1
|
Henke L, Ghorbani A, Mole SE. The use of nanocarriers in treating Batten disease: A systematic review. Int J Pharm 2025; 670:125094. [PMID: 39694161 DOI: 10.1016/j.ijpharm.2024.125094] [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: 09/12/2024] [Revised: 12/09/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
The neuronal ceroid lipofuscinoses, commonly known as Batten disease, are a group of lysosomal storage disorders affecting children. There is extensive central nervous system and retinal degeneration, resulting in seizures, vision loss and a progressive cognitive and motor decline. Enzyme replacement and gene therapies are being developed, and mRNA and oligonucleotide therapies are more recently being considered. Overcoming the challenges of the blood-brain barrier and blood-ocular barrier is crucial for effectively targeting the brain and eye, whatever the therapeutic approach. Nanoparticles and extracellular vesicles are small carriers that can encapsulate a cargo and pass through these cell barriers. They have been investigated as drug carriers for other pathologies and could be a promising treatment strategy for Batten disease. Their use in gene, enzyme, or mRNA replacement therapy of all lysosomal storage disorders, including Mucopolysaccharidoses, Niemann-Pick diseases, and Fabry disease, is investigated in this systematic review. Different nanocarriers can efficiently target the lysosome and cross the barriers into the brain and eyes. This supports continued exploration of nanocarriers as potential future treatment options for Batten disease.
Collapse
Affiliation(s)
- Larissa Henke
- Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Ali Ghorbani
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Sara E Mole
- Great Ormond Street Institute of Child Health, University College London, London WC1E 6BT, UK.
| |
Collapse
|
2
|
Xu Z, He S, Begum MM, Han X. Myelin Lipid Alterations in Neurodegenerative Diseases: Landscape and Pathogenic Implications. Antioxid Redox Signal 2024; 41:1073-1099. [PMID: 39575748 PMCID: PMC11971557 DOI: 10.1089/ars.2024.0676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024]
Abstract
Significance: Lipids, which constitute the highest portion (over 50%) of brain dry mass, are crucial for brain integrity, energy homeostasis, and signaling regulation. Emerging evidence revealed that lipid profile alterations and abnormal lipid metabolism occur during normal aging and in different forms of neurodegenerative diseases. Moreover, increasing genome-wide association studies have validated new targets on lipid-associated pathways involved in disease development. Myelin, the protective sheath surrounding axons, is crucial for efficient neural signaling transduction. As the primary site enriched with lipids, impairments of myelin are increasingly recognized as playing significant and complex roles in various neurodegenerative diseases, beyond simply being secondary effects of neuronal loss. Recent Advances: With advances in the lipidomics field, myelin lipid alterations and their roles in contributing to or reflecting the progression of diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and others, have recently caught great attention. Critical Issues: This review summarizes recent findings of myelin lipid alterations in the five most common neurodegenerative diseases and discusses their implications in disease pathogenesis. Future Directions: By highlighting myelin lipid abnormalities in neurodegenerative diseases, this review aims to encourage further research focused on lipids and the development of new lipid-oriented therapeutic approaches in this area. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ziying Xu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Sijia He
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Mst Marium Begum
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
- Department of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
3
|
Wehn AC, Krestel E, Harapan BN, Klymchenko A, Plesnila N, Khalin I. To see or not to see: In vivo nanocarrier detection methods in the brain and their challenges. J Control Release 2024; 371:216-236. [PMID: 38810705 DOI: 10.1016/j.jconrel.2024.05.044] [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: 02/16/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024]
Abstract
Nanoparticles have a great potential to significantly improve the delivery of therapeutics to the brain and may also be equipped with properties to investigate brain function. The brain, being a highly complex organ shielded by selective barriers, requires its own specialized detection system. However, a significant hurdle to achieve these goals is still the identification of individual nanoparticles within the brain with sufficient cellular, subcellular, and temporal resolution. This review aims to provide a comprehensive summary of the current knowledge on detection systems for tracking nanoparticles across the blood-brain barrier and within the brain. We discuss commonly employed in vivo and ex vivo nanoparticle identification and quantification methods, as well as various imaging modalities able to detect nanoparticles in the brain. Advantages and weaknesses of these modalities as well as the biological factors that must be considered when interpreting results obtained through nanotechnologies are summarized. Finally, we critically evaluate the prevailing limitations of existing technologies and explore potential solutions.
Collapse
Affiliation(s)
- Antonia Clarissa Wehn
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Eva Krestel
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany.
| | - Biyan Nathanael Harapan
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Andrey Klymchenko
- Laboratoire de Biophotonique et Pharmacologie, CNRS UMR 7213, Université de Strasbourg, 74 route du Rhin - CS 60024, 67401 Illkirch Cedex, France.
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany.
| | - Igor Khalin
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), 14 074 Bd Henri Becquerel, 14000 Caen, France.
| |
Collapse
|
4
|
Hartl N, Gabold B, Uhl P, Kromer A, Xiao X, Fricker G, Mier W, Liu R, Merkel OM. ApoE-functionalization of nanoparticles for targeted brain delivery-a feasible method for polyplexes? Drug Deliv Transl Res 2024; 14:1660-1677. [PMID: 38087181 PMCID: PMC11052808 DOI: 10.1007/s13346-023-01482-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 04/28/2024]
Abstract
The blood-brain barrier (BBB) poses a major obstacle in the treatment of all types of central nervous system (CNS) diseases. Small interfering RNA (siRNA) offers in principle a promising therapeutic approach by downregulating disease-related genes via RNA interference. However, the BBB is a formidable barrier for macromolecules such as nucleic acids. In an effort to develop a brain-targeted strategy for siRNA delivery systems formed by electrostatic interactions with cationic polymers (polyplexes (PXs)), we investigated the suitability of the well-known surfactant-based approach for Apolipoprotein E (ApoE)-functionalization of nanoparticles (NPs). The aim of this present work was to investigate if ApoE coating of siRNA PXs formed with cationic branched 25-kDa poly(ethyleneimine) (b-PEI) and nylon-3 polymers without or after precoating with polysorbate 80 (PS 80) would promote successful delivery across the BBB. We utilized highly hydrophobic NM0.2/CP0.8 nylon-3 polymers to evaluate the effects of hydrophobic cyclopentyl (CP) subunits on ApoE binding efficacy and observed successful ApoE binding with and without PS 80 precoating to the nylon-3 but not the PEI polyplexes. Accordingly, ApoE-coated nylon-3 polyplexes showed significantly increased uptake and gene silencing in U87 glioma cells but no benefit in vivo. In conclusion, further optimization of ApoE-functionalized polyplexes and more sophisticated in vitro models are required to achieve more successful in vitro-in vivo translation in future approaches.
Collapse
Affiliation(s)
- Natascha Hartl
- Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Bettina Gabold
- Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Philipp Uhl
- Pharmaceutical Technology and Biopharmaceutics, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120, Heidelberg, Germany
| | - Adrian Kromer
- Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Ximian Xiao
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Gert Fricker
- Pharmaceutical Technology and Biopharmaceutics, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120, Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Olivia M Merkel
- Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
5
|
Moore TL, Pannuzzo G, Costabile G, Palange AL, Spanò R, Ferreira M, Graziano ACE, Decuzzi P, Cardile V. Nanomedicines to treat rare neurological disorders: The case of Krabbe disease. Adv Drug Deliv Rev 2023; 203:115132. [PMID: 37918668 DOI: 10.1016/j.addr.2023.115132] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/04/2023]
Abstract
The brain remains one of the most challenging therapeutic targets due to the low and selective permeability of the blood-brain barrier and complex architecture of the brain tissue. Nanomedicines, despite their relatively large size compared to small molecules and nucleic acids, are being heavily investigated as vehicles to delivery therapeutics into the brain. Here we elaborate on how nanomedicines may be used to treat rare neurodevelopmental disorders, using Krabbe disease (globoid cell leukodystrophy) to frame the discussion. As a monogenetic disorder and lysosomal storage disease affecting the nervous system, the lessons learned from examining nanoparticle delivery to the brain in the context of Krabbe disease can have a broader impact on the treatment of various other neurodevelopmental and neurodegenerative disorders. In this review, we introduce the epidemiology and genetic basis of Krabbe disease, discuss current in vitro and in vivo models of the disease, as well as current therapeutic approaches either approved or at different stage of clinical developments. We then elaborate on challenges in particle delivery to the brain, with a specific emphasis on methods to transport nanomedicines across the blood-brain barrier. We highlight nanoparticles for delivering therapeutics for the treatment of lysosomal storage diseases, classified by the therapeutic payload, including gene therapy, enzyme replacement therapy, and small molecule delivery. Finally, we provide some useful hints on the design of nanomedicines for the treatment of rare neurological disorders.
Collapse
Affiliation(s)
- Thomas Lee Moore
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy.
| | - Giovanna Pannuzzo
- Department of Biomedical and Biotechnological Sciences, Università di Catania, Catania 95123, CT, Italy
| | - Gabriella Costabile
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy; Department of Pharmacy, Università degli Studi di Napoli Federico II, Naples 80131, NA, Italy
| | - Anna Lisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy
| | - Raffaele Spanò
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy
| | - Adriana Carol Eleonora Graziano
- Department of Biomedical and Biotechnological Sciences, Università di Catania, Catania 95123, CT, Italy; Facolta di Medicina e Chirurgia, Università degli Studi di Enna "Kore", Enna 94100, EN, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, Università di Catania, Catania 95123, CT, Italy.
| |
Collapse
|
6
|
Loeck M, Placci M, Muro S. Effect of acid sphingomyelinase deficiency in type A Niemann-Pick disease on the transport of therapeutic nanocarriers across the blood-brain barrier. Drug Deliv Transl Res 2023; 13:3077-3093. [PMID: 37341882 DOI: 10.1007/s13346-023-01374-z] [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] [Accepted: 05/20/2023] [Indexed: 06/22/2023]
Abstract
ASM deficiency in Niemann-Pick disease type A results in aberrant cellular accumulation of sphingomyelin, neuroinflammation, neurodegeneration, and early death. There is no available treatment because enzyme replacement therapy cannot surmount the blood-brain barrier (BBB). Nanocarriers (NCs) targeted across the BBB via transcytosis might help; yet, whether ASM deficiency alters transcytosis remains poorly characterized. We investigated this using model NCs targeted to intracellular adhesion molecule-1 (ICAM-1), transferrin receptor (TfR), or plasmalemma vesicle-associated protein-1 (PV1) in ASM-normal vs. ASM-deficient BBB models. Disease differentially changed the expression of all three targets, with ICAM-1 becoming the highest. Apical binding and uptake of anti-TfR NCs and anti-PV1 NCs were unaffected by disease, while anti-ICAM-1 NCs had increased apical binding and decreased uptake rate, resulting in unchanged intracellular NCs. Additionally, anti-ICAM-1 NCs underwent basolateral reuptake after transcytosis, whose rate was decreased by disease, as for apical uptake. Consequently, disease increased the effective transcytosis rate for anti-ICAM-1 NCs. Increased transcytosis was also observed for anti-PV1 NCs, while anti-TfR NCs remained unaffected. A fraction of each formulation trafficked to endothelial lysosomes. This was decreased in disease for anti-ICAM-1 NCs and anti-PV1 NCs, agreeing with opposite transcytosis changes, while it increased for anti-TfR NCs. Overall, these variations in receptor expression and NC transport resulted in anti-ICAM-1 NCs displaying the highest absolute transcytosis in the disease condition. Furthermore, these results revealed that ASM deficiency can differently alter these processes depending on the particular target, for which this type of study is key to guide the design of therapeutic NCs.
Collapse
Affiliation(s)
- Maximilian Loeck
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain.
- Institution of Catalonia for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
7
|
Bao Y, Lu W. Targeting cerebral diseases with enhanced delivery of therapeutic proteins across the blood-brain barrier. Expert Opin Drug Deliv 2023; 20:1681-1698. [PMID: 36945117 DOI: 10.1080/17425247.2023.2193390] [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: 01/14/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Cerebral diseases have been threatening public physical and psychological health in the recent years. With the existence of the blood-brain barrier (BBB), it is particularly hard for therapeutic proteins like peptides, enzymes, antibodies, etc. to enter the central nervous system (CNS) and function in diagnosis and treatment in cerebral diseases. Fortunately, the past decade has witnessed some emerging strategies of delivering macromolecular therapeutic proteins across the BBB. AREAS COVERED Based on the structure, functions, and substances transport mechanisms, various enhanced delivery strategies of therapeutic proteins were reviewed, categorized by molecule-mediated delivery strategies, carrier-mediated delivery strategies, and other delivery strategies. EXPERT OPINION As for molecule-mediated delivery strategies, development of genetic engineering technology, optimization of protein expression and purification techniques, and mature of quality control systems all help to realize large-scale production of recombinant antibodies, making it possible to apply to the clinical practice. In terms of carrier-mediated delivery strategies and others, although nano-carriers/adeno-associated virus (AAV) are also promising candidates for delivering therapeutic proteins or genes across the BBB, some issues still remain to be further investigated, including safety concerns related to applied materials, large-scale production costs, quality control standards, combination therapies with auxiliary delivery strategies like focused ultrasound, etc.
Collapse
Affiliation(s)
- Yanning Bao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
- Department of Research and Development, Shanghai Tayzen PharmLab Co., Ltd. Lingang of Shanghai, China
| |
Collapse
|
8
|
Placci M, Giannotti MI, Muro S. Polymer-based drug delivery systems under investigation for enzyme replacement and other therapies of lysosomal storage disorders. Adv Drug Deliv Rev 2023; 197:114683. [PMID: 36657645 PMCID: PMC10629597 DOI: 10.1016/j.addr.2022.114683] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/30/2022] [Accepted: 12/25/2022] [Indexed: 01/18/2023]
Abstract
Lysosomes play a central role in cellular homeostasis and alterations in this compartment associate with many diseases. The most studied example is that of lysosomal storage disorders (LSDs), a group of 60 + maladies due to genetic mutations affecting lysosomal components, mostly enzymes. This leads to aberrant intracellular storage of macromolecules, altering normal cell function and causing multiorgan syndromes, often fatal within the first years of life. Several treatment modalities are available for a dozen LSDs, mostly consisting of enzyme replacement therapy (ERT) strategies. Yet, poor biodistribution to main targets such as the central nervous system, musculoskeletal tissue, and others, as well as generation of blocking antibodies and adverse effects hinder effective LSD treatment. Drug delivery systems are being studied to surmount these obstacles, including polymeric constructs and nanoparticles that constitute the focus of this article. We provide an overview of the formulations being tested, the diseases they aim to treat, and the results observed from respective in vitro and in vivo studies. We also discuss the advantages and disadvantages of these strategies, the remaining gaps of knowledge regarding their performance, and important items to consider for their clinical translation. Overall, polymeric nanoconstructs hold considerable promise to advance treatment for LSDs.
Collapse
Affiliation(s)
- Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Marina I Giannotti
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; CIBER-BBN, ISCIII, Barcelona, Spain; Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona 08028, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; Institute of Catalonia for Research and Advanced Studies (ICREA), Barcelona 08010, Spain; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
9
|
Critchley BJ, Gaspar HB, Benedetti S. Targeting the central nervous system in lysosomal storage diseases: Strategies to deliver therapeutics across the blood-brain barrier. Mol Ther 2023; 31:657-675. [PMID: 36457248 PMCID: PMC10014236 DOI: 10.1016/j.ymthe.2022.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/18/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are multisystem inherited metabolic disorders caused by dysfunctional lysosomal activity, resulting in the accumulation of undegraded macromolecules in a variety of organs/tissues, including the central nervous system (CNS). Treatments include enzyme replacement therapy, stem/progenitor cell transplantation, and in vivo gene therapy. However, these treatments are not fully effective in treating the CNS as neither enzymes, stem cells, nor viral vectors efficiently cross the blood-brain barrier. Here, we review the latest advancements in improving delivery of different therapeutic agents to the CNS and comment upon outstanding questions in the field of neurological LSDs.
Collapse
Affiliation(s)
- Bethan J Critchley
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK
| | - H Bobby Gaspar
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; Orchard Therapeutics Ltd., London EC4N 6EU, UK
| | - Sara Benedetti
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
10
|
Del Grosso A, Parlanti G, Mezzena R, Cecchini M. Current treatment options and novel nanotechnology-driven enzyme replacement strategies for lysosomal storage disorders. Adv Drug Deliv Rev 2022; 188:114464. [PMID: 35878795 DOI: 10.1016/j.addr.2022.114464] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/26/2022] [Accepted: 07/19/2022] [Indexed: 11/01/2022]
Abstract
Lysosomal storage disorders (LSDs) are a vast group of more than 50 clinically identified metabolic diseases. They are singly rare, but they affect collectively 1 on 5,000 live births. They result in most of the cases from an enzymatic defect within lysosomes, which causes the subsequent augmentation of unwanted substrates. This accumulation process leads to plenty of clinical signs, determined by the specific substrate and accumulation area. The majority of LSDs present a broad organ and tissue engagement. Brain, connective tissues, viscera and bones are usually afflicted. Among them, brain disease is markedly frequent (two-thirds of LSDs). The most clinically employed approach to treat LSDs is enzyme replacement therapy (ERT), which is practiced by administering systemically the missed or defective enzyme. It represents a healthful strategy for 11 LSDs at the moment, but it solves the pathology only in the case of Gaucher disease. This approach, in fact, is not efficacious in the case of LSDs that have an effect on the central nervous system (CNS) due to the existence of the blood-brain barrier (BBB). Additionally, ERT suffers from several other weak points, such as low penetration of the exogenously administered enzyme to poorly vascularized areas, the development of immunogenicity and infusion-associated reactions (IARs), and, last but not least, the very high cost and lifelong needed. To ameliorate these weaknesses lot of efforts have been recently spent around the development of innovative nanotechnology-driven ERT strategies. They may boost the power of ERT and minimize adverse reactions by loading enzymes into biodegradable nanomaterials. Enzyme encapsulation into biocompatible liposomes, micelles, and polymeric nanoparticles, for example, can protect enzymatic activity, eliminating immunologic reactions and premature enzyme degradation. It can also permit a controlled release of the payload, ameliorating pharmacokinetics and pharmacodynamics of the drug. Additionally, the potential to functionalize the surface of the nanocarrier with targeting agents (antibodies or peptides), could promote the passage through biological barriers. In this review we examined the clinically applied ERTs, highlighting limitations that do not allow to completely cure the specific LSD. Later, we critically consider the nanotechnology-based ERT strategies that have beenin-vitroand/orin-vivotested to improve ERT efficacy.
Collapse
Affiliation(s)
- Ambra Del Grosso
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Gabriele Parlanti
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Roberta Mezzena
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Marco Cecchini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
11
|
Calzoni E, Cesaretti A, Montegiove N, Di Michele A, Pellegrino RM, Emiliani C. HexA-Enzyme Coated Polymer Nanoparticles for the Development of a Drug-Delivery System in the Treatment of Sandhoff Lysosomal Storage Disease. J Funct Biomater 2022; 13:jfb13020037. [PMID: 35466219 PMCID: PMC9036261 DOI: 10.3390/jfb13020037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 01/20/2023] Open
Abstract
Lysosomal storage disorders (LSDs) are a set of metabolic diseases caused by mutations in genes that are in charge of the production of lysosomal enzymes, resulting in the buildup of non-degraded substrates and the consequent systemic damage that mainly involves the Central Nervous System (CNS). One of the most widely used and studied treatments is Enzyme Replacement Therapy, which is based on the administration of the recombinant deficient enzyme. This strategy has often proved fallacious due to the enzyme instability in body fluids and its inability to reach adequate levels in the CNS. In this work, we developed a system based on nanotechnology that allows a stable enzyme to be obtained by its covalent immobilization on nanoparticles (NPs) of polylactic acid, subsequently administered to a cellular model of LSDs, i.e., Sandhoff disease, caused by the absence or deficiency of the β-d-N-acetyl-hexosaminidase A (HexA) enzyme. The HexA enzymes, loaded onto the polymeric NPs through an immobilization procedure that has already been investigated and validated, were found to be stable over time, maintain optimal kinetic parameters, be able to permeate the plasma membrane, hydrolyze HexA’s natural substrate, and restore enzyme activity close to the levels of healthy cells. These results thus lay the foundation for testing the HexA-NPs in animal models of the disease and thus obtaining an efficient drug-delivery system.
Collapse
Affiliation(s)
- Eleonora Calzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (N.M.); (R.M.P.); (C.E.)
| | - Alessio Cesaretti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (N.M.); (R.M.P.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
- Correspondence: ; Tel.: +39-075-585-7436
| | - Nicolò Montegiove
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (N.M.); (R.M.P.); (C.E.)
| | - Alessandro Di Michele
- Department of Physics and Geology, University of Perugia, Via Pascoli, 06123 Perugia, Italy;
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (N.M.); (R.M.P.); (C.E.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (N.M.); (R.M.P.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| |
Collapse
|
12
|
Ndemazie NB, Inkoom A, Morfaw EF, Smith T, Aghimien M, Ebesoh D, Agyare E. Multi-disciplinary Approach for Drug and Gene Delivery Systems to the Brain. AAPS PharmSciTech 2021; 23:11. [PMID: 34862567 PMCID: PMC8817187 DOI: 10.1208/s12249-021-02144-1] [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: 05/26/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Drug delivery into the brain has for long been a huge challenge as the blood–brain barrier (BBB) offers great resistance to entry of foreign substances (with drugs inclusive) into the brain. This barrier in healthy individuals is protective to the brain, disallowing noxious substances present in the blood to get to the brain while allowing for the exchange of small molecules into the brain by diffusion. However, BBB is disrupted under certain disease conditions, such as cerebrovascular diseases including acute ischemic stroke and intracerebral hemorrhage, and neurodegenerative disorders including multiple sclerosis (MS), Alzheimer’s disease (AD), Parkinson’s disease (PD), and cancers. This review aims to provide a broad overview of present-day strategies for brain drug delivery, emphasizing novel delivery systems. Hopefully, this review would inspire scientists and researchers in the field of drug delivery across BBB to uncover new techniques and strategies to optimize drug delivery to the brain. Considering the anatomy, physiology, and pathophysiological functioning of the BBB in health and disease conditions, this review is focused on the controversies drawn from conclusions of recently published studies on issues such as the penetrability of nanoparticles into the brain, and whether active targeted drug delivery into the brain could be achieved with the use of nanoparticles. We also extended the review to cover novel non-nanoparticle strategies such as using viral and peptide vectors and other non-invasive techniques to enhance brain uptake of drugs.
Collapse
|
13
|
Latronico T, Rizzi F, Panniello A, Laquintana V, Arduino I, Denora N, Fanizza E, Milella S, Mastroianni CM, Striccoli M, Curri ML, Liuzzi GM, Depalo N. Luminescent PLGA Nanoparticles for Delivery of Darunavir to the Brain and Inhibition of Matrix Metalloproteinase-9, a Relevant Therapeutic Target of HIV-Associated Neurological Disorders. ACS Chem Neurosci 2021; 12:4286-4301. [PMID: 34726377 PMCID: PMC9297288 DOI: 10.1021/acschemneuro.1c00436] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
![]()
Human
immunodeficiency virus (HIV) can independently replicate
in the central nervous system (CNS) causing neurocognitive impairment
even in subjects with suppressed plasma viral load. The antiretroviral
drug darunavir (DRV) has been approved for therapy of HIV-infected
patients, but its efficacy in the treatment of HIV-associated neurological
disorders (HAND) is limited due to the low penetration through the
blood–brain barrier (BBB). Therefore, innovations in DRV formulations,
based on its encapsulation in optically traceable nanoparticles (NPs),
may improve its transport through the BBB, providing, at the same
time, optical monitoring of drug delivery within the CNS. The aim
of this study was to synthesize biodegradable polymeric NPs loaded
with DRV and luminescent, nontoxic carbon dots (C-Dots) and investigate
their ability to permeate through an artificial BBB and to inhibit in vitro matrix metalloproteinase-9 (MMP-9) that represents
a factor responsible for the development of HIV-related neurological
disorders. Biodegradable poly(lactic-co-glycolic)
acid (PLGA)-based nanoformulations resulted characterized by an average
hydrodynamic size less than 150 nm, relevant colloidal stability in
aqueous medium, satisfactory drug encapsulation efficiency, and retained
emitting optical properties in the visible region of the electromagnetic
spectrum. The assay on the BBB artificial model showed that a larger
amount of DRV was able to cross BBB when incorporated in the PLGA
NPs and to exert an enhanced inhibition of matrix metalloproteinase-9
(MMP-9) expression levels with respect to free DRV. The overall results
reveal the great potential of this class of nanovectors of DRV for
an efficacious treatment of HANDs.
Collapse
Affiliation(s)
- Tiziana Latronico
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126 Bari, Italy
| | - Federica Rizzi
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Annamaria Panniello
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Valentino Laquintana
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari, Via Orabona 4, 70126 Bari, Italy
| | - Ilaria Arduino
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari, Via Orabona 4, 70126 Bari, Italy
| | - Nunzio Denora
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari, Via Orabona 4, 70126 Bari, Italy
| | - Elisabetta Fanizza
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Serafina Milella
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126 Bari, Italy
| | - Claudio M. Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University, AOU Policlinico Umberto 1, 00185 Rome, Italy
| | - Marinella Striccoli
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Maria Lucia Curri
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Grazia M. Liuzzi
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126 Bari, Italy
| | - Nicoletta Depalo
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| |
Collapse
|
14
|
Tween ® Preserves Enzyme Activity and Stability in PLGA Nanoparticles. NANOMATERIALS 2021; 11:nano11112946. [PMID: 34835710 PMCID: PMC8625811 DOI: 10.3390/nano11112946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022]
Abstract
Enzymes, as natural and potentially long-term treatment options, have become one of the most sought-after pharmaceutical molecules to be delivered with nanoparticles (NPs); however, their instability during formulation often leads to underwhelming results. Various molecules, including the Tween® polysorbate series, have demonstrated enzyme activity protection but are often used uncontrolled without optimization. Here, poly(lactic-co-glycolic) acid (PLGA) NPs loaded with β-glucosidase (β-Glu) solutions containing Tween® 20, 60, or 80 were compared. Mixing the enzyme with Tween® pre-formulation had no effect on particle size or physical characteristics, but increased the amount of enzyme loaded. More importantly, NPs made with Tween® 20:enzyme solutions maintained significantly higher enzyme activity. Therefore, Tween® 20:enzyme solutions ranging from 60:1 to 2419:1 mol:mol were further analyzed. Isothermal titration calorimetry analysis demonstrated low affinity and unquantifiable binding between Tween® 20 and β-Glu. Incorporating these solutions in NPs showed no effect on size, zeta potential, or morphology. The amount of enzyme and Tween® 20 in the NPs was constant for all samples, but a trend towards higher activity with higher molar rapports of Tween® 20:β-Glu was observed. Finally, a burst release from NPs in the first hour with Tween®:β-Glu solutions was the same as free enzyme, but the enzyme remained active longer in solution. These results highlight the importance of stabilizers during NP formulation and how optimizing their use to stabilize an enzyme can help researchers design more efficient and effective enzyme loaded NPs.
Collapse
|
15
|
Calzoni E, Cesaretti A, Montegiove N, Di Michele A, Emiliani C. Enhanced Stability of Long-Living Immobilized Recombinant β-d- N-Acetyl-Hexosaminidase A on Polylactic Acid (PLA) Films for Potential Biomedical Applications. J Funct Biomater 2021; 12:jfb12020032. [PMID: 34064736 PMCID: PMC8162980 DOI: 10.3390/jfb12020032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 01/24/2023] Open
Abstract
β-d-N-acetyl-hexosaminidase (Hex, EC 3.2.1.52) is an acid hydrolase that catalyzes the cleavage of the β-1,4 bond in N-acetyl-d-galactosamine (Gal-NAc) and N-acetyl-d-glucosamine (Glc-NAc) from the non-reducing end of oligosaccharides and glycoconjugates. It is widely expressed in both the prokaryotic and eukaryotic world, where it performs multiple and important functions. Hex has antifungal activity in plants, is capable of degrading many biological substrates, and can play an important role in the biomedical field for the treatment of Tay-Sachs and Sandhoff diseases. With the aim being able to obtain a device with a stable enzyme, a method of covalent immobilization on polylactic acid (PLA) films was developed for the A isoform of the β-d-N-acetyl-hexosaminidase enzyme (HexA), produced in a recombinant way from Human Embryonic Kidney-293 (HEK-293) cells and suitably purified. An in-depth biochemical characterization of the immobilized enzyme was carried out, evaluating the optimal temperature, thermal stability, pH parameters, and Km value. Moreover, the stability of the enzymatic activity over time was assessed. The results obtained showed an improvement in terms of kinetic parameters and stability to heat for the enzyme following immobilization and the presence of HexA in two distinct immobilized forms, with an unexpected ability for one of them to maintain its functionality for a long period of time (over a year). The stability and functionality of the enzyme in its immobilized form are therefore extremely promising for potential biotechnological and biomedical applications.
Collapse
Affiliation(s)
- Eleonora Calzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (E.C.); (N.M.); (C.E.)
| | - Alessio Cesaretti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (E.C.); (N.M.); (C.E.)
- Center of Excellence on Innovative Nanostructured Materials—CEMIN, University of Perugia, 06123 Perugia, Italy
- Correspondence: ; Tel.: +39-075-585-7436
| | - Nicolò Montegiove
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (E.C.); (N.M.); (C.E.)
| | | | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (E.C.); (N.M.); (C.E.)
- Center of Excellence on Innovative Nanostructured Materials—CEMIN, University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
16
|
Hartl N, Adams F, Merkel OM. From adsorption to covalent bonding: Apolipoprotein E functionalization of polymeric nanoparticles for drug delivery across the blood-brain barrier. ADVANCED THERAPEUTICS 2021; 4:2000092. [PMID: 33542947 PMCID: PMC7116687 DOI: 10.1002/adtp.202000092] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) is composed of brain endothelial cells, pericytes, and astrocytes, which build a tight cellular barrier. Therapeutic (macro)molecules are not able to transit through the BBB in their free form. This limitation is bypassed by apolipoprotein E (ApoE)-functionalized polymeric nanoparticles (NPs) that are able to transport drugs (e.g. dalargin, loperamide, doxorubicin, nerve growth factor) across the BBB via low density lipoprotein (LDL) receptor mediated transcytosis. Coating with polysorbate 80 or poloxamer 188 facilitates ApoE adsorption onto polymeric NPs enabling recognition by LDL receptors of brain endothelial cells. This effect is even enhanced when NPs are directly coated with ApoE without surfactant anchor. Similarly, covalent coupling of ApoE to NPs that bear reactive groups on their surface leads to significantly improved brain uptake while avoiding the use of surfactants. Several in vitro BBB models using brain endothelial cells or co-cultures with astrocytes/pericytes/glioma cells are described which provide first insights regarding the ability of a drug delivery system to cross this barrier. In vivo models are employed to simulate central nervous system-relevant diseases such as Alzheimer's or Parkinson's disease and cerebral cancer.
Collapse
Affiliation(s)
| | | | - Olivia M. Merkel
- Pharmaceutical Technology and Biopharmaceutics, Department Pharmacy, Ludwig-Maximilians-University, Butenandtstr. 5-13, 81377 Munich, Germany
| |
Collapse
|
17
|
Fonseca-Santos B, Chorilli M. The uses of resveratrol for neurological diseases treatment and insights for nanotechnology based-drug delivery systems. Int J Pharm 2020; 589:119832. [PMID: 32877730 DOI: 10.1016/j.ijpharm.2020.119832] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
Abstract
Neurological disorders have been growing in recent years and are highly prevalent globally. Resveratrol (RES) is a natural product from plant sources such as grape skins. This compound has shown biological activity in many diseases, in particular, those that act on the central nervous system. The mechanism of action and the key points in neurological disorders were described and show the targeted mechanism of action. Due to the insolubility of this compound; the use of nanotechnology-based systems has been proposed for the incorporation of RES and RES-loaded nanocarriers have been designed for intranasal administration, oral or parenteral routes to deliver it to the brain. In general, these nanosystems have shown to be effective in many studies, pharmacological and pharmacokinetic assays, as well as some cell studies. The outcomes show that RES has been reported in human clinical trials for some neurological diseases, although no studies were performed in humans using nanocarriers, animal and/or cellular models have been reported to show good results regarding therapeutics on neurological diseases. Thus, the use of this nutraceutical has shown true for neurological diseases and its loading into nanocarriers displaying good results on the stability, delivery and targeting to the brain.
Collapse
Affiliation(s)
- Bruno Fonseca-Santos
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Department of Drugs and Medicines, Araraquara, São Paulo 14801-903, Brazil
| | - Marlus Chorilli
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Department of Drugs and Medicines, Araraquara, São Paulo 14801-903, Brazil.
| |
Collapse
|
18
|
Simonis H, Yaghootfam C, Sylvester M, Gieselmann V, Matzner U. Evolutionary redesign of the lysosomal enzyme arylsulfatase A increases efficacy of enzyme replacement therapy for metachromatic leukodystrophy. Hum Mol Genet 2020; 28:1810-1821. [PMID: 30657900 DOI: 10.1093/hmg/ddz020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 11/13/2022] Open
Abstract
Protein engineering is a means to optimize protein therapeutics developed for the treatment of so far incurable diseases including cancers and genetic disorders. Here we report on an engineering approach in which we successfully increased the catalytic rate constant of an enzyme that is presently evaluated in enzyme replacement therapies (ERT) of a lysosomal storage disease (LSD). Although ERT is a treatment option for many LSDs, outcomes are lagging far behind expectations for most of them. This has been ascribed to insufficient enzyme activities accumulating in tissues difficult to target such as brain and peripheral nerves. We show for human arylsulfatase A (hARSA) that the activity of a therapeutic enzyme can be substantially increased by reversing activity-diminishing and by inserting activity-promoting amino acid substitutions that had occurred in the evolution of hominids and non-human mammals, respectively. The potential of this approach, here designated as evolutionary redesign, was highlighted by the observation that murinization of only 1 or 3 amino acid positions increased the hARSA activity 3- and 5-fold, with little impact on stability, respectively. The two kinetically optimized hARSA variants showed no immunogenic potential in ERT of a humanized ARSA knockout mouse model of metachromatic leukodystrophy (MLD) and reduced lysosomal storage of kidney, peripheral and central nervous system up to 3-fold more efficiently than wild-type hARSA. Due to their safety profile and higher therapeutic potential the engineered hARSA variants might represent major advances for future enzyme-based therapies of MLD and stimulate analogous approaches for other enzyme therapeutics.
Collapse
Affiliation(s)
- Heidi Simonis
- Institute of Biochemistry and Molecular Biology, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Claudia Yaghootfam
- Institute of Biochemistry and Molecular Biology, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Marc Sylvester
- Institute of Biochemistry and Molecular Biology, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Volkmar Gieselmann
- Institute of Biochemistry and Molecular Biology, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Ulrich Matzner
- Institute of Biochemistry and Molecular Biology, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| |
Collapse
|
19
|
Tosi G, Duskey JT, Kreuter J. Nanoparticles as carriers for drug delivery of macromolecules across the blood-brain barrier. Expert Opin Drug Deliv 2019; 17:23-32. [PMID: 31774000 DOI: 10.1080/17425247.2020.1698544] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Current therapies of neurodegenerative or neurometabolic diseases are, to a large extent, hampered by the inability of drugs to cross the blood-brain barrier (BBB). This very tight barrier severely restricts the entrance of molecules from the blood into the brain, especially macromolecular substances (i.e. neurotrophic factors, enzymes, proteins, as well as genetic materials). Due to their size, physicochemical properties, and instability, the delivery of these materials is particularly difficult.Areas covered: Recent research showed that biocompatible and biodegradable nanoparticles possessing tailored surface properties can enable a delivery of drugs and specifically of macromolecules across the blood-brain barrier by using carrier systems of the brain capillary endothelium (Trojan Horse strategy). In the present review, the state-of-art of nanoparticle-mediated drug delivery of different macromolecular substances into the brain following intravenous injection is summarized, and different nanomedicines that are used to enable the transport of neurotrophic factors and enzymes across the blood-brain barrier into the CNS are critically analyzed.Expert opinion: Brain delivery of macromolecules by an intravenous application using nanomedicines is now a growing area of interest which could be really translated into clinical application if dedicated effort will be given to industrial scale-up production.
Collapse
Affiliation(s)
- Giovanni Tosi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italia
| | - J T Duskey
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italia
| | - Jörg Kreuter
- Laboratory of Drug Delivery Systems, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Institute for Pharmaceutical Technology, Goethe-University Frankfurt, Germany
| |
Collapse
|
20
|
Luque-Michel E, Sebastian V, Larrea A, Marquina C, Blanco-Prieto MJ. Co-encapsulation of superparamagnetic nanoparticles and doxorubicin in PLGA nanocarriers: Development, characterization and in vitro antitumor efficacy in glioma cells. Eur J Pharm Biopharm 2019; 145:65-75. [PMID: 31628997 DOI: 10.1016/j.ejpb.2019.10.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/15/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022]
Abstract
With a very poor prognosis and no clear etiology, glioma is the most aggressive cancer in the brain. Thanks to its versatility, nanomedicine is a promising option to overcome the limitations on chemotherapy imposed by the blood brain barrier (BBB). The objective of this paper was to obtain monitored tumor-targeted therapeutic nanoparticles (NPs). To that end, theranostic surfactant-coated polymer poly-Lactic-co-Glycolic Acid (PLGA) nanoplatform encapsulating doxorubicin hydrochloride (DOX) and superparamagnetic iron oxide NPs (SPIONs) were developed. Different non-ionic surfactants known as BBB crossing enhancers (Tween 80, Brij-35, Pluronic F68 or Vitamin E-TPGS) were used to develop 4 types of theranostic nanoplatforms, which were characterized in terms of size and morphology by DLS, TEM and STEM-HAADF analyses. Moreover, the 3-month stability test, the therapeutic efficacy against different glioma cell lines (U87-MG, 9L/LacZ and patient derived-neuronal stem cells) and the Magnetic Resonance Imaging (MRI) relaxivity were studied. Results showed that the synthesised nanoplatforms were stable at 4 °C after their lyophilization, being that of paramount importance to ensure a long-term stability in a future in vivo application. Furthermore, the theranostic nanoplatforms were efficient in the in vitro treatment of glioma cells, proving to have imaging efficacy as MRI contrast agents. Our results show an efficient loading of drugs and good value of the relaxivity. Therefore, the efficient theranostic hybrid nanoplatform developed here could be used to perform MRI-guided delivery of hydrophobic drugs.
Collapse
Affiliation(s)
- Edurne Luque-Michel
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, C/Irunlarrea 1, E-31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Víctor Sebastian
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC-Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain; Institute of Nanoscience of Aragon (INA) and Department of Chemical, Engineering and Environmental Technology, University of Zaragoza, C/Mariano Esquillor, s/n, I+D+I Building, 50018 Zaragoza, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Centro de Investigación Biomédica en Red, C/ Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain.
| | - Ane Larrea
- Institute of Nanoscience of Aragon (INA) and Department of Chemical, Engineering and Environmental Technology, University of Zaragoza, C/Mariano Esquillor, s/n, I+D+I Building, 50018 Zaragoza, Spain
| | - Clara Marquina
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC-Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain; Departamento de Física de la Materia Condensada, Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - María J Blanco-Prieto
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, C/Irunlarrea 1, E-31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
21
|
Rutherford HA, Hamilton N. Animal models of leukodystrophy: a new perspective for the development of therapies. FEBS J 2019; 286:4176-4191. [DOI: 10.1111/febs.15060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/31/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Holly A. Rutherford
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease University of Sheffield UK
| | - Noémie Hamilton
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease University of Sheffield UK
| |
Collapse
|
22
|
Gigliobianco MR, Di Martino P, Deng S, Casadidio C, Censi R. New Advanced Strategies for the Treatment of Lysosomal Diseases Affecting the Central Nervous System. Curr Pharm Des 2019; 25:1933-1950. [DOI: 10.2174/1381612825666190708213159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/19/2019] [Indexed: 11/22/2022]
Abstract
Lysosomal Storage Disorders (LSDs), also known as lysosomal diseases (LDs) are a group of serious genetic diseases characterized by not only the accumulation of non-catabolized compounds in the lysosomes due to the deficiency of specific enzymes which usually eliminate these compounds, but also by trafficking, calcium changes and acidification. LDs mainly affect the central nervous system (CNS), which is difficult to reach for drugs and biological molecules due to the presence of the blood-brain barrier (BBB). While some therapies have proven highly effective in treating peripheral disorders in LD patients, they fail to overcome the BBB. Researchers have developed many strategies to circumvent this problem, for example, by creating carriers for enzyme delivery, which improve the enzyme’s half-life and the overexpression of receptors and transporters in the luminal or abluminal membranes of the BBB. This review aims to successfully examine the strategies developed during the last decade for the treatment of LDs, which mainly affect the CNS. Among the LD treatments, enzyme-replacement therapy (ERT) and gene therapy have proven effective, while nanoparticle, fusion protein, and small molecule-based therapies seem to offer considerable promise to treat the CNS pathology. This work also analyzed the challenges of the study to design new drug delivery systems for the effective treatment of LDs. Polymeric nanoparticles and liposomes are explored from their technological point of view and for the most relevant preclinical studies showing that they are excellent choices to protect active molecules and transport them through the BBB to target specific brain substrates for the treatment of LDs.
Collapse
Affiliation(s)
- Maria R. Gigliobianco
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Piera Di Martino
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Siyuan Deng
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Cristina Casadidio
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Roberta Censi
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| |
Collapse
|
23
|
Kovacs Z, Jung I, Gurzu S. Arylsulfatases A and B: From normal tissues to malignant tumors. Pathol Res Pract 2019; 215:152516. [PMID: 31262576 DOI: 10.1016/j.prp.2019.152516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/11/2019] [Accepted: 06/25/2019] [Indexed: 02/08/2023]
Abstract
Arylsulfatases are lysosomal enzymes with important roles in the cell metabolism. Several subtypes of arylsulfatase are known, from A to K. Congenital deficiencies of arylsulfatases, especially A (ARSA) and B (ARSB), can induce metabolic disorders such as metachromatic leucodystrophy (ARSA deficiency) and Maroteaux-Lamy syndrome (ARSB deficiency). ARSA and ARSB pseudodeficiencies were recently described but their exact roles are far to be known. The aim of this review was to synthesize the literature data, combined with personal results, regarding the roles of ARSA and ARSB in non-tumor disorders but also carcinogenesis. Few than 50 published papers regard ARSA and ARSB expression in cancer. They suggest decreased activity of these arylsulfatases in most of carcinomas, compared with normal tissues. However, the clinical impact is still unknown. Further complex studies are necessary to be done, to understand the role of ARSA and ARSB expression in cancer.
Collapse
Affiliation(s)
- Zsolt Kovacs
- Department of Pathology, University of Medicine, Pharmacy, Sciences and Technology, Tirgu-Mures, Romania; Department of Biochemistry, University of Medicine, Pharmacy, Sciences and Technology, Tirgu-Mures, Romania
| | - Ioan Jung
- Department of Pathology, University of Medicine, Pharmacy, Sciences and Technology, Tirgu-Mures, Romania
| | - Simona Gurzu
- Department of Pathology, University of Medicine, Pharmacy, Sciences and Technology, Tirgu-Mures, Romania; Department of Pathology, Research Center (CCAMF), Tirgu-Mures, Romania.
| |
Collapse
|
24
|
Targeting Brain Disease in MPSII: Preclinical Evaluation of IDS-Loaded PLGA Nanoparticles. Int J Mol Sci 2019; 20:ijms20082014. [PMID: 31022913 PMCID: PMC6514713 DOI: 10.3390/ijms20082014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 12/17/2022] Open
Abstract
Mucopolysaccharidosis type II (MPSII) is a lysosomal storage disorder due to the deficit of the enzyme iduronate 2-sulfatase (IDS), which leads to the accumulation of glycosaminoglycans in most organ-systems, including the brain, and resulting in neurological involvement in about two-thirds of the patients. The main treatment is represented by a weekly infusion of the functional enzyme, which cannot cross the blood-brain barrier and reach the central nervous system. In this study, a tailored nanomedicine approach based on brain-targeted polymeric nanoparticles (g7-NPs), loaded with the therapeutic enzyme, was exploited. Fibroblasts from MPSII patients were treated for 7 days with NPs loaded with the IDS enzyme; an induced IDS activity like the one detected in healthy cells was measured, together with a reduction of GAG content to non-pathological levels. An in vivo short-term study in MPSII mice was performed by weekly administration of g7-NPs-IDS. Biochemical, histological, and immunohistochemical evaluations of liver and brain were performed. The 6-weeks treatment produced a significant reduction of GAG deposits in liver and brain tissues, as well as a reduction of some neurological and inflammatory markers (i.e., LAMP2, CD68, GFAP), highlighting a general improvement of the brain pathology. The g7-NPs-IDS approach allowed a brain-targeted enzyme replacement therapy. Based on these positive results, the future aim will be to optimize NP formulation further to gain a higher efficacy of the proposed approach.
Collapse
|
25
|
Calzoni E, Cesaretti A, Polchi A, Di Michele A, Tancini B, Emiliani C. Biocompatible Polymer Nanoparticles for Drug Delivery Applications in Cancer and Neurodegenerative Disorder Therapies. J Funct Biomater 2019; 10:jfb10010004. [PMID: 30626094 PMCID: PMC6463038 DOI: 10.3390/jfb10010004] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 12/19/2022] Open
Abstract
Polymer nanoparticles (NPs) represent one of the most innovative non-invasive approaches for drug delivery applications. NPs main objective is to convey the therapeutic molecule be they drugs, proteins, or nucleic acids directly into the target organ or tissue. Many polymers are used for the synthesis of NPs and among the currently most employed materials several biocompatible synthetic polymers, namely polylactic acid (PLA), poly lactic-co-glycolic acid (PLGA), and polyethylene glycol (PEG), can be cited. These molecules are made of simple monomers which are naturally present in the body and therefore easily excreted without being toxic. The present review addresses the different approaches that are most commonly adopted to synthetize biocompatible NPs to date, as well as the experimental strategies designed to load them with therapeutic agents. In fact, drugs may be internalized in the NPs or physically dispersed therein. In this paper the various types of biodegradable polymer NPs will be discussed with emphasis on their applications in drug delivery. Close attention will be devoted to the treatment of cancer, where both active and passive targeting is used to enhance efficacy and reduce systemic toxicity, and to diseases affecting the central nervous system, inasmuch as NPs can be modified to target specific cells or cross membrane barriers.
Collapse
Affiliation(s)
- Eleonora Calzoni
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
- Centro di Eccellenza su Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| | - Alessio Cesaretti
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
- Centro di Eccellenza su Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| | - Alice Polchi
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
- Centro di Eccellenza su Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| | - Alessandro Di Michele
- Department of Physics and Geology, University of Perugia, via Pascoli, 06123 Perugia, Italy.
| | - Brunella Tancini
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
- Centro di Eccellenza su Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
- Centro di Eccellenza su Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| |
Collapse
|
26
|
Khan AR, Yang X, Fu M, Zhai G. Recent progress of drug nanoformulations targeting to brain. J Control Release 2018; 291:37-64. [PMID: 30308256 DOI: 10.1016/j.jconrel.2018.10.004] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 02/08/2023]
|
27
|
González-Mayorga A, López-Dolado E, Gutiérrez MC, Collazos-Castro JE, Ferrer ML, del Monte F, Serrano MC. Favorable Biological Responses of Neural Cells and Tissue Interacting with Graphene Oxide Microfibers. ACS OMEGA 2017; 2:8253-8263. [PMID: 30023578 PMCID: PMC6044865 DOI: 10.1021/acsomega.7b01354] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/01/2017] [Indexed: 05/24/2023]
Abstract
Neural tissue engineering approaches show increasing promise for the treatment of neural diseases including spinal cord injury, for which an efficient therapy is still missing. Encouraged by both positive findings on the interaction of carbon nanomaterials such as graphene with neural components and the necessity of more efficient guidance structures for neural repair, we herein study the potential of reduced graphene oxide (rGO) microfibers as substrates for neural growth in the injured central neural tissue. Compact, bendable, and conductive fibers are obtained. When coated with neural adhesive molecules (poly-l-lysine and N-cadherin), these microfibers behave as supportive substrates of highly interconnected cultures composed of neurons and glial cells for up to 21 days. Synaptic contacts close to rGO are identified. Interestingly, the colonization by meningeal fibroblasts is dramatically hindered by N-cadherin coating. Finally, in vivo studies reveal the feasible implantation of these rGO microfibers as a guidance platform in the injured rat spinal cord, without evident signs of subacute local toxicity. These positive findings boost further investigation at longer implantation times to prove the utility of these substrates as components of advanced therapies for enhancing repair in the damaged central neural tissue including the injured spinal cord.
Collapse
Affiliation(s)
- Ankor González-Mayorga
- Hospital
Nacional de Parapléjicos, Servicio de Salud de Castilla-La
Mancha (HNP-SESCAM), Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Elisa López-Dolado
- Hospital
Nacional de Parapléjicos, Servicio de Salud de Castilla-La
Mancha (HNP-SESCAM), Finca La Peraleda s/n, 45071 Toledo, Spain
- Research
Unit of “Design and Development of Biomaterials for Neural
Regeneration”, Hospital Nacional
de Parapléjicos (HNP-SESCAM), Joint Research Unit with CSIC, 45071 Toledo, Spain
| | - María C. Gutiérrez
- Materials
Science Factory, Instituto de Ciencia de
Materiales de Madrid, Consejo Superior de Investigaciones Científicas
(ICMM-CSIC), Calle Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain
| | - Jorge E. Collazos-Castro
- Hospital
Nacional de Parapléjicos, Servicio de Salud de Castilla-La
Mancha (HNP-SESCAM), Finca La Peraleda s/n, 45071 Toledo, Spain
| | - M. Luisa Ferrer
- Materials
Science Factory, Instituto de Ciencia de
Materiales de Madrid, Consejo Superior de Investigaciones Científicas
(ICMM-CSIC), Calle Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain
| | - Francisco del Monte
- Materials
Science Factory, Instituto de Ciencia de
Materiales de Madrid, Consejo Superior de Investigaciones Científicas
(ICMM-CSIC), Calle Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain
| | - María C. Serrano
- Research
Unit of “Design and Development of Biomaterials for Neural
Regeneration”, Hospital Nacional
de Parapléjicos (HNP-SESCAM), Joint Research Unit with CSIC, 45071 Toledo, Spain
- Materials
Science Factory, Instituto de Ciencia de
Materiales de Madrid, Consejo Superior de Investigaciones Científicas
(ICMM-CSIC), Calle Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain
| |
Collapse
|
28
|
Giugliani R, Vairo F, Kubaski F, Poswar F, Riegel M, Baldo G, Saute JA. Neurological manifestations of lysosomal disorders and emerging therapies targeting the CNS. THE LANCET CHILD & ADOLESCENT HEALTH 2017; 2:56-68. [PMID: 30169196 DOI: 10.1016/s2352-4642(17)30087-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/01/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022]
Abstract
Lysosomal disorders have been an area of interest since intravenous enzyme replacement therapy was successfully introduced for the treatment of Gaucher's disease in the early 1990s. This treatment approach has also been developed for several other lysosomal disorders, including Fabry's disease, Pompe's disease, lysosomal acid lipase deficiency, and five types of mucopolysaccharidosis. Despite the benefits of enzyme replacement therapy, it has limitations-most importantly, its ineffectiveness in treating the neurological components of lysosomal disorders, as only a small proportion of recombinant enzymes can cross the blood-brain barrier. Development of strategies to improve drug delivery to the CNS is now the primary focus in lysosomal disorder research. This Review discusses the neurological manifestations and emerging therapies for the CNS component of these diseases. The therapies in development (which are now in phase 1 or phase 2 clinical trials) might be for specific lysosomal disorders (enzyme replacement therapy via intrathecal or intracerebroventricular routes or with fusion proteins, or gene therapy) or applicable to more than one lysosomal disorder (haemopoietic stem cell transplantation, pharmacological chaperones, substrate reduction therapy, or stop codon readthrough). The combination of early diagnosis with effective therapies should change the outlook for patients with lysosomal disorders with neurological involvement in the next 5-10 years.
Collapse
Affiliation(s)
- Roberto Giugliani
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | | | | | - Fabiano Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mariluce Riegel
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Baldo
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Postgraduate Program in Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jonas Alex Saute
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
29
|
Cai H, Liang Z, Huang W, Wen L, Chen G. Engineering PLGA nano-based systems through understanding the influence of nanoparticle properties and cell-penetrating peptides for cochlear drug delivery. Int J Pharm 2017; 532:55-65. [PMID: 28870763 DOI: 10.1016/j.ijpharm.2017.08.084] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/15/2017] [Accepted: 08/15/2017] [Indexed: 12/13/2022]
Abstract
The properties of poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) and penetration enhancers play a deciding role in the inner ear drug delivery of NPs across the round window membrane (RWM). Thus, PLGA nano-based systems with a variety of particle sizes and surface chemistries and those combined with cell-penetrating peptides (CPPs) as penetration enhancers were devised to explore their impact on the cochlear drug delivery in vivo. First, we demonstrated that the properties of NPs dictated the extent of NP cochlear entry by near-infrared fluorescence imaging. NPs with the sizes of 150 and 300nm had faster entry than that of 80nm NPs. At 0.5h, among the NPs unmodified and modified with chitosan (CS), poloxamer 407 (P407) and methoxy polyethylene glycol, CS-PLGA-NPs (positive surface charge) carried payload to the cochlea fastest, whereas P407-PLGA-NPs (surface hydrophilicity) showed the greatest distribution in the cochlea at 24h. Compared to other CPPs (TAT, penetratin and poly(arginine)8), low molecular weight protamine (LMWP) performed an outstanding enhanced NP cellular uptake in HEI-OC1 cells and cochlear entry. More importantly, NPs with optimized properties and CPPs may be combined to improve RWM penetration. For the first time, we confirmed that the combination of P407-PLGA-NPs (mean diameter: 100-200nm) and LMWP provided a synergistic enhancement in NP entry to the organ of Corti and stria vascularis without inducing pathological alteration of cochlear tissues and RWM. Taken together, we propose an effective PLGA nano-based strategy for enhanced drug delivery to the inner ear tissues that combines hydrophilic molecule-modified NPs and CPPs, ultimately opening an avenue for superior inner ear therapy.
Collapse
Affiliation(s)
- Hui Cai
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhongping Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wenli Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lu Wen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Gang Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|