1
|
Jourdain MA, Eyer J. Recent advances in liposomes and peptide-based therapeutics for glioblastoma treatment. J Control Release 2024; 376:732-752. [PMID: 39437968 DOI: 10.1016/j.jconrel.2024.10.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
In the context of glioblastoma treatment, the penetration of drugs is drastically limited by the blood-brain-barrier (BBB). Emerging therapies have focused on the field of therapeutic peptides for their excellent BBB targeting properties that promote a deep tumor penetration. Peptide-based strategies are also renowned for their abilities of driving cargo such as liposomal system allowing an active targeting of receptors overexpressed on GBM cells. This review provides a detailed description of the internalization mechanisms of specific GBM homing and penetrating peptides as well as the latest in vitro/in vivo studies of liposomes functionalized with them. The purpose of this review is to summarize a selection of promising pre-clinical results that demonstrate the advantages of this nanosystem, including an increase of tumor cell targeting, triggering drug accumulation and thus a strong antitumor effect. Aware of the early stage of these studies, many challenges need to be overcome to promote peptide-directed liposome at clinical level. In particular, the lack of suitable production, the difficulty to characterize the nanosystem and therapeutic competition leaded by antibodies.
Collapse
Affiliation(s)
- M-A Jourdain
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France.
| | - J Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| |
Collapse
|
2
|
Oh CY, Kaur H, Tuteja G, Henderson ER. DNA origami drives gene expression in a human cell culture system. Sci Rep 2024; 14:27364. [PMID: 39521857 PMCID: PMC11550841 DOI: 10.1038/s41598-024-78399-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Self-assembling DNA nanoparticles have the potential to significantly advance the targeted delivery of molecular cargo owing to their chemical and architectural flexibility. Recently, it has been demonstrated that the genetic code embedded in DNA nanoparticles produced by the method of DNA origami or related techniques can be recognized and copied by RNA polymerase in vitro. Further, sculpted DNA nanoparticles can serve as a substrate for Cas9-mediated gene modification and gene expression in cell culture. In the present study, we further investigate the ability of DNA origami nanoparticles to be expressed in a human cell line with emphasis on the impact of single-stranded DNA (ssDNA) domains and the contributions of the architectural disposition of genetic control elements, namely promoter and enhancer sequences. Our findings suggest that while cells possess the remarkable capability to express genes within highly folded architectures, the presence and relative density and location of ssDNA domains appears to influence overall levels of gene expression. These results suggest that it may be possible to nuance folded DNA nanoparticle architecture to regulate the rate and/or level of gene expression. Considering the highly malleable architecture and chemistry of self-assembling DNA nanoparticles, these findings motivate further exploration of their potential as an economic nanotechnology platform for targeted gene editing, nucleic acid-based vaccines, and related biotherapeutic applications.
Collapse
Affiliation(s)
- Chang Yong Oh
- Department of Biochemistry and Molecular Biology, Iowa State University, Ames, IA, 50011, USA.
- Department of Oncology, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Haninder Kaur
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Geetu Tuteja
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Eric R Henderson
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| |
Collapse
|
3
|
Jin Y, Du Q, Song M, Kang R, Zhou J, Zhang H, Ding Y. Amyloid-β-targeting immunotherapies for Alzheimer's disease. J Control Release 2024; 375:346-365. [PMID: 39271059 DOI: 10.1016/j.jconrel.2024.09.012] [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: 06/17/2024] [Revised: 07/24/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024]
Abstract
Recent advances in clinical passive immunotherapy have provided compelling evidence that eliminating amyloid-β (Aβ) slows cognitive decline in Alzheimer's disease (AD). However, the modest benefits and side effects observed in clinical trials indicate that current immunotherapy therapy is not a panacea, highlighting the need for a deeper understanding of AD mechanisms and the significance of early intervention through optimized immunotherapy or immunoprevention. This review focuses on the centrality of Aβ pathology in AD and summarizes recent clinical progress in passive and active immunotherapies targeting Aβ, discussing their lessons and failures to inform future anti-Aβ biotherapeutics design. Various delivery strategies to optimize Aβ-targeting immunotherapies are outlined, highlighting their benefits and drawbacks in overcoming challenges such as poor stability and limited tissue accessibility of anti-Aβ biotherapeutics. Additionally, the perspectives and challenges of immunotherapy and immunoprevention targeting Aβ are concluded in the end, aiming to guide the development of next-generation anti-Aβ immunotherapeutic agents towards improved efficacy and safety.
Collapse
Affiliation(s)
- Yi Jin
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Qiaofei Du
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Mingjie Song
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ruixin Kang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jianping Zhou
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Huaqing Zhang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Yang Ding
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
4
|
Alassaf M, Rajan A. Adipocyte metabolic state regulates glial phagocytic function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614765. [PMID: 39386724 PMCID: PMC11463506 DOI: 10.1101/2024.09.24.614765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Obesity and type 2 diabetes are well-established risk factors for neurodegenerative disorders1-4, yet the underlying mechanisms remain poorly understood. The adipocyte-brain axis is crucial for brain function, as adipocytes secrete signaling molecules, including lipids and adipokines, that impinge on neural circuits to regulate feeding and energy expenditure5. Disruptions in the adipocyte-brain axis are associated with neurodegenerative conditions6, but the causal links are not fully understood. Neural debris accumulates with age and injury, and glial phagocytic function is crucial for clearing this debris and maintaining a healthy brain microenvironment7-9. Using adult Drosophila, we investigate how adipocyte metabolism influences glial phagocytic activity in the brain. We demonstrate that a prolonged obesogenic diet increases adipocyte fatty acid oxidation and ketogenesis. Genetic manipulations that mimic obesogenic diet-induced changes in adipocyte lipid and mitochondrial metabolism unexpectedly reduce the expression of the phagocytic receptor Draper in Drosophila microglia-like cells in the brain. We identify Apolpp-the Drosophila equivalent of human apolipoprotein B (ApoB)-as a critical adipocyte-derived signal that regulates glial phagocytosis. Additionally, we show that Lipoprotein Receptor 1 (LpR1), the LDL receptor on phagocytic glia, is required for glial capacity to clear injury-induced neuronal debris. Our findings establish that adipocyte-brain lipoprotein signaling regulates glial phagocytic function, revealing a novel pathway that links adipocyte metabolic disorders with neurodegeneration.
Collapse
Affiliation(s)
- Mroj Alassaf
- Basic Sciences Division, Fred Hutch, Seattle, WA-98109. The USA
| | - Akhila Rajan
- Basic Sciences Division, Fred Hutch, Seattle, WA-98109. The USA
| |
Collapse
|
5
|
Kim S, Ullah I, Beloor J, Chung K, Kim J, Yi Y, Kang E, Yun G, Heo S, Pyun SH, Kim SH, Kumar P, Lee SK. Systemic Treatment with siRNA Targeting Gamma-Secretase Activating Protein Inhibits Amyloid-β Accumulation in Alzheimer's Disease. Biomater Res 2024; 28:0027. [PMID: 38868092 PMCID: PMC11168191 DOI: 10.34133/bmr.0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/31/2024] [Indexed: 06/14/2024] Open
Abstract
Amyloid-β (Aβ) peptide aggregation in the brain is a key factor in Alzheimer's disease. However, direct inhibition of β-secretase or γ-secretase proves ineffective in reducing Aβ accumulation and improving cognition in Alzheimer's. Recent findings suggest that inhibiting gamma-secretase activating protein (GSAP) can decrease Aβ generation without affecting crucial γ-secretase substrates. Dimerization of Lep9R3LC (diLep9R3LC) was confirmed by Ellman's test. The peptide-small interfering RNA (siRNA) complex ratio, particle size, and surface charge were analyzed using electrophoretic mobility shift assay, and dynamic light scattering, respectively. In a 3xTg mice model of Alzheimer's disease, diLep9R3LC:siRNA complexes were intravenously administered twice a week for 8 weeks. Assessments included gene silencing, protein expression, and behavioral improvement using reverse transcription polymerase chain reaction, quantitative polymerase chain reaction, western blotting, Y-maze, and object recognition tests. The efficacy of Lep9R3LC dimerization was ~80% after a 3-d reaction by Ellman's test. In N2a cells, diLep9R3LC:siGSAP complexes achieved ~70% silencing at 48 h posttransfection. In 7-month-old male 3xTg mice, GSAP knockdown was ~30% in the cortex and ~50% in the hippocampus. The behavior improved in mice treated with diLep9R3LC:siGSAP complexes, showing a 60% increase in entries and an 80% increase object recognition. A novel dipeptide, diLep9R3LC, complexed with siRNA targeting GSAP (siGSAP), efficiently delivers siRNA to the mouse brain, targeting the hippocampus. The treatment inhibits Aβ accumulation, reduces GSK-3β-associated with tau hyperphosphorylation, and improves Alzheimer's behavior. Our findings highlight diLep9R3LC:siGSAP's potential for Alzheimer's and as a siRNA carrier for central nervous system-related diseases.
Collapse
Affiliation(s)
- Sunghwa Kim
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
- Department of Internal Medicine,
Yale University, New Haven, CT, USA
| | - Jagadish Beloor
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
- Department of Internal Medicine,
Yale University, New Haven, CT, USA
| | - Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
- Lerner Research Institute,
Cleveland Clinic, Cleveland, OH, USA
| | - Jongkil Kim
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
- Harvard Medical School, Boston, MA, USA
| | - Yujong Yi
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Eunhwa Kang
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Gyeongju Yun
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Seoyoun Heo
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Seon-Hong Pyun
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine,
Hanyang University, Seoul, Korea
| | - Priti Kumar
- Department of Internal Medicine,
Yale University, New Haven, CT, USA
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| |
Collapse
|
6
|
Niazi SK. Non-Invasive Drug Delivery across the Blood-Brain Barrier: A Prospective Analysis. Pharmaceutics 2023; 15:2599. [PMID: 38004577 PMCID: PMC10674293 DOI: 10.3390/pharmaceutics15112599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Non-invasive drug delivery across the blood-brain barrier (BBB) represents a significant advancement in treating neurological diseases. The BBB is a tightly packed layer of endothelial cells that shields the brain from harmful substances in the blood, allowing necessary nutrients to pass through. It is a highly selective barrier, which poses a challenge to delivering therapeutic agents into the brain. Several non-invasive procedures and devices have been developed or are currently being investigated to enhance drug delivery across the BBB. This paper presents a review and a prospective analysis of the art and science that address pharmacology, technology, delivery systems, regulatory approval, ethical concerns, and future possibilities.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
7
|
Leitão ADG, Ahammad RU, Spencer B, Wu C, Masliah E, Rissman RA. Novel systemic delivery of a peptide-conjugated antisense oligonucleotide to reduce α-synuclein in a mouse model of Alzheimer's disease. Neurobiol Dis 2023; 186:106285. [PMID: 37690676 PMCID: PMC10584037 DOI: 10.1016/j.nbd.2023.106285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023] Open
Abstract
Neurodegenerative disorders of aging are characterized by the progressive accumulation of proteins such as α-synuclein (α-syn) and amyloid beta (Aβ). Misfolded and aggregated α-syn has been implicated in neurological disorders such as Parkinson's disease, and Dementia with Lewy Bodies, but less so in Alzheimer's Disease (AD), despite the fact that accumulation of α-syn has been confirmed in over 50% of postmortem brains neuropathologically diagnosed with AD. To date, no therapeutic strategy has effectively or consistently downregulated α-syn in AD. Here we tested the hypothesis that by using a systemically-delivered peptide (ApoB11) bound to a modified antisense oligonucleotide against α-syn (ASO-α-syn), we can downregulate α-syn expression in an AD mouse model and improve behavioral and neuropathologic phenotypes. Our results demonstrate that monthly systemic treatment with of ApoB11:ASO α-syn beginning at 6 months of age reduces expression of α-synuclein in the brains of 9-month-old AD mice. Downregulation of α-syn led to reduction in Aβ plaque burden, prevented neuronal loss and astrogliosis. Furthermore, we found that AD mice treated with ApoB11:ASO α-syn had greatly improved hippocampal and spatial memory function in comparison to their control counterparts. Collectively, our data supports the reduction of α-syn through use of systemically-delivered ApoB11:ASO α-syn as a promising future disease-modifying therapeutic for AD.
Collapse
Affiliation(s)
- André D G Leitão
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Rijwan U Ahammad
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Brian Spencer
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America; Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA 92121, United States of America
| | - Eliezer Masliah
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD 20892, United States of America
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America; Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA 92121, United States of America; VA San Diego Healthcare System, San Diego, CA 92161, United States of America.
| |
Collapse
|
8
|
Nhàn NTT, Yamada T, Yamada KH. Peptide-Based Agents for Cancer Treatment: Current Applications and Future Directions. Int J Mol Sci 2023; 24:12931. [PMID: 37629112 PMCID: PMC10454368 DOI: 10.3390/ijms241612931] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Peptide-based strategies have received an enormous amount of attention because of their specificity and applicability. Their specificity and tumor-targeting ability are applied to diagnosis and treatment for cancer patients. In this review, we will summarize recent advancements and future perspectives on peptide-based strategies for cancer treatment. The literature search was conducted to identify relevant articles for peptide-based strategies for cancer treatment. It was performed using PubMed for articles in English until June 2023. Information on clinical trials was also obtained from ClinicalTrial.gov. Given that peptide-based strategies have several advantages such as targeted delivery to the diseased area, personalized designs, relatively small sizes, and simple production process, bioactive peptides having anti-cancer activities (anti-cancer peptides or ACPs) have been tested in pre-clinical settings and clinical trials. The capability of peptides for tumor targeting is essentially useful for peptide-drug conjugates (PDCs), diagnosis, and image-guided surgery. Immunomodulation with peptide vaccines has been extensively tested in clinical trials. Despite such advantages, FDA-approved peptide agents for solid cancer are still limited. This review will provide a detailed overview of current approaches, design strategies, routes of administration, and new technological advancements. We will highlight the success and limitations of peptide-based therapies for cancer treatment.
Collapse
Affiliation(s)
- Nguyễn Thị Thanh Nhàn
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Tohru Yamada
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Richard & Loan Hill Department of Biomedical Engineering, University of Illinois College of Engineering, Chicago, IL 60607, USA
| | - Kaori H. Yamada
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Department of Ophthalmology & Visual Sciences, University of Illinois College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
9
|
Ghorai SM, Deep A, Magoo D, Gupta C, Gupta N. Cell-Penetrating and Targeted Peptides Delivery Systems as Potential Pharmaceutical Carriers for Enhanced Delivery across the Blood-Brain Barrier (BBB). Pharmaceutics 2023; 15:1999. [PMID: 37514185 PMCID: PMC10384895 DOI: 10.3390/pharmaceutics15071999] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/25/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Among the challenges to the 21st-century health care industry, one that demands special mention is the transport of drugs/active pharmaceutical agents across the blood-brain barrier (BBB). The epithelial-like tight junctions within the brain capillary endothelium hinder the uptake of most pharmaceutical agents. With an aim to understand more deeply the intricacies of cell-penetrating and targeted peptides as a powerful tool for desirable biological activity, we provide a critical review of both CPP and homing/targeted peptides as intracellular drug delivery agents, especially across the blood-brain barrier (BBB). Two main peptides have been discussed to understand intracellular drug delivery; first is the cell-penetrating peptides (CPPs) for the targeted delivery of compounds of interest (primarily peptides and nucleic acids) and second is the family of homing peptides, which specifically targets cells/tissues based on their overexpression of tumour-specific markers and are thus at the heart of cancer research. These small, amphipathic molecules demonstrate specific physical and chemical modifications aimed at increased ease of cellular internalisation. Because only a limited number of drug molecules can bypass the blood-brain barrier by free diffusion, it is essential to explore all aspects of CPPs that can be exploited for crossing this barrier. Considering siRNAs that can be designed against any target RNA, marking such molecules with high therapeutic potential, we present a synopsis of the studies on synthetic siRNA-based therapeutics using CPPs and homing peptides drugs that can emerge as potential drug-delivery systems as an upcoming requirement in the world of pharma- and nutraceuticals.
Collapse
Affiliation(s)
- Soma Mondal Ghorai
- Department of Zoology, Hindu College, University of Delhi, Delhi 110007, India
| | - Auroni Deep
- Department of Zoology, Hindu College, University of Delhi, Delhi 110007, India
| | - Devanshi Magoo
- Department of Chemistry, Hindu College, University of Delhi, Delhi 110007, India
| | - Chetna Gupta
- Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
| | - Nikesh Gupta
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, WI 53705, USA
| |
Collapse
|
10
|
Mazura AD, Pietrzik CU. Endocrine Regulation of Microvascular Receptor-Mediated Transcytosis and Its Therapeutic Opportunities: Insights by PCSK9-Mediated Regulation. Pharmaceutics 2023; 15:pharmaceutics15041268. [PMID: 37111752 PMCID: PMC10144601 DOI: 10.3390/pharmaceutics15041268] [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: 03/01/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Currently, many neurological disorders lack effective treatment options due to biological barriers that effectively separate the central nervous system (CNS) from the periphery. CNS homeostasis is maintained by a highly selective exchange of molecules, with tightly controlled ligand-specific transport systems at the blood-brain barrier (BBB) playing a key role. Exploiting or modifying these endogenous transport systems could provide a valuable tool for targeting insufficient drug delivery into the CNS or pathological changes in the microvasculature. However, little is known about how BBB transcytosis is continuously regulated to respond to temporal or chronic changes in the environment. The aim of this mini-review is to draw attention to the sensitivity of the BBB to circulating molecules derived from peripheral tissues, which may indicate a fundamental endocrine-operating regulatory system of receptor-mediated transcytosis at the BBB. We present our thoughts in the context of the recent observation that low-density lipoprotein receptor-related protein 1 (LRP1)-mediated clearance of brain amyloid-β (Aβ) across the BBB is negatively regulated by peripheral proprotein convertase subtilisin/kexin type 9 (PCSK9). We hope that our conclusions will inspire future investigations of the BBB as dynamic communication interface between the CNS and periphery, whose peripheral regulatory mechanisms could be easily exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Alexander D Mazura
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55128 Mainz, Germany
| | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55128 Mainz, Germany
| |
Collapse
|
11
|
Zhao D, Liu J, Zhou Y, Zhang L, Zhong Y, Yang Y, Zhao B, Yang M, Wang Y. Penetrating the Blood-Brain Barrier for Targeted Treatment of Neurotoxicant Poisoning by Nanosustained-Released 2-PAM@VB1-MIL-101-NH 2(Fe). ACS APPLIED MATERIALS & INTERFACES 2023; 15:12631-12642. [PMID: 36867458 DOI: 10.1021/acsami.2c18929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
It is very important to establish a sustained-release pralidoxime chloride (2-PAM) drug system with brain targeting function for the treatment of neurotoxicant poisoning. Herein, Vitamin B1 (VB1), also known as thiamine, which can specifically bind to the thiamine transporter on the surface of the blood-brain barrier, was incorporated onto the surface of MIL-101-NH2(Fe) nanoparticles with a size of ∼100 nm. Pralidoxime chloride was further loaded within the interior of the above resulted composite by soaking, and a resulting composite drug (denoted as 2-PAM@VB1-MIL-101-NH2(Fe)) with a loading capacity of 14.8% (wt) was obtained. The results showed that the drug release rate of the composite drug was increased in PBS solution with the increase of pH (2-7.4) and a maximum drug release rate of 77.5% at pH 4. Experiments on the treatment of poisoning by gavage with the nerve agent sarin in mice combined with atropine revealed that sustained release of 2-PAM from the composite drug was achieved for more than 72 h. Sustained and stable reactivation of poisoned acetylcholinesterase (AChE) was observed with an enzyme reactivation rate of 42.7% in the ocular blood samples at 72 h. By using both zebrafish brain and mouse brain as models, we found that the composite drug could effectively cross the blood-brain barrier and restore the AChE activity in the brain of poisoned mice. The composite drug is expected to be a stable therapeutic drug with brain targeting and prolonged drug release properties for nerve agent intoxication in the middle and late stages of treatment.
Collapse
Affiliation(s)
- Dianfa Zhao
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P. R. China
- Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Jie Liu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P. R. China
- Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Yunshan Zhou
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Lijuan Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Yuxu Zhong
- Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Yang Yang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Baoquan Zhao
- Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Mengru Yang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Yong'an Wang
- Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| |
Collapse
|
12
|
Foster D, Larsen J. Polymeric Metal Contrast Agents for T 1-Weighted Magnetic Resonance Imaging of the Brain. ACS Biomater Sci Eng 2023; 9:1224-1242. [PMID: 36753685 DOI: 10.1021/acsbiomaterials.2c01386] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Imaging plays an integral role in diagnostics and treatment monitoring for conditions affecting the brain; enhanced brain imaging capabilities will improve upon both while increasing the general understanding of how the brain works. T1-weighted magnetic resonance imaging is the preferred modality for brain imaging. Commercially available contrast agents, which are often required to render readable brain images, have considerable toxicity concerns. In recent years, much progress has been made in developing new contrast agents based on the magnetic features of gadolinium, iron, or magnesium. Nanotechnological approaches for these systems allow for the protected integration of potentially harmful metals with added benefits like reduced dosage and improved transport. Polymeric enhancement of each design further improves biocompatibility while allowing for specific brain targeting. This review outlines research on polymeric nanomedicine designs for T1-weighted contrast agents that have been evaluated for performance in the brain.
Collapse
|
13
|
Dogan Y, Barese CN, Schindler JW, Yoon JK, Unnisa Z, Guda S, Jacobs ME, Oborski C, Maiwald T, Clarke DL, Schambach A, Pfeifer R, Harper C, Mason C, van Til NP. Screening chimeric GAA variants in preclinical study results in hematopoietic stem cell gene therapy candidate vectors for Pompe disease. Mol Ther Methods Clin Dev 2022; 27:464-487. [PMID: 36419467 PMCID: PMC9676529 DOI: 10.1016/j.omtm.2022.10.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022]
Abstract
Pompe disease is a rare genetic neuromuscular disorder caused by acid α-glucosidase (GAA) deficiency resulting in lysosomal glycogen accumulation and progressive myopathy. Enzyme replacement therapy, the current standard of care, penetrates poorly into the skeletal muscles and the peripheral and central nervous system (CNS), risks recombinant enzyme immunogenicity, and requires high doses and frequent infusions. Lentiviral vector-mediated hematopoietic stem and progenitor cell (HSPC) gene therapy was investigated in a Pompe mouse model using a clinically relevant promoter driving nine engineered GAA coding sequences incorporating distinct peptide tags and codon optimizations. Vectors solely including glycosylation-independent lysosomal targeting tags enhanced secretion and improved reduction of glycogen, myofiber, and CNS vacuolation in key tissues, although GAA enzyme activity and protein was consistently lower compared with native GAA. Genetically modified microglial cells in brains were detected at low levels but provided robust phenotypic correction. Furthermore, an amino acid substitution introduced in the tag reduced insulin receptor-mediated signaling with no evidence of an effect on blood glucose levels in Pompe mice. This study demonstrated the therapeutic potential of lentiviral HSPC gene therapy exploiting optimized GAA tagged coding sequences to reverse Pompe disease pathology in a preclinical mouse model, providing promising vector candidates for further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Chris Mason
- AVROBIO, Inc., Cambridge, MA 02139, USA
- Advanced Centre for Biochemical Engineering, University College London, London WC1E 6AE, UK
- Corresponding author: Chris Mason, Advanced Centre for Biochemical Engineering, University College London, London WC1E 6AE, UK
| | - Niek P. van Til
- AVROBIO, Inc., Cambridge, MA 02139, USA
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1081 HV Amsterdam, the Netherlands
- Corresponding author: Niek P. van Til, Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
14
|
van den Broek SL, Shalgunov V, Herth MM. Transport of nanomedicines across the blood-brain barrier: Challenges and opportunities for imaging and therapy. BIOMATERIALS ADVANCES 2022; 141:213125. [PMID: 36182833 DOI: 10.1016/j.bioadv.2022.213125] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
The blood-brain barrier (BBB) is a protective and semipermeable border of endothelial cells that prevents toxins and foreign bodies to enter and damage the brain. Unfortunately, the BBB also hampers the development of pharmaceuticals targeting receptors, enzymes, or other proteins that lie beyond this barrier. Especially large molecules, such as monoclonal antibodies (mAbs) or nanoparticles, are prevented to enter the brain. The limited passage of these molecules partly explains why nanomedicines - targeting brain diseases - have not made it into the clinic to a great extent. As nanomedicines can target a wide range of targets including protein isoforms and oligomers or potentially deliver cytotoxic drugs safely to their targets, a pathway to smuggle nanomedicines into the brain would allow to treat brain diseases that are currently considered 'undruggable'. In this review, strategies to transport nanomedicines over the BBB will be discussed. Their challenges and opportunities will be highlighted with respect to their use for molecular imaging or therapies. Several strategies have been explored for this thus far. For example, carrier-mediated and receptor-mediated transcytosis (RMT), techniques to disrupt the BBB, nasal drug delivery or administering nanomedicines directly into the brain have been explored. RMT has been the most widely and successfully explored strategy. Recent work on the use of focused ultrasound based BBB opening has shown great promise. For example, successful delivery of mAbs into the brain has been achieved, even in a clinical setting. As nanomedicines bear the potential to treat incurable brain diseases, drug delivery technologies that can deliver nanomedicines into the brain will play an essential role for future treatment options.
Collapse
Affiliation(s)
- Sara Lopes van den Broek
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| |
Collapse
|
15
|
Parrasia S, Szabò I, Zoratti M, Biasutto L. Peptides as Pharmacological Carriers to the Brain: Promises, Shortcomings and Challenges. Mol Pharm 2022; 19:3700-3729. [PMID: 36174227 DOI: 10.1021/acs.molpharmaceut.2c00523] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Central nervous system (CNS) diseases are among the most difficult to treat, mainly because the vast majority of the drugs fail to cross the blood-brain barrier (BBB) or to reach the brain at concentrations adequate to exert a pharmacological activity. The obstacle posed by the BBB has led to the in-depth study of strategies allowing the brain delivery of CNS-active drugs. Among the most promising strategies is the use of peptides addressed to the BBB. Peptides are versatile molecules that can be used to decorate nanoparticles or can be conjugated to drugs, with either a stable link or as pro-drugs. They have been used to deliver to the brain both small molecules and proteins, with applications in diverse therapeutic areas such as brain cancers, neurodegenerative diseases and imaging. Peptides can be generally classified as receptor-targeted, recognizing membrane proteins expressed by the BBB microvessels (e.g., Angiopep2, CDX, and iRGD), "cell-penetrating peptides" (CPPs; e.g. TAT47-57, SynB1/3, and Penetratin), undergoing transcytosis through unspecific mechanisms, or those exploiting a mixed approach. The advantages of peptides have been extensively pointed out, but so far few studies have focused on the potential negative aspects. Indeed, despite having a generally good safety profile, some peptide conjugates may display toxicological characteristics distinct from those of the peptide itself, causing for instance antigenicity, cardiovascular alterations or hemolysis. Other shortcomings are the often brief lifetime in vivo, caused by the presence of peptidases, the vulnerability to endosomal/lysosomal degradation, and the frequently still insufficient attainable increase of brain drug levels, which remain below the therapeutically useful concentrations. The aim of this review is to analyze not only the successful and promising aspects of the use of peptides in brain targeting but also the problems posed by this strategy for drug delivery.
Collapse
Affiliation(s)
- Sofia Parrasia
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| |
Collapse
|
16
|
Kakava S, Schlumpf E, Panteloglou G, Tellenbach F, von Eckardstein A, Robert J. Brain Endothelial Cells in Contrary to the Aortic Do Not Transport but Degrade Low-Density Lipoproteins via Both LDLR and ALK1. Cells 2022; 11:cells11193044. [PMID: 36231005 PMCID: PMC9564369 DOI: 10.3390/cells11193044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
The transport of low-density lipoprotein (LDL) through the endothelium is a key step in the development of atherosclerosis, but it is notorious that phenotypic differences exist between endothelial cells originating from different vascular beds. Endothelial cells forming the blood–brain barrier restrict paracellular and transcellular passage of plasma proteins. Here, we systematically compared brain versus aortic endothelial cells towards their interaction with LDL and the role of proteins known to regulate the uptake of LDL by endothelial cells. Both brain endothelial cells and aortic endothelial cells bind and internalize LDL. However, whereas aortic endothelial cells degrade very small amounts of LDL and transcytose the majority, brain endothelial cells degrade but do not transport LDL. Using RNA interference (siRNA), we found that the LDLR–clathrin pathway leads to LDL degradation in either endothelial cell type. Both loss- and gain-of-function experiments showed that ALK1, which promotes transcellular LDL transport in aortic endothelial cells, also limits LDL degradation in brain endothelial cells. SR-BI and caveolin-1, which promote LDL uptake and transport into aortic endothelial cells, limit neither binding nor association of LDL to brain endothelial cells. Together, these results indicate distinct LDL trafficking by brain microvascular endothelial cells and aortic endothelial cells.
Collapse
Affiliation(s)
- Sofia Kakava
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
- Bio Medicine Program, Life Science Zurich Graduate School, University of Zurich, 8000 Zurich, Switzerland
| | - Eveline Schlumpf
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
| | - Grigorios Panteloglou
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
| | - Flavia Tellenbach
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
- Bio Medicine Program, Life Science Zurich Graduate School, University of Zurich, 8000 Zurich, Switzerland
| | - Jerome Robert
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
- Correspondence: or
| |
Collapse
|
17
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
18
|
Gao X, Xu J, Yao T, Liu X, Zhang H, Zhan C. Peptide-decorated nanocarriers penetrating the blood-brain barrier for imaging and therapy of brain diseases. Adv Drug Deliv Rev 2022; 187:114362. [PMID: 35654215 DOI: 10.1016/j.addr.2022.114362] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
Abstract
Blood-Brain Barrier (BBB) is one of the most important physiological barriers strictly restricting the substance exchange between blood and brain tissues. While the BBB protects the brain from infections and toxins and maintains brain homeostasis, it is also recognized as the main obstacle to the penetration of therapeutics and imaging agents into the brain. Due to high specificity and affinity, peptides are frequently exploited to decorate nanocarriers across the BBB for diagnosis and/or therapy purposes. However, there are still some challenges that restrict their clinical application, such as stability, safety and immunocompatibility. In this review, we summarize the biological and pathophysiological characteristics of the BBB, strategies across the BBB, and recent progress on peptide decorated nanocarriers for brain diseases diagnosis and therapy. The challenges and opportunities for their translation are also discussed.
Collapse
|
19
|
Borrajo ML, Alonso MJ. Using nanotechnology to deliver biomolecules from nose to brain - peptides, proteins, monoclonal antibodies and RNA. Drug Deliv Transl Res 2022; 12:862-880. [PMID: 34731414 PMCID: PMC8888512 DOI: 10.1007/s13346-021-01086-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
There is a growing number of biomolecules, including peptides, proteins, monoclonal antibodies and RNA, that could be potentially used for the treatment of central nervous system (CNS) diseases. However, the realization of their potential is being hampered by the extraordinary difficulties these complex biomolecules have to reach the brain in therapeutically meaningful amounts. Nose-to-brain (N-to-B) delivery is now being investigated as a potential option for the direct transport of biomolecules from the nasal cavity to different brain areas. Here, we discuss how different technological approaches enhance this N-to-B transport, with emphasis on those that have shown a potential for clinical translation. We also analyse how the physicochemical properties of nanocarriers and their modification with cell-penetrating peptides (CPPs) and targeting ligands affect their efficacy as N-to-B carriers for biomolecules.
Collapse
Affiliation(s)
- Mireya L Borrajo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, 15782, Santiago de Compostela, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, 15782, Santiago de Compostela, Spain.
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
20
|
Choi H, Choi K, Kim DH, Oh BK, Yim H, Jo S, Choi C. Strategies for Targeted Delivery of Exosomes to the Brain: Advantages and Challenges. Pharmaceutics 2022; 14:672. [PMID: 35336049 PMCID: PMC8948948 DOI: 10.3390/pharmaceutics14030672] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 02/08/2023] Open
Abstract
Delivering therapeutics to the central nervous system (CNS) is difficult because of the blood-brain barrier (BBB). Therapeutic delivery across the tight junctions of the BBB can be achieved through various endogenous transportation mechanisms. Receptor-mediated transcytosis (RMT) is one of the most widely investigated and used methods. Drugs can hijack RMT by expressing specific ligands that bind to receptors mediating transcytosis, such as the transferrin receptor (TfR), low-density lipoprotein receptor (LDLR), and insulin receptor (INSR). Cell-penetrating peptides and viral components originating from neurotropic viruses can also be utilized for the efficient BBB crossing of therapeutics. Exosomes, or small extracellular vesicles, have gained attention as natural nanoparticles for treating CNS diseases, owing to their potential for natural BBB crossing and broad surface engineering capability. RMT-mediated transport of exosomes expressing ligands such as LDLR-targeting apolipoprotein B has shown promising results. Although surface-modified exosomes possessing brain targetability have shown enhanced CNS delivery in preclinical studies, the successful development of clinically approved exosome therapeutics for CNS diseases requires the establishment of quantitative and qualitative methods for monitoring exosomal delivery to the brain parenchyma in vivo as well as elucidation of the mechanisms underlying the BBB crossing of surface-modified exosomes.
Collapse
Affiliation(s)
- Hojun Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Kyungsun Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Dae-Hwan Kim
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Byung-Koo Oh
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Hwayoung Yim
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Soojin Jo
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Chulhee Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
21
|
Brookes A, Ji L, Bradshaw TD, Stocks M, Gray D, Butler J, Gershkovich P. Is Oral Lipid-Based Delivery for Drug Targeting to the Brain Feasible? Eur J Pharm Biopharm 2022; 172:112-122. [PMID: 35149190 DOI: 10.1016/j.ejpb.2022.02.004] [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: 10/18/2021] [Revised: 01/21/2022] [Accepted: 02/05/2022] [Indexed: 12/12/2022]
Abstract
This review outlines the feasibility of oral lipid-based targeted delivery of drugs to the brain, including permeation of the central nervous system's (CNS) protective blood-brain barrier (BBB). The structure of the BBB and disruption caused by varying disease states highlights the need for disease-specific approaches to alter permeation. Disruption during disease state, and the effects of certain molecules on the barrier, demonstrate the possibility of exploiting such BBB disruption for drug delivery. Many administration methods can be used to target the brain, but oral administration is considered ideal for chronic, long-term illnesses. Several lipids that have been shown to facilitate drug delivery into the brain after systemic administration, but could also be delivered orally are discussed, including oleic acid, triolein, alkylglycerol, and conjugates of linoleic and myristic acids. Current data reveal the potential for the use of such lipids as part of oral formulations for delivery to the brain by reaching sufficient plasma levels after administration to increase the permeability of the BBB. However, gaps in the literature remain regarding the concentrations and form of most lipids required to produce the desired effects. The use of lipids via oral delivery for brain targeting has not been investigated thoroughly enough to determine with certainty if similar permeability-enhancing effects would be observed as for parenteral administration. In conclusion, further research to fill research gaps is needed, but the limited evidence suggests that oral lipid-based drug delivery for brain targeting is potentially feasible.
Collapse
Affiliation(s)
- Alice Brookes
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Liuhang Ji
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Tracey D Bradshaw
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Michael Stocks
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - David Gray
- Division of Food, Nutrition and Dietetics, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, UK, LE12 5RD
| | - James Butler
- GlaxoSmithKline Research and Development, Park Road, Ware, Hertfordshire, UK, SG12 0DP
| | - Pavel Gershkovich
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD.
| |
Collapse
|
22
|
Cantore A, Fraldi A, Meneghini V, Gritti A. In vivo Gene Therapy to the Liver and Nervous System: Promises and Challenges. Front Med (Lausanne) 2022; 8:774618. [PMID: 35118085 PMCID: PMC8803894 DOI: 10.3389/fmed.2021.774618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 12/16/2021] [Indexed: 12/02/2022] Open
Abstract
In vivo genetic engineering has recently shown remarkable potential as a novel effective treatment for an ever-growing number of diseases, as also witnessed by the recent marketing authorization of several in vivo gene therapy products. In vivo genetic engineering comprises both viral vector-mediated gene transfer and the more recently developed genome/epigenome editing strategies, as long as they are directly administered to patients. Here we first review the most advanced in vivo gene therapies that are commercially available or in clinical development. We then highlight the major challenges to be overcome to fully and broadly exploit in vivo gene therapies as novel medicines, discussing some of the approaches that are being taken to address them, with a focus on the nervous system and liver taken as paradigmatic examples.
Collapse
Affiliation(s)
- Alessio Cantore
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- *Correspondence: Alessio Cantore
| | - Alessandro Fraldi
- CEINGE Biotecnologie Avanzate, Naples, Italy
- Department of Translational Medicine, University of Naples “Federico II”, Naples, Italy
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
23
|
Spencer B, Rissman RA, Overk C, Masliah E. Novel Brain-Penetrating Single Chain Antibodies Directed Against 3RTau for the Treatment of Alzheimer's Disease and Related Dementias. Methods Mol Biol 2022; 2383:447-457. [PMID: 34766306 DOI: 10.1007/978-1-0716-1752-6_28] [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] [Indexed: 02/23/2024]
Abstract
Alzheimer's disease (AD), Pick's disease, fronto-temporal lobar degeneration, cortico-basal degeneration, and primary age related tauopathy are examples of neurodegenerative disorders with tau accumulation and jointly referred as "tauopathies." The mechanisms through which tau leads to neurodegeneration are not fully understood but include conversion into toxic oligomers and protofibrils, cell-to-cell propagation, post-transcriptional modifications and as a mediator of cell death signals among others. Potential therapeutics includes reducing tau synthesis (e.g., anti-sense); targeting selective tau species and aggregates or blocking cell-to-cell transmission (e.g., antibodies) or by promoting clearance of tau (e.g., autophagy activators). Among them, immunotherapy is currently one of the approaches most actively explored including active, passive, and cellular. A potential problem with immunotherapy has been the trafficking of the antibodies into the CNS. In this chapter, we describe a method for the production and testing of viral vector driven, brain-penetrating, single chain antibodies that specifically recognize 3RTau. These single chain antibodies are modified by the addition of a fragment of the apoB protein to facilitate trafficking into the brain, once in the CNS these antibody fragments recognize tau with potential value for the treatment of AD and related dementias.
Collapse
Affiliation(s)
- Brian Spencer
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Cassia Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA.
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
24
|
Delivery of Therapeutic Agents to the Central Nervous System and the Promise of Extracellular Vesicles. Pharmaceutics 2021; 13:pharmaceutics13040492. [PMID: 33916841 PMCID: PMC8067091 DOI: 10.3390/pharmaceutics13040492] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
The central nervous system (CNS) is surrounded by the blood–brain barrier (BBB), a semipermeable border of endothelial cells that prevents pathogens, solutes and most molecules from non-selectively crossing into the CNS. Thus, the BBB acts to protect the CNS from potentially deleterious insults. Unfortunately, the BBB also frequently presents a significant barrier to therapies, impeding passage of drugs and biologicals to target cells within the CNS. This review provides an overview of different approaches to deliver therapeutics across the BBB, with an emphasis in extracellular vesicles as delivery vehicles to the CNS.
Collapse
|
25
|
D'Souza A, Dave KM, Stetler RA, S. Manickam D. Targeting the blood-brain barrier for the delivery of stroke therapies. Adv Drug Deliv Rev 2021; 171:332-351. [PMID: 33497734 DOI: 10.1016/j.addr.2021.01.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
A variety of neuroprotectants have shown promise in treating ischemic stroke, yet their delivery to the brain remains a challenge. The endothelial cells lining the blood-brain barrier (BBB) are emerging as a dynamic factor in the response to neurological injury and disease, and the endothelial-neuronal matrix coupling is fundamentally neuroprotective. In this review, we discuss approaches that target the endothelium for drug delivery both across the BBB and to the BBB as a viable strategy to facilitate neuroprotective effects, using the example of brain-derived neurotrophic factor (BDNF). We highlight the advances in cell-derived extracellular vesicles (EVs) used for CNS targeting and drug delivery. We also discuss the potential of engineered EVs as a potent strategy to deliver BDNF or other drug candidates to the ischemic brain, particularly when coupled with internal components like mitochondria that may increase cellular energetics in injured endothelial cells.
Collapse
|
26
|
Anthony DP, Hegde M, Shetty SS, Rafic T, Mutalik S, Rao BSS. Targeting receptor-ligand chemistry for drug delivery across blood-brain barrier in brain diseases. Life Sci 2021; 274:119326. [PMID: 33711385 DOI: 10.1016/j.lfs.2021.119326] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/09/2021] [Accepted: 03/03/2021] [Indexed: 12/18/2022]
Abstract
The blood-brain barrier (BBB) is composed of a layer of endothelial cells that is interspersed with a series of tight junctions and characterized by the absence of fenestrations. The permeability of this barrier is controlled by junctions such as tight junctions and adherent junctions as well as several cells such as astrocytes, pericytes, vascular endothelial cells, neurons, microglia, and efflux transporters with relatively enhanced expression. It plays a major role in maintaining homeostasis in the brain and exerts a protective regulatory control on the influx and efflux of molecules. However, it proves to be a challenge for drug delivery strategies that target brain diseases like Dementia, Parkinson's Disease, Alzheimer's Disease, Brain Cancer or Stroke, Huntington's Disease, Lou Gehrig's Disease, etc. Conventional modes of drug delivery are invasive and have been known to contribute to a "leaky BBB", recent studies have highlighted the efficiency and relative safety of receptor-mediated drug delivery. Several receptors are exhibited on the BBB, and actively participate in nutrient uptake, and recognize specific ligands that modulate the process of endocytosis. The strategy employed in receptor-mediated drug delivery exploits this process of "tricking" the receptors into internalizing ligands that are conjugated to carrier systems like liposomes, nanoparticles, monoclonal antibodies, enzymes etc. These in turn are modified with drug molecules, therefore leading to delivery to desired target cells in brain tissue. This review comprehensively explores each of those receptors that can be modified to serve such purposes as well as the currently employed strategies that have led to increased cellular uptake and transport efficiency.
Collapse
Affiliation(s)
- Danielle Paige Anthony
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Manasa Hegde
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shreya S Shetty
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Thasneema Rafic
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - B S Satish Rao
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
27
|
Seras‐Franzoso J, Díaz‐Riascos ZV, Corchero JL, González P, García‐Aranda N, Mandaña M, Riera R, Boullosa A, Mancilla S, Grayston A, Moltó‐Abad M, Garcia‐Fruitós E, Mendoza R, Pintos‐Morell G, Albertazzi L, Rosell A, Casas J, Villaverde A, Schwartz S, Abasolo I. Extracellular vesicles from recombinant cell factories improve the activity and efficacy of enzymes defective in lysosomal storage disorders. J Extracell Vesicles 2021; 10:e12058. [PMID: 33738082 PMCID: PMC7953474 DOI: 10.1002/jev2.12058] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/16/2020] [Accepted: 01/05/2021] [Indexed: 12/20/2022] Open
Abstract
In the present study the use of extracellular vesicles (EVs) as vehicles for therapeutic enzymes in lysosomal storage disorders was explored. EVs were isolated from mammalian cells overexpressing alpha-galactosidase A (GLA) or N-sulfoglucosamine sulfohydrolase (SGSH) enzymes, defective in Fabry and Sanfilippo A diseases, respectively. Direct purification of EVs from cell supernatants was found to be a simple and efficient method to obtain highly active GLA and SGSH proteins, even after EV lyophilization. Likewise, EVs carrying GLA (EV-GLA) were rapidly uptaken and reached the lysosomes in cellular models of Fabry disease, restoring lysosomal functionality much more efficiently than the recombinant enzyme in clinical use. In vivo, EVs were well tolerated and distributed among all main organs, including the brain. DiR-labelled EVs were localized in brain parenchyma 1 h after intra-arterial (internal carotid artery) or intravenous (tail vein) administrations. Moreover, a single intravenous administration of EV-GLA was able to reduce globotriaosylceramide (Gb3) substrate levels in clinically relevant tissues, such kidneys and brain. Overall, our results demonstrate that EVs from cells overexpressing lysosomal enzymes act as natural protein delivery systems, improving the activity and the efficacy of the recombinant proteins and facilitating their access to organs neglected by conventional enzyme replacement therapies.
Collapse
|
28
|
He P, Schulz P, Sierks MR. A conformation-specific antibody against oligomeric β-amyloid restores neuronal integrity in a mouse model of Alzheimer's disease. J Biol Chem 2021; 296:100241. [PMID: 33376140 PMCID: PMC7948963 DOI: 10.1074/jbc.ra120.015327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/03/2020] [Accepted: 12/29/2020] [Indexed: 11/06/2022] Open
Abstract
Conformationally distinct aggregates of the amyloid β (Aβ) peptide accumulate in brains of patients with Alzheimer's disease (AD), but the roles of the different aggregates in disease progression are not clear. We previously isolated two single-chain variable domain antibody fragments (scFvs), C6T and A4, that selectively bind different toxic conformational variants of oligomeric Aβ. Here, we utilize these scFvs to localize the presence of these Aβ variants in human AD brain and to demonstrate their potential as therapeutic agents for treating AD. Both A4 and C6T label oligomeric Aβ in extracellular amyloid plaques, whereas C6T also labels intracellular oligomeric Aβ in human AD brain tissue and in an AD mouse model. For therapeutic studies, the A4 and C6T scFvs were expressed in the AD mice by viral infection of liver cells. The scFvs were administered at 2 months of age, and mice sacrificed at 9 months. The scFvs contained a peptide tag to facilitate transport across the blood brain barrier. While treatment with C6T only slightly decreased Aβ deposits and plaque-associated inflammation, it restored neuronal integrity to WT levels, significantly promoted growth of new neurons, and impressively rescued survival rates to WT levels. Treatment with A4 on the other hand significantly decreased Aβ deposits but did not significantly decrease neuroinflammation or promote neuronal integrity, neurogenesis, or survival rate. These results suggest that the specific Aβ conformation targeted in therapeutic applications greatly affects the outcome, and the location of the targeted Aβ variants may also play a critical factor.
Collapse
Affiliation(s)
- Ping He
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, USA
| | - Philip Schulz
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, USA
| | - Michael R Sierks
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, USA.
| |
Collapse
|
29
|
Zaghmi A, Drouin-Ouellet J, Brambilla D, Gauthier MA. Treating brain diseases using systemic parenterally-administered protein therapeutics: Dysfunction of the brain barriers and potential strategies. Biomaterials 2020; 269:120461. [PMID: 33218788 DOI: 10.1016/j.biomaterials.2020.120461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022]
Abstract
The parenteral administration of protein therapeutics is increasingly gaining importance for the treatment of human diseases. However, the presence of practically impermeable blood-brain barriers greatly restricts access of such pharmaceutics to the brain. Treating brain disorders with proteins thus remains a great challenge, and the slow clinical translation of these therapeutics may be largely ascribed to the lack of appropriate brain delivery system. Exploring new approaches to deliver proteins to the brain by circumventing physiological barriers is thus of great interest. Moreover, parallel advances in the molecular neurosciences are important for better characterizing blood-brain interfaces, particularly under different pathological conditions (e.g., stroke, multiple sclerosis, Parkinson's disease, and Alzheimer's disease). This review presents the current state of knowledge of the structure and the function of the main physiological barriers of the brain, the mechanisms of transport across these interfaces, as well as alterations to these concomitant with brain disorders. Further, the different strategies to promote protein delivery into the brain are presented, including the use of molecular Trojan horses, the formulation of nanosystems conjugated/loaded with proteins, protein-engineering technologies, the conjugation of proteins to polymers, and the modulation of intercellular junctions. Additionally, therapeutic approaches for brain diseases that do not involve targeting to the brain are presented (i.e., sink and scavenging mechanisms).
Collapse
Affiliation(s)
- A Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada
| | - J Drouin-Ouellet
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - D Brambilla
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - M A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada.
| |
Collapse
|
30
|
Ibeanu N, Egbu R, Onyekuru L, Javaheri H, Tee Khaw P, R. Williams G, Brocchini S, Awwad S. Injectables and Depots to Prolong Drug Action of Proteins and Peptides. Pharmaceutics 2020; 12:E999. [PMID: 33096803 PMCID: PMC7589296 DOI: 10.3390/pharmaceutics12100999] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
Proteins and peptides have emerged in recent years to treat a wide range of multifaceted diseases such as cancer, diabetes and inflammation. The emergence of polypeptides has yielded advancements in the fields of biopharmaceutical production and formulation. Polypeptides often display poor pharmacokinetics, limited permeability across biological barriers, suboptimal biodistribution, and some proclivity for immunogenicity. Frequent administration of polypeptides is generally required to maintain adequate therapeutic levels, which can limit efficacy and compliance while increasing adverse reactions. Many strategies to increase the duration of action of therapeutic polypeptides have been described with many clinical products having been developed. This review describes approaches to optimise polypeptide delivery organised by the commonly used routes of administration. Future innovations in formulation may hold the key to the continued successful development of proteins and peptides with optimal clinical properties.
Collapse
Affiliation(s)
- Nkiruka Ibeanu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Raphael Egbu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Lesley Onyekuru
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Hoda Javaheri
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Peng Tee Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Gareth R. Williams
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Steve Brocchini
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Sahar Awwad
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| |
Collapse
|
31
|
Loupy KM, Lee T, Zambrano CA, Elsayed AI, D'Angelo HM, Fonken LK, Frank MG, Maier SF, Lowry CA. Alzheimer's Disease: Protective Effects of Mycobacterium vaccae, a Soil-Derived Mycobacterium with Anti-Inflammatory and Anti-Tubercular Properties, on the Proteomic Profiles of Plasma and Cerebrospinal Fluid in Rats. J Alzheimers Dis 2020; 78:965-987. [PMID: 33074227 DOI: 10.3233/jad-200568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is an inflammatory neurodegenerative disease that may be associated with prior bacterial infections. Microbial "old friends" can suppress exaggerated inflammation in response to disease-causing infections or increase clearance of pathogens such as Mycobacterium tuberculosis, which causes tuberculosis (TB). One such "old friend" is Mycobacterium vaccae NCTC 11659, a soil-derived bacterium that has been proposed either as a vaccine for prevention of TB, or as immunotherapy for the treatment of TB when used alongside first line anti-TB drug treatment. OBJECTIVE The goal of this study was to use a hypothesis generating approach to explore the effects of M. vaccae on physiological changes in the plasma and cerebrospinal fluid (CSF). METHODS Liquid chromatography-tandem mass spectrometry-based proteomics were performed in plasma and CSF of adult male rats after immunization with a heat-killed preparation of M. vaccae NCTC 11659 or borate-buffered saline vehicle. Gene enrichment analysis and analysis of protein-protein interactions were performed to integrate physiological network changes in plasma and CSF. We used RT-qPCR to assess immune and metabolic gene expression changes in the hippocampus. RESULTS In both plasma and CSF, immunization with M. vaccae increased proteins associated with immune activation and downregulated proteins corresponding to lipid (including phospholipid and cholesterol) metabolism. Immunization with M. vaccae also increased hippocampal expression of interleukin-4 (IL-4) mRNA, implicating anti-inflammatory effects in the central nervous system. CONCLUSION M. vaccae alters host immune activity and lipid metabolism. These data are consistent with the hypothesis that microbe-host interactions may protect against possible infection-induced, inflammation-related cognitive impairments.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Thomas Lee
- Central Analytical Laboratory and Mass Spectrometry Facility, Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Cristian A Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Ahmed I Elsayed
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Heather M D'Angelo
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Senior Fellow, inVIVO Planetary Health, of the Worldwide Universities Network (WUN), West New York, NJ, USA
| |
Collapse
|
32
|
Nonaka M, Suzuki-Anekoji M, Nakayama J, Mabashi-Asazuma H, Jarvis DL, Yeh JC, Yamasaki K, Akama TO, Huang CT, Campos AR, Nagaoka M, Sasai T, Kimura-Takagi I, Suwa Y, Yaegashi T, Shibata TK, Sugihara K, Nishizawa-Harada C, Fukuda M, Fukuda MN. Overcoming the blood-brain barrier by Annexin A1-binding peptide to target brain tumours. Br J Cancer 2020; 123:1633-1643. [PMID: 32921792 PMCID: PMC7686308 DOI: 10.1038/s41416-020-01066-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/12/2020] [Accepted: 08/26/2020] [Indexed: 11/09/2022] Open
Abstract
Background Annexin A1 is expressed specifically on the tumour vasculature surface. Intravenously injected IF7 targets tumour vasculature via annexin A1. We tested the hypothesis that IF7 overcomes the blood–brain barrier and that the intravenously injected IF7C(RR)-SN38 eradicates brain tumours in the mouse. Methods (1) A dual-tumour model was generated by inoculating luciferase-expressing melanoma B16 cell line, B16-Luc, into the brain and under the skin of syngeneic C57BL/6 mice. IF7C(RR)-SN38 was injected intravenously daily at 7.0 μmoles/kg and growth of tumours was assessed by chemiluminescence using an IVIS imager. A similar dual-tumour model was generated with the C6-Luc line in immunocompromised SCID mice. (2) IF7C(RR)-SN38 formulated with 10% Solutol HS15 was injected intravenously daily at 2.5 μmoles/kg into two brain tumour mouse models: B16-Luc cells in C57BL/6 mice, and C6-Luc cells in nude mice. Results (1) Daily IF7C(RR)-SN38 injection suppressed tumour growth regardless of cell lines or mouse strains. (2) Daily injection of Solutol-formulated IF7C(RR)-SN38 led into complete disappearance of B16-Luc brain tumour in C57BL/6 mice, whereas this did not occur in C6-Luc in nude mice. Conclusions IF7C(RR)-SN38 crosses the blood–brain barrier and suppresses growth of brain tumours in mouse models. Solutol HS15-formulated IF7C(RR)-SN38 may have promoted an antitumour immune response.
Collapse
Affiliation(s)
- Motohiro Nonaka
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.,Laboratory for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8568, Japan.,Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Misa Suzuki-Anekoji
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | | | - Donald L Jarvis
- Department of Molecular Biology, University of Wyoming, Laramie, WY, 82071, USA
| | - Jiunn-Chern Yeh
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Kazuhiko Yamasaki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8566, Japan
| | - Tomoya O Akama
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Chun-Teng Huang
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Alexandre Rosa Campos
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Masato Nagaoka
- Yakult Central Institute, Kunitachi, Tokyo, 186-8650, Japan
| | - Toshio Sasai
- Yakult Central Institute, Kunitachi, Tokyo, 186-8650, Japan
| | | | - Yoichi Suwa
- Yakult Central Institute, Kunitachi, Tokyo, 186-8650, Japan
| | | | - Toshiaki K Shibata
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.,Department of Gynecology and Obstetrics, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Kazuhiro Sugihara
- Department of Gynecology and Obstetrics, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Chizuko Nishizawa-Harada
- Laboratory for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8568, Japan
| | - Minoru Fukuda
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Michiko N Fukuda
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA. .,Laboratory for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8568, Japan.
| |
Collapse
|
33
|
Kwon S, Iba M, Kim C, Masliah E. Immunotherapies for Aging-Related Neurodegenerative Diseases-Emerging Perspectives and New Targets. Neurotherapeutics 2020; 17:935-954. [PMID: 32347461 PMCID: PMC7222955 DOI: 10.1007/s13311-020-00853-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neurological disorders such as Alzheimer's disease (AD), Lewy body dementia (LBD), frontotemporal dementia (FTD), and vascular dementia (VCID) have no disease-modifying treatments to date and now constitute a dementia crisis that affects 5 million in the USA and over 50 million worldwide. The most common pathological hallmark of these age-related neurodegenerative diseases is the accumulation of specific proteins, including amyloid beta (Aβ), tau, α-synuclein (α-syn), TAR DNA-binding protein 43 (TDP43), and repeat-associated non-ATG (RAN) peptides, in the intra- and extracellular spaces of selected brain regions. Whereas it remains controversial whether these accumulations are pathogenic or merely a byproduct of disease, the majority of therapeutic research has focused on clearing protein aggregates. Immunotherapies have garnered particular attention for their ability to target specific protein strains and conformations as well as promote clearance. Immunotherapies can also be neuroprotective: by neutralizing extracellular protein aggregates, they reduce spread, synaptic damage, and neuroinflammation. This review will briefly examine the current state of research in immunotherapies against the 3 most commonly targeted proteins for age-related neurodegenerative disease: Aβ, tau, and α-syn. The discussion will then turn to combinatorial strategies that enhance the effects of immunotherapy against aggregating protein, followed by new potential targets of immunotherapy such as aging-related processes.
Collapse
Affiliation(s)
- Somin Kwon
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michiyo Iba
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Changyoun Kim
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Eliezer Masliah
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA.
- Division of Neuroscience, National Institute on Aging/National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
34
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxonl CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Peptide based drug delivery systems to the brain. NANO EXPRESS 2020. [DOI: 10.1088/2632-959x/ab9008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Li Y, Wei J, Wei Y, Cheng L, Guo B, Meng F, Li F, Zhong Z. Apolipoprotein E Peptide-Guided Disulfide-Cross-Linked Micelles for Targeted Delivery of Sorafenib to Hepatocellular Carcinoma. Biomacromolecules 2019; 21:716-724. [DOI: 10.1021/acs.biomac.9b01419] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | | | | | | | | | | | - Feng Li
- Department of Respiratory Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, P. R. China
| | | |
Collapse
|
36
|
Chen YX, Wei CX, Lyu YQ, Chen HZ, Jiang G, Gao XL. Biomimetic drug-delivery systems for the management of brain diseases. Biomater Sci 2019; 8:1073-1088. [PMID: 31728485 DOI: 10.1039/c9bm01395d] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acting as a double-edged sword, the blood-brain barrier (BBB) is essential for maintaining brain homeostasis by restricting the entry of small molecules and most macromolecules from blood. However, it also largely limits the brain delivery of most drugs. Even if a drug can penetrate the BBB, its accumulation in the intracerebral pathological regions is relatively low. Thus, an optimal drug-delivery system (DDS) for the management of brain diseases needs to display BBB permeability, lesion-targeting capability, and acceptable safety. Biomimetic DDSs, developed by directly utilizing or mimicking the biological structures and processes, provide promising approaches for overcoming the barriers to brain drug delivery. The present review summarizes the biological properties and biomedical applications of the biomimetic DDSs including the cell membrane-based DDS, lipoprotein-based DDS, exosome-based DDS, virus-based DDS, protein template-based DDS and peptide template-based DDS for the management of brain diseases.
Collapse
Affiliation(s)
- Yao-Xing Chen
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Chen-Xuan Wei
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Ying-Qi Lyu
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China. and Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201210, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Xiao-Ling Gao
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
37
|
Zou Z, Shao S, Zou R, Qi J, Chen L, Zhang H, Shen Q, Yang Y, Ma L, Guo R, Li H, Tian H, Li P, Yu M, Wang L, Kong W, Li C, Yu Z, Huang Y, Chen L, Shao Q, Gao X, Chen X, Zhang Z, Yan J, Shao X, Pan R, Xu L, Fang J, Zhao L, Huang Y, Li A, Zhang Y, Huang W, Tian K, Hu M, Xie L, Wu L, Wu Y, Luo Z, Xiao W, Ma S, Wang J, Huang K, He S, Yang F, Zhou S, Jia M, Zhang H, Lu H, Wang X, Tan J. Linking the low-density lipoprotein receptor-binding segment enables the therapeutic 5-YHEDA peptide to cross the blood-brain barrier and scavenge excess iron and radicals in the brain of senescent mice. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:717-731. [PMID: 31921964 PMCID: PMC6944740 DOI: 10.1016/j.trci.2019.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Introduction Iron accumulates in the brain during aging, which catalyzes radical formation, causing neuronal impairment, and is thus considered a pathogenic factor in Alzheimer's disease (AD). To scavenge excess iron-catalyzed radicals and thereby protect the brain and decrease the incidence of AD, we synthesized a soluble pro-iron 5-YHEDA peptide. However, the blood-brain barrier (BBB) blocks large drug molecules from entering the brain and thus strongly reduces their therapeutic effects. However, alternative receptor- or transporter-mediated approaches are possible. Methods A low-density lipoprotein receptor (LDLR)-binding segment of Apolipoprotein B-100 was linked to the 5-YHEDA peptide (bs-5-YHEDA) and intracardially injected into senescent (SN) mice that displayed symptoms of cognitive impairment similar to those of people with AD. Results We successfully delivered 5-YHEDA across the BBB into the brains of the SN mice via vascular epithelium LDLR-mediated endocytosis. The data showed that excess brain iron and radical-induced neuronal necrosis were reduced after the bs-5-YHEDA treatment, together with cognitive amelioration in the SN mouse, and that the senescence-associated ferritin and transferrin increase, anemia and inflammation reversed without kidney or liver injury. Discussion bs-5-YHEDA may be a mild and safe iron remover that can cross the BBB and enter the brain to relieve excessive iron- and radical-induced cognitive disorders.
Collapse
Affiliation(s)
- Zhenyou Zou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China.,Medical School of Taizhou University, Taizhou, ZJ, China.,Biochemistry Department, Purdue University, West Lafayette, USA
| | - Shengxi Shao
- Division of Cell and Molecular Biology, Imperial College London, London, United Kingdom
| | - Ruyi Zou
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Jini Qi
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Liguan Chen
- Zhejiang Armed Police Corps, Hangzhou, ZJ, China
| | - Hui Zhang
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, HN, China
| | - Qiqiong Shen
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Yue Yang
- Clinical Laboratory Department, Wenzhou Medical University, ZJ, China
| | - Liman Ma
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Ruzeng Guo
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Hongwen Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Haibo Tian
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Pengxin Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Mingfang Yu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Lu Wang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Wenjuan Kong
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Caiyu Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Zhenhai Yu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Yuping Huang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Li Chen
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Qi Shao
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Xinyan Gao
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Xiaolin Chen
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Zhengbo Zhang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Jianguo Yan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Xiaoyun Shao
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Ru Pan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Lu Xu
- Clinical Laboratory of Jingyou Hospital, Xiaoshan, ZJ, China
| | - Jing Fang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Lei Zhao
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Yaohui Huang
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Anqi Li
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Yuchong Zhang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Wenkao Huang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Kechun Tian
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Minxin Hu
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Linchao Xie
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Lingbin Wu
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Yu Wu
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Zhen Luo
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Wenxin Xiao
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Shanshan Ma
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Jianan Wang
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Kaixin Huang
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Siyuan He
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Fan Yang
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Shuni Zhou
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Mo Jia
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Hui Zhang
- Pathology Department, Affiliated Hospital of Taizhou University, ZJ, China
| | - Hongsheng Lu
- Pathology Department, Affiliated Hospital of Taizhou University, ZJ, China
| | - Xinjuan Wang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| |
Collapse
|
38
|
Sellers DL, Tan JKY, Pineda JMB, Peeler DJ, Porubsky VL, Olden BR, Salipante SJ, Pun SH. Targeting Ligands Deliver Model Drug Cargo into the Central Nervous System along Autonomic Neurons. ACS NANO 2019; 13:10961-10971. [PMID: 31589023 PMCID: PMC7651855 DOI: 10.1021/acsnano.9b01515] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
While biologic drugs such as proteins, peptides, or nucleic acids have shown promise in the treatment of neurodegenerative diseases, the blood-brain barrier (BBB) severely limits drug delivery to the central nervous system (CNS) after systemic administration. Consequently, drug delivery challenges preclude biological drug candidates from the clinical armamentarium. In order to target drug delivery and uptake into to the CNS, we used an in vivo phage display screen to identify peptides able to target drug-uptake by the vast array of neurons of the autonomic nervous system (ANS). Using next-generation sequencing, we identified 21 candidate targeted ANS-to-CNS uptake ligands (TACL) that enriched bacteriophage accumulation and delivered protein-cargo into the CNS after intraperitoneal (IP) administration. The series of TACL peptides were synthesized and tested for their ability to deliver a model enzyme (NeutrAvidin-horseradish peroxidase fusion) to the brain and spinal cord. Three TACL-peptides facilitated significant active enzyme delivery into the CNS, with limited accumulation in off-target organs. Peptide structure and serum stability is increased when internal cysteine residues are cyclized by perfluoroarylation with decafluorobiphenyl, which increased delivery to the CNS further. TACL-peptide was demonstrated to localize in parasympathetic ganglia neurons in addition to neuronal structures in the hindbrain and spinal cord. By targeting uptake into ANS neurons, we demonstrate the potential for TACL-peptides to bypass the blood-brain barrier and deliver a model drug into the brain and spinal cord.
Collapse
Affiliation(s)
- Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - James-Kevin Y. Tan
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Julio Marco B. Pineda
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - David J. Peeler
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Veronica L. Porubsky
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Brynn R. Olden
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington, 98195, USA
| |
Collapse
|
39
|
Hong SS, Oh KT, Choi HG, Lim SJ. Liposomal Formulations for Nose-to-Brain Delivery: Recent Advances and Future Perspectives. Pharmaceutics 2019; 11:pharmaceutics11100540. [PMID: 31627301 PMCID: PMC6835450 DOI: 10.3390/pharmaceutics11100540] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/01/2019] [Accepted: 10/14/2019] [Indexed: 01/07/2023] Open
Abstract
Restricted drug entry to the brain that is closely associated with the existence of the blood brain barrier (BBB) has limited the accessibility of most potential active therapeutic compounds to the brain from the systemic circulation. Recently, evidences for the presence of direct nose-to-brain drug transport pathways have been accumulated by several studies and an intranasal drug administration route has gained attention as a promising way for providing direct access to the brain without the needs to cross to the BBB. Studies aiming for developing nanoparticles as an intranasal drug carrier have shown considerable promise in overcoming the challenges of intranasal drug delivery route. This review gives a comprehensive overview of works having investigated liposomes as a potential vehicle to deliver drugs to the brain through nose-to-brain route while considering the excellent biocompatibility and high potential of liposomes for clinical development. Herein, studies are reviewed with special emphasis on the impact of formulation factors, such as liposome composition and surface modification of liposomes with targeting moieties, in addition to intranasal environmental factors that may affect the extent/site of absorption of intranasally administered, liposome-encapsulated drugs.
Collapse
Affiliation(s)
- Soon-Seok Hong
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Korea.
| | - Kyung Taek Oh
- College of Pharmacy, Chung-ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea.
| | - Han-Gon Choi
- College of Pharmacy, Hangang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Korea.
| | - Soo-Jeong Lim
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Korea.
| |
Collapse
|
40
|
Karakatsani ME, Blesa J, Konofagou EE. Blood-brain barrier opening with focused ultrasound in experimental models of Parkinson's disease. Mov Disord 2019; 34:1252-1261. [PMID: 31361356 PMCID: PMC7213581 DOI: 10.1002/mds.27804] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease has many symptomatic treatments, but there is no neuroprotective therapy currently available. The evolution of this disease is inexorably progressive, and halting or stopping the neurodegenerative process is a major unmet need. Parkinson's disease motor features at onset are typically limited to 1 body segment, that is, focal signs, and the nigrostriatal degeneration is highly asymmetrical and mainly present in the caudal putamen. Thus, clinically and neurobiologically the process is fairly limited early in its evolution. Tentatively, this would allow the possibility of intervening to halt neurodegeneration at the most vulnerable site. The recent use of new technologies such as focused ultrasound provides interesting prospects. In particular, the possibility of transiently opening the blood-brain barrier to facilitate penetrance of putative neuroprotective agents is a highly attractive approach that could be readily applied to Parkinson's disease. However, because there are currently effective treatments available (ie, dopaminergic pharmacological therapy), more experimental evidence is needed to construct a feasible and practical therapeutic approach to be tested early in the evolution of Parkinson's disease patients. In this review, we provide the current evidence for the application of blood-brain barrier opening in experimental models of Parkinson's disease and discuss its potential clinical applicability. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Elisa Evgenia Konofagou
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
- Department of Radiology, Columbia University, New York, New York, USA
| |
Collapse
|
41
|
Pan X, Sands SA, Yue Y, Zhang K, LeVine SM, Duan D. An Engineered Galactosylceramidase Construct Improves AAV Gene Therapy for Krabbe Disease in Twitcher Mice. Hum Gene Ther 2019; 30:1039-1051. [PMID: 31184217 PMCID: PMC6761594 DOI: 10.1089/hum.2019.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 05/16/2019] [Indexed: 12/30/2022] Open
Abstract
Krabbe disease is an inherited neurodegenerative disease caused by mutations in the galactosylceramidase gene. In the infantile form, patients die before 3 years of age. Systemic adeno-associated virus serotype 9 (AAV9) gene therapy was recently shown to reverse the disease course in human patients in another lethal infantile neurodegenerative disease. To explore AAV9 therapy for Krabbe disease, we engineered a codon-optimized AAV9 galactosylceramidase vector. We further incorporated features to allow AAV9-derived galactosylceramidase to more efficiently cross the blood-brain barrier and be secreted from transduced cells. We tested the optimized vector by a single systemic injection in the twitcher mouse, an authentic Krabbe disease model. Untreated twitcher mice showed characteristic neuropathology and motion defects. They died prematurely with a median life span of 41 days. Intravenous injection in 2-day-old twitcher mice reduced central and peripheral neuropathology and significantly improved the gait pattern and body weight. Noticeably, the median life span was extended to 150 days. Intraperitoneal injection in 6- to 12-day-old twitcher mice also significantly improved the motor function, body weight, and median life span (to 104 days). Our results far exceed the ≤70 days median life span seen in all reported stand-alone systemic AAV therapies. Our study highlights the importance of vector engineering for Krabbe disease gene therapy. The engineered vector warrants further development.
Collapse
Affiliation(s)
- Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Scott A. Sands
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Steven M. LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, Missouri
| |
Collapse
|
42
|
Sustained delivery and molecular targeting of a therapeutic monoclonal antibody to metastases in the central nervous system of mice. Nat Biomed Eng 2019; 3:706-716. [PMID: 31384008 PMCID: PMC6736720 DOI: 10.1038/s41551-019-0434-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 06/26/2019] [Indexed: 11/24/2022]
Abstract
Approximately 15–40% of all cancers develop metastases in the
central nervous system (CNS), yet few therapeutic options exist to treat them.
Cancer therapies based on monoclonal antibodies are widely successful, yet have
limited efficacy against CNS metastases, owing to the low levels of the drug
reaching the tumour site. Here, we show that the encapsulation of rituximab
within a crosslinked zwitterionic polymer layer leads to the sustained release
of rituximab as the crosslinkers are gradually hydrolyzed, enhancing by
approximately 10-fold the CNS levels of the antibody with respect to the
administration of naked rituximab. When the nanocapsules are functionalized with
CXCL13, the ligand for the chemokine receptor CXCR5 frequently found on B-cell
lymphoma, a single dose led to improved control of CXCR5-expressing metastases
in a murine xenograft model of non-Hodgkin lymphoma, and eliminated lymphoma in
a xenografted humanized bone-marrow–liver–thymus mouse model.
Encapsulation and molecular targeting of therapeutic antibodies could become an
option for the treatment of cancers with CNS metastases.
Collapse
|
43
|
Spencer B, Trinh I, Rockenstein E, Mante M, Florio J, Adame A, El-Agnaf OMA, Kim C, Masliah E, Rissman RA. Systemic peptide mediated delivery of an siRNA targeting α-syn in the CNS ameliorates the neurodegenerative process in a transgenic model of Lewy body disease. Neurobiol Dis 2019; 127:163-177. [PMID: 30849508 PMCID: PMC6588505 DOI: 10.1016/j.nbd.2019.03.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/05/2019] [Accepted: 03/04/2019] [Indexed: 02/08/2023] Open
Abstract
Neurodegenerative disorders of the aging population are characterized by progressive accumulation of neuronal proteins such as α-synuclein (α-syn) in Parkinson's Disease (PD) and Amyloid ß (Aß) and Tau in Alzheimer's disease (AD) for which no treatments are currently available. The ability to regulate the expression at the gene transcription level would be beneficial for reducing the accumulation of these proteins or regulating expression levels of other genes in the CNS. Short interfering RNA molecules can bind specifically to target RNAs and deliver them for degradation. This approach has shown promise therapeutically in vitro and in vivo in mouse models of PD and AD and other neurological disorders; however, delivery of the siRNA to the CNS in vivo has been achieved primarily through intra-cerebral or intra-thecal injections that may be less amenable for clinical translation; therefore, alternative approaches for delivery of siRNAs to the brain is needed. Recently, we described a small peptide from the envelope protein of the rabies virus (C2-9r) that was utilized to deliver an siRNA targeting α-syn across the blood brain barrier (BBB) following intravenous injection. This approach showed reduced expression of α-syn and neuroprotection in a toxic mouse model of PD. However, since receptor-mediated delivery is potentially saturable, each allowing the delivery of a limited number of molecules, we identified an alternative peptide for the transport of nucleotides across the BBB based on the apolipoprotein B (apoB) protein targeted to the family of low-density lipoprotein receptors (LDL-R). We used an 11-amino acid sequence from the apoB protein (ApoB11) that, when coupled with a 9-amino acid arginine linker, can transport siRNAs across the BBB to neuronal and glial cells. To examine the value of this peptide mediated oligonucleotide delivery system for PD, we delivered an siRNA targeting the α-syn (siα-syn) in a transgenic mouse model of PD. We found that ApoB11 was effective (comparable to C2-9r) at mediating the delivery of siα-syn into the CNS, co-localized to neurons and glial cells and reduced levels of α-syn protein translation and accumulation. Delivery of ApoB11/siα-syn was accompanied by protection from degeneration of selected neuronal populations in the neocortex, limbic system and striato-nigral system and reduced neuro-inflammation. Taken together, these results suggest that systemic delivery of oligonucleotides targeting α-syn using ApoB11 might be an interesting alternative strategy worth considering for the experimental treatment of synucleinopathies.
Collapse
Affiliation(s)
- Brian Spencer
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Ivy Trinh
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Jazmin Florio
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Omar M A El-Agnaf
- Neurological Disorders Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Changyoun Kim
- Laboratory of Neurogenetics National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Laboratory of Neurogenetics National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Division of Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System San Diego, CA, USA.
| |
Collapse
|
44
|
Karakatsani ME, Wang S, Samiotaki G, Kugelman T, Olumolade OO, Acosta C, Sun T, Han Y, Kamimura HAS, Jackson-Lewis V, Przedborski S, Konofagou E. Amelioration of the nigrostriatal pathway facilitated by ultrasound-mediated neurotrophic delivery in early Parkinson's disease. J Control Release 2019; 303:289-301. [PMID: 30953664 PMCID: PMC6618306 DOI: 10.1016/j.jconrel.2019.03.030] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/30/2019] [Accepted: 03/29/2019] [Indexed: 01/01/2023]
Abstract
The blood-brain barrier (BBB) prevents most drugs from gaining access to the brain parenchyma, which is a recognized impediment to the treatment of neurodegenerative disorders like Parkinson's disease (PD). Focused ultrasound (FUS), in conjunction with systemically administered microbubbles, opens the BBB locally, reversibly and non-invasively. Herein, we show that neither FUS applied over both the striatum and the ventral midbrain, without neurotrophic factors, nor intravenous administration of neurotrophic factors (either through protein or gene delivery) without FUS, ameliorates the damage to the nigrostriatal dopaminergic pathway in the sub-acute MPTP mouse model of early-stage PD. Conversely, the combination of FUS and intravenous neurotrophic (protein or gene) delivery attenuates the damage to the nigrostriatal dopaminergic pathway, by allowing the entry of these agents into the brain parenchyma. Our findings provide evidence that the application of FUS at the early stages of PD facilitates critical neurotrophic delivery that can curb the rapid progression of neurodegeneration while improving the neuronal function, seemingly opening new therapeutic avenues for the early treatment of diseases of the central nervous system.
Collapse
Affiliation(s)
| | - Shutao Wang
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Gesthimani Samiotaki
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Tara Kugelman
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Oluyemi O Olumolade
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Camilo Acosta
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Tao Sun
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Yang Han
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Hermes A S Kamimura
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Vernice Jackson-Lewis
- Departments of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA; Departments of Neurology, Columbia University, New York, NY 10032, USA; the Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; the Columbia Translational Neuroscience Initiative, Columbia University, New York, NY 10032, USA
| | - Serge Przedborski
- Departments of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA; Departments of Neurology, Columbia University, New York, NY 10032, USA; the Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; the Columbia Translational Neuroscience Initiative, Columbia University, New York, NY 10032, USA.
| | - Elisa Konofagou
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA; Departments of Radiology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
45
|
Nagree MS, Scalia S, McKillop WM, Medin JA. An update on gene therapy for lysosomal storage disorders. Expert Opin Biol Ther 2019; 19:655-670. [DOI: 10.1080/14712598.2019.1607837] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Murtaza S. Nagree
- Department of Medical Biophysics, University of Toronto, Toronto,
Ontario, Canada
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
WI, USA
| | - Simone Scalia
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
WI, USA
| | | | - Jeffrey A. Medin
- Department of Medical Biophysics, University of Toronto, Toronto,
Ontario, Canada
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee,
WI, USA
| |
Collapse
|
46
|
Bispecific Antibody Fragment Targeting APP and Inducing α-Site Cleavage Restores Neuronal Health in an Alzheimer's Mouse Model. Mol Neurobiol 2019; 56:7420-7432. [PMID: 31041656 DOI: 10.1007/s12035-019-1597-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 04/02/2019] [Indexed: 12/14/2022]
Abstract
The amyloid β (Aβ) peptide, correlated with development of Alzheimer's disease (AD), is produced by sequential proteolytic cleavage of the amyloid precursor protein (APP) by β- and γ-secretases. Alternative proteolytic cleavage of APP by α-secretase prevents formation of Aβ peptide and produces a neuroprotective protein, a soluble fragment of APPα (sAPPα). We previously generated a single-chain variable domain antibody fragment (scFv) that binds APP at the β-secretase cleavage site and blocks cleavage of APP (iBsec1), and a second scFv which has been engineered to have α-secretase-like activity that increases α-secretase cleavage of APP (Asec1a) and showed that a bispecific antibody (Diab) combining both iBsec1 and Asec1a constructs protects mammalian cells from oxidative stress. Here, we show that the diabody is an effective therapeutic agent in a mouse model of AD. An apolipoprotein B (ApoB) binding domain peptide was genetically added to the diabody to facilitate transfer across the blood-brain barrier, and a recombinant human adeno-associated virus 2/8 (rAAV2/8) was used as a vector to express the gene constructs in a APP/PS1 mouse model of AD. The diabody increased levels of sAPPα, decreased Aβ deposits and levels of oligomeric Aβ, increased neuronal health as indicated by MAP2 and synaptophysin staining, increased hippocampal neurogenesis, and most importantly dramatically increased survival rates compared with untreated mice or mice treated only with the β-secretase inhibitor. These results indicate that altering APP processing to inhibit β-site activity while simultaneously promoting α-secretase processing provides substantially increased neuronal benefits compared with inhibition of β-secretase processing alone and represents a promising new therapeutic approach for treating AD.
Collapse
|
47
|
Gleitz HF, Liao AY, Cook JR, Rowlston SF, Forte GM, D'Souza Z, O'Leary C, Holley RJ, Bigger BW. Brain-targeted stem cell gene therapy corrects mucopolysaccharidosis type II via multiple mechanisms. EMBO Mol Med 2019; 10:emmm.201708730. [PMID: 29884617 PMCID: PMC6034129 DOI: 10.15252/emmm.201708730] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The pediatric lysosomal storage disorder mucopolysaccharidosis type II is caused by mutations in IDS, resulting in accumulation of heparan and dermatan sulfate, causing severe neurodegeneration, skeletal disease, and cardiorespiratory disease. Most patients manifest with cognitive symptoms, which cannot be treated with enzyme replacement therapy, as native IDS does not cross the blood-brain barrier. We tested a brain-targeted hematopoietic stem cell gene therapy approach using lentiviral IDS fused to ApoEII (IDS.ApoEII) compared to a lentivirus expressing normal IDS or a normal bone marrow transplant. In mucopolysaccharidosis II mice, all treatments corrected peripheral disease, but only IDS.ApoEII mediated complete normalization of brain pathology and behavior, providing significantly enhanced correction compared to IDS. A normal bone marrow transplant achieved no brain correction. Whilst corrected macrophages traffic to the brain, secreting IDS/IDS.ApoEII enzyme for cross-correction, IDS.ApoEII was additionally more active in plasma and was taken up and transcytosed across brain endothelia significantly better than IDS via both heparan sulfate/ApoE-dependent receptors and mannose-6-phosphate receptors. Brain-targeted hematopoietic stem cell gene therapy provides a promising therapy for MPS II patients.
Collapse
Affiliation(s)
- Hélène Fe Gleitz
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Ai Yin Liao
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - James R Cook
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Samuel F Rowlston
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Gabriella Ma Forte
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Zelpha D'Souza
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Claire O'Leary
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Rebecca J Holley
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Brian W Bigger
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
48
|
Welch JL, Stapleton JT, Okeoma CM. Vehicles of intercellular communication: exosomes and HIV-1. J Gen Virol 2019; 100:350-366. [PMID: 30702421 PMCID: PMC7011712 DOI: 10.1099/jgv.0.001193] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/15/2018] [Indexed: 12/20/2022] Open
Abstract
The terms extracellular vesicles, microvesicles, oncosomes, or exosomes are often used interchangeably as descriptors of particles that are released from cells and comprise a lipid membrane that encapsulates nucleic acids and proteins. Although these entities are defined based on a specific size range and/or mechanism of release, the terminology is often ambiguous. Nevertheless, these vesicles are increasingly recognized as important modulators of intercellular communication. The generic characterization of extracellular vesicles could also be used as a descriptor of enveloped viruses, highlighting the fact that extracellular vesicles and enveloped viruses are similar in both composition and function. Their high degree of similarity makes differentiating between vesicles and enveloped viruses in biological specimens particularly difficult. Because viral particles and extracellular vesicles are produced simultaneously in infected cells, it is necessary to separate these populations to understand their independent functions. We summarize current understanding of the similarities and differences of extracellular vesicles, which henceforth we will refer to as exosomes, and the enveloped retrovirus, HIV-1. Here, we focus on the presence of these particles in semen, as these are of particular importance during HIV-1 sexual transmission. While there is overlap in the terminology and physical qualities between HIV-1 virions and exosomes, these two types of intercellular vehicles may differ depending on the bio-fluid source. Recent data have demonstrated that exosomes from human semen serve as regulators of HIV-1 infection that may contribute to the remarkably low risk of infection per sexual exposure.
Collapse
Affiliation(s)
- Jennifer L. Welch
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242-1109, USA
- Medical Service, Iowa City Veterans Affairs Medical Center, University of Iowa, 604 Highway 6, Iowa City, IA 52246-2208, USA
| | - Jack T. Stapleton
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242-1109, USA
- Medical Service, Iowa City Veterans Affairs Medical Center, University of Iowa, 604 Highway 6, Iowa City, IA 52246-2208, USA
| | - Chioma M. Okeoma
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
- Department of Pharmacologic Sciences, Basic Sciences Tower, Rm 8-142, Stony Brook, University School of Medicine, Stony Brook, NY 11794-8651, USA
| |
Collapse
|
49
|
Abstract
Treatment of certain central nervous system disorders, including different types of cerebral malignancies, is limited by traditional oral or systemic administrations of therapeutic drugs due to possible serious side effects and/or lack of the brain penetration and, therefore, the efficacy of the drugs is diminished. During the last decade, several new technologies were developed to overcome barrier properties of cerebral capillaries. This review gives a short overview of the structural elements and anatomical features of the blood–brain barrier. The various in vitro (static and dynamic), in vivo (microdialysis), and in situ (brain perfusion) blood–brain barrier models are also presented. The drug formulations and administration options to deliver molecules effectively to the central nervous system (CNS) are presented. Nanocarriers, nanoparticles (lipid, polymeric, magnetic, gold, and carbon based nanoparticles, dendrimers, etc.), viral and peptid vectors and shuttles, sonoporation and microbubbles are briefly shown. The modulation of receptors and efflux transporters in the cell membrane can also be an effective approach to enhance brain exposure to therapeutic compounds. Intranasal administration is a noninvasive delivery route to bypass the blood–brain barrier, while direct brain administration is an invasive mode to target the brain region with therapeutic drug concentrations locally. Nowadays, both technological and mechanistic tools are available to assist in overcoming the blood–brain barrier. With these techniques more effective and even safer drugs can be developed for the treatment of devastating brain disorders.
Collapse
|
50
|
Mullin S, Hughes D, Mehta A, Schapira AHV. Neurological effects of glucocerebrosidase gene mutations. Eur J Neurol 2018; 26:388-e29. [PMID: 30315684 PMCID: PMC6492454 DOI: 10.1111/ene.13837] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 10/09/2018] [Indexed: 01/08/2023]
Abstract
The association between Gaucher disease (GD) and Parkinson disease (PD) has been described for almost two decades. In the biallelic state (homozygous or compound heterozygous) mutations in the glucocerebrosidase gene (GBA) may cause GD, in which glucosylceramide, the sphingolipid substrate of the glucocerebrosidase enzyme (GCase), accumulates in visceral organs leading to a number of clinical phenotypes. In the biallelic or heterozygous state, GBA mutations increase the risk for PD. Mutations of the GBA allele are the most significant genetic risk factor for idiopathic PD, found in 5%–20% of idiopathic PD cases depending on ethnicity. The neurological consequences of GBA mutations are reviewed and the proposition that GBA mutations result in a disparate but connected range of clinically and pathologically related neurological features is discussed. The literature relating to the clinical, biochemical and genetic basis of GBA PD, type 1 GD and neuronopathic GD is considered highlighting commonalities and distinctions between them. The evidence for a unifying disease mechanism is considered.
Collapse
Affiliation(s)
- S Mullin
- Department of Clinical Neuroscience, UCL Institute of Neurology, London, UK.,Institute of Translational and Stratified Medicine, University of Plymouth School of Medicine, Plymouth, UK
| | - D Hughes
- LSD Unit/Department of Haematology, Institute of Immunity and Transplantation, UCL, London, UK
| | - A Mehta
- LSD Unit/Department of Haematology, Institute of Immunity and Transplantation, UCL, London, UK
| | - A H V Schapira
- Department of Clinical Neuroscience, UCL Institute of Neurology, London, UK
| |
Collapse
|