1
|
Panghal A, Flora SJS. Nano-based approaches for the treatment of neuro-immunological disorders: a special emphasis on multiple sclerosis. DISCOVER NANO 2024; 19:171. [PMID: 39466516 PMCID: PMC11519283 DOI: 10.1186/s11671-024-04135-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Multiple sclerosis (MS) is a neuroimmunological disorder which causes axonal damage, demyelination and paralysis. Although numerous therapeutics have been developed for the effective treatment of MS and a few have been approved in recent decades, complete remission and treatment of MS remain a matter of concern. Nanotechnology is a potential approach for manipulating the properties of materials at the molecular level to attain desired properties. This approach is effective in the treatment of several CNS disorders by enhancing drug delivery, bioavailability and efficacy. We have briefly discussed the neuroimmunological disorders with a particular emphasis on MS. We also explored nanoengineered drug delivery systems, describing several nano-formulations for the treatment of MS, challenges and future of nanotechnology.
Collapse
Affiliation(s)
- Archna Panghal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-SAS Nagar, Mohali, 160102, India
| | - S J S Flora
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-SAS Nagar, Mohali, 160102, India.
- Era College of Pharmaceutical Sciences, Era Lucknow Medical University, Sarfarajgang, Lucknow, 226002, India.
| |
Collapse
|
2
|
Tang B, Xie X, Lu J, Huang W, Yang J, Tian J, Lei L. Designing biomaterials for the treatment of autoimmune diseases. APPLIED MATERIALS TODAY 2024; 39:102278. [DOI: 10.1016/j.apmt.2024.102278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
3
|
Gao X, Lin X, Wang Q, Chen J. Artemisinins: Promising drug candidates for the treatment of autoimmune diseases. Med Res Rev 2024; 44:867-891. [PMID: 38054758 DOI: 10.1002/med.22001] [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: 09/21/2023] [Revised: 11/02/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Autoimmune diseases are characterized by the immune system's attack on one's own tissues which are highly diverse and diseases differ in severity, causing damage in virtually all human systems including connective tissue (e.g., rheumatoid arthritis), neurological system (e.g., multiple sclerosis) and digestive system (e.g., inflammatory bowel disease). Historically, treatments normally include pain-killing medication, anti-inflammatory drugs, corticosteroids, and immunosuppressant drugs. However, given the above characteristics, treatment of autoimmune diseases has always been a challenge. Artemisinin is a natural sesquiterpene lactone initially extracted and separated from Chinese medicine Artemisia annua L., which has a long history of curing malaria. Artemisinin's derivatives such as artesunate, dihydroartemisinin, artemether, artemisitene, and so forth, are a family of artemisinins with antimalarial activity. Over the past decades, accumulating evidence have indicated the promising therapeutic potential of artemisinins in autoimmune diseases. Herein, we systematically summarized the research regarding the immunoregulatory properties of artemisinins including artemisinin and its derivatives, discussing their potential therapeutic viability toward major autoimmune diseases and the underlying mechanisms. This review will provide new directions for basic research and clinical translational medicine of artemisinins.
Collapse
Affiliation(s)
- Xu Gao
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Xian Lin
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Jian Chen
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| |
Collapse
|
4
|
Leon MM, Maștaleru A, Oancea A, Alexa-Stratulat T, Peptu CA, Tamba BI, Harabagiu V, Grosu C, Alexa AI, Cojocaru E. Lidocaine-Liposomes-A Promising Frontier for Transdermal Pain Management. J Clin Med 2024; 13:271. [PMID: 38202278 PMCID: PMC10779996 DOI: 10.3390/jcm13010271] [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: 11/15/2023] [Revised: 12/16/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
(1) Background: We aim to develop novel gel formulations for transdermal drug delivery systems in acute and inflammatory pain therapy. (2) Methods: We induced inflammation by the injection of λ-carrageenan on the hind paw of 80 Wistar male rats. The animals were randomized into eight groups of 10 rats each: C (placebo gel), E (EMLATM), L (lidocaine 2%), L-CD (lidocaine + cyclodextrin 2.5%), L-LP (lidocaine + liposomes 1.7%), L-CS (lidocaine + chitosan 4%), L-CSh (lidocaine + chitosan hydrochloride), and L-CS-LP (lidocaine + chitosan + liposomes). The behavior response was determined with a hot plate, cold plate, and algesimeter, each being performed at 30, 60, 120, 180, and 240 min after pain induction. At the end of the experiment, tissue samples were collected for histological assessment. (3) Results: L-LP had the greatest anesthetic effects, which was proven on the cold plate test compared to placebo and EMLATM (all p ≤ 0.001). L-CS-LP had a significant effect on cold plate evaluation compared to placebo (p ≤ 0.001) and on hot plate evaluation compared to EMLATM (p = 0.018). (4) Conclusions: L-LP is a new substance with a substantial analgesic effect demonstrated by the cold plate in the first 120 min. Further studies with more animals are needed to determine the maximum doses that can be applied for a better analgesia with minimum side effects.
Collapse
Affiliation(s)
- Maria Magdalena Leon
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaşi, Romania;
| | - Alexandra Maștaleru
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaşi, Romania;
| | - Andra Oancea
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaşi, Romania;
| | - Teodora Alexa-Stratulat
- Department of Medical Oncology–Radiotherapy, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaşi, Romania;
| | - Cătălina Anișoara Peptu
- Department of Natural and Synthetic Polymers, Faculty of Chemical Engineering and Environmental Protection, “Gheorghe Asachi” Technical University of Iasi, 700050 Iasi, Romania;
| | - Bogdan-Ionel Tamba
- CEMEX Laboratory, “Grigore T. Popa” University of Medicine and Pharmacy, 700259 Iaşi, Romania;
| | - Valeria Harabagiu
- “Petru Poni” Institute of Macromolecular Chemistry, 700487 Iaşi, Romania;
| | - Cristina Grosu
- Department of Medical Specialties III, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaşi, Romania;
| | - Anisia Iuliana Alexa
- Department of Surgery II, Discipline of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Elena Cojocaru
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaşi, Romania;
| |
Collapse
|
5
|
Mwema A, Muccioli GG, des Rieux A. Innovative drug delivery strategies to the CNS for the treatment of multiple sclerosis. J Control Release 2023; 364:435-457. [PMID: 37926243 DOI: 10.1016/j.jconrel.2023.10.052] [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: 07/05/2023] [Revised: 10/05/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Disorders of the central nervous system (CNS), such as multiple sclerosis (MS) represent a great emotional, financial and social burden. Despite intense efforts, great unmet medical needs remain in that field. MS is an autoimmune, chronic inflammatory demyelinating disease with no curative treatment up to date. The current therapies mostly act in the periphery and seek to modulate aberrant immune responses as well as slow down the progression of the disease. Some of these therapies are associated with adverse effects related partly to their administration route and show some limitations due to their rapid clearance and inability to reach the CNS. The scientific community have recently focused their research on developing MS therapies targeting different processes within the CNS. However, delivery of therapeutics to the CNS is mainly limited by the presence of the blood-brain barrier (BBB). Therefore, there is a pressing need to develop new drug delivery strategies that ensure CNS availability to capitalize on identified therapeutic targets. Several approaches have been developed to overcome or bypass the BBB and increase delivery of therapeutics to the CNS. Among these strategies, the use of alternative routes of administration, such as the nose-to-brain (N2B) pathway, offers a promising non-invasive option in the scope of MS, as it would allow a direct transport of the drugs from the nasal cavity to the brain. Moreover, the combination of bioactive molecules within nanocarriers bring forth new opportunities for MS therapies, allowing and/or increasing their transport to the CNS. Here we will review and discuss these alternative administration routes as well as the nanocarrier approaches useful to deliver drugs for MS.
Collapse
Affiliation(s)
- Ariane Mwema
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium; Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium
| | - Giulio G Muccioli
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium.
| | - Anne des Rieux
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium.
| |
Collapse
|
6
|
Yathavan B, Ellis A, Jedrzkiewicz J, Subrahmanyam N, Khurana N, Pulsipher A, Alt JA, Ghandehari H. Systemic administration of budesonide in pegylated liposomes for improved efficacy in chronic rhinosinusitis. J Control Release 2023; 360:274-284. [PMID: 37353160 PMCID: PMC11227746 DOI: 10.1016/j.jconrel.2023.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/18/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Chronic rhinosinusitis (CRS) is a chronic inflammatory condition affecting the nasal and paranasal sinuses of approximately 11.5% of the United States adult population. Oral corticosteroids are effective in controlling sinonasal inflammation in CRS, but the associated adverse effects limit their clinical use. Topical budesonide has demonstrated clinical efficacy in patients with CRS. Herein, we investigated the systemic delivery of liposomes tethered with poly(ethylene glycol) (PEG) and loaded with budesonide in a murine model of CRS. PEGylated liposomes encapsulated with budesonide phosphate (L-BudP) were administered via tail vein injection, and the feasibility of L-BudP to reduce sinonasal inflammation was compared to that of free budesonide phosphate (F-BudP) and topical budesonide phosphate (T-BudP) treatment over a 14-day study period. Compared to a single injection of F-BudP and repeat T-BudP administration, a single injection of L-BudP demonstrated increased and prolonged efficacy, resulting in the significant improvement of sinonasal tissue histopathological scores (p < 0.05) with decreased immune cell infiltration (p < 0.05). Toxicities associated with L-BudP and T-BudP treatment, assessed via body and organ weight, as well as peripheral blood liver enzyme and differential white blood cell analyses, were transient and comparable. These data suggest that systemic liposomal budesonide treatment results in improved efficacy over topical treatment.
Collapse
Affiliation(s)
- Bhuvanesh Yathavan
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexa Ellis
- College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Nithya Subrahmanyam
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Nitish Khurana
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Abigail Pulsipher
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA; Department of Otolaryngology - Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA.
| | - Jeremiah A Alt
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA; Department of Otolaryngology - Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Hamidreza Ghandehari
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA; Department of Otolaryngology - Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
7
|
Demirdöğen BC. Theranostic potential of graphene quantum dots for multiple sclerosis. Mult Scler Relat Disord 2022; 68:104232. [PMID: 36244187 DOI: 10.1016/j.msard.2022.104232] [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: 08/23/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022]
Abstract
Nanomedicine offers great promise to solve healthcare problems using nanotechnology. Theranostics provide imaging/diagnosis and therapy simultaneously. Novel agents that target both the neuroinflammation and neurodegeneration component of multiple sclerosis (MS) are required. Progress has been achieved in developing smart, surface decorated nanoparticles that effectively transport the therapeutic drug into the central nervous system (CNS). Graphene quantum dots (GQDs) can be traced in vivo by fluorescence imaging due to their unique optical properties. They can also traverse the blood-brain barrier (BBB) and deliver drugs into the CNS. Moreover, GQDs have low cytotoxicity and higher biocompatibility. Therefore, GQDs can be utilized to design novel multifunctional nanocarrier theranostic tools for MS.
Collapse
Affiliation(s)
- Birsen Can Demirdöğen
- Department of Biomedical Engineering, TOBB University of Economics and Technology, Söğütözü, Ankara 06560, Turkey.
| |
Collapse
|
8
|
Rhaman MM, Islam MR, Akash S, Mim M, Noor alam M, Nepovimova E, Valis M, Kuca K, Sharma R. Exploring the role of nanomedicines for the therapeutic approach of central nervous system dysfunction: At a glance. Front Cell Dev Biol 2022; 10:989471. [PMID: 36120565 PMCID: PMC9478743 DOI: 10.3389/fcell.2022.989471] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022] Open
Abstract
In recent decades, research scientists, molecular biologists, and pharmacologists have placed a strong emphasis on cutting-edge nanostructured materials technologies to increase medicine delivery to the central nervous system (CNS). The application of nanoscience for the treatment of neurodegenerative diseases (NDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), Huntington’s disease (HD), brain cancer, and hemorrhage has the potential to transform care. Multiple studies have indicated that nanomaterials can be used to successfully treat CNS disorders in the case of neurodegeneration. Nanomedicine development for the cure of degenerative and inflammatory diseases of the nervous system is critical. Nanoparticles may act as a drug transporter that can precisely target sick brain sub-regions, boosting therapy success. It is important to develop strategies that can penetrate the blood–brain barrier (BBB) and improve the effectiveness of medications. One of the probable tactics is the use of different nanoscale materials. These nano-based pharmaceuticals offer low toxicity, tailored delivery, high stability, and drug loading capacity. They may also increase therapeutic effectiveness. A few examples of the many different kinds and forms of nanomaterials that have been widely employed to treat neurological diseases include quantum dots, dendrimers, metallic nanoparticles, polymeric nanoparticles, carbon nanotubes, liposomes, and micelles. These unique qualities, including sensitivity, selectivity, and ability to traverse the BBB when employed in nano-sized particles, make these nanoparticles useful for imaging studies and treatment of NDs. Multifunctional nanoparticles carrying pharmacological medications serve two purposes: they improve medication distribution while also enabling cell dynamics imaging and pharmacokinetic study. However, because of the potential for wide-ranging clinical implications, safety concerns persist, limiting any potential for translation. The evidence for using nanotechnology to create drug delivery systems that could pass across the BBB and deliver therapeutic chemicals to CNS was examined in this study.
Collapse
Affiliation(s)
- Md. Mominur Rhaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mobasharah Mim
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Noor alam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Martin Valis
- Department of Neurology, Charles University in Prague, Faculty of Medicine in Hradec Králové and University Hospital, Hradec Králové, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| |
Collapse
|
9
|
van Schaik PEM, Zuhorn IS, Baron W. Targeting Fibronectin to Overcome Remyelination Failure in Multiple Sclerosis: The Need for Brain- and Lesion-Targeted Drug Delivery. Int J Mol Sci 2022; 23:8418. [PMID: 35955549 PMCID: PMC9368816 DOI: 10.3390/ijms23158418] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is a neuroinflammatory and neurodegenerative disease with unknown etiology that can be characterized by the presence of demyelinated lesions. Prevailing treatment protocols in MS rely on the modulation of the inflammatory process but do not impact disease progression. Remyelination is an essential factor for both axonal survival and functional neurological recovery but is often insufficient. The extracellular matrix protein fibronectin contributes to the inhibitory environment created in MS lesions and likely plays a causative role in remyelination failure. The presence of the blood-brain barrier (BBB) hinders the delivery of remyelination therapeutics to lesions. Therefore, therapeutic interventions to normalize the pathogenic MS lesion environment need to be able to cross the BBB. In this review, we outline the multifaceted roles of fibronectin in MS pathogenesis and discuss promising therapeutic targets and agents to overcome fibronectin-mediated inhibition of remyelination. In addition, to pave the way for clinical use, we reflect on opportunities to deliver MS therapeutics to lesions through the utilization of nanomedicine and discuss strategies to deliver fibronectin-directed therapeutics across the BBB. The use of well-designed nanocarriers with appropriate surface functionalization to cross the BBB and target the lesion sites is recommended.
Collapse
Affiliation(s)
- Pauline E. M. van Schaik
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| | - Inge S. Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Wia Baron
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| |
Collapse
|
10
|
Jan Z, Mollazadeh S, Abnous K, Taghdisi SM, Danesh A, Ramezani M, Alibolandi M. Targeted Delivery Platforms for the Treatment of Multiple Sclerosis. Mol Pharm 2022; 19:1952-1976. [PMID: 35501974 DOI: 10.1021/acs.molpharmaceut.1c00892] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is a neurodegenerative condition of the central nervous system (CNS) that presents with varying levels of disability in patients, displaying the significance of timely and effective management of this complication. Though several treatments have been developed to protect nerves, comprehensive improvement of MS is still considered an essential bottleneck. Therefore, the development of innovative treatment methods for MS is one of the core research areas. In this regard, nanoscale platforms can offer practical and ideal approaches to the diagnosis and treatment of various diseases, especially immunological disorders such as MS, to improve the effectiveness of conventional therapies. It should be noted that there is significant progress in the development of neuroprotective strategies through the implementation of various nanoparticles, monoclonal antibodies, peptides, and aptamers. In this study, we summarize different particle systems as well as targeted therapies, such as antibodies, peptides, nucleic acids, and engineered cells for the treatment of MS, and discuss their potential in the treatment of MS in the preclinical and clinical stages. Future advances in targeted delivery of medical supplies may offer new strategies for complete recovery as well as practical treatment of progressive forms of MS.
Collapse
Affiliation(s)
- Zeinab Jan
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Science, 7GJP+VPQ Mashhad, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, F82C+G8V Bojnurd, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, 7GJP+VPQ Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, 7GJP+VPQ Mashhad, Iran
| | - Abolghasem Danesh
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Science, 7GJP+VPQ Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, 7GJP+VPQ Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, 7GJP+VPQ Mashhad, Iran
| |
Collapse
|
11
|
Lin DW, Chang CC, Hsu YC, Lin CL. New Insights into the Treatment of Glomerular Diseases: When Mechanisms Become Vivid. Int J Mol Sci 2022; 23:3525. [PMID: 35408886 PMCID: PMC8998908 DOI: 10.3390/ijms23073525] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/23/2022] Open
Abstract
Treatment for glomerular diseases has been extrapolated from the experience of other autoimmune disorders while the underlying pathogenic mechanisms were still not well understood. As the classification of glomerular diseases was based on patterns of juries instead of mechanisms, treatments were typically the art of try and error. With the advancement of molecular biology, the role of the immune agent in glomerular diseases is becoming more evident. The four-hit theory based on the discovery of gd-IgA1 gives a more transparent outline of the pathogenesis of IgA nephropathy (IgAN), and dysregulation of Treg plays a crucial role in the pathogenesis of minimal change disease (MCD). An epoch-making breakthrough is the discovery of PLA2R antibodies in the primary membranous nephropathy (pMN). This is the first biomarker applied for precision medicine in kidney disease. Understanding the immune system's role in glomerular diseases allows the use of various immunosuppressants or other novel treatments, such as complement inhibitors, to treat glomerular diseases more reasonable. In this era of advocating personalized medicine, it is inevitable to develop precision medicine with mechanism-based novel biomarkers and novel therapies in kidney disease.
Collapse
Affiliation(s)
- Da-Wei Lin
- Department of Internal Medicine, St. Martin De Porres Hospital, Chiayi 60069, Taiwan;
| | - Cheng-Chih Chang
- Department of Surgery, Chang Gung Memorial Hospital, Chiayi 613016, Taiwan;
| | - Yung-Chien Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613016, Taiwan
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613016, Taiwan
| | - Chun-Liang Lin
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613016, Taiwan
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613016, Taiwan
- Division of Chinese Materia Medica Development, National Research Institute of Chinese Medicine, Taipei 613016, Taiwan
- Kidney Research Center, Chang Gung Memorial Hospital, Taipei 613016, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Chang Gung Memorial Hospital, Kaohsiung 833253, Taiwan
| |
Collapse
|
12
|
Xie Y, Papadopoulou P, de Wit B, d’Engelbronner JC, van Hage P, Kros A, Schaaf MJM. Two Types of Liposomal Formulations Improve the Therapeutic Ratio of Prednisolone Phosphate in a Zebrafish Model for Inflammation. Cells 2022; 11:cells11040671. [PMID: 35203318 PMCID: PMC8870436 DOI: 10.3390/cells11040671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/31/2022] [Accepted: 02/12/2022] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoids (GCs) are effective anti-inflammatory drugs, but their clinical use is limited by their side effects. Using liposomes to target GCs to inflammatory sites is a promising approach to improve their therapeutic ratio. We used zebrafish embryos to visualize the biodistribution of liposomes and to determine the anti-inflammatory and adverse effects of the GC prednisolone phosphate (PLP) encapsulated in these liposomes. Our results showed that PEGylated liposomes remained in circulation for long periods of time, whereas a novel type of liposomes (which we named AmbiMACs) selectively targeted macrophages. Upon laser wounding of the tail, both types of liposomes were shown to accumulate near the wounding site. Encapsulation of PLP in the PEGylated liposomes and AmbiMACs increased its potency to inhibit the inflammatory response. However, encapsulation of PLP in either type of liposome reduced its inhibitory effect on tissue regeneration, and encapsulation in PEGylated liposomes attenuated the activation of glucocorticoid-responsive gene expression throughout the body. Thus, by exploiting the unique possibilities of the zebrafish animal model to study the biodistribution as well as the anti-inflammatory and adverse effects of liposomal formulations of PLP, we showed that PEGylated liposomes and AmbiMACs increase the therapeutic ratio of this GC drug.
Collapse
Affiliation(s)
- Yufei Xie
- Institute of Biology, Leiden University, 2333 CC Leiden, The Netherlands; (Y.X.); (B.d.W.); (J.C.d.); (P.v.H.)
| | - Panagiota Papadopoulou
- Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands; (P.P.); (A.K.)
| | - Björn de Wit
- Institute of Biology, Leiden University, 2333 CC Leiden, The Netherlands; (Y.X.); (B.d.W.); (J.C.d.); (P.v.H.)
| | - Jan C. d’Engelbronner
- Institute of Biology, Leiden University, 2333 CC Leiden, The Netherlands; (Y.X.); (B.d.W.); (J.C.d.); (P.v.H.)
| | - Patrick van Hage
- Institute of Biology, Leiden University, 2333 CC Leiden, The Netherlands; (Y.X.); (B.d.W.); (J.C.d.); (P.v.H.)
| | - Alexander Kros
- Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands; (P.P.); (A.K.)
| | - Marcel J. M. Schaaf
- Institute of Biology, Leiden University, 2333 CC Leiden, The Netherlands; (Y.X.); (B.d.W.); (J.C.d.); (P.v.H.)
- Correspondence: ; Tel.: +31-715274975; Fax: +31-715275088
| |
Collapse
|
13
|
Rahiman N, Zamani P, Badiee A, Arabi L, Alavizadeh SH, Jaafari MR. An insight into the role of liposomal therapeutics in the reversion of Multiple Sclerosis. Expert Opin Drug Deliv 2021; 18:1795-1813. [PMID: 34747298 DOI: 10.1080/17425247.2021.2003327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Multiple Sclerosis (MS), as an autoimmune disease, has complicated immunopathology, which makes its management relevant to various factors. Novel pharmaceutical vehicles, especially liposomes, can support efficacious handling of this disease both in early detection and prognosis and also in a therapeutic manner. The most well-known trigger of MS onset is the predominance of cellular to humoral immunity and enhancement of inflammatory cytokines level. The installation of liposomes as nanoparticles to control this disease holds great promise up to now. AREAS COVERED Various types of liposomes with different properties and purposes have been formulated and targeted immune cells with their surface manipulations. They may be encapsulated with anti-inflammatory, MS-related therapeutics, or immunodominant myelin-specific peptides for attaining a higher therapeutic efficacy of the drugs or tolerance induction. Cationic liposomes are also highly applicable for gene delivery of the anti-inflammatory cytokines or silencing the inflammatory cytokines. Liposomes have also been used as biotools for comprehending MS pathomechanisms or as diagnostic agents. EXPERT OPINION The efforts to manage MS through nanomedicine, especially liposomal therapeutics, pave a new avenue to a high-throughput medication of this autoimmune disease and their translation to the clinic in the future for overcoming the challenges that MS patients confront.
Collapse
Affiliation(s)
- Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
Macrophages in heterotopic ossification: from mechanisms to therapy. NPJ Regen Med 2021; 6:70. [PMID: 34702860 PMCID: PMC8548514 DOI: 10.1038/s41536-021-00178-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 09/30/2021] [Indexed: 01/04/2023] Open
Abstract
Heterotopic ossification (HO) is the formation of extraskeletal bone in non-osseous tissues. It is caused by an injury that stimulates abnormal tissue healing and regeneration, and inflammation is involved in this process. It is worth noting that macrophages are crucial mediators of inflammation. In this regard, abundant macrophages are recruited to the HO site and contribute to HO progression. Macrophages can acquire different functional phenotypes and promote mesenchymal stem cell (MSC) osteogenic differentiation, chondrogenic differentiation, and angiogenesis by expressing cytokines and other factors such as the transforming growth factor-β1 (TGF-β1), bone morphogenetic protein (BMP), activin A (Act A), oncostatin M (OSM), substance P (SP), neurotrophin-3 (NT-3), and vascular endothelial growth factor (VEGF). In addition, macrophages significantly contribute to the hypoxic microenvironment, which primarily drives HO progression. Thus, these have led to an interest in the role of macrophages in HO by exploring whether HO is a "butterfly effect" event. Heterogeneous macrophages are regarded as the "butterflies" that drive a sequence of events and ultimately promote HO. In this review, we discuss how the recruitment of macrophages contributes to HO progression. In particular, we review the molecular mechanisms through which macrophages participate in MSC osteogenic differentiation, angiogenesis, and the hypoxic microenvironment. Understanding the diverse role of macrophages may unveil potential targets for the prevention and treatment of HO.
Collapse
|
15
|
Sidoryk-Węgrzynowicz M, Dąbrowska-Bouta B, Sulkowski G, Strużyńska L. Nanosystems and exosomes as future approaches in treating multiple sclerosis. Eur J Neurosci 2021; 54:7377-7404. [PMID: 34561918 DOI: 10.1111/ejn.15478] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system which leads to neurological dysfunctions and severe disabilities. MS pathology is characterised by damage of the blood-brain barrier and infiltration of autoreactive T cells that overactivate glial cells, thereby initiating neuroinflammation accompanied by the formation of demyelinating plaques and neurodegeneration. Clinical deficits in this multifactorial disease depend on the progression of myelin loss, the stage of inflammation, the status of axons and the activity of oligodendrocyte precursor cells (OPCs). Despite significant progress in the treatment of MS, current therapies remain limited and new approaches are highly desirable. Nanosystems based on liposomes and nanoparticles are among some of the more noteworthy therapeutic strategies being investigated. Applications of nanosystems alone or as drug carriers in animal models of MS have been found to successfully alleviate the symptoms of the disease and exert anti-inflammatory potential. Exosomes are a specific type of nanosystem based on nanometre-sized extracellular vesicles released by different cells which exhibit important healing features. Exosomes contain an array of anti-inflammatory and neuroprotective agents which may contribute to modulation of the immune system as well as promoting remyelination and tissue repair. In this review, opportunities to use nanosystems against progression of MS will be discussed in context of cell-specific pathologies associated with MS.
Collapse
Affiliation(s)
- Marta Sidoryk-Węgrzynowicz
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Dąbrowska-Bouta
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Grzegorz Sulkowski
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
16
|
Mahadik N, Bhattacharya D, Padmanabhan A, Sakhare K, Narayan KP, Banerjee R. Targeting steroid hormone receptors for anti-cancer therapy-A review on small molecules and nanotherapeutic approaches. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1755. [PMID: 34541822 DOI: 10.1002/wnan.1755] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
The steroid hormone receptors (SHRs) among nuclear hormone receptors (NHRs) are steroid ligand-dependent transcription factors that play important roles in the regulation of transcription of genes promoted via hormone responsive elements in our genome. Aberrant expression patterns and context-specific regulation of these receptors in cancer, have been routinely reported by multiple research groups. These gave an window of opportunity to target those receptors in the context of developing novel, targeted anticancer therapeutics. Besides the development of a plethora of SHR-targeting synthetic ligands and the availability of their natural, hormonal ligands, development of many SHR-targeted, anticancer nano-delivery systems and theranostics, especially based on small molecules, have been reported. It is intriguing to realize that these cytoplasmic receptors have become a hot target for cancer selective delivery. This is in spite of the fact that these receptors do not fall in the category of conventional, targetable cell surface bound or transmembrane receptors that enjoy over-expression status. Glucocorticoid receptor (GR) is one such exciting SHR that in spite of it being expressed ubiquitously in all cells, we discovered it to behave differently in cancer cells, thus making it a truly druggable target for treating cancer. This review selectively accumulates the knowledge generated in the field of SHR-targeting as a major focus for cancer treatment with various anticancer small molecules and nanotherapeutics on progesterone receptor, mineralocorticoid receptor, and androgen receptor while selectively emphasizing on GR and estrogen receptor. This review also briefly highlights lipid-modification strategy to convert ligands into SHR-targeted cancer nanotherapeutics. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Namita Mahadik
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Dwaipayan Bhattacharya
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Akshaya Padmanabhan
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Kalyani Sakhare
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
17
|
Nuzzo D, Picone P. Multiple Sclerosis: Focus on Extracellular and Artificial Vesicles, Nanoparticles as Potential Therapeutic Approaches. Int J Mol Sci 2021; 22:8866. [PMID: 34445572 PMCID: PMC8396338 DOI: 10.3390/ijms22168866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/02/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the Central Nervous System, characterized by an inflammatory process leading to the destruction of myelin with neuronal death and neurodegeneration. In MS, lymphocytes cross the blood-brain barrier, creating inflammatory demyelinated plaques located primarily in the white matter. MS potential treatments involve various mechanisms of action on immune cells, immunosuppression, inhibition of the passage through the blood-brain barrier, and immunotolerance. Bio-nanotechnology represents a promising approach to improve the treatment of autoimmune diseases by its ability to affect the immune responses. The use of nanotechnology has been actively investigated for the development of new MS therapies. In this review, we summarize the results of the studies on natural and artificial vesicles and nanoparticles, and take a look to the future clinical perspectives for their application in the MS therapy.
Collapse
Affiliation(s)
- Domenico Nuzzo
- Istituto per la Ricerca e l’Innovazione Biomedica, CNR, via U. La Malfa 153, 90146 Palermo, Italy;
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università di Palermo, Viale delle Scienze, 90128 Palermo, Italy
| | - Pasquale Picone
- Istituto per la Ricerca e l’Innovazione Biomedica, CNR, via U. La Malfa 153, 90146 Palermo, Italy;
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università di Palermo, Viale delle Scienze, 90128 Palermo, Italy
| |
Collapse
|
18
|
Liaw J, Hsieh WH, Chiou SH, Huang YS, Chang SF. Assessment of the Oral Delivery of a Myelin Basic Protein Gene Promoter with Antiapoptotic bcl-x L (pMBP-bcl-x L) DNA by Cyclic Peptide Nanotubes with Two Aspect Ratios and Its Biodistribution in the Brain and Spinal Cord. Mol Pharm 2021; 18:2556-2573. [PMID: 34110176 DOI: 10.1021/acs.molpharmaceut.1c00057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cyclo-(D-Trp-Tyr) peptide nanotubes (PNTs) were reported to be potential carriers for oral gene delivery in our previous study; however, the effect of the aspect ratio (AR) of these PNTs on gene delivery in vivo could affect penetration or interception in biological environments. The aim of this study was to assess the feasibility of cyclo-(D-Trp-Tyr) PNTs with two ARs as carriers for oral pMBP-bcl-xL-hRluc delivery to the spinal cord to treat spinal cord injury (SCI). We evaluated the biodistribution of oligodendrocyte (OLG)-specific myelin basic protein gene promoter-driven antiapoptotic DNA (pMBP-bcl-xL) to the brain and spinal cord delivered with cyclo-(D-Trp-Tyr) PNTs with large (L) and small (S) PNTs with two ARs. After complex formation, the length, width, and AR of the L-PNTs/DNA were 77.86 ± 3.30, 6.51 ± 0.28, and 13.75 ± 7.29 μm, respectively, and the length and width of the S-PNTs/DNA were 1.17 ± 0.52 and 0.17 ± 0.05 μm, respectively, giving an AR of 7.12 ± 3.17 as detected by scanning electron microscopy. Each of these three parameters exhibited significant differences (p < 0.05) between L-PNTs/DNA and S-PNTs/DNA. However, there were no significant differences (p > 0.05) between the L-PNTs and S-PNTs for either their DNA encapsulation efficiency (29.72 ± 14.19 and 34.31 ± 16.78%, respectively) or loading efficiency (5.15 ± 2.58 and 5.95 ± 2.91%). The results of the in vitro analysis showed that the S-PNT/DNA complexes had a significantly higher DNA release rate and DNA permeation in the duodenum than the L-PNT/DNA complexes. Using Cy5 and TM-rhodamine to individually and chemically conjugate the PNTs with plasmid DNA, we observed, using laser confocal microscopy, that the PNTs and DNA colocalized in complexes. We further confirmed the complexation between DNA and the PNTs using fluorescence resonance energy transfer (FRET). Data from an in vivo imaging system (IVIS) showed that there was no significant difference (p > 0.05) in PNT distribution between L-PNTs/DNA and S-PNTs/DNA within 4 h. However, the S-PNT/DNA group had a significantly higher DNA distribution (p < 0.05) in several organs, including the ilium, heart, lungs, spleen, kidneys, testes, brain, and spinal cord. Finally, we determined the bcl-xL protein expression levels in the brain and spinal cord regions for the L-PNT/DNA and S-PNT/DNA complex formulations. These results suggested that either L-PNTs or S-PNTs may be used as potential carriers for oral gene delivery to treat SCI.
Collapse
Affiliation(s)
- Jiahorng Liaw
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Wei-Hsien Hsieh
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
| | - Shih-Hsun Chiou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Shan Huang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Shwu-Fen Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 111, Taiwan
| |
Collapse
|
19
|
Shimizu K, Agata K, Takasugi S, Goto S, Narita Y, Asai T, Magata Y, Oku N. New strategy for MS treatment with autoantigen-modified liposomes and their therapeutic effect. J Control Release 2021; 335:389-397. [PMID: 34033858 DOI: 10.1016/j.jconrel.2021.05.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023]
Abstract
As current treatments for multiple sclerosis (MS) remain chemotherapeutic ones directed toward symptoms, the development of a curative treatment is urgently required. Herein, we show an autoreactive immune cell-targetable approach using autoantigen-modified liposomes for the curative treatment of MS. In these experiments, experimental autoimmune encephalomyelitis (EAE) induced by autoantigenic myelin oligodendrocyte glycoprotein (MOG) peptide was used as a model of primary progressive MS, and MOG-modified liposomes encapsulating doxorubicin (MOG-LipDOX) were used as a therapeutic drug. The results showed that the progression of encephalomyelitis symptoms was significantly suppressed by MOG-LipDOX injection, whereas the other samples failed to show any effect. Additionally, invasion of inflammatory immune cells into the spinal cord and demyelination of neurons were clearly suppressed in the MOG-LipDOX-treated mice. FACS analysis revealed that the number of both MOG-recognizable CD4+ T cells in the spleen was obviously decreased after MOG-LipDOX treatment. Furthermore, the number of effector Th17 cells in the spleen was significantly decreased and that of regulatory Treg cells was concomitantly increased. Finally, we demonstrated that myelin proteolipid protein (PLP)-modified liposomes encapsulating DOX (PLP-LipDOX) also showed the therapeutic effect on relapsing-remitting EAE. These findings indicate that autoantigen-modified liposomal drug produced a highly therapeutic effect on EAE by delivering the encapsulated drug to autoantigen-recognizable CD4+ T cells and thus suppressing autoreactive immune responses. The present study suggests that the use of these autoantigen-modified liposomes promises to be a suitable therapeutic approach for the cure of MS.
Collapse
Affiliation(s)
- Kosuke Shimizu
- Department of Molecular Imaging, Institute of Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu City, Shizuoka 431-3192, Japan; Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan.
| | - Kazuki Agata
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Shohei Takasugi
- Department of Molecular Imaging, Institute of Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu City, Shizuoka 431-3192, Japan; Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Shungo Goto
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Yudai Narita
- Department of Molecular Imaging, Institute of Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu City, Shizuoka 431-3192, Japan; Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Yasuhiro Magata
- Department of Molecular Imaging, Institute of Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu City, Shizuoka 431-3192, Japan
| | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan; Faculty of Pharma-Science, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| |
Collapse
|
20
|
Youshia J, Ali ME, Stein V, Lamprecht A. Nanoparticles' properties modify cell type-dependent distribution in immune cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102244. [PMID: 32561256 DOI: 10.1016/j.nano.2020.102244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/25/2020] [Accepted: 06/08/2020] [Indexed: 10/24/2022]
Abstract
Polymeric nanoparticles can passively target inflamed tissues. How their physicochemical properties affect their distribution pattern among the infiltrating immune cells is unknown. Polyvinyl acetate nanoparticles with different particle size (100 and 300 nm) and surface charge (cationic, non-ionic, and anionic) were prepared and incubated with either LPS-activated or unactivated murine splenocytes. Nanoparticle association with macrophages, dendritic cells, neutrophils, B and T cells was investigated using flow cytometry. Cells associated with nanoparticles as follows: cationic>anionic>non-ionic and 300 nm > 100 nm. 40% of ionic nanoparticles were distributed among unactivated macrophages, reduced to 25% for activated macrophages. 60% of 100 nm and 40% of 300 nm non-ionic nanoparticles were distributed among unactivated and LPS-activated macrophages. This study highlights that particles' physicochemical properties impact the number of nanoparticles associating with immune cells more than their distribution pattern, which is principally determined by the cell activation state. This suggests a disease-dependent distribution pattern for therapeutic nanoparticles.
Collapse
Affiliation(s)
- John Youshia
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed Ehab Ali
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Valentin Stein
- Institute of Physiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; PEPITE EA4267, University of Bourgogne Franche-Comté, Besançon, France.
| |
Collapse
|
21
|
Naqvi S, Panghal A, Flora SJS. Nanotechnology: A Promising Approach for Delivery of Neuroprotective Drugs. Front Neurosci 2020; 14:494. [PMID: 32581676 PMCID: PMC7297271 DOI: 10.3389/fnins.2020.00494] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) disorders especially neurodegenerative disorders are the major challenge for public health and demand the great attention of researchers to protect people against them. In past few decades, different treatment strategies have been adopted, but their therapeutic efficacy are not enough and have only shown partial mitigation of symptoms. Blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BSCFB) guard the CNS from harmful substances and pose as the major challenges in delivering drugs into CNS for treatment of CNS complications such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), stroke, epilepsy, brain tumors, multiple sclerosis (MS), and encephalitis, etc. Nanotechnology has come out as an exciting and promising new platform of treating neurological disorders and has shown great potential to overcome problems related to the conventional treatment approaches. Molecules can be nanoengineered to carry out multiple specific functions such as to cross the BBB, target specific cell or signaling pathway, respond to endogenous stimuli, and act as a vehicle for gene delivery, support nerve regeneration and cell survival. In present review, the role of nanocarrier systems such as liposomes, micelles, solid lipid nanoparticles (SLNPs), dendrimers, and nanoemulsions for delivery of various neurotherapeutic agents has been discussed, besides this, their mechanism of action, and nanoformulation of different neuroprotective agents like curcumin, edaravone, nerve growth factors in CNS disorders like Alzheimer’s, Parkinsonism, epilepsy, stroke, and brain tumors has been reviewed.
Collapse
Affiliation(s)
- Saba Naqvi
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - Archna Panghal
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - S J S Flora
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, India
| |
Collapse
|
22
|
Promising Nanotechnology Approaches in Treatment of Autoimmune Diseases of Central Nervous System. Brain Sci 2020; 10:brainsci10060338. [PMID: 32498357 PMCID: PMC7349417 DOI: 10.3390/brainsci10060338] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune, neurodegenerative disease of the central nervous system (CNS) that yields to neuronal axon damage, demyelization, and paralysis. Although several drugs were designed for the treatment of MS, with some of them being approved in the last few decades, the complete remission and the treatment of progressive forms still remain a matter of debate and a medical challenge. Nanotechnology provides a variety of promising therapeutic tools that can be applied for the treatment of MS, overcoming the barriers and the limitations of the already existing immunosuppressive and biological therapies. In the present review, we explore literature case studies on the development of drug delivery nanosystems for the targeted delivery of MS drugs in the pathological tissues of the CNS, providing high bioavailability and enhanced therapeutic efficiency, as well as nanosystems for the delivery of agents to facilitate efficient remyelination. Moreover, we present examples of tolerance-inducing nanocarriers, being used as promising vaccines for antigen-specific immunotherapy of MS. We emphasize on liposomes, as well as lipid- and polymer-based nanoparticles. Finally, we highlight the future perspectives given by the nanotechnology field toward the improvement of the current treatment of MS and its animal model, experimental autoimmune encephalomyelitis (EAE).
Collapse
|
23
|
Kaiser TK, Li H, Roßmann L, Reichardt SD, Bohnenberger H, Feldmann C, Reichardt HM. Glucocorticoids delivered by inorganic-organic hybrid nanoparticles mitigate acute graft-versus-host disease and sustain graft-versus-leukemia activity. Eur J Immunol 2020; 50:1220-1233. [PMID: 32133644 DOI: 10.1002/eji.201948464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/29/2020] [Accepted: 03/03/2020] [Indexed: 12/19/2022]
Abstract
Glucocorticoids (GCs) are widely used to treat acute graft-versus-host disease (aGvHD) due to their immunosuppressive activity, but they also reduce the beneficial graft-versus-leukemia (GvL) effect of the allogeneic T cells contained in the graft. Here, we tested whether aGvHD therapy could be improved by delivering GCs with the help of inorganic-organic hybrid nanoparticles (IOH-NPs) that preferentially target myeloid cells. IOH-NPs containing the GC betamethasone (BMP-NPs) efficiently reduced morbidity, mortality, and tissue damage in a totally MHC mismatched mouse model of aGvHD. Therapeutic activity was lost in mice lacking the GC receptor (GR) in myeloid cells, confirming the cell type specificity of our approach. BMP-NPs had no relevant systemic activity but suppressed cytokine and chemokine gene expression locally in the small intestine, which presumably explains their mode of action. Most importantly, BMP-NPs delayed the development of an adoptively transferred B cell lymphoma better than the free drug, although the overall incidence was unaffected. Our findings thus suggest that employing IOH-NPs could diminish the risk of relapse associated with GC therapy of aGvHD patients while still allowing to efficiently ameliorate the disease.
Collapse
Affiliation(s)
- Tina K Kaiser
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Hu Li
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Laura Roßmann
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Sybille D Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Claus Feldmann
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Holger M Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
24
|
Highly selective organ distribution and cellular uptake of inorganic-organic hybrid nanoparticles customized for the targeted delivery of glucocorticoids. J Control Release 2020; 319:360-370. [DOI: 10.1016/j.jconrel.2020.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/11/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022]
|
25
|
Jiang S, Prozeller D, Pereira J, Simon J, Han S, Wirsching S, Fichter M, Mottola M, Lieberwirth I, Morsbach S, Mailänder V, Gehring S, Crespy D, Landfester K. Controlling protein interactions in blood for effective liver immunosuppressive therapy by silica nanocapsules. NANOSCALE 2020; 12:2626-2637. [PMID: 31939969 DOI: 10.1039/c9nr09879h] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Immunosuppression with glucocorticoids is a common treatment for autoimmune liver diseases and after liver transplant, which is however associated with severe side-effects. Targeted delivery of glucocorticoids to inflammatory cells, e.g. liver macrophages and Kupffer cells, is a promising approach for minimizing side effects. Herein, we prepare core-shell silica nanocapsules (SiO2 NCs) via a sol-gel process confined in nanodroplets for targeted delivery of dexamethasone (DXM) for liver immunosuppressive therapy. DXM with concentrations up to 100 mg mL-1 in olive oil are encapsulated while encapsulation efficiency remains over 95% after 15 days. Internalization of NCs by non-parenchymal murine liver cells significantly reduces the release of inflammatory cytokines, indicating an effective suppression of inflammatory response of liver macrophages. Fluorescent and magnetic labeling of the NCs allows for monitoring their intracellular trafficking and biodegradation. Controlled interaction with blood proteins and good colloidal stability in blood plasma are achieved via PEGylation of the NCs. Specific proteins responsible for stealth effect, such as apolipoprotein A-I, apolipoprotein A-IV, and clusterin, are present in large amounts on the PEGylated NCs. In vivo biodistribution investigations prove an efficient accumulation of NCs in the liver, underlining the suitability of the SiO2 NCs as a dexamethasone carrier for treating inflammatory liver diseases.
Collapse
Affiliation(s)
- Shuai Jiang
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Domenik Prozeller
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Jorge Pereira
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Johanna Simon
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. and Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Shen Han
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Sebastian Wirsching
- Children's Hospital, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Michael Fichter
- Children's Hospital, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Milagro Mottola
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Ingo Lieberwirth
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. and Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Stephan Gehring
- Children's Hospital, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Daniel Crespy
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. and Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| |
Collapse
|
26
|
Teixeira MI, Lopes CM, Amaral MH, Costa PC. Current insights on lipid nanocarrier-assisted drug delivery in the treatment of neurodegenerative diseases. Eur J Pharm Biopharm 2020; 149:192-217. [PMID: 31982574 DOI: 10.1016/j.ejpb.2020.01.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/16/2019] [Accepted: 01/08/2020] [Indexed: 12/12/2022]
Abstract
The central nervous system (CNS) is vulnerable to pathologic processes that lead to the development of neurodegenerative disorders like Alzheimer's, Parkinson's and Huntington's diseases, Multiple sclerosis or Amyotrophic lateral sclerosis. These are chronic and progressive pathologies characterized by the loss of neurons and the formation of misfolded proteins. Additionally, neurodegenerative diseases are accompanied by a structural and functional dysfunction of the blood-brain barrier (BBB). Although serving as a protection for the CNS, the existence of physiological barriers, especially the BBB, limits the access of several therapeutic agents to the brain, constituting a major hindrance in neurotherapeutics advancement. In this regard, nanotechnology-based approaches have arisen as a promising strategy to not only improve drug targeting to the brain, but also to increase bioavailability. Lipid nanocarriers such as liposomes, solid lipid nanoparticles (SLN), nanostructured lipid carriers (NLC), microemulsions and nanoemulsions, have already proven their potential for enhancing brain transport, crossing more easily into the CNS and allowing the administration of medicines that could benefit the treatment of neurological pathologies. Given the socioeconomic impact of such conditions and the advent of nanotechnology that inevitably leads to more effective and superior therapeutics for their management, it is imperative to constantly update on the current knowledge of these topics. Herein, we provide insight on the BBB and the pathophysiology of the main neurodegenerative disorders. Moreover, this review seeks to highlight the several approaches that can be used to improve the delivery of therapeutic agents to the CNS, while also offering an extensive overview of the latest efforts regarding the use of lipid-based nanocarriers in the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- M I Teixeira
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - C M Lopes
- FP-ENAS/CEBIMED, Fernando Pessoa Energy, Environment and Health Research Unit/Biomedical Research Centre, Faculty of Health Sciences, Fernando Pessoa University, Rua Carlos da Maia, 296, 4200-150 Porto, Portugal
| | - M H Amaral
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - P C Costa
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
27
|
Ghosh S, Lalani R, Patel V, Bhowmick S, Misra A. Surface engineered liposomal delivery of therapeutics across the blood brain barrier: recent advances, challenges and opportunities. Expert Opin Drug Deliv 2019; 16:1287-1311. [DOI: 10.1080/17425247.2019.1676721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Rohan Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Vivek Patel
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Subhas Bhowmick
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
28
|
Targeted Transport as a Promising Method of Drug Delivery to the Central Nervous System (Review). Pharm Chem J 2019. [DOI: 10.1007/s11094-019-02088-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
29
|
Gallez A, Palazzo C, Blacher S, Tskitishvili E, Noël A, Foidart JM, Evrard B, Pequeux C, Piel G. Liposomes and drug-in-cyclodextrin-in-liposomes formulations encapsulating 17β-estradiol: An innovative drug delivery system that prevents the activation of the membrane-initiated steroid signaling (MISS) of estrogen receptor α. Int J Pharm 2019; 573:118861. [PMID: 31765774 DOI: 10.1016/j.ijpharm.2019.118861] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 01/05/2023]
Abstract
The encapsulation into liposomes of several types of molecules presents the advantages to protect the activity of these molecules and to target specific tissues. Nevertheless, a major obstacle remains the incomplete understanding of nano-bio interactions. Specifically, the impact that inclusion of drug into liposomes or of drug-in-cyclodextrin-in liposomes (DCL) could have on the molecular and cellular mechanism of drug action is largely unknown. As a proof of concept, we evaluated the impact of 17β-estradiol (E2) included into liposomes or DCL on estrogen receptor (ER)α signaling pathways. Indeed, ERα relays the pleiotropic actions of E2 in physiology and pathophysiology through two major pathways: (1) the genomic/nuclear effects associated to the transcriptional activity of the ERα and (2) the rapid/nongenomic/membrane-initiated steroid signaling (MISS) effects related to the induction of fast signaling pathways occurring when ERα is anchored to the plasma membrane. We evidenced that the inclusion of E2 into liposomes (Lipo-E2) or into DCL (DCL-E2) prevented the activation of the rapid/nongenomic/extranuclear/MISS pathway of ERα, while the activation of the genomic/nuclear pathway was maintained. These results support that Lipo-E2 and DCL-E2 could be a useful tool to delineate the complex molecular mechanisms associated to ERα. In conclusion, this study supports the notion that inclusion of drugs into liposomes or DCL could modify some specific pathways of their molecular and cellular mechanisms of action. These results emphasized that attention should be paid to nano-bio interactions induced by the use of nanovectors in medicine.
Collapse
Affiliation(s)
- Anne Gallez
- Laboratory of Tumor and Development Biology (LBTD), GIGA-Cancer, University of Liège, Quartier hôpital, B23, Avenue Hippocrate 13, B-4000 Liege, Belgium
| | - Claudio Palazzo
- Laboratory of Pharmaceutical Technology and Biopharmacy (LTPB), Nanomedicine Development, CIRM, University of Liege, Liege, Belgium
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology (LBTD), GIGA-Cancer, University of Liège, Quartier hôpital, B23, Avenue Hippocrate 13, B-4000 Liege, Belgium
| | - Ekaterine Tskitishvili
- Laboratory of Tumor and Development Biology (LBTD), GIGA-Cancer, University of Liège, Quartier hôpital, B23, Avenue Hippocrate 13, B-4000 Liege, Belgium
| | - Agnès Noël
- Laboratory of Tumor and Development Biology (LBTD), GIGA-Cancer, University of Liège, Quartier hôpital, B23, Avenue Hippocrate 13, B-4000 Liege, Belgium
| | - Jean-Michel Foidart
- Laboratory of Tumor and Development Biology (LBTD), GIGA-Cancer, University of Liège, Quartier hôpital, B23, Avenue Hippocrate 13, B-4000 Liege, Belgium
| | - Brigitte Evrard
- Laboratory of Pharmaceutical Technology and Biopharmacy (LTPB), Nanomedicine Development, CIRM, University of Liege, Liege, Belgium
| | - Christel Pequeux
- Laboratory of Tumor and Development Biology (LBTD), GIGA-Cancer, University of Liège, Quartier hôpital, B23, Avenue Hippocrate 13, B-4000 Liege, Belgium
| | - Geraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy (LTPB), Nanomedicine Development, CIRM, University of Liege, Liege, Belgium.
| |
Collapse
|
30
|
Johnsen KB, Burkhart A, Thomsen LB, Andresen TL, Moos T. Targeting the transferrin receptor for brain drug delivery. Prog Neurobiol 2019; 181:101665. [DOI: 10.1016/j.pneurobio.2019.101665] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/10/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
|
31
|
Nally FK, De Santi C, McCoy CE. Nanomodulation of Macrophages in Multiple Sclerosis. Cells 2019; 8:cells8060543. [PMID: 31195710 PMCID: PMC6628349 DOI: 10.3390/cells8060543] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022] Open
Abstract
Multiple Sclerosis (MS) is a chronic demyelinating autoimmune disease primarily affecting young adults. Despite an unclear causal factor, symptoms and pathology arise from the infiltration of peripheral immune cells across the blood brain barrier. Accounting for the largest fraction of this infiltrate, macrophages are functionally heterogeneous innate immune cells capable of adopting either a pro or an anti-inflammatory phenotype, a phenomenon dependent upon cytokine milieu in the CNS. This functional plasticity is of key relevance in MS, where the pro-inflammatory state dominates the early stage, instructing demyelination and axonal loss while the later anti-inflammatory state holds a key role in promoting tissue repair and regeneration in later remission. This review highlights a potential therapeutic benefit of modulating macrophage polarisation to harness the anti-inflammatory and reparative state in MS. Here, we outline the role of macrophages in MS and look at the role of current FDA approved therapeutics in macrophage polarisation. Moreover, we explore the potential of particulate carriers as a novel strategy to manipulate polarisation states in macrophages, whilst examining how optimising macrophage uptake via nanoparticle size and functionalisation could offer a novel therapeutic approach for MS.
Collapse
Affiliation(s)
- Frances K Nally
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| | - Chiara De Santi
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| | - Claire E McCoy
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| |
Collapse
|
32
|
The availability of drug by liposomal drug delivery : Individual kinetics and tissue distribution of encapsulated and released drug in mice after administration of PEGylated liposomal prednisolone phosphate. Invest New Drugs 2018; 37:890-901. [PMID: 30547315 PMCID: PMC6736927 DOI: 10.1007/s10637-018-0708-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/30/2018] [Indexed: 12/14/2022]
Abstract
Lately, the usefulness of liposomal drug delivery systems has been debated. To better understand the underlying pharmacokinetics of the targeted drug delivery by liposomes, individual encapsulated and non-encapsulated drug concentrations in blood, tumor, liver, spleen and kidneys were quantified after i.v. administration of liposomal prednisolone phosphate in mice. Kinetic analysis shows that the tumor influx of encapsulated drug is not dominant compared to the uptake by the other tissues. Further, from a quantitative point of view, the availability of non-encapsulated drug in the tumor tissue after liposomal delivery is not pronounced as compared to the other tissues studied. However, drug release in the tumor seems more extended than in the other tissues and the non-encapsulated drug concentration decreases more slowly in the tumor than in the liver and spleen. The spleen shows a high affinity for the uptake of encapsulated drug as well as the release of drug from the liposomes. Subsequently, released drug in the spleen, and possibly also in other tissues, is probably quickly redistributed towards the blood and other tissues. This also impairs the drug delivery effect of the liposomes. In contrast to the released drug in the central circulation, liver and spleen, the released drug concentration in the tumor remains at a fairly constant level likely due to the extended release kinetics from the liposomes. These extended release characteristics in the tumor most probably contribute to the beneficial effect. Nevertheless, it should be noted that larger released drug concentrations are formed in healthy tissues.
Collapse
|
33
|
Nanodelivery systems for overcoming limited transportation of therapeutic molecules through the blood-brain barrier. Future Med Chem 2018; 10:2659-2674. [PMID: 30499740 DOI: 10.4155/fmc-2018-0208] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Due to the impermeable structure and barrier function of the blood-brain barrier (BBB), the delivery of therapeutic molecules into the CNS is extremely limited. Nanodelivery systems are regarded as the most effective and versatile carriers for the CNS, as they can transport cargo molecules across the BBB via various mechanisms. This review emphasizes the multi-functionalization strategies of nanodelivery systems and combinatorial approaches for the delivery of therapeutic drugs and genes into the CNS. The characteristics and functions of the BBB and underlying mechanisms of molecular translocation across the BBB are also described.
Collapse
|
34
|
Palazzo C, Laloy J, Delvigne AS, Nys G, Fillet M, Dogne JM, Pequeux C, Foidart JM, Evrard B, Piel G. Development of injectable liposomes and drug-in-cyclodextrin-in-liposome formulations encapsulating estetrol to prevent cerebral ischemia of premature babies. Eur J Pharm Sci 2018; 127:52-59. [PMID: 30308312 DOI: 10.1016/j.ejps.2018.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/25/2018] [Accepted: 10/07/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Claudio Palazzo
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liege, Liege, Belgium
| | - Julie Laloy
- Département de Pharmacie, Namur Nanosafety Centre, NARILIS, University of Namur, Namur, Belgium
| | - Anne-Sophie Delvigne
- Département de Pharmacie, Namur Nanosafety Centre, NARILIS, University of Namur, Namur, Belgium
| | - Gwenael Nys
- Analysis of Medicines Laboratory, CIRM, University of Liege, Liege, Belgium
| | - Marianne Fillet
- Analysis of Medicines Laboratory, CIRM, University of Liege, Liege, Belgium
| | - Jean-Michel Dogne
- Département de Pharmacie, Namur Nanosafety Centre, NARILIS, University of Namur, Namur, Belgium
| | - Christel Pequeux
- Tumor and Development Biology Laboratory, University of Liege, Liege, Belgium
| | - Jean-Michel Foidart
- Tumor and Development Biology Laboratory, University of Liege, Liege, Belgium
| | - Brigitte Evrard
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liege, Liege, Belgium
| | - Geraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liege, Liege, Belgium.
| |
Collapse
|
35
|
Vandewalle J, Luypaert A, De Bosscher K, Libert C. Therapeutic Mechanisms of Glucocorticoids. Trends Endocrinol Metab 2018; 29:42-54. [PMID: 29162310 DOI: 10.1016/j.tem.2017.10.010] [Citation(s) in RCA: 312] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 12/20/2022]
Abstract
Glucocorticoids (GCs) have been used clinically for decades as potent anti-inflammatory and immunosuppressive agents. Nevertheless, their use is severely hampered by the risk of developing side effects and the occurrence of glucocorticoid resistance (GCR). Therefore, efforts to understand the complex mechanisms underlying GC function and GCR are ongoing. The goal is to generate new glucocorticoid receptor (GR) ligands that can dissociate anti-inflammatory from metabolic side effects and/or overcome GCR. In this review paper we discuss recent insights into GR-mediated actions in GCR and novel therapeutic strategies for acute and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jolien Vandewalle
- Center for Inflammation Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Astrid Luypaert
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-University of Ghent (UGent) Center for Medical Biotechnology, Ghent, Belgium; Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-University of Ghent (UGent) Center for Medical Biotechnology, Ghent, Belgium; Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Claude Libert
- Center for Inflammation Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
36
|
Durymanov M, Kamaletdinova T, Lehmann SE, Reineke J. Exploiting passive nanomedicine accumulation at sites of enhanced vascular permeability for non-cancerous applications. J Control Release 2017. [DOI: 10.1016/j.jconrel.2017.06.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
37
|
Novel Drug Delivery Systems Tailored for Improved Administration of Glucocorticoids. Int J Mol Sci 2017; 18:ijms18091836. [PMID: 28837059 PMCID: PMC5618485 DOI: 10.3390/ijms18091836] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/18/2017] [Accepted: 08/21/2017] [Indexed: 12/11/2022] Open
Abstract
Glucocorticoids (GC) are one of the most popular and versatile classes of drugs available to treat chronic inflammation and cancer, but side effects and resistance constrain their use. To overcome these hurdles, which are often related to the uniform tissue distribution of free GC and their short half-life in biological fluids, new delivery vehicles have been developed including PEGylated liposomes, polymeric micelles, polymer-drug conjugates, inorganic scaffolds, and hybrid nanoparticles. While each of these nanoformulations has individual drawbacks, they are often superior to free GC in many aspects including therapeutic efficacy when tested in cell culture or animal models. Successful application of nanomedicines has been demonstrated in various models of neuroinflammatory diseases, cancer, rheumatoid arthritis, and several other disorders. Moreover, investigations using human cells and first clinical trials raise the hope that the new delivery vehicles may have the potential to make GC therapies more tolerable, specific and efficient in the future.
Collapse
|
38
|
Sellner S, Kocabey S, Zhang T, Nekolla K, Hutten S, Krombach F, Liedl T, Rehberg M. Dexamethasone-conjugated DNA nanotubes as anti-inflammatory agents in vivo. Biomaterials 2017; 134:78-90. [DOI: 10.1016/j.biomaterials.2017.04.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 04/12/2017] [Accepted: 04/15/2017] [Indexed: 12/24/2022]
|
39
|
Gaurab R, Dattatrya S, Amit Y, Gopal C K. Nanomedicine. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Nanomedicine, an offshoot of nanotechnology, is considered as one of the most promising technologies of the 21st century. Due to their minute size, nanomedicines can easily target difficult-to-reach sites with improved solubility and bioavailability and reduced adverse effects. They also act as versatile delivery systems, carrying both chemotherapeutics and imaging agents to targeted sites. Hence, nanomedicine can be used to achieve the same therapeutic effect at smaller doses than their conventional counterparts and can offer impressive resolutions for various life-threatening diseases. Although certain issues have been raised about the potential toxicities of nanomaterials, it is anticipated that the advances in nanomedicine will furnish clarifications to many of modern medicine's unsolved problems. This chapter aims to provide a comprehensive and contemporary survey of various nanomedicine products along with the major risks and side effects associated with the nanoparticles.
Collapse
Affiliation(s)
- Roy Gaurab
- National Center for Cell Science (NCCS) – Pune, India
| | | | - Yadav Amit
- National Center for Cell Science (NCCS) – Pune, India
| | - Kundu Gopal C
- National Center for Cell Science (NCCS) – Pune, India
| |
Collapse
|
40
|
Montes-Cobos E, Ring S, Fischer HJ, Heck J, Strauß J, Schwaninger M, Reichardt SD, Feldmann C, Lühder F, Reichardt HM. Targeted delivery of glucocorticoids to macrophages in a mouse model of multiple sclerosis using inorganic-organic hybrid nanoparticles. J Control Release 2016; 245:157-169. [PMID: 27919626 DOI: 10.1016/j.jconrel.2016.12.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 11/23/2016] [Accepted: 12/01/2016] [Indexed: 11/25/2022]
Abstract
Glucocorticoids (GC) are widely used to treat acute relapses in multiple sclerosis (MS) patients, but their application is accompanied by side effects due to their broad spectrum of action. Here, we report on the therapeutic option to apply GC via inorganic-organic hybrid nanoparticles (IOH-NP) with the composition [ZrO]2+[(BMP)0.9(FMN)0.1]2- (designated BMP-NP with BMP: betamethasone phosphate; FMN: flavinmononucleotide). We found that these BMP-NP have an increased cell type-specificity compared to free GC while retaining full therapeutic efficacy in a mouse model of MS. BMP-NP were preferentially taken up by phagocytic cells and modulated macrophages in vivo more efficiently than T cells. When GC were applied in the form of BMP-NP, treatment of neuroinflammatory disease in mice exclusively depended on the control of macrophage function whereas effects on T cells and brain endothelial cells were dispensable for therapeutic efficacy. Importantly, BMP-NP were not only active in mice but also showed strong activity towards monocytes isolated from healthy human volunteers. We conclude that application of GC via IOH-NP has the potential to improve MS therapy in the future.
Collapse
Affiliation(s)
- Elena Montes-Cobos
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Sarah Ring
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Henrike J Fischer
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Joachim Heck
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Judith Strauß
- Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562 Lübeck, Germany
| | - Sybille D Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Claus Feldmann
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Fred Lühder
- Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Holger M Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany.
| |
Collapse
|
41
|
Turjeman K, Barenholz Y. Liposomal nano-drugs based on amphipathic weak acid steroid prodrugs for treatment of inflammatory diseases. J Drug Target 2016; 24:805-820. [PMID: 27750439 DOI: 10.1080/1061186x.2016.1236262] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Steroids are the most efficacious anti-inflammatory agents. However, their toxicities and side-effects compromise their clinical application. Various strategies and major efforts were dedicated for formulating viable liposomal glucocorticosteroids (GCs), so far none of these were approved. OBJECTIVES To evaluate these approaches for formulating GC-delivery systems, especially liposomes, and with focus on the Barenholz Lab experience. METHODS We developed PEGylated nano-liposomes (NSSL) remotely loaded with water-soluble amphipathic weak acid GC-prodrugs. Their remote loading results in high, efficient and stable loading to the level that enables human clinical use. We characterized them for their physical chemistry and stability. We demonstrated their therapeutic efficacy in relevant animal models and studied their pharmacokinetics (PK), biodistribution (BD) and pharmacodynamics advantages over the free pro-drugs. RESULTS Our steroidal nano-drugs demonstrate much superior PK, BD, tolerability and therapeutic efficacies compared to the free pro-drugs and to most drugs currently used to treat these diseases. These nano-drugs act as robust immune-suppressors, affecting cytokines secretion and diminishing hemorrhage and edema. CONCLUSIONS The combination of improved physical-chemistry, PK, BD, tolerability and therapeutic efficacy of these steroidal nano-drugs over the pro-drugs "as-is" support their further clinical development as potential therapeutic agents for treating inflammatory diseases.
Collapse
Affiliation(s)
- Keren Turjeman
- a Department of Biochemistry and Molecular Biology, Laboratory of Membrane and Liposome Research , Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School , Jerusalem , Israel
| | - Yechezkel Barenholz
- a Department of Biochemistry and Molecular Biology, Laboratory of Membrane and Liposome Research , Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School , Jerusalem , Israel
| |
Collapse
|
42
|
Cao M, Qi P, Chen C, Song L, Wang X, Li N, Wu D, Hu G, Zhao J. A Vector-Based Short Hairpin RNA Targeting Aurora B Suppresses Human Prostatic Carcinoma Growth. Technol Cancer Res Treat 2016; 16:112-119. [PMID: 28100163 PMCID: PMC5616121 DOI: 10.1177/1533034616673534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aurora kinase B, playing a vital, important role in mitosis, is frequently detected to be overexpressed in many cancer cell lines and various tumor tissues, including prostatic carcinoma. Given the essential function of Aurora kinase B in mitosis and its association with tumorigenesis, it might be a drug target for prostatic carcinoma treatment. In our study, short hairpin RNA targeting Aurora kinase B was cloned into a pGPU6 plasmid vector and then transfected into human prostatic carcinoma cells. The expression level of Aurora kinase B was verified by reverse transcription-polymerase chain reaction and Western blot. At the same time, cell apoptosis was detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide, fluorescent staining, and flow cytometric analysis. Furthermore, prostate carcinoma cells were injected into mice to establish a tumor xenograft model. Previous studies have shown the effect of pGPU6-shAURKB plasmid on tumor growth in a prostate carcinoma xenogenic implantation model. From the study, we knew that the Aurora kinase B was significantly downregulated in prostate carcinoma cells, and cell apoptosis was also detected higher in treated groups than that in control groups. Moreover, in the prostate carcinoma xenogenic implantation model, compared with the control groups, the tumor growth was inhibited about 78.7% in the pGPU6-shAURKB plasmid–treated group, and cell apoptosis in the experimental group was notably higher than that in control groups. The average duration of tumor-bearing mice was prolonged to about 35 days. The results of experiment indicated that specific knockdown of Aurora kinase B led to prostate carcinoma cells apoptosis and inhibited tumor growth. Our data clearly confirmed that specific knockdown of Aurora kinase B expression by vector-based short hairpin RNA/liposome may be a potential new approach to treat human prostatic carcinoma.
Collapse
Affiliation(s)
- Mei Cao
- 1 Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China.,2 Core Laboratory, School of Medicine, Sichuan Provincial People's Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Panpan Qi
- 1 Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Chong Chen
- 1 Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Liju Song
- 1 Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Xuege Wang
- 1 Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Ningzhe Li
- 1 Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Daoyan Wu
- 1 Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Guoku Hu
- 2 Core Laboratory, School of Medicine, Sichuan Provincial People's Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, People's Republic of China.,3 Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jian Zhao
- 1 Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
43
|
Wong C, Bezhaeva T, Rothuizen TC, Metselaar JM, de Vries MR, Verbeek FPR, Vahrmeijer AL, Wezel A, van Zonneveld AJ, Rabelink TJ, Quax PHA, Rotmans JI. Liposomal prednisolone inhibits vascular inflammation and enhances venous outward remodeling in a murine arteriovenous fistula model. Sci Rep 2016; 6:30439. [PMID: 27460883 PMCID: PMC4962038 DOI: 10.1038/srep30439] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/01/2016] [Indexed: 12/20/2022] Open
Abstract
Arteriovenous fistulas (AVF) for hemodialysis access have a 1-year primary patency rate of only 60%, mainly as a result of maturation failure that is caused by insufficient outward remodeling and intimal hyperplasia. The exact pathophysiology remains unknown, but the inflammatory vascular response is thought to play an important role. In the present study we demonstrate that targeted liposomal delivery of prednisolone increases outward remodeling of the AVF in a murine model. Liposomes accumulate in the post-anastomotic area of the venous outflow tract in which the vascular pathology is most prominent in failed AVFs. On a histological level, we observed a reduction of lymphocytes and granulocytes in the vascular wall. In addition, a strong anti-inflammatory effect of liposomal prednisolone on macrophages was demonstrated in vitro. Therefore, treatment with liposomal prednisolone might be a valuable strategy to improve AVF maturation.
Collapse
Affiliation(s)
- ChunYu Wong
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden Medical Center, Leiden, The Netherlands
| | - Taisiya Bezhaeva
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden Medical Center, Leiden, The Netherlands
| | - Tonia C Rothuizen
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden Medical Center, Leiden, The Netherlands
| | - Josbert M Metselaar
- Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, The Netherlands.,Enceladus Pharmaceuticals BV, The Netherlands
| | - Margreet R de Vries
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden Medical Center, Leiden, The Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Floris P R Verbeek
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Anouk Wezel
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Anton-Jan van Zonneveld
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden Medical Center, Leiden, The Netherlands
| | - Ton J Rabelink
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden Medical Center, Leiden, The Netherlands
| | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden Medical Center, Leiden, The Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Joris I Rotmans
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden Medical Center, Leiden, The Netherlands
| |
Collapse
|
44
|
Ghalamfarsa G, Hojjat-Farsangi M, Mohammadnia-Afrouzi M, Anvari E, Farhadi S, Yousefi M, Jadidi-Niaragh F. Application of nanomedicine for crossing the blood–brain barrier: Theranostic opportunities in multiple sclerosis. J Immunotoxicol 2016; 13:603-19. [DOI: 10.3109/1547691x.2016.1159264] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ghasem Ghalamfarsa
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene Therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
- Department of Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mousa Mohammadnia-Afrouzi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Enayat Anvari
- Department of Physiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Shohreh Farhadi
- Department of Agricultural Engineering, Islamic Azad University, Tehran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Grabrucker AM, Ruozi B, Belletti D, Pederzoli F, Forni F, Vandelli MA, Tosi G. Nanoparticle transport across the blood brain barrier. Tissue Barriers 2016; 4:e1153568. [PMID: 27141426 DOI: 10.1080/21688370.2016.1153568] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/02/2016] [Accepted: 02/04/2016] [Indexed: 01/13/2023] Open
Abstract
While the role of the blood-brain barrier (BBB) is increasingly recognized in the (development of treatments targeting neurodegenerative disorders, to date, few strategies exist that enable drug delivery of non-BBB crossing molecules directly to their site of action, the brain. However, the recent advent of Nanomedicines may provide a potent tool to implement CNS targeted delivery of active compounds. Approaches for BBB crossing are deeply investigated in relation to the pathology: among the main important diseases of the CNS, this review focuses on the application of nanomedicines to neurodegenerative disorders (Alzheimer, Parkinson and Huntington's Disease) and to other brain pathologies as epilepsy, infectious diseases, multiple sclerosis, lysosomal storage disorders, strokes.
Collapse
Affiliation(s)
- Andreas M Grabrucker
- WG Molecular Analysis of Synaptopathies, Neurology Dept, Neurocenter of Ulm University , Ulm, Germany
| | - Barbara Ruozi
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Daniela Belletti
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Francesca Pederzoli
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Flavio Forni
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Maria Angela Vandelli
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Giovanni Tosi
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| |
Collapse
|
46
|
Jatczak-Pawlik I, Książek-Winiarek D, Wojkowska D, Jóźwiak K, Jastrzębski K, Pietruczuk M, Głąbiński A. The impact of multiple sclerosis relapse treatment on migration of effector T cells--Preliminary study. Neurol Neurochir Pol 2016; 50:155-62. [PMID: 27154441 DOI: 10.1016/j.pjnns.2016.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/25/2016] [Accepted: 02/10/2016] [Indexed: 11/18/2022]
Abstract
UNLABELLED Migration of inflammatory cells from the blood to the central nervous system (CNS) is crucial for development of multiple sclerosis (MS). Inhibition of this process would allow to control disease activity. The first step confirming this approach would be the analysis of the impact of effective MS relapse therapy on migration of effector T cells. The aim of the study was to analyze the influence of methylprednisolone (MP) on the migratory activity of effector CD4+ T cells from MS patients. Moreover, to study the potential mechanism of this process we studied expression of chemokine receptors on migrating cells. MATERIAL AND METHODS Peripheral blood samples were obtained from relapsing-remitting MS (RR-MS) patients during relapse (n=23) and from control group (n=23). After isolation CD4+ T cells were incubated with various concentrations of MP. Then they were stimulated in chemotaxis assay with chemokines CCL3 or CXCL10 or were used to CCR1 and CXCR3 expression analysis. RESULTS CXCL10- and CCL3-stimulated migration of CD4+ T cells was significantly increased in MS. MP was able to reduce in vitro migration of effector T cells induced by CXCL10, but not by CCL3. Inhibition by MP was dose-dependent. Expression of analyzed chemokine receptors was unaltered after MP incubation. CONCLUSIONS MP reduced CD4+ T cells migration induced by CXCL10 without affecting CXCR3 expression. These observations demonstrate one of the potential mechanisms of MP action in MS, distinct from inducing cell apoptosis, and suggests the new targets for development of more effective MS treatments.
Collapse
Affiliation(s)
| | - Dominika Książek-Winiarek
- Department of Propedeutics of Neurology, Medical University of Lodz, Lodz, Poland; Department of Neurology and Stroke, Medical University of Lodz, Lodz, Poland.
| | - Dagmara Wojkowska
- Department of Propedeutics of Neurology, Medical University of Lodz, Lodz, Poland; Department of Neurology and Stroke, Medical University of Lodz, Lodz, Poland
| | - Krzysztof Jóźwiak
- Department of Propedeutics of Neurology, Medical University of Lodz, Lodz, Poland; Department of Neurology and Stroke, Medical University of Lodz, Lodz, Poland
| | - Karol Jastrzębski
- Department of Neurology and Stroke, Medical University of Lodz, Lodz, Poland
| | | | - Andrzej Głąbiński
- Department of Propedeutics of Neurology, Medical University of Lodz, Lodz, Poland; Department of Neurology and Stroke, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
47
|
Hosseini SH, Maleki A, Eshraghi HR, Hamidi M. Preparation and in vitro/pharmacokinetic/pharmacodynamic evaluation of a slow-release nano-liposomal form of prednisolone. Drug Deliv 2016; 23:3008-3016. [DOI: 10.3109/10717544.2016.1138341] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Seyed Hojjat Hosseini
- Department of Veterinary Basic Science, Tehran Science and Research Branch, Islamic Azad University, Tehran, Iran and
| | - Aziz Maleki
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamid Reza Eshraghi
- Department of Veterinary Basic Science, Tehran Science and Research Branch, Islamic Azad University, Tehran, Iran and
| | - Mehrdad Hamidi
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
48
|
Johnsen KB, Moos T. Revisiting nanoparticle technology for blood–brain barrier transport: Unfolding at the endothelial gate improves the fate of transferrin receptor-targeted liposomes. J Control Release 2016; 222:32-46. [DOI: 10.1016/j.jconrel.2015.11.032] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/28/2015] [Accepted: 11/30/2015] [Indexed: 12/25/2022]
|
49
|
Turjeman K, Bavli Y, Kizelsztein P, Schilt Y, Allon N, Katzir TB, Sasson E, Raviv U, Ovadia H, Barenholz Y. Nano-Drugs Based on Nano Sterically Stabilized Liposomes for the Treatment of Inflammatory Neurodegenerative Diseases. PLoS One 2015; 10:e0130442. [PMID: 26147975 PMCID: PMC4492950 DOI: 10.1371/journal.pone.0130442] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 05/20/2015] [Indexed: 12/28/2022] Open
Abstract
The present study shows the advantages of liposome-based nano-drugs as a novel strategy of delivering active pharmaceutical ingredients for treatment of neurodegenerative diseases that involve neuroinflammation. We used the most common animal model for multiple sclerosis (MS), mice experimental autoimmune encephalomyelitis (EAE). The main challenges to overcome are the drugs’ unfavorable pharmacokinetics and biodistribution, which result in inadequate therapeutic efficacy and in drug toxicity (due to high and repeated dosage). We designed two different liposomal nano-drugs, i.e., nano sterically stabilized liposomes (NSSL), remote loaded with: (a) a “water-soluble” amphipathic weak acid glucocorticosteroid prodrug, methylprednisolone hemisuccinate (MPS) or (b) the amphipathic weak base nitroxide, Tempamine (TMN). For the NSSL-MPS we also compared the effect of passive targeting alone and of active targeting based on short peptide fragments of ApoE or of β-amyloid. Our results clearly show that for NSSL-MPS, active targeting is not superior to passive targeting. For the NSSL-MPS and the NSSL-TMN it was demonstrated that these nano-drugs ameliorate the clinical signs and the pathology of EAE. We have further investigated the MPS nano-drug’s therapeutic efficacy and its mechanism of action in both the acute and the adoptive transfer EAE models, as well as optimizing the perfomance of the TMN nano-drug. The highly efficacious anti-inflammatory therapeutic feature of these two nano-drugs meets the criteria of disease-modifying drugs and supports further development and evaluation of these nano-drugs as potential therapeutic agents for diseases with an inflammatory component.
Collapse
Affiliation(s)
- Keren Turjeman
- Laboratory of Membrane and Liposome Research, Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
- * E-mail:
| | - Yaelle Bavli
- Laboratory of Membrane and Liposome Research, Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Pablo Kizelsztein
- Laboratory of Membrane and Liposome Research, Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yaelle Schilt
- The Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem, Israel
| | - Nahum Allon
- Laboratory of Membrane and Liposome Research, Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | - Efrat Sasson
- BioImage MRI Research & Consulting, Tel Aviv, Israel
| | - Uri Raviv
- The Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem, Israel
| | - Haim Ovadia
- Department of Neurology, Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
50
|
Edagwa BJ, Zhou T, McMillan JM, Liu XM, Gendelman HE. Development of HIV reservoir targeted long acting nanoformulated antiretroviral therapies. Curr Med Chem 2015; 21:4186-98. [PMID: 25174930 DOI: 10.2174/0929867321666140826114135] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/19/2014] [Accepted: 08/22/2014] [Indexed: 11/22/2022]
Abstract
Human immunodeficiency virus (HIV) infection commonly results in a myriad of comorbid conditions secondary to immune deficiency. Infection also affects broad organ system function. Although current antiretroviral therapy (ART) reduces disease morbidity and mortality through effective control of peripheral viral load, restricted infection in HIV reservoirs including gut, lymphoid and central nervous system tissues, is not eliminated. What underlies these events is, in part, poor ART penetrance into each organ across tissue barriers, viral mutation and the longevity of infected cells. We posit that one means to improve these disease outcomes is through nanotechnology. To this end, this review discusses a broad range of cutting-edge nanomedicines and nanomedicine platforms that are or can be used to improve ART delivery. Discussion points include how polymer-drug conjugates, dendrimers, micelles, liposomes, solid lipid nanoparticles and polymeric nanoparticles can be harnessed to best yield cell-based delivery systems. When completely developed, such nanomedicine platforms have the potential to clear reservoirs of viral infection.
Collapse
Affiliation(s)
| | | | | | | | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|