1
|
Meng JL, Dong ZX, Chen YR, Lin MH, Liu YC, Roffler SR, Lin WW, Chang CY, Tzou SC, Cheng TL, Huang HC, Li ZQ, Lin YC, Su YC. pH-Responsive Polyethylene Glycol Engagers for Enhanced Brain Delivery of PEGylated Nanomedicine to Treat Glioblastoma. ACS NANO 2025. [PMID: 39749925 DOI: 10.1021/acsnano.4c05906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The blood-brain barrier (BBB) remains a major obstacle for effective delivery of therapeutics to treat central nervous system (CNS) disorders. Although transferrin receptor (TfR)-mediated transcytosis is widely employed for brain drug delivery, the inefficient release of therapeutic payload hinders their efficacy from crossing the BBB. Here, we developed a pH-responsive anti-polyethylene glycol (PEG) × anti-TfR bispecific antibody (pH-PEG engagerTfR) that can complex with PEGylated nanomedicine at physiological pH to trigger TfR-mediated transcytosis in the brain microvascular endothelial cells, while rapidly dissociating from PEGylated nanomedicine at acidic endosomes for efficient release of PEGylated nanomedicine to cross the BBB. The pH-PEG engagerTfR significantly increased the accumulation of PEGylated nanomedicine in the mouse brain compared to wild-type PEG engagerTfR (WT-PEG engagerTfR). pH-PEG engagerTfR-decorated PEGylated liposomal doxorubicin exhibited an enhanced antitumor effect and extended survival in a human glioblastoma (GBM) orthotopic xenograft mice model. Conditional release of PEGylated nanomedicine during BBB-related receptor-mediated transcytosis by pH-PEG engagerTfR is promising for enhanced brain drug delivery to treat CNS disorders.
Collapse
Affiliation(s)
- Jun-Lun Meng
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Zi-Xuan Dong
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yan-Ru Chen
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Meng-Hsuan Lin
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yu-Ching Liu
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wen-Wei Lin
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chin-Yuan Chang
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Shey-Cherng Tzou
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department of Biomedical Science and Environmental Biology, Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hsiao-Chen Huang
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Zhi-Qin Li
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yen-Cheng Lin
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yu-Cheng Su
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department of Biomedical Science and Environmental Biology, Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
2
|
Liu Y, Wang Y, Kulkarni RA, Wegiel LA, Lee B, Bedingfield SK, Weitz D. Syringable Microcapsules for Sustained, Localized, and Controllable siRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39705128 DOI: 10.1021/acsami.4c12805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
The clinical use of small interfering RNA (siRNA) and antisense oligonucleotides often requires invasive routes of administration, including intrathecal or intraocular injection. Additionally, these treatments often necessitate repeated injections. While nanoparticle formulation and chemical modifications have extended siRNA therapeutic durability, challenges persist, such as the side effects of bolus injections with high toxicity and maximum exposure in the acute phase. We present a microcapsule-based method to extend the activity of cholesterol-conjugated siRNA locally. Using microfluidics, microcapsules with well-defined size distribution and shell thickness are fabricated with poly(lactic-co-glycolic acid) (PLGA) with varying molecular weights and compositions. The microcapsules show a remarkably high drug encapsulation efficiency of nearly 100% and a high loading capacity (8900 μg siRNA/1 mg polymer). Additionally, microcapsules with an average diameter of 40 μm show superior syringeability when tested with needles ranging from gauge sizes of 27 to 32 G. This makes them suitable for various injection routes. Two sustained-release formulations were selected based on a 3-month in vitro release test. Subsequently, these formulations were injected subcutaneously into mice to verify their in vivo release profiles. The findings demonstrate that the microcapsules effectively shield the siRNAs from being cleared and enable them to be released constantly over 3 months. In contrast, unencapsulated siRNAs are rapidly cleared.
Collapse
Affiliation(s)
- Yan Liu
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, Massachusetts 02138, United States
| | - Yang Wang
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, Massachusetts 02138, United States
| | | | | | - Byungkook Lee
- Eli Lilly & Company, Indianapolis, Indiana 46285, United States
| | | | - David Weitz
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, Massachusetts 02138, United States
- Department of Physics, Harvard University, Cambridge, Massachusetts 02138, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
3
|
Kang JH, Yang JK, Cho KH, Lee OH, Kwon H, Kim SY, Kim S, Ko YT. Intracalvariosseous administration of donepezil microspheres protects against cognitive impairment by virtue of long-lasting brain exposure in mice. Theranostics 2024; 14:6708-6725. [PMID: 39479440 PMCID: PMC11519799 DOI: 10.7150/thno.100986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Rationale: Recent studies have demonstrated the direct connections between the skull bone marrow, meninges, and brain. In an effort to explore these connections for the purpose of brain drug delivery, we previously proposed the direct application of CNS drugs into the diploic space between the outer and inner cortex of the skull, namely, intracalvariosseous administration (ICO). It was successfully demonstrated that small molecular to large colloidal drugs can readily reach the brain after ICO in mice and rabbits. Here, we report that a single ICO of donepezil microspheres protects cognitive impairment in Alzheimer mouse models over a month-long period. Methods: Donepezil-loaded long-acting microspheres (DPZ@LAM) were prepared with biodegradable poly(DL-lactide-co-glycolide) (PLGA). Pharmacokinetic study and behavioral test were performed to determine the brain exposure and therapeutic effects after ICO of DPZ@LAM in scopolamine-induced memory-deficient mice. Results: DPZ@LAM were capable of a month-long and precisely controlled drug release. After a single ICO of DPZ@LAM, DPZ concentration in brain sustained above the effective therapeutic levels for four weeks. The long-lasting brain exposure also led to significantly recovered cognitive function in scopolamine-induced memory-deficient mice, along with decreased acetylcholinesterase activity and increased brain-derived neurotrophic factor. Conclusions: ICO allows for BBB-bypassing brain drug delivery through the direct connection between the skull bone marrow and brain, providing an alternative approach for the treatment of neurodegenerative diseases with otherwise BBB impermeable CNS drugs.
Collapse
Affiliation(s)
- Ji Hee Kang
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Jin-Kyoung Yang
- Department of Chemical Engineering, Dong-Eui University, Busan, 47340, Republic of Korea
| | - Kyo Hee Cho
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - O Hyun Lee
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Hayoon Kwon
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Sehoon Kim
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Young Tag Ko
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| |
Collapse
|
4
|
Benatti HR, Anagnostakou V, Taghian T, Hall EF, Nath S, Heilman CB, Beneduce BM, Leporati A, Raskett C, Epshtein M, King R, Gounis MJ, Malek AM, Gray-Edwards HL. A minimally invasive endovascular approach to the cerebellopontine angle cistern enables broad CNS biodistribution of scAAV9-CB-GFP. Mol Ther 2024; 32:3346-3355. [PMID: 39192584 PMCID: PMC11489529 DOI: 10.1016/j.ymthe.2024.08.024] [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: 11/29/2023] [Revised: 07/31/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
Neurological disorders pose a challenge for targeted therapy due to restricted access of therapeutic agents to the central nervous system (CNS). Current methods are limited by procedure-related risks, invasiveness, and insufficient CNS biodistribution. A novel percutaneous transvenous technology, currently in clinical trials for communicating hydrocephalus, offers a minimally invasive approach by providing endovascular access to the cerebrospinal fluid-filled cerebellopontine angle (CPA) cistern. We hypothesized that drug delivery to the CPA cistern could yield widespread CNS distribution. Using an ovine model, we compared the biodistribution of scAAV9-CB-GFP following CPA cistern infusion with previously reported cisterna magna (CM) administration. Targeting both the CPA cistern and CM in sheep, we employed a lumbar spine-inserted microcatheter under fluoroscopy. CPA delivery of AAV9 demonstrated biodistribution and transduction in the cerebral cortices, striatum, thalamus, midbrain, cerebellum, and spinal cord, with minor liver distribution comparable to CM. The favorable safety profile in humans with hydrocephalus suggests that percutaneous endovascular injection into the CPA could offer a clinically safer and minimally invasive delivery system for CNS gene and cell-based therapies.
Collapse
Affiliation(s)
- Hector Ribeiro Benatti
- Horae Gene Therapy Center, UMass Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Vania Anagnostakou
- New England Center for Stroke Research, Department of Radiology, UMass Chan Medical School, 55 N Lake Avenue, Worcester, MA 01655, USA
| | - Toloo Taghian
- Horae Gene Therapy Center, UMass Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA; New England Center for Stroke Research, Department of Radiology, UMass Chan Medical School, 55 N Lake Avenue, Worcester, MA 01655, USA
| | - Erin F Hall
- Horae Gene Therapy Center, UMass Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Sarah Nath
- Horae Gene Therapy Center, UMass Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Carl B Heilman
- Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | | | - Anita Leporati
- New England Center for Stroke Research, Department of Radiology, UMass Chan Medical School, 55 N Lake Avenue, Worcester, MA 01655, USA
| | - Christopher Raskett
- New England Center for Stroke Research, Department of Radiology, UMass Chan Medical School, 55 N Lake Avenue, Worcester, MA 01655, USA
| | - Mark Epshtein
- New England Center for Stroke Research, Department of Radiology, UMass Chan Medical School, 55 N Lake Avenue, Worcester, MA 01655, USA
| | - Robert King
- New England Center for Stroke Research, Department of Radiology, UMass Chan Medical School, 55 N Lake Avenue, Worcester, MA 01655, USA
| | - Matthew J Gounis
- New England Center for Stroke Research, Department of Radiology, UMass Chan Medical School, 55 N Lake Avenue, Worcester, MA 01655, USA
| | - Adel M Malek
- Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA.
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, UMass Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA; New England Center for Stroke Research, Department of Radiology, UMass Chan Medical School, 55 N Lake Avenue, Worcester, MA 01655, USA.
| |
Collapse
|
5
|
Lin L, Huang L, Huang S, Chen W, Huang H, Chi L, Su F, Liu X, Yuan K, Jiang Q, Li C, Smith WW, Fu Q, Pei Z. MSC-Derived Extracellular Vesicles Alleviate NLRP3/GSDMD-Mediated Neuroinflammation in Mouse Model of Sporadic Alzheimer's Disease. Mol Neurobiol 2024; 61:5494-5509. [PMID: 38200351 DOI: 10.1007/s12035-024-03914-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, with sporadic form being the predominant type. Neuroinflammation plays a critical role in accelerating pathogenic processes in AD. Mesenchymal stem cell (MSC)-derived small extracellular vesicles (MSC-sEVs) regulate inflammatory responses and show great promise for treating AD. Induced pluripotent stem cell (iPSC)-derived MSCs are similar to MSCs and exhibit low immunogenicity and heterogeneity, making them promising cell sources for clinical applications. This study examined the anti-inflammatory effects of MSC-sEVs in a streptozotocin-induced sporadic mouse model of AD (sAD). The intracisternal administration of iPSC-MSC-sEVs alleviated NLRP3/GSDMD-mediated neuroinflammation, decreased amyloid deposition and neuronal apoptosis, and mitigated cognitive dysfunction. Furthermore, it explored the role of miR-223-3p in the iPSC-MSC-sEVs-mediated anti-inflammatory effects in vitro. miR-223-3p directly targeted NLRP3, whereas inhibiting miR-223-3p almost completely reversed the suppression of NLRP3 by MSC-sEVs, suggesting that miR-223-3p may, at least partially, account for MSC-sEVs-mediated anti-inflammation. Results obtained suggest that intracisternal administration of iPSC-MSC-sEVs can reduce cognitive impairment by inhibiting NLRP3/GSDMD neuroinflammation in a sAD mouse model. Therefore, the present study provides a proof-of-principle for applying iPSC-MSC-sEVs to target neuroinflammation in sAD.
Collapse
Affiliation(s)
- Lishan Lin
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Longxin Huang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Sen Huang
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Weineng Chen
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Heng Huang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Li Chi
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Fengjuan Su
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiaoqing Liu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Kang Yuan
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Qiuhong Jiang
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Changu Li
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Wanli W Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Qingling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
6
|
Koller BH, Nguyen M, Snouwaert JN, Gabel CA, Ting JPY. Species-specific NLRP3 regulation and its role in CNS autoinflammatory diseases. Cell Rep 2024; 43:113852. [PMID: 38427558 DOI: 10.1016/j.celrep.2024.113852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
The NLRP3 inflammasome is essential for caspase-1 activation and the release of interleukin (IL)-1β, IL-18, and gasdermin-D in myeloid cells. However, research on species-specific NLRP3's physiological impact is limited. We engineer mice with the human NLRP3 gene, driven by either the human or mouse promoter, via syntenic replacement at the mouse Nlrp3 locus. Both promoters facilitate hNLRP3 expression in myeloid cells, but the mouse promoter responds more robustly to LPS. Investigating the disease impact of differential NLRP3 regulation, we introduce the D305N gain-of-function mutation into both humanized lines. Chronic inflammation is evident with both promoters; however, CNS outcomes vary significantly. Despite poor response to LPS, the human promoter results in D305N-associated aseptic meningitis, mirroring human pathology. The mouse promoter, although leading to increased CNS expression post-LPS, does not induce meningitis in D305N mutants. Therefore, human-like NLRP3 expression may be crucial for accurate modeling of its role in disease pathogenesis.
Collapse
Affiliation(s)
- Beverly H Koller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - MyTrang Nguyen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John N Snouwaert
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Jenny P-Y Ting
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
7
|
Salomonsson SE, Clelland CD. Building CRISPR Gene Therapies for the Central Nervous System: A Review. JAMA Neurol 2024; 81:283-290. [PMID: 38285472 PMCID: PMC11164426 DOI: 10.1001/jamaneurol.2023.4983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Importance Gene editing using clustered regularly interspaced short palindromic repeats (CRISPR) holds the promise to arrest or cure monogenic disease if it can be determined which genetic change to create without inducing unintended cellular dysfunction and how to deliver this technology to the target organ reliably and safely. Clinical trials for blood and liver disorders, for which delivery of CRISPR is not limiting, show promise, yet no trials have begun for central nervous system (CNS) indications. Observations The CNS is arguably the most challenging target given its innate exclusion of large molecules and its defenses against bacterial invasion (from which CRISPR originates). Herein, the types of CRISPR editing (DNA cutting, base editing, and templated repair) and how these are applied to different genetic variants are summarized. The challenges of delivering genome editors to the CNS, including the viral and nonviral delivery vehicles that may ultimately circumvent these challenges, are discussed. Also, ways to minimize the potential in vivo genotoxic effects of genome editors through delivery vehicle design and preclinical off-target testing are considered. The ethical considerations of germline editing, a potential off-target outcome of any gene editing therapy, are explored. The unique regulatory challenges of a human-specific therapy that cannot be derisked solely in animal models are also discussed. Conclusions and Relevance An understanding of both the potential benefits and challenges of CRISPR gene therapy better informs the scientific, clinical, regulatory, and timeline considerations of developing CRISPR gene therapy for neurologic diseases.
Collapse
Affiliation(s)
- Sally E Salomonsson
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco
- Department of Neurology, Memory and Aging Center, University of California, San Francisco
| | - Claire D Clelland
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco
- Department of Neurology, Memory and Aging Center, University of California, San Francisco
| |
Collapse
|
8
|
Rouse CJ, Jensen VN, Heldermon CD. Mucopolysaccharidosis type IIIB: a current review and exploration of the AAV therapy landscape. Neural Regen Res 2024; 19:355-359. [PMID: 37488890 PMCID: PMC10503619 DOI: 10.4103/1673-5374.377606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/29/2023] [Accepted: 04/24/2023] [Indexed: 07/26/2023] Open
Abstract
Mucopolysaccharidoses type IIIB is a rare genetic disorder caused by mutations in the gene that encodes for N-acetyl-alpha-glucosaminidase. This results in the aggregation of heparan sulfate polysaccharides within cell lysosomes that leads to progressive and severe debilitating neurological dysfunction. Current treatment options are expensive, limited, and presently there are no approved cures for mucopolysaccharidoses type IIIB. Adeno-associated virus gene therapy has significantly advanced the field forward, allowing researchers to successfully design, enhance, and improve potential cures. Our group recently published an effective treatment using a codon-optimized triple mutant adeno-associated virus 8 vector that restores N-acetyl-alpha-glucosaminidase levels, auditory function, and lifespan in the murine model for mucopolysaccharidoses type IIIB to that seen in healthy mice. Here, we review the current state of the field in relation to the capsid landscape, adeno-associated virus gene therapy and its successes and challenges in the clinic, and how novel adeno-associated virus capsid designs have evolved research in the mucopolysaccharidoses type IIIB field.
Collapse
|
9
|
Burgio F, Gaiser C, Brady K, Gatta V, Class R, Schrage R, Suter-Dick L. A Perfused In Vitro Human iPSC-Derived Blood-Brain Barrier Faithfully Mimics Transferrin Receptor-Mediated Transcytosis of Therapeutic Antibodies. Cell Mol Neurobiol 2023; 43:4173-4187. [PMID: 37698826 PMCID: PMC10661771 DOI: 10.1007/s10571-023-01404-x] [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: 04/24/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023]
Abstract
Delivering biologics to elicit a therapeutic response in the central nervous system (CNS) remains challenging due to the presence of the blood-brain barrier (BBB). Receptor-mediated transcytosis is a strategy to improve brain exposure after systemic drug administration. The availability of a clinically relevant in vitro BBB model is crucial to investigate transcytosis pathways and to predict the penetration of biologics into the CNS. We created a perfused human in vitro BBB model made of induced pluripotent stem cells (iPSC)-derived brain microvascular endothelial cells (BMEC) for studying transferrin receptor-mediated transcytosis. iPSC-derived BMEC were seeded in the top channel of a three-lane microfluidic device (OrganoPlate®). After 2 days in culture, the established cell model exhibited relevant BBB features, including physiological transendothelial electrical resistance in a transwell setting (1500 Ω*cm2), reduced apparent permeability (Papp) to the fluorescence tracer Lucifer yellow (20-fold less than cell-free chips), expression of key BBB markers such as tight junctions proteins, transporters, receptors and functional P-gp efflux pump. Moreover, the model exhibited functional transferrin receptor-mediated uptake and transcytosis. To assess selective transferrin receptor-mediated transcytosis, a mixture of anti-human transferrin receptor (MEM-189) and control (sheep IgG anti-bovine serum albumin) antibodies was perfused in the top channel for 2 h. The Papp of MEM-189 was 11-fold higher than that of the control antibody, demonstrating facilitated receptor-mediated transcytosis. Compared to published work reporting a 2-fold ratio, this result is remarkable and establishes the suitability of our model for exploring receptor-mediated transcytosis and screening of antibodies for putative brain shuttle application. A perfused in vitro human model made of iPSC-derived BMEC with the chief characteristics (barrier tightness, functionality) of the human BBB can be applied to study transferrin receptor (TfR)-mediated transcytosis of therapeutic antibodies. This may bring critical advances in drug shuttle technology. Graphical abstract generated with biorender.com.
Collapse
Affiliation(s)
- Floriana Burgio
- University of Applied Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland
| | - Carine Gaiser
- University of Applied Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland
| | - Kevin Brady
- Development Sciences, UCB Biopharma SRL, Braine L'Alleud, Belgium
| | - Viviana Gatta
- Neuroscience Therapeutic Area, UCB Biopharma SRL, Braine L'Alleud, Belgium
| | - Reiner Class
- Development Sciences, UCB Biopharma SRL, Braine L'Alleud, Belgium
| | - Ramona Schrage
- Neuroscience Therapeutic Area, UCB Biopharma SRL, Braine L'Alleud, Belgium
| | - Laura Suter-Dick
- University of Applied Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland.
| |
Collapse
|
10
|
Shi S, Ren H, Xie Y, Yu M, Chen Y, Yang L. Engineering advanced nanomedicines against central nervous system diseases. MATERIALS TODAY 2023; 69:355-392. [DOI: 10.1016/j.mattod.2023.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
11
|
Kaur A, Singh N, Kaur H, Kakoty V, Sharma DS, Khursheed R, Babu MR, Harish V, Gupta G, Gulati M, Kumar P, Dureja H, Alharthi NS, Khan FR, Rehman ZU, Hakami MA, Patel M, Patel R, Zandi M, Vishwas S, Dua K, Singh SK. Neurodegenerative diseases and brain delivery of therapeutics: Bridging the gap using dendrimers. J Drug Deliv Sci Technol 2023; 87:104868. [DOI: 10.1016/j.jddst.2023.104868] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Mitchell NL, Murray SJ, Wellby MP, Barrell GK, Russell KN, Deane AR, Wynyard JR, Palmer MJ, Pulickan A, Prendergast PM, Casy W, Gray SJ, Palmer DN. Long-term safety and dose escalation of intracerebroventricular CLN5 gene therapy in sheep supports clinical translation for CLN5 Batten disease. Front Genet 2023; 14:1212228. [PMID: 37614821 PMCID: PMC10442658 DOI: 10.3389/fgene.2023.1212228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023] Open
Abstract
CLN5 neuronal ceroid lipofuscinosis (NCL, Batten disease) is a rare, inherited fatal neurodegenerative disorder caused by mutations in the CLN5 gene. The disease is characterised by progressive neuronal loss, blindness, and premature death. There is no cure. This study evaluated the efficacy of intracerebroventricular (ICV) delivery of an adeno-associated viral vector encoding ovine CLN5 (scAAV9/oCLN5) in a naturally occurring sheep model of CLN5 disease. CLN5 affected (CLN5-/-) sheep received low, moderate, or high doses of scAAV9/oCLN5 at three disease stages. The treatment delayed disease progression, extended survival and slowed stereotypical brain atrophy in most animals. Of note, one high dose treated animal only developed mild disease symptomology and survived to 60.1 months of age, triple the natural life expectancy of an untreated CLN5-/- sheep. Eyesight was not preserved at any administration age or dosage. Histopathologic examination revealed that greater transduction efficiency was achieved through higher ICV doses, and this resulted in greater amelioration of disease pathology. Together with other pre-clinical data from CLN5-/- sheep, the safety and efficacy data from these investigational new drug (IND)-enabling studies supported the initiation of the first in-human CLN5 gene therapy clinical study using the ICV delivery route for the treatment of CLN5 NCL. Clinical Trial Registration: https://clinicaltrials.gov/, identifier NCT05228145.
Collapse
Affiliation(s)
- Nadia L. Mitchell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
- Department of Radiology, University of Otago, Christchurch, New Zealand
| | - Samantha J. Murray
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Martin P. Wellby
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Graham K. Barrell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Katharina N. Russell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Ashley R. Deane
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - John R. Wynyard
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Madeleine J. Palmer
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Anila Pulickan
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | | | - Widler Casy
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Steven J. Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David N. Palmer
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
- Department of Radiology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
13
|
Ajeeb R, Clegg JR. Intrathecal delivery of Macromolecules: Clinical status and emerging technologies. Adv Drug Deliv Rev 2023; 199:114949. [PMID: 37286086 DOI: 10.1016/j.addr.2023.114949] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/09/2023]
Abstract
The proximity and association of cerebrospinal fluid (CSF) and the intrathecal (IT) space with deep targets in the central nervous system (CNS) parenchyma makes IT injection an attractive route of administration for brain drug delivery. However, the extent to which intrathecally administered macromolecules are effective in treating neurological diseases is a question of both clinical debate and technological interest. We present the biological, chemical, and physical properties of the intrathecal space that are relevant to drug absorption, distribution, metabolism, and elimination from CSF. We then analyze the evolution of IT drug delivery in clinical trials over the last 20 years. Our analysis revealed that the percentage of clinical trials assessing IT delivery for the delivery of biologics (i.e., macromolecules, cells) for treatment of chronic conditions (e.g., neurodegeneration, cancer, and metabolic diseases) has steadily increased. Clinical trials exploring cell or macromolecular delivery within the IT space have not evaluated engineering technologies, such as depots, particles, or other delivery systems. Recent pre-clinical studies have evaluated IT macromolecule delivery in small animals, postulating that delivery efficacy can be assisted by external medical devices, micro- or nanoparticles, bulk biomaterials, and viral vectors. Further studies are necessary to evaluate the extent to which engineering technologies and IT administration improve CNS targeting and therapeutic outcome.
Collapse
Affiliation(s)
- Rana Ajeeb
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK, United States
| | - John R Clegg
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK, United States; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Institute for Biomedical Engineering, Science, and Technology, University of Oklahoma, Norman, OK, United States.
| |
Collapse
|
14
|
Duan S, Wang S, Qiao L, Yu X, Wang N, Chen L, Zhang X, Zhao X, Liu H, Wang T, Wu Y, Li N, Liu F. Oncolytic Virus-Driven Biotherapies from Bench to Bedside. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206948. [PMID: 36879416 DOI: 10.1002/smll.202206948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/17/2023] [Indexed: 06/08/2023]
Abstract
With advances in cancer biology and an ever-deepening understanding of molecular virology, oncolytic virus (OV)-driven therapies have developed rapidly and become a promising alternative to traditional cancer therapies. In recent years, satisfactory results for oncolytic virus therapy (OVT) are achieved at both the cellular and organismal levels, and efforts are being increasingly directed toward clinical trials. Unfortunately, OVT remains ineffective in these trials, especially when performed using only a single OV reagent. In contrast, integrated approaches, such as using immunotherapy, chemotherapy, or radiotherapy, alongside OVT have demonstrated considerable efficacy. The challenges of OVT in clinical efficacy include the restricted scope of intratumoral injections and poor targeting of intravenous administration. Further optimization of OVT delivery is needed before OVs become a viable therapy for tumor treatment. In this review, the development process and antitumor mechanisms of OVs are introduced. The advances in OVT delivery routes to provide perspectives and directions for the improvement of OVT delivery are highlighted. This review also discusses the advantages and limitations of OVT monotherapy and combination therapy through the lens of recent clinical trials and aims to chart a course toward safer and more effective OVT strategies.
Collapse
Affiliation(s)
- Shijie Duan
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Qiao
- Colorectal and Henia Minimally Invasive Surgery Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xinbo Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Nan Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Liting Chen
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xinyuan Zhang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xu Zhao
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Hongyu Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Tianye Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ying Wu
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Department of General Practice, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
15
|
Law LH, Huang J, Xiao P, Liu Y, Chen Z, Lai JHC, Han X, Cheng GWY, Tse KH, Chan KWY. Multiple CEST contrast imaging of nose-to-brain drug delivery using iohexol liposomes at 3T MRI. J Control Release 2023; 354:208-220. [PMID: 36623695 DOI: 10.1016/j.jconrel.2023.01.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
Image guided nose-to-brain drug delivery provides a non-invasive way to monitor drug delivered to the brain, and the intranasal administration could increase effective dose via bypassing Blood Brain Barrier (BBB). Here, we investigated the imaging of liposome-based drug delivery to the brain via intranasal administration, in which the liposome could penetrate mucus and could be detected by chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) at 3T field strength. Liposomes were loaded with a computed tomography (CT) contrast agent, iohexol (Ioh-Lipo), which has specific amide protons exchanging at 4.3 ppm of Z-spectrum (or CEST spectrum). Ioh-Lipo generated CEST contrasts of 35.4% at 4.3 ppm, 1.8% at -3.4 ppm and 20.6% at 1.2 ppm in vitro. After intranasal administration, these specific CEST contrasts were observed in both olfactory bulb (OB) and frontal lobe (FL) in the case of 10% polyethylene glycol (PEG) Ioh-Lipo. We observed obvious increases in CEST contrast in OB half an hour after the injection of 10% PEG Ioh-Lipo, with a percentage increase of 62.0% at 4.3 ppm, 10.9% at -3.4 ppm and 25.7% at 1.2 ppm. Interestingly, the CEST map at 4.3 ppm was distinctive from that at -3.4 pm and 1.2 ppm. The highest contrast of 4.3 ppm was at the external plexiform layer (EPL) and the region between left and right OB (LROB), while the CEST contrast at -3.4 ppm had no significant difference among all investigated regions with slightly higher signal in olfactory limbus (OL, between OB and FL) and FL, as validated with histology. While no substantial increase of CEST contrast at 4.3 ppm, -3.4 ppm or 1.2 ppm was observed in OB and FL when 1% PEG Ioh-Lipo was administered. We demonstrated for the first time the feasibility of non-invasively detecting the nose-to-brain delivery of liposomes using CEST MRI. This multiple-contrast approach is necessary to image the specific distribution of iohexol and liposome simultaneously and independently, especially when designing drug carriers for nose-to-brain drug delivery.
Collapse
Affiliation(s)
- Lok Hin Law
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Peng Xiao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Yang Liu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Joseph H C Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Xiongqi Han
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Gerald W Y Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Kai-Hei Tse
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Kannie W Y Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States; City University of Hong Kong Shenzhen Research Institute, Shenzhen, China; Tung Biomedical Science Centre, City University of Hong Kong, Hong Kong, China; Hong Kong Centre for Cerebro-cardiovascular Health Engineering, Hong Kong, China.
| |
Collapse
|
16
|
CNS Delivery of Nucleic Acid Therapeutics: Beyond the Blood-Brain Barrier and Towards Specific Cellular Targeting. Pharm Res 2023; 40:77-105. [PMID: 36380168 DOI: 10.1007/s11095-022-03433-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
Nucleic acid-based therapeutic molecules including small interfering RNA (siRNA), microRNA(miRNA), antisense oligonucleotides (ASOs), messenger RNA (mRNA), and DNA-based gene therapy have tremendous potential for treating diseases in the central nervous system (CNS). However, achieving clinically meaningful delivery to the brain and particularly to target cells and sub-cellular compartments is typically very challenging. Mediating cell-specific delivery in the CNS would be a crucial advance that mitigates off-target effects and toxicities. In this review, we describe these challenges and provide contemporary evidence of advances in cellular and sub-cellular delivery using a variety of delivery mechanisms and alternative routes of administration, including the nose-to-brain approach. Strategies to achieve subcellular localization, endosomal escape, cytosolic bioavailability, and nuclear transfer are also discussed. Ultimately, there are still many challenges to translating these experimental strategies into effective and clinically viable approaches for treating patients.
Collapse
|
17
|
Kumagai S, Nakajima T, Shimazaki K, Kakiuchi T, Harada N, Ohba H, Onuki Y, Takino N, Ito M, Sato M, Nakamura S, Osaka H, Yamagata T, Kawai K, Muramatsu SI. Early distribution of 18 F-labeled AAV9 vectors in the cerebrospinal fluid after intracerebroventricular or intracisternal magna infusion in non-human primates. J Gene Med 2023; 25:e3457. [PMID: 36278965 DOI: 10.1002/jgm.3457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/16/2022] [Accepted: 10/15/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The delivery of adeno-associated virus (AAV) vectors via the cerebrospinal fluid (CSF) has emerged as a valuable method for widespread transduction in the central nervous system. Although infusion into the cerebral ventricles is a common protocol in preclinical studies of small animals, the cisterna magna has been recognized as an alternative target for clinical studies because it can be reached in a less invasive manner using an intrathecal catheter via the subarachnoid space from a lumbar puncture. METHODS We evaluated the early distribution of fluorine-18-labeled AAV9 vectors infused into the lateral ventricle or cisterna magna of four non-human primates using positron emission tomography. The expression of the green fluorescent protein was immunohistochemically determined. RESULTS In both approaches, the labeled vectors diffused into the broad arachnoid space around the brain stem and cervical spinal cord within 30 min. Both infusion routes efficiently transduced neurons in the cervical spinal cord. CONCLUSIONS For gene therapy that primarily targets the cervical spinal cord and brainstem, such as amyotrophic lateral sclerosis, cisterna magna infusion would be a feasible and effective administration method.
Collapse
Affiliation(s)
- Shinichi Kumagai
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Takeshi Nakajima
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Kuniko Shimazaki
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Takeharu Kakiuchi
- Central Research Laboratory, Hamamatsu Photonics K.K., Shizuoka, Japan
| | - Norihiro Harada
- Central Research Laboratory, Hamamatsu Photonics K.K., Shizuoka, Japan
| | - Hiroyuki Ohba
- Central Research Laboratory, Hamamatsu Photonics K.K., Shizuoka, Japan
| | - Yoshiyuki Onuki
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Naomi Takino
- Division of Neurological Gene Therapy, Jichi Medical University, Tochigi, Japan
| | - Mika Ito
- Division of Neurological Gene Therapy, Jichi Medical University, Tochigi, Japan
| | - Makoto Sato
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Sachie Nakamura
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | | | - Kensuke Kawai
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Shin-Ichi Muramatsu
- Division of Neurological Gene Therapy, Jichi Medical University, Tochigi, Japan.,Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Blanco MJ, Gardinier KM, Namchuk MN. Advancing New Chemical Modalities into Clinical Studies. ACS Med Chem Lett 2022; 13:1691-1698. [PMID: 36385931 PMCID: PMC9661701 DOI: 10.1021/acsmedchemlett.2c00375] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/17/2022] [Indexed: 11/29/2022] Open
Abstract
Drug discovery and development has experienced an incredible paradigm shift in the past two decades. What once was considered a predominant R&D landscape of small molecules within a prescribed properties and mechanism space now includes an innovative wave of new chemical modalities. Scientists in the pharmaceutical industry can now strategize across a variety of modalities to find the best option to modulate a given target and provide treatment for a specific disease. We have witnessed a remarkable change not only in molecular design but also in creative approaches to drug delivery that have enabled advancement of novel modalities to clinical studies. In this Microperspective, we evaluate the critical differences between traditional small molecules and beyond rule of 5 compounds, peptides, oligonucleotides, and biologics for advancing into development, particularly their pharmacokinetic profiles and drug delivery strategies.
Collapse
Affiliation(s)
- Maria-Jesus Blanco
- Chemical
Sciences, Atavistik Bio, 75 Sidney Street, Cambridge Massachusetts 02139, United States
| | - Kevin M. Gardinier
- Discovery
Research, Karuna Therapeutics, 99 High Street Boston, Massachusetts 02110, United States
| | - Mark N. Namchuk
- Department
of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, 25 Shattuck Street Boston, Massachusetts 02115, United States
| |
Collapse
|
19
|
Minichmayr IK, Ravenstijn P, van der Graaf PH, Vamvakas S. Therapeutic Innovations in Neuroscience: What's New on the Horizon? Clin Pharmacol Ther 2022; 111:715-717. [PMID: 35313013 DOI: 10.1002/cpt.2547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 11/11/2022]
Affiliation(s)
| | | | - Piet H van der Graaf
- Certara, Canterbury, UK.,Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Spiros Vamvakas
- Human Division, European Medicines Agency, Amsterdam, The Netherlands
| |
Collapse
|