1
|
Furusho T, Das R, Hakui H, Sairavi A, Adachi K, Galbraith-Liss MS, Rajagopal P, Horikawa M, Luo S, Li L, Yamada K, Andeen N, Dissen GA, Nakai H. Enhancing gene transfer to renal tubules and podocytes by context-dependent selection of AAV capsids. Nat Commun 2024; 15:10728. [PMID: 39737896 PMCID: PMC11685967 DOI: 10.1038/s41467-024-54475-9] [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: 01/17/2024] [Accepted: 11/13/2024] [Indexed: 01/01/2025] Open
Abstract
AAV vectors show promise for gene therapy; however, kidney gene transfer remains challenging. Here we conduct a barcode-seq-based comparison of 47 AAV capsids administered through different routes in mice, followed by individual validation. We find that local delivery of AAV-KP1, but not AAV9, via the renal vein or pelvis effectively transduces proximal tubules with minimal off-target liver transduction, while systemic AAV9, but not AAV-KP1, enhances proximal tubule and podocyte transduction in chronic kidney disease. We demonstrate that these contrasting observations are partly due to differences in their pharmacokinetics. Importantly, we show that renal pelvis injection overcomes pre-existing immunity, leading to robust and exclusive proximal tubule transduction, in non-human primates (NHPs). In addition, we highlight drastic differences in renal transduction profiles between mice and NHPs. Thus, this study provides mechanistic insights and underscores importance of context-dependent selection of AAV capsids to overcome challenges in gene delivery to the kidney.
Collapse
Affiliation(s)
- Taisuke Furusho
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Ranjan Das
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Hideyuki Hakui
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Anusha Sairavi
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Kei Adachi
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Mia S Galbraith-Liss
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Pratheppa Rajagopal
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Masahiro Horikawa
- Dotter Department of Interventional Radiology, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Shuhua Luo
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Lena Li
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Kentaro Yamada
- Dotter Department of Interventional Radiology, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Nicole Andeen
- Department of Pathology, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Gregory A Dissen
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
- Molecular Virology Core, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Hiroyuki Nakai
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA.
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA.
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University School of Medicine, Portland, OR, USA.
| |
Collapse
|
2
|
Guibinga GH, Do J, Chu B, Gu Y, Kikkawa R, Li X, Ozsolak F, MacLachlan T. Comparative assessment of the transduction efficiency and safety associated with the delivery of AAV9-GFP vector via lumbar puncture to cynomolgus macaques with and without anti-AAV9 pre-existing antibodies. Mol Ther Methods Clin Dev 2024; 32:101371. [PMID: 39717225 PMCID: PMC11664412 DOI: 10.1016/j.omtm.2024.101371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024]
Abstract
Administration of AAV-based gene therapies into the intra-cerebrospinal fluid (CSF) compartments via routes such as lumbar puncture (LP) has been implemented as an alternative to intravenous dosing to target the CNS regions. This route enables lower doses, decreases systemic toxicity, and circumvents intravascular pre-existing anti-AAV antibodies. In this study, AAV9-GFP vectors were administered via LP to juvenile cynomolgus macaques with and without pre-existing serum anti-AAV9 antibodies at a 5.0 × 1013 vector genomes per mL (vg/mL) dose and examined for 28 days. CNS and peripheral tissues were surveyed for vector genome, mRNA, and protein expression. Histopathology, clinical pathology, and humoral immune response to the viral capsid and transgene were also assessed. In addition, serum and CSF samples were analyzed to examine 276 proteomic markers curated to evaluate neural injury, organ damage, and inflammatory response. This study reveals no noticeable difference in AAV9-mediated gene transfer in the CNS tissues in the two groups; however, differences were observed for endpoints such as liver enzyme activities, histopathology, and levels of protein markers in the serum and CSF. These findings provide a view into vector transduction efficiency and safety following LP-delivered AAV9 to juvenile cynomolgus macaques with and without pre-existing anti-AAV9 antibodies.
Collapse
Affiliation(s)
- Ghiabe H. Guibinga
- Novartis Gene Therapies, San Diego, CA, USA
- Biologics Research Center (BRC), Novartis Biomedical Research, San Diego, CA, USA
| | - Janet Do
- Novartis Gene Therapies, San Diego, CA, USA
| | - Binh Chu
- Novartis Gene Therapies, San Diego, CA, USA
| | - Yin Gu
- Novartis Gene Therapies, San Diego, CA, USA
- Biologics Research Center (BRC), Novartis Biomedical Research, San Diego, CA, USA
| | - Rie Kikkawa
- Preclinical Safety (PCS), Novartis Biomedical Research, East Hanover, NJ, USA
| | - Xiaoguang Li
- Biologics Research Center (BRC), Novartis Biomedical Research, San Diego, CA, USA
| | - Fatih Ozsolak
- Novartis Gene Therapies, San Diego, CA, USA
- Biologics Research Center (BRC), Novartis Biomedical Research, San Diego, CA, USA
| | - Timothy MacLachlan
- Preclinical Safety (PCS), Novartis Biomedical Research, Cambridge, MA, USA
| |
Collapse
|
3
|
Wiggins KB, Winston SM, Reeves IL, Gaevert J, Spence Y, Brimble MA, Livingston B, Morton CL, Thomas PG, Sant AJ, Ross TM, Davidoff AM, Schultz-Cherry S. rAAV expressing a COBRA-designed influenza hemagglutinin generates a protective and durable adaptive immune response with a single dose. J Virol 2024; 98:e0078124. [PMID: 39078191 PMCID: PMC11338075 DOI: 10.1128/jvi.00781-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 07/31/2024] Open
Abstract
Influenza remains a worldwide public health threat. Although seasonal influenza vaccines are currently the best means of preventing severe disease, the standard-of-care vaccines require frequent updating due to antigenic drift and can have low efficacy, particularly in vulnerable populations. Here, we demonstrate that a single administration of a recombinant adenovirus-associated virus (rAAV) vector expressing a computationally optimized broadly reactive antigen (COBRA)-derived influenza H1 hemagglutinin (HA) induces strongly neutralizing and broadly protective antibodies in naïve mice and ferrets with pre-existing influenza immunity. Following a lethal viral challenge, the rAAV-COBRA vaccine allowed for significantly reduced viral loads in the upper and lower respiratory tracts and complete protection from morbidity and mortality that lasted for at least 5 months post-vaccination. We observed no signs of antibody waning during this study. CpG motif enrichment of the antigen can act as an internal adjuvant to further enhance the immune responses to allow for lower vaccine dosages with the induction of unique interferon-producing CD4+ and CD8+ T cells specific to HA head and stem peptide sequences. Our studies highlight the utility of rAAV as an effective platform to improve seasonal influenza vaccines. IMPORTANCE Developing an improved seasonal influenza vaccine remains an ambitious goal of researchers and clinicians alike. With influenza routinely causing severe epidemics with the potential to rise to pandemic levels, it is critical to create an effective, broadly protective, and durable vaccine to improve public health worldwide. As a potential solution, we created a rAAV viral vector expressing a COBRA-optimized influenza hemagglutinin antigen with modestly enriched CpG motifs to evoke a robust and long-lasting immune response after a single intramuscular dose without needing boosts or adjuvants. Importantly, the rAAV vaccine boosted antibody breadth to future strains in ferrets with pre-existing influenza immunity. Together, our data support further investigation into the utility of viral vectors as a potential avenue to improve our seasonal influenza vaccines.
Collapse
Affiliation(s)
- Kristin B. Wiggins
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Stephen M. Winston
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Isaiah L. Reeves
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Jessica Gaevert
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Yunyu Spence
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Mark A. Brimble
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Brandi Livingston
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Christopher L. Morton
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Paul G. Thomas
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Andrea J. Sant
- David H. Smith Center
for Vaccine Biology and Immunology, Department of Microbiology and
Immunology, University of Rochester Medical
Center, Rochester, New
York, USA
| | - Ted M. Ross
- Department of
Infectious Biology, Cleveland Clinic,
Cleveland, Ohio, USA
- Cleveland Clinic,
Florida Research and Innovation Center,
Port St. Lucie, Florida,
USA
| | - Andrew M. Davidoff
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Stacey Schultz-Cherry
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| |
Collapse
|
4
|
Werner MS, Aras S, Morgan AR, Roamer J, Param NJ, Olagbegi K, Lamontagne RJ, Greig JA, Wilson JM. Adeno-associated virus-mediated trastuzumab delivery to the central nervous system for human epidermal growth factor receptor 2+ brain metastasis. Cancer Gene Ther 2024; 31:766-777. [PMID: 38480976 DOI: 10.1038/s41417-024-00751-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 05/19/2024]
Abstract
Trastuzumab improves overall survival for HER2+ breast cancer, but its short half-life in the cerebrospinal fluid (~2-4 days) and delivery limitations restrict the ability to target HER2+ central nervous system (CNS) disease. We developed an adeno-associated virus (AAV) vector expressing a codon-optimized, ubiquitin C (UbC)-promoter-driven trastuzumab sequence (AAV9.UbC.trastuzumab) for intrathecal administration. Transgene expression was evaluated in adult Rag1 knockout mice and rhesus nonhuman primates (NHPs) after a single intracerebroventricular (ICV) or intra-cisterna magna (ICM) AAV9.UbC.trastuzumab injection, respectively, using real-time PCR, ELISA, Western blot, in situ hybridization, single-nucleus RNA sequencing, and liquid chromatography-mass spectrometry; antitumor efficacy was evaluated in brain xenografts using HER2+ breast cancer cell lines (BT-474, MDA-MB-453). Transgene expression was detected in brain homogenates of Rag1 knockout mice following a single ICV injection of AAV9.UbC.trastuzumab (1 × 1011 vector genome copies [GC]/mouse) and tumor progression was inhibited in xenograft models of breast-to-brain metastasis. In NHPs, ICM delivery of AAV9.UbC.trastuzumab (3 × 1013 GC/animal) was well tolerated (36-37 days in-life) and resulted in transgene expression in CNS tissues and cerebrospinal fluid at levels sufficient to induce complete tumor remission in MDA-MB-453 brain xenografts. With AAV9's proven clinical safety record, this gene therapy may represent a viable approach for targeting HER2 + CNS malignancies.
Collapse
Affiliation(s)
- Marcela S Werner
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shweta Aras
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ashleigh R Morgan
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jillian Roamer
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nesteene J Param
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kanyin Olagbegi
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - R Jason Lamontagne
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jenny A Greig
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
5
|
Wang JH, Gessler DJ, Zhan W, Gallagher TL, Gao G. Adeno-associated virus as a delivery vector for gene therapy of human diseases. Signal Transduct Target Ther 2024; 9:78. [PMID: 38565561 PMCID: PMC10987683 DOI: 10.1038/s41392-024-01780-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Adeno-associated virus (AAV) has emerged as a pivotal delivery tool in clinical gene therapy owing to its minimal pathogenicity and ability to establish long-term gene expression in different tissues. Recombinant AAV (rAAV) has been engineered for enhanced specificity and developed as a tool for treating various diseases. However, as rAAV is being more widely used as a therapy, the increased demand has created challenges for the existing manufacturing methods. Seven rAAV-based gene therapy products have received regulatory approval, but there continue to be concerns about safely using high-dose viral therapies in humans, including immune responses and adverse effects such as genotoxicity, hepatotoxicity, thrombotic microangiopathy, and neurotoxicity. In this review, we explore AAV biology with an emphasis on current vector engineering strategies and manufacturing technologies. We discuss how rAAVs are being employed in ongoing clinical trials for ocular, neurological, metabolic, hematological, neuromuscular, and cardiovascular diseases as well as cancers. We outline immune responses triggered by rAAV, address associated side effects, and discuss strategies to mitigate these reactions. We hope that discussing recent advancements and current challenges in the field will be a helpful guide for researchers and clinicians navigating the ever-evolving landscape of rAAV-based gene therapy.
Collapse
Affiliation(s)
- Jiang-Hui Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurological Surgery, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Thomas L Gallagher
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
6
|
Zhu M, Lan Z, Park J, Gong S, Wang Y, Guo F. Regulation of CNS pathology by Serpina3n/SERPINA3: The knowns and the puzzles. Neuropathol Appl Neurobiol 2024; 50:e12980. [PMID: 38647003 PMCID: PMC11131959 DOI: 10.1111/nan.12980] [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: 01/09/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024]
Abstract
Neuroinflammation, blood-brain barrier (BBB) dysfunction, neuron and glia injury/death and myelin damage are common central nervous system (CNS) pathologies observed in various neurological diseases and injuries. Serine protease inhibitor (Serpin) clade A member 3n (Serpina3n), and its human orthologue SERPINA3, is an acute-phase inflammatory glycoprotein secreted primarily by the liver into the bloodstream in response to systemic inflammation. Clinically, SERPINA3 is dysregulated in brain cells, cerebrospinal fluid and plasma in various neurological conditions. Although it has been widely accepted that Serpina3n/SERPINA3 is a reliable biomarker of reactive astrocytes in diseased CNS, recent data have challenged this well-cited concept, suggesting instead that oligodendrocytes and neurons are the primary sources of Serpina3n/SERPINA3. The debate continues regarding whether Serpina3n/SERPINA3 induction represents a pathogenic or a protective mechanism. Here, we propose possible interpretations for previously controversial data and present perspectives regarding the potential role of Serpina3n/SERPINA3 in CNS pathologies, including demyelinating disorders where oligodendrocytes are the primary targets. We hypothesise that the 'good' or 'bad' aspects of Serpina3n/SERPINA3 depend on its cellular sources, its subcellular distribution (or mis-localisation) and/or disease/injury types. Furthermore, circulating Serpina3n/SERPINA3 may cross the BBB to impact CNS pathologies. Cell-specific genetic tools are critically important to tease out the potential roles of cell type-dependent Serpina3n in CNS diseases/injuries.
Collapse
Affiliation(s)
- Meina Zhu
- Department of Neurology, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, California, USA
| | - Zhaohui Lan
- Center for Brain Health and Brain Technology, Global Institute of Future Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Joohyun Park
- Department of Neurology, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, California, USA
| | | | - Yan Wang
- Department of Neurology, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, California, USA
| | - Fuzheng Guo
- Department of Neurology, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, California, USA
| |
Collapse
|
7
|
Perera A, Brock O, Ahmed A, Shaw C, Ashkan K. Taking the knife to neurodegeneration: a review of surgical gene therapy delivery to the CNS. Acta Neurochir (Wien) 2024; 166:136. [PMID: 38483631 PMCID: PMC10940433 DOI: 10.1007/s00701-024-06028-8] [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: 01/11/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024]
Abstract
Gene supplementation and editing for neurodegenerative disorders has emerged in recent years as the understanding of the genetic mechanisms underlying several neurodegenerative disorders increases. The most common medium to deliver genetic material to cells is via viral vectors; and with respect to the central nervous system, adeno-associated viral (AAV) vectors are a popular choice. The most successful example of AAV-based gene therapy for neurodegenerative disorders is Zolgensma© which is a transformative intravenous therapy given to babies with spinal muscular atrophy. However, the field has stalled in achieving safe drug delivery to the central nervous system in adults for which treatments for disorders such as amyotrophic lateral sclerosis are desperately needed. Surgical gene therapy delivery has been proposed as a potential solution to this problem. While the field of the so-called regenerative neurosurgery has yielded pre-clinical optimism, several challenges have emerged. This review seeks to explore the field of regenerative neurosurgery with respect to AAV-based gene therapy for neurodegenerative diseases, its progress so far and the challenges that need to be overcome.
Collapse
Affiliation(s)
- Andrea Perera
- Maurice Wohl Institute of Neuroscience, Department of Basic Clinical Neuroscience, King's College London, Cutcombe Road, Denmark Hill, London, SE5 9RS, UK.
- Department of Neurosurgery, King's College Hospital NHS Trust, London, UK.
| | - Olivier Brock
- Maurice Wohl Institute of Neuroscience, Department of Basic Clinical Neuroscience, King's College London, Cutcombe Road, Denmark Hill, London, SE5 9RS, UK
| | - Aminul Ahmed
- Department of Neurosurgery, King's College Hospital NHS Trust, London, UK
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Chris Shaw
- Maurice Wohl Institute of Neuroscience, Department of Basic Clinical Neuroscience, King's College London, Cutcombe Road, Denmark Hill, London, SE5 9RS, UK
- Centre for Brain Research, University of Auckland, 85 Park Road Grafton, Auckland, 1023, New Zealand
| | - Keyoumars Ashkan
- Maurice Wohl Institute of Neuroscience, Department of Basic Clinical Neuroscience, King's College London, Cutcombe Road, Denmark Hill, London, SE5 9RS, UK
- Department of Neurosurgery, King's College Hospital NHS Trust, London, UK
| |
Collapse
|