential to alleviate significant PD symptoms and cause fewer adverse effects than conventional therapies. DA replacement approaches and DA enzyme expression do not slow disease progression. However, DA replacement gene therapies, such as adeno-associated virus (AAV)-glutamic acid decarboxylase (GAD) and L-amino acid decarboxylase (AADC) gene therapies, which increase DA transmitter levels, have been demonstrated to be safe and efficient in early-phase clinical trials. Disease-modifying strategies, which aim to slow disease progression, appear to be potent. These include therapies targeting downstream pathways, neurotrophic factors, and midbrain DAnergic neuronal factors, all of which have shown potential in preclinical and clinical trials. These approaches focus on maintaining the integrity of DAnergic neurons, not just targeting the DA transmitter level itself. In particular, critical midbrain developmental and maintenance factors, such as Nurr1 and Foxa2, can interact synergistically with neighboring glia, in a paracrine mode of action, to protect DAnergic neurons against various toxic factors. Similar outcomes could be achieved by targeting both DAnergic neurons and glial cells with other candidate gene therapies, but in-depth research is needed. Neurotrophic factors, such as neurturin, the glial-cell-line-derived neurotrophic factor (GDNF), the brain-derived neurotrophic factor (BDNF), and the vascular endothelial growth factor (VEGF), are also being investigated for their potential to support DAnergic neuron survival. Additionally, gene therapies targeting key downstream pathways, such as the autophagy-lysosome pathway, mitochondrial function, and endoplasmic reticulum (ER) stress, offer promising avenues. Gene editing and delivery techniques continue to evolve, presenting new opportunities to develop effective gene therapies for PD.
Collapse
Affiliation(s)
- Jintae Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Goyang 10326, Republic of Korea;
| | - Mi-Yoon Chang
- Department of Premedicine, College of Medicine, Hanyang University, FTC12, 222 Wangsimni-ro, Seoul 04763, Republic of Korea
- Biomedical Research Institute, Hanyang University, Seoul 04763, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul 04763, Republic of Korea
| |
Collapse
|
4
|
Barker RA, Saarma M, Svendsen CN, Morgan C, Whone A, Fiandaca MS, Luz M, Bankiewicz KS, Fiske B, Isaacs L, Roach A, Phipps T, Kordower JH, Lane EL, Huttunen HJ, Sullivan A, O'Keeffe G, Yartseva V, Federoff H. Neurotrophic factors for Parkinson's disease: Current status, progress, and remaining questions. Conclusions from a 2023 workshop. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1659-1676. [PMID: 39957193 DOI: 10.1177/1877718x241301041] [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: 02/18/2025]
Abstract
In 2023, a workshop was organized by the UK charity Cure Parkinson's with The Michael J Fox Foundation for Parkinson's Research and Parkinson's UK to review the field of growth factors (GFs) for Parkinson's disease (PD). This was a follow up to a previous meeting held in 2019.1 This 2023 workshop reviewed new relevant data that has emerged in the intervening 4 years around the development of new GFs and better models for studying them including the merit of combining treatments as well as therapies that can be modulated. We also discussed new insights into GF delivery and trial design that have emerged from the analyses of completed GDNF trials, including the patient voice, as well as the recently completed CDNF trial.2 We then concluded with our recommendations on how GF studies in PD should develop going forward.
Collapse
Affiliation(s)
- Roger A Barker
- Department of Clinical Neurosciences and Cambridge Stem Cell Institute, John van Geest Centre for Brain Repair, Forvie Site, Cambridge, UK
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Catherine Morgan
- Movement Disorders Group, Bristol Brain Centre, North Bristol NHS Trust, Southmead Hospital, Southmead Road, Bristol, UK
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Alan Whone
- Movement Disorders Group, Bristol Brain Centre, North Bristol NHS Trust, Southmead Hospital, Southmead Road, Bristol, UK
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Massimo S Fiandaca
- Asklepios BioPharmaceutical, Inc. (AskBio), Research Triangle Park, NC, USA
| | - Matthias Luz
- Asklepios BioPharmaceutical, Inc. (AskBio), Research Triangle Park, NC, USA
| | - Krystof S Bankiewicz
- Asklepios BioPharmaceutical, Inc. (AskBio), Research Triangle Park, NC, USA
- The Ohio State University, College of Medicine, Pelotonia Research Center, Columbus, OH, USA
| | - Brian Fiske
- The Michael J Fox Foundation for Parkinson's Research, Grand Central Station, New York, NY, USA
| | | | | | | | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Emma L Lane
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | | | - Aideen Sullivan
- Department of Pharmacology and Therapeutics, School of Medicine, University College Cork, Cork, Ireland
| | - Gerard O'Keeffe
- Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland
| | | | - Howard Federoff
- Kenai Therapeutics, San Diego, CA, USA
- Neurology, School of Medicine, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
5
|
Xu L, Yao S, Ding YE, Xie M, Feng D, Sha P, Tan L, Bei F, Yao Y. Designing and optimizing AAV-mediated gene therapy for neurodegenerative diseases: from bench to bedside. J Transl Med 2024; 22:866. [PMID: 39334366 PMCID: PMC11429861 DOI: 10.1186/s12967-024-05661-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Recombinant adeno-associated viruses (rAAVs) have emerged as an attractive tool for gene delivery, and demonstrated tremendous promise in gene therapy and gene editing-therapeutic modalities with potential "one-and-done" treatment benefits compared to conventional drugs. Given their tropisms for the central nervous system (CNS) across various species including humans, rAAVs have been extensively investigated in both pre-clinical and clinical studies targeting neurodegenerative disease. However, major challenges remain in the application of rAAVs for CNS gene therapy, such as suboptimal vector design, low CNS transduction efficiency and specificity, and therapy-induced immunotoxicity. Therefore, continuing efforts are being made to optimize the rAAV vectors from their "core" genetic payloads to their "coat" or capsid structure. In this review, we describe current approaches for rAAV vector design tailored for transgene expression in the CNS, summarize the development of CNS-targeting AAV serotypes, and highlight recent advancements in AAV capsid engineering, aimed at generating a new generation of rAAVs with improved CNS tropism. Additionally, we discuss various administration routes for delivering rAAVs to the CNS and provide an overview of AAV-mediated gene therapies currently under investigation in clinical trials for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Liang Xu
- Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shun Yao
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Yifan Evan Ding
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mengxiao Xie
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dingqi Feng
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215123, China
| | - Pengfei Sha
- Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Lu Tan
- Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fengfeng Bei
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Yizheng Yao
- Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.
| |
Collapse
|
6
|
Wu T, Hu Y, Tang LV. Gene therapy for polygenic or complex diseases. Biomark Res 2024; 12:99. [PMID: 39232780 PMCID: PMC11375922 DOI: 10.1186/s40364-024-00618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
Gene therapy utilizes nucleic acid drugs to treat diseases, encompassing gene supplementation, gene replacement, gene silencing, and gene editing. It represents a distinct therapeutic approach from traditional medications and introduces novel strategies for genetic disorders. Over the past two decades, significant advancements have been made in the field of gene therapy, leading to the approval of various gene therapy drugs. Gene therapy was initially employed for treating genetic diseases and cancers, particularly monogenic conditions classified as orphan diseases due to their low prevalence rates; however, polygenic or complex diseases exhibit higher incidence rates within populations. Extensive research on the etiology of polygenic diseases has unveiled new therapeutic targets that offer fresh opportunities for their treatment. Building upon the progress achieved in gene therapy for monogenic diseases and cancers, extending its application to polygenic or complex diseases would enable targeting a broader range of patient populations. This review aims to discuss the strategies of gene therapy, methods of gene editing (mainly CRISPR-CAS9), and carriers utilized in gene therapy, and highlight the applications of gene therapy in polygenic or complex diseases focused on applications that have either entered clinical stages or are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Tingting Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| | - Liang V Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| |
Collapse
|
7
|
Tuominen RK, Renko JM. Biomarkers of Parkinson's disease in perspective of early diagnosis and translation of neurotrophic therapies. Basic Clin Pharmacol Toxicol 2024; 135:271-284. [PMID: 38973499 DOI: 10.1111/bcpt.14042] [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: 10/27/2023] [Revised: 04/15/2024] [Accepted: 05/28/2024] [Indexed: 07/09/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by progressive loss of dopamine neurons and aberrant deposits of alpha-synuclein (α-syn) in the brain. The symptomatic treatment is started after the onset of motor manifestations in a late stage of the disease. Preclinical studies with neurotrophic factors (NTFs) show promising results of disease-modifying neuroprotective or even neurorestorative effects. Four NTFs have entered phase I-II clinical trials with inconclusive outcomes. This is not surprising because the preclinical evidence is from acute early-stage disease models, but the clinical trials included advanced PD patients. To conclude the value of NTF therapies, clinical studies should be performed in early-stage patients with prodromal symptoms, that is, before motor manifestations. In this review, we summarize currently available diagnostic and prognostic biomarkers that could help identify at-risk patients benefiting from NTF therapies. Focus is on biochemical and imaging biomarkers, but also other modalities are discussed. Neuroimaging is the most important diagnostic tool today, but α-syn imaging is not yet viable. Modern techniques allow measuring various forms of α-syn in cerebrospinal fluid, blood, saliva, and skin. Digital biomarkers and artificial intelligence offer new means for early diagnosis and longitudinal follow-up of degenerative brain diseases.
Collapse
Affiliation(s)
- Raimo K Tuominen
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Juho-Matti Renko
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
8
|
Ye D, Chukwu C, Yang Y, Hu Z, Chen H. Adeno-associated virus vector delivery to the brain: Technology advancements and clinical applications. Adv Drug Deliv Rev 2024; 211:115363. [PMID: 38906479 PMCID: PMC11892011 DOI: 10.1016/j.addr.2024.115363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Adeno-associated virus (AAV) vectors have emerged as a promising tool in the development of gene therapies for various neurological diseases, including Alzheimer's disease and Parkinson's disease. However, the blood-brain barrier (BBB) poses a significant challenge to successfully delivering AAV vectors to the brain. Strategies that can overcome the BBB to improve the AAV delivery efficiency to the brain are essential to successful brain-targeted gene therapy. This review provides an overview of existing strategies employed for AAV delivery to the brain, including direct intraparenchymal injection, intra-cerebral spinal fluid injection, intranasal delivery, and intravenous injection of BBB-permeable AAVs. Focused ultrasound has emerged as a promising technology for the noninvasive and spatially targeted delivery of AAV administered by intravenous injection. This review also summarizes each strategy's current preclinical and clinical applications in treating neurological diseases. Moreover, this review includes a detailed discussion of the recent advances in the emerging focused ultrasound-mediated AAV delivery. Understanding the state-of-the-art of these gene delivery approaches is critical for future technology development to fulfill the great promise of AAV in neurological disease treatment.
Collapse
Affiliation(s)
- Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Chinwendu Chukwu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Zhongtao Hu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA; Department of Neurosurgery, Washington University School of Medicine, Saint Louis, MO 63110 USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
9
|
Harkins AL, Ambegaokar PP, Keeler AM. Immune responses to central nervous system directed adeno-associated virus gene therapy: Does direct CNS delivery make a difference? Neurotherapeutics 2024; 21:e00435. [PMID: 39180957 PMCID: PMC11386282 DOI: 10.1016/j.neurot.2024.e00435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/11/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
Adeno-associated virus (AAV) mediated gene therapy is a leading gene delivery platform with potential to transform the landscape of treatment for neurological disorders. While AAV is deemed non-immunogenic compared to other viral vectors, adverse immune reactions have been observed in the clinic, raising concerns. As the central nervous system (CNS) has a tightly regulated immune system, characterized by a degree of tolerance, it has been considered a unique target for AAV gene therapy. AAV vectors have shown promising results for the treatment of several CNS disorders including Spinal Muscular Atrophy, Giant Axonal Neuropathy, Amyotrophic Lateral Sclerosis, Tay Sachs Disease, Parkinson's Disease, and others, demonstrating safety and success. The Food and Drug Administration (FDA) approval of Zolgensma and European Medicines Agency (EMA) approval of Upstaza, for Spinal Muscular Atrophy (SMA) and Aromatic l-amino acid decarboxylase deficiency (AADC) respectively, represent this success, all while highlighting significant differences in immune responses to AAV, particularly with regards to therapeutic administration route. AAV therapies like Upstaza that are injected directly into the immune-specialized brain have been characterized by mild immune response profiles and minor adverse events, whereas therapies like Zolgensma that are injected systemically demonstrate more robust immune stimulation and off-target toxicities. Despite these contrasting parallels, these therapeutics and others in the clinic have demonstrated clinical benefit for patients, warranting further exploration of immune responses to CNS-directed AAV clinical trials. Thus, in this review, we discuss effects of different routes of AAV administration on eliciting local and peripheral immune responses specifically observed in CNS-targeted trials.
Collapse
Affiliation(s)
- Ashley L Harkins
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, United States; Department of Neurology, University of Massachusetts Chan Medical School, United States; Horae Gene Therapy Center, University of Massachusetts Chan Medical School, United States
| | - Prajakta P Ambegaokar
- Graduate Program in Translational Science, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, United States; Horae Gene Therapy Center, University of Massachusetts Chan Medical School, United States
| | - Allison M Keeler
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, United States; Graduate Program in Translational Science, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, United States; NeuroNexus Institute, University of Massachusetts Chan Medical School, United States; Department of Pediatrics, University of Massachusetts Chan Medical School, United States; Horae Gene Therapy Center, University of Massachusetts Chan Medical School, United States.
| |
Collapse
|
10
|
Lopez-Gordo E, Chamberlain K, Riyad JM, Kohlbrenner E, Weber T. Natural Adeno-Associated Virus Serotypes and Engineered Adeno-Associated Virus Capsid Variants: Tropism Differences and Mechanistic Insights. Viruses 2024; 16:442. [PMID: 38543807 PMCID: PMC10975205 DOI: 10.3390/v16030442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 05/23/2024] Open
Abstract
Today, adeno-associated virus (AAV)-based vectors are arguably the most promising in vivo gene delivery vehicles for durable therapeutic gene expression. Advances in molecular engineering, high-throughput screening platforms, and computational techniques have resulted in a toolbox of capsid variants with enhanced performance over parental serotypes. Despite their considerable promise and emerging clinical success, there are still obstacles hindering their broader use, including limited transduction capabilities, tissue/cell type-specific tropism and penetration into tissues through anatomical barriers, off-target tissue biodistribution, intracellular degradation, immune recognition, and a lack of translatability from preclinical models to clinical settings. Here, we first describe the transduction mechanisms of natural AAV serotypes and explore the current understanding of the systemic and cellular hurdles to efficient transduction. We then outline progress in developing designer AAV capsid variants, highlighting the seminal discoveries of variants which can transduce the central nervous system upon systemic administration, and, to a lesser extent, discuss the targeting of the peripheral nervous system, eye, ear, lung, liver, heart, and skeletal muscle, emphasizing their tissue and cell specificity and translational promise. In particular, we dive deeper into the molecular mechanisms behind their enhanced properties, with a focus on their engagement with host cell receptors previously inaccessible to natural AAV serotypes. Finally, we summarize the main findings of our review and discuss future directions.
Collapse
|
11
|
Nouraein S, Lee S, Saenz VA, Del Mundo HC, Yiu J, Szablowski JO. Acoustically targeted noninvasive gene therapy in large brain volumes. Gene Ther 2024; 31:85-94. [PMID: 37696982 DOI: 10.1038/s41434-023-00421-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/23/2023] [Accepted: 08/31/2023] [Indexed: 09/13/2023]
Abstract
Focused Ultrasound Blood-Brain Barrier Opening (FUS-BBBO) can deliver adeno-associated viral vectors (AAVs) to treat genetic disorders of the brain. However, such disorders often affect large brain regions. Moreover, the applicability of FUS-BBBO in the treatment of brain-wide genetic disorders has not yet been evaluated. Herein, we evaluated the transduction efficiency and safety of opening up to 105 sites simultaneously. Increasing the number of targeted sites increased gene delivery efficiency at each site. We achieved transduction of up to 60% of brain cells with comparable efficiency in the majority of the brain regions. Furthermore, gene delivery with FUS-BBBO was safe even when all 105 sites were targeted simultaneously without negative effects on animal weight or neuronal loss. To evaluate the application of multi-site FUS-BBBO for gene therapy, we used it for gene editing using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9) system and found effective gene editing, but also a loss of neurons at the targeted sites. Overall, this study provides a brain-wide map of transduction efficiency, shows the synergistic effect of multi-site targeting on transduction efficiency, and is the first example of large brain volume gene editing after noninvasive gene delivery with FUS-BBBO.
Collapse
Affiliation(s)
- Shirin Nouraein
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
- Rice Neuroengineering Initiative, Rice University, Houston, TX, 77030, USA
- Synthetic, Systems, and Physical Biology Program, Rice University, Houston, TX, 77005, USA
| | - Sangsin Lee
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
- Rice Neuroengineering Initiative, Rice University, Houston, TX, 77030, USA
| | - Vidal A Saenz
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | | | - Joycelyn Yiu
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Jerzy O Szablowski
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
- Rice Neuroengineering Initiative, Rice University, Houston, TX, 77030, USA.
- Synthetic, Systems, and Physical Biology Program, Rice University, Houston, TX, 77005, USA.
- Applied Physics Program, Rice University, Houston, TX, 77005, USA.
| |
Collapse
|
12
|
Liu N, Liang X, Chen Y, Xie L. Recent trends in treatment strategies for Alzheimer 's disease and the challenges: A topical advancement. Ageing Res Rev 2024; 94:102199. [PMID: 38232903 DOI: 10.1016/j.arr.2024.102199] [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: 11/14/2023] [Accepted: 01/12/2024] [Indexed: 01/19/2024]
Abstract
Alzheimer's Disease (AD) is an irreversible and progressive neurological disease that has affected at least 50 million people around the globe. Considering the severity of the disease and the continuous increase in the number of patients, the development of new effective drugs or intervention strategies for AD has become urgent. AD is caused by a combination of genetic, environmental, and lifestyle factors, but its exact cause has not yet been clarified. Given the current challenges being faced in the clinical treatment of AD, such as complex AD pathological network and insufficient early diagnosis, herein, we have focused on the three core pathological features of AD, including amyloid-β (Aβ) aggregation, tau phosphorylation and tangles, and activation of inflammatory factors. In this review, we have briefly underscored the primary evidence supporting each pathology and discuss AD pathological network among Aβ, tau, and inflammation. We have also comprehensively summarized the most instructive drugs and their treatment strategies against Aβ, tau, or neuroinflammation used in basic research and clinical trials. Finally, we have discussed and outlined the pros and cons of each pathological approach and looked forward to potential personalized diagnosis and treatment strategies that are beneficial to AD patients.
Collapse
Affiliation(s)
- Ni Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Xiaohan Liang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Yu Chen
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Lihang Xie
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Gupta DP, Bhusal A, Rahman MH, Kim JH, Choe Y, Jang J, Jung HJ, Kim UK, Park JS, Maeng LS, Suk K, Song GJ. EBP50 is a key molecule for the Schwann cell-axon interaction in peripheral nerves. Prog Neurobiol 2023; 231:102544. [PMID: 37940033 DOI: 10.1016/j.pneurobio.2023.102544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023]
Abstract
Peripheral nerve injury disrupts the Schwann cell-axon interaction and the cellular communication between them. The peripheral nervous system has immense potential for regeneration extensively due to the innate plastic potential of Schwann cells (SCs) that allows SCs to interact with the injured axons and exert specific repair functions essential for peripheral nerve regeneration. In this study, we show that EBP50 is essential for the repair function of SCs and regeneration following nerve injury. The increased expression of EBP50 in the injured sciatic nerve of control mice suggested a significant role in regeneration. The ablation of EBP50 in mice resulted in delayed nerve repair, recovery of behavioral function, and remyelination following nerve injury. EBP50 deficiency led to deficits in SC functions, including proliferation, migration, cytoskeleton dynamics, and axon interactions. The adeno-associated virus (AAV)-mediated local expression of EBP50 improved SCs migration, functional recovery, and remyelination. ErbB2-related proteins were not differentially expressed in EBP50-deficient sciatic nerves following injury. EBP50 binds and stabilizes ErbB2 and activates the repair functions to promote regeneration. Thus, we identified EBP50 as a potent SC protein that can enhance the regeneration and functional recovery driven by NRG1-ErbB2 signaling, as well as a novel regeneration modulator capable of potential therapeutic effects.
Collapse
Affiliation(s)
- Deepak Prasad Gupta
- Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic of Korea; Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Anup Bhusal
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jae-Hong Kim
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jaemyung Jang
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyun Jin Jung
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Un-Kyung Kim
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jin-Sung Park
- Department of Neurology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Lee-So Maeng
- Department of Hospital Pathology, Incheon St. Mary's Hospital College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Gyun Jee Song
- Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic of Korea; Department of Medicine, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea.
| |
Collapse
|
16
|
Ling Q, Herstine JA, Bradbury A, Gray SJ. AAV-based in vivo gene therapy for neurological disorders. Nat Rev Drug Discov 2023; 22:789-806. [PMID: 37658167 DOI: 10.1038/s41573-023-00766-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2023] [Indexed: 09/03/2023]
Abstract
Recent advancements in gene supplementation therapy are expanding the options for the treatment of neurological disorders. Among the available delivery vehicles, adeno-associated virus (AAV) is often the favoured vector. However, the results have been variable, with some trials dramatically altering the course of disease whereas others have shown negligible efficacy or even unforeseen toxicity. Unlike traditional drug development with small molecules, therapeutic profiles of AAV gene therapies are dependent on both the AAV capsid and the therapeutic transgene. In this rapidly evolving field, numerous clinical trials of gene supplementation for neurological disorders are ongoing. Knowledge is growing about factors that impact the translation of preclinical studies to humans, including the administration route, timing of treatment, immune responses and limitations of available model systems. The field is also developing potential solutions to mitigate adverse effects, including AAV capsid engineering and designs to regulate transgene expression. At the same time, preclinical research is addressing new frontiers of gene supplementation for neurological disorders, with a focus on mitochondrial and neurodevelopmental disorders. In this Review, we describe the current state of AAV-mediated neurological gene supplementation therapy, including critical factors for optimizing the safety and efficacy of treatments, as well as unmet needs in this field.
Collapse
Affiliation(s)
- Qinglan Ling
- Department of Paediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jessica A Herstine
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Paediatrics, The Ohio State University, Columbus, OH, USA
| | - Allison Bradbury
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Paediatrics, The Ohio State University, Columbus, OH, USA
| | - Steven J Gray
- Department of Paediatrics, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
17
|
Ng J, Barral S, Waddington SN, Kurian MA. Gene Therapy for Dopamine Dyshomeostasis: From Parkinson's to Primary Neurotransmitter Diseases. Mov Disord 2023; 38:924-936. [PMID: 37147851 PMCID: PMC10946997 DOI: 10.1002/mds.29416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/23/2023] [Accepted: 04/05/2023] [Indexed: 05/07/2023] Open
Abstract
Neurological disorders encompass a broad range of neurodegenerative and neurodevelopmental diseases that are complex and almost universally without disease modifying treatments. There is, therefore, significant unmet clinical need to develop novel therapeutic strategies for these patients. Viral gene therapies are a promising approach, where gene delivery is achieved through viral vectors such as adeno-associated virus and lentivirus. The clinical efficacy of such gene therapies has already been observed in two neurological disorders of pediatric onset; for spinal muscular atrophy and aromatic L-amino acid decarboxylase (AADC) deficiency, gene therapy has significantly modified the natural history of disease in these life-limiting neurological disorders. Here, we review recent advances in gene therapy, focused on the targeted delivery of dopaminergic genes for Parkinson's disease and the primary neurotransmitter disorders, AADC deficiency and dopamine transporter deficiency syndrome (DTDS). Although recent European Medicines Agency and Medicines and Healthcare products Regulatory Agency approval of Upstaza (eladocagene exuparvovec) signifies an important landmark, numerous challenges remain. Future research will need to focus on defining the optimal therapeutic window for clinical intervention, better understanding of the duration of therapeutic efficacy, and improved brain targeting. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Joanne Ng
- Gene Transfer Technology Group, EGA‐Institute for Women's HealthUniversity College LondonLondonUnited Kingdom
- Genetic Therapy Accelerator Centre, Department of Neurodegenerative Disease, Queen Square Institute of NeurologyUniversity College LondonLondonUnited Kingdom
| | - Serena Barral
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS‐Institute of Child HealthUniversity College LondonLondonUnited Kingdom
| | - Simon N. Waddington
- Gene Transfer Technology Group, EGA‐Institute for Women's HealthUniversity College LondonLondonUnited Kingdom
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Manju A. Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS‐Institute of Child HealthUniversity College LondonLondonUnited Kingdom
- Department of NeurologyGreat Ormond Street Hospital for ChildrenLondonUnited Kingdom
| |
Collapse
|
18
|
Aly AEE, Sun T, Zhang Y, Li Z, Kyada M, Ma Q, Padegimas L, Sesenoglu-Laird O, Cooper MJ, McDannold NJ, Waszczak BL. Focused ultrasound enhances transgene expression of intranasal hGDNF DNA nanoparticles in the sonicated brain regions. J Control Release 2023; 358:498-509. [PMID: 37127076 DOI: 10.1016/j.jconrel.2023.04.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/03/2023]
Abstract
The therapeutic potential of many gene therapies is limited by their inability to cross the blood brain barrier (BBB). While intranasal administration of plasmid DNA nanoparticles (NPs) offers a non-invasive approach to bypass the BBB, it is not targeted to disease-relevant brain regions. Here, our goal was to determine whether focused ultrasound (FUS) can enrich intranasal delivery of our plasmid DNA NPs to target deeper brain regions, in this case the regions most affected in Parkinson's disease. Combining FUS with intranasal administration resulted in enhanced delivery of DNA NPs to the rodent brain, by recruitment and transfection of microglia. FUS increased transgene expression by over 3-fold after intranasal administration compared to intravenous administration. Additionally, FUS with intranasal delivery increased transgene expression in the sonicated hemisphere by over 80%, altered cellular transfection patterns at the sonication sites, and improved penetration of plasmid NPs into the brain parenchyma (with a 1-fold and 3-fold increase in proximity of transgene expression to neurons in the forebrain and midbrain respectively, and a 40% increase in proximity of transgene expression to dopaminergic neurons in the substantia nigra). These results provide evidence in support of using FUS to improve transgene expression after intranasal delivery of non-viral gene therapies.
Collapse
Affiliation(s)
- Amirah E-E Aly
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Tao Sun
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yongzhi Zhang
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zejun Li
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Margee Kyada
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Qingxi Ma
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | | | | | | | - Nathan J McDannold
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Barbara L Waszczak
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA.
| |
Collapse
|
19
|
Campos LJ, Arokiaraj CM, Chuapoco MR, Chen X, Goeden N, Gradinaru V, Fox AS. Advances in AAV technology for delivering genetically encoded cargo to the nonhuman primate nervous system. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 4:100086. [PMID: 37397806 PMCID: PMC10313870 DOI: 10.1016/j.crneur.2023.100086] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/05/2023] [Accepted: 03/17/2023] [Indexed: 07/04/2023] Open
Abstract
Modern neuroscience approaches including optogenetics, calcium imaging, and other genetic manipulations have facilitated our ability to dissect specific circuits in rodent models to study their role in neurological disease. These approaches regularly use viral vectors to deliver genetic cargo (e.g., opsins) to specific tissues and genetically-engineered rodents to achieve cell-type specificity. However, the translatability of these rodent models, cross-species validation of identified targets, and translational efficacy of potential therapeutics in larger animal models like nonhuman primates remains difficult due to the lack of efficient primate viral vectors. A refined understanding of the nonhuman primate nervous system promises to deliver insights that can guide the development of treatments for neurological and neurodegenerative diseases. Here, we outline recent advances in the development of adeno-associated viral vectors for optimized use in nonhuman primates. These tools promise to help open new avenues for study in translational neuroscience and further our understanding of the primate brain.
Collapse
Affiliation(s)
- Lillian J. Campos
- Department of Psychology and the California National Primate Research Center, University of California, Davis, CA, 05616, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Cynthia M. Arokiaraj
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Miguel R. Chuapoco
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Nick Goeden
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Capsida Biotherapeutics, Thousand Oaks, CA, 91320, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Andrew S. Fox
- Department of Psychology and the California National Primate Research Center, University of California, Davis, CA, 05616, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
20
|
Issa SS, Shaimardanova AA, Solovyeva VV, Rizvanov AA. Various AAV Serotypes and Their Applications in Gene Therapy: An Overview. Cells 2023; 12:785. [PMID: 36899921 PMCID: PMC10000783 DOI: 10.3390/cells12050785] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Despite scientific discoveries in the field of gene and cell therapy, some diseases still have no effective treatment. Advances in genetic engineering methods have enabled the development of effective gene therapy methods for various diseases based on adeno-associated viruses (AAVs). Today, many AAV-based gene therapy medications are being investigated in preclinical and clinical trials, and new ones are appearing on the market. In this article, we present a review of AAV discovery, properties, different serotypes, and tropism, and a following detailed explanation of their uses in gene therapy for disease of different organs and systems.
Collapse
Affiliation(s)
- Shaza S. Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Alisa A. Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
21
|
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
|
22
|
Protective mechanisms by glial cell line-derived neurotrophic factor and cerebral dopamine neurotrophic factor against the α-synuclein accumulation in Parkinson's disease. Biochem Soc Trans 2023; 51:245-257. [PMID: 36794783 DOI: 10.1042/bst20220770] [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: 10/13/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/17/2023]
Abstract
Synucleinopathies constitute a disease family named after alpha-synuclein protein, which is a significant component of the intracellular inclusions called Lewy bodies. Accompanying the progressive neurodegeneration, Lewy bodies and neurites are the main histopathologies of synucleinopathies. The complicated role of alpha-synuclein in the disease pathology makes it an attractive therapeutic target for disease-modifying treatments. GDNF is one of the most potent neurotrophic factors for dopamine neurons, whereas CDNF is protective and neurorestorative with entirely different mechanisms of action. Both have been in the clinical trials for the most common synucleinopathy, Parkinson's disease. With the AAV-GDNF clinical trials ongoing and the CDNF trial being finalized, their effects on abnormal alpha-synuclein accumulation are of great interest. Previous animal studies with an alpha-synuclein overexpression model have shown that GDNF was ineffective against alpha-synuclein accumulation. However, a recent study with cell culture and animal models of alpha-synuclein fibril inoculation has demonstrated the opposite by revealing that the GDNF/RET signaling cascade is required for the protective effect of GDNF on alpha-synuclein aggregation. CDNF, an ER resident protein, was shown to bind alpha-synuclein directly. CDNF reduced the uptake of alpha-synuclein fibrils by the neurons and alleviated the behavioral deficits induced by fibrils injected into the mouse brain. Thus, GDNF and CDNF can modulate different symptoms and pathologies of Parkinson's disease, and perhaps, similarly for other synucleinopathies. Their unique mechanisms for preventing alpha-synuclein-related pathology should be studied more carefully to develop disease-modifying therapies.
Collapse
|
23
|
Horton RH, Saade D, Markati T, Harriss E, Bönnemann CG, Muntoni F, Servais L. A systematic review of adeno-associated virus gene therapies in neurology: the need for consistent safety monitoring of a promising treatment. J Neurol Neurosurg Psychiatry 2022; 93:1276-1288. [PMID: 36190933 DOI: 10.1136/jnnp-2022-329431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
Adeno-associated virus (AAV) gene therapies are generating much excitement in the rare disease field, particularly for previously untreatable neurological conditions. Efficacy has been claimed for several gene therapy products and the number of trials is rapidly increasing. However, reports of severe treatment-related adverse reactions are emerging, including death. There is still insufficient knowledge about their aetiology, prevention and treatment. We therefore undertook to systematically review publicly available data on AAV gene therapies in order to collate existing information on both safety and efficacy. Here, we review emerging efficacy reports of these novel therapies, many of which show promise. We also collate an increasing number of adverse reactions. Overwhelmingly, these results make a case for unified reporting of adverse events. This is likely to be critical for improving the safety of these promising treatments.
Collapse
Affiliation(s)
| | - Dimah Saade
- Division of Neurology, University of Iowa, Iowa, USA
| | | | - Elinor Harriss
- Bodleian Health Care Libraries, University of Oxford, Oxford, UK
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Francesco Muntoni
- Dubowtiz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, UK, London, UK
| | - Laurent Servais
- Department of Paediatrics, University of Oxford, Oxford, UK .,Neuromuscular Reference Center, Department of Paediatrics, CHU of Liège, Liège, Belgium
| |
Collapse
|
24
|
Gomez Limia C, Baird M, Schwartz M, Saxena S, Meyer K, Wein N. Emerging Perspectives on Gene Therapy Delivery for Neurodegenerative and Neuromuscular Disorders. J Pers Med 2022; 12:1979. [PMID: 36556200 PMCID: PMC9788053 DOI: 10.3390/jpm12121979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 12/05/2022] Open
Abstract
Neurodegenerative disorders (NDDs), such as Alzheimer's disease (AD) and Parkinson's Disease (PD), are a group of heterogeneous diseases that mainly affect central nervous system (CNS) functions. A subset of NDDs exhibit CNS dysfunction and muscle degeneration, as observed in Gangliosidosis 1 (GM1) and late stages of PD. Neuromuscular disorders (NMDs) are a group of diseases in which patients show primary progressive muscle weaknesses, including Duchenne Muscular Dystrophy (DMD), Pompe disease, and Spinal Muscular Atrophy (SMA). NDDs and NMDs typically have a genetic component, which affects the physiological functioning of critical cellular processes, leading to pathogenesis. Currently, there is no cure or efficient treatment for most of these diseases. More than 200 clinical trials have been completed or are currently underway in order to establish safety, tolerability, and efficacy of promising gene therapy approaches. Thus, gene therapy-based therapeutics, including viral or non-viral delivery, are very appealing for the treatment of NDDs and NMDs. In particular, adeno-associated viral vectors (AAV) are an attractive option for gene therapy for NDDs and NMDs. However, limitations have been identified after systemic delivery, including the suboptimal capacity of these therapies to traverse the blood-brain barrier (BBB), degradation of the particles during the delivery, high reactivity of the patient's immune system during the treatment, and the potential need for redosing. To circumvent these limitations, several preclinical and clinical studies have suggested intrathecal (IT) delivery to target the CNS and peripheral organs via cerebrospinal fluid (CSF). CSF administration can vastly improve the delivery of small molecules and drugs to the brain and spinal cord as compared to systemic delivery. Here, we review AAV biology and vector design elements, different therapeutic routes of administration, and highlight CSF delivery as an attractive route of administration. We discuss the different aspects of neuromuscular and neurodegenerative diseases, such as pathogenesis, the landscape of mutations, and the biological processes associated with the disease. We also describe the hallmarks of NDDs and NMDs as well as discuss current therapeutic approaches and clinical progress in viral and non-viral gene therapy and enzyme replacement strategies for those diseases.
Collapse
Affiliation(s)
- Cintia Gomez Limia
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Megan Baird
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Maura Schwartz
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Smita Saxena
- Department of Neurology, Inselspital, 3010 Bern, Switzerland
| | - Kathrin Meyer
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| | - Nicolas Wein
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| |
Collapse
|
25
|
Hayes G, Pinto J, Sparks SN, Wang C, Suri S, Bulte DP. Vascular smooth muscle cell dysfunction in neurodegeneration. Front Neurosci 2022; 16:1010164. [PMID: 36440263 PMCID: PMC9684644 DOI: 10.3389/fnins.2022.1010164] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/24/2022] [Indexed: 09/01/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the key moderators of cerebrovascular dynamics in response to the brain's oxygen and nutrient demands. Crucially, VSMCs may provide a sensitive biomarker for neurodegenerative pathologies where vasculature is compromised. An increasing body of research suggests that VSMCs have remarkable plasticity and their pathophysiology may play a key role in the complex process of neurodegeneration. Furthermore, extrinsic risk factors, including environmental conditions and traumatic events can impact vascular function through changes in VSMC morphology. VSMC dysfunction can be characterised at the molecular level both preclinically, and clinically ex vivo. However the identification of VSMC dysfunction in living individuals is important to understand changes in vascular function at the onset and progression of neurological disorders such as dementia, Alzheimer's disease, and Parkinson's disease. A promising technique to identify changes in the state of cerebral smooth muscle is cerebrovascular reactivity (CVR) which reflects the intrinsic dynamic response of blood vessels in the brain to vasoactive stimuli in order to modulate regional cerebral blood flow (CBF). In this work, we review the role of VSMCs in the most common neurodegenerative disorders and identify physiological systems that may contribute to VSMC dysfunction. The evidence collected here identifies VSMC dysfunction as a strong candidate for novel therapeutics to combat the development and progression of neurodegeneration, and highlights the need for more research on the role of VSMCs and cerebrovascular dynamics in healthy and diseased states.
Collapse
Affiliation(s)
- Genevieve Hayes
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Joana Pinto
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Sierra N. Sparks
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Congxiyu Wang
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Sana Suri
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Daniel P. Bulte
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
26
|
Gene-Based Therapeutics for Parkinson’s Disease. Biomedicines 2022; 10:biomedicines10081790. [PMID: 35892690 PMCID: PMC9331241 DOI: 10.3390/biomedicines10081790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a complex multifactorial disorder that is not yet fully surmised, and it is only when such a disease is tackled on multiple levels simultaneously that we should expect to see fruitful results. Gene therapy is a modern medical practice that theoretically and, so far, practically, has demonstrated its capability in joining the battle against PD and other complex disorders on most if not all fronts. This review discusses how gene therapy can efficiently replace current forms of therapy such as drugs, personalized medicine or invasive surgery. Furthermore, we discuss the importance of enhancing delivery techniques to increase the level of transduction and control of gene expression or tissue specificity. Importantly, the results of current trials establish the safety, efficacy and applicability of gene therapy for PD. Gene therapy’s variety of potential in interfering with PD’s pathology by improving basal ganglial circuitry, enhancing dopamine synthesis, delivering neuroprotection or preventing neurodegeneration may one day achieve symptomatic benefit, disease modification and eradication.
Collapse
|
27
|
Wu Y, Rakotoarisoa M, Angelov B, Deng Y, Angelova A. Self-Assembled Nanoscale Materials for Neuronal Regeneration: A Focus on BDNF Protein and Nucleic Acid Biotherapeutic Delivery. NANOMATERIALS 2022; 12:nano12132267. [PMID: 35808102 PMCID: PMC9268293 DOI: 10.3390/nano12132267] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023]
Abstract
Enabling challenging applications of nanomedicine and precision medicine in the treatment of neurodegenerative disorders requires deeper investigations of nanocarrier-mediated biomolecular delivery for neuronal targeting and recovery. The successful use of macromolecular biotherapeutics (recombinant growth factors, antibodies, enzymes, synthetic peptides, cell-penetrating peptide–drug conjugates, and RNAi sequences) in clinical developments for neuronal regeneration should benefit from the recent strategies for enhancement of their bioavailability. We highlight the advances in the development of nanoscale materials for drug delivery in neurodegenerative disorders. The emphasis is placed on nanoformulations for the delivery of brain-derived neurotrophic factor (BDNF) using different types of lipidic nanocarriers (liposomes, liquid crystalline or solid lipid nanoparticles) and polymer-based scaffolds, nanofibers and hydrogels. Self-assembled soft-matter nanoscale materials show favorable neuroprotective characteristics, safety, and efficacy profiles in drug delivery to the central and peripheral nervous systems. The advances summarized here indicate that neuroprotective biomolecule-loaded nanoparticles and injectable hydrogels can improve neuronal survival and reduce tissue injury. Certain recently reported neuronal dysfunctions in long-COVID-19 survivors represent early manifestations of neurodegenerative pathologies. Therefore, BDNF delivery systems may also help in prospective studies on recovery from long-term COVID-19 neurological complications and be considered as promising systems for personalized treatment of neuronal dysfunctions and prevention or retarding of neurodegenerative disorders.
Collapse
Affiliation(s)
- Yu Wu
- CNRS, Institut Galien Paris-Saclay, Université Paris-Saclay, F-92290 Châtenay-Malabry, France; (Y.W.); (M.R.)
| | - Miora Rakotoarisoa
- CNRS, Institut Galien Paris-Saclay, Université Paris-Saclay, F-92290 Châtenay-Malabry, France; (Y.W.); (M.R.)
| | - Borislav Angelov
- Institute of Physics, ELI Beamlines, Academy of Sciences of the Czech Republic, Na Slovance 2, CZ-18221 Prague, Czech Republic;
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, No. 1, Jinlian Road, Longwan District, Wenzhou 325001, China;
| | - Angelina Angelova
- CNRS, Institut Galien Paris-Saclay, Université Paris-Saclay, F-92290 Châtenay-Malabry, France; (Y.W.); (M.R.)
- Correspondence:
| |
Collapse
|
28
|
Paul A, Collins MG, Lee HY. Gene Therapy: The Next-Generation Therapeutics and Their Delivery Approaches for Neurological Disorders. Front Genome Ed 2022; 4:899209. [PMID: 35832929 PMCID: PMC9272754 DOI: 10.3389/fgeed.2022.899209] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/05/2022] [Indexed: 12/14/2022] Open
Abstract
Neurological conditions like neurodevelopmental disorders and neurodegenerative diseases are quite complex and often exceedingly difficult for patients. Most of these conditions are due to a mutation in a critical gene. There is no cure for the majority of these neurological conditions and the availability of disease-modifying therapeutics is quite rare. The lion's share of the treatments that are available only provide symptomatic relief, as such, we are in desperate need of an effective therapeutic strategy for these conditions. Considering the current drug development landscape, gene therapy is giving us hope as one such effective therapeutic strategy. Consistent efforts have been made to develop gene therapy strategies using viral and non-viral vectors of gene delivery. Here, we have discussed both of these delivery methods and their properties. We have summarized the relative advantages and drawbacks of viral and non-viral vectors from the perspectives of safety, efficiency, and productivity. Recent developments such as clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated gene editing and its use in vivo have been described here as well. Given recent advancements, gene therapy shows great promise to emerge as a next-generation therapeutic for many of the neurodevelopmental and neurodegenerative conditions.
Collapse
|
29
|
Sola P, Krishnamurthy PT, Kumari M, Byran G, Gangadharappa HV, Garikapati KK. Neuroprotective approaches to halt Parkinson's disease progression. Neurochem Int 2022; 158:105380. [PMID: 35718278 DOI: 10.1016/j.neuint.2022.105380] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023]
Abstract
One of the most significant threats in Parkinson's disease (PD) is neurodegeneration. Neurodegeneration at both nigral as well as non-nigral regions of the brain is considered responsible for disease progression in PD. The key factors that initiate neurodegeneration are oxidative stress, neuroinflammation, mitochondrial complex-1 inhibition, and abnormal α-synuclein (SNCA) protein aggregations. Nigral neurodegeneration results in motor symptoms (tremor, bradykinesia, rigidity, shuffling gait, and postural instability) whereas; non-nigral neurodegeneration is responsible for non-motor symptoms (depression, cognitive dysfunctions, sleep disorders, hallucination, and psychosis). The available therapies for PD aim at increasing dopamine levels. The medications such as Monoamine oxidase B (MAO-B) inhibitors, catechol o-methyltransferase (COMT) inhibitors, Dopamine precursor (Levodopa), dopamine agonists, and dopamine reuptake inhibitors drastically improve the motor symptoms and quality of life only in the early stages of the disease. However, dopa resistant motor symptoms (abnormality in posture, speech impediment, gait, and balance problems), dopa resistant non-motor signs (sleep problems, autonomic dysfunction, mood, and cognitive impairment, pain), and drug-related side effects (motor fluctuations, psychosis, and dyskinesias) are considered responsible for the failure of these therapies. Further, none of the treatments, alone or in combination, are capable of halting the disease progression in the long run. Therefore, there is a need to develop safe and efficient neuroprotective agents, which can slow or stop the disease progression for the better management of PD. In this review, an effort has been made to discuss the various mechanisms responsible for progressive neurodegeneration (disease progression) in PD and also multiple strategies available for halting disease progression.
Collapse
Affiliation(s)
- Piyong Sola
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India.
| | - Mamta Kumari
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Gowramma Byran
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | | | - Kusuma Kumari Garikapati
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| |
Collapse
|
30
|
Stefani A, Pierantozzi M, Cardarelli S, Stefani L, Cerroni R, Conti M, Garasto E, Mercuri NB, Marini C, Sucapane P. Neurotrophins as Therapeutic Agents for Parkinson’s Disease; New Chances From Focused Ultrasound? Front Neurosci 2022; 16:846681. [PMID: 35401084 PMCID: PMC8990810 DOI: 10.3389/fnins.2022.846681] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/31/2022] [Indexed: 01/02/2023] Open
Abstract
Magnetic Resonance–guided Focused Ultrasound (MRgFUS) represents an effective micro-lesioning approach to target pharmaco-resistant tremor, mostly in patients afflicted by essential tremor (ET) and/or Parkinson’s disease (PD). So far, experimental protocols are verifying the clinical extension to other facets of the movement disorder galaxy (i.e., internal pallidus for disabling dyskinesias). Aside from those neurosurgical options, one of the most intriguing opportunities of this technique relies on its capability to remedy the impermeability of blood–brain barrier (BBB). Temporary BBB opening through low-intensity focused ultrasound turned out to be safe and feasible in patients with PD, Alzheimer’s disease, and amyotrophic lateral sclerosis. As a mere consequence of the procedures, some groups described even reversible but significant mild cognitive amelioration, up to hippocampal neurogenesis partially associated to the increased of endogenous brain-derived neurotrophic factor (BDNF). A further development elevates MRgFUS to the status of therapeutic tool for drug delivery of putative neurorestorative therapies. Since 2012, FUS-assisted intravenous administration of BDNF or neurturin allowed hippocampal or striatal delivery. Experimental studies emphasized synergistic modalities. In a rodent model for Huntington’s disease, engineered liposomes can carry glial cell line–derived neurotrophic factor (GDNF) plasmid DNA (GDNFp) to form a GDNFp-liposome (GDNFp-LPs) complex through pulsed FUS exposures with microbubbles; in a subacute MPTP-PD model, the combination of intravenous administration of neurotrophic factors (either through protein or gene delivery) plus FUS did curb nigrostriatal degeneration. Here, we explore these arguments, focusing on the current, translational application of neurotrophins in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alessandro Stefani
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
- *Correspondence: Alessandro Stefani,
| | | | - Silvia Cardarelli
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Lucrezia Stefani
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Rocco Cerroni
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Matteo Conti
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Elena Garasto
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Nicola B. Mercuri
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Carmine Marini
- UOC Neurology and Stroke Unit, University of L’Aquila, L’Aquila, Italy
| | | |
Collapse
|
31
|
Fajardo-Serrano A, Rico AJ, Roda E, Honrubia A, Arrieta S, Ariznabarreta G, Chocarro J, Lorenzo-Ramos E, Pejenaute A, Vázquez A, Lanciego JL. Adeno-Associated Viral Vectors as Versatile Tools for Neurological Disorders: Focus on Delivery Routes and Therapeutic Perspectives. Biomedicines 2022; 10:biomedicines10040746. [PMID: 35453499 PMCID: PMC9025350 DOI: 10.3390/biomedicines10040746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/10/2022] [Accepted: 03/21/2022] [Indexed: 11/24/2022] Open
Abstract
It is without doubt that the gene therapy field is currently in the spotlight for the development of new therapeutics targeting unmet medical needs. Thus, considering the gene therapy scenario, neurological diseases in general and neurodegenerative disorders in particular are emerging as the most appealing choices for new therapeutic arrivals intended to slow down, stop, or even revert the natural progressive course that characterizes most of these devastating neurodegenerative processes. Since an extensive coverage of all available literature is not feasible in practical terms, here emphasis was made in providing some advice to beginners in the field with a narrow focus on elucidating the best delivery route available for fulfilling any given AAV-based therapeutic approach. Furthermore, it is worth nothing that the number of ongoing clinical trials is increasing at a breath-taking speed. Accordingly, a landscape view of preclinical and clinical initiatives is also provided here in an attempt to best illustrate what is ongoing in this quickly expanding field.
Collapse
Affiliation(s)
- Ana Fajardo-Serrano
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Correspondence: (A.F.-S.); (J.L.L.)
| | - Alberto J. Rico
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Elvira Roda
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Adriana Honrubia
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Sandra Arrieta
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Goiaz Ariznabarreta
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Julia Chocarro
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Elena Lorenzo-Ramos
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Alvaro Pejenaute
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Alfonso Vázquez
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Department of Neurosurgery, Servicio Navarro de Salud, Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
| | - José Luis Lanciego
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Correspondence: (A.F.-S.); (J.L.L.)
| |
Collapse
|
32
|
Li H, Sun B, Huang Y, Zhang J, Xu X, Shen Y, Chen Z, Yang J, Shen L, Hu Y, Gu H. Gene therapy of yeast NDI1 on mitochondrial complex I dysfunction in rotenone-induced Parkinson’s disease models in vitro and vivo. Mol Med 2022; 28:29. [PMID: 35255803 PMCID: PMC8900322 DOI: 10.1186/s10020-022-00456-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/18/2022] [Indexed: 01/18/2023] Open
Abstract
Abstract
Purpose
Parkinson's disease (PD) is the second most common neurodegenerative disease without cure or effective treatment. This study explores whether the yeast internal NADH-quinone oxidoreductase (NDI1) can functionally replace the defective mammalian mitochondrial complex I, which may provide a gene therapy strategy for treating sporadic PD caused by mitochondrial complex I dysfunction.
Method
Recombinant lentivirus expressing NDI1 was transduced into SH-SY5Y cells, or recombinant adeno-associated virus type 5 expressing NDI1 was transduced into the right substantia nigra pars compacta (SNpc) of mouse. PD cell and mouse models were established by rotenone treatment. The therapeutic effects of NDI1 on rotenone-induced PD models in vitro and vivo were assessed in neurobehavior, neuropathology, and mitochondrial functions, by using the apomorphine-induced rotation test, immunohistochemistry, immunofluorescence, western blot, complex I enzyme activity determination, oxygen consumption detection, ATP content determination and ROS measurement.
Results
NDI1 was expressed and localized in mitochondria in SH-SY5Y cells. NDI1 resisted rotenone-induced changes in cell morphology, loss of cell viability, accumulation of α-synuclein and pS129 α-synuclein, mitochondrial ROS production and mitochondria-mediated apoptosis. The basal and maximal oxygen consumption, mitochondrial coupling efficiency, basal and oligomycin-sensitive ATP and complex I activity in cell model were significantly increased in rotenone + NDI1 group compared to rotenone + vector group. NDI1 was efficiently expressed in dopaminergic neurons in the right SNpc without obvious adverse effects. The rotation number to the right side (NDI1-treated side) was significantly increased compared to that to the left side (untreated side) in mouse model. The number of viable dopaminergic neurons, the expression of tyrosine hydroxylase, total and maximal oxygen consumption, mitochondrial coupling efficiency and complex I enzyme activity in right substantia nigra, and the content of dopamine in right striatum were significantly increased in rotenone + NDI1 group compared to rotenone + vector group.
Conclusion
Yeast NDI1 can rescue the defect of oxidative phosphorylation in rotenone-induced PD cell and mouse models, and ameliorate neurobehavioral and neuropathological damages. The results may provide a basis for the yeast NDI1 gene therapy of sporadic PD caused by mitochondrial complex I dysfunction.
Collapse
|
33
|
Beiki R, Khaghani M, Esmaeili F, Dehghanian F. Synergistic Effects of Combined Nurr1 Overexpression and Natural Inducers on the More Efficient Production of Dopaminergic Neuron-Like Cells From Stem Cells. Front Cell Neurosci 2022; 15:803272. [PMID: 35087379 PMCID: PMC8787052 DOI: 10.3389/fncel.2021.803272] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/16/2021] [Indexed: 11/21/2022] Open
Abstract
The development of dopaminergic (DA) neurons is a very complex process, and a combination of extrinsic and intrinsic factors involves their differentiation. Transcription factor, Nurr1 plays an essential role in the differentiation and maintenance of midbrain DA neurons. Nurr1-based therapies may restore DA function in Parkinson's disease (PD) by replacing damaged cells with differentiated cells derived from stem cells. Providing tissue-specific microenvironments such as brain extract can effectively induce dopaminergic gene expression in stem cells. The present study aimed to investigate the combined effects of Nurr1 gene overexpression and a neonatal rat brain extract (NRBE) induction on dopaminergic differentiation of P19 stem cells. In order to neural differentiation induction, stably Nurr1-transfected cells were treated with 100 μg/ml of NRBE. The differentiation potential of the cells was then evaluated during a period of 1-3 weeks via various methods. The initial evaluation of the cells by direct observation under a light microscope and cresyl violet specific staining, confirmed neuron-like morphology in the differentiated cells. In addition, different molecular and cellular techniques, including real-time PCR, immunofluorescence, and flow cytometry, demonstrated that the treated cells expressed pan-neuronal and dopaminergic markers. In all experimental groups, neuronal phenotype with dopaminergic neuron-like cells characteristics mainly appeared in the second week of the differentiation protocol. Overall, the results of the present study revealed for the first time the synergistic effects of Nurr1 gene overexpression and possible soluble factors that existed in NRBE on the differentiation of P19 stem cells into dopaminergic neuron-like cells.
Collapse
Affiliation(s)
| | | | - Fariba Esmaeili
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | | |
Collapse
|
34
|
Chau MJ, Quintero JE, Monje PV, Voss SR, Welleford AS, Gerhardt GA, van Horne CG. Using a Transection Paradigm to Enhance the Repair Mechanisms of an Investigational Human Cell Therapy. Cell Transplant 2022; 31:9636897221123515. [PMID: 36169034 PMCID: PMC9523845 DOI: 10.1177/09636897221123515] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/16/2022] [Indexed: 12/02/2022] Open
Abstract
One promising strategy in cell therapies for Parkinson's disease (PD) is to harness a patient's own cells to provide neuroprotection in areas of the brain affected by neurodegeneration. No treatment exists to replace cells in the brain. Thus, our goal has been to support sick neurons and slow neurodegeneration by transplanting living repair tissue from the peripheral nervous system into the substantia nigra of those with PD. Our group has pioneered the transplantation of transection-activated sural nerve fascicles into the brain of human subjects with PD. Our experience in sural nerve transplantation has supported the safety and feasibility of this approach. As part of a paradigm to assess the reparative properties of human sural nerve following a transection injury, we collected nerve tissue approximately 2 weeks after sural nerve transection for immunoassays from 15 participants, and collected samples from two additional participants for single nuclei RNA sequencing. We quantified the expression of key neuroprotective and select anti-apoptotic genes along with their corresponding protein levels using immunoassays. The single nuclei data clustered into 10 distinctive groups defined on the basis of previously published cell type-specific genes. Transection-induced reparative peripheral nerve tissue showed RNA expression of neuroprotective factors and anti-apoptotic factors across multiple cell types after nerve injury induction. Key proteins of interest (BDNF, GDNF, beta-NGF, PDGFB, and VEGF) were upregulated in reparative tissue. These results provide insight on this repair tissue's utility as a neuroprotective cell therapy.
Collapse
Affiliation(s)
- Monica J. Chau
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jorge E. Quintero
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Paula V. Monje
- Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephen Randal Voss
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Andrew S. Welleford
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Greg A. Gerhardt
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Craig G. van Horne
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
35
|
Machtakova M, Thérien-Aubin H, Landfester K. Polymer nano-systems for the encapsulation and delivery of active biomacromolecular therapeutic agents. Chem Soc Rev 2021; 51:128-152. [PMID: 34762084 DOI: 10.1039/d1cs00686j] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biomacromolecular therapeutic agents, particularly proteins, antigens, enzymes, and nucleic acids are emerging as powerful candidates for the treatment of various diseases and the development of the recent vaccine based on mRNA highlights the enormous potential of this class of drugs for future medical applications. However, biomacromolecular therapeutic agents present an enormous delivery challenge compared to traditional small molecules due to both a high molecular weight and a sensitive structure. Hence, the translation of their inherent pharmaceutical capacity into functional therapies is often hindered by the limited performance of conventional delivery vehicles. Polymer drug delivery systems are a modular solution able to address those issues. In this review, we discuss recent developments in the design of polymer delivery systems specifically tailored to the delivery challenges of biomacromolecular therapeutic agents. In the future, only in combination with a multifaceted and highly tunable delivery system, biomacromolecular therapeutic agents will realize their promising potential for the treatment of diseases and for the future of human health.
Collapse
Affiliation(s)
- Marina Machtakova
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Héloïse Thérien-Aubin
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. .,Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada.
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| |
Collapse
|
36
|
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
|
37
|
Larson PS. Improved Delivery Methods for Gene Therapy and Cell Transplantation in Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2021; 11:S199-S206. [PMID: 34366372 PMCID: PMC8543258 DOI: 10.3233/jpd-212710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A number of cell transplantation and gene therapy trials have been performed over the last three decades in an effort to restore function in Parkinson’s disease. Much has been learned about optimizing delivery methods for these therapeutics. This is particularly true in gene therapy, which has predominated the clinical trial landscape in recent years; however, cell transplantation for Parkinson’s disease is currently undergoing a renaissance. Innovations such as cannula design, iMRI-guided surgery and an evolution in delivery strategy has radically changed the way investigators approach clinical trial design. Future therapeutic strategies may employ newer delivery methods such as chronically implanted infusion devices and focal opening of the blood brain barrier with focused ultrasound.
Collapse
Affiliation(s)
- Paul S Larson
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
38
|
Van Laar AD, Van Laar VS, San Sebastian W, Merola A, Bradley Elder J, Lonser RR, Bankiewicz KS. An Update on Gene Therapy Approaches for Parkinson's Disease: Restoration of Dopaminergic Function. JOURNAL OF PARKINSONS DISEASE 2021; 11:S173-S182. [PMID: 34366374 PMCID: PMC8543243 DOI: 10.3233/jpd-212724] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
At present there is a significant unmet need for clinically available treatments for Parkinson’s disease (PD) patients to stably restore balance to dopamine network function, leaving patients with inadequate management of symptoms as the disease progresses. Gene therapy is an attractive approach to impart a durable effect on neuronal function through introduction of genetic material to reestablish dopamine levels and/or functionally recover dopaminergic signaling by improving neuronal health. Ongoing clinical gene therapy trials in PD are focused on enzymatic enhancement of dopamine production and/or the restoration of the nigrostriatal pathway to improve dopaminergic network function. In this review, we discuss data from current gene therapy trials for PD and recent advances in study design and surgical approaches.
Collapse
Affiliation(s)
- Amber D Van Laar
- Asklepios BioPharmaceutical, Inc., Columbus, OH, USA.,Department of Neurology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Victor S Van Laar
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, OH, USA
| | - Waldy San Sebastian
- Asklepios BioPharmaceutical, Inc., Columbus, OH, USA.,Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Aristide Merola
- Department of Neurology, College of Medicine, the Ohio State University, Columbus, OH, USA
| | - J Bradley Elder
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, OH, USA
| | - Russell R Lonser
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, OH, USA
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, OH, USA.,Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
39
|
Current Therapies in Clinical Trials of Parkinson's Disease: A 2021 Update. Pharmaceuticals (Basel) 2021; 14:ph14080717. [PMID: 34451813 PMCID: PMC8398928 DOI: 10.3390/ph14080717] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/15/2021] [Accepted: 07/22/2021] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that currently has no cure, but treatments are available to improve PD symptoms and maintain quality of life. In 2020, about 10 million people worldwide were living with PD. In 1970, the United States Food and Drug Administration approved the drug levodopa as a dopamine replacement to manage PD motor symptoms; levodopa-carbidopa combination became commercialized in 1975. After over 50 years of use, levodopa is still the gold standard for PD treatment. Unfortunately, levodopa therapy-induced dyskinesia and OFF symptoms remain unresolved. Therefore, we urgently need to analyze each current clinical trial's status and therapeutic strategy to discover new therapeutic approaches for PD treatment. We surveyed 293 registered clinical trials on ClinicalTrials.gov from 2008 to 16 June 2021. After excluded levodopa/carbidopa derivative add-on therapies, we identified 47 trials as PD treatment drugs or therapies. Among them, 19 trials are in phase I (41%), 25 trials are in phase II (53%), and 3 trials are in phase III (6%). The three phase-III trials use embryonic dopamine cell implant, 5-HT1A receptor agonist (sarizotan), and adenosine A2A receptor antagonist (caffeine). The therapeutic strategy of each trial shows 29, 5, 1, 5, 5, and 2 trials use small molecules, monoclonal antibodies, plasma therapy, cell therapy, gene therapy, and herbal extract, respectively. Additionally, we discuss the most potent drug or therapy among these trials. By systematically updating the current trial status and analyzing the therapeutic strategies, we hope this review can provide new ideas and insights for PD therapy development.
Collapse
|
40
|
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
|
41
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
42
|
Zhu D, Schieferecke AJ, Lopez PA, Schaffer DV. Adeno-Associated Virus Vector for Central Nervous System Gene Therapy. Trends Mol Med 2021; 27:524-537. [PMID: 33895085 DOI: 10.1016/j.molmed.2021.03.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022]
Abstract
The past several years have witnessed significant advances in the development of therapeutic gene delivery for neurological disorders of the central nervous system (CNS). In particular, genome-wide sequencing analysis has deepened our understanding of mutations that underlie many monogenic disorders, which in turn has contributed to clinical advances involving adeno-associated virus (AAV) vector delivery of replacement genes to treat recessive disorders. Moreover, gene therapy has been further bolstered with advances in genome editing tools that allow researchers to silence, repair, and amend endogenous genes. However, despite strong preclinical and clinical progress, challenges remain, including delivery and safety. Here, we discuss advances in AAV engineering, recent developments in cargo design, and translation of these technologies towards clinical progress.
Collapse
Affiliation(s)
- Danqing Zhu
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA, 94720, USA
| | - Adam J Schieferecke
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Paola A Lopez
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - David V Schaffer
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA, 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA; Department of Bioengineering, University of California, Berkeley, CA, 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
43
|
Merola A, Kobayashi N, Romagnolo A, Wright BA, Artusi CA, Imbalzano G, Litvan I, Van Laar AD, Bankiewicz K. Gene Therapy in Movement Disorders: A Systematic Review of Ongoing and Completed Clinical Trials. Front Neurol 2021; 12:648532. [PMID: 33889127 PMCID: PMC8056023 DOI: 10.3389/fneur.2021.648532] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/26/2021] [Indexed: 01/14/2023] Open
Abstract
Introduction: We sought to provide an overview of the published and currently ongoing movement disorders clinical trials employing gene therapy, defined as a technology aiming to modulate the expression of one or more genes to achieve a therapeutic benefit. Methods: We systematically reviewed movement disorders gene therapy clinical trials from PubMed and ClinicalTrials.gov using a searching strategy that included Parkinson disease (PD), Huntington disease (HD), amino acid decarboxylase (AADC) deficiency, multiple system atrophy (MSA), progressive supranuclear palsy (PSP), dystonia, tremor, ataxia, and other movement disorders. Data extracted included study characteristics, investigational product, route of administration, safety/tolerability, motor endpoints, and secondary outcomes (i.e., neuroimaging, biomarkers). Results: We identified a total of 46 studies focusing on PD (21 published and nine ongoing), HD (2 published and 5 ongoing), AADC deficiency (4 published and 2 ongoing), MSA (2 ongoing), and PSP (1 ongoing). In PD, intraparenchymal infusion of viral vector-mediated gene therapies demonstrated to be safe and showed promising preliminary data in trials aiming at restoring the synthesis of dopamine, enhancing the production of neurotrophic factors, or modifying the functional interaction between different nodes of the basal ganglia. In HD, monthly intrathecal delivery of an antisense oligonucleotide (ASO) targeting the huntingtin protein (HTT) mRNA proved to be safe and tolerable, and demonstrated a dose-dependent reduction of the cerebrospinal fluid levels of mutated HTT, while a small phase-I study testing implantable capsules of cells engineered to synthesize ciliary neurotrophic factor failed to show consistent drug delivery. In AADC deficiency, gene replacement studies demonstrated to be relatively safe in restoring catecholamine and serotonin synthesis, with promising outcomes. Ongoing movement disorders clinical trials are focusing on a variety of gene therapy approaches including alternative viral vector serotypes, novel recombinant genes, novel delivery techniques, and ASOs for the treatment of HD, MSA, and distinct subtypes of PD (LRRK2 mutation or GBA1 mutation carriers). Conclusion: Initial phase-I and -II studies tested the safety and feasibility of gene therapy in PD, HD, and AADC deficiency. The ongoing generation of clinical trials aims to test the efficacy of these approaches and explore additional applications for gene therapy in movement disorders.
Collapse
Affiliation(s)
- Aristide Merola
- Department of Neurology, Madden Center for Parkinson Disease and Other Movement Disorders, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | | | - Alberto Romagnolo
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Brenton A. Wright
- Department of Neurosciences, Parkinson and Other Movement Disorders Center, University of California, San Diego, La Jolla, CA, United States
| | - Carlo Alberto Artusi
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Gabriele Imbalzano
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Irene Litvan
- Department of Neurosciences, Parkinson and Other Movement Disorders Center, University of California, San Diego, La Jolla, CA, United States
| | - Amber D. Van Laar
- Asklepios BioPharmaceutical Inc., Research Triangle, NC, United States
- Department of Neurology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Krystof Bankiewicz
- Department of Neurological Surgery, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
44
|
Sun J, Roy S. Gene-based therapies for neurodegenerative diseases. Nat Neurosci 2021; 24:297-311. [PMID: 33526943 PMCID: PMC8394447 DOI: 10.1038/s41593-020-00778-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Gene therapy is making a comeback. With its twin promise of targeting disease etiology and 'long-term correction', gene-based therapies (defined here as all forms of genome manipulation) are particularly appealing for neurodegenerative diseases, for which conventional pharmacologic approaches have been largely disappointing. The recent success of a viral-vector-based gene therapy in spinal muscular atrophy-promoting survival and motor function with a single intravenous injection-offers a paradigm for such therapeutic intervention and a platform to build on. Although challenges remain, the newfound optimism largely stems from advances in the development of viral vectors that can diffusely deliver genes throughout the CNS, as well as genome-engineering tools that can manipulate disease pathways in ways that were previously impossible. Surely spinal muscular atrophy cannot be the only neurodegenerative disease amenable to gene therapy, and one can imagine a future in which the toolkit of a clinician will include gene-based therapeutics. The goal of this Review is to highlight advances in the development and application of gene-based therapies for neurodegenerative diseases and offer a prospective look into this emerging arena.
Collapse
Affiliation(s)
- Jichao Sun
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Subhojit Roy
- Department of Pathology, University of California, San Diego, La Jolla, CA,Department of Neurosciences, University of California, San Diego, La Jolla, CA,Correspondence:
| |
Collapse
|
45
|
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
|
46
|
Weber-Adrian D, Kofoed RH, Silburt J, Noroozian Z, Shah K, Burgess A, Rideout S, Kügler S, Hynynen K, Aubert I. Systemic AAV6-synapsin-GFP administration results in lower liver biodistribution, compared to AAV1&2 and AAV9, with neuronal expression following ultrasound-mediated brain delivery. Sci Rep 2021; 11:1934. [PMID: 33479314 PMCID: PMC7820310 DOI: 10.1038/s41598-021-81046-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/20/2020] [Indexed: 02/06/2023] Open
Abstract
Non-surgical gene delivery to the brain can be achieved following intravenous injection of viral vectors coupled with transcranial MRI-guided focused ultrasound (MRIgFUS) to temporarily and locally permeabilize the blood-brain barrier. Vector and promoter selection can provide neuronal expression in the brain, while limiting biodistribution and expression in peripheral organs. To date, the biodistribution of adeno-associated viruses (AAVs) within peripheral organs had not been quantified following intravenous injection and MRIgFUS delivery to the brain. We evaluated the quantity of viral DNA from the serotypes AAV9, AAV6, and a mosaic AAV1&2, expressing green fluorescent protein (GFP) under the neuron-specific synapsin promoter (syn). AAVs were administered intravenously during MRIgFUS targeting to the striatum and hippocampus in mice. The syn promoter led to undetectable levels of GFP expression in peripheral organs. In the liver, the biodistribution of AAV9 and AAV1&2 was 12.9- and 4.4-fold higher, respectively, compared to AAV6. The percentage of GFP-positive neurons in the FUS-targeted areas of the brain was comparable for AAV6-syn-GFP and AAV1&2-syn-GFP. In summary, MRIgFUS-mediated gene delivery with AAV6-syn-GFP had lower off-target biodistribution in the liver compared to AAV9 and AAV1&2, while providing neuronal GFP expression in the striatum and hippocampus.
Collapse
Affiliation(s)
- Danielle Weber-Adrian
- grid.410356.50000 0004 1936 8331Present Address: Faculty of Health Sciences, School of Medicine, Queen′s University, Kingston, ON Canada ,grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Rikke Hahn Kofoed
- grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Joseph Silburt
- grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Zeinab Noroozian
- grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Kairavi Shah
- grid.17063.330000 0001 2157 2938Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Alison Burgess
- grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, ON Canada
| | - Shawna Rideout
- grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, ON Canada
| | - Sebastian Kügler
- grid.411984.10000 0001 0482 5331Department of Neurology, Center Nanoscale Microscopy and Physiology of the Brain (CNMPB) at University Medical Center Göttingen, Göttingen, Germany
| | - Kullervo Hynynen
- grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Isabelle Aubert
- grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| |
Collapse
|
47
|
Jarrin S, Hakami A, Newland B, Dowd E. Growth Factor Therapy for Parkinson's Disease: Alternative Delivery Systems. JOURNAL OF PARKINSON'S DISEASE 2021; 11:S229-S236. [PMID: 33896851 PMCID: PMC8543245 DOI: 10.3233/jpd-212662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 12/30/2022]
Abstract
Despite decades of research and billions in global investment, there remains no preventative or curative treatment for any neurodegenerative condition, including Parkinson's disease (PD). Arguably, the most promising approach for neuroprotection and neurorestoration in PD is using growth factors which can promote the growth and survival of degenerating neurons. However, although neurotrophin therapy may seem like the ideal approach for neurodegenerative disease, the use of growth factors as drugs presents major challenges because of their protein structure which creates serious hurdles related to accessing the brain and specific targeting of affected brain regions. To address these challenges, several different delivery systems have been developed, and two major approaches-direct infusion of the growth factor protein into the target brain region and in vivo gene therapy-have progressed to clinical trials in patients with PD. In addition to these clinically evaluated approaches, a range of other delivery methods are in various degrees of development, each with their own unique potential. This review will give a short overview of some of these alternative delivery systems, with a focus on ex vivo gene therapy and biomaterial-aided protein and gene delivery, and will provide some perspectives on their potential for clinical development and translation.
Collapse
Affiliation(s)
- Sarah Jarrin
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Abrar Hakami
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Eilís Dowd
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| |
Collapse
|
48
|
Richardson RM, Bankiewicz KS, Christine CW, Van Laar AD, Gross RE, Lonser R, Factor SA, Kostyk SK, Kells AP, Ravina B, Larson PS. Data-driven evolution of neurosurgical gene therapy delivery in Parkinson's disease. J Neurol Neurosurg Psychiatry 2020; 91:1210-1218. [PMID: 32732384 PMCID: PMC7569395 DOI: 10.1136/jnnp-2020-322904] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/28/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
Loss of nigrostriatal dopaminergic projection neurons is a key pathology in Parkinson's disease, leading to abnormal function of basal ganglia motor circuits and the accompanying characteristic motor features. A number of intraparenchymally delivered gene therapies designed to modify underlying disease and/or improve clinical symptoms have shown promise in preclinical studies and subsequently were evaluated in clinical trials. Here we review the challenges with surgical delivery of gene therapy vectors that limited therapeutic outcomes in these trials, particularly the lack of real-time monitoring of vector administration. These challenges have recently been addressed during the evolution of novel techniques for vector delivery that include the use of intraoperative MRI. The preclinical development of these techniques are described in relation to recent clinical translation in an adeno-associated virus serotype 2-mediated human aromatic L-amino acid decarboxylase gene therapy development programme. This new paradigm allows visualisation of the accuracy and adequacy of viral vector delivery within target structures, enabling intertrial modifications in surgical approaches, cannula design, vector volumes and dosing. The rapid, data-driven evolution of these procedures is unique and has led to improved vector delivery.
Collapse
Affiliation(s)
- R Mark Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA .,Harvard Medical School, Boston, Massachusetts, USA
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA.,Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Chadwick W Christine
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Amber D Van Laar
- Department of Neurology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Brain Neurotherapy Bio, Inc, Columbus, Ohio, USA
| | - Robert E Gross
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA.,Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Russell Lonser
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Stewart A Factor
- Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Sandra K Kostyk
- Departments of Neuroscience and Neurology, Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | - Bernard Ravina
- Praxis Precision Medicines, Inc, Cambridge, Massachusetts, USA
| | - Paul S Larson
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
49
|
Sidorova YA, Saarma M. Can Growth Factors Cure Parkinson's Disease? Trends Pharmacol Sci 2020; 41:909-922. [PMID: 33198924 DOI: 10.1016/j.tips.2020.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 01/03/2023]
Abstract
Growth factors (GFs) hold considerable promise for disease modification in neurodegenerative disorders because they can protect and restore degenerating neurons and also enhance their functional activity. However, extensive efforts applied to utilize their therapeutic potential in humans have achieved limited success so far. Multiple clinical trials with GFs were performed in Parkinson's disease (PD) patients, in whom diagnostic symptoms of the disease are caused by advanced degeneration of nigrostriatal dopamine neurons (DNs), but the results of these trials are controversial. This review discusses recent developments in the field of therapeutic use of GFs, problems and obstacles related to this use, suggests the ways to overcome these issues, and alternative approaches that can be used to utilize the potential ofGFsin PD management.
Collapse
Affiliation(s)
- Yulia A Sidorova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
50
|
Buttery PC, Barker RA. Gene and Cell-Based Therapies for Parkinson's Disease: Where Are We? Neurotherapeutics 2020; 17:1539-1562. [PMID: 33128174 PMCID: PMC7598241 DOI: 10.1007/s13311-020-00940-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that carries large health and socioeconomic burdens. Current therapies for PD are ultimately inadequate, both in terms of symptom control and in modification of disease progression. Deep brain stimulation and infusion therapies are the current mainstay for treatment of motor complications of advanced disease, but these have very significant drawbacks and offer no element of disease modification. In fact, there are currently no agents that are established to modify the course of the disease in clinical use for PD. Gene and cell therapies for PD are now being trialled in the clinic. These treatments are diverse and may have a range of niches in the management of PD. They hold great promise for improved treatment of symptoms as well as possibly slowing progression of the disease in the right patient group. Here, we review the current state of the art for these therapies and look to future strategies in this fast-moving field.
Collapse
Affiliation(s)
- Philip C Buttery
- Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, CB2 0XY, Cambridge, UK.
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Hills Road, CB2 0QQ, Cambridge, UK.
| | - Roger A Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Hills Road, CB2 0QQ, Cambridge, UK.
- John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, CB2 0PY, Cambridge, UK.
| |
Collapse
|