1
|
Yue X, Guo H, Wang G, Li J, Zhai Z, Wang Z, Wang W, Zhao Z, Xia X, Chen C, Cui Y, Wu C, Huang Z, Zhang X. A tailored phytosomes based nose-to-brain drug delivery strategy: Silver bullet for Alzheimer's disease. Bioact Mater 2025; 44:97-115. [DOI: 10.1016/j.bioactmat.2024.09.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
|
2
|
Kalugin PN, Soden PA, Massengill CI, Amsalem O, Porniece M, Guarino DC, Tingley D, Zhang SX, Benson JC, Hammell MF, Tong DM, Ausfahl CD, Lacey TE, Courtney Y, Hochstetler A, Lutas A, Wang H, Geng L, Li G, Li B, Li Y, Lehtinen MK, Andermann ML. Simultaneous, real-time tracking of many neuromodulatory signals with Multiplexed Optical Recording of Sensors on a micro-Endoscope. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.26.634931. [PMID: 39896634 PMCID: PMC11785251 DOI: 10.1101/2025.01.26.634931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Dozens of extracellular molecules jointly impact a given neuron, yet we lack methods to simultaneously record many such signals in real time. We developed a probe to track ten or more neuropeptides and neuromodulators using spatial multiplexing of genetically encoded fluorescent sensors. Cultured cells expressing one sensor at a time are immobilized at the front of a gradient refractive index (GRIN) lens for 3D two-photon imaging in vitro and in vivo . The sensor identity and detection sensitivity of each cell are determined via robotic dipping of the probe into wells containing various ligands and concentrations. Using this probe, we detected stimulation-evoked release of multiple neuromodulators in acute brain slices. We also tracked endogenous and drug-evoked changes in cerebrospinal fluid composition in the awake mouse lateral ventricle, which triggered downstream activation of the choroid plexus epithelium. Our approach offers a first step towards quantitative, real-time, high-dimensional tracking of brain fluid composition.
Collapse
|
3
|
Ge K, Bai Z, Wang J, Li Z, Gao F, Liu S, Zhang L, Gao F, Xie C. Engineering EVs-Mediated mRNA Delivery Regulates Microglia Function and Alleviates Depressive-Like Behaviors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2418872. [PMID: 39838773 DOI: 10.1002/adma.202418872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/13/2025] [Indexed: 01/23/2025]
Abstract
The development of new non-neurotransmitter drugs is an important supplement to the clinical treatment of major depressive disorder. The latest development of mRNA therapy provides the possibility for the treatment of some major diseases. The endoplasmic reticulum (ER) and mitochondria constitute a highly interconnected set of fundamental organelles within cells. The interconnection between them forms specific microdomains that play pivotal roles in calcium signaling, mitochondrial dynamics, inflammation, and autophagy. Perturbations in ER-mitochondrial connections may contribute to the progression of neurological disorders and other diseases. Herein, an extracellular vesicles-based delivery system, grounded in mRNA gene therapy and integrated with nanomedicine technology is devised. This system is engineered to traverse the blood-brain barrier and specifically target the central nervous system (CNS), facilitating the simultaneous delivery of mRNA drugs and metallic nanozymes into the brain. This dual-pronged approach, targeting ER and mitochondrial crosstalk, inhibits microglial overactivation, promotes M2 polarization of microglia, and suppresses the NF-κB signaling pathway. Consequently, it significantly alleviates Lipopolysaccharides-induced neuroinflammatory responses and ameliorates anxiety- and depression-like behaviors. This study demonstrates a novel antidepressant therapeutic strategy and establishes a new paradigm for mRNA gene therapy in CNS diseases.
Collapse
Affiliation(s)
- Kezhen Ge
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Jiangsu, 210009, P. R. China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Zetai Bai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Jiwei Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Jiangsu, 210009, P. R. China
| | - Zheng Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Fenfang Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Sangni Liu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Jiangsu, 210009, P. R. China
| | - Ling Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Jiangsu, 210009, P. R. China
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Chunming Xie
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Jiangsu, 210009, P. R. China
| |
Collapse
|
4
|
Hawley ZCE, Pardo ID, Cao S, Zavodszky MI, Casey F, Ferber K, Luo Y, Hana S, Chen SK, Doherty J, Costa R, Cullen P, Liu Y, Carlile TM, Chowdhury T, Doyle B, Clarner P, Mangaudis K, Guilmette E, Bourque S, Koske D, Nadella MVP, Trapa P, Hawes ML, Raitcheva D, Lo SC. Dorsal root ganglion toxicity after AAV intra-CSF delivery of a RNAi expression construct into non-human primates and mice. Mol Ther 2025; 33:215-234. [PMID: 39563026 PMCID: PMC11764093 DOI: 10.1016/j.ymthe.2024.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/24/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
Dorsal root ganglion (DRG) toxicity has been consistently reported as a potential safety concern after delivery of adeno-associated viruses (AAVs) containing gene-replacement vectors but has yet to be reported for RNAi-based vectors. Here, we report DRG toxicity after AAV intra-CSF delivery of an RNAi expression construct-artificial microRNA targeting superoxide dismutase 1 (SOD1)-in non-human primates (NHPs) and provide evidence that this can be recapitulated within mice. Histopathology evaluation showed that NHPs and mice develop DRG toxicity after AAV delivery, including DRG neuron degeneration and necrosis and nerve-fiber degeneration that were associated with increases in cerebrospinal fluid (CSF) and serum phosphorylated neurofilament heavy chain (pNF-H). RNA-sequencing analysis of DRGs showed that dysregulated pathways were preserved between NHPs and mice, including increases in innate/adaptive immune responses and decreases in mitochondrial- and neuronal-related genes, following AAV treatment. Finally, endogenous miR-21-5p was upregulated in DRGs of AAV-treated NHPs and mice. Increases in miR-21-5p were also identified within the CSF of NHPs, which significantly correlated with pNF-H, implicating miR-21-5p as a potential biomarker of DRG toxicity in conjunction with other molecular analytes. This work highlights the importance of assessing safety concerns related to DRG toxicity when developing RNAi-based AAV vectors for therapeutic purposes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yi Luo
- Biogen, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Nadim N, Khan AA, Khan S, Parveen R, Ali J. A narrative review on potential applications of spanlastics for nose-to-brain delivery of therapeutically active agents. Adv Colloid Interface Sci 2025; 335:103341. [PMID: 39566150 DOI: 10.1016/j.cis.2024.103341] [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: 04/19/2024] [Revised: 09/28/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024]
Abstract
Spanlastics, which are commonly referred to as elastic niosomes, presents a modified advancement in the area of colloidal system based drug delivery carriers. They are different from niosomes, which are non-ionic surfactant vesicles in having an edge activator. Initially, they were described as ocular drug delivery systems in 2011 by Kakkar and Kaur. Spanlastics have discovered a wide range of applications via different routes of administration. The purpose of this article is to provide information about spanlastics, a newly developed drug delivery system for the management of diseases pertaining to the Central Nervous System (CNS) via intranasal route. The article begins with the details on spanlastics and their composition, their benefits over traditional niosomes, and the mechanism underlying their enhanced absorption. Their applications through various routes of administration in a variety of diseases for a variety of drugs have been discussed. Furthermore, the article explains the nose to brain delivery channels and the advantages that this route offers over conventional delivery routes. Finally, the article discusses the studies encompassing the drug candidates that have been formulated as intranasal spanlastics for the management of different diseased conditions along with the future prospects of this emerging drug delivery system.
Collapse
Affiliation(s)
- Noorain Nadim
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Ayub Ahmad Khan
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Saba Khan
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Rabea Parveen
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India.
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
6
|
Wahengbam GS, Nirmal S, Nandwana J, Kar S, Kumari V, Mishra R, Singh A. Polymeric Nanoparticles Revolutionizing Brain Cancer Therapy: A Comprehensive Review of Strategies and Advances. Crit Rev Ther Drug Carrier Syst 2025; 42:73-106. [PMID: 39819464 DOI: 10.1615/critrevtherdrugcarriersyst.2024051822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Brain cancer continues to be one of the most formidable malignancies to manage, mainly attributable to the presence of the blood-brain barrier (BBB) limiting the permeability of drugs and the diverse characteristics of brain tumors complicating treatment. The management of brain tumors has been hampered by many different factors, including the impermeability of the BBB, which restricts the delivery of chemotherapeutic agents to the tumor site, as well as intertumoral heterogeneity and the influence of brain tumor stem cells. In addition, small molecular weight drugs cannot specifically accumulate in malignant cells and have a limited circulation half-life. Nanoparticles (NPs) can be engineered to traverse the BBB and transport therapeutic medications directly into the brain, enhancing their efficacy compared with the conventional delivery of unbound drugs. Surface modifications of NPs can boost their efficiency by increasing their selectivity towards tumor receptors. This review covers treatment methods for malignant gliomas, associated risk factors, and improvements in brain drug administration, emphasizing the future potential of polymeric NPs and their mechanism for crossing the BBB. To surmount these obstacles, the newly formulated drug-delivery approach utilizing NPs, particularly those coated with cell membranes, has demonstrated potential in treating brain cancer. These NPs provide targeted tumor specificity, biocompatibility, extended circulation, enhanced BBB penetration, and immune evasion. This review focuses on coating strategies for PLGA NPs, particularly dual-targeting methods, to enhance BBB permeability and tumor-targeted delivery of drugs in brain cancer.
Collapse
Affiliation(s)
| | - Sakshi Nirmal
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Jai Nandwana
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Swatileena Kar
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Vandana Kumari
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Rajeev Mishra
- Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, India
| | | |
Collapse
|
7
|
Eades W, Abdolmohammadpourbonab S, Dinh L, Yan B. Ionic liquids and their potential use in development and improvement of drug delivery systems: evidence of their tendency to promote drug accumulation in the brain. Pharm Dev Technol 2024; 29:1065-1074. [PMID: 39403783 DOI: 10.1080/10837450.2024.2417004] [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: 05/21/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Ionic liquids (ILs) are considered salt in liquid state, which is composed of organic cations and anions with low melting points (<100 °C). ILs have become a major scientific area with an extensive range of applications including chemistry, electrochemistry, and pharmaceutics. ILs have received great research interest in the pharmaceutical field as solvents, anti-solvents, co-solvents, and reagents in synthesis and formulation. While therapeutic ILs have been investigated for oral and trans-dermal drug delivery systems showing promising compatibility with a wide range of therapeutics, enhanced drug permeation through the skin, and cell membrane solvation to open channels to facilitate molecular passage, their potential to cross the challenging blood-brain barrier (BBB) remains an unanswered question. IL-based therapies could potentially be a game changer for improving drug delivery to cellular targets both at and across the BBB. In this review, we discuss (1) the tunable physicochemical properties of ILs; (2) the vast and various applications of ILs in the development and improvement of drug delivery systems; and (3) ILs as a potential approach for increasing drug accumulation in the brain tissue.
Collapse
Affiliation(s)
- William Eades
- Division of Pharmaceutical Sciences, University of Cincinnati, Cincinnati, OH, USA
| | | | - Linh Dinh
- Division of Pharmaceutical Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Bingfang Yan
- Division of Pharmaceutical Sciences, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
8
|
Krishnamurthy S, Oh JY, Gautham S, Li J, Shen Y. Optimizing Drug Delivery to the Brain for Breast Metastasis: A Novel Method for Tumor Targeting. Cureus 2024; 16:e73598. [PMID: 39677180 PMCID: PMC11645177 DOI: 10.7759/cureus.73598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
INTRODUCTION Brain metastases are difficult to treat due to the blood-brain barrier limiting the delivery of therapeutic agents to the brain effectively. Intraventricular drug delivery has not been well studied for intra-axial pathologies. However, our prior work demonstrated that intraventricular drug delivery in a hyperosmolar vehicle showed preferential accumulation of drug within breast cancer tissue compared to surrounding brain parenchyma. The focus of this study was to explore the molecular parameters of intraventricular drug administration that may optimize drug delivery to intra-axial brain metastases. Our hypothesis was that a low molecular weight drug with a high osmolarity solution would increase drug delivery to tumor tissue. METHODS We used an intracerebral breast cancer tumor model in adult female nude rats divided into six experimental groups. We examined three iron-labeled dextran molecules (3 kD, 5 kD, and 10 kD) in 337 mOsm/L solution and three different osmolarities of delivery solution (307, 353, and 368 mOsm/L) with 10 kD dextran. 7T magnetic resonance imaging (MRI) was used to analyze dextran distribution at different time points. All animals were sacrificed after two hours, and the quantity of dextran particles was determined by histopathology. RESULTS Breast cancer tumor cells were successfully implanted in all rats. The MRI quantification of dextran concentration was well corroborated by histopathology. Varying the molecular size of dextran resulted in the smallest molecule reaching peak levels in tumor tissue earlier than the larger molecules, but the larger molecules remained concentrated in tumor tissue for a longer time. Varying the osmolarity of the delivery solution resulted in the preferential accumulation of 10 kD dextran in tumor tissue except for when dextran was delivered in 368 mOsm/L solution where no preferential distribution was seen. CONCLUSION Hyperosmolar intraventricular delivery of chemotherapeutic drugs could be effective in preferentially delivering drugs to abnormal tumor tissues.
Collapse
Affiliation(s)
- Satish Krishnamurthy
- Neurological Surgery, State University of New York Upstate Medical University, Syracuse, USA
| | - Justin Y Oh
- Neurological Surgery, State University of New York Upstate Medical University, Syracuse, USA
| | - Shruti Gautham
- Neurological Surgery, State University of New York Upstate Medical University, Syracuse, USA
| | - Jie Li
- Neurological Surgery, State University of New York Upstate Medical University, Syracuse, USA
| | - Yimin Shen
- Radiology, Wayne State University, Detroit, USA
| |
Collapse
|
9
|
Butola M, Nainwal N. Non-Invasive Techniques of Nose to Brain Delivery Using Nanoparticulate Carriers: Hopes and Hurdles. AAPS PharmSciTech 2024; 25:256. [PMID: 39477829 DOI: 10.1208/s12249-024-02946-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/15/2024] [Indexed: 12/12/2024] Open
Abstract
Intranasal drug delivery route has emerged as a promising non-invasive method of administering drugs directly to the brain, bypassing the blood-brain barrier (BBB) and blood-cerebrospinal fluid barriers (BCSF). BBB and BCSF prevent many therapeutic molecules from entering the brain. Intranasal drug delivery can transport drugs from the nasal mucosa to the brain, to treat a variety of Central nervous system (CNS) diseases. Intranasal drug delivery provides advantages over invasive drug delivery techniques such as intrathecal or intraparenchymal which can cause infection. Many strategies, including nanocarriers liposomes, solid-lipid NPs, nano-emulsion, nanostructured lipid carriers, dendrimers, exosomes, metal NPs, nano micelles, and quantum dots, are effective in nose-to-brain drug transport. However, the biggest obstacles to the nose-to-brain delivery of drugs include mucociliary clearance, poor drug retention, enzymatic degradation, poor permeability, bioavailability, and naso-mucosal toxicity. The current review aims to compile current approaches for drug delivery to the CNS via the nose, focusing on nanotherapeutics and nasal devices. Along with a brief overview of the related pathways or mechanisms, it also covers the advantages of nasal drug delivery as a potential method of drug administration. It also offers several possibilities to improve drug penetration across the nasal barrier. This article overviews various in-vitro, ex-vivo, and in-vivo techniques to assess drug transport from the nasal epithelium into the brain.
Collapse
Affiliation(s)
- Mansi Butola
- Department of Pharmaceutics, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248001, India
| | - Nidhi Nainwal
- Department of Pharmaceutics, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248001, India.
| |
Collapse
|
10
|
Ma J, Huang L, Han C. Expert consensus on the use of third-generation EGFR-TKIs in EGFR-mutated advanced non-small cell lung cancer with various T790M mutations post-resistance to first-/second-generation EGFR-TKIs. Ther Adv Med Oncol 2024; 16:17588359241289648. [PMID: 39434954 PMCID: PMC11492187 DOI: 10.1177/17588359241289648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
Third-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have emerged as the mainstay of treatment for advanced EGFR-mutant advanced non-small cell lung cancer (NSCLC), effectively overcoming the problems of acquired threonine-to-methionine (T790M) mutations associated with the first- or second-generation TKIs. Evidence from several studies suggests that these agents, including osimertinib and aumolertinib, also show potential benefits in T790M-negative or unknown populations, particularly those with brain metastases, where the high permeability of the blood-brain barrier allows effective control of intracranial lesions. Despite the encouraging results, further high-quality research, including prospective trials, is warranted to fully elucidate the efficacy profiles of these third-generation TKIs in T790M-negative or unknown NSCLC patients after first- or second-line TKI failure. The present expert consensus highlights the evolving role of third-generation EGFR-TKIs in overcoming therapeutic resistance and optimizing patient outcomes.
Collapse
Affiliation(s)
- Jietao Ma
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Letian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chengbo Han
- Department of Oncology, Shengjing Hospital of China Medical University, No. 39, Huaxiang Road, Tiexi District, Shenyang 110022 China
| |
Collapse
|
11
|
Tong X, Remsik J, Brook J, Petrova B, Xu L, Li MJ, Snyder J, Chabot K, Estrera R, Osei-Gyening I, Nobre AR, Wang H, Osman AM, Wong AYL, Sidharta M, Piedrafita-Ortiz S, Manoranjan B, Zhou T, Murali R, Hamard PJ, Koche R, He Y, Kanarek N, Boire A. Retinoid X Receptor Signaling Mediates Cancer Cell Lipid Metabolism in the Leptomeninges. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618083. [PMID: 39464048 PMCID: PMC11507812 DOI: 10.1101/2024.10.13.618083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Cancer cells metastatic to the leptomeninges encounter a metabolically-challenging extreme microenvironment. To understand adaptations to this space, we subjected leptomeningeal-metastatic (LeptoM) mouse breast and lung cancers isolated from either the leptomeninges or orthotopic primary sites to ATAC-and RNA-sequencing. When inhabiting the leptomeninges, the LeptoM cells demonstrated transcription downstream of retinoid-X-receptors (RXRs). We found evidence of local retinoic acid (RA) generation in both human leptomeningeal metastasis and mouse models in the form of elevated spinal fluid retinol and expression of RA-generating dehydrogenases within the leptomeningeal microenvironment. Stimulating LeptoM cells with RA induced expression of transcripts encoding de novo fatty acid synthesis pathway enzymes in vitro . In vivo , while deletion of Stra6 did not alter cancer cell leptomeningeal growth, knockout of Rxra/b/g interrupted cancer cell lipid biosynthesis and arrested cancer growth. These observations illustrate a mechanism whereby metastatic cancer cells awake locally-generated developmental cues for metabolically reprograming, suggesting novel therapeutic approaches.
Collapse
|
12
|
Yun C, Woo HC, Lovatt D, Parish CA, Spellman DS, Shen H. Development of a Versatile High-through-put Oligonucleotide LC-MS Method to Accelerate Drug Discovery. AAPS PharmSciTech 2024; 25:239. [PMID: 39390148 DOI: 10.1208/s12249-024-02934-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Liquid chromatography-mass spectrometry (LC-MS) is an effective tool for high-throughput quantification of oligonucleotides that is crucial for understanding their biological roles and developing diagnostic tests. This paper presents a high-throughput LC-MS/MS method that may be versatilely applied for a wide range of oligonucleotides, making it a valuable tool for rapid screening and discovery. The method is demonstrated using an in-house synthesized MALAT-1 Antisense oligonucleotide (ASO) as a test case. Biological samples were purified using a reversed liquid-liquid extraction process automated by a liquid handling workstation and analyzed with ion-pairing LC-MS/MS. The assay was evaluated for sensitivity (LLOQ = 2 nM), specificity, precision, accuracy, recovery, matrix effect, and stability in rat cerebrospinal fluid (CSF) and plasma. Besides some existing considerations such as column selection, ion-pairing reagent, and sample purification, our work focused on the following four subtopics: 1) selecting the appropriate Multiple Reaction Monitoring (MRM) transition to maximize sensitivity for trace-level ASO in biological samples; 2) utilizing a generic risk-free internal standard (tenofovir) to avoid crosstalk interference from the oligo internal standard commonly utilized in the LC-MS assay; 3) automating the sample preparation process to increase precision and throughput; and 4) comparing liquid-liquid extraction (LLE) and solid-phase extraction (SPE) as sample purification methods in oligo method development. The study quantified the concentration of MALAT-1 ASO in rat CSF and plasma after intrathecal injection and used the difference between the two matrices to evaluate the injection technique. The results provide a solid foundation for further internal oligonucleotide discovery and development.
Collapse
Affiliation(s)
- Changhong Yun
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc., Boston, Massachusetts, 02115, USA
| | - Hyun Chong Woo
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc., Boston, Massachusetts, 02115, USA
| | - Ditte Lovatt
- Biology-Discovery, Merck & Co., Inc., West Point, Pennsylvania, 19446, USA
| | - Craig A Parish
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey, 07065, USA
| | - Daniel S Spellman
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc., West Point, Pennsylvania, 19446, USA
| | - Honglue Shen
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc., Boston, Massachusetts, 02115, USA.
| |
Collapse
|
13
|
Ferreira MDA, Lückemeyer DD, Martins F, Schran RG, da Silva AM, Gambeta E, Zamponi GW, Ferreira J. Pronociceptive role of spinal Ca v2.3 (R-type) calcium channels in a mouse model of postoperative pain. Br J Pharmacol 2024; 181:3594-3609. [PMID: 38812100 DOI: 10.1111/bph.16407] [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: 08/07/2023] [Revised: 03/05/2024] [Accepted: 03/28/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND More than 80% of patients may experience acute pain after a surgical procedure, and this is often refractory to pharmacological intervention. The identification of new targets to treat postoperative pain is necessary. There is an association of polymorphisms in the Cav2.3 gene with postoperative pain and opioid consumption. Our study aimed to identify Cav2.3 as a potential target to treat postoperative pain and to reduce opioid-related side effects. EXPERIMENTAL APPROACH A plantar incision model was established in adult male and female C57BL/6 mice. Cav2.3 expression was detected by qPCR and suppressed by siRNA treatment. The antinociceptive efficacy and safety of a Cav2.3 blocker-alone or together with morphine-was also assessed after surgery. KEY RESULTS Paw incision in female and male mice caused acute nociception and increased Cav2.3 mRNA expression in the spinal cord but not in the incised tissue. Intrathecal treatment with siRNA against Cav2.3, but not with a scrambled siRNA, prevented the development of surgery-induced nociception in both male and female mice, with female mice experiencing long-lasting effects. High doses of i.t. SNX-482, a Cav2.3 channel blocker, or morphine injected alone, reversed postoperative nociception but also induced side effects. A combination of lower doses of morphine and SNX-482 mediated a long-lasting reversal of postsurgical pain in female and male mice. CONCLUSION Our results demonstrate that Cav2.3 has a pronociceptive role in the induction of postoperative pain, indicating that it is a potential target for the development of therapeutic approaches for the treatment of postoperative pain.
Collapse
Affiliation(s)
- Marcella de Amorim Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Debora Denardin Lückemeyer
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
- Department of Anesthesiology, Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fernanda Martins
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Roberta Giusti Schran
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Ana Merian da Silva
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Eder Gambeta
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Juliano Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
14
|
Lu Z, Ngan MP, Liu JYH, Yang L, Tu L, Chan SW, Giuliano C, Lovati E, Pietra C, Rudd JA. The growth hormone secretagogue receptor 1a agonists, anamorelin and ipamorelin, inhibit cisplatin-induced weight loss in ferrets: Anamorelin also exhibits anti-emetic effects via a central mechanism. Physiol Behav 2024; 284:114644. [PMID: 39043357 DOI: 10.1016/j.physbeh.2024.114644] [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: 05/31/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024]
Abstract
This study investigated whether ghrelin mimetics, namely anamorelin and ipamorelin, can alleviate weight loss and inhibition of feeding observed during acute and delayed phases of cisplatin-induced emesis in ferrets. The potential of anamorelin to inhibit electrical field stimulation (EFS)-induced contractions of isolated ferret ileum was compared with ipamorelin. In other experiments, ferrets were administered anamorelin (1-3 mg/kg), ipamorelin (1-3 mg/kg), or vehicle intraperitoneally (i.p.) 30 s before cisplatin (5 mg/kg, i.p.) and then every 24 h, and their behaviour was recorded for up to 72 h. Food and water consumption was measured every 24 h. The effect of anamorelin (10 µg) was also assessed following intracerebroventricular administration. Anamorelin and ipamorelin inhibited EFS-induced contractions of isolated ileum by 94.4 % (half-maximal inhibitory concentration [IC50]=14.0 µM) and 54.4 % (IC50=11.7 µM), respectively. Neither of compounds administered i.p. had any effect on cisplatin-induced acute or delayed emesis, but both inhibited associated cisplatin-induced weight loss on the last day of delayed phase (48-72 h) by approximately 24 %. Anamorelin (10 µg) administered intracerebroventricularly reduced cisplatin-induced acute emesis by 60 % but did not affect delayed emesis. It also improved food and water consumption by approximately 20 %-40 % during acute phase, but not delayed phase, and reduced associated cisplatin-induced weight loss during delayed phase by ∼23 %. In conclusion, anamorelin and ipamorelin administered i.p. had beneficial effects in alleviating cisplatin-induced weight loss during delayed phase, and these effects were seen when centrally administered anamorelin. Anamorelin inhibited cisplatin-induced acute emesis following intracerebroventricular but not intraperitoneal administration, suggesting that brain penetration is important for its anti-emetic mechanism of action.
Collapse
Affiliation(s)
- Zengbing Lu
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Man P Ngan
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Julia Y H Liu
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Lingqing Yang
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Longlong Tu
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, USA
| | - Sze Wa Chan
- School of Health Sciences, Saint Francis University, Tseung Kwan O, New Territories, Hong Kong
| | | | | | | | - John A Rudd
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong.
| |
Collapse
|
15
|
Yuan M, Zong M, Ren C, Zong W, Li Z. Ceftazidime/avibactam combined with colistimethate sodium successfully cures carbapenem-resistant Pseudomonas aeruginosa-induced brain abscess in a child post-craniotomy: a case report. Front Oncol 2024; 14:1444172. [PMID: 39364315 PMCID: PMC11446902 DOI: 10.3389/fonc.2024.1444172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024] Open
Abstract
The treatment of brain abscess induced by carbapenem-resistant Pseudomonas aeruginosa (CRPA) is a clinical challenge around the world. Apart from novel β-lactam/β-lactamase inhibitors and polymyxins, there are few sufficiently powerful antibiotics that are effective against CRPA-induced infections. Considering the blood-brain barrier factor, there are even fewer drugs that can be used to treat intracranial CRPA-induced infections. In this article, we reported a case of CRPA-induced brain abscess that was successfully treated with intravenous ceftazidime/avibactam and intrathecal colistimethate sodium in a child after intracranial tumor resection.
Collapse
Affiliation(s)
- Minglu Yuan
- Department of Pharmacy, Beijing Electric Power Hospital of State Grid Co. of China, Capital Medical University Electric Teaching Hospital, Beijing, China
| | - Miao Zong
- Department of Neurosurgery, Beijing Electric Power Hospital of State Grid Co. of China, Capital Medical University Electric Teaching Hospital, Beijing, China
| | - Cong Ren
- Department of Neurosurgery, Beijing Electric Power Hospital of State Grid Co. of China, Capital Medical University Electric Teaching Hospital, Beijing, China
| | - Wenjing Zong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhongdong Li
- Department of Pharmacy, Beijing Electric Power Hospital of State Grid Co. of China, Capital Medical University Electric Teaching Hospital, Beijing, China
| |
Collapse
|
16
|
Schwinghamer K, Kopec BM, Ayewoh E, Tao X, Sadekar S, Sreedhara A, Kelley RF, Tesar DB, Siahaan TJ. Exploring How Antibody Format Drives Clearance from the Brain. Mol Pharm 2024; 21:4416-4429. [PMID: 39058284 PMCID: PMC11368618 DOI: 10.1021/acs.molpharmaceut.4c00354] [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: 07/28/2024]
Abstract
Monoclonal antibodies (mAbs) have high binding specificity and affinity, making them attractive for treating brain diseases. However, their effectiveness is limited by poor blood-brain barrier (BBB) penetration and rapid central nervous system (CNS) clearance. Our group identified blood-brain barrier modulator (BBBM) peptides that improved mAb penetration across the BBB into the brain. In this study, we investigated the pharmacokinetics of a mAb delivered to the brain using BBBMs after intravenous (IV) administration and explored the impact of antibody format (size, neonatal Fc receptor (FcRn) binding, hyaluronic acid binding) on brain clearance following direct injection into the central nervous system (CNS) via intracerebroventricular (ICV) injection. IRDye800CW-labeled antibodies were administered into C57BL/6 mice via ICV or IV injection, and organ concentrations were measured after various time points. When a mAb was coadministered with a BBBM peptide, the permeation of mAb across the BBB was increased compared to mAb alone at early time points; however, the mAb was cleared within 2 h from the brain. ICV experiments revealed that an antibody Fab fragment had a higher brain exposure than a mAb, and that a Fab fused to a hyaluronic acid binding domain (Fab-VG1) showed remarkable improvement in brain exposure. These findings suggest that BBBMs and antibody format optimization may be promising strategies for enhancing brain retention of therapeutic antibodies.
Collapse
Affiliation(s)
- Kelly Schwinghamer
- Department of Pharmaceutical Chemistry, The University of Kansas, 2093 Constant Ave., Lawrence, Kansas 66046, United States
| | - Brian M Kopec
- Department of Pharmaceutical Chemistry, The University of Kansas, 2093 Constant Ave., Lawrence, Kansas 66046, United States
| | - Ebehiremen Ayewoh
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Xun Tao
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shraddha Sadekar
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Alavattam Sreedhara
- Department of Pharmaceutical Development, 1 DNA Way, South San Francisco, California 94080, United States
| | - Robert F Kelley
- Department of Pharmaceutical Development, 1 DNA Way, South San Francisco, California 94080, United States
| | - Devin B Tesar
- Department of Pharmaceutical Development, 1 DNA Way, South San Francisco, California 94080, United States
| | - Teruna J Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, 2093 Constant Ave., Lawrence, Kansas 66046, United States
| |
Collapse
|
17
|
van Osch MJP, Wåhlin A, Scheyhing P, Mossige I, Hirschler L, Eklund A, Mogensen K, Gomolka R, Radbruch A, Qvarlander S, Decker A, Nedergaard M, Mori Y, Eide PK, Deike K, Ringstad G. Human brain clearance imaging: Pathways taken by magnetic resonance imaging contrast agents after administration in cerebrospinal fluid and blood. NMR IN BIOMEDICINE 2024; 37:e5159. [PMID: 38634301 DOI: 10.1002/nbm.5159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 04/19/2024]
Abstract
Over the last decade, it has become evident that cerebrospinal fluid (CSF) plays a pivotal role in brain solute clearance through perivascular pathways and interactions between the brain and meningeal lymphatic vessels. Whereas most of this fundamental knowledge was gained from rodent models, human brain clearance imaging has provided important insights into the human system and highlighted the existence of important interspecies differences. Current gold standard techniques for human brain clearance imaging involve the injection of gadolinium-based contrast agents and monitoring their distribution and clearance over a period from a few hours up to 2 days. With both intrathecal and intravenous injections being used, which each have their own specific routes of distribution and thus clearance of contrast agent, a clear understanding of the kinetics associated with both approaches, and especially the differences between them, is needed to properly interpret the results. Because it is known that intrathecally injected contrast agent reaches the blood, albeit in small concentrations, and that similarly some of the intravenously injected agent can be detected in CSF, both pathways are connected and will, in theory, reach the same compartments. However, because of clear differences in relative enhancement patterns, both injection approaches will result in varying sensitivities for assessment of different subparts of the brain clearance system. In this opinion review article, the "EU Joint Programme - Neurodegenerative Disease Research (JPND)" consortium on human brain clearance imaging provides an overview of contrast agent pharmacokinetics in vivo following intrathecal and intravenous injections and what typical concentrations and concentration-time curves should be expected. This can be the basis for optimizing and interpreting contrast-enhanced MRI for brain clearance imaging. Furthermore, this can shed light on how molecules may exchange between blood, brain, and CSF.
Collapse
Affiliation(s)
- Matthias J P van Osch
- C. J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Anders Wåhlin
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
- Department of Applied Physics and Electronics, Umeå University, Umeå, Sweden
- Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Paul Scheyhing
- Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ingrid Mossige
- Division of Radiology and Nuclear Medicine, Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, The Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lydiane Hirschler
- C. J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Anders Eklund
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
- Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Klara Mogensen
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Ryszard Gomolka
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Radbruch
- Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Sara Qvarlander
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Andreas Decker
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Yuki Mori
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Per Kristian Eide
- Department of Neurosurgery, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- KG Jebsen Centre for Brain Fluid Research, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Katerina Deike
- Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Geir Ringstad
- Department of Radiology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Department of Geriatrics and Internal Medicine, Sorlandet Hospital, Arendal, Norway
| |
Collapse
|
18
|
Mehta RI, Mehta RI. Understanding central nervous system fluid networks: Historical perspectives and a revised model for clinical neurofluid imaging. NMR IN BIOMEDICINE 2024; 37:e5149. [PMID: 38584002 PMCID: PMC11531858 DOI: 10.1002/nbm.5149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/09/2024]
Abstract
The central nervous system (CNS) lacks traditionally defined lymphatic vasculature. However, CNS tissues and barriers compartmentalize the brain, spinal cord, and adjacent spaces, facilitating the transmittal of fluids, metabolic wastes, immune cells, and vital signals, while more conventional lymphatic pathways in the meninges, cervicofacial and paraspinal regions transmit efflux fluid and molecules to peripheral lymph and lymph nodes. Thus, a unique and highly organized fluid circulation network encompassing intraparenchymal, subarachnoid, dural, and extradural segments functions in unison to maintain CNS homeostasis. Pathways involved in this system have been under investigation for centuries and continue to be the source of considerable interest and debate. Modern imaging and microscopy technologies have led to important breakthroughs pertaining to various elements of CNS fluid circuitry and exchange over the past decade, thus enhancing knowledge on mechanisms of mammalian CNS maintenance and disease. Yet, to better understand precise anatomical routes, the physiology and clinical significance of these CNS pathways, and potential therapeutic targets in humans, fluid conduits, flow-regulating factors, and tissue effects must be analyzed systematically and in a global manner in persons across age, demographical factors, and disease states. Here, we illustrate the system-wide nature of intermixing CNS fluid networks, summarize historical and clinical studies, and discuss anatomical and physiological similarities and differences that are relevant for translation of evidence from mice to humans. We also review Cushing's classical model of cerebrospinal fluid flow and present a new framework of this "third circulation" that emphasizes previously unexplained complexities of CNS fluid circulation in humans. Finally, we review future directions in the field, including emerging theranostic techniques and MRI studies required in humans.
Collapse
Affiliation(s)
- Rupal I. Mehta
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Rashi I. Mehta
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
19
|
Qu Z, Luo J, Li Z, Yang R, Zhao J, Chen X, Yu S, Shu H. Advancements in strategies for overcoming the blood-brain barrier to deliver brain-targeted drugs. Front Aging Neurosci 2024; 16:1353003. [PMID: 39253614 PMCID: PMC11381257 DOI: 10.3389/fnagi.2024.1353003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
The blood-brain barrier is known to consist of a variety of cells and complex inter-cellular junctions that protect the vulnerable brain from neurotoxic compounds; however, it also complicates the pharmacological treatment of central nervous system disorders as most drugs are unable to penetrate the blood-brain barrier on the basis of their own structural properties. This dramatically diminished the therapeutic effect of the drug and compromised its biosafety. In response, a number of drugs are often delivered to brain lesions in invasive ways that bypass the obstruction of the blood-brain barrier, such as subdural administration, intrathecal administration, and convection-enhanced delivery. Nevertheless, these intrusive strategies introduce the risk of brain injury, limiting their clinical application. In recent years, the intensive development of nanomaterials science and the interdisciplinary convergence of medical engineering have brought light to the penetration of the blood-brain barrier for brain-targeted drugs. In this paper, we extensively discuss the limitations of the blood-brain barrier on drug delivery and non-invasive brain-targeted strategies such as nanomedicine and blood-brain barrier disruption. In the meantime, we analyze their strengths and limitations and provide outlooks on the further development of brain-targeted drug delivery systems.
Collapse
Affiliation(s)
- Zhichuang Qu
- Department of Neurosurgery, Meishan City People's Hospital, Meishan, China
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Juan Luo
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zheng Li
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rong Yang
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiaxi Zhao
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xin Chen
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Sixun Yu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| | - Haifeng Shu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
20
|
Kumar J, Karim A, Sweety UH, Sarma H, Nurunnabi M, Narayan M. Bioinspired Approaches for Central Nervous System Targeted Gene Delivery. ACS APPLIED BIO MATERIALS 2024; 7:4975-4997. [PMID: 38100377 DOI: 10.1021/acsabm.3c00842] [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] [Indexed: 12/17/2023]
Abstract
Disorders of the central nervous system (CNS) which include a wide range of neurodegenerative and neurological conditions have become a serious global issue. The presence of CNS barriers poses a significant challenge to the progress of designing effective therapeutic delivery systems, limiting the effectiveness of drugs, genes, and other therapeutic agents. Natural nanocarriers present in biological systems have inspired researchers to design unique delivery systems through biomimicry. As natural resource derived delivery systems are more biocompatible, current research has been focused on the development of delivery systems inspired by bacteria, viruses, fungi, and mammalian cells. Despite their structural potential and extensive physiological function, making them an excellent choice for biomaterial engineering, the delivery of nucleic acids remains challenging due to their instability in biological systems. Similarly, the efficient delivery of genetic material within the tissues of interest remains a hurdle due to a lack of selectivity and targeting ability. Considering that gene therapies are the holy grail for intervention in diseases, including neurodegenerative disorders such as Alzheimer's disease, Parkinson's Disease, and Huntington's disease, this review centers around recent advances in bioinspired approaches to gene delivery for the prevention of CNS disorders.
Collapse
Affiliation(s)
- Jyotish Kumar
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Afroz Karim
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Ummy Habiba Sweety
- Environmental Science and Engineering, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Hemen Sarma
- Bioremediation Technology Research Group, Department of Botany, Bodoland University, Rangalikhata, Deborgaon, 783370, Kokrajhar (BTR), Assam, India
| | - Md Nurunnabi
- The Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| |
Collapse
|
21
|
Goodlet KJ, Nailor MD. Central Nervous System Infections and Antibiotic Selection: All Infection Sites Are Not Created Equal. Clin Infect Dis 2024; 79:573-574. [PMID: 38113485 DOI: 10.1093/cid/ciad772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Affiliation(s)
- Kellie J Goodlet
- Department of Pharmacy Practice, Midwestern University College of Pharmacy, Glendale, Arizona, USA
| | - Michael D Nailor
- Department of Pharmacy Services, St.Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| |
Collapse
|
22
|
Barker SJ, Thayer MB, Kim C, Tatarakis D, Simon MJ, Dial R, Nilewski L, Wells RC, Zhou Y, Afetian M, Akkapeddi P, Chappell A, Chew KS, Chow J, Clemens A, Discenza CB, Dugas JC, Dwyer C, Earr T, Ha C, Ho YS, Huynh D, Lozano EI, Jayaraman S, Kwan W, Mahon C, Pizzo M, Robles-Colmenares Y, Roche E, Sanders L, Stergioulis A, Tong R, Tran H, Zuchero Y, Estrada AA, Gadkar K, Koth CMM, Sanchez PE, Thorne RG, Watts RJ, Sandmann T, Kane LA, Rigo F, Dennis MS, Lewcock JW, DeVos SL. Targeting the transferrin receptor to transport antisense oligonucleotides across the mammalian blood-brain barrier. Sci Transl Med 2024; 16:eadi2245. [PMID: 39141703 DOI: 10.1126/scitranslmed.adi2245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/24/2024] [Indexed: 08/16/2024]
Abstract
Antisense oligonucleotides (ASOs) are promising therapeutics for treating various neurological disorders. However, ASOs are unable to readily cross the mammalian blood-brain barrier (BBB) and therefore need to be delivered intrathecally to the central nervous system (CNS). Here, we engineered a human transferrin receptor 1 (TfR1) binding molecule, the oligonucleotide transport vehicle (OTV), to transport a tool ASO across the BBB in human TfR knockin (TfRmu/hu KI) mice and nonhuman primates. Intravenous injection and systemic delivery of OTV to TfRmu/hu KI mice resulted in sustained knockdown of the ASO target RNA, Malat1, across multiple mouse CNS regions and cell types, including endothelial cells, neurons, astrocytes, microglia, and oligodendrocytes. In addition, systemic delivery of OTV enabled Malat1 RNA knockdown in mouse quadriceps and cardiac muscles, which are difficult to target with oligonucleotides alone. Systemically delivered OTV enabled a more uniform ASO biodistribution profile in the CNS of TfRmu/hu KI mice and greater knockdown of Malat1 RNA compared with a bivalent, high-affinity TfR antibody. In cynomolgus macaques, an OTV directed against MALAT1 displayed robust ASO delivery to the primate CNS and enabled more uniform biodistribution and RNA target knockdown compared with intrathecal dosing of the same unconjugated ASO. Our data support systemically delivered OTV as a potential platform for delivering therapeutic ASOs across the BBB.
Collapse
Affiliation(s)
| | - Mai B Thayer
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Chaeyoung Kim
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | - Rebekah Dial
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | - Yinhan Zhou
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | - Kylie S Chew
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Johann Chow
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | - Jason C Dugas
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Timothy Earr
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Connie Ha
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Yvonne S Ho
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - David Huynh
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | - Wanda Kwan
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Cathal Mahon
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | - Elysia Roche
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Laura Sanders
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Raymond Tong
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Hai Tran
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Y Zuchero
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Kapil Gadkar
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | - Ryan J Watts
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Lesley A Kane
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | - Mark S Dennis
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Sarah L DeVos
- Denali Therapeutics Inc., South San Francisco, CA, USA
| |
Collapse
|
23
|
Ramos Ferrer P, Sakiyama-Elbert S. Affinity-based drug delivery systems for the central nervous system: exploiting molecular interactions for local, precise targeting. J Neural Eng 2024; 21:041004. [PMID: 39059438 DOI: 10.1088/1741-2552/ad680a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 07/26/2024] [Indexed: 07/28/2024]
Abstract
Objective: The effective treatment of central nervous system (CNS) disorders remains a significant challenge, primarily due to its molecular and structural complexity. Clinical translation of promising therapeutic agents is limited by the lack of optimal drug delivery systems capable of targeted, localized release of drugs to the brain and spinal cord.Approach: This review provides an overview of the potential of affinity-based drug delivery systems, which leverage molecular interactions to enhance the delivery and efficacy of therapeutic agents within the CNS.Main results: Various approaches, including hydrogels, micro- and nanoparticles, and functionalized biomaterials, are examined for their ability to provide local, sustained release of proteins, growth factors and other drugs.Significance: Furthermore, we present a detailed analysis of design considerations for developing effective affinity-based systems, incorporating insights from both existing literature and our group's research. These considerations include the biochemical modification of delivery vehicles and the optimization of physical and chemical properties to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Pablo Ramos Ferrer
- Department of Chemical Engineering, University of Washington, Seattle, WA, United States of America
| | - Shelly Sakiyama-Elbert
- Department of Chemical Engineering, University of Washington, Seattle, WA, United States of America
- Department of Bioengineering, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
24
|
Ma J, Wang B, Wei X, Tian M, Bao X, Zhang Y, Qi H, Zhang Y, Hu M. Accumulation of extracellular elastin-derived peptides disturbed neuronal morphology and neuron-microglia crosstalk in aged brain. J Neurochem 2024; 168:1460-1474. [PMID: 38168728 DOI: 10.1111/jnc.16039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024]
Abstract
Extracellular elastin-derived peptides (EDPs) accumulate in the aging brain and have been associated with vascular dementia and Alzheimer's disease (AD). The activation of inflammatory processes in glial cells with EDP treatment has received attention, but not in neurons. To properly understand EDPs' pathogenic significance, the impact on neuronal function and neuron-microglia crosstalk was explored further. Among the EDP molecules, Val-Gly-Val-Ala-Pro-Gly (VGVAPG) is a typical repeating hexapeptide. Here, we observed that EDPs-VGVAPG influenced neuronal survival and morphology in a dose-dependent manner. High concentrations of VGVAPG induced synapse loss and microglia hyperactivation in vivo and in vitro. Following EDP incubation, galectin 3 (Gal-3) released by neurons served as a chemokine, attracting microglial engulfment. Blocking Gal-3 and EDP binding remedied synapse loss in neurons and phagocytosis in microglia. In response to the accumulation of EDPs, proteomics in matrix remodeling and cytoskeleton dynamics, such as a disintegrin and metalloproteinase (ADAM) family, were engaged. These findings in extracellular EDPs provided more evidence for the relationship between aging and neuron dysfunction, increasing the insight of neuroinflammatory responses and the development of new specialized extracellular matrix remolding-targeted therapy options for dementia or other neurodegenerative disease.
Collapse
Affiliation(s)
- Jun Ma
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Bingqian Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiaoxi Wei
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Meng Tian
- Key Laboratory of Molecular Epigenetics, Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Xingfu Bao
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yifan Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Huichuan Qi
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yi Zhang
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
25
|
Rhea EM, Leclerc M, Yassine HN, Capuano AW, Tong H, Petyuk VA, Macauley SL, Fioramonti X, Carmichael O, Calon F, Arvanitakis Z. State of the Science on Brain Insulin Resistance and Cognitive Decline Due to Alzheimer's Disease. Aging Dis 2024; 15:1688-1725. [PMID: 37611907 PMCID: PMC11272209 DOI: 10.14336/ad.2023.0814] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is common and increasing in prevalence worldwide, with devastating public health consequences. While peripheral insulin resistance is a key feature of most forms of T2DM and has been investigated for over a century, research on brain insulin resistance (BIR) has more recently been developed, including in the context of T2DM and non-diabetes states. Recent data support the presence of BIR in the aging brain, even in non-diabetes states, and found that BIR may be a feature in Alzheimer's disease (AD) and contributes to cognitive impairment. Further, therapies used to treat T2DM are now being investigated in the context of AD treatment and prevention, including insulin. In this review, we offer a definition of BIR, and present evidence for BIR in AD; we discuss the expression, function, and activation of the insulin receptor (INSR) in the brain; how BIR could develop; tools to study BIR; how BIR correlates with current AD hallmarks; and regional/cellular involvement of BIR. We close with a discussion on resilience to both BIR and AD, how current tools can be improved to better understand BIR, and future avenues for research. Overall, this review and position paper highlights BIR as a plausible therapeutic target for the prevention of cognitive decline and dementia due to AD.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Manon Leclerc
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
| | - Hussein N Yassine
- Departments of Neurology and Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Ana W Capuano
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Han Tong
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA.
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA.
| | - Xavier Fioramonti
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France.
| | - Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Frederic Calon
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
26
|
Moyer H, Mellett R, Vigneault K, McKeown M, Karlawish J, Augustine E, Schneider L, Kimmelman J. Prevalence and Impact of Bypassing or Overriding Phase 2 Trials in Neurologic Drug Development. Neurology 2024; 103:e209533. [PMID: 38833654 PMCID: PMC11226307 DOI: 10.1212/wnl.0000000000209533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/09/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Pivotal trials for neurologic drugs in clinical development are often initiated without a phase 2 trial ("bypass") or despite a negative phase 2 efficacy result ("override"). Such practices may degrade the risk/benefit ratio of phase 3 trials. The aim of this study is to estimate the proportion of phase 3 trials for 10 neurologic diseases started without a positive phase 2 trial, to identify factors associated with this practice, and to investigate any association with unfavorable phase 3 trial outcomes. METHODS We searched ClinicalTrials.gov for phase 3 trials completed during 2011-2021, with at least 1 research site in the United States, Canada, the European Union, the United Kingdom, or Australia, and investigating drugs or biologics for treatment of 10 neurologic conditions. Our primary objective was to assess the prevalence of phase 2 bypass/override by searching for preceding phase 2 trials. We used Fisher exact tests to determine whether phase 3 trial characteristics and trial results were associated with phase 2 bypass/override. RESULTS Of the 1,188 phase 3 trials captured in our search, 113 met eligibility for inclusion. Of these, 46% were not preceded by a phase 2 trial that was positive on an efficacy endpoint (31% bypassed and 15% overrode phase 2 trial). Phase 2 bypass/override was not associated with industry funding (77% vs 89%, 95% CI 0.75-7.55, p = 0.13) or testing already approved interventions (23% vs 15%, 95% CI 0.60-5.14, p = 0.33). Overall, phase 3 trials based on phase 2 bypassed/override were statistically significantly less likely to be positive on their primary outcome (31% vs 57%, respectively, 95% CI 1.21-6.92, p = 0.01). This effect disappeared when indications characterized by nearly universal positive or negative results were excluded. Trials that bypassed/overrode phase 2 trials were not statistically significantly more likely to be terminated early because of safety or futility (29% vs 15%, respectively, 95% CI 0.15-1.18, p = 0.11) and did not show increased risk of adverse events in experimental arms (RR = 1.46, 95% CI 1.19-1.79, vs RR = 1.36, 95% CI 1.10-1.69, respectively, p = 0.65). DISCUSSION Almost half of the neurologic disease phase 3 trials were initiated without the support of a positive phase 2 trial. Although our analysis does not establish harm with bypass/override, its prevalence and the scientific rationale for phase 2 trial testing favor development of criteria defining when phase 2 bypass/override is justified.
Collapse
Affiliation(s)
- Hannah Moyer
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Robyn Mellett
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Karine Vigneault
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Maya McKeown
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Jason Karlawish
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Erika Augustine
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Lon Schneider
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Jonathan Kimmelman
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| |
Collapse
|
27
|
Zhao A, Xu W, Han R, Wei J, Yu Q, Wang M, Li H, Li M, Chi G. Role of histone modifications in neurogenesis and neurodegenerative disease development. Ageing Res Rev 2024; 98:102324. [PMID: 38762100 DOI: 10.1016/j.arr.2024.102324] [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: 12/10/2023] [Revised: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 05/20/2024]
Abstract
Progressive neuronal dysfunction and death are key features of neurodegenerative diseases; therefore, promoting neurogenesis in neurodegenerative diseases is crucial. With advancements in proteomics and high-throughput sequencing technology, it has been demonstrated that histone post-transcriptional modifications (PTMs) are often altered during neurogenesis when the brain is affected by disease or external stimuli and that the degree of histone modification is closely associated with the development of neurodegenerative diseases. This review aimed to show the regulatory role of histone modifications in neurogenesis and neurodegenerative diseases by discussing the changing patterns and functional significance of histone modifications, including histone methylation, acetylation, ubiquitination, phosphorylation, and lactylation. Finally, we explored the control of neurogenesis and the development of neurodegenerative diseases by artificially modulating histone modifications.
Collapse
Affiliation(s)
- Anqi Zhao
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Rui Han
- Department of Neurovascular Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Junyuan Wei
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Qi Yu
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Haokun Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| |
Collapse
|
28
|
Nonnenbroich LF, Bouchal SM, Millesi E, Rechberger JS, Khatua S, Daniels DJ. H3K27-Altered Diffuse Midline Glioma of the Brainstem: From Molecular Mechanisms to Targeted Interventions. Cells 2024; 13:1122. [PMID: 38994974 PMCID: PMC11240752 DOI: 10.3390/cells13131122] [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: 06/03/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024] Open
Abstract
Pediatric high-grade gliomas are a devastating subset of brain tumors, characterized by their aggressive pathophysiology and limited treatment options. Among them, H3 K27-altered diffuse midline gliomas (DMG) of the brainstem stand out due to their distinct molecular features and dismal prognosis. Recent advances in molecular profiling techniques have unveiled the critical role of H3 K27 alterations, particularly a lysine-to-methionine mutation on position 27 (K27M) of the histone H3 tail, in the pathogenesis of DMG. These mutations result in epigenetic dysregulation, which leads to altered chromatin structure and gene expression patterns in DMG tumor cells, ultimately contributing to the aggressive phenotype of DMG. The exploration of targeted therapeutic avenues for DMG has gained momentum in recent years. Therapies, including epigenetic modifiers, kinase inhibitors, and immunotherapies, are under active investigation; these approaches aim to disrupt aberrant signaling cascades and overcome the various mechanisms of therapeutic resistance in DMG. Challenges, including blood-brain barrier penetration and DMG tumor heterogeneity, require innovative approaches to improve drug delivery and personalized treatment strategies. This review aims to provide a comprehensive overview of the evolving understanding of DMG, focusing on the intricate molecular mechanisms driving tumorigenesis/tumor progression and the current landscape of emerging targeted interventions.
Collapse
Affiliation(s)
- Leo F. Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Hopp Children’s Cancer Center, Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - Samantha M. Bouchal
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Elena Millesi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Research Laboratory of the Division of Plastic and Reconstructive Surgery, Department of Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Soumen Khatua
- Department of Pediatric Hematology/Oncology, Section of Neuro-Oncology, Mayo Clinic, Rochester, MN 55905, USA;
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
29
|
Baltira C, Aronica E, Elmquist WF, Langer O, Löscher W, Sarkaria JN, Wesseling P, de Gooijer MC, van Tellingen O. The impact of ATP-binding cassette transporters in the diseased brain: Context matters. Cell Rep Med 2024; 5:101609. [PMID: 38897176 PMCID: PMC11228798 DOI: 10.1016/j.xcrm.2024.101609] [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: 08/22/2023] [Revised: 02/20/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024]
Abstract
ATP-binding cassette (ABC) transporters facilitate the movement of diverse molecules across cellular membranes, including those within the CNS. While most extensively studied in microvascular endothelial cells forming the blood-brain barrier (BBB), other CNS cell types also express these transporters. Importantly, disruptions in the CNS microenvironment during disease can alter transporter expression and function. Through this comprehensive review, we explore the modulation of ABC transporters in various brain pathologies and the context-dependent consequences of these changes. For instance, downregulation of ABCB1 may exacerbate amyloid beta plaque deposition in Alzheimer's disease and facilitate neurotoxic compound entry in Parkinson's disease. Upregulation may worsen neuroinflammation by aiding chemokine-mediated CD8 T cell influx into multiple sclerosis lesions. Overall, ABC transporters at the BBB hinder drug entry, presenting challenges for effective pharmacotherapy. Understanding the context-dependent changes in ABC transporter expression and function is crucial for elucidating the etiology and developing treatments for brain diseases.
Collapse
Affiliation(s)
- Chrysiida Baltira
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Neuroscience, Department of (Neuro)Pathology, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Löscher
- Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands; Laboratory for Childhood Cancer Pathology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Mark C de Gooijer
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Faculty of Biology, Medicine and Health, University of Manchester; The Christie NHS Foundation Trust, Manchester, UK.
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
30
|
Stavrou M, Georgiou E, Kleopa KA. Lumbar Intrathecal Injection in Adult and Neonatal Mice. Curr Protoc 2024; 4:e1091. [PMID: 38923413 DOI: 10.1002/cpz1.1091] [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] [Indexed: 06/28/2024]
Abstract
This article describes a step-by-step process of lumbar intrathecal injection of Evans blue dye and AAV9-EGFP in adult (2-month-old) and neonatal (postnatal day 10) mice. Intrathecal injection is a clinically translatable technique that has already been extensively applied in humans. In mice, intrathecal injection is considered a challenging procedure that requires a trained and experienced researcher. For both adult and neonatal mice, lumbar intrathecal injection is directed into the L5-L6 intervertebral space. Intrathecally injected material enters the cerebrospinal fluid (CSF) within the intrathecal space from where it can directly access the central nervous system (CNS) parenchyma. Simultaneously, intrathecally injected material exits the CSF with pressure gradient and enters the endoneurial fluid and ultimately the peripheral nerves. While in the CSF, the injectable material also enters the bloodstream and systemic circulation through the arachnoid villi. A successful lumbar intrathecal injection results in adequate biodistribution of the injectable material in the CNS, PNS, and peripheral organs. When correctly applied, this technique is considered as minimally invasive and non-disruptive and can be used for the lumbar delivery of any solute. © 2024 Wiley Periodicals LLC. Basic Protocol 1: C57BL/6 adult and P10 mice lumbar intrathecal injection Basic Protocol 2: Tissue collection and preparation for evaluating Evans blue dye diffusion Basic Protocol 3: Tissue collection and preparation for immunohistochemistry staining Basic Protocol 4: Tissue collection and vector genome copy number analysis.
Collapse
Affiliation(s)
- Marina Stavrou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Elena Georgiou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Center for Neuromuscular Disorders, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
31
|
Tan LY, Cunliffe G, Hogan MP, Yeo XY, Oh C, Jin B, Kang J, Park J, Kwon MS, Kim M, Jung S. Emergence of the brain-border immune niches and their contribution to the development of neurodegenerative diseases. Front Immunol 2024; 15:1380063. [PMID: 38863704 PMCID: PMC11165048 DOI: 10.3389/fimmu.2024.1380063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
Historically, the central nervous system (CNS) was regarded as 'immune-privileged', possessing its own distinct immune cell population. This immune privilege was thought to be established by a tight blood-brain barrier (BBB) and blood-cerebrospinal-fluid barrier (BCSFB), which prevented the crossing of peripheral immune cells and their secreted factors into the CNS parenchyma. However, recent studies have revealed the presence of peripheral immune cells in proximity to various brain-border niches such as the choroid plexus, cranial bone marrow (CBM), meninges, and perivascular spaces. Furthermore, emerging evidence suggests that peripheral immune cells may be able to infiltrate the brain through these sites and play significant roles in driving neuronal cell death and pathology progression in neurodegenerative disease. Thus, in this review, we explore how the brain-border immune niches may contribute to the pathogenesis of neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). We then discuss several emerging options for harnessing the neuroimmune potential of these niches to improve the prognosis and treatment of these debilitative disorders using novel insights from recent studies.
Collapse
Affiliation(s)
- Li Yang Tan
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Grace Cunliffe
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Michael Patrick Hogan
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Xin Yi Yeo
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chansik Oh
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Bohwan Jin
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Junmo Kang
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Junho Park
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - MinYoung Kim
- Rehabilitation and Regeneration Research Center, CHA University School of Medicine, Seongnam, Republic of Korea
- Department of Biomedical Science, CHA University School of Medicine, Seongnam, Republic of Korea
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sangyong Jung
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
32
|
Avila A, Zhang SL. A circadian clock regulates the blood-brain barrier across phylogeny. VITAMINS AND HORMONES 2024; 126:241-287. [PMID: 39029975 DOI: 10.1016/bs.vh.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
As the central regulatory system of an organism, the brain is responsible for overseeing a wide variety of physiological processes essential for an organism's survival. To maintain the environment necessary for neurons to function, the brain requires highly selective uptake and elimination of specific molecules through the blood-brain barrier (BBB). As an organism's activities vary throughout the day, how does the BBB adapt to meet the changing needs of the brain? A mechanism is through temporal regulation of BBB permeability via its circadian clock, which will be the focal point of this chapter. To comprehend the circadian clock's role within the BBB, we will first examine the anatomy of the BBB and the transport mechanisms enabling it to fulfill its role as a restrictive barrier. Next, we will define the circadian clock, and the discussion will encompass an introduction to circadian rhythms, the Transcription-Translation Feedback Loop (TTFL) as the mechanistic basis of circadian timekeeping, and the organization of tissue clocks found in organisms. Then, we will cover the role of the circadian rhythms in regulating the cellular mechanisms and functions of the BBB. We discuss the implications of this regulation in influencing sleep behavior, the progression of neurodegenerative diseases, and finally drug delivery for treatment of neurological diseases.
Collapse
Affiliation(s)
- Ashley Avila
- Cell Biology Department, Emory University, Atlanta, GA, United States
| | - Shirley L Zhang
- Cell Biology Department, Emory University, Atlanta, GA, United States.
| |
Collapse
|
33
|
Di Francesco V, Chua AJ, Huang D, D'Souza A, Yang A, Bleier BS, Amiji MM. RNA therapies for CNS diseases. Adv Drug Deliv Rev 2024; 208:115283. [PMID: 38494152 DOI: 10.1016/j.addr.2024.115283] [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: 12/17/2023] [Revised: 03/06/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024]
Abstract
Neurological disorders are a diverse group of conditions that pose an increasing health burden worldwide. There is a general lack of effective therapies due to multiple reasons, of which a key obstacle is the presence of the blood-brain barrier, which limits drug delivery to the central nervous system, and generally restricts the pool of candidate drugs to small, lipophilic molecules. However, in many cases, these are unable to target key pathways in the pathogenesis of neurological disorders. As a group, RNA therapies have shown tremendous promise in treating various conditions because they offer unique opportunities for specific targeting by leveraging Watson-Crick base pairing systems, opening up possibilities to modulate pathological mechanisms that previously could not be addressed by small molecules or antibody-protein interactions. This potential paradigm shift in disease management has been enabled by recent advances in synthesizing, purifying, and delivering RNA. This review explores the use of RNA-based therapies specifically for central nervous system disorders, where we highlight the inherent limitations of RNA therapy and present strategies to augment the effectiveness of RNA therapeutics, including physical, chemical, and biological methods. We then describe translational challenges to the widespread use of RNA therapies and close with a consideration of future prospects in this field.
Collapse
Affiliation(s)
- Valentina Di Francesco
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway Building, Boston, MA 02115, USA; Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA
| | - Andy J Chua
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway Building, Boston, MA 02115, USA; Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA; Department of Otorhinolaryngology - Head and Neck Surgery, Sengkang General Hospital, 110 Sengkang E Way, 544886, Singapore
| | - Di Huang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway Building, Boston, MA 02115, USA; Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA
| | - Anisha D'Souza
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway Building, Boston, MA 02115, USA; Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA
| | - Alicia Yang
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Benjamin S Bleier
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway Building, Boston, MA 02115, USA; Department of Chemical Engineering, College of Engineering, Northeastern University, 360 Huntington Avenue, 140 The Fenway Building, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Werner MS, Aras S, Morgan AR, Roamer J, Param NJ, Olagbegi K, Lamontagne RJ, Greig JA, Wilson JM. Adeno-associated virus-mediated trastuzumab delivery to the central nervous system for human epidermal growth factor receptor 2+ brain metastasis. Cancer Gene Ther 2024; 31:766-777. [PMID: 38480976 DOI: 10.1038/s41417-024-00751-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 05/19/2024]
Abstract
Trastuzumab improves overall survival for HER2+ breast cancer, but its short half-life in the cerebrospinal fluid (~2-4 days) and delivery limitations restrict the ability to target HER2+ central nervous system (CNS) disease. We developed an adeno-associated virus (AAV) vector expressing a codon-optimized, ubiquitin C (UbC)-promoter-driven trastuzumab sequence (AAV9.UbC.trastuzumab) for intrathecal administration. Transgene expression was evaluated in adult Rag1 knockout mice and rhesus nonhuman primates (NHPs) after a single intracerebroventricular (ICV) or intra-cisterna magna (ICM) AAV9.UbC.trastuzumab injection, respectively, using real-time PCR, ELISA, Western blot, in situ hybridization, single-nucleus RNA sequencing, and liquid chromatography-mass spectrometry; antitumor efficacy was evaluated in brain xenografts using HER2+ breast cancer cell lines (BT-474, MDA-MB-453). Transgene expression was detected in brain homogenates of Rag1 knockout mice following a single ICV injection of AAV9.UbC.trastuzumab (1 × 1011 vector genome copies [GC]/mouse) and tumor progression was inhibited in xenograft models of breast-to-brain metastasis. In NHPs, ICM delivery of AAV9.UbC.trastuzumab (3 × 1013 GC/animal) was well tolerated (36-37 days in-life) and resulted in transgene expression in CNS tissues and cerebrospinal fluid at levels sufficient to induce complete tumor remission in MDA-MB-453 brain xenografts. With AAV9's proven clinical safety record, this gene therapy may represent a viable approach for targeting HER2 + CNS malignancies.
Collapse
Affiliation(s)
- Marcela S Werner
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shweta Aras
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ashleigh R Morgan
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jillian Roamer
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nesteene J Param
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kanyin Olagbegi
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - R Jason Lamontagne
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jenny A Greig
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
35
|
Wu D, Luo R, Chen Y, Zheng Z, Gui S, He N. Preparation, characterisation, pharmacokinetics and distribution of esculin microspheres administered via intravitreal injection into rabbit brain. Xenobiotica 2024; 54:233-247. [PMID: 38638108 DOI: 10.1080/00498254.2024.2341402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/07/2024] [Indexed: 04/20/2024]
Abstract
This study explored the distribution of esculin microspheres in rabbit brain tissue following intravitreal injection and investigated the possibility of direct entry of the drug into the brain through the eye, to develop a formulation with enhanced therapeutic efficacy against Parkinson's disease.Chitosan microspheres of esculin were prepared via an emulsification cross-linking method and their characteristics were evaluated, including angle of repose, bulk density, and swelling ratio. Furthermore, the pharmacokinetic parameters and brain tissue distribution in rabbits were compared among groups administered esculin eye drops, intravitreal esculin solution, and intravitreal esculin microspheres, to determine whether esculin could enter the brain through an ocular route.The results showed that the prepared esculin microspheres were spherical and had good fluidity. Notably, intravitreal administration enhanced the area under the curve (AUC) of esculin in the thalamus. Delivery through microspheres prolonged the drug retention time in both rabbit plasma and brain tissues, as well as the brain-targeting efficiency of esculin.The collective findings indicated that there may be a direct eye-brain pathway facilitating enter of esculin microspheres into brain tissue after intravitreal injection, supporting the utility of intravitreal esculin microspheres as an effective therapeutic formulation for Parkinson's disease, a long-term chronic condition.
Collapse
Affiliation(s)
- Danqing Wu
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Rui Luo
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yangnan Chen
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Zhiyun Zheng
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medical Sciences, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
| | - Shuangying Gui
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medical Sciences, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
| | - Ning He
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medical Sciences, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
| |
Collapse
|
36
|
Seo Y, Chang KW, Lee J, Kong C, Shin J, Chang JW, Na YC, Chang WS. Optimal timing for drug delivery into the hippocampus by focused ultrasound: A comparison of hydrophilic and lipophilic compounds. Heliyon 2024; 10:e29480. [PMID: 38644896 PMCID: PMC11033133 DOI: 10.1016/j.heliyon.2024.e29480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024] Open
Abstract
Aims Previous studies have reported that focused ultrasound (FUS) helps modulate the blood-brain barrier (BBB). These studies have generally used the paracellular pathway owing to tight junction proteins (TJPs) regulation. However, BBB transport pathways also include diffusion and transcytosis. Few studies have examined transcellular transport across endothelial cells. We supposed that increased BBB permeability caused by FUS may affect transcytosis. We investigated drug delivery through transcytosis and paracellular transport to the brain after BBB modulation using FUS. Main methods FUS and microbubbles were applied to the hippocampus of rats, and were euthanized at 1, 4, 24, and 48 h after sonication. To investigate paracellular transport, we analyzed TJPs, including zona occludens-1 (ZO-1) and occludin. We also investigated caveola-mediated transcytosis by analyzing caveola formation and major facilitator superfamily domain-containing 2a (Mfsd2a) levels, which inhibit caveola vesicle formation. Key findings One hour after FUS, ZO-1 and occludin expression was the lowest and gradually increased over time, returning to baseline 24 h after FUS treatment. Compared with that of TJPs, caveola formation started to increase 1 h after FUS treatment and peaked at 4 h after FUS treatment before returning to baseline by 48 h after FUS treatment. Decreased Mfsd2a levels were observed at 1 h and 4 h after FUS treatment, indicating increased caveola formation. Significance FUS induces BBB permeability changes and regulates both paracellular transport and caveola-mediated transcytosis. However, a time difference was observed between these two mechanisms. Hence, when delivering drugs into the brain after FUS, the optimal drug administration timing should be determined by the mechanism by which each drug passes through the BBB.
Collapse
Affiliation(s)
- Younghee Seo
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyung Won Chang
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Chanho Kong
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Jaewoo Shin
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu, 41061, South Korea
| | - Jin Woo Chang
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Young Cheol Na
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- Department of Neurosurgery, Catholic Kwandong University College of Medicine, International St. Mary's Hospital, Incheon Metropolitan City, South Korea
| | - Won Seok Chang
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
37
|
Li J, Wu A, Kim S. Mechanistic Modeling of Intrathecal Chemotherapy Pharmacokinetics in the Human Central Nervous System. Clin Cancer Res 2024; 30:1397-1408. [PMID: 38289997 PMCID: PMC10984761 DOI: 10.1158/1078-0432.ccr-23-3062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/05/2023] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
PURPOSE The pharmacokinetics of intrathecally administered antibody or small-molecule drugs in the human central nervous system (CNS) remains poorly understood. This study aimed to provide mechanistic and quantitative perspectives on the CNS pharmacokinetics of intrathecal chemotherapy, by using a physiologically based pharmacokinetic (PBPK) modeling approach. EXPERIMENTAL DESIGN A novel CNS PBPK model platform was developed and verified, which accounted for the human CNS general anatomy and physiologic processes governing drug distribution and disposition. The model was used to predict CNS pharmacokinetics of antibody (trastuzumab) and small-molecule drugs (methotrexate, abemaciclib, tucatinib) following intraventricular injection or intraventricular 24-hour infusion, and to assess the key determinants of drug penetration into the deep brain parenchyma. RESULTS Intraventricularly administered antibody and small-molecule drugs exhibited distinct temporal and spatial distribution and disposition in human CNS. Both antibody and small-molecule drugs achieved supratherapeutic or therapeutic concentrations in the cerebrospinal fluid (CSF) compartments and adjacent brain tissue. While intrathecal small-molecule drugs penetrated the deep brain parenchyma to a negligible extent, intrathecal antibodies may achieve therapeutic concentrations in the deep brain parenchyma. Intraventricular 24-hour infusion enabled prolonged CNS exposure to therapeutically relevant concentrations while avoiding excessively high and potentially neurotoxic drug concentrations. CONCLUSIONS CNS PBPK modeling, in line with available clinical efficacy data, confirms the therapeutic value of intrathecal chemotherapy with antibody or small-molecule drugs for treating neoplastic meningitis and warrants further clinical investigation of intrathecal antibody drugs to treat brain parenchyma tumors. Compared with intraventricular injection, intraventricular 24-hour infusion may mitigate neurotoxicity while retaining potential efficacy.
Collapse
Affiliation(s)
- Jing Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 482012
| | - Andrew Wu
- Northville High School, 45700 Six Mile Rd, Northville, MI 48168
| | - Seongho Kim
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 482012
| |
Collapse
|
38
|
Luan X, Xing H, Guo F, Liu W, Jiao Y, Liu Z, Wang X, Gao S. The role of ncRNAs in depression. Heliyon 2024; 10:e27307. [PMID: 38496863 PMCID: PMC10944209 DOI: 10.1016/j.heliyon.2024.e27307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/19/2024] Open
Abstract
Depressive disorders have a significant impact on public health, and depression have an unsatisfactory recurrence rate and are challenging to treat. Non-coding RNAs (ncRNAs) are RNAs that do not code protein, which have been shown to be crucial for transcriptional regulation. NcRNAs are important to the onset, progress and treatment of depression because they regulate various physiological functions. This makes them distinctively useful as biomarkers for diagnosing and tracking responses to therapy among individuals with depression. It is important to seek out and summarize the research findings on the impact of ncRNAs on depression since significant advancements have been made in this area recently. Hence, we methodically outlined the findings of published researches on ncRNAs and depression, focusing on microRNAs. Above all, this review aims to improve our understanding of ncRNAs and provide new insights of the diagnosis and treatment of depression.
Collapse
Affiliation(s)
- Xinchi Luan
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Han Xing
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Feifei Guo
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Weiyi Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Yang Jiao
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Zhenyu Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Xuezhe Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Shengli Gao
- Biomedical Center, Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
39
|
Ueno M, Chiba Y, Murakami R, Miyai Y, Matsumoto K, Wakamatsu K, Nakagawa T, Takebayashi G, Uemura N, Yanase K, Ogino Y. Transporters, Ion Channels, and Junctional Proteins in Choroid Plexus Epithelial Cells. Biomedicines 2024; 12:708. [PMID: 38672064 PMCID: PMC11048166 DOI: 10.3390/biomedicines12040708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
The choroid plexus (CP) plays significant roles in secreting cerebrospinal fluid (CSF) and forming circadian rhythms. A monolayer of epithelial cells with tight and adherens junctions of CP forms the blood-CSF barrier to control the movement of substances between the blood and ventricles, as microvessels in the stroma of CP have fenestrations in endothelial cells. CP epithelial cells are equipped with several kinds of transporters and ion channels to transport nutrient substances and secrete CSF. In addition, junctional components also contribute to CSF production as well as blood-CSF barrier formation. However, it remains unclear how junctional components as well as transporters and ion channels contribute to the pathogenesis of neurodegenerative disorders. In this manuscript, recent findings regarding the distribution and significance of transporters, ion channels, and junctional proteins in CP epithelial cells are introduced, and how changes in expression of their epithelial proteins contribute to the pathophysiology of brain disorders are reviewed.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Ryuta Murakami
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Yumi Miyai
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Koichi Matsumoto
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Keiji Wakamatsu
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Toshitaka Nakagawa
- Division of Research Instrument and Equipment, Research Facility Center, Kagawa University, Kagawa 761-0793, Japan;
| | - Genta Takebayashi
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| | - Naoya Uemura
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| | - Ken Yanase
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| | - Yuichi Ogino
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| |
Collapse
|
40
|
Abstract
The blood-brain barrier (BBB) is a critical interface separating the central nervous system from the peripheral circulation, ensuring brain homeostasis and function. Recent research has unveiled a profound connection between the BBB and circadian rhythms, the endogenous oscillations synchronizing biological processes with the 24-hour light-dark cycle. This review explores the significance of circadian rhythms in the context of BBB functions, with an emphasis on substrate passage through the BBB. Our discussion includes efflux transporters and the molecular timing mechanisms that regulate their activities. A significant focus of this review is the potential implications of chronotherapy, leveraging our knowledge of circadian rhythms for improving drug delivery to the brain. Understanding the temporal changes in BBB can lead to optimized timing of drug administration, to enhance therapeutic efficacy for neurological disorders while reducing side effects. By elucidating the interplay between circadian rhythms and drug transport across the BBB, this review offers insights into innovative therapeutic interventions.
Collapse
Affiliation(s)
- Mari Kim
- Cell Biology Department, Emory University, Atlanta, GA, USA (M.K., S.L.Z.)
| | - Richard F Keep
- Neurosurgery, University of Michigan, Ann Arbor, MI, USA (R.F.K.)
| | - Shirley L Zhang
- Cell Biology Department, Emory University, Atlanta, GA, USA (M.K., S.L.Z.)
| |
Collapse
|
41
|
Lim SH, Yee GT, Khang D. Nanoparticle-Based Combinational Strategies for Overcoming the Blood-Brain Barrier and Blood-Tumor Barrier. Int J Nanomedicine 2024; 19:2529-2552. [PMID: 38505170 PMCID: PMC10949308 DOI: 10.2147/ijn.s450853] [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: 12/05/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024] Open
Abstract
The blood-brain barrier (BBB) and blood-tumor barrier (BTB) pose substantial challenges to efficacious drug delivery for glioblastoma multiforme (GBM), a primary brain tumor with poor prognosis. Nanoparticle-based combinational strategies have emerged as promising modalities to overcome these barriers and enhance drug penetration into the brain parenchyma. This review discusses various nanoparticle-based combinatorial approaches that combine nanoparticles with cell-based drug delivery, viral drug delivery, focused ultrasound, magnetic field, and intranasal drug delivery to enhance drug permeability across the BBB and BTB. Cell-based drug delivery involves using engineered cells as carriers for nanoparticles, taking advantage of their intrinsic migratory and homing capabilities to facilitate the transport of therapeutic payloads across BBB and BTB. Viral drug delivery uses engineered viral vectors to deliver therapeutic genes or payloads to specific cells within the GBM microenvironment. Focused ultrasound, coupled with microbubbles or nanoparticles, can temporarily disrupt the BBB to increase drug permeability. Magnetic field-guided drug delivery exploits magnetic nanoparticles to facilitate targeted drug delivery under an external magnetic field. Intranasal drug delivery offers a minimally invasive avenue to bypass the BBB and deliver therapeutic agents directly to the brain via olfactory and trigeminal pathways. By combining these strategies, synergistic effects can enhance drug delivery efficiency, improve therapeutic efficacy, and reduce off-target effects. Future research should focus on optimizing nanoparticle design, exploring new combination strategies, and advancing preclinical and clinical investigations to promote the translation of nanoparticle-based combination therapies for GBM.
Collapse
Affiliation(s)
- Su Hyun Lim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Gi Taek Yee
- Department of Neurosurgery, Gil Medical Center, Gachon University, School of Medicine, Incheon, 21565, South Korea
| | - Dongwoo Khang
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
- Department of Physiology, School of Medicine, Gachon University, Incheon, 21999, South Korea
| |
Collapse
|
42
|
Nonaka H, Sakamoto S, Shiraiwa K, Ishikawa M, Tamura T, Okuno K, Kondo T, Kiyonaka S, Susaki EA, Shimizu C, Ueda HR, Kakegawa W, Arai I, Yuzaki M, Hamachi I. Bioorthogonal chemical labeling of endogenous neurotransmitter receptors in living mouse brains. Proc Natl Acad Sci U S A 2024; 121:e2313887121. [PMID: 38294939 PMCID: PMC10861872 DOI: 10.1073/pnas.2313887121] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/16/2023] [Indexed: 02/02/2024] Open
Abstract
Neurotransmitter receptors are essential components of synapses for communication between neurons in the brain. Because the spatiotemporal expression profiles and dynamics of neurotransmitter receptors involved in many functions are delicately governed in the brain, in vivo research tools with high spatiotemporal resolution for receptors in intact brains are highly desirable. Covalent labeling by chemical reaction (chemical labeling) of proteins without genetic manipulation is now a powerful method for analyzing receptors in vitro. However, selective target receptor labeling in the brain has not yet been achieved. This study shows that ligand-directed alkoxyacylimidazole (LDAI) chemistry can be used to selectively tether synthetic probes to target endogenous receptors in living mouse brains. The reactive LDAI reagents with negative charges were found to diffuse well over the whole brain and could selectively label target endogenous receptors, including AMPAR, NMDAR, mGlu1, and GABAAR. This simple and robust labeling protocol was then used for various applications: three-dimensional spatial mapping of endogenous receptors in the brains of healthy and disease-model mice; multi-color receptor imaging; and pulse-chase analysis of the receptor dynamics in postnatal mouse brains. Here, results demonstrated that bioorthogonal receptor modification in living animal brains may provide innovative molecular tools that contribute to the in-depth understanding of complicated brain functions.
Collapse
Affiliation(s)
- Hiroshi Nonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto615-8510, Japan
- Hamachi Innovative Molecular Technology for Neuroscience, Exploratory Research for Advanced Technology, Japan Science and Technology Agency, Kyoto615-8530, Japan
| | - Seiji Sakamoto
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto615-8510, Japan
- Hamachi Innovative Molecular Technology for Neuroscience, Exploratory Research for Advanced Technology, Japan Science and Technology Agency, Kyoto615-8530, Japan
| | - Kazuki Shiraiwa
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto615-8510, Japan
| | - Mamoru Ishikawa
- Hamachi Innovative Molecular Technology for Neuroscience, Exploratory Research for Advanced Technology, Japan Science and Technology Agency, Kyoto615-8530, Japan
| | - Tomonori Tamura
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto615-8510, Japan
- Hamachi Innovative Molecular Technology for Neuroscience, Exploratory Research for Advanced Technology, Japan Science and Technology Agency, Kyoto615-8530, Japan
| | - Kyohei Okuno
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto615-8510, Japan
| | - Takumi Kondo
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya464-8603, Japan
| | - Shigeki Kiyonaka
- Hamachi Innovative Molecular Technology for Neuroscience, Exploratory Research for Advanced Technology, Japan Science and Technology Agency, Kyoto615-8530, Japan
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya464-8603, Japan
| | - Etsuo A. Susaki
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo113-8421, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka 565-5241, Japan
| | - Chika Shimizu
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka 565-5241, Japan
| | - Hiroki R. Ueda
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka 565-5241, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| | - Wataru Kakegawa
- Hamachi Innovative Molecular Technology for Neuroscience, Exploratory Research for Advanced Technology, Japan Science and Technology Agency, Kyoto615-8530, Japan
- Department of Neurophysiology, Keio University School of Medicine, Tokyo160-8582, Japan
| | - Itaru Arai
- Department of Neurophysiology, Keio University School of Medicine, Tokyo160-8582, Japan
| | - Michisuke Yuzaki
- Department of Neurophysiology, Keio University School of Medicine, Tokyo160-8582, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto615-8510, Japan
- Hamachi Innovative Molecular Technology for Neuroscience, Exploratory Research for Advanced Technology, Japan Science and Technology Agency, Kyoto615-8530, Japan
| |
Collapse
|
43
|
Wang H, Yang J, Li X, Zhao H. Current state of immune checkpoints therapy for glioblastoma. Heliyon 2024; 10:e24729. [PMID: 38298707 PMCID: PMC10828821 DOI: 10.1016/j.heliyon.2024.e24729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Glioblastoma (GBM), one of the most aggressive forms of brain cancer, has limited treatment options. Recent years have witnessed the remarkable success of checkpoint inhibitor immunotherapy across various cancer types. Against this backdrop, several clinical trials investigating checkpoint inhibitors for GBM are underway in multiple countries. Furthermore, the integration of immunotherapy with traditional treatment approaches is now emerging as a highly promising strategy. This review summarizes the latest advancements in checkpoint inhibitor immunotherapy for GBM treatment. We provide a concise yet comprehensive overview of current GBM immunotherapy options. Additionally, this review underscores combination strategies and potential biomarkers for predicting response and resistance in GBM immunotherapies.
Collapse
Affiliation(s)
- He Wang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Jing Yang
- Department of Emergency Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Xiangjun Li
- School of medicine, Department of Breast surgery, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| |
Collapse
|
44
|
Sharma P, Otto M. Multifunctional nanocomposites modulating the tumor microenvironment for enhanced cancer immunotherapy. Bioact Mater 2024; 31:440-462. [PMID: 37701452 PMCID: PMC10494322 DOI: 10.1016/j.bioactmat.2023.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer immunotherapy has gained momentum for treating malignant tumors over the past decade. Checkpoint blockade and chimeric antigen receptor cell therapy (CAR-T) have shown considerable potency against liquid and solid cancers. However, the tumor microenvironment (TME) is highly immunosuppressive and hampers the effect of currently available cancer immunotherapies on overall treatment outcomes. Advancements in the design and engineering of nanomaterials have opened new avenues to modulate the TME. Progress in the current nanocomposite technology can overcome immunosuppression and trigger robust immunotherapeutic responses by integrating synergistic functions of different molecules. We will review recent advancements in nanomedical applications and discuss specifically designed nanocomposites modulating the TME for cancer immunotherapy. In addition, we provide information on the current landscape of clinical-stage nanocomposites for cancer immunotherapy.
Collapse
Affiliation(s)
- Prashant Sharma
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
| | - Mario Otto
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
- Center for Cancer and Blood Disorders (CCBD), Phoenix Children's, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| |
Collapse
|
45
|
Debori Y, Igari T, Nakakariya M, Hirabayashi H, Aoyama K, Amano N, Kurosawa T, Kubo Y, Deguchi Y. Involvement of Proton-Coupled Organic Cation Antiporter in Human Blood-Brain Barrier Transport of Mesoridazine and Metoclopramide. Biol Pharm Bull 2024; 47:1662-1667. [PMID: 39414441 DOI: 10.1248/bpb.b24-00329] [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] [Indexed: 10/18/2024]
Abstract
Mesoridazine and metoclopramide are cationic drugs that are distributed in the human brain despite being substrates of multidrug resistance protein 1 (MDR1), an efflux transporter expressed at the blood-brain barrier (BBB). We investigated their transport mechanisms at the BBB using hCMEC/D3, a human cerebral microvascular endothelial cell line often used as an in vitro BBB model. The cells exhibited time- and concentration-dependent uptake of mesoridazine and metoclopramide, with Km values of 34 and 277 µM, respectively. The uptake of both drugs significantly decreased in the presence of typical inhibitors and/or substrates of the H+-coupled organic cation (H+/OC) antiporter but not in the presence of inhibitors or substrates of organic cation transporters (OCTs), OCTN2, OATPs, SLC35F2, or the plasma membrane monoamine transporter (PMAT). Furthermore, metoclopramide uptake by hCMEC/D3 cells was pH- and energy-dependent, whereas mesoridazine uptake was unaffected by intracellular acidification and treatment with metabolic inhibitors. These results suggest that the H+/OC antiporter is involved in the influx of mesoridazine and metoclopramide into the brain across the BBB.
Collapse
Affiliation(s)
- Yasuyuki Debori
- Faculty of Pharmaceutical Sciences, Teikyo University
- Discovery DM and Toxicology, Axcelead Drug Discovery Partners, Inc
| | - Tomoko Igari
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited
| | - Masanori Nakakariya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited
| | - Kazunobu Aoyama
- Discovery DM and Toxicology, Axcelead Drug Discovery Partners, Inc
| | - Nobuyuki Amano
- Discovery DM and Toxicology, Axcelead Drug Discovery Partners, Inc
| | | | | | | |
Collapse
|
46
|
Ronaldson PT, Davis TP. Blood-brain barrier transporters: a translational consideration for CNS delivery of neurotherapeutics. Expert Opin Drug Deliv 2024; 21:71-89. [PMID: 38217410 PMCID: PMC10842757 DOI: 10.1080/17425247.2024.2306138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/12/2024] [Indexed: 01/15/2024]
Abstract
INTRODUCTION Successful neuropharmacology requires optimization of CNS drug delivery and, by extension, free drug concentrations at brain molecular targets. Detailed assessment of blood-brain barrier (BBB) physiological characteristics is necessary to achieve this goal. The 'next frontier' in CNS drug delivery is targeting BBB uptake transporters, an approach that requires evaluation of brain endothelial cell transport processes so that effective drug accumulation and improved therapeutic efficacy can occur. AREAS COVERED BBB permeability of drugs is governed by tight junction protein complexes (i.e., physical barrier) and transporters/enzymes (i.e., biochemical barrier). For most therapeutics, a component of blood-to-brain transport involves passive transcellular diffusion. Small molecule drugs that do not possess acceptable physicochemical characteristics for passive permeability may utilize putative membrane transporters for CNS uptake. While both uptake and efflux transport mechanisms are expressed at the brain microvascular endothelium, uptake transporters can be targeted for optimization of brain drug delivery and improved treatment of neurological disease states. EXPERT OPINION Uptake transporters represent a unique opportunity to optimize brain drug delivery by leveraging the endogenous biology of the BBB. A rigorous understanding of these transporters is required to improve translation from the bench to clinical trials and stimulate the development of new treatment paradigms for neurological diseases.
Collapse
Affiliation(s)
| | - Thomas P. Davis
- Department of Pharmacology, University of Arizona College of Medicine
| |
Collapse
|
47
|
Kumar N, Khurana B, Arora D. Nose-to-brain drug delivery for the treatment of glioblastoma multiforme: nanotechnological interventions. Pharm Dev Technol 2023; 28:1032-1047. [PMID: 37975846 DOI: 10.1080/10837450.2023.2285506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive malignant brain tumor with a short survival rate. Extensive research is underway for the last two decades to find an effective treatment for GBM but the tortuous pathophysiology, development of chemoresistance, and presence of BBB are the major challenges, prompting scientists to look for alternative targets and delivery strategies. Therefore, the nose to brain delivery emerged as an unorthodox and non-invasive route, which delivers the drug directly to the brain via the olfactory and trigeminal pathways and also bypasses the BBB and hepatic metabolism of the drug. However, mucociliary clearance, low administration volume, and less permeability of nasal mucosa are the obstacles retrenching the brain drug concentration. Thus, nanocarrier delivery through this route may conquer these limitations because of their unique surface characteristics and smaller size. In this review, we have emphasized the advantages and limitations of nanocarrier technologies such as polymeric, lipidic, inorganic, and miscellaneous nanoparticles used for nose-to-brain drug delivery against GBM in the past 10 years. Furthermore, recent advances, patents, and clinical trials are highlighted. However, most of these studies are in the early stages, so translating their outcomes into a marketed formulation would be a milestone in the better progression and survival of glioma patients.
Collapse
Affiliation(s)
- Nitish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Bharat Khurana
- Department of Pharmaceutics, Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, India
| | - Daisy Arora
- Department of Pharmacy, Panipat Institute of Engineering and Technology, Panipat, Haryana, India
| |
Collapse
|
48
|
Schwinghamer K, Siahaan TJ. Enhancing Antibody Exposure in the Central Nervous System: Mechanisms of Uptake, Clearance, and Strategies for Improved Brain Delivery. JOURNAL OF NANOTHERANOSTICS 2023; 4:463-479. [PMID: 39897432 PMCID: PMC11784990 DOI: 10.3390/jnt4040020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
Abstract
Antibodies (mAbs) are attractive molecules for their application as a diagnostic and therapeutic agent for diseases of the central nervous system (CNS). mAbs can be generated to have high affinity and specificity to target molecules in the CNS. Unfortunately, only a very small number of mAbs have been specifically developed and approved for neurological indications. This is primarily attributed to their low exposure within the CNS, hindering their ability to reach and effectively engage their potential targets in the brain. This review discusses aspects of various barriers such as the blood-brain barrier (BBB) and blood-cerebrospinal fluid (CSF) barrier (BCSFB) that regulate the entry and clearance of mAbs into and from the brain. The roles of the glymphatic system on brain exposure and clearance are being described. We also discuss the proposed mechanisms of the uptake of mAbs into the brain and for clearance. Finally, several methods of enhancing the exposure of mAbs in the CNS were discussed, including receptor-mediated transcytosis, osmotic BBB opening, focused ultrasound (FUS), BBB-modulating peptides, and enhancement of mAb brain retention.
Collapse
Affiliation(s)
- Kelly Schwinghamer
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66046, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66046, USA
| |
Collapse
|
49
|
Gao X, Diep JK, Norris DA, Yu RZ, Geary RS. Predicting the pharmacokinetics and pharmacodynamics of antisense oligonucleotides: an overview of various approaches and opportunities for PBPK/PD modelling. Expert Opin Drug Metab Toxicol 2023; 19:979-990. [PMID: 37970635 DOI: 10.1080/17425255.2023.2283524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
INTRODUCTION Advances in research and development (R&D) have enabled many approvals of antisense oligonucleotides (ASOs). Its administration expanded from systemic to local for treating various diseases, where predicting target tissue exposures and pharmacokinetics (PK) and pharmacodynamics (PD) in human can be critical. AREAS COVERED A literature search for PBPK/PD models of ASOs was conducted using PubMed and Embase (to 1 April 2023). ASO PK and PD in animals and humans and modeling approaches including physiologically based (PB) are summarized; and relevance and impacts of PBPK/PD modeling are assessed. EXPERT OPINION Allometric scaling and compartmental PK/PD modeling have been successful to predict human ASO PK/PD, addressing most R&D needs. Understanding tissue distribution of ASOs can be crucial for their efficacy and safety especially for intrathecal (IT), pulmonary, or other local routes. PBPK/PD modeling is expected to improve such understanding, for which, efforts have been sporadic. However, developing a PBPK/PD model requires careful review of known biology/pharmacology and thoughtful experimental designs. Resulting models have the potential to predict target/specified tissue exposures and responses in human adults and pediatrics. Ultimately, a PBPK/PD modeling approach can lead to more efficient and rational clinical development, resulting in well-informed decision making and a shortened timeline.
Collapse
Affiliation(s)
- Xiang Gao
- Preclinical Development, Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | - John K Diep
- Preclinical Development, Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | - Daniel A Norris
- Preclinical Development, Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | - Rosie Z Yu
- Preclinical Development, Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | - Richard S Geary
- Preclinical Development, Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| |
Collapse
|
50
|
Mwema A, Muccioli GG, des Rieux A. Innovative drug delivery strategies to the CNS for the treatment of multiple sclerosis. J Control Release 2023; 364:435-457. [PMID: 37926243 DOI: 10.1016/j.jconrel.2023.10.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/05/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Disorders of the central nervous system (CNS), such as multiple sclerosis (MS) represent a great emotional, financial and social burden. Despite intense efforts, great unmet medical needs remain in that field. MS is an autoimmune, chronic inflammatory demyelinating disease with no curative treatment up to date. The current therapies mostly act in the periphery and seek to modulate aberrant immune responses as well as slow down the progression of the disease. Some of these therapies are associated with adverse effects related partly to their administration route and show some limitations due to their rapid clearance and inability to reach the CNS. The scientific community have recently focused their research on developing MS therapies targeting different processes within the CNS. However, delivery of therapeutics to the CNS is mainly limited by the presence of the blood-brain barrier (BBB). Therefore, there is a pressing need to develop new drug delivery strategies that ensure CNS availability to capitalize on identified therapeutic targets. Several approaches have been developed to overcome or bypass the BBB and increase delivery of therapeutics to the CNS. Among these strategies, the use of alternative routes of administration, such as the nose-to-brain (N2B) pathway, offers a promising non-invasive option in the scope of MS, as it would allow a direct transport of the drugs from the nasal cavity to the brain. Moreover, the combination of bioactive molecules within nanocarriers bring forth new opportunities for MS therapies, allowing and/or increasing their transport to the CNS. Here we will review and discuss these alternative administration routes as well as the nanocarrier approaches useful to deliver drugs for MS.
Collapse
Affiliation(s)
- Ariane Mwema
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium; Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium
| | - Giulio G Muccioli
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium.
| | - Anne des Rieux
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium.
| |
Collapse
|