1
|
Zhang M, Xiang C, Niu R, He X, Luo W, Liu W, Gu R. Liposomes as versatile agents for the management of traumatic and nontraumatic central nervous system disorders: drug stability, targeting efficiency, and safety. Neural Regen Res 2025; 20:1883-1899. [PMID: 39254548 DOI: 10.4103/nrr.nrr-d-24-00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/28/2024] [Indexed: 09/11/2024] Open
Abstract
Various nanoparticle-based drug delivery systems for the treatment of neurological disorders have been widely studied. However, their inability to cross the blood-brain barrier hampers the clinical translation of these therapeutic strategies. Liposomes are nanoparticles composed of lipid bilayers, which can effectively encapsulate drugs and improve drug delivery across the blood-brain barrier and into brain tissue through their targeting and permeability. Therefore, they can potentially treat traumatic and nontraumatic central nervous system diseases. In this review, we outlined the common properties and preparation methods of liposomes, including thin-film hydration, reverse-phase evaporation, solvent injection techniques, detergent removal methods, and microfluidics techniques. Afterwards, we comprehensively discussed the current applications of liposomes in central nervous system diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, traumatic brain injury, spinal cord injury, and brain tumors. Most studies related to liposomes are still in the laboratory stage and have not yet entered clinical trials. Additionally, their application as drug delivery systems in clinical practice faces challenges such as drug stability, targeting efficiency, and safety. Therefore, we proposed development strategies related to liposomes to further promote their development in neurological disease research.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | | | | | | | | | | | | |
Collapse
|
2
|
Yang Z, Shi X, Qiu L. Tunable supramolecular self-assemblies based on cyclodextrin polymer as a loading platform for water-soluble drugs. Carbohydr Polym 2025; 347:122743. [PMID: 39486972 DOI: 10.1016/j.carbpol.2024.122743] [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: 07/12/2024] [Revised: 08/31/2024] [Accepted: 09/10/2024] [Indexed: 11/04/2024]
Abstract
Drug loading capacity is a crucial character of nano-scaled drug carriers to achieve high quality pharmaceutical preparations. However, efficient encapsulation of water-soluble small molecular drugs still faces large obstacles in many cases. Herein, we designed a novel supramolecular delivery system constructed by poly(β-cyclodextrin) containing benzoic acid groups (PCD-PA) and adamantyl terminated poly(ethylene glycol) (PEG-AD) to provide multiple intermolecular interactions for competent loading of water-soluble small-molecular drugs. PCD-PA had multiple host molecules, and PEG-AD could be inserted via host-guest interaction in different proportion to adjust the composition of supramolecular carrier. Meanwhile, π-π stacking and electrostatic interaction furnished by benzoic acid groups served as binding force for drug entrapment, which led to considerable loading capacity for several water-soluble drugs. Among the drugs with different chemical structures, mitoxantrone hydrochloride and doxorubicin hydrochloride bearing anthraquinone rings and several protonable amino groups acquired the highest loading content as about 14 % in PCD-PA3/PEG-AD supramolecular self-assemblies. Further computational simulations investigated the mechanism of drug loading based on the interactions between the carrier materials and the payloads. In addition, the weakly acidic environment obviously accelerated the release of certain drugs. All in all, this self-assembled supramolecular nano-system displayed great potentials as a delivery platform for diverse water-soluble drugs.
Collapse
Affiliation(s)
- Zhuting Yang
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Xuezhang Shi
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Liyan Qiu
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
3
|
Mochalova EN, Cherkasov VR, Sizikov AA, Litvinenko AV, Vorobeva TS, Norvillo NB, Gopanenko AV, Ivashchenko IA, Nikitin MP, Ivashchenko AA. Liposome-encapsulated aprotinin biodistribution in mice: Side-by-side comparison with free drug formulation. Biochem Biophys Res Commun 2024; 734:150636. [PMID: 39250873 DOI: 10.1016/j.bbrc.2024.150636] [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: 05/14/2024] [Revised: 08/21/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024]
Abstract
Injuries of the respiratory system caused by viral infections (e.g., by influenza virus, respiratory syncytial virus, metapneumovirus, or coronavirus) can lead to long-term complications or even life-threatening conditions. The challenges of treatment of such diseases have become particularly pronounced during the recent pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). One promising drug is the anti-fibrinolytic and anti-inflammatory protease inhibitor aprotinin, which has demonstrated considerable inhibition of the replication of some viruses. Encapsulation of aprotinin in liposomes can significantly improve the effectiveness of the drug, however, the use of nanoparticles as carriers of aprotinin can radically change its biodistribution in the body. Here we show that the liposomal form of aprotinin accumulates more efficiently in the lungs, heart, and kidneys than the molecular form by side-by-side comparison of the ex vivo biodistribution of these two fluorescently labeled formulations in mice using bioimaging. In particular, we synthesized liposomes of different compositions and studied their accumulation in various organs and tissues. Direct comparison of the biodistributions of liposomal and free aprotinin showed that liposomes accumulated in the lungs 1.82 times more effectively, and in the heart and kidneys - 3.56 and 2.00 times, respectively. This suggests that the liposomal formulation exhibits a longer residence time in the target organ and, thus, has the potential for a longer therapeutic effect. The results reveal the great potential of the aprotinin-loaded liposomes for the treatment of respiratory system injuries and heart- and kidney-related complications of viral infections.
Collapse
Affiliation(s)
- Elizaveta N Mochalova
- Moscow Institute of Physics and Technology, 1А Kerchenskaya St, 117303, Moscow, Russia; Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340, Sirius, Krasnodar region, Russia; Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St, 119991, Moscow, Russia
| | - Vladimir R Cherkasov
- Moscow Institute of Physics and Technology, 1А Kerchenskaya St, 117303, Moscow, Russia; Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St, 119991, Moscow, Russia
| | - Artem A Sizikov
- Moscow Institute of Physics and Technology, 1А Kerchenskaya St, 117303, Moscow, Russia
| | | | - Tatiana S Vorobeva
- Moscow Institute of Physics and Technology, 1А Kerchenskaya St, 117303, Moscow, Russia
| | - Natalia B Norvillo
- Moscow Institute of Physics and Technology, 1А Kerchenskaya St, 117303, Moscow, Russia
| | - Alexander V Gopanenko
- Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340, Sirius, Krasnodar region, Russia
| | - Ilya A Ivashchenko
- Moscow Institute of Physics and Technology, 1А Kerchenskaya St, 117303, Moscow, Russia
| | - Maxim P Nikitin
- Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340, Sirius, Krasnodar region, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St, 117997, Moscow, Russia; Moscow Center for Advanced Studies, 20 Kulakova St, 123592, Moscow, Russia.
| | | |
Collapse
|
4
|
Costa F, Giorgini G, Minnelli C, Mobbili G, Guardiani C, Giacomello A, Galeazzi R. Membrane Composition Allows the Optimization of Berberine Encapsulation in Liposomes. Mol Pharm 2024; 21:5818-5826. [PMID: 39425686 DOI: 10.1021/acs.molpharmaceut.4c00830] [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: 10/21/2024]
Abstract
Berberine (BBR) is a natural molecule with noteworthy pharmacological properties, including the prevention of antibiotic resistance in Gram-negative bacteria. However, its oral bioavailability is poor, thus resulting in an impaired absorption and efficacy in humans. In combination with other drugs, liposomes have been shown to enhance the availability of the drug, representing a smart delivery system to target tissues and reduce negative side effects. To date, there is a lack of studies on BBR and liposomes that enable the rationalization and molecular-based design of such formulations for future use in humans. In this work, the encapsulation of BBR into liposomes is proposed to overcome current limitations using a combination of experimental and computational assays to rationalize the membrane composition of liposomes that maximizes BBR encapsulation. First, the encapsulation efficiency was measured for several membrane compositions, revealing that it is enhanced by cholesteryl hemisuccinate and, to a lesser extent, by cholesterol. The physical basis of the BBR encapsulation efficiency and permeability was clarified using molecular dynamics simulation: using the lipid composition, one can tune the capability of membranes to attract, i.e., to adsorb, the molecules onto their surface. Overall, these findings suggest a rational strategy to maximize the encapsulation efficiency of liposomes by using negatively charged lipids, thus representing the basis for designing delivery systems for BBR, useful to treat, e.g., antibiotic resistance.
Collapse
Affiliation(s)
- Flavio Costa
- Dipartimento di Ingegneria Meccanica e Aerospaziale, Sapienza Università di Roma, Via Eudossiana 18, 00184 Rome, Italy
| | - Giorgia Giorgini
- Department of Life and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, 60131 Ancona, Italy
| | - Cristina Minnelli
- Department of Life and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, 60131 Ancona, Italy
| | - Giovanna Mobbili
- Department of Life and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, 60131 Ancona, Italy
| | - Carlo Guardiani
- Dipartimento di Ingegneria Meccanica e Aerospaziale, Sapienza Università di Roma, Via Eudossiana 18, 00184 Rome, Italy
| | - Alberto Giacomello
- Dipartimento di Ingegneria Meccanica e Aerospaziale, Sapienza Università di Roma, Via Eudossiana 18, 00184 Rome, Italy
| | - Roberta Galeazzi
- Department of Life and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, 60131 Ancona, Italy
| |
Collapse
|
5
|
Setyawati DR, Azzahra K, Mardliyati E, Tarwadi, Maharani BY, Nurmeilis. Box-Behnken design assisted approach in optimizing lipid composition for cationic liposome formulation as gene carrier. Biochim Biophys Acta Gen Subj 2024; 1868:130705. [PMID: 39178921 DOI: 10.1016/j.bbagen.2024.130705] [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: 12/19/2023] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Cationic liposomes represent a promising non-viral carrier platform for gene delivery. The successful intracellular delivery of genes to the target cell is highly influenced by lipid compositions in the liposomal formulation. In the present study, a Box-Behnken design was applied to investigate the optimal lipid composition for the liposome-based transfection agent. METHODS The concentrations of DOTAP, DSPE-PEG, and cholesterol were set as independent factors. A total of 15 lipid compositions were generated and tested for specific responses, including particle size, encapsulation efficiency, cell viability, and cell transfection. The data were then analyzed to predict the optimal composition using response surface methodology (RSM). RESULTS The results for particle size, encapsulation efficiency, cell viability and fluorescence intensity ranged from 158.7 to 2064 nm, 48.19-95.72%, 81.50-122.67%, and 0.0-9.08, respectively. Compositions of liposome-based transfection agent without DOTAP, those without cholesterol, and those containing DSPE-PEG2000 with a molar ratio equal to or greater than that of cholesterol tended to exhibit low encapsulation efficiency. The ability of the liposome to complex DNA, as determined through electrophoresis gel retardation assay, showed that the composition without DOTAP produced DNA bands, indicating that the prepared liposomes had a less ability to complex DNA. The cytotoxicity test results indicated that all lipid compositions were considered non-toxic, as they exhibited >80% cell viability. The cell transfection assay demonstrated that the lipid composition containing a combination of DOTAP and cholesterol was able to transfect DNA into cells. According to response analysis, RSM predicted that the optimal lipid composition consisted of 2.75 μmol DOTAP and 0.91 μmol cholesterol, with a desirability value of 0.85. CONCLUSIONS Although the equation model is still acceptable for predicting the optimal lipid composition, further study is needed to obtain a model with higher desirability, such as by using more lipid compositions, increased replications, and different variable responses.
Collapse
Affiliation(s)
- Damai Ria Setyawati
- Research Center for Vaccine and Drugs, National Research and Innovation Agency, Jakarta, Indonesia
| | - Khairunnisa Azzahra
- Department of Pharmacy, Faculty of Health and Sciences, Universitas Islam Negeri Syarif Hidayatullah, Jakarta, Indonesia; Nano Center Indonesia, South Tangerang 15314, Indonesia
| | - Etik Mardliyati
- Research Center for Vaccine and Drugs, National Research and Innovation Agency, Jakarta, Indonesia.
| | - Tarwadi
- Research Center for Vaccine and Drugs, National Research and Innovation Agency, Jakarta, Indonesia
| | - Bismi Yasinta Maharani
- Research Center for Vaccine and Drugs, National Research and Innovation Agency, Jakarta, Indonesia
| | - Nurmeilis
- Department of Pharmacy, Faculty of Health and Sciences, Universitas Islam Negeri Syarif Hidayatullah, Jakarta, Indonesia
| |
Collapse
|
6
|
Moon S, Jung M, Go S, Hong J, Sohn HS, Kim C, Kang M, Lee BJ, Kim J, Lim J, Kim BS. Engineered Nanoparticles for Enhanced Antitumoral Synergy Between Macrophages and T Cells in the Tumor Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410340. [PMID: 39252658 DOI: 10.1002/adma.202410340] [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: 07/17/2024] [Revised: 08/22/2024] [Indexed: 09/11/2024]
Abstract
T cells and macrophages have the potential to collaborate to eliminate tumor cells efficiently. Macrophages can eliminate tumor cells through phagocytosis and subsequently activate T cells by presenting tumor antigens. The activated T cells, in turn, can kill tumor cells and redirect tumor-associated macrophages toward an antitumoral M1 phenotype. However, checkpoint molecules expressed on tumor cells impede the collaborative action of these immune cells. Meanwhile, monotherapy with a single immune checkpoint inhibitor (ICI) for either macrophages or T cells yields suboptimal efficacy in cancer patients. To address this challenge, here a nanoparticle capable of efficiently delivering dual ICIs to tumors for both macrophages and T cells is developed. These programmed cell death protein 1 (PD-1)-transfected macrophage membrane-derived nanoparticles (PMMNPs) can target tumors and provide signal-regulatory protein alpha and PD-1 to block CD47 and programmed cell death-ligand 1 (PD-L1), respectively, on tumor cells. PMMNPs enhance macrophage-mediated cancer cell phagocytosis and antigen presentation, promote T cell activation, and induce the reprogramming of macrophages toward an antitumoral phenotype. In syngeneic tumor-bearing mice, PMMNPs demonstrate superior therapeutic efficacy compared to nanoparticles delivering single ICIs and non-targeted delivery of anti-CD47 and anti-PD-L1 antibodies. PMMNPs capable of augmenting the antitumoral interplay between macrophages and T cells may offer a promising avenue for cancer immunotherapy.
Collapse
Affiliation(s)
- Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee Su Sohn
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mikyung Kang
- School of Health and Environmental Science, Korea University, Seoul, 02841, Republic of Korea
| | - Byung Joon Lee
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jungwoo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinwoong Lim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Institute of Chemical Processes, and BioMAX, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
7
|
Luo B, Xuan S, Wang X, Ding K, Jin P, Zheng Y, Wu Z. Liposome/chitosan coating film bioplastic packaging for Litchi fruit preservation. Food Chem 2024; 464:141850. [PMID: 39489124 DOI: 10.1016/j.foodchem.2024.141850] [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: 06/23/2024] [Revised: 10/12/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Chitosan is an ideal coating film for food preservation, but the performance of a single chitosan coating film is not good. Herein, the liposome was prepared by embedding copper nanoparticles (CuNPs) and thyme essential oil (TEO) in the hydrophilic and hydrophobic double-domain structure formed by phospholipids, and combining with chitosan to obtain a chitosan-based coating film for litchi preservation. The liposome was well-dispersed and stable with an average particle size of about 190 nm. The liposome showed excellent controllable release properties, and the cumulative release rate of TEO was 65.17 % and that of CuNPs was 15.17 % after 7 days. Furthermore, the oxygen and water vapor barrier properties of the coating film were greatly improved. Importantly, the film possessed effective antioxidant, antibacterial activity and excellent safety, which presents a better fresh-keeping effect on litchi. This study provides insights into the design and manufacture of food packaging for controllable and long-lasting preservation.
Collapse
Affiliation(s)
- Bodan Luo
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Simin Xuan
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaotong Wang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Keying Ding
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Peng Jin
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yonghua Zheng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Zhengguo Wu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
8
|
Carney RP, Mizenko RR, Bozkurt BT, Lowe N, Henson T, Arizzi A, Wang A, Tan C, George SC. Harnessing extracellular vesicle heterogeneity for diagnostic and therapeutic applications. NATURE NANOTECHNOLOGY 2024:10.1038/s41565-024-01774-3. [PMID: 39468355 DOI: 10.1038/s41565-024-01774-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/11/2024] [Indexed: 10/30/2024]
Abstract
Extracellular vesicles (EVs) are diverse nanoparticles with large heterogeneity in size and molecular composition. Although this heterogeneity provides high diagnostic value for liquid biopsy and confers many exploitable functions for therapeutic applications in cancer detection, wound healing and neurodegenerative and cardiovascular diseases, it has also impeded their clinical translation-hence heterogeneity acts as a double-edged sword. Here we review the impact of subpopulation heterogeneity on EV function and identify key cornerstones for addressing heterogeneity in the context of modern analytical platforms with single-particle resolution. We outline concrete steps towards the identification of key active biomolecules that determine EV mechanisms of action across different EV subtypes. We describe how such knowledge could accelerate EV-based therapies and engineering approaches for mimetic artificial nanovesicle formulations. This approach blunts one edge of the sword, leaving only a single razor-sharp edge on which EV heterogeneity can be exploited for therapeutic applications across many diseases.
Collapse
Affiliation(s)
- Randy P Carney
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA.
| | - Rachel R Mizenko
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Batuhan T Bozkurt
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Neona Lowe
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Tanner Henson
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA, USA
| | - Alessandra Arizzi
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Aijun Wang
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA, USA
| | - Cheemeng Tan
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA.
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
9
|
Juang V, Gan J, Xia Z, Wang Y, Schwendeman A. Development and optimization of an in vitro release assay for evaluation of liposomal irinotecan formulation. Int J Pharm 2024; 667:124854. [PMID: 39442767 DOI: 10.1016/j.ijpharm.2024.124854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/13/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Onivyde®, a pegylated irinotecan liposomal formulation, is approved by the USFDA for treating metastatic pancreatic adenocarcinoma. Despite the substantial interest in developing its generic versions, the unique structural and manufacturing complexities of liposomal formulations pose challenges. In this study, we address this gap by developing a robust in vitro release test (IVRT) using dialysis membrane techniques. The release of Onivyde® is influenced by several key factors, including the composition of the release medium, temperature, initial formulation concentration, the materials and molecular weight cut-offs of dialysis bags, and the pH of the release medium. Our optimized IVRT for Onivyde® incorporates a release medium containing 5 mM ammonium bicarbonate in a HEPES solution with a pH of 7.4. Additionally, the method includes an initial formulation concentration of 4.6 µg/mL and 50 kDa dialysis bags, while maintaining a temperature of 37 °C with continuous agitation at 80 rpm. This optimized IVR assay effectively differentiates between varying qualities of irinotecan liposomal formulations. Our findings contribute to optimizing IVRT for liposomal irinotecan formulations, enabling better quality control procedures. This assay serves as a reliable tool for evaluating generic irinotecan liposomal formulations, aiding in their development and ensuring in vitro comparability.
Collapse
Affiliation(s)
- Vivian Juang
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jingyao Gan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Ziyun Xia
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Yan Wang
- Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
10
|
Wang Z, Kulkarni S, Nong J, Zamora M, Ebrahimimojarad A, Hood E, Shuvaeva T, Zaleski M, Gullipalli D, Wolfe E, Espy C, Arguiri E, Wang Y, Marcos-Contreras OA, Song W, Muzykantov VR, Fu J, Radhakrishnan R, Myerson JW, Brenner JS. A percolation-type criticality threshold controls immune protein coating of surfaces. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618530. [PMID: 39464129 PMCID: PMC11507815 DOI: 10.1101/2024.10.15.618530] [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
When a material enters the body, it is immediately attacked by hundreds of proteins, organized into complex networks of binding interactions and reactions. How do such complex systems interact with a material, "deciding" whether to attack? We focus on the "complement" system of ∼40 blood proteins that bind microbes, nanoparticles, and medical devices, initiating inflammation. We show a sharp threshold for complement activation upon varying a fundamental material parameter, the surface density of potential complement attachment points. This sharp threshold manifests at scales spanning single nanoparticles to macroscale pathologies, shown here for diverse engineered and living materials. Computational models show these behaviors arise from a minimal subnetwork of complement, manifesting percolation-type critical transitions in the complement response. This criticality switch explains the "decision" of a complex signaling network to interact with a material, and elucidates the evolution and engineering of materials interacting with the body.
Collapse
|
11
|
Iversen A, Utterström J, Khare LP, Aili D. Influence of lipid vesicle properties on the function of conjugation dependent membrane active peptides. J Mater Chem B 2024; 12:10320-10331. [PMID: 39291919 PMCID: PMC11409839 DOI: 10.1039/d4tb01107d] [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: 05/21/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
Membrane active peptides (MAPs) can provide novel means to trigger the release of liposome encapsulated drugs to improve the efficacy of liposomal drug delivery systems. Design of MAP-based release strategies requires possibilities to carefully tailor the interactions between the peptides and the lipid bilayer. Here we explore the influence of lipid vesicle properties on the function of conjugation-dependent MAPs, specifically focusing on two de novo designed peptides, JR2KC and CKV4. Utilizing liposomes with differences in size, lipid composition, and surface charge, we investigated the mechanisms and abilities of the peptides to induce controlled release of encapsulated cargo. Our findings indicate that liposome size modestly affects the structural changes and function of the peptides, with larger vesicles facilitating a minor increase in drug release efficiency due to higher peptide-to-liposome ratios. Notably, the introduction of negatively charged lipids significantly enhanced the release efficiency, predominantly through electrostatic interactions that favor peptide accumulation at the lipid bilayer interface and subsequent membrane disruption. The incorporation of cholesterol and a mix of saturated and unsaturated lipids was shown to alter the vesicle's phase behavior, thus modulating the membrane activity of the peptides. This was particularly evident in the cholesterol-enriched liposomes, where JR2KC induced lipid phase separation, markedly enhancing cargo release. Our results underscore the critical role of lipid vesicle composition in the design of MAP-based drug delivery systems, suggesting that precise tuning of lipid characteristics can significantly influence their performance.
Collapse
Affiliation(s)
- Alexandra Iversen
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry, and Biology, Linköping University, 581 83 Linköping, Sweden.
| | - Johanna Utterström
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry, and Biology, Linköping University, 581 83 Linköping, Sweden.
| | - Lalit Pramod Khare
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry, and Biology, Linköping University, 581 83 Linköping, Sweden.
| | - Daniel Aili
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry, and Biology, Linköping University, 581 83 Linköping, Sweden.
| |
Collapse
|
12
|
Zhao L, Li L, Zhang Y, He Z, Chen X, Liu Y, Shi B, Liu Y. Targeting Synovial Macrophages with Astaxanthin-Loaded Liposomes for Antioxidant Treatment of Osteoarthritis. ACS Biomater Sci Eng 2024. [PMID: 39413302 DOI: 10.1021/acsbiomaterials.4c00998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease highly associated with an imbalance in the network of inflammatory factors and typically characterized by oxidative stress and cartilage damage. Moreover, the specificity of the joint structure makes it difficult for drugs to achieve good penetration and effective enrichment in the joint cavity. Therefore, therapeutic strategies that increase the specific targeting of drugs to inflammatory joint and incorporate antioxidative stress effects are important to improve the efficacy of OA. Here, we developed a folic acid-modified liposomal nanoparticle (AST@Lip-FA) loaded with the antioxidant astaxanthin (AST) to enhance the water solubility and stability of AST and to target the delivery of AST to the site of OA, leading to a significant improvement in therapeutic efficacy. In vitro experiments demonstrated that, due to the recognition by FA of the receptor folate receptor β on the surface of activated macrophages, the cellular uptake efficiency of AST@Lip-FA was increased. Meanwhile, intracellularly overexpressed inflammatory mediators such as reactive oxygen species and nitric oxide were efficiently removed by AST@Lip-FA. In addition, in the ACLT-induced OA mouse model, AST@Lip-FA was precisely enriched in the inflamed joints and achieved long-term retention, fully utilizing the anti-inflammatory, antioxidant, and cartilage-protecting effects of AST to effectively alleviate the progression of OA. In summary, AST@Lip-FA has an important prospect as a potential and effective therapeutic strategy for OA.
Collapse
Affiliation(s)
- Linlin Zhao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Liangxiao Li
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Yingyu Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Ziye He
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xin Chen
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Yingying Liu
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Bin Shi
- Department of Traditional Chinese Medicine Orthopedics, Neck-Shoulder and Lumbocrural Pain Hospital Affiliated to Shandong First Medical University, Jinan 250014, Shandong, China
| | - Yajun Liu
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| |
Collapse
|
13
|
Rafati N, Zarepour A, Bigham A, Khosravi A, Naderi-Manesh H, Iravani S, Zarrabi A. Nanosystems for targeted drug Delivery: Innovations and challenges in overcoming the Blood-Brain barrier for neurodegenerative disease and cancer therapy. Int J Pharm 2024; 666:124800. [PMID: 39374818 DOI: 10.1016/j.ijpharm.2024.124800] [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: 07/18/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
The evolution of sophisticated nanosystems has revolutionized biomedicine, notably in treating neurodegenerative diseases and cancer. These systems show potential in delivering medication precisely to affected tissues, improving treatment effectiveness while minimizing side effects. Nevertheless, a major hurdle in targeted drug delivery is breaching the blood-brain barrier (BBB), a selective shield separating the bloodstream from the brain and spinal cord. The tight junctions between endothelial cells in brain capillaries create a formidable physical barrier, alongside efflux transporters that expel harmful molecules. This presents a notable challenge for brain drug delivery. Nanosystems present distinct advantages in overcoming BBB challenges, offering enhanced drug efficacy, reduced side effects, improved stability, and controlled release. Despite their promise, challenges persist, such as the BBB's regional variability hindering uniform drug distribution. Efflux transporters can also limit therapeutic agent efficacy, while nanosystem toxicity necessitates rigorous safety evaluations. Understanding the long-term impact of nanomaterials on the brain remains crucial. Additionally, addressing nanosystem scalability, cost-effectiveness, and safety profiles is vital for widespread clinical implementation. This review delves into the advancements and obstacles of advanced nanosystems in targeted drug delivery for neurodegenerative diseases and cancer therapy, with a focus on overcoming the BBB.
Collapse
Affiliation(s)
- Nesa Rafati
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Ashkan Bigham
- Institute of Polymers, Composites, and Biomaterials, National Research Council (IPCB-CNR), Naples 80125, Italy; Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Turkiye
| | - Hossein Naderi-Manesh
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran; Departments of Biophysics, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran.
| | - Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Ave, Isfahan, Iran.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan.
| |
Collapse
|
14
|
Greco G, Sarpietro MG. Liposome-Assisted Drug Delivery in the Treatment of Multiple Sclerosis. Molecules 2024; 29:4689. [PMID: 39407617 PMCID: PMC11477494 DOI: 10.3390/molecules29194689] [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: 07/08/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the nervous system that leads to neurological dysfunctions and severe disabilities. It is worth noting that conventional pharmacotherapy is poorly selective and causes toxicity problems and several systemic side effects. Thus, there is a need to develop new approaches to this medical challenge. The use of nanocarriers for drug delivery represents a good strategy to overcome several issues such as high therapeutic drug doses with side effects, such as diarrhea, nausea, and abdominal pain, and drug degradation processes; in addition, nanocarriers can provide controlled and targeted drug release. This review describes the application of liposomes for the delivery of pharmaceutical actives to target MS. Firstly, MS is explained. Then, liposomes are described along with their preparation, characterization, and stability. The literature about the use of liposomes for the treatment of MS is then analyzed.
Collapse
Affiliation(s)
- Giuliana Greco
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
- NANOMED-Research Center on Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95125 Catania, Italy
| | - Maria Grazia Sarpietro
- NANOMED-Research Center on Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95125 Catania, Italy
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| |
Collapse
|
15
|
Brugnera M, Vicario-de-la-Torre M, González-Cela Casamayor MA, López-Cano JJ, Bravo-Osuna I, Huete-Toral F, González Rubio ML, Carracedo G, Molina-Martínez IT, Andrés-Guerrero V, Herrero-Vanrell R. Enhancing the hypotensive effect of latanoprost by combining synthetic phosphatidylcholine liposomes with hyaluronic acid and osmoprotective agents. Drug Deliv Transl Res 2024; 14:2804-2822. [PMID: 38602615 PMCID: PMC11385046 DOI: 10.1007/s13346-024-01584-z] [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] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
The first line of glaucoma treatment focuses on reducing intraocular pressure (IOP) through the prescription of topical prostaglandin analogues, such as latanoprost (LAT). Topical ophthalmic medicines have low bioavailability due to their rapid elimination from the ocular surface. Nanotechnology offers innovative ways of enhancing the ocular bioavailability of antiglaucoma agents while reducing administration frequency. This study aims to combine LAT-loaded synthetic phosphatidylcholine liposomes with hyaluronic acid (0.2% w/v) and the osmoprotectants betaine (0.40% w/v) and leucine (0.90% w/v) (LAT-HA-LIP) to extend the hypotensive effect of LAT while protecting the ocular surface. LAT-HA-LIP was prepared as a mixture of 1,2-dioleoyl-sn-glycero-3-phosphocholine and 1,2-dimyristoyl-sn-glycero-3-phosphocholine, cholesterol and α-tocopherol acetate. LAT-HA-LIP exhibited high drug-loading capacity (104.52 ± 4.10%), unimodal vesicle sizes (195.14 ± 14.34 nm) and a zeta potential of -13.96 ± 0.78 mV. LAT-HA-LIP was isotonic (284.00 ± 1.41 mOsm L-1), had neutral pH (7.63 ± 0.01) and had suitable surface tension (44.07 ± 2.70 mN m-1) and viscosity (2.69 ± 0.15 mPa s-1) for topical ophthalmic administration. LAT-HA-LIP exhibited optimal in vitro tolerance in human corneal and conjunctival epithelial cells. No signs of ocular alteration or discomfort were observed when LAT-HA-LIP was instilled in albino male New Zealand rabbits. Hypotensive studies revealed that, after a single eye drop, the effect of LAT-HA-LIP lasted 24 h longer than that of a marketed formulation and that relative ocular bioavailability was almost three times higher (p < 0.001). These findings indicate the potential ocular protection and hypotensive effect LAT-HA-LIP offers in glaucoma treatment.
Collapse
Affiliation(s)
- Marco Brugnera
- Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, Complutense University of Madrid (UCM), Madrid, Spain
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, UCM; IdISSC, Madrid, Spain
- University Institute of Industrial Pharmacy (IUFI), Faculty of Pharmacy, UCM, Madrid, Spain
| | - Marta Vicario-de-la-Torre
- Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, Complutense University of Madrid (UCM), Madrid, Spain
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, UCM; IdISSC, Madrid, Spain
- University Institute of Industrial Pharmacy (IUFI), Faculty of Pharmacy, UCM, Madrid, Spain
| | - Miriam Ana González-Cela Casamayor
- Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, Complutense University of Madrid (UCM), Madrid, Spain
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, UCM; IdISSC, Madrid, Spain
| | - José Javier López-Cano
- Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, Complutense University of Madrid (UCM), Madrid, Spain
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, UCM; IdISSC, Madrid, Spain
| | - Irene Bravo-Osuna
- Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, Complutense University of Madrid (UCM), Madrid, Spain
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, UCM; IdISSC, Madrid, Spain
- University Institute of Industrial Pharmacy (IUFI), Faculty of Pharmacy, UCM, Madrid, Spain
| | - Fernando Huete-Toral
- Ocupharm Research Group, Department of Optometry and Vision, Faculty of Optics and Optometry, UCM, Madrid, Spain
| | - María Luisa González Rubio
- Ocupharm Research Group, Department of Optometry and Vision, Faculty of Optics and Optometry, UCM, Madrid, Spain
| | - Gonzalo Carracedo
- Ocupharm Research Group, Department of Optometry and Vision, Faculty of Optics and Optometry, UCM, Madrid, Spain
| | - Irene Teresa Molina-Martínez
- Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, Complutense University of Madrid (UCM), Madrid, Spain
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, UCM; IdISSC, Madrid, Spain
- University Institute of Industrial Pharmacy (IUFI), Faculty of Pharmacy, UCM, Madrid, Spain
| | - Vanessa Andrés-Guerrero
- Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, Complutense University of Madrid (UCM), Madrid, Spain.
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, UCM; IdISSC, Madrid, Spain.
- University Institute of Industrial Pharmacy (IUFI), Faculty of Pharmacy, UCM, Madrid, Spain.
| | - Rocío Herrero-Vanrell
- Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, Complutense University of Madrid (UCM), Madrid, Spain.
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, UCM; IdISSC, Madrid, Spain.
- University Institute of Industrial Pharmacy (IUFI), Faculty of Pharmacy, UCM, Madrid, Spain.
| |
Collapse
|
16
|
Jin X, Zhou Q, Cao L, Tie X, Ouyang H, Pan X, Diao J, Zhu Y, Li Y, Liu X, Zheng Y. Improved therapeutic index of the liposomal docetaxel-glutathione prepared by active click loading. Eur J Pharm Biopharm 2024; 203:114435. [PMID: 39103002 DOI: 10.1016/j.ejpb.2024.114435] [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: 03/17/2024] [Revised: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
The clinical usage of docetaxel (DTX) is severely hindered by the dose-limiting neutropenia and peripheral neurotoxicity of polysorbate 80-solubilized DTX injection, and there are no alternative formulations until now. In this study, we developed a new liposomal formulation of DTX to reduce its toxicities, accompanying with the greatly improved antitumor activity. The DTX was encapsulated into liposomes in the form of hydrophilic glutathione (GSH)-conjugated prodrugs using a click drug loading method, which achieved a high encapsulation efficiency (∼95 %) and loading capacity (∼30 % wt). The resulting liposomal DTX-GSH provided a sustained and efficient DTX release (∼50 % within 48 h) in plasma, resulting in a greatly improved antitumor activities as compared with that of polysorbate 80-solubilized DTX injection in the subcutaneous and orthotopic 4T1 breast tumor bearing mice. Even large tumors > 500 mm3 could be effectively inhibited and shrunk after the administration of liposomal DTX-GSH. More importantly, the liposomal DTX-GSH significantly decreased the neutropenia and peripheral neurotoxicity as compared with that of polysorbate 80-solubilized DTX injection at the equivalent dose. These data suggested that the liposomal DTX-GSH might become a superior alternative formulation to the commercial DTX injection.
Collapse
Affiliation(s)
- XueLi Jin
- Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Qing Zhou
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, China
| | - Lei Cao
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Xiaoru Tie
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Huihui Ouyang
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Xiao Pan
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Jing Diao
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Yuting Zhu
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Yang Li
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoxue Liu
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China.
| | - Yaxin Zheng
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China.
| |
Collapse
|
17
|
Zhang S, Zhang X, Ren Y, Huang L, Xu W, Wang H, Lu Q. Regorafenib enhances the efficacy of photodynamic therapy in hepatocellular carcinoma through MAPK signaling pathway suppression. Photodiagnosis Photodyn Ther 2024; 49:104319. [PMID: 39181490 DOI: 10.1016/j.pdpdt.2024.104319] [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: 06/22/2024] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Photodynamic therapy (PDT) is a promising and innovative approach for treating tumors. The synergistic effect of PDT and chemotherapy can enhance the anti-tumor efficacy by leveraging their complementing benefits. In this study, we created lipid vesicles to deliver a photosensitizer (chlorin e6, Ce6) and Regorafenib into tumors for the purpose of examining the effectiveness and mechanism of Lipo-Ce6@Rego-PDT (LCR-P) on Hepatocellular carcinoma (HCC) both in vitro and in vivo. We found that the cytotoxicity on HCC caused by LCR-P was significantly stronger than that caused by Lipo-Ce6-PDT (LC-P). Cellular ROS production in the LCR-P group was approximately higher than that in the LC-P group, and Regorafenib significantly inhibited the phosphorylation of JNK, ERK, and P38 of Lipo-Ce6-PDT group in vitro and in vivo. Furthermore, Regorafenib significantly downregulated the expression of Bcl-2 and upregulated the expression of Bax and cleaved caspase-3 of LC-P group in vitro and in vivo. Compared with LC-P, LCR-P significantly increased cell apoptosis rate. The body weight and HE staining of normal organs primarily indicated the safety of this combined strategy. These results indicate that the combination of Regorafenib and Lipo-Ce6 can significantly enhance the anti-tumor efficiency of PDT for HCC and exhibits good biosafety.
Collapse
Affiliation(s)
- Song Zhang
- Postdoctoral Research Station, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Xiao Zhang
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Yali Ren
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Lu Huang
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Weitian Xu
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Haiping Wang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China.
| | - Qiping Lu
- Postdoctoral Research Station, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China.
| |
Collapse
|
18
|
Xu F, Ni Q, Gong N, Xia B, Zhang J, Guo W, Hu Z, Li J, Liang XJ. Delivery Systems Developed for Treatment Combinations to Improve Adoptive Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407525. [PMID: 39165065 DOI: 10.1002/adma.202407525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/26/2024] [Indexed: 08/22/2024]
Abstract
Adoptive cell therapy (ACT) has shown great success in the clinic for treating hematologic malignancies. However, solid tumor treatment with ACT monotherapy is still challenging, owing to insufficient expansion and rapid exhaustion of adoptive cells, tumor antigen downregulation/loss, and dense tumor extracellular matrix. Delivery strategies for combination cell therapy have great potential to overcome these hurdles. The delivery of vaccines, immune checkpoint inhibitors, cytokines, chemotherapeutics, and photothermal reagents in combination with adoptive cells, have been shown to improve the expansion/activation, decrease exhaustion, and promote the penetration of adoptive cells in solid tumors. Moreover, the delivery of nucleic acids to engineer immune cells directly in vivo holds promise to overcome many of the hurdles associated with the complex ex vivo cell engineering strategies. Here, these research advance, as well as the opportunities and challenges for integrating delivery technologies into cell therapy s are discussed, and the outlook for these emerging areas are criticlly analyzed.
Collapse
Affiliation(s)
- Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qiankun Ni
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Ningqiang Gong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Weisheng Guo
- College of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
19
|
Savitri CMA, Fauzia KA, Alfaray RI, Aftab H, Syam AF, Lubis M, Yamaoka Y, Miftahussurur M. Opportunities for Helicobacter pylori Eradication beyond Conventional Antibiotics. Microorganisms 2024; 12:1986. [PMID: 39458296 PMCID: PMC11509656 DOI: 10.3390/microorganisms12101986] [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: 07/31/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Helicobacter pylori (H. pylori) is a bacterium known to be associated with a significant risk of gastric cancer in addition to chronic gastritis, peptic ulcer, and MALT lymphoma. Although only a small percentage of patients infected with H. pylori develop gastric cancer, Gastric cancer causes more than 750,000 deaths worldwide, with 90% of cases being caused by H. pylori. The eradication of this bacterium rests on multiple drug regimens as guided by various consensus. However, the efficacy of empirical therapy is decreasing due to antimicrobial resistance. In addition, biofilm formation complicates eradication. As the search for new antibiotics lags behind the bacterium's ability to mutate, studies have been directed toward finding new anti-H. pylori agents while also optimizing current drug functions. Targeting biofilm, repurposing outer membrane vesicles that were initially a virulence factor of the bacteria, phage therapy, probiotics, and the construction of nanoparticles might be able to complement or even be alternatives for H. pylori treatment. This review aims to present reports on various compounds, either new or combined with current antibiotics, and their pathways to counteract H. pylori resistance.
Collapse
Affiliation(s)
- Camilia Metadea Aji Savitri
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Oita, Japan; (C.M.A.S.); (R.I.A.)
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
| | - Kartika Afrida Fauzia
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
- Research Centre for Preclinical and Clinical Medicine, National Research and Innovation Agency, Cibinong Science Center, Bogor 16915, Indonesia
| | - Ricky Indra Alfaray
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Oita, Japan; (C.M.A.S.); (R.I.A.)
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
| | - Hafeza Aftab
- Department of Gastroenterology, Dhaka Medical College and Hospital, Dhaka 1000, Bangladesh;
| | - Ari Fahrial Syam
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Masrul Lubis
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Oita, Japan; (C.M.A.S.); (R.I.A.)
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX 77030, USA
- Division of Genome-Wide Microbiology, Research Center for Global and Local Infectious Diseases (RCGLID), Oita University, Yufu 879-5593, Oita, Japan
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine—Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya 60286, Indonesia
| | - Muhammad Miftahussurur
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine—Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya 60286, Indonesia
| |
Collapse
|
20
|
Zhao M, Liu C, Liu Z, Zuo Y, Chen C, Shi S, Shi X, Xie Y, Yang H, Chen Y. Myocardium-targeted liposomal delivery of the antioxidant peptide 8P against doxorubicin-induced myocardial injury. Int J Pharm 2024; 663:124569. [PMID: 39127172 DOI: 10.1016/j.ijpharm.2024.124569] [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: 04/10/2024] [Revised: 07/27/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Doxorubicin (Dox) is a broad-spectrum antineoplastic chemotherapeutic agent used in clinical settings, yet it exhibits significant cardiotoxicity, which in severe cases can lead to heart failure. Research indicates that oxidative stress plays a pivotal role in Dox -induced cardiomyocyte injury. Therefore, the application of antioxidants represents an effective strategy to mitigate the cardiotoxic effects of doxorubicin. In preliminary studies, we isolated an antioxidative peptide, PHWWEYRR (8P). This study utilizes a PCM cardiomyocyte-targeting peptide-modified liposome as a carrier to deliver 8P into cardiomyocytes, aiming to prevent Dox-induced cardiac injury through its antioxidative mechanism. The results demonstrated that we prepared the 8P-loaded and PCM-targeting peptide-modified liposome (P-P-8P), which exhibited good dispersibility, encapsulation efficiency, drug loading capacity, and in vitro release, along with myocardial targeting capability. In vitro experiments showed that P-P-8P could prevent oxidative stress injury in H9C2 cells, protect mitochondrial functions, and inhibit cell apoptosis through a mitochondria-dependent pathway. In vivo experiments indicated that P-P-8P could prevent abnormalities in serum biochemical indicators, cardiac dysfunction, and myocardial pathological changes in mice. In conclusion, P-P-8P effectively delivers 8P to cardiomyocytes, offering protection against the cardiotoxic effects of Dox, and holds potential as a future preventative or therapeutic agent for drug-induced cardiomyopathy.
Collapse
Affiliation(s)
- Meijun Zhao
- Department of Clinical Pharmacy, Affiliated Hospital of Jilin Medical College, Jilin, Jilin 132013, PR China.
| | - Chang Liu
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Zhenye Liu
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Yuanyuan Zuo
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Chen Chen
- Affiliated Hospital of Yanbian University, Yanji, Jilin 133002, PR China
| | - Shuai Shi
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, PR China
| | - Xinlin Shi
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Yining Xie
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Huiying Yang
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Yutong Chen
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| |
Collapse
|
21
|
Zhang Z, Lin S, Yu X, Jing J, Zhang Y, Chen L, Han J, Meng Z, Chen J, Meng Q. HI-6-Loaded Vehicle of Liposomes Mediated by an Amphiphilic Pillar[5]arene against Paraoxon Poisoning. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50474-50483. [PMID: 39287334 DOI: 10.1021/acsami.4c11893] [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: 09/19/2024]
Abstract
Organophosphate (OP) intoxication has become a severe common health matter all over the world. For the treatment of acute OP poisoning, the effective intracerebral delivery of acetylcholinesterase reactivators is crucial. Here, an amphiphilic hydrazide-pillar[5]arene (HP5A-6C), which could be readily integrated into liposomal bilayers' zwitterionic disaturated phosphatidylcholine (DSPC), was synthesized. A T7 peptide-containing guest (G) was attached on the surface via a noncovalent interaction to make mixed liposomes a particularly appealing candidate for brain-targeting delivery. Such coassembly could remain stable at room temperature for up to 6 weeks, and safety evaluations initially verified its fine biological compatibility. The hydrophilic interiors of T7/HP5A-6C@DSPC could further load HI-6 with 89.70% encapsulation efficiency. Support for brain-targeting potency came from imaging results. Notably, intravenous injection of HI-6-loaded vesicles exhibited a remarkable therapeutic effect on paraoxon (POX)-poisoned mice, effectively alleviating seizures and brain damage and significantly increasing the improving survival rate to 60% over the course of 7 days.
Collapse
Affiliation(s)
- Ziliang Zhang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Shujie Lin
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Xiang Yu
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Jie Jing
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Yahan Zhang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Longming Chen
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Jiaqi Han
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Zhao Meng
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Junyi Chen
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
- Key Laboratory of Inorganic-Organic Hybrid Functional Material Chemistry, Ministry of Education, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China
| | - Qingbin Meng
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| |
Collapse
|
22
|
Ge K, Ren Y, Hong Z, Mao Z, Yao B, Ye K, Jia C. Microchip Based Isolation and Drug Delivery of Patient-Derived Extracellular Vesicles Against Their Homologous Tumor. Adv Healthc Mater 2024:e2401990. [PMID: 39221674 DOI: 10.1002/adhm.202401990] [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: 05/30/2024] [Revised: 07/19/2024] [Indexed: 09/04/2024]
Abstract
Extracellular vesicles (EVs) have demonstrated significant potential in drug delivery and anti-tumor therapy. Despite this promising strategy, challenges such as specific targeting, EVs purification persist. In this study, a personalized nanodrug delivery platform using patient-derived tumor EVs (PT-EVs) based on a microchip is presented. The microchip integrates multiple functions, including capture, enrichment, drug loading, and elution of PT-EVs. The isolation and drug-carrying procedures are completed within a 12 h timeframe, achieving a recovery rate of 65%, significantly surpassing the conventional ultracentrifuge (UC) method. Furthermore, PT-EVs derived from patient tumor models are first utilized as natural drug carriers, capitalizing on their inherent homing ability to precisely target homologous tumors. Lenvatinib and doxorubicin (DOX), two commonly utilized drugs in the clinical treatment of hepatocellular carcinoma (HCC), are loaded into PT-EVs and delivered to a matched in vitro tumor model that recapitulates original tumors for drug susceptibility testing. As is proven, PT-EVs exhibit robust tumor cell targeting and efficient receptor-mediated cellular uptake, and the efficacy of chemotherapeutic drugs is improved significantly. These results suggest that this platform could be a valuable tool for efficient isolation of PT-EVs and personalized drug customization, particularly when working with limited clinical samples, thus supporting personalized and precision medicine.
Collapse
Affiliation(s)
- Ke Ge
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital Affiliated to Medical School of Westlake University, Hangzhou, 310006, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Yongan Ren
- Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Zichen Hong
- Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Zhenjun Mao
- Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yao
- Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Kai Ye
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Changku Jia
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital Affiliated to Medical School of Westlake University, Hangzhou, 310006, China
| |
Collapse
|
23
|
Chen XA, Chuang CC, Chen CC, Lee CY, Chin CY, Young JJ, Bai MY, Chuang CC. Polyelectrolyte-coated liposomes microfluidically assembled in one-step for enhancing cell endocytosis and in-vivo immune responses. Colloids Surf B Biointerfaces 2024; 241:114030. [PMID: 38901267 DOI: 10.1016/j.colsurfb.2024.114030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/14/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024]
Abstract
To enhance the cellular uptake of liposomes, we prepared conventional liposomes with targeting molecules and surface-charged liposomes and evaluated their potential as nano-carriers and vaccine adjuvants by comparing their endocytosis efficiencies using immune cells. Surface-charged liposomes were synthesized via a one-step microfluidic method, which provided a novel, simple, fast, and highly reproducible method for preparing liposomes. Flow cytometry revealed that cationic polyelectrolyte-coated liposomes exhibited higher endocytosis efficiencies (of up to a factor of 100) in A774A.1 cells and JAWs II cells compared with uncoated liposomes or those coated with anionic polyelectrolytes. Positively charged liposomes exhibited some cytotoxicity at quaternary-chitosan coating concentrations higher than 6 mg/mL; however, significantly lower cytotoxicities (by a factor of almost ten) were obtained by protein mixing. Furthermore, BALB/c mice vaccinated with a mixture of Anthrax vaccine adsorbed (AVA) and quaternary chitosan-coated liposomes showed faster and stronger anti-PA IgG inductions compared to those vaccinated with AVA alone, with titers positively correlating with the amount of cationic liposome used. This finding clearly reveals that quaternary chitosan-coated liposomes act as both nano-carriers and vaccine adjuvants that significantly enhance in-vivo immune responses to vaccines with low immunogenicities.
Collapse
Affiliation(s)
- Xin-An Chen
- Biomedical Engineering Program, Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei City 10607, Taiwan, ROC
| | - Chuan-Chang Chuang
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC; Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei City 11490, Taiwan, ROC
| | - Cheng-Cheung Chen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City 11490, Taiwan, ROC
| | - Chia-Ying Lee
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Chia-Ying Chin
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Jenn-Jong Young
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC.
| | - Meng-Yi Bai
- Biomedical Engineering Program, Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei City 10607, Taiwan, ROC; Adjunct Appointment to the Department of Biomedical Engineering, National Defense Medical Center, Taipei City 11490, Taiwan, ROC.
| | - Chuan-Chung Chuang
- School of Dentistry and Graduate Institute of Dental Science, National Defense Medical Center, Taipei City 11490, Taiwan, ROC; Department of dentistry, Tri-Service General Hospital, Taipei City 11490, Taiwan, ROC.
| |
Collapse
|
24
|
Du Z, Sui D, Xin D, Tang X, Li M, Liu X, Deng Y, Song Y. Sialic acid-modified doxorubicin liposomes target tumor-related immune cells to relieve multiple inhibitions of CD8 + T cells. J Liposome Res 2024; 34:464-474. [PMID: 38196168 DOI: 10.1080/08982104.2023.2298901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/18/2023] [Indexed: 01/11/2024]
Abstract
In different types of cancer treatments, cancer-specific T cells are required for effective anticancer immunity, which has a central role in cancer immunotherapy. However, due to the multiple inhibitions of CD8+ T cells by tumor-related immune cells, CD8+ T-cell mediated antitumor immunotherapy has not achieved breakthrough progress in the treatment of solid tumors. Receptors for sialic acid (SA) are highly expressed in tumor-associated immune cells, so SA-modified nanoparticles are a drug delivery nanoplatform using tumor-associated immune cells as vehicles. To relieve the multiple inhibitions of CD8+ T cells by tumor-associated immune cells, we prepared SA-modified doxorubicin liposomes (SL-DOX, Scheme 1A). In our study, free SA decreased the toxicity of SL-DOX to tumor-associated immune cells. Compared with common liposomes, SL-DOX could inhibit tumor growth more effectively. It is worth noting that SL-DOX could not only kill tumor-related neutrophils and monocytes to relieve the multiple inhibitions of CD8+ T cells but also induce immunogenic death of tumor cells to promote the infiltration and differentiation of CD8+ T cells (Scheme 1B). Therefore, SL-DOX has potential value for the clinical therapeutic effect of CD8+ T cells mediating anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Zhouchunxiao Du
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Dezhi Sui
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Dongzhe Xin
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xueying Tang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Mingze Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
25
|
Yang S, Raza F, Li K, Qiu Y, Su J, Qiu M. Maximizing arsenic trioxide's anticancer potential: Targeted nanocarriers for solid tumor therapy. Colloids Surf B Biointerfaces 2024; 241:114014. [PMID: 38850742 DOI: 10.1016/j.colsurfb.2024.114014] [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: 03/24/2024] [Revised: 05/18/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Arsenic trioxide (ATO) has gained significant attention due to its promising therapeutic effects in treating different diseases, particularly acute promyelocytic leukemia (APL). Its potent anticancer mechanisms have been extensively studied. Despite the great efficacy ATO shows in fighting cancers, drawbacks in the clinical use are obvious, especially for solid tumors, which include rapid renal clearance and short half-life, severe adverse effects, and high toxicity to normal cells. Recently, the emergence of nanomedicine offers a potential solution to these limitations. The enhanced biocompatibility, excellent targeting capability, and desirable effectiveness have attracted much interest. Therefore, we summarized various nanocarriers for targeted delivery of ATO to solid tumors. We also provided detailed anticancer mechanisms of ATO in treating cancers, its clinical trials and shortcomings as well as the combination therapy of ATO and other chemotherapeutic agents for reduced drug resistance and synergistic effects. Finally, the future study direction and prospects were also presented.
Collapse
Affiliation(s)
- Shiqi Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kunwei Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yujiao Qiu
- The Wharton School and School of Nursing, University of Pennsylvania, Philadelphia 19104, USA
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
26
|
Skrinda-Melne M, Locs J, Grava A, Dubnika A. Calcium phosphates enhanced with liposomes - the future of bone regeneration and drug delivery. J Liposome Res 2024; 34:507-522. [PMID: 37988074 DOI: 10.1080/08982104.2023.2285973] [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: 06/03/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Effective healing and regeneration of various bone defects is still a major challenge and concern in modern medicine. Calcium phosphates have emerged as extensively studied bone substitute materials due to their structural and chemical resemblance to the mineral phase of bone, along with their versatile properties. Calcium phosphates present promising biological characteristics that make them suitable for bone substitution, but a critical limitation lies in their low osteoinductivity. To supplement these materials with properties that promote bone regeneration, prevent infections, and cure bone diseases locally, calcium phosphates can be biologically and therapeutically modified. A promising approach involves combining calcium phosphates with drug-containing liposomes, renowned for their high biocompatibility and ability to provide controlled and sustained drug delivery. Surprisingly, there is a lack of research focused on liposome-calcium phosphate composites, where liposomes are dispersed within a calcium phosphate matrix. This raises the question of why such studies are limited. In order to provide a comprehensive overview of existing liposome and calcium phosphate composites as bioactive substance delivery systems, the authors review the literature exploring the interactions between calcium phosphates and liposomes. Additionally, it seeks to identify potential interactions between calcium ions and liposomes, which may impact the feasibility of developing liposome-containing calcium phosphate composite materials. Liposome capacity to protect bioactive compounds and facilitate localized treatment can be particularly valuable in scenarios involving bone regeneration, infection prevention, and the management of bone diseases. This review explores the implications of liposomes and calcium phosphate material containing liposomes on drug delivery, bioavailability, and stability, offering insights into their advantages.
Collapse
Affiliation(s)
- Marite Skrinda-Melne
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Janis Locs
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Andra Grava
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Arita Dubnika
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
27
|
Carnero Canales CS, Marquez Cazorla JI, Marquez Cazorla RM, Roque-Borda CA, Polinário G, Figueroa Banda RA, Sábio RM, Chorilli M, Santos HA, Pavan FR. Breaking barriers: The potential of nanosystems in antituberculosis therapy. Bioact Mater 2024; 39:106-134. [PMID: 38783925 PMCID: PMC11112550 DOI: 10.1016/j.bioactmat.2024.05.013] [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: 01/31/2024] [Revised: 04/17/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, continues to pose a significant threat to global health. The resilience of TB is amplified by a myriad of physical, biological, and biopharmaceutical barriers that challenge conventional therapeutic approaches. This review navigates the intricate landscape of TB treatment, from the stealth of latent infections and the strength of granuloma formations to the daunting specters of drug resistance and altered gene expression. Amidst these challenges, traditional therapies often fail, contending with inconsistent bioavailability, prolonged treatment regimens, and socioeconomic burdens. Nanoscale Drug Delivery Systems (NDDSs) emerge as a promising beacon, ready to overcome these barriers, offering better drug targeting and improved patient adherence. Through a critical approach, we evaluate a spectrum of nanosystems and their efficacy against MTB both in vitro and in vivo. This review advocates for the intensification of research in NDDSs, heralding their potential to reshape the contours of global TB treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Cesar Augusto Roque-Borda
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Giulia Polinário
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | | | - Rafael Miguel Sábio
- School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, the Netherlands
| | - Marlus Chorilli
- School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Hélder A. Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Fernando Rogério Pavan
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| |
Collapse
|
28
|
Nie S, Zhou J, Zheng X, Wei X, Zhang J, Shen X, Zhang W. CD133 ligand-enhanced etoposide-liposome complex for targeted killing of lung cancer cells. Biotechnol Prog 2024; 40:e3460. [PMID: 38558545 DOI: 10.1002/btpr.3460] [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: 01/12/2024] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
Lung cancer has a high incidence rate and a low cure rate, hence the urgent need for effective treatment methods. Current lung cancer drugs have several drawbacks, including low specificity, poor targeting, drug resistance, and irreversible damage to normal tissues. Therefore, there is a need to develop a safe and effective new drug that can target and kill tumor cells. In this study, we combined nanotechnology and biotechnology to develop a CD133 ligand-modified etoposide-liposome complex (Lipo@ETP-CD133) for targeted therapy of lung cancer. The CD133 ligand targeted lung cancer stem cells, causing the composite material to aggregate at the tumor site, where high levels of ETP liposomes could exert a strong tumor-killing effect. Our research results demonstrated that this nano-drug had efficient targeting and tumor-killing effects, indicating its potential for clinical application.
Collapse
Affiliation(s)
- Shiwei Nie
- Department of Thoracic Surgery, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang City, Henan Province, China
| | - Junzheng Zhou
- Department of Thoracic Surgery, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang City, Henan Province, China
| | - Xiaodong Zheng
- Department of Thoracic Surgery, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang City, Henan Province, China
| | - Xudong Wei
- Department of Thoracic Surgery, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang City, Henan Province, China
| | - Jinrui Zhang
- Supply House, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang City, Henan Province, China
| | - Xiaojuan Shen
- Department of Thoracic Surgery, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang City, Henan Province, China
| | - Weimin Zhang
- Department of Thoracic Surgery, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang City, Henan Province, China
| |
Collapse
|
29
|
Sia CS, Tey BT, Goh BH, Low LE. Controlled assembly of superparamagnetic iron oxide nanoparticle into nanoliposome for Pickering emulsion preparation. Colloids Surf B Biointerfaces 2024; 241:114051. [PMID: 38954935 DOI: 10.1016/j.colsurfb.2024.114051] [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: 03/13/2024] [Revised: 04/22/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
There has been a surge in effort in the development of various solid nanoparticles as Pickering emulsion stabilizers in the past decades. Regardless, the exploration of stabilizers that simultaneously stabilize and deliver bioactive has been limited. For this, liposomes with amphiphilic nature have been introduced as Pickering emulsion stabilizers but these nano-sized vesicles lack targeting specificity. Therefore in this study, superparamagnetic iron oxide nanoparticles (SPION) encapsulated within liposomes (MLP) were used as Pickering emulsion stabilizers to prepare pH and magnetic-responsive Pickering emulsions. A stable MLP-stabilized Pickering emulsion formulation was established by varying the MLP pH, concentration, and oil loading during the emulsification process. The primary stabilization mechanism of the emulsion under pH variation was identified to be largely associated with the MLP phosphate group deprotonation. When subjected to sequential pH adjustment to imitate the gastrointestinal digestion pH environment, a recovery in Pickering emulsion integrity was observed as the pH changes from acidic to alkaline. By incorporating SPION, the Pickering emulsion can be guided to the targeted site under the influence of a magnetic field without compromising emulsion stability. Overall, the results demonstrated the potential of MLP-stabilized Pickering emulsion as a dual pH- and magnetic-responsive drug delivery carrier with the ability to co-encapsulate hydrophobic and hydrophilic bioactive.
Collapse
Affiliation(s)
- Chin Siew Sia
- Department of Chemical Engineering, School of Engineering, Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia; Medical Engineering and Technology (MET) Hub, School of Engineering, Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia
| | - Beng Ti Tey
- Department of Chemical Engineering, School of Engineering, Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia
| | - Bey-Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Subang Jaya, Selangor, 47500, Malaysia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo 2007, NSW, Australia; Biofunctional Molecule Exploratory Research (BMEX) Group, School of Pharmacy, Monash University Malaysia, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Liang Ee Low
- Department of Chemical Engineering, School of Engineering, Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia; Medical Engineering and Technology (MET) Hub, School of Engineering, Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia; Monash-Industry Plant Oils Research Laboratory (MIPO), Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia.
| |
Collapse
|
30
|
Jiang K, Wang Q, Chen XL, Wang X, Gu X, Feng S, Wu J, Shang H, Ba X, Zhang Y, Tang K. Nanodelivery Optimization of IDO1 Inhibitors in Tumor Immunotherapy: Challenges and Strategies. Int J Nanomedicine 2024; 19:8847-8882. [PMID: 39220190 PMCID: PMC11366248 DOI: 10.2147/ijn.s458086] [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: 01/30/2024] [Accepted: 07/13/2024] [Indexed: 09/04/2024] Open
Abstract
Tryptophan (Trp) metabolism plays a vital role in cancer immunity. Indoleamine 2.3-dioxygenase 1 (IDO1), is a crucial enzyme in the metabolic pathway by which Trp is degraded to kynurenine (Kyn). IDO1-mediated Trp metabolites can inhibit tumor immunity and facilitate immune evasion by cancer cells; thus, targeting IDO1 is a potential tumor immunotherapy strategy. Recently, numerous IDO1 inhibitors have been introduced into clinical trials as immunotherapeutic agents for cancer treatment. However, drawbacks such as low oral bioavailability, slow onset of action, and high toxicity are associated with these drugs. With the continuous development of nanotechnology, medicine is gradually entering an era of precision healthcare. Nanodrugs carried by inorganic, lipid, and polymer nanoparticles (NPs) have shown great potential for tumor therapy, providing new ways to overcome tumor diversity and improve therapeutic efficacy. Compared to traditional drugs, nanomedicines offer numerous significant advantages, including a prolonged half-life, low toxicity, targeted delivery, and responsive release. Moreover, based on the physicochemical properties of these nanomaterials (eg, photothermal, ultrasonic response, and chemocatalytic properties), various combination therapeutic strategies have been developed to synergize the effects of IDO1 inhibitors and enhance their anticancer efficacy. This review is an overview of the mechanism by which the Trp-IDO1-Kyn pathway acts in tumor immune escape. The classification of IDO1 inhibitors, their clinical applications, and barriers for translational development are discussed, the use of IDO1 inhibitor-based nanodrug delivery systems as combination therapy strategies is summarized, and the issues faced in their clinical application are elucidated. We expect that this review will provide guidance for the development of IDO1 inhibitor-based nanoparticle nanomedicines that can overcome the limitations of current treatments, improve the efficacy of cancer immunotherapy, and lead to new breakthroughs in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Kehua Jiang
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Qing Wang
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Xiao-Long Chen
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Xiaodong Wang
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Xiaoya Gu
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Shuangshuang Feng
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yanlong Zhang
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
31
|
Wang C, Zhang Y, Kong W, Rong X, Zhong Z, Jiang L, Chen S, Li C, Zhang F, Jiang J. Delivery of miRNAs Using Nanoparticles for the Treatment of Osteosarcoma. Int J Nanomedicine 2024; 19:8641-8660. [PMID: 39188861 PMCID: PMC11346496 DOI: 10.2147/ijn.s471900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
Osteosarcoma is the predominant primary malignant bone tumor that poses a significant global health challenge. MicroRNAs (miRNAs) that regulate gene expression are associated with osteosarcoma pathogenesis. Thus, miRNAs are potential therapeutic targets for osteosarcoma. Nanoparticles, widely used for targeted drug delivery, facilitate miRNA-based osteosarcoma treatment. Numerous studies have focused on miRNA delivery using nanoparticles to inhibit the progress of osteosarcoma. Polymer-based, lipid-based, inorganic-based nanoparticles and extracellular vesicles were used to deliver miRNAs for the treatment of osteosarcoma. They can be modified to enhance drug loading and delivery capabilities. Also, miRNA delivery was combined with traditional therapies, for example chemotherapy, to treat osteosarcoma. Consequently, miRNA delivery offers promising therapeutic avenues for osteosarcoma, providing renewed hope for patients. This review emphasizes the studies utilizing nanoparticles for miRNA delivery in osteosarcoma treatment, then introduced and summarized the nanoparticles in detail. And it also discusses the prospects for clinical applications.
Collapse
Affiliation(s)
- Chengran Wang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Yihong Zhang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Weihui Kong
- Department of Stomatology, the First Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Xin’ao Rong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Ziming Zhong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Lei Jiang
- Department of Geriatric Medicine, Changchun Central Hospital, Changchun, Jilin Province, People’s Republic of China
| | - Shuhan Chen
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Chuang Li
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Fuqiang Zhang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Jinlan Jiang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| |
Collapse
|
32
|
Silli EK, Zheng Z, Zhou X, Li M, Tang J, Guo R, Tan C, Wang Y. Design optimization of Fucoidan-coating Cationic Liposomes for enhance Gemcitabine delivery. Invest New Drugs 2024:10.1007/s10637-024-01455-x. [PMID: 39154300 DOI: 10.1007/s10637-024-01455-x] [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: 04/01/2024] [Accepted: 06/26/2024] [Indexed: 08/19/2024]
Abstract
Obstacles facing chemotherapeutic drugs for cancers led scientists to load Gemcitabine (GEM) into nanocarriers like liposomes, known for their nontoxicity profile and targeting capacity. The liposomal nanostructures containing GEM were coated with Fucoidan (FU) due to its anti-tumor properties by targeting cancer cells. Thus four different cationic liposomes formulations were prepared by thin-film hydration method in optimal conditions: DOTAP (formulation A); DPPC/DOTAP (4:1 molar ratio, formulation B), DPPC/DMPC/DOTAP (4:1:1 molar ratio, formulation C) and DPPC/DMPC/DOTAP/DSPE-mPEG2000 (4:1:1:0.1 molar ratio, formulation D). They were studied to identify lipid-compositions offering effective GEM-entrapment and successful coating of FU on the liposome surface. Additional qualitative characteristics, such as particle size, polydispersity index, zeta potential, stability and in vitro drug release were then evaluated. Formulation C gave the best GEM-entrapment efficiency (EE) but formed aggregates when coated with FU, giving non-homogenous large size particles then not suitable for effective delivery. It was the same situation with formulation A and B. Only the formulation D showed a good GEM-EE (> 80%) and affinity by successful coating FU from three different algae species. The PEGylated formulation D coated of FU, with regard to storage stability and drug release studies, revealed to be a promising approach on design of optimal drug delivery system.
Collapse
Affiliation(s)
- Epiphane K Silli
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | | | - Xintao Zhou
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Mengfei Li
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jiali Tang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ruizhe Guo
- School of Chinese Medicine Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ying Wang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
33
|
Zhao F, Fan M, Jing Z, Zhang Y, Wang Y, Zhou C, Liu Y, Aitken RJ, Xia X. Engineered nanoparticles potentials in male reproduction. Andrology 2024. [PMID: 39120563 DOI: 10.1111/andr.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/06/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND The escalating prevalence of fertility problems in the aging population necessitates a comprehensive exploration of contributing factors, extending beyond environmental concerns, work-related stress, and unhealthy lifestyles. Among these, the rising incidence of testicular disorders emerges as a pivotal determinant of fertility issues. Current treatment challenges are underscored by the limitations of high-dose and frequent drug administration, coupled with substantial side effects and irreversible trauma inflicted by surgical interventions on testicular tissue. MATERIAL AND METHODS The formidable barrier posed by the blood-testis barrier compounds the complexities of treating testicular diseases, presenting a significant therapeutic obstacle. The advent of nanocarriers, with their distinctive attributes, holds promise in overcoming this impediment. These nanocarriers exhibit exceptional biocompatibility, and membrane penetration capabilities, and can strategically target the blood-testis barrier through surface ligand modification, thereby augmenting drug bioavailability and enhancing therapeutic efficacy. RESULTS AND DISCUSSION This review concentrates on the transformative potential of nanocarriers in the delivery of therapeutic agents to testicular tissue. By summarizing key applications, we illuminate the strides made in utilizing nanocarriers as a novel avenue to effectively treat testicular diseases. CONCLUSIONS Nanocarriers are critical in delivering therapeutic agents to testicular tissue.
Collapse
Affiliation(s)
- Feifei Zhao
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengyu Fan
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Zhiyang Jing
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yanxu Zhang
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yanlin Wang
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Congli Zhou
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yang Liu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Robert John Aitken
- School of Environmental and Life Sciences, College of Engineering, Science and Environmental Science, University of Newcastle, Callaghan, Australia
| | - Xue Xia
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, China
| |
Collapse
|
34
|
Mendanha D, Casanova MR, Gimondi S, Ferreira H, Neves NM. Microfluidic-Derived Docosahexaenoic Acid Liposomes for Targeting Glioblastoma and Its Inflammatory Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40543-40554. [PMID: 39042828 PMCID: PMC11310905 DOI: 10.1021/acsami.4c01368] [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: 04/17/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/25/2024]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor, characterized by limited treatment options and a poor prognosis. Its aggressiveness is attributed not only to the uncontrolled proliferation and invasion of tumor cells but also to the complex interplay between these cells and the surrounding microenvironment. Within the tumor microenvironment, an intricate network of immune cells, stromal cells, and various signaling molecules creates a pro-inflammatory milieu that supports tumor growth and progression. Docosahexaenoic acid (DHA), an essential ω3 polyunsaturated fatty acid for brain function, is associated with anti-inflammatory and anticarcinogenic properties. Therefore, in this work, DHA liposomes were synthesized using a microfluidic platform to target and reduce the inflammatory environment of GBM. The liposomes were rapidly taken up by macrophages in a time-dependent manner without causing cytotoxicity. Moreover, DHA liposomes successfully downregulated the expression of inflammatory-associated genes (IL-6; IL-1β; TNFα; NF-κB, and STAT-1) and the secretion of key cytokines (IL-6 and TNFα) in stimulated macrophages and GBM cells. Conversely, no significant differences were observed in the expression of IL-10, an anti-inflammatory gene expressed in alternatively activated macrophages. Additionally, DHA liposomes were found to be more efficient in regulating the inflammatory profile of these cells compared with a free formulation of DHA. The nanomedicine platform established in this work opens new opportunities for developing liposomes incorporating DHA to target GBM and its inflammatory milieu.
Collapse
Affiliation(s)
- Daniel Mendanha
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Marta R. Casanova
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Sara Gimondi
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Helena Ferreira
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Nuno M. Neves
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| |
Collapse
|
35
|
Gupta J, Sharma G. Nanogel: A versatile drug delivery system for the treatment of various diseases and their future perspective. Drug Deliv Transl Res 2024:10.1007/s13346-024-01684-w. [PMID: 39103593 DOI: 10.1007/s13346-024-01684-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
Nanogel (NG) drug delivery systems have emerged as promising tools for targeted and controlled drug release, revolutionizing treatment approaches across various diseases. Their unique physicochemical properties, such as nano size, high surface area, biocompatibility, stability, and tunable drug release, make them ideal carriers for a wide range of therapeutic agents. Nanogels (NGs), characterized by their 3D network of crosslinked polymers, offer unique edges like high drug loading capacity, controlled release, and targeted delivery. Additionally, the diverse applications of NGs in medical therapeutics highlight their versatility and potential impact on improving patient outcomes. Their application spans cancer treatment, infectious diseases, and chronic conditions, allowing for precise drug delivery to specific tissues or cells, minimizing side effects, and enhancing therapeutic efficacy. Despite their potential, challenges such as scalability, manufacturing reproducibility, and regulatory hurdles must be addressed. Achieving clinical translation requires overcoming these obstacles to ensure therapeutic payloads' safe and efficient delivery. Strategies such as surface modification and incorporating stimuli-responsive elements enhanced NG performance and addressed specific therapeutic challenges. Advances in nanotechnology, biomaterials, and targeted drug design offer opportunities to improve the performance of NGs and address current limitations. Tailoring NGs for exploring combination therapies and integrating diagnostics for real-time monitoring represent promising avenues for future research. In conclusion, NG drug delivery systems have demonstrated tremendous potential in diverse disease applications. Overcoming challenges and leveraging emerging technologies will pave the way for their widespread clinical implementation, ushering in a new era of precision medicine and improved patient care.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India.
| | - Gaurang Sharma
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India
| |
Collapse
|
36
|
de Roode KE, Hashemi K, Verdurmen WPR, Brock R. Tumor-On-A-Chip Models for Predicting In Vivo Nanoparticle Behavior. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402311. [PMID: 38700060 DOI: 10.1002/smll.202402311] [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: 03/23/2024] [Indexed: 05/05/2024]
Abstract
Nanosized drug formulations are broadly explored for the improvement of cancer therapy. Prediction of in vivo nanoparticle (NP) behavior, however, is challenging, given the complexity of the tumor and its microenvironment. Microfluidic tumor-on-a-chip models are gaining popularity for the in vitro testing of nanoparticle targeting under conditions that simulate the 3D tumor (microenvironment). In this review, following a description of the tumor microenvironment (TME), the state of the art regarding tumor-on-a-chip models for investigating nanoparticle delivery to solid tumors is summarized. The models are classified based on the degree of compartmentalization (single/multi-compartment) and cell composition (tumor only/tumor microenvironment). The physiological relevance of the models is critically evaluated. Overall, microfluidic tumor-on-a-chip models greatly improve the simulation of the TME in comparison to 2D tissue cultures and static 3D spheroid models and contribute to the understanding of nanoparticle behavior. Interestingly, two interrelated aspects have received little attention so far which are the presence and potential impact of a protein corona as well as nanoparticle uptake through phagocytosing cells. A better understanding of their relevance for the predictive capacity of tumor-on-a-chip systems and development of best practices will be a next step for the further refinement of advanced in vitro tumor models.
Collapse
Affiliation(s)
- Kim E de Roode
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Khadijeh Hashemi
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Roland Brock
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, 329, Bahrain
| |
Collapse
|
37
|
Safarbalou A, Abbasi A. Oral administration of liposome-encapsulated thymol could alleviate the inflammatory parameters in serum and hippocampus in a rat model of Alzheimer's disease. Exp Gerontol 2024; 193:112473. [PMID: 38801839 DOI: 10.1016/j.exger.2024.112473] [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: 04/17/2024] [Revised: 05/19/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Neuroinflammation is closely related to Alzheimer's Disease (AD) pathology, hence supplements with anti-inflammatory property could help attenuate the progression of AD. This study was conducted to evaluate the potential anti-inflammatory effects of liposome encapsulated thymol (LET), administered orally, in prevention of Alzheimer in a rat model by anti-inflammatory mechanisms. METHODS The rats were grouped into six groups (n = 10 animals per group), including Control healthy (Con), Alzheimer's disease (AD) model, AD model treated with free thymol in 40 and 80 mg/kg body weight (TH40 and TH80), AD model treated with LET in 40 and 80 mg/kg of body weight (LET40 and LET80). The behavioral response of step through latency (Passive Avoidance Test), concentrations of interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) and cyclooxygenase-2 (COX-2) and brain-derived neurotrophic factor (BDNF) were assessed in serum and hippocampus. RESULTS The results showed that significant increase in concentrations of IL-1β (P = 0.001), IL-6 (P = 0.001), TNF-α (P = 0.001) and COX-2 (P = 0.001) in AD group compared with healthy control rats. AD induction significantly reduced step through latency and revealed deficits in passive avoidance performance. The results also showed the treatment with free thymol especially in higher concentrations and also LTE could decrease serum concentrations of IL-1β (P < 0.05), IL-6 (P < 0.05), TNF-α (P < 0.05), and COX-2 (P < 0.05) and increase BDNF (P < 0.05) compared with control Alzheimer rats in hippocampus and serum. There were also significant correlations between serum and hippocampus concentrations of IL-1β (r2 = 0.369, P = 0.001), IL-6 (r2 = 0.386, P = 0.001), TNF-α (r2 = 0.412, P = 0.001), and COX-2 (r2 = 0.357, P = 0.001). It means a closed and positive relation between serum and hippocampus concentrations of IL-1β, IL-6, TNF-α, and COX-2. CONCLUSIONS LET demonstrates its ability to attenuate neuroinflammatory reaction in AD model through suppression of IL-1β, IL-6, and TNF-α and COX-2 indicators. Hence, it can ameliorate AD pathogenesis by declining inflammatory reaction.
Collapse
Affiliation(s)
- Asal Safarbalou
- Department of Biomedical Research, Institute for Intelligent Research, Tbilisi, Georgia
| | - Adeel Abbasi
- Department of Biomedical Research, Institute for Intelligent Research, Tbilisi, Georgia.
| |
Collapse
|
38
|
Zheng B, Wang L, Yi Y, Yin J, Liang A. Design strategies, advances and future perspectives of colon-targeted delivery systems for the treatment of inflammatory bowel disease. Asian J Pharm Sci 2024; 19:100943. [PMID: 39246510 PMCID: PMC11375318 DOI: 10.1016/j.ajps.2024.100943] [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: 03/31/2024] [Revised: 05/02/2024] [Accepted: 05/21/2024] [Indexed: 09/10/2024] Open
Abstract
Inflammatory bowel diseases (IBD) significantly contribute to high mortality globally and negatively affect patients' qualifications of life. The gastrointestinal tract has unique anatomical characteristics and physiological environment limitations. Moreover, certain natural or synthetic anti-inflammatory drugs are associated with poor targeting, low drug accumulation at the lesion site, and other side effects, hindering them from exerting their therapeutic effects. Colon-targeted drug delivery systems represent attractive alternatives as novel carriers for IBD treatment. This review mainly discusses the treatment status of IBD, obstacles to drug delivery, design strategies of colon-targeted delivery systems, and perspectives on the existing complementary therapies. Moreover, based on recent reports, we summarized the therapeutic mechanism of colon-targeted drug delivery. Finally, we addressed the challenges and future directions to facilitate the exploitation of advanced nanomedicine for IBD therapy.
Collapse
Affiliation(s)
- Baoxin Zheng
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liping Wang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yan Yi
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jun Yin
- School of Traditional Chinese Material, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Aihua Liang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
39
|
Yueh PF, Chiang CS, Tsai IJ, Tseng YL, Chen HR, Lan KL, Hsu FT. A multifunctional PEGylated liposomal-encapsulated sunitinib enhancing autophagy, immunomodulation, and safety in renal cell carcinoma. J Nanobiotechnology 2024; 22:459. [PMID: 39085911 PMCID: PMC11293195 DOI: 10.1186/s12951-024-02664-5] [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: 01/26/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Sunitinib is a multikinase inhibitor used to treat patients with advanced renal cell carcinoma (RCC). However, sunitinib toxicity makes it a double-edged sword. Potent immune modulation by sunitinib extends to nuclear interactions. To address these issues, there is an urgent need for delivery vectors suitable for sunitinib treatment. METHODS We developed PEGylated liposomes as delivery vectors to precisely target sunitinib (lipo-sunitinib) to RCC tumors. Further investigations, including RNA sequencing (RNA-seq), were performed to evaluate transcriptomic changes in these pathways. DiI/DiR-labeled lipo-sunitinib was used for the biodistribution analysis. Flow cytometry and immunofluorescence (IF) were used to examine immune modulation in orthotopic RCC models. RESULTS The evaluation of results indicated that lipo-sunitinib precisely targeted the tumor site to induce autophagy and was readily taken up by RCC tumor cells. In addition, transcriptomic assays revealed that following lipo-sunitinib treatment, autophagy, antigen presentation, cytokine, and chemokine production pathways were upregulated, whereas the epithelial-mesenchymal transition (EMT) pathway was downregulated. In vivo data provided evidence supporting the inhibitory effect of lipo-sunitinib on RCC tumor progression and metastasis. Flow cytometry further demonstrated that liposunitinib increased the infiltration of effector T cells (Teffs) and conventional type 1 dendritic cells (cDC1s) into the tumor. Furthermore, systemic immune organs such as the tumor-draining lymph nodes, spleen, and bone marrow exhibited upregulated anticancer immunity following lipo-sunitinib treatment. CONCLUSION Our findings demonstrated that lipo-sunitinib is distributed at the RCC tumor site, concurrently inducing potent autophagy, elevating antigen presentation, activating cytokine and chemokine production pathways, and downregulating EMT in RCC cells. This comprehensive approach significantly enhanced tumor inhibition and promoted anticancer immune modulation.
Collapse
Affiliation(s)
- Po-Fu Yueh
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, 6th Floor, Shouren Building, No. 155, Section 2, Linong Street, Beitou District, Taipei, 112, Taiwan, ROC
| | - Chih-Sheng Chiang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan, ROC
| | - I-Jung Tsai
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan, ROC
| | | | - He-Ru Chen
- Taiwan Liposome Company, Ltd., Taipei, Taiwan, ROC
| | - Keng-Li Lan
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, 6th Floor, Shouren Building, No. 155, Section 2, Linong Street, Beitou District, Taipei, 112, Taiwan, ROC.
- Department of Heavy Ion and Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.
- Department of Heavy Particles & Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Fei-Ting Hsu
- Department of Biology Science and Technology, China Medical University, 7F, Research Building, No. 100, Jingmao 1st Rd., Beitun Dist., Taichung City, 406, Taiwan, ROC.
| |
Collapse
|
40
|
Chen R, Wang P, Xie J, Tang Z, Fu J, Ning Y, Zhong Q, Wang D, Lei M, Mai H, Li H, Shi Z, Wang J, Cheng H. A multifunctional injectable, self-healing, and adhesive hydrogel-based wound dressing stimulated diabetic wound healing with combined reactive oxygen species scavenging, hyperglycemia reducing, and bacteria-killing abilities. J Nanobiotechnology 2024; 22:444. [PMID: 39068417 PMCID: PMC11283728 DOI: 10.1186/s12951-024-02687-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
The proficient handling of diabetic wounds, a rising issue coinciding with the global escalation of diabetes cases, poses significant clinical difficulties. A range of biofunctional dressings have been engineered and produced to expedite the healing process of diabetic wounds. This study proposes a multifunctional hydrogel dressing for diabetic wound healing, which is composed of Polyvinyl Alcohol (PVA) and N1-(4-boronobenzyl)-N3-(4-boronophenyl)-N1, N1, N3, N3-teramethylpropane-1, 3-diaminium (TSPBA), and a dual-drug loaded Gelatin methacryloyl (GM) microgel. The GM microgel is loaded with sodium fusidate (SF) and nanoliposomes (LP) that contain metformin hydrochloride (MH). Notably, adhesive and self-healing properties the hydrogel enhance their therapeutic potential and ease of application. In vitro assessments indicate that SF-infused hydrogel can eliminate more than 98% of bacteria within 24 h and maintain a sustained release over 15 days. Additionally, MH incorporated within the hydrogel has demonstrated effective glucose level regulation for a duration exceeding 15 days. The hydrogel demonstrates a sustained ability to neutralize ROS throughout the entire healing process, predominantly by electron donation and sequestration. This multifunctional hydrogel dressing, which integrated biological functions of efficient bactericidal activity against both MSSA and MRSA strains, blood glucose modulation, and control of active oxygen levels, has successfully promoted the healing of diabetic wounds in rats in 14 days. The hydrogel dressing exhibited significant effectiveness in facilitating the healing process of diabetic wounds, highlighting its considerable promise for clinical translation.
Collapse
Affiliation(s)
- Rong Chen
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Pinkai Wang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Jiajun Xie
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Zinan Tang
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jinlang Fu
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanhong Ning
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiang Zhong
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ding Wang
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Mingyuan Lei
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huaming Mai
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hao Li
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhanjun Shi
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Jian Wang
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Hao Cheng
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
41
|
Su Q, Pan J, Wang C, Zhang M, Cui H, Zhao X. Curcumin and Baicalin Co-Loaded Nanoliposomes for Synergistic Treatment of Non-Small Cell Lung Cancer. Pharmaceutics 2024; 16:973. [PMID: 39204318 PMCID: PMC11359521 DOI: 10.3390/pharmaceutics16080973] [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: 06/04/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Currently, the treatment of patients with advanced non-small cell lung cancer (NSCLC) mainly relies on traditional chemotherapeutic drugs; however, most of them have limited therapeutic effects and high toxicity. Some natural products with good therapeutic efficacy and low toxicity and side effects are limited in clinical application due to their low solubility and bioavailability. In this study, a nanoliposome drug-carrying system (Lip-Cur/Ba) was developed for the co-delivery of curcumin (Cur) and baicalin (Ba) using the thin-film hydration method. In vitro experiments demonstrated that Lip-Cur/Ba had a strong killing effect on A549 cells, and the inhibitory effect of Lip-Cur/Ba on A549 cells was enhanced by 67.8% and 51.9% relative to that of the single-carrier system, which could reduce the use of a single-drug dose (Lip-Cur and Lip-Ba), delay the release rate of the drug and improve the bioavailability. In vivo experiments demonstrated the antitumor activity of Lip-Cur/Ba by intravitreal injection in BALB/c mice, and there were no obvious toxic side effects. This study provides a new idea for curcumin and baicalin to be used in the co-treatment of NSCLC by constructing a new vector.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiang Zhao
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.S.); (J.P.); (C.W.); (M.Z.); (H.C.)
| |
Collapse
|
42
|
Li Y, Sun Q, Hao L, Shan H, Jiang Z, Wang Y, Chen Z, Zhu W, Zhao S. Liposomes Loaded with 5-Fluorouracil Can Improve the Efficacy in Pathological Scars. Int J Nanomedicine 2024; 19:7353-7365. [PMID: 39050869 PMCID: PMC11268756 DOI: 10.2147/ijn.s466221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Pathological scars, such as hypertrophic scars and keloids, are characterized by the proliferation of fibroblasts and the deposition of collagen that often cause pruritus, pain, and disfigurement. Due to their high incidence and deformity, pathological scars have resulted in severe physical and psychological trauma for patients. Intralesional injection of 5-fluorouracil (5-Fu) is a recommended option for treating pathological scars. However, the efficacy of 5-Fu injection was limited and unstable due to limited drug penetration and short retention time. Methods Liposomes are promising carriers that have advantages, such as high biocompatibility, controlled release property, and enhanced clinical efficacy. Here, we constructed a transdermal 5-Fu-loaded liposome (5-Fu-Lip) to provide a more effective and safer modality to scar treatment. Results Compared to 5-Fu, 5-Fu-Lip showed superior ability in inhibiting primary keloid fibroblasts proliferation, migration, and collagen deposition, and also significantly inhibited human umbilical vein endothelial cells (HUVECs) proliferation and microvessel construction. In vivo experiments demonstrated that 5-Fu-Lip can significantly reduce the severity of hypertrophic scars in a rabbit ear wounding model. Discussion 5-Fu-Lip provides a promising strategy to improve drug efficacy, which has great potential in the treatment of pathological scars.
Collapse
Affiliation(s)
- Yixin Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Furong Laboratory (Precision Medicine), Changsha, 410008, People’s Republic of China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| | - Qi Sun
- Furong Laboratory (Precision Medicine), Changsha, 410008, People’s Republic of China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- School of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, People’s Republic of China
| | - Lingjia Hao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Xiangya School of Medicine, Central South University, Changsha, 410083, People’s Republic of China
| | - Han Shan
- Furong Laboratory (Precision Medicine), Changsha, 410008, People’s Republic of China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- School of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, People’s Republic of China
| | - Zixi Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Furong Laboratory (Precision Medicine), Changsha, 410008, People’s Republic of China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| | - Ying Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Furong Laboratory (Precision Medicine), Changsha, 410008, People’s Republic of China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| | - Zeyu Chen
- Furong Laboratory (Precision Medicine), Changsha, 410008, People’s Republic of China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- School of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, People’s Republic of China
| | - Wu Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Furong Laboratory (Precision Medicine), Changsha, 410008, People’s Republic of China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Furong Laboratory (Precision Medicine), Changsha, 410008, People’s Republic of China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| |
Collapse
|
43
|
Chen X, Wu Y, Jia R, Fang Y, Cao K, Yang X, Qu X, Xia H. Antioxidant Activity and the Therapeutic Effect of Sinomenine Hydrochloride-Loaded Liposomes-in-Hydrogel on Atopic Dermatitis. Int J Mol Sci 2024; 25:7676. [PMID: 39062919 PMCID: PMC11276951 DOI: 10.3390/ijms25147676] [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: 06/05/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Sinomenine hydrochloride is an excellent drug with anti-inflammatory, antioxidant, immune-regulatory, and other functions. Atopic dermatitis is an inherited allergic inflammation that causes itchiness, redness, and swelling in the affected area, which can have a significant impact on the life of the patient. There are many therapeutic methods for atopic dermatitis, and sinomenine with immunomodulatory activity might be effective in the treatment of atopic dermatitis. In this study, the atopic dermatitis model was established in experimental mice, and physical experiments were carried out on the mice. In the experiment, sinomenine hydrochloride liposomes-in-hydrogel as a new preparation was selected for delivery. In this case, liposomes were dispersed in the colloidal hydrogel on a mesoscopic scale and could provide specific transfer properties. The results showed that the sinomenine hydrochloride-loaded liposomes-in-hydrogel system could effectively inhibit atopic dermatitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hongmei Xia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (X.C.); (Y.W.); (R.J.); (Y.F.); (K.C.); (X.Y.); (X.Q.)
| |
Collapse
|
44
|
Zhao F, Wang J, Zhang Y, Hu J, Li C, Liu S, Li R, Du R. In vivo Fate of Targeted Drug Delivery Carriers. Int J Nanomedicine 2024; 19:6895-6929. [PMID: 39005963 PMCID: PMC11246094 DOI: 10.2147/ijn.s465959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
This review aimed to systematically investigate the intracellular and subcellular fate of various types of targeting carriers. Upon entering the body via intravenous injection or other routes, a targeting carrier that can deliver therapeutic agents initiates their journey. If administered intravenously, the carrier initially faces challenges presented by the blood circulation before reaching specific tissues and interacting with cells within the tissue. At the subcellular level, the car2rier undergoes processes, such as drug release, degradation, and metabolism, through specific pathways. While studies on the fate of 13 types of carriers have been relatively conclusive, these studies are incomplete and lack a comprehensive analysis. Furthermore, there are still carriers whose fate remains unclear, underscoring the need for continuous research. This study highlights the importance of comprehending the in vivo and intracellular fate of targeting carriers and provides valuable insights into the operational mechanisms of different carriers within the body. By doing so, researchers can effectively select appropriate carriers and enhance the successful clinical translation of new formulations.
Collapse
Affiliation(s)
- Fan Zhao
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Jitong Wang
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Yu Zhang
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Jinru Hu
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Chenyang Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Shuainan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Key Laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
- Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Ruixiang Li
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Ruofei Du
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| |
Collapse
|
45
|
Wang D, Li Q, Xiao C, Wang H, Dong S. Nanoparticles in Periodontitis Therapy: A Review of the Current Situation. Int J Nanomedicine 2024; 19:6857-6893. [PMID: 39005956 PMCID: PMC11246087 DOI: 10.2147/ijn.s465089] [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: 02/20/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Periodontitis is a disease of inflammation that affects the tissues supporting the periodontium. It is triggered by an immunological reaction of the gums to plaque, which leads to the destruction of periodontal attachment structures. Periodontitis is one of the most commonly recognized dental disorders in the world and a major factor in the loss of adult teeth. Scaling and root planing remain crucial for managing patients with persistent periodontitis. Nevertheless, exclusive reliance on mechanical interventions like periodontal surgery, extractions, and root planning is insufficient to halt the progression of periodontitis. In response to the problem of bacterial resistance, some researchers are committed to finding alternative therapies to antibiotics. In addition, some scholars focus on finding new materials to provide a powerful microenvironment for periodontal tissue regeneration and promote osteogenic repair. Nanoparticles possess distinct therapeutic qualities, including exceptional antibacterial, anti-inflammatory, and antioxidant properties, immunomodulatory capacities, and the promotion of bone regeneration ability, which made them can be used for the treatment of periodontitis. However, there are many problems that limit the clinical translation of nanoparticles, such as toxic accumulation in cells, poor correlation between in vitro and in vivo, and poor animal-to-human transmissibility. In this paper, we review the present researches on nanoparticles in periodontitis treatment from the perspective of three main categories: inorganic nanoparticles, organic nanoparticles, and nanocomposites (including nanofibers, hydrogels, and membranes). The aim of this review is to provide a comprehensive and recent update on nanoparticles-based therapies for periodontitis. The conclusion section summarizes the opportunities and challenges in the design and clinical translation of nanoparticles for the treatment of periodontitis.
Collapse
Affiliation(s)
- Di Wang
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Qiqi Li
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Hao Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
46
|
Mesut B, Al-Mohaya M, Gholap AD, Yeşilkaya E, Das U, Akhtar MS, Sah R, Khan S, Moin A, Faiyazuddin M. Demystifying the potential of lipid-based nanocarriers in targeting brain malignancies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03212-6. [PMID: 38963550 DOI: 10.1007/s00210-024-03212-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/02/2024] [Indexed: 07/05/2024]
Abstract
Drug targeting for brain malignancies is restricted due to the presence of the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB), which act as barriers between the blood and brain parenchyma. Certainly, the limited therapeutic options for brain malignancies have made notable progress with enhanced biological understanding and innovative approaches, such as targeted therapies and immunotherapies. These advancements significantly contribute to improving patient prognoses and represent a promising shift in the landscape of brain malignancy treatments. A more comprehensive understanding of the histology and pathogenesis of brain malignancies is urgently needed. Continued research focused on unraveling the intricacies of brain malignancy biology holds the key to developing innovative and tailored therapies that can improve patient outcomes. Lipid nanocarriers are highly effective drug delivery systems that significantly improve their solubility, bioavailability, and stability while also minimizing unwanted side effects. Surface-modified lipid nanocarriers (liposomes, niosomes, solid lipid nanoparticles, nanostructured lipid carriers, lipid nanocapsules, lipid-polymer hybrid nanocarriers, lipoproteins, and lipoplexes) are employed to improve BBB penetration and uptake through various mechanisms. This systematic review illuminates and covers various topics related to brain malignancies. It explores the different methods of drug delivery used in treating brain malignancies and delves into the benefits, limitations, and types of brain-targeted lipid-based nanocarriers. Additionally, this review discusses ongoing clinical trials and patents related to brain malignancy therapies and provides a glance into future perspectives for treating this condition.
Collapse
Affiliation(s)
- Burcu Mesut
- Pharmaceutical Technology Department, Faculty of Pharmacy, Istanbul University, Istanbul, 34216, Turkey
| | - Mazen Al-Mohaya
- Institute of Health Sciences, Istanbul University, Istanbul, 34216, Turkey
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, 401404, Maharashtra, India
| | - Eda Yeşilkaya
- Institute of Health Sciences, Istanbul University, Istanbul, 34216, Turkey
| | - Ushasi Das
- Pharmaceutical Technology Department, Jadavpur University, Kolkata, West Bengal, India
| | - Mohammad Shabib Akhtar
- Department of Clinical Pharmacy, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Ranjit Sah
- Department of Microbiology, Institute of Medicine, Tribhuvan University Teaching Hospital, Kathmandu, 44600, Nepal.
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, 411018, Maharashtra, India.
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, 411018, Maharashtra, India.
| | | | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Hail, 2440, Hail, Saudi Arabia
| | - Md Faiyazuddin
- School of Pharmacy, Al - Karim University, Katihar, 854106, Bihar, India.
- Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| |
Collapse
|
47
|
Wei Y, Lv J, Zhu S, Wang S, Su J, Xu C. Enzyme-responsive liposomes for controlled drug release. Drug Discov Today 2024; 29:104014. [PMID: 38705509 DOI: 10.1016/j.drudis.2024.104014] [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: 12/16/2023] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
Compared to other nanovectors, liposomes exhibit unique advantages, such as good biosafety and high drug-loading capacity. However, slow drug release from conventional liposomes makes most payloads unavailable, restricting the therapeutic efficacy. Therefore, in the last ∼20 years, enzyme-responsive liposomes have been extensively investigated, which liberate drugs under the stimulation of enzymes overexpressed at disease sites. In this review, we elaborate on the research progress on enzyme-responsive liposomes. The involved enzymes mainly include phospholipases, particularly phospholipase A2, matrix metalloproteinases, cathepsins, and esterases. These enzymes can cleave ester bonds or specific peptide sequences incorporated in the liposomes for controlled drug release by disrupting the primary structure of liposomes, detaching protective polyethylene glycol shells, or activating liposome-associated prodrugs. Despite decades of efforts, there are still a lack marketed products of enzyme-responsive liposomes. Therefore, more efforts should be made to improve the safety and effectiveness of enzyme-responsive liposomes and address the issues associated with production scale-up.
Collapse
Affiliation(s)
- Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Jiajing Lv
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Shiyu Zhu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Can Xu
- Department of Gastroenterology, Changhai Hospital, Shanghai 200433, China.
| |
Collapse
|
48
|
Xing H, Pan X, Hu Y, Yang Y, Zhao Z, Peng H, Wang J, Li S, Hu Y, Li G, Ma D. High molecular weight hyaluronic acid-liposome delivery system for efficient transdermal treatment of acute and chronic skin photodamage. Acta Biomater 2024; 182:171-187. [PMID: 38759743 DOI: 10.1016/j.actbio.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/21/2024] [Accepted: 05/12/2024] [Indexed: 05/19/2024]
Abstract
Photodamage is one of the most common causes of skin injury. High molecular weight hyaluronic acid (HHA) has shown immense potential in the treatment of skin photodamage by virtue of its anti-inflammatory, reparative, and antioxidative properties. However, due to its large molecular structure of HHA, HHA solution could only form a protective film on the skin surface in conventional application, failing to effectively penetrate the skin, which necessitates the development of new delivery strategies. Liposomes, with a structure similar to biological membranes, have garnered extensive attention as transdermal drug delivery carriers because of their advantages in permeability, dermal compatibility, and biosafety. Herein, we have developed a HHA-liposome transdermal system (HHL) by embedding HHA into the liposome structure using reverse evaporation, high-speed homogenization, and micro-jet techniques. The effective penetration and long-term residence of HHA in skin tissue were multidimensionally verified, and the kinetics of HHA in the skin were extensively studied. Moreover, it was demonstrated that HHL significantly strengthened the activity of human keratinocytes and effectively inhibits photo-induced cellular aging in vitro. Furthermore, a murine model of acute skin injury induced by laser ablation was established, where the transdermal system showed significant anti-inflammatory and immunosuppressive properties, promoting skin proliferation and scar repair, thereby demonstrating immense potential in accelerating skin wound healing. Meanwhile, HHL significantly ameliorated skin barrier dysfunction caused by simulated sunlight exposure, inhibited skin erythema, inflammatory responses, and oxidative stress, and promoted collagen expression in a chronic photodamage skin model. Therefore, this transdermal delivery system with biocompatibility represents a promising new strategy for the non-invasive application of HHA in skin photodamage, revealing the significant potential for clinical translation and broad application prospects. STATEMENT OF SIGNIFICANCE: The transdermal system utilizing hyaluronic acid-based liposomes enhances skin permeability and retains high molecular weight hyaluronic acid (HHL). In vitro experiments with human keratinocytes demonstrate significant skin repair effects of HHL and its effective inhibition of cellular aging. In an acute photodamage model, HHL exhibits stronger anti-inflammatory and immunosuppressive properties, promoting skin proliferation and scar repair. In a chronic photodamage model, HHL significantly improves skin barrier dysfunction, reduces oxidative stress induced by simulated sunlight, and enhances collagen expression.
Collapse
Affiliation(s)
- Hui Xing
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China; Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Xiangjun Pan
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China
| | - Yihan Hu
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China; Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yuhui Yang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Ziyi Zhao
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Huanqi Peng
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Jianjin Wang
- Honest Medical China Co., Ltd, Zhuhai, 519000, China
| | - Shanying Li
- Honest Medical China Co., Ltd, Zhuhai, 519000, China
| | - Yunfeng Hu
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China.
| | - Guowei Li
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China; Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China.
| | - Dong Ma
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
49
|
Tokura D, Konarita K, Suzuki M, Ogata K, Honda Y, Miura Y, Nishiyama N, Nomoto T. Active control of pharmacokinetics using light-responsive polymer-drug conjugates for boron neutron capture therapy. J Control Release 2024; 371:445-454. [PMID: 38844180 DOI: 10.1016/j.jconrel.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/03/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
In boron neutron capture therapy (BNCT), boron drugs should exhibit high intratumoral boron concentrations during neutron irradiation, while being cleared from the blood and normal organs. However, it is usually challenging to achieve such tumor accumulation and quick clearance simultaneously in a temporally controlled manner. Here, we developed a polymer-drug conjugate that can actively control the clearance of the drugs from the blood. This polymer-drug conjugate is based on a biocompatible polymer that passively accumulates in tumors. Its side chains were conjugated with the low-molecular-weight boron drugs, which are immediately excreted by the kidneys, via photolabile linkers. In a murine subcutaneous tumor model, the polymer-drug conjugate could accumulate in the tumor with the high boron concentration ratio of the tumor to the surrounding normal tissue (∼10) after intravenous injection while a considerable amount remained in the bloodstream as well. Photoirradiation to blood vessels through the skin surface cleaved the linker to release the boron drug in the blood, allowing for its rapid clearance from the bloodstream. Meanwhile, the boron concentration in the tumor which was not photoirradiated could be maintained high, permitting strong BNCT effects. In clinical BNCT, the dose of thermal neutrons to solid tumors is determined by the maximum radiation exposure to normal organs. Thus, our polymer-drug conjugate may enable us to increase the therapeutic radiation dose to tumors in such a practical situation.
Collapse
Affiliation(s)
- Daiki Tokura
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Kakeru Konarita
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Minoru Suzuki
- Division of Particle Radiation Oncology, Particle Radiation Oncology Research Center, Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2-1010 Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Keisuke Ogata
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Yuto Honda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Yutaka Miura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Takahiro Nomoto
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.
| |
Collapse
|
50
|
Duraloglu C, Baysal I, Yabanoglu-Ciftci S, Arica B. Nintedanib and miR-29b co-loaded lipoplexes in idiopathic pulmonary fibrosis: formulation, characterization, and in vitro evaluation. Drug Dev Ind Pharm 2024; 50:671-686. [PMID: 39099436 DOI: 10.1080/03639045.2024.2387166] [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: 03/21/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/06/2024]
Abstract
OBJECTIVE This study was aimed to develop a cationic lipoplex formulation loaded with Nintedanib and miR-29b (LP-NIN-miR) as an alternative approach in the combination therapy of idiopathic pulmonary dibrosis (IPF) by proving its additive anti-fibrotic therapeutic effects through in vitro lung fibrosis model. SIGNIFICANCE This is the first research article reported that the LP-NIN-MIR formulations in the treatment of IPF. METHODS To optimize cationic liposomes (LPs), quality by design (QbD) approach was carried out. Optimized blank LP formulation was prepared with DOTAP, CHOL, DOPE, and DSPE-mPEG 2000 at the molar ratio of 10:10:1:1. Nintedanib loaded LP (LPs-NIN) were produced by microfluidization method and were incubated with miR-29b at room temperature for 30 min to obtain LP-NIN-miR. To evaluate the cellular uptake of LP-NIN-miR, NIH/3T3 cells were treated with 20 ng.mL-1 transforming growth factor-β1 (TGF-β1) for 96 h to establish the in vitro IPF model and incubated with LP-NIN-miR for 48 h. RESULTS The hydrodynamic diameter, polydispersity index (PDI), and zeta potential of the LP-NIN-miR were 87.3 ± 0.9 nm, 0.184 ± 0.003, and +24 ± 1 mV, respectively. The encapsulation efficiencies of Nintedanib and miR-29b were 99.8% ± 0.08% and 99.7% ± 1.2%, respectively. The results of the cytotoxicity study conducted with NIH/3T3 cells indicated that LP-NIN-miR is a safe delivery system. CONCLUSIONS The outcome of the transfection study proved the additive anti-fibrotic therapeutic effect of LP-NIN-miR and suggested that lipoplexes are effective delivery systems for drug and nucleic acid to the NIH/3T3 cells in the treatment of IPF.
Collapse
Affiliation(s)
- Ceren Duraloglu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Ipek Baysal
- Vocational School of Health Services, Hacettepe University, Ankara, Turkey
| | | | - Betul Arica
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|