1
|
Shi H, Wang B, Shi Z, Ma H, Li Y, Liu Y, Zhao Y, Xia N, Wu C, Gao Y. Paclitaxel-Ang-2-functionalized bionic mesoporous selenium nanoparticles for targeted therapy of glioma. Pharmacol Res 2025; 216:107783. [PMID: 40378941 DOI: 10.1016/j.phrs.2025.107783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/19/2025]
Abstract
Glioma, the most prevalent primary intracranial tumor, presents significant clinical treatment challenges due to its high invasiveness and therapeutic resistance. Therefore, the development of a targeted therapeutic agent that is both highly effective and low in toxicity is crucial. In this research, we aimed to design a bionic mesoporous selenium nanoparticle (ACMLMSeP) functionalized with paclitaxel and Ang-2 for nasal administration as a targeted treatment approach for glioma. Nasal administration facilitates direct delivery of drugs to the brain through the olfactory nerve, thereby circumventing the protective mechanisms of the blood-brain barrier. Mesoporous selenium (MSe) significantly enhances the loading capacity for insoluble drugs while improving their water solubility. The functionalization of MSe enables slow drug release and facilitates targeted drug accumulation. Moreover, accumulated nano-selenium promotes reactive oxygen species (ROS) production, induces autophagy, and synergizes with drugs to accelerate apoptosis in tumor cells. Analysis using Transmission Electron Microscopy (TEM) images and Dynamic Light Scattering (DLS) indicated that ACMLMSe has an average particle size of roughly 135 nm. Results from in vitro release assessments indicated that the ACMLMSeP sustained the release of the drug, reaching a total release rate of 74.96 ± 2.34 % within 24 h. Cellular uptake studies and in vivo imaging showed the strong targeting capabilities of the ACMLMSeP nanoparticles. Furthermore, the results obtained from the MTT assays, flow cytometry analysis, immunofluorescence staining, and in vivo antitumor evaluations collectively revealed that ACMLMSeP effectively inhibited proliferation while promoting apoptosis in C6 cells. In summary, these experimental findings clearly suggest that ACMLMSeP may serve as a promising biomimetic nanosystem for the targeted treatment of brain glioma.
Collapse
Affiliation(s)
- Huan Shi
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Biaobiao Wang
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Zhiwei Shi
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Huilin Ma
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Yunmei Li
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Ying Liu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Yu Zhao
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Nan Xia
- Xuzhou Central Hospital, Quanshan District, Xuzhou, Jiangsu, China.
| | - Chao Wu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Yu Gao
- Department of Medical Oncology, the First Affiliated Hospital of Jinzhou Medical University, No.2, the Fifth Section of Renmin Street, Guta District, Jinzhou, Liaoning 121001, China.
| |
Collapse
|
2
|
Qu M, Wang Q, Wang X, Tang J, Dong X, Zhao C, Guan Q. Recent advances in nanomaterial-based brain-targeted delivery systems for glioblastoma therapy. Nanomedicine (Lond) 2025:1-17. [PMID: 40353316 DOI: 10.1080/17435889.2025.2503694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Glioblastoma (GBM) poses a formidable challenge because of its high morbidity and mortality. The therapeutic efficacy of GBM is significantly hampered by the intricate blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB). Nanomaterial-based brain-targeted delivery systems have shown great potential for effectively delivering therapeutic agents for GBM treatment by overcoming the limitations of conventional drugs, such as poor BBB penetration, a short half-life, and low bioavailability. This review focuses on an in-depth analysis of the interplay between the BBB/BBTB and drug transport kinetics while analyzing innovative nanoparticle-mediated strategies for enhanced GBM treatment. Moreover, the delivery strategies of nanoparticle-based brain-targeted systems are emphasized, with particular attention given to biomimetic nanoparticles (BMNPs), whose unique advantages. The current challenges, translational potential, and future research directions in this rapidly evolving field are comprehensively discussed, highlighting advances in nanomaterial applications. This review aims to stimulate further research into GBM delivery systems, offering promising avenues for maximizing the therapeutic effects of gene drugs or chemotherapeutic agents in practical applications.
Collapse
Affiliation(s)
- Mingyue Qu
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Quan Wang
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xinying Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Jie Tang
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Xiyao Dong
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Chaoyue Zhao
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Qingxiang Guan
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
3
|
Wang R, Wang K, Li Z, Long H, Zhang D, Li Y, Xia Z, Guo X, Chen W, Cao F, Jiang F. Blood-Brain Barrier-Permeable, Reactive Oxygen Species-Producing, and Mitochondria-Targeting Nanosystem Amplifies Glioblastoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27434-27447. [PMID: 40289326 DOI: 10.1021/acsami.5c02384] [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: 04/30/2025]
Abstract
Gemcitabine (GTB), a clinically approved nucleoside analogue for cancer treatment, faces therapeutic limitations due to rapid enzymatic deactivation by cytidine deaminase (CDA) in tumor microenvironments. Over 90% of systemically administered GTB undergoes catalytic conversion to inactive 2'-deoxy-2',2'-difluorouracil metabolites through CDA-mediated deamination. To address this pharmacological challenge, we developed a multifunctional codelivery nanosystem through strategic engineering of reactive oxygen species (ROS)-generating, mitochondria-targeting CPUL1-TPP (CT) nanoaggregates. These self-assembling CT/GTB complexes were further optimized with DSPE-MPEG2k (DP) and Angiopep-2-conjugated DSPE-MPEG2k (Ang-DP) to create blood-brain barrier (BBB)-penetrating Ang-DP@CT/GTB nanoparticles, enhancing both physiological stability and low-density lipoprotein receptor-related protein 1 (LRP1)-mediated glioma targeting. Comparative analyses revealed that Ang-DP@CT/GTB nanoparticles significantly enhanced GTB's antiglioblastoma efficacy compared to free drug administration in both in vitro and in vivo models. Mechanistic investigations demonstrated that the nanosystem upregulates heme oxygenase-1 (HO-1), subsequently downregulating CDA expression to mitigate GTB metabolism. This coordinated molecular modulation prolongs GTB's therapeutic activity while leveraging the ROS-generating capacity of CT components for synergistic tumor suppression. The BBB-permeable codelivery platform exemplifies a rational design paradigm for multifunctional carrier-free pure nanodrugs (PNDs), demonstrating how clinical drug reformulation can overcome inherent pharmacokinetic limitations. This nanotechnology-driven approach provides critical insights for optimizing chemotherapeutic performance through metabolic pathway regulation and targeted delivery engineering.
Collapse
Affiliation(s)
- Rong Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Ke Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zhuolin Li
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Haoping Long
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Dongyu Zhang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yanting Li
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zhuolu Xia
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Xindong Guo
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Feng Cao
- Department of Pharmaceutical, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Feng Jiang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| |
Collapse
|
4
|
Refaat A, Thomas P, Zhang W, Esser L, Tong Z, Beer M, Mawdsley D, Thirkettle-Watts D, Shields KA, Nicolazzo JA, Voelcker NH. An In Vitro-In Vivo Comparative Study Using Highly Sensitive Radioisotopic Assays to Assess the Predictive Power of Emerging Blood-Brain Barrier Models. SMALL METHODS 2025; 9:e2401400. [PMID: 39663724 DOI: 10.1002/smtd.202401400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/07/2024] [Indexed: 12/13/2024]
Abstract
Microfluidic BBB-on-a-chip models (μBBB) aim to recapitulate the organotypic features of the human BBB with great potential to model CNS diseases and advance CNS therapeutics. Nevertheless, their predictive capacity for drug uptake into the brain remains uncertain due to limited evaluation with only a small number of model drugs. Here, the in vivo brain uptake of a panel of nine radiolabeled compounds is evaluated in Swiss-outbred mice following a single intravenously administered dose and compared against results from the microfluidic μBBB platform and the conventional Transwell BBB model. Radioisotopic measurements are employed to calculate brain-to-plasma concentration ratios (B/P) of the compounds both in vivo and in vitro. The in vitro-in vivo correlation plots of the B/P ratios revealed a strong positive correlation (r = 0.8081, R2 = 0.6530) for the μBBB, suggesting a high degree of predictive ability for drug permeability into the brain. In contrast, the Transwell assay showed a weaker in vitro-in vivo correlation (r = 0.6467, R2 = 0.4182). Finally, brain uptake of radiolabeled, brain-targeted, angiopep2-conjugated nanoparticles (ANG2-NP) is assessed in the μBBB and results mirrored the in vivo uptake, while the Transwell model failed to resolve the differences between the targeted and non-targeted NPs.
Collapse
Affiliation(s)
- Ahmed Refaat
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
| | - Patrick Thomas
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - Weisen Zhang
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organization, Research Way, Melbourne, VIC, 3168, Australia
| | - Lars Esser
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organization, Research Way, Melbourne, VIC, 3168, Australia
| | - Ziqiu Tong
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
| | - Michael Beer
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - David Mawdsley
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - David Thirkettle-Watts
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - Katherine A Shields
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - Joseph A Nicolazzo
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
| | - Nicolas H Voelcker
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Rd, Melbourne, VIC, 3168, Australia
- Department of Materials Science & Engineering, Faculty of Engineering, Monash University, 14 Alliance Ln, Melbourne, VIC, 3168, Australia
| |
Collapse
|
5
|
Tian Z, Fu Y, Dang Z, Guo T, Li W, Zhang J. Utilizing Nanomaterials in Microfluidic Devices for Disease Detection and Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:434. [PMID: 40137607 PMCID: PMC11946687 DOI: 10.3390/nano15060434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
Microfluidic technology has gained widespread application in the field of biomedical research due to its exceptional sensitivity and high specificity. Particularly when combined with nanomaterials, the synergy between the two has significantly advanced fields such as precision medicine, drug delivery, disease detection, and treatment. This article aims to provide an overview of the latest research achievements of microfluidic nanomaterials in disease detection and treatment. It delves into the applications of microfluidic nanomaterials in detecting blood parameters, cardiovascular disease markers, neurological disease markers, and tumor markers. Special emphasis is placed on their roles in disease treatment, including models such as blood vessels, the blood-brain barrier, lung chips, and tumors. The development of microfluidic nanomaterials in emerging medical technologies, particularly in skin interactive devices and medical imaging, is also introduced. Additionally, the challenges and future prospects of microfluidic nanomaterials in current clinical applications are discussed. In summary, microfluidic nanomaterials play an indispensable role in disease detection and treatment. With the continuous advancement of technology, their applications in the medical field will become even more profound and extensive.
Collapse
Affiliation(s)
- Zhibiao Tian
- College of Basic Medicine, Hebei University, Baoding 071000, China; (Z.T.); (Z.D.)
| | - Yatian Fu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China;
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
| | - Zhiyong Dang
- College of Basic Medicine, Hebei University, Baoding 071000, China; (Z.T.); (Z.D.)
| | - Tao Guo
- College of Basic Medicine, Hebei University, Baoding 071000, China; (Z.T.); (Z.D.)
- Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-Autoimmune Diseases in Hebei Province, Hebei University, Baoding 071000, China
| | - Wenjuan Li
- College of Basic Medicine, Hebei University, Baoding 071000, China; (Z.T.); (Z.D.)
- Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-Autoimmune Diseases in Hebei Province, Hebei University, Baoding 071000, China
| | - Jing Zhang
- College of Basic Medicine, Hebei University, Baoding 071000, China; (Z.T.); (Z.D.)
- Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-Autoimmune Diseases in Hebei Province, Hebei University, Baoding 071000, China
| |
Collapse
|
6
|
Mi Y, Jiang P, Luan J, Feng L, Zhang D, Gao X. Peptide‑based therapeutic strategies for glioma: Current state and prospects. Peptides 2025; 185:171354. [PMID: 39922284 DOI: 10.1016/j.peptides.2025.171354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Glioma is a prevalent form of primary malignant central nervous system tumor, characterized by its cellular invasiveness, rapid growth, and the presence of the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB). Current therapeutic approaches, such as chemotherapy and radiotherapy, have shown limited efficacy in achieving significant antitumor effects. Therefore, there is an urgent demand for new treatments. Therapeutic peptides represent an innovative class of pharmaceutical agents with lower immunogenicity and toxicity. They are easily modifiable via chemical means and possess deep tissue penetration capabilities which reduce side effects and drug resistance. These unique pharmacokinetic characteristics make peptides a rapidly growing class of new therapeutics that have demonstrated significant progress in glioma treatment. This review outlines the efforts and accomplishments in peptide-based therapeutic strategies for glioma. These therapeutic peptides can be classified into four types based on their anti-tumor function: tumor-homing peptides, inhibitor/antagonist peptides targeting cell surface receptors, interference peptides, and peptide vaccines. Furthermore, we briefly summarize the results from clinical trials of therapeutic peptides in glioma, which shows that peptide-based therapeutic strategies exhibit great potential as multifunctional players in glioma therapy.
Collapse
Affiliation(s)
- Yajing Mi
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Pengtao Jiang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Jing Luan
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lin Feng
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Dian Zhang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Xingchun Gao
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China.
| |
Collapse
|
7
|
Gawel AM, Betkowska A, Gajda E, Godlewska M, Gawel D. Current Non-Metal Nanoparticle-Based Therapeutic Approaches for Glioblastoma Treatment. Biomedicines 2024; 12:1822. [PMID: 39200286 PMCID: PMC11351974 DOI: 10.3390/biomedicines12081822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/24/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
The increase in the variety of nano-based tools offers new possibilities to approach the therapy of poorly treatable tumors, which includes glioblastoma multiforme (GBM; a primary brain tumor). The available nanocomplexes exhibit great potential as vehicles for the targeted delivery of anti-GBM compounds, including chemotherapeutics, nucleic acids, and inhibitors. The main advantages of nanoparticles (NPs) include improved drug stability, increased penetration of the blood-brain barrier, and better precision of tumor targeting. Importantly, alongside their drug-delivery ability, NPs may also present theranostic properties, including applications for targeted imaging or photothermal therapy of malignant brain cells. The available NPs can be classified into two categories according to their core, which can be metal or non-metal based. Among non-metal NPs, the most studied in regard to GBM treatment are exosomes, liposomes, cubosomes, polymeric NPs, micelles, dendrimers, nanogels, carbon nanotubes, and silica- and selenium-based NPs. They are characterized by satisfactory stability and biocompatibility, limited toxicity, and high accumulation in the targeted tumor tissue. Moreover, they can be easily functionalized for the improved delivery of their cargo to GBM cells. Therefore, the non-metal NPs discussed here, offer a promising approach to improving the treatment outcomes of aggressive GBM tumors.
Collapse
Affiliation(s)
- Agata M. Gawel
- Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland;
| | - Anna Betkowska
- Department of Cell Biology and Immunology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland; (A.B.); (E.G.); (M.G.)
| | - Ewa Gajda
- Department of Cell Biology and Immunology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland; (A.B.); (E.G.); (M.G.)
| | - Marlena Godlewska
- Department of Cell Biology and Immunology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland; (A.B.); (E.G.); (M.G.)
| | - Damian Gawel
- Department of Cell Biology and Immunology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland; (A.B.); (E.G.); (M.G.)
| |
Collapse
|
8
|
Susa F, Arpicco S, Pirri CF, Limongi T. An Overview on the Physiopathology of the Blood-Brain Barrier and the Lipid-Based Nanocarriers for Central Nervous System Delivery. Pharmaceutics 2024; 16:849. [PMID: 39065547 PMCID: PMC11279990 DOI: 10.3390/pharmaceutics16070849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The state of well-being and health of our body is regulated by the fine osmotic and biochemical balance established between the cells of the different tissues, organs, and systems. Specific districts of the human body are defined, kept in the correct state of functioning, and, therefore, protected from exogenous or endogenous insults of both mechanical, physical, and biological nature by the presence of different barrier systems. In addition to the placental barrier, which even acts as a linker between two different organisms, the mother and the fetus, all human body barriers, including the blood-brain barrier (BBB), blood-retinal barrier, blood-nerve barrier, blood-lymph barrier, and blood-cerebrospinal fluid barrier, operate to maintain the physiological homeostasis within tissues and organs. From a pharmaceutical point of view, the most challenging is undoubtedly the BBB, since its presence notably complicates the treatment of brain disorders. BBB action can impair the delivery of chemical drugs and biopharmaceuticals into the brain, reducing their therapeutic efficacy and/or increasing their unwanted bioaccumulation in the surrounding healthy tissues. Recent nanotechnological innovation provides advanced biomaterials and ad hoc customized engineering and functionalization methods able to assist in brain-targeted drug delivery. In this context, lipid nanocarriers, including both synthetic (liposomes, solid lipid nanoparticles, nanoemulsions, nanostructured lipid carriers, niosomes, proniosomes, and cubosomes) and cell-derived ones (extracellular vesicles and cell membrane-derived nanocarriers), are considered one of the most successful brain delivery systems due to their reasonable biocompatibility and ability to cross the BBB. This review aims to provide a complete and up-to-date point of view on the efficacy of the most varied lipid carriers, whether FDA-approved, involved in clinical trials, or used in in vitro or in vivo studies, for the treatment of inflammatory, cancerous, or infectious brain diseases.
Collapse
Affiliation(s)
- Francesca Susa
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| | - Candido Fabrizio Pirri
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Tania Limongi
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| |
Collapse
|
9
|
ter Linden E, Abels ER, van Solinge TS, Neefjes J, Broekman MLD. Overcoming Barriers in Glioblastoma-Advances in Drug Delivery Strategies. Cells 2024; 13:998. [PMID: 38920629 PMCID: PMC11201826 DOI: 10.3390/cells13120998] [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: 05/08/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
The world of cancer treatment is evolving rapidly and has improved the prospects of many cancer patients. Yet, there are still many cancers where treatment prospects have not (or hardly) improved. Glioblastoma is the most common malignant primary brain tumor, and even though it is sensitive to many chemotherapeutics when tested under laboratory conditions, its clinical prospects are still very poor. The blood-brain barrier (BBB) is considered at least partly responsible for the high failure rate of many promising treatment strategies. We describe the workings of the BBB during healthy conditions and within the glioblastoma environment. How the BBB acts as a barrier for therapeutic options is described as well as various approaches developed and tested for passing or opening the BBB, with the ultimate aim to allow access to brain tumors and improve patient perspectives.
Collapse
Affiliation(s)
- Esther ter Linden
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.t.L.); (E.R.A.)
| | - Erik R. Abels
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.t.L.); (E.R.A.)
| | - Thomas S. van Solinge
- Department of Neurosurgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Jacques Neefjes
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.t.L.); (E.R.A.)
| | - Marike L. D. Broekman
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.t.L.); (E.R.A.)
- Department of Neurosurgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Department of Neurosurgery, Haaglanden Medical Center, 2512 VA The Hague, The Netherlands
| |
Collapse
|