1
|
Ismail M, Liu J, Wang N, Zhang D, Qin C, Shi B, Zheng M. Advanced nanoparticle engineering for precision therapeutics of brain diseases. Biomaterials 2025; 318:123138. [PMID: 39914193 DOI: 10.1016/j.biomaterials.2025.123138] [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: 09/18/2024] [Revised: 12/31/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
Despite the increasing global prevalence of neurological disorders, the development of nanoparticle (NP) technologies for brain-targeted therapies confronts considerable challenges. One of the key obstacles in treating brain diseases is the blood-brain barrier (BBB), which restricts the penetration of NP-based therapies into the brain. To address this issue, NPs can be installed with specific ligands or bioengineered to boost their precision and efficacy in targeting brain-diseased cells by navigating across the BBB, ultimately improving patient treatment outcomes. At the outset of this review, we highlighted the critical role of ligand-functionalized or bioengineered NPs in treating brain diseases from a clinical perspective. We then identified the key obstacles and challenges NPs encounter during brain delivery, including immune clearance, capture by the reticuloendothelial system (RES), the BBB, and the complex post-BBB microenvironment. Following this, we overviewed the recent progress in NPs engineering, focusing on ligand-functionalization or bionic designs to enable active BBB transcytosis and targeted delivery to brain-diseased cells. Lastly, we summarized the critical challenges hindering clinical translation, including scalability issues and off-target effects, while outlining future opportunities for designing cutting-edge brain delivery technologies.
Collapse
Affiliation(s)
- Muhammad Ismail
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiayi Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Ningyang Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Dongya Zhang
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Changjiang Qin
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China.
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, 2109, Australia.
| | - Meng Zheng
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
2
|
Guo Z, Li Y, Xu L, Wang J, Lyu J, Yang G, Liu Y, Hui Y, Zhao CX. Design and optimization of bifunctional peptides for controlled core-shell nanocapsule formation. J Colloid Interface Sci 2025; 686:599-612. [PMID: 39914305 DOI: 10.1016/j.jcis.2025.01.250] [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: 10/28/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Nanocapsules with core-shell structures hold significant potential across diverse applications. Biomimetic templating offers a benign approach for synthesizing inorganic nanostructures using biomolecules, leveraging amino acid sequences from natural sources and combinatorial biology in a process known as biomineralization. This study investigates the design and functionality of bifunctional peptides for controlled interfacial biosilicification. Five bifunctional peptides were designed and compared for their surface activity, structural behavior, and biosilicification capability under benign conditions. AM1 and SurSi-G1 demonstrate rapid adsorption, lower interfacial tension, and higher surface activity. In contrast, SurSi and its variants show slower adsorption due to higher molecular charge, resulting in high interfacial tension. Biosilicification assays confirmed that peptide charge strongly influences particle morphology, with SurSi and SurSi-R3 yielding well-dispersed silica nanoparticles, while AM1, SurSi-R2, and SurSi-G1 formed larger aggregates. Low ionic strength and sufficient surface charge enhance electrostatic interaction between positively charged bifunctional peptides and negatively charged hydrolyzed silicic acid, facilitating controlled biosilicification at interface and enabling the precise formation of core-shell nanocapsules. These findings highlight the pivotal role of peptide sequence and charge distribution in determining surface activity and interfacial biosilicification, providing insights for optimizing nanocapsule synthesis.
Collapse
Affiliation(s)
- Zichao Guo
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Yang Li
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Letao Xu
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Jiaqi Wang
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Jitong Lyu
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Guangze Yang
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Yun Liu
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Yue Hui
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia.
| | - Chun-Xia Zhao
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia.
| |
Collapse
|
3
|
Robles-Fernández A, Jiménez-Boland D, Leon-Cecilla A, Villegas-Montoya M, Traverso JÁ, Cuadros MA, Martín-Rodríguez A, Lopez-Lopez MT, Bramini M, Moraila-Martínez CL, Sánchez-Moreno P. Tuning lipid nanocarrier mechanical properties to improve glioblastoma targeting and blood brain barrier penetration. NANOSCALE 2025. [PMID: 40293789 DOI: 10.1039/d5nr00984g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Nanocarrier lipid systems (NLSs) have emerged as versatile platforms for diagnostic and therapeutic applications, including drug delivery, gene therapy, and vaccine development. Recent advancements highlight their potential in targeting infectious diseases and treating pathological conditions like tumors, largely due to their ability to effectively encapsulate and deliver therapeutic agents. This study focuses on the synthesis and characterization of NLSs with varying lipid compositions to understand their physicochemical and mechanical properties, which are crucial for their performance in biomedical applications. NLSs were prepared using a solvent displacement method, resulting in formulations with different ratios of olive oil and stearic acid. These formulations were characterized to determine their size, polydispersity index, and surface charge. Dynamic Light Scattering and Nanoparticle Tracking Analysis revealed that the size of the NLSs increased with higher stearic acid content. The NLSs demonstrated stability across a range of pH levels and in cell culture medium. The biomolecular corona formation and its impact on surface charge were also evaluated, showing significant effects on NLS stability. Mechanical properties, including rigidity and deformability, were assessed using Atomic Force Microscopy and rheological tests. The study found that increasing stearic acid content enhanced NLS rigidity and adhesion strength, which is crucial for their behaviour in biological systems such as blood circulation, tumor targeting, and cellular uptake. Biological evaluations demonstrated that these mechanical properties significantly influenced bio-interactions. Softer NLSs with a pure olive oil core displayed enhanced translocation across an in vitro blood-brain barrier model, underscoring their potential for drug delivery to the brain. Conversely, glioblastoma cell uptake studies revealed that the more rigid NLSs were internalized more efficiently by U87-MG cells, suggesting a role for stiffness in cellular entry. These findings provide insights into optimizing NLSs for specific therapeutic applications, particularly in overcoming barriers like the blood-brain barrier and targeting cerebral diseases.
Collapse
Affiliation(s)
- Ana Robles-Fernández
- Universidad de Granada, Departamento de Biología Celular, E-18071 Granada, Spain.
- Universidad de Granada, Departamento de Física Aplicada, E-18071, Granada, Spain.
| | - Daniel Jiménez-Boland
- Universidad de Granada, Departamento de Biología Celular, E-18071 Granada, Spain.
- Universidad de Granada, Departamento de Física Aplicada, E-18071, Granada, Spain.
| | - Alberto Leon-Cecilla
- Universidad de Granada, Departamento de Física Aplicada, E-18071, Granada, Spain.
- Instituto de Investigación Biosanitaria Ibs.GRANADA, E-18014, Granada, Spain
| | - Martín Villegas-Montoya
- Universidad de Granada, Departamento de Física Aplicada, E-18071, Granada, Spain.
- Universidad Autónoma de Sinaloa, Facultad de Biología, 80040, Culiacán, Mexico
| | - José Ángel Traverso
- Universidad de Granada, Departamento de Biología Celular, E-18071 Granada, Spain.
| | - Miguel A Cuadros
- Universidad de Granada, Departamento de Biología Celular, E-18071 Granada, Spain.
| | | | - Modesto T Lopez-Lopez
- Universidad de Granada, Departamento de Física Aplicada, E-18071, Granada, Spain.
- Instituto de Investigación Biosanitaria Ibs.GRANADA, E-18014, Granada, Spain
| | - Mattia Bramini
- Universidad de Granada, Departamento de Biología Celular, E-18071 Granada, Spain.
| | | | - Paola Sánchez-Moreno
- Universidad de Granada, Departamento de Física Aplicada, E-18071, Granada, Spain.
| |
Collapse
|
4
|
Karmaker S, Rosales PD, Tirumuruhan B, Viravalli A, Boehnke N. More than a delivery system: the evolving role of lipid-based nanoparticles. NANOSCALE 2025. [PMID: 40293317 DOI: 10.1039/d4nr04508d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Lipid-based nanoparticles, including liposomes and lipid nanoparticles (LNPs), make up an important class of drug delivery systems. Their modularity enables encapsulation of a wide range of therapeutic cargoes, their ease of functionalization allows for incorporation of targeting motifs and anti-fouling coatings, and their scalability facilitates rapid translation to the clinic. While the discovery and early understanding of lipid-based nanoparticles is heavily rooted in biology, formulation development has largely focused on materials properties, such as how liposome and lipid nanoparticle composition can be altered to maximize drug loading, stability and circulation. To achieve targeted delivery and enable improved accumulation of therapeutics at target tissues or disease sites, emphasis is typically placed on the use of external modifications, such as peptide, protein, and polymer motifs. However, these approaches can increase the complexity of the nanocarrier and complicate scale up. In this review, we focus on how our understanding of lipid structure and function in biological contexts can be used to design intrinsically functional and targeted nanocarriers. We highlight formulation-based strategies, such as the incorporation of bioactive lipids, that have been used to modulate liposome and lipid nanoparticle properties and improve their functionality while retaining simple nanocarrier designs. We also highlight classes of naturally occurring lipids, their functions, and how they have been incorporated into lipid-based nanoparticles. We will additionally position these approaches into the historical context of both liposome and LNP development.
Collapse
Affiliation(s)
- Senjuti Karmaker
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Plinio D Rosales
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Barath Tirumuruhan
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Amartya Viravalli
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Natalie Boehnke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| |
Collapse
|
5
|
Wan H, Jiao Z, Li J, Dai X, Li J, Yan LT. Dynamic Interplay between Deformability and Activity in Cell Entry of Soft Active Nanoparticles. NANO LETTERS 2025; 25:6797-6802. [PMID: 40227871 DOI: 10.1021/acs.nanolett.5c01445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Deformability has been recognized as a prime important characteristic influencing cellular uptake. But little is known about whether it controls cell-nanoparticle interfaces driven out of equilibrium. Here, we report on soft elastic active nanoparticles whose deformability due to the rigidity regulates the nonequilibrium interaction and dynamics in their endocytosis process. Simulations demonstrate a definitely nonmonotonic feature for the dependence of uptake efficiency on nanoparticle rigidity, in striking contrast to their passive counterpart. There exists a minimum activity for certain cellular uptake, which turns to a larger rigidity for a more vertical orientation of the nanoparticle. We analyze these results by developing analytical theories that reveal the physical origin of various energetic contributions and dissipations governed by the dynamic interplay between nanoparticle deformability and activity. Altogether, the present findings provide new insights into the nonequilibrium physics at cellular interfaces and might be of immediate interest to designing soft systems for the desired biomedical applications.
Collapse
Affiliation(s)
- Haixiao Wan
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Zheng Jiao
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Jiaqi Li
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Xiaobin Dai
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Jianfeng Li
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Li-Tang Yan
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
6
|
Zhao Y, Tian M, Tong X, Yang X, Gan L, Yong T. Emerging strategies in lymph node-targeted nano-delivery systems for tumor immunotherapy. Essays Biochem 2025; 69:EBC20253008. [PMID: 40159756 DOI: 10.1042/ebc20253008] [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: 01/13/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
The emergence of immunotherapy has led to the clinical approval of several related drugs. However, their efficacy against solid tumors remains limited. As the hub of immune activation, lymph nodes (LNs) play a critical role in tumor immunotherapy by initiating and amplifying immune responses. Nevertheless, the intricate physiological structure and barriers within LNs, combined with the immunosuppressive microenvironment induced by tumor cells, significantly impede the therapeutic efficacy of immunotherapy. Engineered nanoparticles (NPs) have shown great potential in overcoming these challenges by facilitating targeted drug transport to LNs and directly or indirectly activating T cells. This review systematically examines the structural features of LNs, key factors influencing the targeting efficiency of NPs, and current strategies for remodeling the immunosuppressive microenvironment of LNs. Additionally, it discusses future opportunities for optimizing NPs to enhance tumor immunotherapy, addressing challenges in clinical translation and safety evaluation.
Collapse
Affiliation(s)
- Yaoli Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Muzi Tian
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xin Tong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
7
|
Zhou S, Lyu P, Huang Y, Man X, Xiao S. Nonmonotonous Translocation Dynamics of Highly Deformable Particles across Channels. ACS NANO 2025; 19:10807-10815. [PMID: 40073271 DOI: 10.1021/acsnano.4c11319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
The translocation dynamics of cells and particles through geometric constrictions are critical in biological and biomedical processes from splenic filtration to tumor metastasis. While particle stiffness plays a key role, its role in highly nonequilibrium states remains poorly understood. Here, we present a multiscale model to investigate the impact of particle stiffness on the translocation dynamics in microfluidic channels. We find that semielastic particles exhibit superior translocation capabilities compared to both softer and more rigid particles, with a nonmonotonic stiffness dependence observed for highly deformable particles. Additionally, we identify crossover behaviors in translocation time driven by variations in the flow rate, particle size, and particle-plate interactions. Excessive particle deformation significantly regulates these dynamics, with stiffness-induced shape transitions from pancake-like to ellipsoidal forms, controlling frictional forces at the particle-channel interface and the sieve plate. The balance between these forces explains the observed nonmonotonic translocation dynamics. Our work provides insights into the relationship between particle deformability and flow dynamics, highlighting the importance of elasticity in translocation behavior. These findings have implications for designing microfluidic devices for efficient separation and analysis of cells with varying elasticities, advancing applications in human health diagnostics.
Collapse
Affiliation(s)
- Shenrong Zhou
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Peihan Lyu
- School of Physics, Beihang University, Beijing 100191, China
| | - Yu Huang
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xingkun Man
- School of Physics, Beihang University, Beijing 100191, China
- Peng Huanwu Collaborative Center for Research and Education, Beihang University, Beijing 100191, China
| | - Shiyan Xiao
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
- State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
8
|
Larreina Vicente N, Srinivas M, Tagit O. Perfluorocarbon-Loaded Poly(lactide- co-glycolide) Nanoparticles from Core to Crust: Multifaceted Impact of Surfactant on Particle Ultrastructure, Stiffness, and Cell Uptake. ACS APPLIED POLYMER MATERIALS 2025; 7:2864-2878. [PMID: 40110246 PMCID: PMC11915196 DOI: 10.1021/acsapm.4c03360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Poly(lactide-co-glycolide) nanoparticles (PLGA NPs) loaded with Perfluoro-15-crown-5-ether (PFCE) have been developed for imaging applications. A slight modification of the formulation led to the formation of two distinct particle ultrastructures: multicore particles (MCPs) and core-shell particles (CSPs), where poly(vinyl alcohol) (PVA), a nonionic surfactant, and sodium cholate (NaCh), an anionic surfactant, were used, respectively. Despite their similar composition and colloidal characteristics, these particles have previously demonstrated significant differences in their in vivo distribution and clearance. We hypothesize that these differences are collectively driven by variations in their structural, chemical, and mechanical properties, which are investigated in this study. Nanomechanical characterizations of MCPs and CSPs by atomic force microscopy (AFM) revealed elastic modulus values of 54 and 270 MPa in water, respectively, indicating a better permeability and deformability of the multicore ultrastructure. The impact of the surfactant on the NP surface chemistry was evidenced by their protein corona, which was significantly greater in the CSPs. Additionally, an important amount of residual NaCh was found on the surface of CSPs, which formed strong interactions with bovine serum albumin (BSA), accounting for the difference in protein coronas and surface chemistry. Surprisingly, in vitro cell uptake studies showed a higher uptake of MCPs by RAW macrophages but a preference for CSPs by HeLa cells. We conclude that for this specific formulation and in this stiffness range, mechanical differences have a stronger impact in HeLa cells, while surface properties and chemical recognition play a more important role in uptake by macrophages. Overall, the extent to which a physical factor impacts cell uptake is highly dependent on the specific uptake mechanism. With this study, we provide an integrated perspective on the role of different surfactants in the particle formation process, their impact on particle ultrastructure, mechanical properties, and surface chemistry, and the overall effect on cell uptake in vitro.
Collapse
Affiliation(s)
- Naiara Larreina Vicente
- Cell Biology and Immunology (CBI), Wageningen University, De Elst 1, Wageningen 6708 WD, Netherlands
| | - Mangala Srinivas
- Cell Biology and Immunology (CBI), Wageningen University, De Elst 1, Wageningen 6708 WD, Netherlands
| | - Oya Tagit
- Group of Biointerfaces, Institute for Chemistry and Bioanalytics, FHNW University of Applied Sciences and Arts Northwestern Switzerland, Hofackerstrasse 30, Muttenz 4132, Switzerland
| |
Collapse
|
9
|
Lu C, Li C, Gu N, Yang F. Emerging Elastic Micro-Nano Materials for Diagnosis and Treatment of Thrombosis. RESEARCH (WASHINGTON, D.C.) 2025; 8:0614. [PMID: 40028043 PMCID: PMC11868703 DOI: 10.34133/research.0614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025]
Abstract
Thrombus is a blood clot that forms in a blood vessel at the point of flaking. Thrombosis is closely associated with cardiovascular diseases caused by different sources and factors. However, the current clinical methods of thrombus diagnosis and treatment still have problems with targeting, permeability, stability, and biosafety. Therefore, in recent years, based on the development of micro/nano technology, researchers have tried to develop some new strategies for the diagnosis and treatment of thrombosis. Due to the unique structural characteristics, the micro-nano materials in physiological environments show excellent transport and delivery properties such as better in vivo circulation, longer life span, better targeting ability, and controllable cellular internalization. Especially, elasticity and stiffness are inherent mechanical properties of some well-designed micro-nano materials, which can make them better adapted to the needs of thrombosis diagnosis and treatment. Herein, this review first introduces the thrombotic microenvironment to characterize the thrombus development process. Then, to fine-tune the pathological occurrence and development of thrombosis, the role of elastic micro-nano materials for thrombus diagnosis and treatment is summarized. The properties, preparation methods, and biological fate of these materials have been discussed in detail. Following, the applications of elastic micro-nano materials in biomedical imaging, drug delivery, and therapy of thrombosis are highlighted. Last, the shortcomings and future design strategies of elastic micro-nano materials in diagnosis and treatment of clinical thrombosis are discussed. This review will provide new ideas for the use of nanotechnology in clinical diagnosis and treatment of thrombus in the future.
Collapse
Affiliation(s)
- Chenxin Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, P. R. China
| | - Chunjian Li
- Department of Cardiology,
The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Ning Gu
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Institute of Clinical Medicine, Nanjing Drum Tower Hospital, Medical School,
Nanjing University, Nanjing 210093, P. R. China
| | - Fang Yang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
10
|
Kuang L, Wu L, Li Y. Extracellular vesicles in tumor immunity: mechanisms and novel insights. Mol Cancer 2025; 24:45. [PMID: 39953480 PMCID: PMC11829561 DOI: 10.1186/s12943-025-02233-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/17/2025] Open
Abstract
Extracellular vesicles (EVs), nanoscale vesicles secreted by cells, have attracted considerable attention in recent years due to their role in tumor immunomodulation. These vesicles facilitate intercellular communication by transporting proteins, nucleic acids, and other biologically active substances, and they exhibit a dual role in tumor development and immune evasion mechanisms. Specifically, EVs can assist tumor cells in evading immune surveillance and attack by impairing immune cell function or modulating immunosuppressive pathways, thereby promoting tumor progression and metastasis. Conversely, they can also transport and release immunomodulatory factors that stimulate the activation and regulation of the immune system, enhancing the body's capacity to combat malignant diseases. This dual functionality of EVs presents promising avenues and targets for tumor immunotherapy. By examining the biological characteristics of EVs and their influence on tumor immunity, novel therapeutic strategies can be developed to improve the efficacy and relevance of cancer treatment. This review delineates the complex role of EVs in tumor immunomodulation and explores their potential implications for cancer therapeutic approaches, aiming to establish a theoretical foundation and provide practical insights for the advancement of future EVs-based cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Liwen Kuang
- School of Medicine, Chongqing University, Chongqing, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Li
- School of Medicine, Chongqing University, Chongqing, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
11
|
Xu J, Adepoju S, Pandey S, Pérez Tetuán J, Williams M, Abdelmessih RG, Auguste DT, Hung FR. Effects of Lipid Headgroups on the Mechanical Properties and In Vitro Cellular Internalization of Liposomes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:2600-2618. [PMID: 39834158 PMCID: PMC11803717 DOI: 10.1021/acs.langmuir.4c04363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
We performed all-atom and coarse-grained simulations of lipid bilayer mixtures of the unsaturated lipid DOPC, with saturated lipids having the same 18-carbon acyl tails and different headgroups, to understand their mechanical properties. The secondary lipids were DSPG, DSPA, DSPS, DSPC, and DSPE. The DOPC:DSPG system with 65:35 molar ratio was the softest, with area compressibility modulus KA ∼ 22% smaller than the pure DOPC value. Raising the mole % of DOPC leads to increases in KA, yet at any given composition the KA trend is DSPG < DSPA < DSPS < DSPC < DSPE. Lipid-lipid interactions are weaker in DOPC:DSPG mixtures and stronger in DSPE systems. The head and phosphate groups of the secondary lipids DSPG, DSPA, and DSPS interact strongly with salt ions. Adding secondary lipids leads to DOPC having more ordered acyl tails relative to pure DOPC systems. No evidence of phase separation or inhomogeneities was observed in our simulations. We synthesized three liposomal formulations, L-DOPC (pure DOPC) and L-DOPC/DSPG and L-DOPC/DSPA, both with 15 mol % of secondary lipid. L-DOPC/DSPA had approximately 3- and 2-times higher in vitro internalization by normal epithelial (EpH4-Ev) and metastatic breast cancer (4T1) cells, compared with L-DOPC. The uptake of L-DOPC/DSPG by EpH4-Ev cells was almost 2-fold compared to L-DOPC, but both liposomes had similar uptakes by cancer cells. As L-DOPC/DSPG and L-DOPC/DSPA have similar KA values, we presumed that the mechanical properties, possibly in combination with the higher negative surface charges in L-DOPC/DSPA and differences in effective liposome diameters and diffusivities, contributed to these observations.
Collapse
Affiliation(s)
- Jiaming Xu
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Stephen Adepoju
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Simran Pandey
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Jimena Pérez Tetuán
- Department
of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Mary Williams
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Rudolf G. Abdelmessih
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Debra T. Auguste
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
- Department
of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Francisco R. Hung
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
12
|
Austria E, Bilek M, Varamini P, Akhavan B. Breaking biological barriers: Engineering polymeric nanoparticles for cancer therapy. NANO TODAY 2025; 60:102552. [DOI: 10.1016/j.nantod.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
13
|
Sun Q, Yang W, Song Z, Lu H, Shang W, Li H, Yang Z, Gao W, Li Y, Xu Y, Luo M, Liu K, Wu Q, Xuan Z, Shen W, Yang Y, Yin D. Precisely Controlling the Activation of an Iron-Locked Drug Generator in the Liver Sinusoid to Enhance Barrier Penetration and Reduction of Liver Fibrosis. J Am Chem Soc 2024; 146:33784-33803. [PMID: 39584725 DOI: 10.1021/jacs.4c11988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Complex physical barriers and the nanomaterial's clearance mechanism in the liver greatly hinder the feasibility of using a conventional liver-targeting nanoplatform to deliver antifibrotic drugs to pathological sites for the treatment of liver fibrosis. Here, a novel drug delivery strategy was designed to overcome drug penetration barriers in a fibrotic liver and cooperated with oral nattokinase (NKase)-mediated antifibrosis therapy as a proof of concept, which relies on the coadministration of a nanosized iron-locked drug generator (named Pro-HAase) and orally absorbed iron chelator deferasirox (DFX). Such a strategy starts from the rapid accumulation of intravenously injected Pro-HAase in the microcapillaries of the fibrotic liver followed by disrupting the polyphenol-iron coordination inside Pro-HAase by DFX, liberating antifibrotic components, including procyanidine (PA) and hyaluronidase (HAase). Attractively, absorption of DFX requires the sequential processes of traversing the intestinal mucosa and targeting the liver, which enable DFX to preferentially disassemble Pro-HAase accumulated in the liver sinusoid rather than in systemic circulation or other organs, thus avoiding the off-target activation of Pro-HAase and depletion of the normal iron pool. The in situ disassembly process decreases the sequestration of Pro-HAase by cells of the mononuclear phagocyte system and promotes gradient-driven permeation of therapeutic components to surrounding liver tissues within 2 h, accompanied by biliary excretion of the inactive iron-DFX complex. As a result, the cooperation of Pro-HAase and DFX not only allows NKase-mediated therapy to completely reverse liver fibrosis but also suppresses the chronic hepatotoxicity of residual liver iron after multiple doses of Pro-HAase. The high spatiotemporal precision, unique barrier-penetration mechanism, and self-detoxification ability of this strategy will inspire the rational design of analogous iron-locked nanosystems to improve the therapeutic outcomes of liver fibrosis or other liver diseases.
Collapse
Affiliation(s)
- Quanwei Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wenshuo Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Zhengwei Song
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Huiyu Lu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wencui Shang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Huihui Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Zexin Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wenheng Gao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Yunlong Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Yujing Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Min Luo
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Kang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Qinghua Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Zihua Xuan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230031, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM),, Hefei 230012, China
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230031, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM),, Hefei 230012, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
- Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei 230021, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM),, Hefei 230012, China
| |
Collapse
|
14
|
Bal T, Anjrini N, Zeroual M. Recent Advances and Challenges in Targeted Drug Delivery Using Biofunctional Coatings. MEDICAL APPLICATIONS FOR BIOCOMPATIBLE SURFACES AND COATINGS 2024:41-75. [DOI: 10.1039/9781837675555-00041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Globally, clinics are overwhelmed by drugs targeting undesired cells and organs, causing adverse systemic effects on the body. This shortfall in targeting specificity, safety, and efficiency has noticeably contributed to the failure of the bench-to-bedside transition. Activation or impairment of immune activity due to a misdirected drug and its carrier fuels complications, extending the range of destruction which can convert the course of disease into a life-threatening route. To address these great challenges, advanced coatings as indispensable components of future medicine have been investigated over the last few decades for precisely targeted drug delivery to achieve favorable prognoses in the treatment of a broad spectrum of diseases. Complemented by advancements in the pharmacological parameters, these systems hold great promise for the field. This chapter aims to discuss recent progress on new coatings for targeted drug delivery and the parameters for manufacturing these platforms for their cargo based on major determinants such as biocompatibility and bioactivity. A brief overview of the various applications of targeted drug delivery with functional coatings is also provided to offer a new perspective on the field.
Collapse
Affiliation(s)
- Tugba Bal
- aDepartment of Bioengineering, Graduate School of Sciences, Uskudar University, 34662, Istanbul, Turkiye
- bDepartment of Bioengineering, Faculty of Engineering and Natural Sciences, Uskudar University, 34662, Istanbul, Turkiye
| | - Nasma Anjrini
- aDepartment of Bioengineering, Graduate School of Sciences, Uskudar University, 34662, Istanbul, Turkiye
| | - Meryem Zeroual
- aDepartment of Bioengineering, Graduate School of Sciences, Uskudar University, 34662, Istanbul, Turkiye
| |
Collapse
|
15
|
Gao L, Dai X, Wu Y, Wang Y, Cheng L, Yan LT. Self-Assembly at Curved Biointerfaces. ACS NANO 2024; 18:30184-30210. [PMID: 39453716 DOI: 10.1021/acsnano.4c09675] [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: 10/27/2024]
Abstract
Most of the biological interfaces are curved. Understanding the organizational structures and interaction patterns at such curved biointerfaces is therefore crucial not only for deepening our comprehension of the principles that govern life processes but also for designing and developing targeted drugs aimed at diseased cells and tissues. Despite the considerable efforts dedicated to this area of research, our understanding of curved biological interfaces is still limited. Many aspects of these interfaces remain elusive, presenting both challenges and opportunities for further exploration. In this review, we summarize the structural characteristics of biological interfaces found in nature, the current research status of materials associated with curved biointerfaces, and the theoretical advancements achieved to date. Finally, we outline future trends and challenges in the theoretical and technological development of curved biointerfaces. By addressing these challenges, people could bridge the knowledge gap and unlock the full potential of curved biointerfaces for scientific and technological advancements, ultimately benefiting various fields and improving human health and well-being.
Collapse
Affiliation(s)
- Lijuan Gao
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Xiaobin Dai
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Yibo Wu
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Yuming Wang
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Linghe Cheng
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Li-Tang Yan
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
16
|
Chen X, Xu S, Chu B, Guo J, Zhang H, Sun S, Song L, Feng XQ. Applying Spatiotemporal Modeling of Cell Dynamics to Accelerate Drug Development. ACS NANO 2024; 18:29311-29336. [PMID: 39420743 DOI: 10.1021/acsnano.4c12599] [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: 10/19/2024]
Abstract
Cells act as physical computational programs that utilize input signals to orchestrate molecule-level protein-protein interactions (PPIs), generating and responding to forces, ultimately shaping all of the physiological and pathophysiological behaviors. Genome editing and molecule drugs targeting PPIs hold great promise for the treatments of diseases. Linking genes and molecular drugs with protein-performed cellular behaviors is a key yet challenging issue due to the wide range of spatial and temporal scales involved. Building predictive spatiotemporal modeling systems that can describe the dynamic behaviors of cells intervened by genome editing and molecular drugs at the intersection of biology, chemistry, physics, and computer science will greatly accelerate pharmaceutical advances. Here, we review the mechanical roles of cytoskeletal proteins in orchestrating cellular behaviors alongside significant advancements in biophysical modeling while also addressing the limitations in these models. Then, by integrating generative artificial intelligence (AI) with spatiotemporal multiscale biophysical modeling, we propose a computational pipeline for developing virtual cells, which can simulate and evaluate the therapeutic effects of drugs and genome editing technologies on various cell dynamic behaviors and could have broad biomedical applications. Such virtual cell modeling systems might revolutionize modern biomedical engineering by moving most of the painstaking wet-laboratory effort to computer simulations, substantially saving time and alleviating the financial burden for pharmaceutical industries.
Collapse
Affiliation(s)
- Xindong Chen
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
- BioMap, Beijing 100144, China
| | - Shihao Xu
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Bizhu Chu
- School of Pharmacy, Shenzhen University, Shenzhen 518055, China
- Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jing Guo
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Huikai Zhang
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Shuyi Sun
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Le Song
- BioMap, Beijing 100144, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| |
Collapse
|
17
|
Wang Y, Wang C, Lu Y. Spleen Targeting Nucleic Acid Delivery Vector Based on Metal-Organic Frameworks. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56826-56836. [PMID: 39390629 DOI: 10.1021/acsami.4c13519] [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: 10/12/2024]
Abstract
Nucleic acids have attracted increasing attention as drugs due to their fascinating advantages, such as long-term efficacy and ease of preparation compared to proteins. The nucleic acid therapy relies heavily on delivery vectors, which can prevent the degradation of nucleic acids while assisting them in cellular internalization. However, commonly used nonviral vector liposomes easily accumulate in the liver, which can limit their application in extrahepatic diseases. Herein, a potential spleen targeting vector for nucleic acids is developed based on the metal-organic frameworks. The plasmids are encapsulated inside the nanoscale zeolitic imidazolate framework (ZIF) via coprecipitation. The co-encapsulation of the cationic polymer poly(ether imide) (PEI) and the stabilizer polyvinylpyrrolidone (PVP) can significantly improve particle dispersion and stability. The prepared nanoparticles allow efficient transfection in vitro, mainly through clathrin-mediated and caveolae-mediated endocytosis. The biodistribution in mice shows that 46% of the nanoparticles accumulate in the spleen, which is much higher than that of the liposomes. The vector can successfully deliver plasmids to extrahepatic organs for protein synthesis and even induce an immune response. The elaborate ZIF-based nanoparticle may offer a new route for extrahepatic, especially spleen targeting delivery for the nucleic acids.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Chen Wang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| |
Collapse
|
18
|
Archer PA, Heiler AJ, Bourque AR, Alapan Y, Thomas SN. Different leukocyte subsets are targeted by systemic and locoregional administration despite conserved nanomaterial characteristics optimal for lymph node delivery. Biomater Sci 2024; 12:5582-5597. [PMID: 39318195 PMCID: PMC11422756 DOI: 10.1039/d4bm00910j] [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: 07/09/2024] [Accepted: 09/07/2024] [Indexed: 09/26/2024]
Abstract
Lymph nodes (LNs) house a large proportion of the body's leukocytes. Accordingly, engineered nanomaterials are increasingly developed to direct therapeutics to LNs to enhance their efficacy. Yet while lymphatic delivery of nanomaterials to LNs upon locoregional injection has been extensively evaluated, nanomaterial delivery to LN-localized leukocytes after intravenous administration has not been systematically explored nor benchmarked. In this work, a panel of inert, fluorescent nanoscale tracers and drug delivery vehicles were utilized to interrogate intravenous versus locoregionally administered nanomaterial access to LNs and leukocyte subsets therein. Hydrodynamic size and material effects on LN accumulation extents were similar between intravenous versus intradermal injection routes. Nanomaterial distribution to various LN leukocyte subsets differed substantially with injection route, however, in a manner not proportional to total LN accumulation. While intravenously administered nanomaterials accumulated in LNs lowly compared to systemic tissues, in sharp contrast to locoregional delivery, they exhibited size-dependent but material-independent access to immune cells within the LN parenchyma, which are not easily accessed with locoregional delivery.
Collapse
Affiliation(s)
- Paul A Archer
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310, 315 Ferst Drive NW, Atlanta, GA 30332, USA.
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alexander J Heiler
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310, 315 Ferst Drive NW, Atlanta, GA 30332, USA.
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alisyn R Bourque
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310, 315 Ferst Drive NW, Atlanta, GA 30332, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yunus Alapan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310, 315 Ferst Drive NW, Atlanta, GA 30332, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
19
|
Gadalla HH, Yuan Z, Chen Z, Alsuwayyid F, Das S, Mitra H, Ardekani AM, Wagner R, Yeo Y. Effects of nanoparticle deformability on multiscale biotransport. Adv Drug Deliv Rev 2024; 213:115445. [PMID: 39222795 DOI: 10.1016/j.addr.2024.115445] [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: 06/13/2024] [Revised: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Deformability is one of the critical attributes of nanoparticle (NP) drug carriers, along with size, shape, and surface properties. It affects various aspects of NP biotransport, ranging from circulation and biodistribution to interactions with biological barriers and target cells. Recent studies report additional roles of NP deformability in biotransport processes, including protein corona formation, intracellular trafficking, and organelle distribution. This review focuses on the literature published in the past five years to update our understanding of NP deformability and its effect on NP biotransport. We introduce different methods of modulating and evaluating NP deformability and showcase recent studies that compare a series of NPs in their performance in biotransport events at all levels, highlighting the consensus and disagreement of the findings. It concludes with a perspective on the intricacy of systematic investigation of NP deformability and future opportunities to advance its control toward optimal drug delivery.
Collapse
Affiliation(s)
- Hytham H Gadalla
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Zhongyue Yuan
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Ziang Chen
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Faisal Alsuwayyid
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Department of Pharmaceutical Sciences, College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Subham Das
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN, 47907, USA
| | - Harsa Mitra
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN, 47907, USA
| | - Arezoo M Ardekani
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN, 47907, USA
| | - Ryan Wagner
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN, 47907, USA
| | - Yoon Yeo
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Purdue University Institute for Cancer Research, 201 South University Street, West Lafayette, IN, 47907, USA; Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
20
|
Sun T, Li C, Luan J, Zhao F, Zhang Y, Liu J, Shao L. Black phosphorus for bone regeneration: Mechanisms involved and influencing factors. Mater Today Bio 2024; 28:101211. [PMID: 39280114 PMCID: PMC11402231 DOI: 10.1016/j.mtbio.2024.101211] [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: 05/18/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
BP has shown good potential for promoting bone regeneration. However, the understanding of the mechanisms of BP-enhanced bone regeneration is still limited. This review first summarizes the recent advances in applications of BP in bone regeneration. We further highlight the possibility that BP enhances bone regeneration by regulating the behavior of mesenchymal stem cells (MSCs), osteoblasts, vascular endothelial cells (VECs), and macrophages, mainly through the regulation of cytoskeletal remodeling, energy metabolism, oxidation resistance and surface adsorption properties, etc. In addition, moderating the physicochemical properties of BP (i.e., shape, size, and surface charge) can alter the effects of BP on bone regeneration. This review reveals the underlying mechanisms of BP-enhanced bone regeneration and provides strategies for further material design of BP-based materials for bone regeneration.
Collapse
Affiliation(s)
- Ting Sun
- Foshan Stomatology Hospital & School of Medicine, Foshan University, Foshan, 528000, China
- School of Dentistry, Jinan University, Guangzhou, 510630, China
| | - Chufeng Li
- School of Dentistry, Jinan University, Guangzhou, 510630, China
| | - Jiayi Luan
- Foshan Stomatology Hospital & School of Medicine, Foshan University, Foshan, 528000, China
| | - Fujian Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
21
|
Fan N, Zhao F, Meng Y, Chen L, Miao L, Wang P, Tang M, Wu X, Li Y, Li Y, Gao Z. Metal complex lipid-based nanoparticles deliver metabolism-regulating lomitapide to overcome CTC immune evasion via activating STING pathway. Eur J Pharm Biopharm 2024; 203:114467. [PMID: 39173934 DOI: 10.1016/j.ejpb.2024.114467] [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: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Activating the cGAS-STING pathway of circulating tumor cell clusters (CTC clusters) represents a promising strategy to mitigate metastases. To fully exploit the potential of cholesterol-regulating agents in activating CTCs' STING levels, we developed a nanoparticle (NP) composed of metal complex lipid (MCL). This design includes MCL-miriplatin to increase NP stiffness and loads lomitapide (lomi) modulating cholesterol levels, resulting in the creation of PLTs@Pt-lipid@lomi NPs. MCL-miriplatin not only enhances lomi's eliciting efficacy on STING pathway but also increases NPs' stiffness, thus a vital factor affecting the penetration into CTC clusters to further boost lomi's ability. Demonstrated by cy5 tracking experiments, PLTs@Pt-lipid@lomi NPs quickly attach to cancer cell via platelet membrane anchorage, penetrate deep into the spheres, and reach the subcellular endoplasmic reticulum where lomi regulates cholesterol. Additionally, these NPs have been shown to track CTCs in the bloodstream, a capability not demonstrated by the free drug. PLTs@Pt-lipid@lomi NPs more efficiently activate the STING pathway and reduce CTC stemness compared to free lomi. Ultimately, PLTs@Pt-lipid@lomi NPs reduce metastasis in a post-surgery animal model. While cholesterol-regulating agents are limited in efficacy when being repositioned as immunomodulatory agents, this MCL-composing NP strategy demonstrates the potential to effectively deliver these agents to target CTC clusters.
Collapse
Affiliation(s)
- Ni Fan
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Feng Zhao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuanyuan Meng
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lin Miao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ping Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Manqing Tang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xuanjun Wu
- Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Qingdao, Shandong University, Shandong 266237, China
| | - Yingpeng Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yunfei Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
22
|
Wang J, Brugnoli B, Foglietta F, Andreana I, Longo G, Dinarelli S, Girasole M, Serpe L, Arpicco S, Francolini I, Di Meo C, Matricardi P. Tuning stiffness of hyaluronan-cholesterol nanogels by mussel-inspired dopamine-Fe 3+ coordination: Preparation and properties evaluation. Int J Biol Macromol 2024; 280:135553. [PMID: 39276885 DOI: 10.1016/j.ijbiomac.2024.135553] [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: 03/22/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
In the evolving field of nanomedicine, tailoring the mechanical properties of nanogels to fine-tune their biological performance is a compelling avenue of research. This work investigates an innovative method for modulating the stiffness of hyaluronan-cholesterol (HACH) nanogels, an area that remains challenging. By grafting dopamine (DOPA) onto the HA backbone, characterized through UV, 1H NMR, and FT-IR analyses, we synthesized a novel polymer that spontaneously forms nanogels in aqueous environments. These HACH-DOPA nanogels are characterized by their small size (~170 nm), negative charge (around -32 mV), high stability, efficient drug encapsulation, and potent antioxidant activities (measured by ABTS test). Leveraging mussel-inspired metal coordination chemistry, the DOPA moieties enable stiffness modulation of the nanogels through catechol-Fe3+ interactions. This modification leads to increased crosslinking and, consequently, nanogels with a significantly increased stiffness, as measured by atomic force microscopy (AFM), with the formation of the HACH-DOPA@Fe3+ complex being pH-dependent and reversible. The cytocompatibility was evaluated via WST-1 cell proliferation assays on HUVEC and HDF cell lines, showing no evident cytotoxicity. Furthermore, the modified nanogels demonstrated enhanced cellular uptake, suggesting their substantial potential for intracellular drug delivery applications, a hypothesis supported by confocal microscopy assays. This work not only provides valuable insight into modulating nanogel stiffness but also advances new nanosystems for promising biomedical applications.
Collapse
Affiliation(s)
- Ju Wang
- Departments of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Benedetta Brugnoli
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Federica Foglietta
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria, 9, 10125, Turin, Italy
| | - Ilaria Andreana
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria, 9, 10125, Turin, Italy
| | - Giovanni Longo
- Institute for the Structure of the Matter (ISM), Italian National Research Council (CNR), Via del fosso del Cavaliere 100, 00133, Rome, Italy
| | - Simone Dinarelli
- Institute for the Structure of the Matter (ISM), Italian National Research Council (CNR), Via del fosso del Cavaliere 100, 00133, Rome, Italy
| | - Marco Girasole
- Institute for the Structure of the Matter (ISM), Italian National Research Council (CNR), Via del fosso del Cavaliere 100, 00133, Rome, Italy
| | - Loredana Serpe
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria, 9, 10125, Turin, Italy
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria, 9, 10125, Turin, Italy
| | - Iolanda Francolini
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Chiara Di Meo
- Departments of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Pietro Matricardi
- Departments of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy.
| |
Collapse
|
23
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [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: 11/01/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
| | | | | | - Weiqi Hong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
24
|
Zheng Y, Luo S, Xu M, He Q, Xie J, Wu J, Huang Y. Transepithelial transport of nanoparticles in oral drug delivery: From the perspective of surface and holistic property modulation. Acta Pharm Sin B 2024; 14:3876-3900. [PMID: 39309496 PMCID: PMC11413706 DOI: 10.1016/j.apsb.2024.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 09/25/2024] Open
Abstract
Despite the promising prospects of nanoparticles in oral drug delivery, the process of oral administration involves a complex transportation pathway that includes cellular uptake, intracellular trafficking, and exocytosis by intestinal epithelial cells, which are necessary steps for nanoparticles to enter the bloodstream and exert therapeutic effects. Current researchers have identified several crucial factors that regulate the interaction between nanoparticles and intestinal epithelial cells, including surface properties such as ligand modification, surface charge, hydrophilicity/hydrophobicity, intestinal protein corona formation, as well as holistic properties like particle size, shape, and rigidity. Understanding these properties is essential for enhancing transepithelial transport efficiency and designing effective oral drug delivery systems. Therefore, this review provides a comprehensive overview of the surface and holistic properties that influence the transepithelial transport of nanoparticles, elucidating the underlying principles governing their impact on transepithelial transport. The review also outlines the chosen of parameters to be considered for the subsequent design of oral drug delivery systems.
Collapse
Affiliation(s)
- Yaxian Zheng
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Shiqin Luo
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Min Xu
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin He
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiang Xie
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiawei Wu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
25
|
Jain M, Yu X, Schneck JP, Green JJ. Nanoparticle Targeting Strategies for Lipid and Polymer-Based Gene Delivery to Immune Cells In Vivo. SMALL SCIENCE 2024; 4:2400248. [PMID: 40212067 PMCID: PMC11935263 DOI: 10.1002/smsc.202400248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/15/2024] [Indexed: 04/13/2025] Open
Abstract
Lipid nanoparticles and polymeric nanoparticles are promising biomaterial platforms for robust intracellular DNA and mRNA delivery, highlighted by the widespread use of nanoparticle- (NP) based mRNA vaccines to help end the COVID-19 pandemic. Recent research has sought to adapt this nanotechnology to transfect and engineer immune cells in vivo. The immune system is an especially appealing target due to its involvement in many different diseases, and ex vivo-engineered immune cell therapies like chimeric antigen receptor (CAR) T therapy have already demonstrated remarkable clinical success in certain blood cancers. Although gene delivery can potentially address some of the cost and manufacturing concerns associated with current autologous immune cell therapies, transfecting immune cells in vivo is challenging. Not only is extrahepatic NP delivery to lymphoid organs difficult, but immune cells like T cells have demonstrated particular resistance to transfection. Despite these challenges, the modular nature of NPs allows researchers to examine critical structure-function relationships between a particle's properties and its ability to specifically engineer immune cells in vivo. Herein, several nanomaterial components are outlined, including targeting ligands, nucleic acid cargo, chemical properties, physical properties, and the route of administration to specifically target NPs to immune cells for optimal in vivo transfection.
Collapse
Affiliation(s)
- Manav Jain
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
| | - Xinjie Yu
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Department of Chemical & Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jonathan P. Schneck
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Departments of Pathology and MedicineJohns Hopkins University School of MedicineBaltimoreMD21231USA
| | - Jordan J. Green
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Department of Chemical & Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of OncologyThe Sidney Kimmel Comprehensive Cancer CenterThe Bloomberg∼Kimmel Institute for Cancer ImmunotherapyJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Departments of Ophthalmology, Neurosurgery, and Materials Science & EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| |
Collapse
|
26
|
Paats JWD, Hamelmann NM, Paulusse JMJ. Dual-reactive single-chain polymer nanoparticles for orthogonal functionalization through active ester and click chemistry. J Control Release 2024; 373:117-127. [PMID: 38968970 DOI: 10.1016/j.jconrel.2024.07.003] [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: 09/20/2023] [Revised: 06/09/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Glucose has been extensively studied as a targeting ligand on nanoparticles for biomedical nanoparticles. A promising nanocarrier platform are single-chain polymer nanoparticles (SCNPs). SCNPs are well-defined 5-20 nm semi-flexible nano-objects, formed by intramolecularly crosslinked linear polymers. Functionality can be incorporated by introducing labile pentafluorophenyl (PFP) esters in the polymer backbone, which can be readily substituted by functional amine-ligands. However, not all ligands are compatible with PFP-chemistry, requiring different ligation strategies for increasing versatility of surface functionalization. Here, we combine active PFP-ester chemistry with copper(I)-catalyzed azide alkyne cycloaddition (CuAAC) click chemistry to yield dual-reactive SCNPs. First, the SCNPs are functionalized with increasing amounts of 1-amino-3-butyne groups through PFP-chemistry, leading to a range of butyne-SCNPs with increasing terminal alkyne-density. Subsequently, 3-azido-propylglucose is conjugated through the glucose C1- or C6-position by CuAAC click chemistry, yielding two sets of glyco-SCNPs. Cellular uptake is evaluated in HeLa cancer cells, revealing increased uptake upon higher glucose-surface density, with no apparent positional dependance. The general conjugation strategy proposed here can be readily extended to incorporate a wide variety of functional molecules to create vast libraries of multifunctional SCNPs.
Collapse
Affiliation(s)
- Jan-Willem D Paats
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500, AE, Enschede, the Netherlands
| | - Naomi M Hamelmann
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500, AE, Enschede, the Netherlands
| | - Jos M J Paulusse
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500, AE, Enschede, the Netherlands.
| |
Collapse
|
27
|
Yuan P, Yan X, Zong X, Li X, Yang C, Chen X, Li Y, Wen Y, Zhu T, Xue W, Dai J. Modulating Elasticity of Liposome for Enhanced Cancer Immunotherapy. ACS NANO 2024; 18:23797-23811. [PMID: 39140567 DOI: 10.1021/acsnano.4c09094] [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: 08/15/2024]
Abstract
Cancer immunotherapy has emerged as a promising approach to cancer treatment in recent years. The physical and chemical properties of nanocarriers are critical factors that regulate the immune activation of antigen-presenting cells (APCs) in the tumor microenvironment (TME). Herein, we extensively investigated the behavior of liposome nanoparticles (Lipo-NPs) with different elasticities, focusing on their interaction with immune cells and their transport mechanisms from tumors to tumor-draining lymph nodes (tdLNs). Successfully preparing Lipo-NPs with distinct elastic properties, their varied behaviors were observed, concerning immune cell interaction. Soft Lipo-NPs exhibited an affinity to cell membranes, while those with medium elasticity facilitated the cargo delivery to macrophages through membrane fusion. Conversely, hard Lipo-NPs enter macrophages via classical cellular uptake pathways. Additionally, it was noted that softer Lipo-NPs displayed superior transport to tdLNs in vivo, attributed to their deformable nature with lower elasticity. As a result, the medium elastic Lipo-NPs with agonists (cGAMP), by activating the STING pathway and enhancing transport to tdLNs, promoted abundant infiltration of tumor-infiltrating lymphocytes (TILs), leading to notable antitumor effects and extended survival in a melanoma mouse model. Furthermore, this study highlighted the potential synergistic effect of medium elasticity Lipo-NPs with immune checkpoint blockade (ICB) therapy in preventing tumor immune evasion. These findings hold promise for guiding immune-targeted delivery systems in cancer immunotherapy, particularly in vaccine design for tdLNs targeting and eradicating metastasis within tdLNs.
Collapse
Affiliation(s)
- Pengfei Yuan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Xiaodie Yan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Xiaoqing Zong
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Xiaodi Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Caiqi Yang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Xinjie Chen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yuchao Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yaoqi Wen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Tianci Zhu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Jian Dai
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
28
|
Gao Z, Sun H, Yang S, Li M, Qi N, Cui J. Red Blood Cell-Like Poly(ethylene glycol) Particles: Influence of Particle Stiffness on Biological Behaviors. ACS Macro Lett 2024; 13:966-971. [PMID: 39038183 DOI: 10.1021/acsmacrolett.4c00330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Cell-like particles represent a category of synthetic particles designed to emulate the structures or functions of natural cells. Herein, we present the assembly of cell-like poly(ethylene glycol) (PEG) particles with different stiffnesses and shapes via replication of animal cells and investigate the impact of particle stiffness on their biological behaviors. As a proof of concept, we fabricate red blood cell-like and spherical PEG particles with varying cross-linking densities. A systematic exploration of their properties, encompassing morphology, stiffness, deformability, and biodistribution, reveal the vital influence of particle stiffness on in vivo fate, elucidating its role in governing the traversal of capillaries and the dynamic interactions with phagocytic cells.
Collapse
Affiliation(s)
- Zhiliang Gao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Hongning Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Shuang Yang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Mengqi Li
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Na Qi
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, Shandong University, Jinan, Shandong 250100, China
| |
Collapse
|
29
|
Yang Z, Zhao Y, Zhang X, Huang L, Wang K, Sun J, Chen N, Yin W, Chen S, Zhi H, Xue L, An L, Li R, Dong H, Xu J, Li Y, Li Y. Nano-mechanical Immunoengineering: Nanoparticle Elasticity Reprograms Tumor-Associated Macrophages via Piezo1. ACS NANO 2024; 18:21221-21235. [PMID: 39079080 DOI: 10.1021/acsnano.4c04614] [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: 09/26/2024]
Abstract
The mechanical properties of nanoparticles play a crucial role in regulating nanobiointeractions, influencing processes such as blood circulation, tumor accumulation/penetration, and internalization into cancer cells. Consequently, they have a significant impact on drug delivery and therapeutic efficacy. However, it remains unclear whether and how macrophages alter their biological function in response to nanoparticle elasticity. Here, we report on the nano-mechanical biological effects resulting from the interactions between elastic silica nanoparticles (SNs) and macrophages. The SNs with variational elasticity Young's moduli ranging from 81 to 837 MPa were synthesized, and it was demonstrated that M2 [tumor-associated macrophages (TAMs)] could be repolarized to M1 by the soft SNs. Additionally, our findings revealed that cell endocytosis, membrane tension, the curvature protein Baiap2, and the cytoskeleton were all influenced by the elasticity of SNs. Moreover, the mechanically sensitive protein Piezo1 on the cell membrane was activated, leading to calcium ion influx, activation of the NF-κB pathway, and the initiation of an inflammatory response. In vivo experiments demonstrated that the softest 81 MPa SNs enhanced tumor penetration and accumulation and repolarized TAMs in intratumoral hypoxic regions, ultimately resulting in a significant inhibition of tumor growth. Taken together, this study has established a cellular feedback mechanism in response to nanoparticle elasticity, which induces plasma membrane deformation and subsequent activation of mechanosensitive signals. This provides a distinctive "nano-mechanical immunoengineering" strategy for reprogramming TAMs to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Zichen Yang
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuge Zhao
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xiaoyou Zhang
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Li Huang
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Kun Wang
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jiuyuan Sun
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Nana Chen
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Weimin Yin
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Shiyu Chen
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Hui Zhi
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Liangyi Xue
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Lulu An
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Rongjie Li
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Haiqing Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Tongji Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, P. R. China
| | - Jinfu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yan Li
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yongyong Li
- Shanghai Skin Disease Hospital, the Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
30
|
Cai R, Baimanov D, Yuan H, Xie H, Yu S, Zhang Z, Yang J, Zhao F, You Y, Guan Y, Zheng P, Xu M, Qi M, Zhang Z, Zhong S, Li YF, Wang L. Protein Corona-Directed Cellular Recognition and Uptake of Polyethylene Nanoplastics by Macrophages. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:14158-14168. [PMID: 39088650 DOI: 10.1021/acs.est.4c05215] [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: 08/03/2024]
Abstract
The widespread use of plastic products in daily life has raised concerns about the health hazards associated with nanoplastics (NPs). When exposed, NPs are likely to infiltrate the bloodstream, interact with plasma proteins, and trigger macrophage recognition and clearance. In this study, we focused on establishing a correlation between the unique protein coronal signatures of high-density (HDPE) and low-density (LDPE) polyethylene (PE) NPs with their ultimate impact on macrophage recognition and cytotoxicity. We observed that low-density and high-density lipoprotein receptors (LDLR and SR-B1), facilitated by apolipoproteins, played an essential role in PE-NP recognition. Consequently, PE-NPs activated the caspase-3/GSDME pathway and ultimately led to pyroptosis. Advanced imaging techniques, including label-free scattered light confocal imaging and cryo-soft X-ray transmission microscopy with 3D-tomographic reconstruction (nano-CT), provided powerful insights into visualizing NPs-cell interactions. These findings underscore the potential risks of NPs to macrophages and introduce analytical methods for studying the behavior of NPs in biological systems.
Collapse
Affiliation(s)
- Rui Cai
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, PR China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Didar Baimanov
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Hao Yuan
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, PR China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Hongxin Xie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, PR China
| | - Shengtao Yu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zehao Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jiacheng Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Feng Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yue You
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yong Guan
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei 230029, PR China
| | - Pingping Zheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ming Xu
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| | - Mengying Qi
- University of Chinese Academy of Sciences, Beijing 100049, PR China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
| | - Zhiyong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shengliang Zhong
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, PR China
| | - Yu-Feng Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Liming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
31
|
Fan L, Tong W, Wei A, Mu X. Progress of proteolysis-targeting chimeras (PROTACs) delivery system in tumor treatment. Int J Biol Macromol 2024; 275:133680. [PMID: 38971291 DOI: 10.1016/j.ijbiomac.2024.133680] [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: 05/03/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) can use the intrinsic protein degradation system in cells to degrade pathogenic target proteins, and are currently a revolutionary frontier of development strategy for tumor treatment with small molecules. However, the poor water solubility, low cellular permeability, and off-target side effects of most PROTACs have prevented them from passing the preclinical research stage of drug development. This requires the use of appropriate delivery systems to overcome these challenging hurdles and ensure precise delivery of PROTACs towards the tumor site. Therefore, the combination of PROTACs and multifunctional delivery systems will open up new research directions for targeted degradation of tumor proteins. In this review, we systematically reviewed the design principles and the most recent advances of various PROTACs delivery systems. Moreover, the constructive strategies for developing multifunctional PROTACs delivery systems were proposed comprehensively. This review aims to deepen the understanding of PROTACs drugs and promote the further development of PROTACs delivery system.
Collapse
Affiliation(s)
- Lianlian Fan
- Department of Pharmacy, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Weifang Tong
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130021, China
| | - Anhui Wei
- Jilin University School of Pharmaceutical Sciences, Changchun 130021, China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
32
|
Reddy Baddam S, Ganta S, Nalla S, Banoth C, Vudari B, Akkiraju PC, Srinivas E, Tade RS. Polymeric nanomaterials-based theranostic platforms for triple-negative breast cancer (TNBC) treatment. Int J Pharm 2024; 660:124346. [PMID: 38889853 DOI: 10.1016/j.ijpharm.2024.124346] [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: 04/02/2024] [Revised: 06/05/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer, the second leading global cause of death, affects 2.1 million women annually, with an alarming 15 percent mortality rate. Among its diverse forms, Triple-negative breast cancer (TNBC) emerges as the deadliest, characterized by the absence of hormone receptors. This article underscores the urgent need for innovative treatment approaches in tackling TNBC, emphasizing the transformative potential of polymeric nanomaterials (PNMs). Evolved through nanotechnology, PNMs offer versatile biomedical applications, particularly in addressing the intricate challenges of TNBC. The synthesis methods of PNMs, explored within the tumor microenvironment using cellular models, showcase their dynamic nature in cancer treatment. The article anticipates the future of TNBC therapeutics through the optimization of PNMs-based strategies, integrating them into photothermal (PT), photodynamic (PT), and hyperthermia therapy (HTT), drug delivery, and active tumor targeting strategies. Advancements in synthetic methods, coupled with a nuanced understanding of the tumor microenvironment, hold promise for personalized interventions. Comparative investigations of therapeutic models and a thorough exploration of polymeric nanoplatforms toxicological perspectives become imperative for ensuring efficacy and safety. We have explored the interdisciplinary collaboration between nanotechnology, oncology, and molecular biology as pivotal in translating PNMs innovations into tangible benefits for TNBC patients.
Collapse
Affiliation(s)
- Sudhakar Reddy Baddam
- University of Massachusetts, Chan Medical School, RNA Therapeutic Institute, Worcester, MA 01655, USA
| | | | | | - Chandrasekhar Banoth
- Department of Microbiology, Army College of Dental Sciences, Chennapur, Secunderabad 500087, India
| | - Balaraju Vudari
- Sreenidhi Institute of Science and Technology, Hyderabad, Telangana 501301, India
| | - Pavan C Akkiraju
- Department of Medical Biotechnology, School of Allied Healthcare Sciences, Malla Reddy University, Hyderabad 500014, India
| | - Enaganti Srinivas
- Averinbiotech Laboratories, Windsor Plaza, Nallakunta, Hyderabad 500044, India
| | - Rahul S Tade
- Department of Pharmaceutics, H.R. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405, India.
| |
Collapse
|
33
|
Zhao Z, Liu M, Duan L, Lin R, Wang L, Zhang P, Li J, Ma B, Yang Y, Bu F, Wang R, Zhou W, Chao D, Zhao Y, Yin S, Tang L, Zhang W, Li X, Zhao D. Ultrafine Asymmetric Soft/Stiff Nanohybrids with Tunable Patchiness via a Dynamic Surface-Mediated Assembly. J Am Chem Soc 2024. [PMID: 39025826 DOI: 10.1021/jacs.4c05072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Asymmetric soft-stiff patch nanohybrids with small size, spatially separated organics and inorganics, controllable configuration, and appealing functionality are important in applications, while the synthesis remains a great challenge. Herein, based on polymeric single micelles (the smallest assembly subunit of mesoporous materials), we report a dynamic surface-mediated anisotropic assembly approach to fabricate a new type of small asymmetric organic/inorganic patch nanohybrid for the first time. The size of this asymmetric organic/inorganic nanohybrid is ∼20 nm, which contains dual distinct subunits of a soft organic PS-PVP-PEO single micelle nanosphere (12 nm in size and 632 MPa in Young' modulus) and stiff inorganic SiO2 nanobulge (∼8 nm, 2275 MPa). Moreover, the number of SiO2 nanobulges anchored on each micelle can be quantitatively controlled (from 1 to 6) by dynamically tuning the density (fluffy or dense state) of the surface cap organic groups. This small asymmetric patch nanohybrid also exhibits a dramatically enhanced uptake level of which the total amount of intracellular endocytosis is about three times higher than that of the conventional nanohybrids.
Collapse
Affiliation(s)
- Zaiwang Zhao
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Mengli Liu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Linlin Duan
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Runfeng Lin
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Lipeng Wang
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Pengfei Zhang
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Jun Li
- Henan Institute of Advanced Technology, College of Chemistry, Zhengzhou University, Zhengzhou 450052, China
| | - Bing Ma
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Yang Yang
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Fanxing Bu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Ruicong Wang
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Wanhai Zhou
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Dongliang Chao
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Yujuan Zhao
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Sixing Yin
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Lei Tang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| | - Weian Zhang
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Xiaomin Li
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Dongyuan Zhao
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
34
|
Wang X, Yu H, Liu D, Hu B, Zhang R, Hu L, Hu G, Li C. The application of nanomaterials in tumor therapy based on the regulation of mechanical properties. NANOSCALE 2024; 16:13386-13398. [PMID: 38967103 DOI: 10.1039/d4nr01812e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Mechanical properties, as crucial physical properties, have a significant impact on the occurrence, development, and metastasis of tumors. Regulating the mechanical properties of tumors to enhance their sensitivity to radiotherapy and chemotherapy has become an important strategy in the field of cancer treatment. Over the past few decades, nanomaterials have made remarkable progress in cancer therapy, either based on their intrinsic properties or as drug delivery carriers. However, the investigation of nanomaterials of mechanical regulation in tumor therapy is currently in its initial stages. The mechanical properties of nanomaterials themselves, drug carrier targeting, and regulation of the mechanical environment of tumor tissue have far-reaching effects on the efficient uptake of drugs and clinical tumor treatment. Therefore, this review aims to comprehensively summarize the applications and research progress of nanomaterials in tumor therapy based on the regulation of mechanical properties, in order to provide strong support for further research and the development of treatment strategies in this field.
Collapse
Affiliation(s)
- Xiaolei Wang
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beihang University, Beijing 100191, China.
| | - Hongxi Yu
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beihang University, Beijing 100191, China.
| | - Dan Liu
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beihang University, Beijing 100191, China.
| | - Boxian Hu
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beihang University, Beijing 100191, China.
| | - Ruihang Zhang
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beihang University, Beijing 100191, China.
| | - Lihua Hu
- Department of Cardiology, Peking University First Hospital, Beijing 100034, China
| | - Guiping Hu
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beihang University, Beijing 100191, China.
| | - Cheng Li
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beihang University, Beijing 100191, China.
| |
Collapse
|
35
|
Wang S, Lv Y. Silica-coated liquid metal nanoparticles with different stiffness for cellular uptake-enhanced tumor photothermal therapy. BIOMATERIALS ADVANCES 2024; 161:213872. [PMID: 38733802 DOI: 10.1016/j.bioadv.2024.213872] [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: 01/06/2024] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024]
Abstract
Cells can sense the mechanical stimulation of nanoparticles (NPs) and then regulate the cellular uptake process. The enhanced endocytosis efficiency can improve the concentration of NPs in tumor cells significantly, which is the key prerequisite for achieving efficient biological performance. However, the preparation methods of NPs with flexible and tunable stiffness are relatively limited, and the impact of stiffness property on their interaction with tumor cells remains unclear. In this study, soft liquid metal (LM) core was coated with hard silica layer, the obtained core-shell NPs with a wide range of Young's modulus (130.5 ± 25.6 MPa - 1729.2 ± 146.7 MPa) were prepared by adjusting the amount of silica. It was found that the NPs with higher stiffness exhibited superior cellular uptake efficiency and lysosomal escape ability compared to the NPs with lower stiffness. The silica layer not only affected the stiffness, but also improved the photothermal stability of the LM NPs. Both in vitro and in vivo results demonstrated that the NPs with higher stiffness displayed significantly enhanced tumor hyperthermia capability. This work may provide a paradigm for the preparation of NPs with varying stiffness and offer insights into the role of the mechanical property of NPs in their delivery.
Collapse
Affiliation(s)
- Shuai Wang
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, PR China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, PR China.
| |
Collapse
|
36
|
Pileni MP. "Nano-egg" superstructures of hydrophobic nanocrystals dispersed in water. Phys Chem Chem Phys 2024; 26:16931-16941. [PMID: 38835199 DOI: 10.1039/d4cp01299b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
In this feature article, we use hydrophobic ferrite (Fe3O4) nanocrystal shells filled with Au nanocrystals self-assembled into 3D superlattices and dispersed in water. These superstructures act as nano-heaters. The stability of such superstructures is very high, even for several years, when stored at room temperature. When subjected to an electron beam, the inverted structure of Fe3O4 structures is gradually dissolved due to the formation of hydrated electrons and hydroxyl radicals.
Collapse
Affiliation(s)
- M P Pileni
- Sorbonne Université, Department of Chemistry, 4 Place Jussieu, 75005 Paris, France.
| |
Collapse
|
37
|
Wang KN, Zhou K, Zhong NN, Cao LM, Li ZZ, Xiao Y, Wang GR, Huo FY, Zhou JJ, Liu B, Bu LL. Enhancing cancer therapy: The role of drug delivery systems in STAT3 inhibitor efficacy and safety. Life Sci 2024; 346:122635. [PMID: 38615745 DOI: 10.1016/j.lfs.2024.122635] [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: 01/16/2024] [Revised: 03/14/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
The signal transducer and activator of transcription 3 (STAT3), a member of the STAT family, resides in the nucleus to regulate genes essential for vital cellular functions, including survival, proliferation, self-renewal, angiogenesis, and immune response. However, continuous STAT3 activation in tumor cells promotes their initiation, progression, and metastasis, rendering STAT3 pathway inhibitors a promising avenue for cancer therapy. Nonetheless, these inhibitors frequently encounter challenges such as cytotoxicity and suboptimal biocompatibility in clinical trials. A viable strategy to mitigate these issues involves delivering STAT3 inhibitors via drug delivery systems (DDSs). This review delineates the regulatory mechanisms of the STAT3 signaling pathway and its association with cancer. It offers a comprehensive overview of the current application of DDSs for anti-STAT3 inhibitors and investigates the role of DDSs in cancer treatment. The conclusion posits that DDSs for anti-STAT3 inhibitors exhibit enhanced efficacy and reduced adverse effects in tumor therapy compared to anti-STAT3 inhibitors alone. This paper aims to provide an outline of the ongoing research and future prospects of DDSs for STAT3 inhibitors. Additionally, it presents our insights on the merits and future outlook of DDSs in cancer treatment.
Collapse
Affiliation(s)
- Kang-Ning Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Fang-Yi Huo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jun-Jie Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial, Anyang Sixth People's Hospital, Anyang 45500, China.
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
38
|
Li ZZ, Zhong NN, Cao LM, Cai ZM, Xiao Y, Wang GR, Liu B, Xu C, Bu LL. Nanoparticles Targeting Lymph Nodes for Cancer Immunotherapy: Strategies and Influencing Factors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308731. [PMID: 38327169 DOI: 10.1002/smll.202308731] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/07/2024] [Indexed: 02/09/2024]
Abstract
Immunotherapy has emerged as a potent strategy in cancer treatment, with many approved drugs and modalities in the development stages. Despite its promise, immunotherapy is not without its limitations, including side effects and suboptimal efficacy. Using nanoparticles (NPs) as delivery vehicles to target immunotherapy to lymph nodes (LNs) can improve the efficacy of immunotherapy drugs and reduce side effects in patients. In this context, this paper reviews the development of LN-targeted immunotherapeutic NP strategies, the mechanisms of NP transport during LN targeting, and their related biosafety risks. NP targeting of LNs involves either passive targeting, influenced by NP physical properties, or active targeting, facilitated by affinity ligands on NP surfaces, while alternative methods, such as intranodal injection and high endothelial venule (HEV) targeting, have uncertain clinical applicability and require further research and validation. LN targeting of NPs for immunotherapy can reduce side effects and increase biocompatibility, but risks such as toxicity, organ accumulation, and oxidative stress remain, although strategies such as biodegradable biomacromolecules, polyethylene glycol (PEG) coating, and impurity addition can mitigate these risks. Additionally, this work concludes with a future-oriented discussion, offering critical insights into the field.
Collapse
Affiliation(s)
- Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Ze-Min Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Chun Xu
- School of Dentistry, The University of Queensland, 288 Herston Road, Brisbane, 4066, Australia
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| |
Collapse
|
39
|
Liang G, Cao W, Tang D, Zhang H, Yu Y, Ding J, Karges J, Xiao H. Nanomedomics. ACS NANO 2024; 18:10979-11024. [PMID: 38635910 DOI: 10.1021/acsnano.3c11154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Nanomaterials have attractive physicochemical properties. A variety of nanomaterials such as inorganic, lipid, polymers, and protein nanoparticles have been widely developed for nanomedicine via chemical conjugation or physical encapsulation of bioactive molecules. Superior to traditional drugs, nanomedicines offer high biocompatibility, good water solubility, long blood circulation times, and tumor-targeting properties. Capitalizing on this, several nanoformulations have already been clinically approved and many others are currently being studied in clinical trials. Despite their undoubtful success, the molecular mechanism of action of the vast majority of nanomedicines remains poorly understood. To tackle this limitation, herein, this review critically discusses the strategy of applying multiomics analysis to study the mechanism of action of nanomedicines, named nanomedomics, including advantages, applications, and future directions. A comprehensive understanding of the molecular mechanism could provide valuable insight and therefore foster the development and clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Ganghao Liang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Wanqing Cao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
40
|
Kim J, Lee E, Lee ES. Development of 5-Fluorouracil/pH-Responsive Adjuvant-Embedded Extracellular Vesicles for Targeting α vβ 3 Integrin Receptors in Tumors. Pharmaceutics 2024; 16:599. [PMID: 38794261 PMCID: PMC11125367 DOI: 10.3390/pharmaceutics16050599] [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: 03/21/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
To selectively target and treat murine melanoma B16BL6 tumors expressing αvβ3 integrin receptors, we engineered tumor-specific functional extracellular vesicles (EVs) tailored for the targeted delivery of antitumor drugs. This objective was achieved through the incorporation of a pH-responsive adjuvant, cyclic arginine-glycine-aspartic acid peptide (cRGD, serving as a tumor-targeting ligand), and 5-fluorouracil (5-FU, employed as a model antitumor drug). The pH-responsive adjuvant, essential for modulating drug release, was synthesized by chemically conjugating 3-(diethylamino)propylamine (DEAP) to deoxycholic acid (DOCA, a lipophilic substance capable of integrating into EVs' membranes), denoted as DEAP-DOCA. The DOCA, preactivated using N-(2-aminoethyl)maleimide (AEM), was chemically coupled with the thiol group of the cRGD-DOCA through the thiol-maleimide click reaction, resulting in the formation of cRGD-DOCA. Subsequently, DEAP-DOCA, cRGD-DOCA, and 5-FU were efficiently incorporated into EVs using a sonication method. The resulting tumor-targeting EVs, expressing cRGD ligands, demonstrated enhanced in vitro/in vivo cellular uptake specifically for B16BL6 tumors expressing αvβ3 integrin receptors. The ionization characteristics of the DEAP in DEAP-DOCA induced destabilization of the EVs membrane at pH 6.5 through protonation of the DEAP substance, thereby expediting 5-FU release. Consequently, an improvement in the in vivo antitumor efficacy was observed for B16BL6 tumors. Based on these comprehensive in vitro/in vivo findings, we anticipate that this EV system holds substantial promise as an exceptionally effective platform for antitumor therapeutic delivery.
Collapse
Affiliation(s)
- Jiseung Kim
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si 1462, Gyeonggi-do, Republic of Korea; (J.K.); (E.L.)
| | - Eunsol Lee
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si 1462, Gyeonggi-do, Republic of Korea; (J.K.); (E.L.)
| | - Eun Seong Lee
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si 1462, Gyeonggi-do, Republic of Korea
| |
Collapse
|
41
|
Tian Y, Cheng T, Sun F, Zhou Y, Yuan C, Guo Z, Wang Z. Effect of biophysical properties of tumor extracellular matrix on intratumoral fate of nanoparticles: Implications on the design of nanomedicine. Adv Colloid Interface Sci 2024; 326:103124. [PMID: 38461766 DOI: 10.1016/j.cis.2024.103124] [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: 10/23/2023] [Revised: 02/11/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
Nanomedicine has a profound impact on various research domains including drug delivery, diagnostics, theranostics, and regenerative medicine. Nevertheless, the clinical translation of nanomedicines for solid cancer remains limited due to the abundant physiological and pathological barriers in tumor that hinder the intratumoral penetration and distribution of these nanomedicines. In this article, we review the dynamic remodeling of tumor extracellular matrix during the tumor progression, discuss the impact of biophysical obstacles within tumors on the penetration and distribution of nanomedicines within the solid tumor and collect innovative approaches to surmount these obstacles for improving the penetration and accumulation of nanomedicines in tumor. Furthermore, we dissect the challenges and opportunities of the respective approaches, and propose potential avenues for future investigations. The purpose of this review is to provide a perspective guideline on how to effectively enhance the penetration of nanomedicines within tumors using promising methods.
Collapse
Affiliation(s)
- Yachao Tian
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China; Heilongjiang Guoru Biotechnology Co., Ltd., Xiangfang District, Harbin City 150030, China; School of Food Science and Engineering, Qilu University of Technology, Jinan, Shandong 250353, China
| | - Tianfu Cheng
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Fuwei Sun
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Yaxin Zhou
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Chao Yuan
- School of Food Science and Engineering, Qilu University of Technology, Jinan, Shandong 250353, China
| | - Zengwang Guo
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| | - Zhongjiang Wang
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| |
Collapse
|
42
|
Hou F, Guo Z, Ho MT, Hui Y, Zhao CX. Particle-Based Artificial Antigen-Presenting Cell Systems for T Cell Activation in Adoptive T Cell Therapy. ACS NANO 2024; 18:8571-8599. [PMID: 38483840 DOI: 10.1021/acsnano.3c10180] [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: 03/27/2024]
Abstract
T cell-based adoptive cell therapy (ACT) has emerged as a promising treatment for various diseases, particularly cancers. Unlike other immunotherapy modalities, ACT involves directly transferring engineered T cells into patients to eradicate diseased cells; hence, it necessitates methods for effectively activating and expanding T cells in vitro. Artificial antigen-presenting cells (aAPCs) have been widely developed based on biomaterials, particularly micro- and nanoparticles, and functionalized with T cell stimulatory antibodies to closely mimic the natural T cell-APC interactions. Due to their vast clinical utility, aAPCs have been employed as an off-the-shelf technology for T cell activation in FDA-approved ACTs, and the development of aAPCs is constantly advancing with the emergence of aAPCs with more sophisticated designs and additional functionalities. Here, we review the recent advancements in particle-based aAPCs for T cell activation in ACTs. Following a brief introduction, we first describe the manufacturing processes of ACT products. Next, the design and synthetic strategies for micro- and nanoparticle-based aAPCs are discussed separately to emphasize their features, advantages, and limitations. Then, the impact of design parameters of aAPCs, such as size, shape, ligand density/mobility, and stiffness, on their functionality and biomedical performance is explored to provide deeper insights into the design concepts and principles for more efficient and safer aAPCs. The review concludes by discussing current challenges and proposing future perspectives for the development of more advanced aAPCs.
Collapse
Affiliation(s)
- Fei Hou
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Zichao Guo
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Minh Trang Ho
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Yue Hui
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Chun-Xia Zhao
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
43
|
Ghosh R, Roy L, Mukherjee D, Sarker S, Mondal J, Pan N, Hasan MN, Ghosh S, Chattopadhyay A, Adhikary A, Bhattacharyya M, Mallick AK, Biswas R, Das R, Pal SK. Structurally Dynamic Monocyte-Liposome Hybrid Vesicles as an Anticancer Drug Delivery Vehicle: A Crucial Correlation of Microscopic Elasticity and Ultrafast Dynamics. J Phys Chem Lett 2024; 15:3078-3088. [PMID: 38467015 DOI: 10.1021/acs.jpclett.3c03192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
A biomimetic cell-based carrier system based on monocyte membranes and liposomes has been designed to create a hybrid "Monocyte-LP" which inherits the surface antigens of the monocytes along with the drug encapsulation property of the liposome. Förster resonance energy transfer (FRET) and polarization gated anisotropy measurements show the stiffness of the vesicles obtained from monocyte membranes (Mons), phosphatidylcholine membranes (LP), and Monocyte-LP to follow an increasing order of Mons > Monocyte-LP > LP. The dynamics of interface bound water molecules plays a key role in the elasticity of the vesicles, which in turn imparts higher delivery efficacy to the hybrid Monocyte-LP for a model anticancer drug doxorubicin than the other two vesicles, indicating a critical balance between flexibility and rigidity for an efficient cellular uptake. The present work provides insight on the influence of elasticity of delivery vehicles for enhanced drug delivery.
Collapse
Affiliation(s)
- Ria Ghosh
- Department of Biochemistry, University of Calcutta 35 Ballygunge Circular Road, Ballygunge, Kolkata 700019, India
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Lopamudra Roy
- Department of Applied Optics and Photonics, University of Calcutta, Block-JD, Sector-III, Saltlake, Kolkata 700106, India
| | - Dipanjan Mukherjee
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Sushmita Sarker
- Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, Salt Lake, Kolkata 700106, India
| | - Jayanta Mondal
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Nivedita Pan
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Md Nur Hasan
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Subhajit Ghosh
- Department of Life Science & Biotechnology, Jadavpur University, 188, Raja Subodh Chandra Mallick Rd, Jadavpur, Kolkata, West Bengal 700032, India
| | - Arpita Chattopadhyay
- Department of Basic science and Humanities, Techno International New Town Block, DG 1/1, Action Area 1, New Town, Rajarhat, Kolkata 700156, India
| | - Arghya Adhikary
- Department of Life Science & Biotechnology, Jadavpur University, 188, Raja Subodh Chandra Mallick Rd, Jadavpur, Kolkata, West Bengal 700032, India
| | - Maitree Bhattacharyya
- Department of Biochemistry, University of Calcutta 35 Ballygunge Circular Road, Ballygunge, Kolkata 700019, India
| | - Asim Kumar Mallick
- Department of Paediatric Medicine, Nil RatanSircar Medical College & Hospital, 138, AJC Bose Road, Sealdah, Raja Bazar, Kolkata 700014, India
| | - Ranjit Biswas
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Ranjan Das
- Department of Chemistry, West Bengal State University, Barasat, Kolkata 700126, India
| | - Samir Kumar Pal
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, West Bengal 700106, India
| |
Collapse
|
44
|
Afrifa MAO, Kim JH, Pitton KA, Olelewe C, Arojojoye AS, Strachan DR, Suckow MA, Awuah SG. Auranofin-Loaded Chitosan Nanoparticles Demonstrate Potency against Triple-Negative Breast Cancer. ACS APPLIED BIO MATERIALS 2024; 7:2012-2022. [PMID: 38450675 PMCID: PMC11214827 DOI: 10.1021/acsabm.4c00184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Triple-negative breast cancer (TNBC) remains a clinical challenge due to molecular, metabolic, and genetic heterogeneity as well as the lack of validated drug targets. Thus, therapies or delivery paradigms are needed. Gold-derived compounds including the FDA-approved drug, auranofin have shown promise as effective anticancer agents against several tumors. To improve the solubility and bioavailability of auranofin, we hypothesized that the nanodelivery of auranofin using biodegradable chitosan modified polyethylene glycol (PEG) nanoparticles (NPs) will enhance anticancer activity against TNBC by comparing the best nanoformulation with the free drug. The selection of the nanoformulation was based on synthesis of various chitosan PEG copolymers via formaldehyde-mediated engraftment of PEG onto chitosan to form [chitosan-g-PEG] copolymer. Furthermore, altered physiochemical properties of the copolymer was based on the formaldehyde ratio towards nanoparticles (CP 1-4 NPs). Following the recruitment of PEG onto the chitosan polymer surface, we explored how this process influenced the stiffness of the nanoparticle using atomic force microscopy (AFM), a factor crucial for in vitro and in vivo studies. Our objective was to ensure the full functionality and inherent properties of chitosan as the parent polymer was maintained without allowing PEG to overshadow chitosan's unique cationic properties while improving solubility in neutral pH. Hence, CP 2 NP was chosen. To demonstrate the efficacy of CP 2 NP as a good delivery carrier for auranofin, we administered a dose of 3 mg/kg of auranofin, in contrast to free auranofin, which was given at 5 mg/kg. In vivo studies revealed the potency of encapsulated auranofin against TNBC cells with a severe necrotic effect following treatment superior to that of free auranofin. In conclusion, chitosan-g-PEG nanoparticles have the potential to be an excellent delivery system for auranofin, increasing its effectiveness and potentially reducing its clinical limitations.
Collapse
Affiliation(s)
- Maame Abena O. Afrifa
- Department of Biomedical Engineering, University of Kentucky; Lexington, Kentucky, 40506, USA
| | - Jong H. Kim
- Department of Chemistry, University of Kentucky; Lexington, Kentucky, 40506, USA
| | - Kathryn A. Pitton
- Department of Chemistry, University of Kentucky; Lexington, Kentucky, 40506, USA
| | - Chibuzor Olelewe
- Department of Chemistry, University of Kentucky; Lexington, Kentucky, 40506, USA
| | | | - Douglas R. Strachan
- Department of Astronomy and Physics, University of Kentucky; Lexington, Kentucky, 40506, USA
| | - Mark A. Suckow
- Department of Biomedical Engineering, University of Kentucky; Lexington, Kentucky, 40506, USA
- Attending Veterinarian, University of Kentucky, Lexington, Kentucky, 40506, USA
| | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky; Lexington, Kentucky, 40506, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
- University of Kentucky Markey NCI Comprehensive Cancer Center, Lexington, Kentucky 40536, United States
- University of Kentucky Bioelectronics and Nanomedicine Research Center, Lexington, Kentucky 40506, United States
| |
Collapse
|
45
|
Xie H, Zhang C. Potential of the nanoplatform and PROTAC interface to achieve targeted protein degradation through the Ubiquitin-Proteasome system. Eur J Med Chem 2024; 267:116168. [PMID: 38310686 DOI: 10.1016/j.ejmech.2024.116168] [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: 11/03/2023] [Revised: 01/11/2024] [Accepted: 01/21/2024] [Indexed: 02/06/2024]
Abstract
In eukaryotic cells, the ubiquitin-proteasome system (UPS) plays a crucial role in selectively breaking down specific proteins. The ability of the UPS to target proteins effectively and expedite their removal has significantly contributed to the evolution of UPS-based targeted protein degradation (TPD) strategies. In particular, proteolysis targeting chimeras (PROTACs) are an immensely promising tool due to their high efficiency, extensive target range, and negligible drug resistance. This breakthrough has overcome the limitations posed by traditionally "non-druggable" proteins. However, their high molecular weight and constrained solubility impede the delivery of PROTACs. Fortunately, the field of nanomedicine has experienced significant growth, enabling the delivery of PROTACs through nanoscale drug-delivery systems, which effectively improves the stability, solubility, drug distribution, tissue-specific accumulation, and stimulus-responsive release of PROTACs. This article reviews the mechanism of action attributed to PROTACs and their potential implications for clinical applications. Moreover, we present strategies involving nanoplatforms for the effective delivery of PROTACs and evaluate recent advances in targeting nanoplatforms to the UPS. Ultimately, an assessment is conducted to determine the feasibility of utilizing PROTACs and nanoplatforms for UPS-based TPD. The primary aim of this review is to provide innovative, reliable solutions to overcome the current challenges obstructing the effective use of PROTACs in the management of cancer, neurodegenerative diseases, and metabolic syndrome. Therefore, this is a promising technology for improving the treatment status of major diseases.
Collapse
Affiliation(s)
- Hanshu Xie
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Chao Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
46
|
Cooley MB, Wegierak D, Exner AA. Using imaging modalities to predict nanoparticle distribution and treatment efficacy in solid tumors: The growing role of ultrasound. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1957. [PMID: 38558290 PMCID: PMC11006412 DOI: 10.1002/wnan.1957] [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: 04/26/2023] [Revised: 12/22/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
Nanomedicine in oncology has not had the success in clinical impact that was anticipated in the early stages of the field's development. Ideally, nanomedicines selectively accumulate in tumor tissue and reduce systemic side effects compared to traditional chemotherapeutics. However, this has been more successful in preclinical animal models than in humans. The causes of this failure to translate may be related to the intra- and inter-patient heterogeneity of the tumor microenvironment. Predicting whether a patient will respond positively to treatment prior to its initiation, through evaluation of characteristics like nanoparticle extravasation and retention potential in the tumor, may be a way to improve nanomedicine success rate. While there are many potential strategies to accomplish this, prediction and patient stratification via noninvasive medical imaging may be the most efficient and specific strategy. There have been some preclinical and clinical advances in this area using MRI, CT, PET, and other modalities. An alternative approach that has not been studied as extensively is biomedical ultrasound, including techniques such as multiparametric contrast-enhanced ultrasound (mpCEUS), doppler, elastography, and super-resolution processing. Ultrasound is safe, inexpensive, noninvasive, and capable of imaging the entire tumor with high temporal and spatial resolution. In this work, we summarize the in vivo imaging tools that have been used to predict nanoparticle distribution and treatment efficacy in oncology. We emphasize ultrasound imaging and the recent developments in the field concerning CEUS. The successful implementation of an imaging strategy for prediction of nanoparticle accumulation in tumors could lead to increased clinical translation of nanomedicines, and subsequently, improved patient outcomes. This article is categorized under: Diagnostic Tools In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery Emerging Technologies.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Radiology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| |
Collapse
|
47
|
Wang L, Quine S, Frickenstein AN, Lee M, Yang W, Sheth VM, Bourlon MD, He Y, Lyu S, Garcia-Contreras L, Zhao YD, Wilhelm S. Exploring and Analyzing the Systemic Delivery Barriers for Nanoparticles. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2308446. [PMID: 38828467 PMCID: PMC11142462 DOI: 10.1002/adfm.202308446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Indexed: 06/05/2024]
Abstract
Most nanomedicines require efficient in vivo delivery to elicit diagnostic and therapeutic effects. However, en route to their intended tissues, systemically administered nanoparticles often encounter delivery barriers. To describe these barriers, we propose the term "nanoparticle blood removal pathways" (NBRP), which summarizes the interactions between nanoparticles and the body's various cell-dependent and cell-independent blood clearance mechanisms. We reviewed nanoparticle design and biological modulation strategies to mitigate nanoparticle-NBRP interactions. As these interactions affect nanoparticle delivery, we studied the preclinical literature from 2011-2021 and analyzed nanoparticle blood circulation and organ biodistribution data. Our findings revealed that nanoparticle surface chemistry affected the in vivo behavior more than other nanoparticle design parameters. Combinatory biological-PEG surface modification improved the blood area under the curve by ~418%, with a decrease in liver accumulation of up to 47%. A greater understanding of nanoparticle-NBRP interactions and associated delivery trends will provide new nanoparticle design and biological modulation strategies for safer, more effective, and more efficient nanomedicines.
Collapse
Affiliation(s)
- Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Skyler Quine
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Michael Lee
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Vinit M. Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Margaret D. Bourlon
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yuxin He
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Shanxin Lyu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Lucila Garcia-Contreras
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yan D. Zhao
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73012, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), Norman, Oklahoma, 73019, USA
| |
Collapse
|
48
|
Zhou W, Lew B, Choi H, Kim K, Anakk S. Chenodeoxycholic Acid-Loaded Nanoparticles Are Sufficient to Decrease Adipocyte Size by Inducing Mitochondrial Function. NANO LETTERS 2024; 24:1642-1649. [PMID: 38278518 PMCID: PMC10854752 DOI: 10.1021/acs.nanolett.3c04352] [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: 11/13/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/28/2024]
Abstract
Excess fat accumulation is not only associated with metabolic diseases but also negatively impacts physical appearance and emotional well-being. Bile acid, the body's natural emulsifier, is one of the few FDA-approved noninvasive therapeutic options for double chin (submental fat) reduction. Synthetic sodium deoxycholic acid (NaDCA) causes adipose cell lysis; however, its side effects include inflammation, bruising, and necrosis. Therefore, we investigated if an endogenous bile acid, chenodeoxycholic acid (CDCA), a well-known signaling molecule, can be beneficial without many of the untoward effects. We first generated CDCA-loaded nanoparticles to achieve sustained and localized delivery. Then, we injected them into the subcutaneous fat depot and monitored adipocyte size and mitochondrial function. Unlike NaDCA, CDCA did not cause cytolysis. Instead, we demonstrate that a single injection of CDCA-loaded nanoparticles into the subcutaneous fat reduced the adipocyte size by promoting fat burning and mitochondrial respiration, highlighting their potential for submental fat reduction.
Collapse
Affiliation(s)
- Weinan Zhou
- Department
of Molecular and Integrative Physiology, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Benjamin Lew
- Department
of Electrical and Computer Engineering, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Hyungsoo Choi
- Department
of Electrical and Computer Engineering, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Kyekyoon Kim
- Department
of Electrical and Computer Engineering, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
- Department
of Bioengineering, University of Illinois
Urbana−Champaign, Urbana, Illinois 61801, United States
- Beckman
Institute for Advanced Science and Technology, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Sayeepriyadarshini Anakk
- Department
of Molecular and Integrative Physiology, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
- Beckman
Institute for Advanced Science and Technology, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
- Division
of Nutritional Sciences, University of Illinois
Urbana−Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
49
|
Ma B, Li Q, Mi Y, Zhang J, Tan W, Guo Z. pH-responsive nanogels with enhanced antioxidant and antitumor activities on drug delivery and smart drug release. Int J Biol Macromol 2024; 257:128590. [PMID: 38056756 DOI: 10.1016/j.ijbiomac.2023.128590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/12/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
pH-responsive nanogels have played an increasingly momentous role in tumor treatment. The focus of this study is to design and develop pH-responsive benzimidazole-chitosan quaternary ammonium salt (BIMIXHAC) nanogels for the controlled release of doxorubicin hydrochloride (DOX) while enhancing its hydrophilicity. BIMIXHAC is crosslinked with carboxymethyl chitosan (CMC), hyaluronic acid sodium salt (HA), and sodium alginates (SA) using an ion crosslinking method. The chemical structure of chitosan derivatives was verified by 1H NMR and FT-IR techniques. Compared to hydroxypropyl trimethyl ammonium chloride chitosan (HACC)-based nanogels, BIMIXHAC-based nanogels exhibit better drug encapsulation efficiency and loading capacity (BIMIXHAC-D-HA 91.76 %, and 32.23 %), with pH-responsive release profiles and accelerated release in vitro. The series of nanogels formed by crosslinking with three different polyanionic crosslinkers have different particle size potentials and antioxidant properties. BIMIXHAC-HA, BIMIXHAC-SA and BIMIXHAC-CMC demonstrate favorable antioxidant capability. In addition, cytotoxicity tests showed that BIMIXHAC-based nanogels have high biocompatibility. BIMIXHAC-based nanogels exhibit preferable anticancer effects on MCF-7 and A549 cells. Furthermore, the BIMIXHAC-D-HA nanogel was 2.62 times less toxic than DOX to L929 cells. These results suggest that BIMIXHAC-based nanogels can serve as pH-responsive nanoplatforms for the delivery of anticancer drugs.
Collapse
Affiliation(s)
- Bing Ma
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Qing Li
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Yingqi Mi
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Jingjing Zhang
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Wenqiang Tan
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Zhanyong Guo
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| |
Collapse
|
50
|
Liu R, Zhang Z, Liu L, Li X, Duan R, Ren Y, Du B, Zhang Q, Zhou Z. The effects of stiffness on the specificity and avidity of antibody-coated microcapsules with target cells are strongly shape dependent. Colloids Surf B Biointerfaces 2024; 234:113752. [PMID: 38219638 DOI: 10.1016/j.colsurfb.2024.113752] [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: 10/19/2023] [Revised: 12/29/2023] [Accepted: 01/07/2024] [Indexed: 01/16/2024]
Abstract
Antibody modification is a common method for endowing drug carriers with the ability to target specific cells. Recent studies suggest that the efficacy of these antibody-modified drug carriers is closely related to their physicochemical properties, such as size, shape, stiffness, charge, and surface chemistry. In this study, we functionalized microcapsules with antibodies to investigate the combined effect of shape and stiffness on their targeting ability. We synthesized hollow microcapsules, both spherical and rod-shaped, with adjustable stiffness using calcium carbonate particles as templates and silk fibroin (SF) as the shell material. These microcapsules were then functionalized with trastuzumab (TTZ) to enhance targeting capabilities. Our analysis revealed that increasing stiffness significantly improved the specificity and avidity of TTZ-coated rod-shaped microcapsules, but not spherical ones, indicating a strong shape-dependent influence of stiffness on these properties. Additionally, we explored the mechanisms of endocytosis using various inhibitors and found that both macropinocytosis and clathrin played critical roles in the cellular uptake of microcapsules. Furthermore, we loaded microcapsules with doxorubicin (DOX) to evaluate their anti-tumor efficacy. The stiffest TTZ-coated, DOX-loaded rod-shaped microcapsules demonstrated the most potent anti-tumor effects on BT-474 cells and the highest uptake in BT-474 3D spheroids. This research contributes to the development of more effective microcapsule-based target delivery systems and the realization of the full potential of microcapsule drug delivery systems.
Collapse
Affiliation(s)
- Rui Liu
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Zhe Zhang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmaceutical Sciences; Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Lingrong Liu
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Xuemin Li
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Ruiping Duan
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Ying Ren
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Bo Du
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Qiqing Zhang
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China; Fujian Bote Biotechnology Co. Ltd, Fuzhou, Fujian 350013, China; Institute of Biomedical Engineering, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China.
| | - Zhimin Zhou
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|