1
|
Cunha J, Latocheski E, Fidalgo ACD, Gerola AP, Marin CFDF, Ribeiro AJ. Core-shell hybrid liposomes: Transforming imaging diagnostics and therapeutic strategies. Colloids Surf B Biointerfaces 2025; 251:114597. [PMID: 40043539 DOI: 10.1016/j.colsurfb.2025.114597] [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: 01/09/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 04/15/2025]
Abstract
For the last few years, researchers and industry have intensified efforts to develop a diverse array of diagnostic and therapeutic approaches to fight diseases such as cancer, diabetes, and viral infections. Among the emerging technologies, hybrid liposomes (HLs) stand out for their ability to address key limitations of conventional liposomes and deliver multifunctional solutions more effectively. While several novel nanosystems, including polymerlipid conjugates and inorganic nanoparticles (NPs), have shown great potential in the preclinical and clinical phases for the diagnosis and treatment of diseases, particularly cancer, HLs can integrate the best of both worlds, combining drug delivery properties with imaging capabilities. HLs, particularly those with core-shell structures, can surpass conventional liposomes by offering improved physicochemical properties, multifunctionality, and the capacity to overcome critical delivery challenges. The integration of natural and synthetic polymers has rapidly emerged as a preferred strategy in the development of HLs, providing significant advantages, such as enhanced stability, stimuli-responsive drug release, prolonged circulation, and improved therapeutic efficacy. Additionally, the customizable structure of HLs allows the incorporation of diverse materials, such as metals, ligands, and functional lipids, improving diagnosis and enhancing targeted delivery and cellular uptake far beyond what conventional liposomes offer. This review provides a critical and updated analysis of core-shell structure exhibiting HLs, with a focus on their preparation, characterization, and functional enhancements. We also examine in vitro/in vivo outcomes in imaging diagnosis and drug delivery while addressing the current barriers to clinical translation and future prospects for these versatile nanoplatforms.
Collapse
Affiliation(s)
- Joana Cunha
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra 3000-548, Portugal
| | - Eloah Latocheski
- Department of Chemistry, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | | | | | | | - António José Ribeiro
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra 3000-548, Portugal; Group Genetics of Cognitive Dysfunction, I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto 4169-007, Portugal.
| |
Collapse
|
2
|
Yu D, Liu M, Ding Q, Wu Y, Wang T, Song L, Li X, Qian K, Cheng Z, Gu M, Li Z. Molecular imaging-guided diagnosis and treatment integration for brain diseases. Biomaterials 2025; 316:123021. [PMID: 39705925 DOI: 10.1016/j.biomaterials.2024.123021] [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: 09/02/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
In practical clinical scenarios, improved diagnostic methods have been developed for the precise visualization of molecular targets using molecular imaging in brain diseases. Recently, the introduction of innovative molecular imaging modalities across both macroscopic and mesoscopic dimensions, with remarkable specificity and spatial resolution, has expanded the scope of applications beyond diagnostic testing, with the potential to guide therapeutic interventions, offering real-time feedback in the context of brain therapy. The molecular imaging-guided integration of diagnosis and treatment holds the potential to revolutionize disease management by enabling the real-time monitoring of treatment responses and therapy adjustments. Given the vibrant and ever-evolving nature of this field, this review provides an integrated picture on molecular image-guided diagnosis and treatment integration for brain diseases involving the basic concepts, significant breakthroughs, and recent trends. In addition, based on the current achievements, some critical challenges are also discussed.
Collapse
Affiliation(s)
- Donghu Yu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Menghao Liu
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul, 02841, South Korea.
| | - Youxian Wu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tianqing Wang
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Litong Song
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaoyu Li
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kun Qian
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Meijia Gu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Zhiqiang Li
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
3
|
Ijaz M, Hasan I, Aslam B, Yan Y, Zeng W, Gu J, Jin J, Zhang Y, Wang S, Xing L, Guo B. Diagnostics of brain tumor in the early stage: current status and future perspectives. Biomater Sci 2025. [PMID: 40200902 DOI: 10.1039/d4bm01503g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Early diagnosis of brain tumors is challenging due to their complexity and delicate structure. Conventional imaging techniques like MRI, CT, and PET are unable to provide detailed visualization of early-stage brain tumors. Early-stage detection of brain tumors is vital for enhancing patient outcomes and survival rates. So far, several scientists have dedicated their efforts to innovating advanced diagnostic probes to efficiently cross the BBB and selectively target brain tumors for optimal imaging. The integration of these techniques presents a viable pathway for non-invasive, accurate, and early-stage tumor identification. Herein, we provide a timely update on the various imaging probes and potential challenges for the diagnosis of early-stage brain tumors. Furthermore, this review highlights the significance of integrating advanced imaging probes for improving the early detection of brain tumors, ultimately enhancing treatment outcomes. Hopefully, this review will stimulate the interest of researchers to accelerate the development of new imaging probes and even their clinical translation for improving the early diagnosis of brain tumors.
Collapse
Affiliation(s)
- Muhammad Ijaz
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| | - Ikram Hasan
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Bilal Aslam
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Yuqian Yan
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| | - Wenjun Zeng
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| | - Jingsi Gu
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen 518055, China
| | - Jian Jin
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yinghe Zhang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| | - Shaohua Wang
- Diagnostic Center of Infectious Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China.
| | - Lu Xing
- Department of Sleep Medicine, Shenzhen Kangning Hospital, No. 1080 Cuizhu Road, Guangdong 518020, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| |
Collapse
|
4
|
Lu C, Bao Y, Fei Z, Wang Z, Ma J, Ren R, Xu X, Zhang Y. Multielement Doping Engineered Iron Oxide Nanoparticles: Enabling the Shift from Negative to Positive MRI Contrast for Enhanced Diagnostic Precision. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410414. [PMID: 39981967 DOI: 10.1002/smll.202410414] [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: 11/04/2024] [Revised: 01/23/2025] [Indexed: 02/22/2025]
Abstract
Contrast-enhanced magnetic resonance imaging (CE-MRI) is a crucial tool for the diagnosis and management of various diseases globally. Iron oxide nanoparticles with sizes less than 5 nm are expected to address the long-term toxicity and brain accumulation issues associated with clinical gadolinium-based T1 contrast agents (GBCAs) due to their non-toxicity and biodegradability. However, synthesizing sub-5-nanometer particles presents significant challenges that complicate their clinical translation. Herein, traditional iron oxide-based negative (T2) agents into positive (T1) agents are transformed and an all-in-one multielement doping strategy is developed. Multiple elements into iron oxide crystals are introduced to form multielement doping engineered iron oxide nanoparticles (MDE-IONPs) and their surfaces with flexible hydrophilic ligands are subsequently modified. It is shown that Ni (II) and Gd (III) doping engineered nanoparticles can effectively enhance imaging efficacy, reducing clearance rates, and enabling controlled synthesis. Ultimately, the implementation of Ni (II) and Gd (III) co-engineering yield longitudinal relaxivity of up to 14.7 mM-1s-1 even for particles as large as 9 nm, an improvement of approximately 300% over GBCAs. Combined with the stability, biosafety, both in vitro and in vivo results suggest that all-in-one multielement doping is a favorable strategy for advancing the development of next-generation safe MRI contrast agents.
Collapse
Affiliation(s)
- Chichong Lu
- Department of Chemistry, College of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, 100048, China
| | - Yingjie Bao
- Department of Chemistry, College of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, 100048, China
| | - Zihan Fei
- Department of Chemistry, College of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, 100048, China
| | - Zhijie Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junjie Ma
- Department of Chemistry, College of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, 100048, China
| | - Ruimin Ren
- Department of Chemistry, College of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, 100048, China
| | - Xue Xu
- Department of Chemistry, College of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, 100048, China
| | - Yang Zhang
- Department of Chemistry, College of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, 100048, China
| |
Collapse
|
5
|
Lu Z, Yan J, Zeng J, Zhang R, Xu M, Liu J, Sun L, Zu G, Chen X, Zhang Y, Pei R, Cao Y. Time-resolved T 1 and T 2 contrast for enhanced accuracy in MRI tumor detection. Biomaterials 2025; 321:123313. [PMID: 40187097 DOI: 10.1016/j.biomaterials.2025.123313] [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: 01/26/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Stimuli-responsive contrast agents (CAs) have shown great promise in enhancing magnetic resonance imaging (MRI) for more accurate tumor diagnosis. However, current CAs still face challenges in achieving high accuracy due to their low specificity and contrast signals being confounded by potential endogenous MRI artifacts. Herein, an extremely small iron oxide nanoparticle (ESIONP)-based smart responsive MRI contrast agent (LESPH) is proposed, which is meticulously designed with sequential dual biochemical stimuli-initiated, time-resolved T1 and T2 contrast presentation, ensuring high tumor specificity while minimizing interference from endogenous artifacts. LESPH is constructed using emulsion solvent evaporation by assembling poly(2-(hexamethyleneimino) ethyl methacrylate) terminally conjugated with a disulfide bond-linked catechol group (DSPH)-modified ESIONPs, with lauryl betaine serving as a surfactant. When LESPH undergoes sequential responses to the weak acidity and high-concentration glutathione (GSH) in the tumor microenvironment, it experiences an extremely rapid transition from sparse ESIONP assemblies to dispersed ESIONPs, followed by a slower transition to closely aggregated ones, concomitantly providing distinguishable brightening and darkening contrast enhancement at the tumor location on different time scales. By virtue of its sequential dual responsiveness and time-resolved distinguishable contrast enhancements, LESPH successfully detects tumors with extremely high accuracy, providing a novel paradigm for the precise medical diagnosis of cancer.
Collapse
Affiliation(s)
- Zhongzhong Lu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230000, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jincong Yan
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230000, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jianxian Zeng
- Department of Radiology, First Affiliated Hospital of Soochow University, Suzhou, 215026, China
| | - Ruihao Zhang
- Department of Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, 215026, China
| | - Mingsheng Xu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jihuan Liu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Lina Sun
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Guangyue Zu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xiaomin Chen
- Department of Stomatology, Traditional Chinese Medicine Hospital of Kunshan, Kunshan, 215300, China
| | - Ye Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230000, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yi Cao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230000, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| |
Collapse
|
6
|
Moiseeva EO, Skribitsky VA, Finogenova YA, German SV, Shpakova KE, Sergeev IS, Terentyeva DA, Sindeeva OA, Kulikov OA, Lipengolts AA, Grigorieva EY, Gorin DA. Ultrasmall maghemite nanoparticles as MRI contrast agent: Unique combination of aggregation stability, low toxicity, and tumor visualization. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2025; 65:102811. [PMID: 40024489 DOI: 10.1016/j.nano.2025.102811] [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: 12/26/2024] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 03/04/2025]
Abstract
Iron oxide nanoparticles are a promising candidate for the dual-mode MRI contrast agent, however most of them have limited circulation time and predominant negative contrast. We developed citric acid stabilized superparamagnetic maghemite nanoparticles (CA-SPMNs) with size 3.2 ± 0.7 nm with intense positive contrast. Co-precipitation reactions under well-controlled conditions in the automatic chemical reactor have carried out the synthesis. We found an encouraging correlation between aggregate formation kinetics in biological media and in vitro cytotoxicity results and in vivo circulation time. A cytotoxicity test showed the mouse fibroblast viability over 80 % for iron doses exceeding 1 mg/mL. CA-SPMNs have a low r2/r1 ratio, exhibiting positive contrast. Using in vivo MRI we demonstrated that CA-SPMNs circulate in the blood for 12-24 h, enabling blood vessel and tumor visualization, and partial renal clearance. Finally, CA-SPMNs show promise as effective MRI contrast agents, enabling differentiation between normal and pathological tissues.
Collapse
Affiliation(s)
- Ekaterina O Moiseeva
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, 143026 Moscow, Russia
| | | | - Yulia A Finogenova
- N.N. Blokhin National Medical Research Center of Oncology, 115522 Моscow, Russia
| | - Sergei V German
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, 143026 Moscow, Russia.
| | - Kristina E Shpakova
- N.N. Blokhin National Medical Research Center of Oncology, 115522 Моscow, Russia
| | - Igor S Sergeev
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, 143026 Moscow, Russia
| | - Daria A Terentyeva
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, 143026 Moscow, Russia
| | - Olga A Sindeeva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, 143026 Moscow, Russia
| | - Oleg A Kulikov
- Institute of Medicine, National Research Ogarev Mordovia State University, 430005 Saransk, Russia
| | - Alexey A Lipengolts
- N.N. Blokhin National Medical Research Center of Oncology, 115522 Моscow, Russia
| | - Elena Yu Grigorieva
- N.N. Blokhin National Medical Research Center of Oncology, 115522 Моscow, Russia
| | - Dmitry A Gorin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, 143026 Moscow, Russia
| |
Collapse
|
7
|
Wang Q, Gong Y, Li J, Luo D, Zeng X, Ling Y, Zhou Y, Chen Z. Topology-dependent T2 relaxivity in Fe 3O cluster-based MOFs for enhanced tumor monitoring via MRI. J Mater Chem B 2025. [PMID: 40163109 DOI: 10.1039/d4tb02858a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Metal-organic frameworks (MOFs) are crystalline porous materials with tunable structures, where metal ions or clusters serve as magnetic centers and organic ligands offer spatial separation. These characteristics, combined with their diverse topologies, make MOFs promising candidates for contrast agents (CAs) in magnetic resonance imaging (MRI). Herein we synthesized four MOFs based on the same triangular Fe3O clusters with different topologies: MIL-101(Fe) (moo net), MIL-100(Fe) (mtn net), MIL-59(Fe) (pcu net), and MIL-88B(Fe) (acs net). To clarify the relationship between topologies and T2 relaxivities, the MOFs were tailored into uniform, nanoscale spherical morphologies. Notably, the value of T2 relaxivity for MIL-88B(Fe) with acs topology is nearly three times that for MIL-101(Fe) with moo topology at 7.0 T. By comparing the magnetic properties of Fe3O molecular clusters and Ga-doped MIL-88B(Fe), our analysis demonstrated the significant advantage of MOFs with fixed arrays, adjustable components and diverse topologies in enhancing magnetic relaxation. Cellular MRI experiments further revealed that MIL-88B(Fe) could differentiate between M1 and M2 macrophages, highlighting its potential for monitoring tumor progression. These findings offer valuable insights into how MOF topology can be strategically utilized to enhance T2 relaxivities for MRI applications.
Collapse
Affiliation(s)
- Qiao Wang
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai, 200433, China.
| | - Yimin Gong
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai, 200433, China.
| | - Jianing Li
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai, 200433, China.
| | - Dan Luo
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai, 200433, China.
| | - Xin Zeng
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai, 200433, China.
| | - Yun Ling
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai, 200433, China.
| | - Yaming Zhou
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai, 200433, China.
| | - Zhenxia Chen
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
8
|
Jacinto C, Javed Y, Lavorato G, Tarraga WA, Conde BIC, Orozco JM, Picco AS, Garcia J, Dias CSB, Malik S, Sharma SK. Biotransformation and biological fate of magnetic iron oxide nanoparticles for biomedical research and clinical applications. NANOSCALE ADVANCES 2025:d5na00195a. [PMID: 40255989 PMCID: PMC12004083 DOI: 10.1039/d5na00195a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/15/2025] [Indexed: 04/22/2025]
Abstract
Safe implementation of nanotechnology-based products in biomedical applications necessitates an extensive understanding of the (bio)transformations that nanoparticles undergo in living organisms. The long-term fate in the body is a crucial consideration because it governs potential risks for human health. To accurately predict the life cycle of nanoparticles, their fate after administration into the body-including their (bio)transformations, persistence, and biodegradation-needs to be thoroughly evaluated. Magnetic iron oxide nanoparticles (MIONPs) can enter the body through various routes, including inhalation, ingestion, dermal absorption, and injection. Microscale and nanoscale studies are performed to observe nanomaterial biotransformations and their effect on clinically relevant properties. Researchers are utilizing high-resolution TEM for nanoscale monitoring of the nanoparticles while microscale follow-up approaches comprise quantification tools at the whole organism level and the molecular level. Nanoparticle-cell interactions, including cellular uptake and intracellular trafficking, are key to understanding nanoparticle accumulation in cells and organs. Prolonged accumulation may induce cell stress and nanoparticle toxicity, often mediated through oxidative stress and inflammation. In this review article, the journey of nanoparticles in the body is depicted and their biotransformations and final fate are discussed. Immunohistochemical techniques are particularly valuable in tracking nanoparticle distribution within tissues and assessing their impact at the cellular level. A thorough description of a wide range of characterization techniques is provided to unveil the fate and biotransformations of clinically relevant nanoparticles and to assist in their design for successful biomedical applications.
Collapse
Affiliation(s)
- Carlos Jacinto
- Nano-Photonics and Imaging Group, Institute of Physics, Universidade Federal de Alagoas 57072-900 Maceió AL Brazil
| | - Yasir Javed
- Department of Physics, University of Agriculture Faisalabad Pakistan
| | - Gabriel Lavorato
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | - Wilson A Tarraga
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | | | - Juan Manuel Orozco
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | - Agustin S Picco
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | - Joel Garcia
- Department of Chemistry, De La Salle University Manila Philippines
| | - Carlos Sato Baraldi Dias
- Institute for Photon Science and Synchrotron Radiation (IPS), Karlsruhe Institute of Technology (KIT) Hermann-von-Helmholtz-Platz 1 Eggenstein-Leopoldshafen 76344 Germany
| | - Sonia Malik
- Physiology, Ecology & Environmental Laboratory (P2e), University of Orléans 45067 France
- Department of Biotechnology, Baba Farid College Bathinda 151001 India
| | - Surender Kumar Sharma
- Department of Physics, Central University of Punjab Bathinda 151401 India
- Department of Physics, Federal University of Maranhão São Luís 65080-805 Brazil
| |
Collapse
|
9
|
Yuan X, Yu H, Wang L, Uddin MA, Ouyang C. Nitroxide radical contrast agents for safe magnetic resonance imaging: progress, challenges, and perspectives. MATERIALS HORIZONS 2025; 12:1726-1756. [PMID: 39757847 DOI: 10.1039/d4mh00995a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Magnetic resonance imaging (MRI) is considered one of the most valuable diagnostic technologies in the 21st century. To enhance the image contrast of anatomical features, MRI contrast agents have been widely used in clinical MRI diagnosis, especially those based on gadolinium, manganese, and iron oxide. However, these metal-based MRI contrast agents show potential toxicity to patients, which urges researchers to develop novel MRI contrast agents that can replace metal-based MRI contrast agents. Metal-free nitroxide radical contrast agents (NRCAs) effectively overcome the shortcomings of metal-based contrast agents and also have many advantages, including good biocompatibility, prolonged systemic circulation time, and easily functionalized structures. Importantly, since NRCAs acquire MRI signals with standard tissue water 1H relaxation mechanisms, they have great potential to realize clinical translation among many metal-free MRI contrast agents. At present, NRCAs have been proposed as an effective substitute for metal-based MRI contrast agents. Herein, this review first briefly introduces NRCAs, including their composition, classification, mechanism of action, application performances and advantages. Then, this review highlights the progress of NRCAs, including small molecule-based NRCAs and polymer-based NRCAs. Finally, this review also discusses the challenges and future perspectives of NRCAs.
Collapse
Affiliation(s)
- Xunchun Yuan
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Md Alim Uddin
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Chenguang Ouyang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| |
Collapse
|
10
|
Wu C, Zhong J, Li J, Luo Y, Wang J, Zeng X, Mao J, Lu J, Xu J, Wu C, Wang Z. Facile construction of manganese-based contrast agent with high T 1 relaxivity for magnetic resonance imaging via flash technology-based self-assembly. Regen Biomater 2025; 12:rbaf009. [PMID: 40270577 PMCID: PMC12017619 DOI: 10.1093/rb/rbaf009] [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: 11/18/2024] [Revised: 01/24/2025] [Accepted: 02/21/2025] [Indexed: 04/25/2025] Open
Abstract
To address the limitations of low relaxivity and physiological toxicity in commercial gadolinium-based contrast agents for magnetic resonance imaging (MRI), a novel manganese chelate macromolecular system was developed using a flash nanopreparation technique. Herein, the approach applying an instantaneous fluid device incorporated gallic acid, dopamine and Mn2+ to perform in situ polymerization of dopamine and covalent binding with albumin in a nanoconfined environment. This controllable self-assembly process characterized by its scalability and reproducibility was suitable for industrial-scale production. Under optimized flow rates and material ratios, the synthesized ultrasmall protein-based system, Mn-GA@BSA@DA, exhibited excellent aqueous dispersion with an average size of approximately 18 nm, allowing for long-term lyophilized powder storage. More importantly, the nanosystem demonstrated superior MRI-T 1 relaxivity, significantly surpassing that of clinical gadopentetate dimeglumine, with a high value around 18.5 mM-1 s-1 and a low r 2/r 1 ratio (<5 at 3.0 T). Furthermore, this Mn-GA@BSA@DA contrast agent was endowed with tumor-targeting effects and a long MRI monitoring window period for the liver, gallbladder and renal tubules. The metal chelation within the nanoagent minimizes Mn2+ release; importantly, the antioxidant components, gallic acid and dopamine, significantly inhibit the Fenton reaction-induced toxicity, enhancing biocompatibility. Therefore, this study presents a simple and scalable production technique for a kind of MRI-T 1-weighted contrast agent with high relaxivity and biocompatibility, offering a promising alternative to commercial Gd chelates.
Collapse
Affiliation(s)
- Chunwei Wu
- School of Materials Science and Engineering, Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Jie Zhong
- Medical Imaging Key Laboratory of Sichuan Province and School of Medical Imaging, North Sichuan Medical College, Nanchong 637000, P. R. China
| | - Jianing Li
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Yande Luo
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511495, P. R. China
| | - Junyao Wang
- School of Materials Science and Engineering, Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Xiaodie Zeng
- School of Materials Science and Engineering, Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Jiaji Mao
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Jianping Lu
- School of Materials Science and Engineering, Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Junyao Xu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, P. R. China
| | - Changqiang Wu
- Medical Imaging Key Laboratory of Sichuan Province and School of Medical Imaging, North Sichuan Medical College, Nanchong 637000, P. R. China
| | - Zhiyong Wang
- School of Materials Science and Engineering, Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, P. R. China
| |
Collapse
|
11
|
Zhao LX, Fan YG, Zhang X, Li C, Cheng XY, Guo F, Wang ZY. Graphdiyne biomaterials: from characterization to properties and applications. J Nanobiotechnology 2025; 23:169. [PMID: 40038692 DOI: 10.1186/s12951-025-03227-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/11/2025] [Indexed: 03/06/2025] Open
Abstract
Graphdiyne (GDY), the sole synthetic carbon allotrope with sp-hybridized carbon atoms, has been extensively researched that benefit from its pore structure, fully conjugated surfaces, wide band gaps, and more reactive C≡C bonds. In addition to the intrinsic features of GDY, engineering at the nanoscale, including metal/transition metal ion modification, chemical elemental doping, and other biomolecular modifications, endowed GDY with a broader functionality. This has led to its involvement in biomedical applications, including enzyme catalysis, molecular assays, targeted drug delivery, antitumor, and sensors. These promising research developments have been made possible by the rational design and critical characterization of GDY biomaterials. In contrast to other research areas, GDY biomaterials research has led to the development of characterization techniques and methods with specific patterns and some innovations based on the integration of materials science and biology, which are crucial for the biomedical applications of GDY. The objective of this review is to provide a comprehensive overview of the biomedical applications of GDY and the characterization techniques and methods that are essential in this process. Additionally, a general strategy for the biomedical research of GDY will be proposed, which will be of limited help to researchers in the field of GDY or nanomedicine.
Collapse
Affiliation(s)
- Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| | - Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| | - Xue Zhang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Chan Li
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| | - Xue-Yan Cheng
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| | - Feng Guo
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China.
| |
Collapse
|
12
|
Zhang Y, Lu Z, Guo J, Wang Q, Zhang X, Yang H, Li X. Advanced Carriers for Precise Delivery and Therapeutic Mechanisms of Traditional Chinese Medicines: Integrating Spatial Multi-Omics and Delivery Visualization. Adv Healthc Mater 2025; 14:e2403698. [PMID: 39828637 DOI: 10.1002/adhm.202403698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/01/2024] [Indexed: 01/22/2025]
Abstract
The complex composition of traditional Chinese medicines (TCMs) has posed challenges for in-depth study and global application, despite their abundance of bioactive compounds that make them valuable resources for disease treatment. To overcome these obstacles, it is essential to modernize TCMs by focusing on precise disease treatment. This involves elucidating the structure-activity relationships within their complex compositions, ensuring accurate in vivo delivery, and monitoring the delivery process. This review discusses the research progress of TCMs in precision disease treatment from three perspectives: spatial multi-omics technology for precision therapeutic activity, carrier systems for precise in vivo delivery, and medical imaging technology for visualizing the delivery process. The aim is to establish a novel research paradigm that advances the precision therapy of TCMs.
Collapse
Affiliation(s)
- Yusheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| | - Zhiguo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Jing Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Qing Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, P. R. China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Hongjun Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, China Academy of Chinese Medical Sciences, Beijing, 100029, P. R. China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| |
Collapse
|
13
|
Asad S, Ahl D, Suárez-López YDC, Erdélyi M, Phillipson M, Teleki A. Click Chemistry-Based Bioconjugation of Iron Oxide Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407883. [PMID: 39924809 PMCID: PMC11922026 DOI: 10.1002/smll.202407883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/19/2025] [Indexed: 02/11/2025]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) exhibit unique properties for diverse biomedical applications, including drug delivery and diagnostic imaging. Actively targeted SPIONs enhance delivery to diseased sites, reducing side effects and enhancing treatment efficacy. However, the development of reproducible functionalization protocols is challenged by the erratic behavior of nanoparticles in suspensions, such as agglomeration and sedimentation. In this study, a functionalization method is developed and systematically optimized to attach the Fc-region of antibodies onto silica-coated SPIONs via click chemistry, ensuring controlled ligand orientation on the particle surface. The synthesis and successive modifications of silica-coated SPIONs with organic moieties are presented resulting in the final click conjugation with antibodies targeting intercellular adhesion molecule 1 (ICAM1). This protein is upregulated on epithelial cell surfaces during gastrointestinal inflammation. Thermogravimetric analysis and infrared spectroscopy confirm successful SPION functionalization after each modification step. Cell viability assessment indicates no adverse effects of bioconjugated particles. Quantitative elemental analysis reveals significantly higher iron concentration in inflammation-induced Caco-2 cells exposed to ICAM1-modified particles compared to non-conjugated counterparts. Furthermore, laser scanning confocal microscopy of these cells suggests surface interaction and internalization of bioconjugated SPIONs, underscoring their potential for targeted imaging and therapy in inflammatory diseases.
Collapse
Affiliation(s)
- Shno Asad
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, SE-75123, Sweden
| | - David Ahl
- Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, Uppsala, SE-75123, Sweden
| | | | - Máté Erdélyi
- Department of Chemistry - BMC, Uppsala University, Uppsala, SE-75123, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, Uppsala, SE-75123, Sweden
| | - Alexandra Teleki
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, SE-75123, Sweden
| |
Collapse
|
14
|
Wang Y, Ma X, Zhang Y, Yang Y, Wang P, Chen T, Gao C, Dong C, Zheng J, Wu A. Insights into Non-Metallic Magnetic Resonance Imaging Contrast Agents: Advances and Perspectives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2411875. [PMID: 39901535 DOI: 10.1002/smll.202411875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/15/2025] [Indexed: 02/05/2025]
Abstract
Traditional metal-based magnetic resonance imaging contrast agents (MRI CAs), such as gadolinium, iron, and manganese, have made significant advancements in diagnosing major diseases. However, their potential toxicity due to long-term accumulation in the brain and bones raises safety concerns. In contrast, non-metallic MRI CAs, which can produce a nuclear magnetic resonance effect, show great promise in MRI applications due to their adaptable structure and function, good biocompatibility, and excellent biodegradability. Nevertheless, the development of non-metallic MRI CAs is slow due to the inherent low magnetic sensitivity of organic compounds, their rapid metabolism, and susceptibility to reduction. Designing effective multifunctional organic compounds for high-sensitivity MRI remains a challenge. In this discussion, the mechanisms of various non-metallic MRI CAs are explored and an overview of their current status, highlighting both their advantages and potential drawbacks, is provided. The key strategies for creating high-performance MRI CAs are summarized and how different synthetic approaches affect the performance of non-metallic MRI Cas is evaluated. Last, the challenges and future prospects for these promising non-metallic MRI CAs are addressed.
Collapse
Affiliation(s)
- Yanan Wang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Cixi, 315300, China
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Xuehua Ma
- Cixi Biomedical Research Institute, Wenzhou Medical University, Cixi, 315300, China
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yunhao Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Yanqiang Yang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Pengyu Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianxiang Chen
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Changyong Gao
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Chen Dong
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Jianjun Zheng
- Department of Radiology, Ningbo No.2 Hospital, Ningbo, 3l5010, China
| | - Aiguo Wu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Cixi, 315300, China
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| |
Collapse
|
15
|
Wang Z, Wang C, Ji Y, Yang M, Li C, Li M, Yang J, Tang H, Luo X, Hao H, Liu Z, Chen K, Chang Y, Yuan H, Feng L, Xing G, Li J. Magnetically driven bionic nanorobots enhance chemotherapeutic efficacy and the tumor immune response via precise targeting. Innovation (N Y) 2025; 6:100777. [PMID: 39991478 PMCID: PMC11846086 DOI: 10.1016/j.xinn.2024.100777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 12/23/2024] [Indexed: 02/25/2025] Open
Abstract
We developed magnetically driven bionic drug-loaded nanorobots (MDNs) to accurately target tumors and deliver chemotherapy agents using a customized three-dimensional (3D) magnetic manipulation platform (MMP) system to precisely control their movement mode. MDNs were based on polyethylene glycol-modified homogeneous ultrasmall iron oxide nanoparticles (7.02 ± 0.18 nm). Doxorubicin (12% ± 2% [w/w]) was encapsulated in MDNs by an imide bond. MDNs could imitate the movement mode of a school of wild herrings (e.g., re-dispersion/arrangement/vortex/directional movement) to adapt to the changing and complex physiological environment through the 3D MMP system. MDNs overcame blood flow resistance and biological barriers using optimized magnetic driving properties according to in vivo imaging (magnetic resonance imaging and fluorescence) and histopathology. The performance of fabricated MDNs was verified through cells and tumor-bearing mouse models. The MDNs showed high efficiency of drug delivery and targeting at the tumor site (>10-fold), lower toxicity than free doxorubicin (5 mg/kg body weight), activated immune response in the tumor site, and significantly lengthened survival for mice. The synergistic interaction between MDNs and the 3D MMP system underscores the immense potential of this drug delivery system, indicating a potential revolution in the field of tumor chemotherapy.
Collapse
Affiliation(s)
- Zhijie Wang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chutian Wang
- School of Mechanical Engineering and Automation, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China
| | - Ying Ji
- Institute of Textiles and Clothing, School of Fashion and Textiles, Research Institute for Intelligent Wearable Systems, The Hong Kong Polytechnic University, Hong Kong SAR 999077, China
| | - Mingxin Yang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chan Li
- School of Mechanical Engineering and Automation, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China
| | - Mengyao Li
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingru Yang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongyu Tang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianwei Luo
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haoyang Hao
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhicai Liu
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kui Chen
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanan Chang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Yuan
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Feng
- School of Mechanical Engineering and Automation, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China
| | - Gengmei Xing
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juan Li
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
16
|
Innuan P, Kongkarnka S, Thongtharb A, Kantapan J, Dechsupa N. Iron(III)-Quercetin Complex: In Vivo Acute Toxicity and Biodistribution of Novel MRI Agent. Int J Nanomedicine 2025; 20:1303-1320. [PMID: 39906526 PMCID: PMC11792624 DOI: 10.2147/ijn.s496015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/24/2025] [Indexed: 02/06/2025] Open
Abstract
Background The iron(III)-quercetin complex, known as "IronQ", is an innovative MRI contrast agent composed of one Fe(III) ion and two quercetin molecules. IronQ is efficiently internalized by cells, enabling T1-weighted MRI tracking. It has demonstrated therapeutic benefits in reducing inflammation in an intracerebral hemorrhage (ICH) mouse model and offers a safer alternative to gadolinium-based agents by avoiding cytotoxicity and genotoxicity. These properties make IronQ a promising candidate for safe and effective MRI contrast enhancement. Purpose This study aims to further the development of IronQ as an MRI contrast agent by investigating its biodistribution, pharmacokinetics, and acute toxicity in a preclinical animal model. Methods The relaxivity of IronQ was measured in water and whole blood phantoms. Acute toxicity was evaluated in Sprague Dawley rats administered single intraperitoneal doses of IronQ (75, 150, and 225 µmol Fe/kg BW) over a 14-day period. Pharmacokinetic studies were performed at a dose of 150 µmol Fe/kg BW, with blood iron content analyzed using ICP-OES. For in vivo biodistribution, SD rats were administered an intravenous dose of IronQ (225 µmol Fe/kg BW), followed by MR imaging using a 1.5 T scanner and subsequent tissue-ICP analysis. Results The longitudinal relaxivity (r1) of IronQ was measured to be 2.17 mm⁻¹s⁻¹ in ultrapure water and 3.56 mm⁻¹s⁻¹ in whole blood. Acute toxicity studies showed no mortality, morbidity, or significant biochemical changes, with histopathology confirming no irreversible organ damage. Pharmacokinetics revealed peak blood iron content at 1.1 hours post-administration and clearance within 24 hours. MRI demonstrated enhanced T1 signal intensity, particularly in the liver and kidney. Conclusion These findings provide valuable insights into the safety, pharmacokinetics, and imaging efficacy of IronQ, highlighting its potential as a robust and biocompatible MRI contrast agent.
Collapse
Affiliation(s)
- Phattarawadee Innuan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sarawut Kongkarnka
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Atigan Thongtharb
- Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
17
|
Lu K, Zhang R, Wang H, Li C, Yang Z, Xu K, Cao X, Wang N, Cai W, Zeng J, Gao M. PEGylated Ultrasmall Iron Oxide Nanoparticles as MRI Contrast Agents for Vascular Imaging and Real-Time Monitoring. ACS NANO 2025; 19:3519-3530. [PMID: 39818797 DOI: 10.1021/acsnano.4c13356] [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: 01/19/2025]
Abstract
Accurate imaging evaluations of pre- and post-treatment of cardiovascular diseases are pivotal for effective clinical interventions and improved patient outcomes. However, current imaging methods lack real-time monitoring capabilities with a high contrast and resolution during treatments. This study introduces PEGylated ultrasmall iron oxide nanoparticles (PUSIONPs), which have undergone comprehensive safety evaluations, boasting an r1 value of 6.31 mM-1 s-1, for contrast-enhanced magnetic resonance angiography (MRA). Systematic comparisons against common clinical methods in rabbits reveal that PUSIONPs-enhanced MRA exhibited improved vascular contrast, clearer vascular boundaries, and superior vessel resolution. Moreover, owing to their nanosize, PUSIONPs demonstrate significantly prolonged blood circulation compared to small molecular contrast agents such as Magnevist and Ultravist. This extended circulation enables captivating real-time monitoring of thrombolysis treatment for up to 4 h in rabbit models postsingle contrast agent injection. Additionally, in larger animal models such as beagles and Bama minipigs, PUSIONPs-enhanced MRA also showcases superior contrast effects, boundary delineation, and microvessel visualization, underscoring their potential to transform cardiovascular imaging, particularly in real-time monitoring and high-resolution visualization during treatment processes.
Collapse
Affiliation(s)
- Kuan Lu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
- The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Ruru Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Hongzhao Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Cang Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Zhe Yang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
- The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Keyang Xu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Xiaoyi Cao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Ning Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Wu Cai
- The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Mingyuan Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
- The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
- School of life Sciences, Soochow University, Suzhou, 215123, China
| |
Collapse
|
18
|
Jyoti D, Reeves D, Gordon-Wylie S, Eskey C, Weaver J. Improving Stroke Treatment Using Magnetic Nanoparticle Sensors to Monitor Brain Thrombus Extraction. SENSORS (BASEL, SWITZERLAND) 2025; 25:672. [PMID: 39943310 PMCID: PMC11820568 DOI: 10.3390/s25030672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/12/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025]
Abstract
(1) Background: Mechanical thrombectomy (MT) successfully treats ischemic strokes by extracting the thrombus, or clot, using a stent retriever to pull it through the blood vessel. However, clot slippage and/or fragmentation can occur. Real-time feedback to a clinician about attachment between the stent and clot could enable more complete removal. We propose a system whereby antibody-targeted magnetic nanoparticles (NPs) are injected via a microcatheter to coat the clot, oscillating magnetic fields excite the particles, and a small coil attached to the catheter picks up a signal that determines the proximity of the clot to the stent. (2) Methods: We used existing simulation code to model the signal from NPs distributed on a hemispherical clot with three orthogonally applied magnetic fields. An in vitro apparatus was built that applied fields and read out signals from a 1.5 mm pickup coil at a variable distance and orientation angle from a sample of 100 nm iron oxide core/shell NPs. (3) Results: Our simulations suggest that the sum of the voltages induced in the pickup coil from three orthogonal applied fields could localize a clot to within 180 µm, regardless of the exact orientation of the pickup coil, with further precision added via rotation-correction formulae. Our experimental system validated simulations; we estimated an in vitro distance recovery precision of 41 µm with a pickup coil 1 mm from the clot. (4) Conclusions: Magnetic NP sensing could be a safe and real-time method to estimate whether a clot is attached to the stent retriever during MT.
Collapse
Affiliation(s)
- Dhrubo Jyoti
- LCD Nanotech, Hanover, NH 03755, USA;
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Daniel Reeves
- Fred Hutch Cancer Center, University of Washington, Seattle, WA 98195, USA;
| | - Scott Gordon-Wylie
- LCD Nanotech, Hanover, NH 03755, USA;
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Flow Aluminum, Albuquerque, NM 87123, USA
| | - Clifford Eskey
- Department of Radiology, Dartmouth Health, Lebanon, NH 03756, USA
| | - John Weaver
- LCD Nanotech, Hanover, NH 03755, USA;
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Department of Radiology, Dartmouth Health, Lebanon, NH 03756, USA
| |
Collapse
|
19
|
Navarrete-León C, Doherty A, Strimaite M, Bear JC, Olivo A, Endrizzi M, Patrick PS. Nanoparticle Contrast Agents for Dark-Field X-ray Imaging. NANO LETTERS 2025; 25:1036-1042. [PMID: 39601295 PMCID: PMC11760164 DOI: 10.1021/acs.nanolett.4c04878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
The poor soft tissue contrast of X-ray CT necessitates contrast agent use to improve diagnosis across disease applications, yet their poor detection sensitivity requires high injected doses, which restrict use in at-risk populations. Dark-field X-ray imaging is emerging as a more sensitive alternative to traditional attenuation-based imaging, leveraging scattered radiation to produce contrast. Yet aside from large, short-lived microbubbles, the alternate physics of dark-field detection has yet to be exploited for contrast agent development. Here we demonstrate that high-Z nanoparticles can provide a new means to producing dark-field image contrast, promoting scatter via a higher rather than lower electron density compared to microbubbles, increasing detection sensitivity compared to attenuation-based detection of a clinical iodine-based agent at an equivalent X-ray dose. As the use of dark-field X-ray imaging expands into more common clinical usage, this will support the development of a new class of nanoparticulate contrast agents.
Collapse
Affiliation(s)
- Carlos Navarrete-León
- Department
of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, United Kingdom
- X-ray
microscopy and tomography lab, The Francis
Crick Institute, London, NW1 1AT, United Kingdom
| | - Adam Doherty
- Department
of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, United Kingdom
- X-ray
microscopy and tomography lab, The Francis
Crick Institute, London, NW1 1AT, United Kingdom
| | - Margarita Strimaite
- Centre
for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
- UCL
School of Pharmacy, Faculty of Life Sciences, University College London, London, WC1N 1AX, United Kingdom
| | - Joseph C. Bear
- School
of Life Sciences, Pharmacy & Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, United Kingdom
| | - Alessandro Olivo
- Department
of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, United Kingdom
| | - Marco Endrizzi
- Department
of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, United Kingdom
- X-ray
microscopy and tomography lab, The Francis
Crick Institute, London, NW1 1AT, United Kingdom
| | - P. Stephen Patrick
- Centre
for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| |
Collapse
|
20
|
Na L, Song X, Luo P, Su J, Yao Z. Innovative applications of advanced nanomaterials in cerebrovascular imaging. Front Bioeng Biotechnol 2025; 12:1456704. [PMID: 39911816 PMCID: PMC11794002 DOI: 10.3389/fbioe.2024.1456704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/24/2024] [Indexed: 02/07/2025] Open
Abstract
Cerebrovascular imaging is essential for the diagnosis, treatment, and prognosis of cerebrovascular disease, including stroke, aneurysms, and vascular malformations. Conventional imaging techniques such as MRI, CT, DSA and ultrasound have their own strengths and limitations, particularly in terms of resolution, contrast and safety. Recent advances in nanotechnology offer new opportunities for improved cerebrovascular imaging. Nanomaterials, including metallic nanoparticles, magnetic nanoparticles, quantum dots, carbon-based nanomaterials, and polymer nanoparticles, show great potential due to their unique physical, chemical, and biological properties. This review summarizes recent advances in advanced nanomaterials for cerebrovascular imaging and their applications in various imaging techniques, and discusses challenges and future research directions. The aim is to provide valuable insights for researchers to facilitate the development and clinical application of these innovative nanomaterials in cerebrovascular imaging.
Collapse
Affiliation(s)
- Li Na
- Department of Neurology, Liaoning Provincial People’s Hospital, Shenyang, China
| | - Xiaofu Song
- Department of Neurology, Liaoning Provincial People’s Hospital, Shenyang, China
| | - Ping Luo
- Liaoning Provincial People’s Hospital, China Medical University, Shenyang, China
| | - Jingqi Su
- Liaoning Provincial People’s Hospital, China Medical University, Shenyang, China
| | - Zhicheng Yao
- Department of Neurology, Liaoning Provincial People’s Hospital, Shenyang, China
| |
Collapse
|
21
|
Nian L, Liu Z, Cai X, Wang B, Zhang Q, Lei J, Xiao J. A Single-Chain Peptide Probe Targeting Pathological Collagen for Precise Staging of Hepatic Fibrosis by MR Imaging. Anal Chem 2025; 97:1117-1124. [PMID: 39772523 DOI: 10.1021/acs.analchem.4c03601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Hepatic fibrosis, a chronic liver response to injury with potential severe outcomes like cirrhosis and liver cancer, necessitates urgent noninvasive diagnostic techniques to halt disease progression. We herein for the first time developed a single-chain peptide probe targeting pathological collagen for in vivo magnetic resonance imaging (MRI) of hepatic fibrosis. The novel (GhypO)10 probe, distinguished by its unique monomeric conformation achieved through Pro to (2S,4S)-hydroxyproline (hyp) substitution and subsequent disruption of hydrogen bonding, exhibits selectivity for pathological collagen over its intact counterpart in connective tissues. Fluorescence imaging of liver specimens from fibrotic models displayed a discernible relationship between pathological collagen levels and fibrosis stage. Moreover, T1-weighted MR images post Gd-GhypO administration revealed progressive signal enhancement congruent with fibrosis severity, corroborated by a corresponding increase in the contrast-to-noise ratio (ΔCNR). Biodistribution analysis indicates that Gd-GhypO has low Gd retention in the main organs 24 h postinjection, ensuring the probe's safety for molecular imaging. The Gd-GhypO probe therefore emerges as a potent tool for the precise, noninvasive delineation of hepatic fibrosis stages, offering significant implications for the diagnosis and management of liver fibrosis.
Collapse
Affiliation(s)
- Linge Nian
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Zhao Liu
- The First Hospital of Lanzhou University, Lanzhou 730000, P. R. China
| | - Xiangdong Cai
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Bo Wang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Qianqian Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P. R. China
| | - Junqiang Lei
- The First Hospital of Lanzhou University, Lanzhou 730000, P. R. China
| | - Jianxi Xiao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| |
Collapse
|
22
|
Luo T, Wang B, Chen R, Qi Q, Wu R, Xie S, Chen H, Han J, Wu D, Cao S. Research progress of nitroxide radical-based MRI contrast agents: from structure design to application. J Mater Chem B 2025; 13:372-398. [PMID: 39565110 DOI: 10.1039/d4tb02272f] [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: 11/21/2024]
Abstract
Magnetic resonance imaging (MRI) remains a cornerstone of diagnostic imaging, offering unparalleled insights into anatomical structures and pathological conditions. Gadolinium-based contrast agents have long been the standard in MRI enhancement, yet concerns over nephrogenic systemic fibrosis have spurred interest in metal-free alternatives. Nitroxide radical-based MRI contrast agents (NO-CAs) have emerged as promising candidates, leveraging their biocompatibility and imaging capabilities. This review summaries the latest advancements in NO-CAs, focusing on synthesis methodologies, influencing effects of structures of NO-CAs on relaxation efficiency and their applications across various clinical contexts. Comprehensive discussions encompass small molecular, polymeric, and nano-sized NO-CAs, detailing their unique properties and potential clinical utilities. Despite challenges, NO-CAs represent a dynamic area of research poised to revolutionize MRI diagnostics. This review serves as a critical resource for researchers and practitioners seeking to navigate the evolving landscape of MRI contrast agents.
Collapse
Affiliation(s)
- Tao Luo
- School of Biomedical Engineering, Sun Yat-Sen University of Shenzhen Campus, Shenzhen, China.
| | - Bo Wang
- School of Biomedical Engineering, Sun Yat-Sen University of Shenzhen Campus, Shenzhen, China.
| | - Runxin Chen
- Shenzhen University General Hospital, Shenzhen, China
| | - Qi Qi
- Shenzhen University General Hospital, Shenzhen, China
| | - Ruodai Wu
- Shenzhen University General Hospital, Shenzhen, China
| | - Shunzi Xie
- School of Biomedical Engineering, Sun Yat-Sen University of Shenzhen Campus, Shenzhen, China.
| | - Hanbing Chen
- School of Biomedical Engineering, Sun Yat-Sen University of Shenzhen Campus, Shenzhen, China.
| | - Jialei Han
- School of Biomedical Engineering, Sun Yat-Sen University of Shenzhen Campus, Shenzhen, China.
| | - Dalin Wu
- School of Biomedical Engineering, Sun Yat-Sen University of Shenzhen Campus, Shenzhen, China.
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, Sun Yat-Sen University, Shenzhen, China
| | | |
Collapse
|
23
|
Liu H, Zhen Z, Chen F, Chen J, Chen W. Advancements in Iron Oxide Nanoparticles for Multimodal Imaging and Tumor Theranostics. Curr Med Chem 2025; 32:301-321. [PMID: 39005127 DOI: 10.2174/0109298673301359240705063544] [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: 02/29/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 07/16/2024]
Abstract
The emergence of nanomedicine offers renewed promise in the diagnosis and treatment of diseases. Due to their unique physical and chemical properties, iron oxide nanoparticles (IONPs) exhibit widespread application in the diagnosis and treatment of various ailments, particularly tumors. IONPs have magnetic resonance (MR) T1/T2 imaging capabilities due to their different sizes. In addition, IONPs also have biocatalytic activity (nanozymes) and magnetocaloric effects. They are widely used in chemodynamic therapy (CDT), magnetic hyperthermia treatment (MHT), photodynamic therapy (PDT), and drug delivery. This review outlines the synthesis, modification, and biomedical applications of IONPs, emphasizing their role in enhancing diagnostic imaging (including single-mode and multimodal imaging) and their potential in cancer therapies (including chemotherapy, radiotherapy, CDT, and PDT). Furthermore, we briefly explore the challenges in the clinical application of IONPs, such as surface modification and protein adsorption, and put forward opinions on the clinical transformation of IONPs.
Collapse
Affiliation(s)
- He Liu
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhiming Zhen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fengxi Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiafei Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wei Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
24
|
Li X, Liu Q, Wu M, Wang H, Yang J, Mu X, Zhang XD. Artificially Engineered Nanoprobes for Ultrasensitive Magnetic Resonance Imaging. Adv Healthc Mater 2025; 14:e2403099. [PMID: 39562174 DOI: 10.1002/adhm.202403099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/11/2024] [Indexed: 11/21/2024]
Abstract
Magnetic resonance imaging (MRI) is a noninvasive and radiation-free technique used for soft tissue. However, there are some limitations of the MRI modality, such as low sensitivity and poor image resolution. Artificially engineered magnetic nanoprobes have been extensively explored as a versatile platform for ultrasensitive MRI contrast agents due to their unique physiochemical characteristics and tunable magnetic properties. In this review, the emphasis is on recent progress in MRI nanoprobes with different structures and elements, including gadolinium-, iron-, manganese-based and metal-free nanoprobes. The key influencing factors and advanced engineering strategies for modulating the relaxation ratio of MRI nanoprobes are systematically condensed. Furthermore, the widespread and noninvasive visualization applications of MRI nanoprobes for real time monitoring of major organs and accurate disease diagnosing, such as cerebrovascular, ischemia, Alzheimer's disease, liver fibrosis, whole-body tumors, inflammation, as well as multi-mode imaging applications are summarized. Finally, the challenges and prospects for the future development of MRI nanoprobes are discussed, and promising strategies are specifically emphasized for improving biocompatibility, precisely engineering of optimal size, AI-driven prediction and design, and multifunctional self-assembly to enhance diagnostics. This review will provide new inspiration for artificial engineering and nanotechnology-based molecular probes for medical diagnosis and therapy with ultrasensitive MRI.
Collapse
Affiliation(s)
- Xuyan Li
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Qingshan Liu
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Menglin Wu
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Hao Wang
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jiang Yang
- School of Medicine, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xiaoyu Mu
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Science, Tianjin University, Tianjin, 300072, China
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Science, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
25
|
Butt A, Bach H. Nanomedicine and clinical diagnostics part I: applications in conventional imaging (MRI, X-ray/CT, and ultrasound). Nanomedicine (Lond) 2025; 20:167-182. [PMID: 39661327 PMCID: PMC11731363 DOI: 10.1080/17435889.2024.2439776] [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: 09/08/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024] Open
Abstract
Integrating nanotechnologies in diagnostic imaging presents a promising step forward compared to traditional methods, which carry certain limitations. Conventional imaging routes, such as X-ray/computed tomography and magnetic resonance imaging, derive significant advantages from nanoparticles (NPs), which allow researchers and clinicians to overcome some of the limitations of traditional imaging agents. In this literature review, we explore recent advancements in nanomaterials being applied in conventional diagnostic imaging techniques by exploring relevant reviews and original research papers (e.g. experimental models and theoretical model studies) in the literature. Collectively, there are numerous nanomaterials currently being examined for use in conventional imaging modalities, and each imaging technique has unique NPs with properties that can be manipulated to answer an array of clinical questions specific to that imaging modality. There are still challenges to consider, including getting regulatory approval for clinical research and routine use about long-term biocompatibility, which collectively emphasize the need for continued research to facilitate the integration of nanotechnology into routine clinical practice. Most importantly, there is a continued need for strong, collaborative efforts between researchers, biomedical engineers, clinicians, and industry stakeholders, which are necessary to bridge the persistent gap between translational ideas and implementation in clinical settings.
Collapse
Affiliation(s)
- Ahmad Butt
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Horacio Bach
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Faculty of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
26
|
Solak K, Atiş M, Kasapoğlu AE, Karaman A, Mavi A. Metal Nanoparticles for Simultaneous Use in AC Magnetic Field Hyperthermia and Magnetic Resonance Imaging. J Biomed Mater Res A 2025; 113:e37817. [PMID: 39474677 DOI: 10.1002/jbm.a.37817] [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: 03/28/2024] [Revised: 09/19/2024] [Accepted: 10/07/2024] [Indexed: 12/26/2024]
Abstract
Magnetic nanoparticles (MNPs) are produced for both diagnosis and treatment due to their simultaneous availability in magnetic resonance imaging (MRI) and magnetic hyperthermia (MHT). Extensive investigations focus on developing MNPs for individual MHT or MRI applications, but the development of MNPs for theragnostic applications has received very little attention. In this study, through efficient examination of synthesis conditions such as metal precursors, reaction parameters, and solvent choices, we aimed to optimize MNP production for effective utilization for MHT and MRI simultaneously. MNPs were synthesized by thermal decomposition under 17 different conditions and deeply characterized by transmission electron microscopy (TEM), x-ray diffraction (XRD), and x-ray photoelectron spectroscopy (XPS). The heating efficiency of MNPs under an alternating current (AC) magnetic field was quantified, while MRI performance was evaluated through agar phantom experiments. Our findings highlight the crucial role of benzyl ether in metal ion reduction and size control. Metal-doped iron oxide MNPs displayed promise for MHT, whereas Mn-doped iron oxide MNPs exhibited enhanced MRI capabilities. Consequently, five engineered MNPs were considered potential candidates for further studies, demonstrating their dual ability in MRI and MHT.
Collapse
Affiliation(s)
- Kübra Solak
- Department of Nanoscience and Nanoengineering, Graduate School of Natural and Applied Sciences, Atatürk University, Erzurum, Türkiye
| | - Mustafa Atiş
- School of Medicine, Atatürk University, Erzurum, Türkiye
- School of Medicine, Ağrı İbrahim Çeçen University, Ağrı, Türkiye
| | - Ahmet Emre Kasapoğlu
- East Anatolia High Technology Application and Research Center, Atatürk University, Erzurum, Türkiye
| | - Adem Karaman
- Faculty of Medicine, Department of Radiology, Atatürk University, Erzurum, Türkiye
| | - Ahmet Mavi
- Department of Nanoscience and Nanoengineering, Graduate School of Natural and Applied Sciences, Atatürk University, Erzurum, Türkiye
- Department of Mathematics and Science Education, Kâzım Karabekir Faculty of Education, Atatürk University, Erzurum, Türkiye
| |
Collapse
|
27
|
Kaviani M, Geramizadeh B. Nanoparticles Perspective in Skin Tissue Engineering: Current Concepts and Future Outlook. Curr Stem Cell Res Ther 2025; 20:2-8. [PMID: 38284717 DOI: 10.2174/011574888x291345240110102648] [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: 11/05/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024]
Abstract
Nanotechnology seems to provide solutions to the unresolved complications in skin tissue engineering. According to the broad function of nanoparticles, this review article is intended to build a perspective for future success in skin tissue engineering. In the present review, recent studies were reviewed, and essential benefits and challenging issues regarding the application of nanoparticles in skin tissue engineering were summarized. Previous studies indicated that nanoparticles can play essential roles in the improvement of engineered skin. Bio-inspired design of an engineered skin structure first needs to understand the native tissue and mimic that in laboratory conditions. Moreover, a fundamental comprehension of the nanoparticles and their related effects on the final structure can guide researchers in recruiting appropriate nanoparticles. Attention to essential details, including the designation of nanoparticle type according to the scaffold, how to prepare the nanoparticles, and what concentration to use, is critical for the application of nanoparticles to become a reality. In conclusion, nanoparticles were applied to promote scaffold characteristics and angiogenesis, improve cell behavior, provide antimicrobial conditions, and cell tracking.
Collapse
Affiliation(s)
- Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bita Geramizadeh
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
28
|
Zuo X, Wang X, Si G, Zhang D, Yu X, Guo Z, Gu N. Size-Dependent Oxygen Vacancy of Iron Oxide Nanoparticles. SMALL METHODS 2025; 9:e2400685. [PMID: 39031906 DOI: 10.1002/smtd.202400685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/09/2024] [Indexed: 07/22/2024]
Abstract
Prior research has highlighted the reduction of iron oxide nanoparticle (IONPs) sizes to the "ultra-small" dimension as a pivotal approach in developing T1-MRI contrast agents, and the enhancement in T1 contrast performance with the reducing size is usually attributed to the increased specific surface area and weakened magnetization. Nonetheless, as the size decreases, the variation in surface defects, particularly oxygen vacancy (VO) defects, significantly impacts the T1 imaging efficacy. In this study, the VO on IONPs is meticulously investigated through XPS, Raman, and EPR spectroscopy. As the nanoparticle size decreased, the VO concentration rose initially but subsequently declined, with the peak concentration observed in the size of 8.27 nm. Further insights gained from synchrotron XAS analysis and DFT calculations indicate that both surface tension and phase transition in IONPs contribute to alterations in the Fe─O bond length, thereby influencing the VO formation energy across varying nanoparticle sizes. The MRI tests reveal that the VO in IONPs serve as pivotal sites for the attachment of water molecules to iron ions, and IONPs with fewer VO exhibited a deterioration in T1-MRI contrast effects. This research may provide a deeper understanding of the relationship between T1 contrast performance and the size of IONPs.
Collapse
Affiliation(s)
- Xudong Zuo
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Xinyu Wang
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou, 213100, P. R. China
| | - Guangxiang Si
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Dongmei Zhang
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou, 213100, P. R. China
| | - Xiaogang Yu
- Xinyu Key Laboratory of Materials Technology and Application for Intelligent Manufacturing, School of Mechanical and Electrical Engineering, Xinyu University, Xinyu, 338004, P. R. China
| | - Zhanhang Guo
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, 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
| |
Collapse
|
29
|
Zhang D, Zhang J, Bian X, Zhang P, Wu W, Zuo X. Iron Oxide Nanoparticle-Based T 1 Contrast Agents for Magnetic Resonance Imaging: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 15:33. [PMID: 39791792 PMCID: PMC11722098 DOI: 10.3390/nano15010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
This review highlights recent progress in utilizing iron oxide nanoparticles (IONPs) as a safer alternative to gadolinium-based contrast agents (GBCAs) for magnetic resonance imaging (MRI). It consolidates findings from multiple studies, discussing current T1 contrast agents (CAs), the synthesis techniques for IONPs, the theoretical principles for designing IONP-based MRI CAs, and the key factors that impact their T1 contrast efficacy, such as nanoparticle size, morphology, surface modifications, valence states, and oxygen vacancies. Furthermore, we summarize current strategies to achieve IONP-based responsive CAs, including self-assembly/disassembly and distance adjustment. This review also evaluates the biocompatibility, organ accumulation, and clearance pathways of IONPs for clinical applications. Finally, the challenges associated with the clinical translation of IONP-based T1 CAs are included.
Collapse
Affiliation(s)
- Dongmei Zhang
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Jing Zhang
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Xianglin Bian
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Pei Zhang
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Weihua Wu
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Xudong Zuo
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
- The Jiangsu Key Laboratory of Clean Energy Storage and Conversion, Jiangsu University of Technology, Changzhou 213100, China
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
30
|
Zhang P, Li Y, Li X, Wang Y, Lin H, Zhang N, Li W, Jing L, Jiao M, Luo X, Hou Y. Shedding light on vascular imaging: the revolutionary role of nanotechnology. J Nanobiotechnology 2024; 22:757. [PMID: 39695727 DOI: 10.1186/s12951-024-03042-x] [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: 03/07/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Vascular dysfunction, characterized by changes in anatomy, hemodynamics, and molecular expressions of vasculatures, is closely linked to the onset and development of diseases, emphasizing the importance of its detection. In clinical practice, medical imaging has been utilized as a significant tool in the assessment of vascular dysfunction, however, traditional imaging techniques still lack sufficient resolution for visualizing the complex microvascular systems. Over the past decade, with the rapid advancement of nanotechnology and the emergence of corresponding detection facilities, engineered nanomaterials offer new alternatives to traditional contrast agents. Compared with conventional small molecule counterparts, nanomaterials possess numerous advantages for vascular imaging, holding the potential to significantly advance related technologies. In this review, the latest developments in nanotechnology-assisted vascular imaging research across different imaging modalities, including contrast-enhanced magnetic resonance (MR) angiography, susceptibility-weighted imaging (SWI), and fluorescence imaging in the second near-infrared window (NIR-II) are summarized. Additionally, the advancements of preclinical and clinical studies related to these nanotechnology-enhanced vascular imaging approaches are outlined, with subsequent discussion on the current challenges and future prospects in both basic research and clinical translation.
Collapse
Affiliation(s)
- Peisen Zhang
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yao Li
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Xiaoqi Li
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Yudong Wang
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Hua Lin
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ni Zhang
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Wenyue Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lihong Jing
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Colloid and Interface and Thermodynamics, CAS Research/Education Center for Excellence in Molecular Sciences, Center for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China
| | - Mingxia Jiao
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China.
| | - Xiliang Luo
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China.
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| |
Collapse
|
31
|
Tan Y, Wang J, Wan Q, Yang J, Huang J, Zhou Z, Dong H, Zhang X. A switchable magnetic resonance imaging nanoplatform for in situ microRNA imaging. Chem Sci 2024; 16:199-204. [PMID: 39600495 PMCID: PMC11587794 DOI: 10.1039/d4sc04675g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Aberrant microRNA (miRNA) expression is associated with various types of carcinogenesis, making miRNA a promising candidate for diagnostic and therapeutic biomarkers. However, in situ miRNA diagnostics remains a significant challenge owing to the various biological barriers. Herein, we report a novel miRNA imaging probe consisting of PEG-polylysine-PNIPAM polymer matrix-modified small Fe3O4 (PAA-Fe3O4-DNA@PPP) nanoparticles with an improved circulatory half-life, efficient tissue permeability, and enhanced tumor accumulation, for in situ miRNA magnetic resonance imaging (MRI). In this strategy, we employed large size PAA-Fe3O4-DNA@PPP to improve circulatory time and utilized PEG-polylysine-PNIPAM as a GSH-responsive moiety to dissociate PAA-Fe3O4-DNA@PPP and release small size PAA-Fe3O4-DNA for enhanced tumor permeability. Specifically, the target miRNA acts as a cross-linker for PAA-Fe3O4-DNA, forming larger assemblies that not only amplify the MRI signal for detection but also enhance retention for prolonged observation. Both the in vitro and in vivo results validate that the imaging probe exhibits an enhanced MRI signal with 3.69-fold amplification for tumor interior miRNA detection, allowing the dynamic changes in miRNA to be monitored by the probe. Given its long circulation, efficient penetration, and enhanced tumor accumulation, the PAA-Fe3O4-DNA@PPP probe holds great promise for in situ miRNA imaging and spatial genomics analysis in situ.
Collapse
Affiliation(s)
- Yan Tan
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Research Center for Biosensor and Nanotheranostic, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University Shenzhen 518060 China
| | - Junren Wang
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Research Center for Biosensor and Nanotheranostic, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University Shenzhen 518060 China
| | - Qian Wan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advance Technology, Chinese Academy of Sciences Shenzhen 518055 P. R. China
| | - Jinlong Yang
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Research Center for Biosensor and Nanotheranostic, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University Shenzhen 518060 China
| | - Jinkun Huang
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Research Center for Biosensor and Nanotheranostic, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University Shenzhen 518060 China
| | - Zijia Zhou
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Research Center for Biosensor and Nanotheranostic, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University Shenzhen 518060 China
| | - Haifeng Dong
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Research Center for Biosensor and Nanotheranostic, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University Shenzhen 518060 China
| | - Xueji Zhang
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Research Center for Biosensor and Nanotheranostic, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University Shenzhen 518060 China
- Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ) Shenzhen 518060 China
| |
Collapse
|
32
|
Maier A, Jia Q, Shukla K, Dugulan AI, Hagedoorn PL, van Oossanen R, van Rhoon G, Denkova AG, Djanashvili K. Enhancing Magnetic Hyperthermia Efficiency in Pd/Fe-Oxide Hybrid Nanoparticles through Mn-Doping. ACS APPLIED NANO MATERIALS 2024; 7:27465-27475. [PMID: 39697527 PMCID: PMC11650597 DOI: 10.1021/acsanm.4c05452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024]
Abstract
Multifunctional, biocompatible magnetic materials, such as iron oxide nanoparticles (IONPs), hold great potential for biomedical applications including diagnostics (e.g., MRI) and cancer therapy. In particular, they can play a crucial role in advancing cancer thermotherapy by generating heat when administered intratumorally and when exposed to an alternating magnetic field. This heat application is often combined with radio- (chemo)therapy and/or imaging. Consequently, the design of materials for such a multimodal approach requires hybrid nanoparticles that retain their magnetic properties while integrating additional functionalities. This work introduces synthesis and investigation of magnetically enhanced nanoparticles with a palladium core (envisioned for future radiolabeling with therapeutic 103Pd) and a magnetic iron oxide shell containing paramagnetic manganese (Pd/Fe|(nMn)-oxide, n = 0.25 and 0.5). Doping the iron oxide lattice with Mn significantly increases magnetic saturation, boosting specific loss power up to 1.7 times compared to that of undoped analogs. Interestingly, higher Mn-content in Pd/Fe|(0.5Mn)-oxide leads to a pronounced Mn outer rim, enhancing the heating efficiency at 346 kHz and 23 mT and contributing to the water exchange on the surface of the paramagnetically doped nanoparticles, resulting in additional T 1 MRI contrast. The enhanced magnetic properties of the hybrid Pd/Fe|Mn-oxide nanoparticles enable effective therapeutic outcomes with injection of only small quantities of the material, offering great potential for effective cancer treatment strategies that combine hyperthermia/thermal ablation with radiotherapy while allowing for real-time monitoring via MRI.
Collapse
Affiliation(s)
- Alexandra Maier
- Department
of Biotechnology, Delft University of Technology, 2628 HZ Delft, The Netherlands
- Department
of Radiation Science and Technology, Delft
University of Technology, 2629 JB Delft, The
Netherlands
| | - Qi Jia
- Department
of Biotechnology, Delft University of Technology, 2628 HZ Delft, The Netherlands
- Department
of Radiation Science and Technology, Delft
University of Technology, 2629 JB Delft, The
Netherlands
| | - Keshav Shukla
- Department
of Biotechnology, Delft University of Technology, 2628 HZ Delft, The Netherlands
- Department
of Radiation Science and Technology, Delft
University of Technology, 2629 JB Delft, The
Netherlands
| | - Achim Iulian Dugulan
- Department
of Radiation Science and Technology, Delft
University of Technology, 2629 JB Delft, The
Netherlands
| | - Peter-Leon Hagedoorn
- Department
of Biotechnology, Delft University of Technology, 2628 HZ Delft, The Netherlands
| | - Rogier van Oossanen
- Department
of Radiation Science and Technology, Delft
University of Technology, 2629 JB Delft, The
Netherlands
- Department
of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center, 3008 AE Rotterdam, The Netherlands
| | - Gerard van Rhoon
- Department
of Radiation Science and Technology, Delft
University of Technology, 2629 JB Delft, The
Netherlands
- Department
of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center, 3008 AE Rotterdam, The Netherlands
| | - Antonia G. Denkova
- Department
of Radiation Science and Technology, Delft
University of Technology, 2629 JB Delft, The
Netherlands
| | - Kristina Djanashvili
- Department
of Biotechnology, Delft University of Technology, 2628 HZ Delft, The Netherlands
- Department
of Radiation Science and Technology, Delft
University of Technology, 2629 JB Delft, The
Netherlands
| |
Collapse
|
33
|
Shu G, Zhang C, Wen Y, Pan J, Zhang X, Sun SK. Bismuth drug-inspired ultra-small dextran coated bismuth oxide nanoparticles for targeted computed tomography imaging of inflammatory bowel disease. Biomaterials 2024; 311:122658. [PMID: 38901130 DOI: 10.1016/j.biomaterials.2024.122658] [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: 01/23/2024] [Revised: 05/03/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024]
Abstract
Bismuth (Bi)-based computed tomography (CT) imaging contrast agents (CAs) hold significant promise for diagnosing gastrointestinal diseases due to their cost-effectiveness, heightened sensitivity, and commendable biocompatibility. Nevertheless, substantial challenges persist in achieving an easy synthesis process, remarkable water solubility, and effective targeting ability for the potential clinical transformation of Bi-based CAs. Herein, we show Bi drug-inspired ultra-small dextran coated bismuth oxide nanoparticles (Bi2O3-Dex NPs) for targeted CT imaging of inflammatory bowel disease (IBD). Bi2O3-Dex NPs are synthesized through a simple alkaline precipitation reaction using bismuth salts and dextran as the template. The Bi2O3-Dex NPs exhibit ultra-small size (3.4 nm), exceptional water solubility (over 200 mg mL-1), high Bi content (19.75 %), excellent biocompatibility and demonstrate higher X-ray attenuation capacity compared to clinical iohexol. Bi2O3-Dex NPs not only enable clear visualization of the GI tract outline and intestinal loop structures in CT imaging but also specifically target and accumulate at the inflammatory site in colitis mice after oral administration, facilitating a precise diagnosis and enabling targeted CT imaging of IBD. Our study introduces a novel and clinically promising strategy for synthesizing high-performance Bi2O3-Dex NPs for diagnosing gastrointestinal diseases.
Collapse
Affiliation(s)
- Gang Shu
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, 300203, China; Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Cai Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Ya Wen
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Jinbin Pan
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xuening Zhang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, 300203, China.
| |
Collapse
|
34
|
Curcio A, Curé G, Espinosa A, Menguy N, Galarreta-Rodriguez I, Abou-Hassan A, Piquet B, Motte L, Lalatonne Y, Wilhelm C, Van de Walle A. Elucidating the Dynamics of Biodegradation and Biosynthesis of Magnetic Nanoparticles in Human Stem Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2407034. [PMID: 39439159 DOI: 10.1002/smll.202407034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Iron oxide nanoparticles, due to their magnetic properties, are versatile tools for biomedical applications serving both diagnostic and therapeutic roles. Their performance is intricately intertwined with their fate in the demanding biological environment. Once inside cells, these nanoparticles can be degraded, implying a loss of magnetic efficacy, but also transformed into neo-synthesized magnetic nanoparticles, potentially restoring functionality. This study aims to delineate biological features governing these processes. Magnetic nanoparticles are internalized in human mesenchymal stem cells (hMSCs), and their biotransformations are investigated from nano- to micro-scale using electron microscopy (STEM-HAADF, HRTEM, SAED), a benchtop magnetic sensor, and fine structural characterizations (synchrotron XRD, VSM). Results evidence a delicate equilibrium between the biodegradation and biosynthesis of magnetic nanoparticles, with biotransformation kinetics depending on cell density at magnetic labeling and on spatial cell configuration (monolayers vs spheroids). The biotransformed nanoparticles, composed of magnetite or maghemite, are localized within endosomal/lysosomal compartments and associated with the recruitment of ferritin proteins.
Collapse
Affiliation(s)
- Alberto Curcio
- Laboratoire Physique des Cellules et Cancer, Institut Curie, CNRS, Université PSL, Paris, 75005, France
| | - Guilhem Curé
- Laboratoire Physique des Cellules et Cancer, Institut Curie, CNRS, Université PSL, Paris, 75005, France
| | - Ana Espinosa
- Instituto de Ciencia de Materiales de Madrid, ICMM-CSIC, Madrid, 28049, Spain
| | - Nicolas Menguy
- Sorbonne Université, UMR CNRS 7590, MNHN, de physique des matériaux et de cosmochimie (IMPMC), Paris, 75005, France
| | - Itziar Galarreta-Rodriguez
- Instituto de Ciencia de Materiales de Madrid, ICMM-CSIC, Madrid, 28049, Spain
- Spanish CRG beamline at the European Synchrotron (ESRF), B.P. 220, Grenoble, F-38043, France
| | - Ali Abou-Hassan
- CNRS, Physicochimie des Électrolytes et Nanosystèmes InterfaciauX (PHENIX), Sorbonne Université, Paris, F-75005, France
- Institut Universitaire de France (IUF), Paris, 75231 Cedex 05, France
| | - Bérénice Piquet
- Electron Microscopy Platform, Muséum National d'Histoire Naturelle, CP 39, 12 rue Buffon, Paris, 75231 CEDEX 05, France
| | - Laurence Motte
- INSERM, LVTS, Université Sorbonne Paris Nord and Université Paris Cité, Paris, F-75018, France
| | - Yoann Lalatonne
- INSERM, LVTS, Université Sorbonne Paris Nord and Université Paris Cité, Paris, F-75018, France
- Service de Biophysique et Médecine Nucléaire, Hôpital Avicenne AP-HP, Bobigny, F-93009, France
| | - Claire Wilhelm
- Laboratoire Physique des Cellules et Cancer, Institut Curie, CNRS, Université PSL, Paris, 75005, France
| | - Aurore Van de Walle
- Laboratoire Physique des Cellules et Cancer, Institut Curie, CNRS, Université PSL, Paris, 75005, France
| |
Collapse
|
35
|
Bi J, Zeng J, Liu X, Mo C, Yao M, Zhang J, Yuan P, Jia B, Xu S. Drug delivery for age-related bone diseases: From therapeutic targets to common and emerging therapeutic strategies. Saudi Pharm J 2024; 32:102209. [PMID: 39697472 PMCID: PMC11653637 DOI: 10.1016/j.jsps.2024.102209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
With the accumulation of knowledge on aging, people have gradually realized that among the many factors that cause individual aging, the accumulation of aging cells is an essential cause of organ degeneration and, ultimately, age-related diseases. Most cells present in the bone microenvironment gradually age over time, leading to an imbalance of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis. This imbalance contributes to age-related bone loss and the development of age-related bone diseases, such as osteoporosis. Bone aging can prolong the lifespan and delay the development of age-related diseases. Nanoparticles have controllable and stable physical and chemical properties and can precisely target different tissues and organs. By preparing multiple easily modified and biocompatible nanoparticles as different drug delivery carriers, specifically targeting various diseased tissues for controlled-release and sustained-release administration, the delivery efficiency of drugs can be significantly improved, and the toxicity and side effects of drugs can be substantially reduced, thereby improving the therapeutic effect of age-related bone diseases. In addition, other novel anti-aging strategies (such as stem cell exosomes) also have significant scientific and practical significance in anti-aging research on age-related bone diseases. This article reviews the research progress of various nano-drug-loaded particles and emerging anti-aging methods for treating age-related bone diseases, offering new insights and directions for precise targeted clinical therapies.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohao Liu
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Jing Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
36
|
Rama E, Mohapatra SR, Sugimura Y, Suzuki T, Siebert S, Barmin R, Hermann J, Baier J, Rix A, Lemainque T, Koletnik S, Elshafei AS, Pallares RM, Dadfar SM, Tolba RH, Schulz V, Jankowski J, Apel C, Akhyari P, Jockenhoevel S, Kiessling F. In vitro and in vivo evaluation of biohybrid tissue-engineered vascular grafts with transformative 1H/ 19F MRI traceable scaffolds. Biomaterials 2024; 311:122669. [PMID: 38906013 DOI: 10.1016/j.biomaterials.2024.122669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024]
Abstract
Biohybrid tissue-engineered vascular grafts (TEVGs) promise long-term durability due to their ability to adapt to hosts' needs. However, the latter calls for sensitive non-invasive imaging approaches to longitudinally monitor their functionality, integrity, and positioning. Here, we present an imaging approach comprising the labeling of non-degradable and degradable TEVGs' components for their in vitro and in vivo monitoring by hybrid 1H/19F MRI. TEVGs (inner diameter 1.5 mm) consisted of biodegradable poly(lactic-co-glycolic acid) (PLGA) fibers passively incorporating superparamagnetic iron oxide nanoparticles (SPIONs), non-degradable polyvinylidene fluoride scaffolds labeled with highly fluorinated thermoplastic polyurethane (19F-TPU) fibers, a smooth muscle cells containing fibrin blend, and endothelial cells. 1H/19F MRI of TEVGs in bioreactors, and after subcutaneous and infrarenal implantation in rats, revealed that PLGA degradation could be faithfully monitored by the decreasing SPIONs signal. The 19F signal of 19F-TPU remained constant over weeks. PLGA degradation was compensated by cells' collagen and α-smooth-muscle-actin deposition. Interestingly, only TEVGs implanted on the abdominal aorta contained elastin. XTT and histology proved that our imaging markers did not influence extracellular matrix deposition and host immune reaction. This concept of non-invasive longitudinal assessment of cardiovascular implants using 1H/19F MRI might be applicable to various biohybrid tissue-engineered implants, facilitating their clinical translation.
Collapse
Affiliation(s)
- Elena Rama
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Saurav Ranjan Mohapatra
- Department of Biohybrid & Medical Textiles, AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Yukiharu Sugimura
- Department of Cardiac Surgery, Medical Faculty and RWTH University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Tomoyuki Suzuki
- Department of Cardiac Surgery, Medical Faculty and RWTH University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stefan Siebert
- Department of Biohybrid & Medical Textiles, AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Roman Barmin
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Juliane Hermann
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Jasmin Baier
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Teresa Lemainque
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany; Department of Diagnostic and Interventional Radiology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Susanne Koletnik
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Asmaa Said Elshafei
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Roger Molto Pallares
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Seyed Mohammadali Dadfar
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany; Ardena Oss, 5349 AB Oss, the Netherlands
| | - René H Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, Medical Faculty, RWTH Aachen International University, Aachen, Germany
| | - Volkmar Schulz
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, the Netherlands
| | - Christian Apel
- Department of Biohybrid & Medical Textiles, AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Payam Akhyari
- Department of Cardiac Surgery, Medical Faculty and RWTH University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles, AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany.
| |
Collapse
|
37
|
Dydak K, Zalewski T, Kempka M, Florczak P, Nowaczyk G, Przysiecka Ł, Jagielski J, Loppinet B, Banaszak M, Flak D. Nanoassemblies with Gd-chelating lipids (GMO@DTPA-BSA-Gd) as a potential new type of high molecular weight contrast agents. J Mater Chem B 2024; 12:12017-12029. [PMID: 39451020 DOI: 10.1039/d4tb01684j] [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: 10/26/2024]
Abstract
Self-assembled lipid nanoparticles containing Gd-chelating lipids are a new type of positive magnetic resonance imaging contrast agents (MRI CAs). High molecular weight imposes reduced molecular reorientation (τr) and corresponding longer reorientation correlation times (τc), finally resulting in overall high relaxivity (r1) of such contrast agents. Therefore, we report nanoassemblies based on two types of amphiphile molecules: glyceryl monooleate (GMO) as a matrix embedded with DTPA-bis(stearylamide) and its gadolinium salt (DTPA-BSA-Gd) as a Gd-chelating lipid, stabilized by surfactant Pluronic F127 molecules. The loading of DTPA-BSA-Gd into the GMO matrix was investigated at low (5% w/w) and high (30, 40, 50% w/w) contents. Small angle X-ray scattering (SAXS), cryogenic transmission electron microscopy (cryo-TEM) and dynamic light scattering (DLS) results show that although the nanoassembly of both amphiphile molecules within the nanoparticle is disturbed in terms of the formed phases, this composition ensures their colloidal stability. In nanoparticles with low DTPA-BSA-Gd contents, the assembly results in a cubic diamond phase that is co-existing with a fraction of liposomes. For high DTPA-BSA-Gd contents, swelling of the structure occurs such that the initially formed primitive cubic phase transforms toward a lamellar phase in the nanoassemblies. Results from inductively coupled plasma mass spectrometry (ICP-MS) indicate that for almost all systems, the loading efficiency (LE) of DTPA-BSA-Gd is high (reaching up to approx. 85%), and the nanoassembly provides strong entrapment of Gd3+ ions, which are then efficiently uptaken by cells. Moreover, the higher the surfactant content, the higher the LE. The viability studies demonstrate that the prepared nanoassemblies preserve high biocompatibility towards both cancer (HeLa) and normal cells (MSU 1.1). Nuclear magnetic resonance relaxometry studies (NMR relaxometry) followed by MRI on the prepared nanoassembly dispersions proved that the formation of GMO@DTPA-BSA-Gd nanoassemblies, considered as high molecular weight CAs, results in high relaxivity parameters (e.g., r1 = 19.72 mM-1 s-1 for 2GMO-40DTPA-10F127) that are superior to commercially developed ones (e.g., Magnevist or Gadovist). These comprehensive studies imply that a high degree of internal ordering of nanoassemblies with a higher content of Gd-chelating lipid is not a decisive factor in determining the increase in relaxivity, thus confirming their potential as positive MRI CAs.
Collapse
Affiliation(s)
- Karolina Dydak
- Faculty of Physics, Adam Mickiewicz University, Uniwersytetu Poznańskiego 2, 61-614 Poznań, Poland
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland.
| | - Tomasz Zalewski
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland.
| | - Marek Kempka
- Faculty of Physics, Adam Mickiewicz University, Uniwersytetu Poznańskiego 2, 61-614 Poznań, Poland
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland.
| | - Patryk Florczak
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland.
| | - Grzegorz Nowaczyk
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland.
| | - Łucja Przysiecka
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland.
| | - Jakub Jagielski
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland.
| | - Benoit Loppinet
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, P.O. Box 1385, 711 10 Heraklion, Crete, Greece
| | - Michał Banaszak
- Faculty of Physics, Adam Mickiewicz University, Uniwersytetu Poznańskiego 2, 61-614 Poznań, Poland
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland.
| | - Dorota Flak
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland.
| |
Collapse
|
38
|
Wang W, Liu Z, Zhu J, Zhen H, Qi M, Luo J, Zhen J. Macrophage tracking with USPIO imaging and T2 mapping predicts immune rejection of transplanted stem cells. Sci Rep 2024; 14:29162. [PMID: 39587241 PMCID: PMC11589617 DOI: 10.1038/s41598-024-80750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024] Open
Abstract
To develop a clinical imaging method for monitoring macrophage migration to the defect site after implantation of various stem cells and evaluating immune responses in the context of knee arthritis, T2 mapping was correlated with CD68-positive cell densities in defects and the bone marrow. This study, which was approved by the Institutional Animal Care and Use Committee, used 32 New Zealand white rabbits preloaded with ultrasmall superparamagnetic iron oxide particles (USPIOs). They were divided into groups that received different stem cell implants after osteochondral defect induction. T2 imaging was performed using a 3.0 T MR scanner, and the data were analysed via one-way ANOVA, with CD68 expression assessed via immunohistochemistry. After implantation, the T2 signal intensity increased across groups, with subgroup D1 (implantation of rat bone marrow stem cells (BMSCs)) showing the lowest T2 value early and the steepest increase in T2 values. Notable differences in CD68-positive cell density were found between Subgroup D1 and the other groups and between Subgroups A1 and C1 post-surgery. A moderate negative correlation was observed between T2 signals and CD68-positive cell density in defects (r = -0.468, p = 0.001), whereas a weak correlation was detected in the bone marrow (r = 0.096, p = 0.313). A significant link was identified between CD68-positive cell density in the bone marrow and in defects (r = -0.255, p = 0.001). This study revealed significant differences in immune responses to stem cells from different origin tissues in the context of cartilage repair. Adipose-derived stem cells (ADSCs) were found to be more likely to provoke immune rejection than were BMSCs in the repair of femoral condyle cartilage defects. Compared with allogeneic transplants, xenogeneic mesenchymal stem cell transplants were associated with prolonged immune rejection. T2 mapping technology was effective in predicting the density of CD68-positive cells, providing a valuable tool for immune monitoring in stem cell therapy.
Collapse
Affiliation(s)
- Wenhui Wang
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, 116000, Liaoning, China
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Zhenyu Liu
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jiahong Zhu
- Department of Radiology, Taiyuan Hospital of Traditional Chinese Medicine, Taiyuan, 030001, Shanxi, China
| | - Haocheng Zhen
- Clinical and Basic Medical College, Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Meiling Qi
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jing Luo
- Department of Rheumatology and immunology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Junping Zhen
- Department of Rheumatology and immunology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Department of Magnetic Resonance, Faculty of Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
39
|
Miao L, Zhu Y, Chang H, Zhang X. Nanotheranostics in Breast Cancer Bone Metastasis: Advanced Research Progress and Future Perspectives. Pharmaceutics 2024; 16:1491. [PMID: 39771471 PMCID: PMC11676679 DOI: 10.3390/pharmaceutics16121491] [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: 09/30/2024] [Revised: 10/27/2024] [Accepted: 11/14/2024] [Indexed: 01/11/2025] Open
Abstract
Breast cancer is the leading cause of cancer-related morbidity and mortality among women worldwide, with bone being the most common site of all metastatic breast cancer. Bone metastases are often associated with pain and skeletal-related events (SREs), indicating poor prognosis and poor quality of life. Most current therapies for breast cancer bone metastasis primarily serve palliative purposes, focusing on pain management, mitigating the risk of bone-related complications, and inhibiting tumor progression. The emergence of nanodelivery systems offers novel insights and potential solutions for the diagnosis and treatment of breast cancer-related bone metastasis. This article reviews the recent advancements and innovative applications of nanodrug delivery systems in the context of breast cancer bone metastasis and explores future directions in nanotheranostics.
Collapse
Affiliation(s)
- Lin Miao
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
- Graduate School, China Medical University, Shenyang 110122, China
| | - Yidan Zhu
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Hong Chang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
| | - Xinfeng Zhang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
- Graduate School, China Medical University, Shenyang 110122, China
| |
Collapse
|
40
|
Guedes G, Uribe KB, Martínez-Parra L, Aires A, Beraza M, Ruiz-Cabello J, Cortajarena AL. Engineering Protein-Nanoparticle Hybrids as Targeted Contrast Agents. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59849-59861. [PMID: 39444371 DOI: 10.1021/acsami.4c12799] [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: 10/25/2024]
Abstract
Iron oxide nanoparticles (IONPs) have shown great promise in biomedical applications, particularly as MRI contrast agents due to their magnetic properties and biocompatibility. Although several IONPs have been approved by regulatory agencies as MRI contrast agents, their primary application as negative contrast agents limits their usage. Additionally, there is an emerging need for the development of molecular contrast agents that can specifically target biomarkers, enabling more accurate and sensitive diagnostics. To address these challenges, we exploited the engineerability of proteins to stabilize IONPs with tailored magnetic properties, creating protein-stabilized iron oxide nanoparticles (Prot-IONPs) and leveraged the chemical diversity of proteins to functionalize Prot-IONPs with targeting moieties. As a proof-of-concept, we used alendronate (Ald) to target atherosclerotic plaques in the aorta. Simple protein functionalization allowed targeting while maintaining the stability and relaxation properties of the Prot-IONPs. Prot-IONPs-Ald successfully enabled positive contrast imaging of atherosclerotic plaques in vivo in an atherosclerotic mouse model (ApoE-/- mice on a high-fat diet). This study demonstrates the potential of engineering protein-nanoparticle hybrids as versatile platforms for developing targeted in vivo MRI contrast agents.
Collapse
Affiliation(s)
- Gabriela Guedes
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Parque Tecnológico de San Sebastian Paseo Miramón 194, 20014 Donostia-San Sebastian, Spain
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain
| | - Kepa B Uribe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Parque Tecnológico de San Sebastian Paseo Miramón 194, 20014 Donostia-San Sebastian, Spain
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain
| | - Lydia Martínez-Parra
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Parque Tecnológico de San Sebastian Paseo Miramón 194, 20014 Donostia-San Sebastian, Spain
| | - Antonio Aires
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Parque Tecnológico de San Sebastian Paseo Miramón 194, 20014 Donostia-San Sebastian, Spain
| | - Marta Beraza
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Parque Tecnológico de San Sebastian Paseo Miramón 194, 20014 Donostia-San Sebastian, Spain
| | - Jesús Ruiz-Cabello
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Parque Tecnológico de San Sebastian Paseo Miramón 194, 20014 Donostia-San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- Ciber Enfermedades Respiratorias (Ciberes), 28029 Madrid, Spain
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Aitziber L Cortajarena
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Parque Tecnológico de San Sebastian Paseo Miramón 194, 20014 Donostia-San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
41
|
Ning Y, Yuwen Zhou I, Caravan P. Quantitative in Vivo Molecular MRI. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407262. [PMID: 39279542 PMCID: PMC11530320 DOI: 10.1002/adma.202407262] [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: 05/21/2024] [Revised: 07/29/2024] [Indexed: 09/18/2024]
Abstract
Molecular magnetic resonance imaging (MRI) combines chemistry, chemical biology, and imaging techniques to track molecular events non-invasively. Quantitative molecular MRI aims to provide meaningful, reproducible numerical measurements of molecular processes or biochemical targets within the body. In this review, the classifications of molecular MRI probes based on their signal-generating mechanism and functionality are first described. From there, the primary considerations for in vitro characterization and in vivo validation of molecular MRI probes, including how to avoid pitfalls and biases are discussed. Then, recommendations on imaging acquisition protocols and analysis methods to establish quantitative relationships between MRI signal change induced by the probes and the molecular processes of interest are provided. Finally, several representative case studies are highlighted that incorporate these features. Quantitative molecular MRI is a multidisciplinary research area incorporating expertise in chemical biology, inorganic chemistry, molecular probes, imaging physics, drug development, pathobiology, and medicine. The purpose of this review is to provide guidance to chemists developing MR imaging probes and methods in terms of in vitro and in vivo validation to accelerate the translation of these new quantitative tools for non-invasive imaging of biological processes.
Collapse
Affiliation(s)
- Yingying Ning
- Spin-X Institute, School of Chemistry and Chemical Engineering, School of Biomedical Sciences and Engineering, State Key Laboratory of Luminescent Materials and Devices, Guangdong-Hong Kong-Macao Joint Laboratory of Optoelectronic and Magnetic Functional Materials, South China University of Technology, Guangzhou 510641, China
| | - Iris Yuwen Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
42
|
Cheng Q, Chang Y, Zhang D, Zhao X, Xiao Z, Chen T, Shi C, Luo L. Biomineralization Synthesis of HoMn Nanoparticles for Ultrahigh-Field-Tailored and T1-T2 Dual-Mode MRI-Guided Cancer Theranostics. ACS NANO 2024; 18:27853-27868. [PMID: 39370780 DOI: 10.1021/acsnano.4c00516] [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: 10/08/2024]
Abstract
Ultrahigh field magnetic resonance imaging (UHF-MRI) (≥7 T) can dramatically boost image resolution and signal-to-noise ratio, which have distinct advantages in multifunctional imaging. However, their research and application are currently limited by the absence of high-field contrast agents (CAs) and the low sensitivity and accuracy of T1/T2 single-modality CAs. Therefore, the development of T1-T2 dual-mode CAs that respond to UHF-MRI and nanoformulations with therapeutic sensitization can bring ideas for the integrated application of precise and synchronous tumor theranostics. Herein, we present a biomimetic mineralization strategy for synthesizing holmium/manganese oxide-bovine serum albumin-photosensitizer chlorin e6 nanohybrids. The hybrid nanoparticles exhibited better tumor accumulation, a suitable time imaging window, and excellent pH-response T1-T2 dual-mode UHF-MRI performance. The antitumor effect comes from the amelioration of the hypoxic tumor microenvironment to promote the synergistic effect of photodynamic therapy and radiotherapy, along with negligible acute toxicity. Undoubtedly, this work not only provides a different perspective for developing multifunctional nanotherapeutics but also promotes the potential clinical exploitation and translation of UHF CAs.
Collapse
Affiliation(s)
- Qingqing Cheng
- Department of Medical Imaging Center, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou 510630, China
| | - Yanzhou Chang
- Department of Medical Imaging Center, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou 510630, China
| | - Dong Zhang
- Department of Medical Imaging Center, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou 510630, China
- The Shunde Affiliated Hospital, Jinan University, Foshan 528300, China
| | - Xiangsheng Zhao
- Department of Radiology, Wuyi Hospital of Traditional Chinese Medicine, Jiangmen 529099, China
| | - Zeyu Xiao
- Department of Medical Imaging Center, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou 510630, China
| | - Tianfeng Chen
- Department of Medical Imaging Center, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou 510630, China
- Guangdong No. 2 Provincial People's Hospital, Jinan University, Guangzhou 510310, China
| | - Changzheng Shi
- Department of Medical Imaging Center, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou 510630, China
| | - Liangping Luo
- Department of Medical Imaging Center, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou 510630, China
- Guangdong No. 2 Provincial People's Hospital, Jinan University, Guangzhou 510310, China
| |
Collapse
|
43
|
Ayyami Y, Ghorbani M, Dastgir M, Malekzadeh R, Mortezazadeh T. Chitosan-modified manganese oxide-conjugated methotrexate nanoparticles delivering 5-aminolevulinic acid as a dual-modal T1-T2* MRI contrast agent in U87MG cell detection. MAGMA (NEW YORK, N.Y.) 2024; 37:909-924. [PMID: 38795276 DOI: 10.1007/s10334-024-01169-4] [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: 01/23/2024] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 05/27/2024]
Abstract
OBJECTIVE Glioblastoma multiforme is a highly aggressive form of brain cancer, and early diagnosis plays a pivotal role in improving patient survival rates. In this regard, molecular magnetic resonance imaging has emerged as a promising imaging modality due to its exceptional sensitivity to minute tissue changes and the ability to penetrate deep into the brain. This study aimed to assess the efficacy of a novel contrast agent in detecting gliomas during MRI scans. MATERIALS AND METHODS The contrast agent utilized modified chitosan coating on manganese oxide nanoparticles. The modification included adding methotrexate and 5-aminolevulinic acid (MnO2/CS@5-ALA-MTX) to target cells with overexpressed folate receptors and breaking down excess hydrogen peroxide in tumor tissue, resulting in enhanced signal intensity in T1-weighted MR images but diminished signal intensity in T2*-weighted MR images. RESULTS The nanosystem was characterized and evaluated in MR imaging, safety, and ability to target cells both in vivo and in vitro. MTX-free nanoparticles (MnO2/CS@5-ALA NPs) had no obvious cytotoxicity on cell lines U87MG and NIH3T3 after 24/48 h at a concentration of up to 160 µgr/mL (cell viability more than 80%). In this system, methotrexate enables tumor targeting and the MnO2/5-ALA improves T1-T2*-weighted MRI. In addition, MRI scans of mice with M109 carcinoma indicated significant tumor uptake and NP capacity to improve the positive contrast effect. CONCLUSION This developed MnO2/CS@5-ALA-MTX nanoparticle system may exhibit great potential in the accurate diagnosis of folate receptor over-expressing cancers such as glioblastoma.
Collapse
Affiliation(s)
- Yasin Ayyami
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marjan Ghorbani
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Iran Polymer and Petrochemical Institute, P.O.Box: 14965/115, Tehran, Iran
| | - Masoumeh Dastgir
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Malekzadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Tohid Mortezazadeh
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
44
|
Shen S, Koonjoo N, Boele T, Lu J, Waddington DEJ, Zhang M, Rosen MS. Enhancing organ and vascular contrast in preclinical ultra-low field MRI using superparamagnetic iron oxide nanoparticles. Commun Biol 2024; 7:1197. [PMID: 39342051 PMCID: PMC11438998 DOI: 10.1038/s42003-024-06884-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) are characterized by their exceptional susceptibility and relaxivity at ultra-low field (ULF) regimes, make them a promising contrast agent (CA) for ULF MRI. Despite their distinct advantages, the translation of these properties into clinically valuable image contrast in ULF MRI remains underexplored. In this study, we investigate the use of SPIONs to generate in vivo MRI contrast at 6.5 mT within the organs and vascular system of rodents. This investigation includes comprehensive SPION characterization and phantom imaging experiments to validate the utility of SPIONs to produce positive image contrast and to facilitate phase-sensitive imaging at ULF. Optimized balanced steady-state free precession (bSSFP) and spoiled gradient echo (SPGR) MRI sequences are used to generate in vivo contrast by leveraging the distinctive properties of SPIONs at ULF. Imaging studies in rodents reveal positive organ contrast attainable in magnitude images, and MRI phase maps can be used to visualize the vascular system. This work demonstrates the effectiveness of SPIONs in enhancing preclinical organ and vascular imaging at ULF; it bridges the gap between the study of the distinctive physical properties of SPIONs and the demonstration of in vivo image contrast.
Collapse
Affiliation(s)
- Sheng Shen
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Neha Koonjoo
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Thomas Boele
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jiaqi Lu
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
- Department of Chemistry, New York University, New York, NY, USA
| | - David E J Waddington
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | | | - Matthew S Rosen
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Physics, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
45
|
Blasiak B, MacDonald D, Jasiński K, Cheng FY, Tomanek B. Application of H 2N-Fe 3O 4 Nanoparticles for Prostate Cancer Magnetic Resonance Imaging in an Animal Model. Int J Mol Sci 2024; 25:10334. [PMID: 39408664 PMCID: PMC11477031 DOI: 10.3390/ijms251910334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
This paper presents the efficacy of a contrast agent based on H2N-Fe3O4 nanoparticles for the detection of prostate cancer in an animal model using a preclinical 9.4 T MRI system. The relaxivities r1 and r2 of the nanoparticles were 6.31 mM-1s-1 and 8.33 mM-1s-1, respectively. Nanoparticles injected in a concentration of 2 mg Fe/mL decreased the tumor-relative T1 relaxation across all animals from 100 to 76 ± 26, 85 ± 27, 89 ± 20, and 97 ± 16 12 min 1 h, 2 h, and 24 h post injection, respectively. The corresponding T1 decrease in muscle tissues was 90 ± 20, 94 ± 23, 99 ± 12, and 99 ± 14. The relative T2 changes in the tumor were 82 ± 17, 89 ± 19, 97 ± 14, and 99 ± 8 12 min, 1 h, 2 h, and 24 h post injection, respectively, while, for muscle tissues, these values were 95 ± 11, 95 ± 8, 97 ± 6, and 95 ± 10 at the corresponding time points. The differences in the relative T1 and T2 were only significant 12 min after injection (p < 0.05), although a decrease was visible at each time point, but it was statistically insignificant (p > 0.05). The results showed the potential application of H2N-Fe3O4 nanoparticles as contrast agents for enhanced prostate cancer MRI.
Collapse
Affiliation(s)
- Barbara Blasiak
- The Henryk Niewodniczanski Institute of Nuclear Physics Polish Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland; (D.M.); (K.J.); (B.T.)
| | - David MacDonald
- The Henryk Niewodniczanski Institute of Nuclear Physics Polish Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland; (D.M.); (K.J.); (B.T.)
| | - Krzysztof Jasiński
- The Henryk Niewodniczanski Institute of Nuclear Physics Polish Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland; (D.M.); (K.J.); (B.T.)
| | - Fong-Yu Cheng
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan;
| | - Boguslaw Tomanek
- The Henryk Niewodniczanski Institute of Nuclear Physics Polish Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland; (D.M.); (K.J.); (B.T.)
- Division of Medical Physics, Department of Oncology, University of Alberta, 8303 112 St. NW, Edmonton, AB T6G 2T4, Canada
| |
Collapse
|
46
|
Seredina YV, Oreshonkov AS, Molokeev MS, Sedykh AE, Aleksandrovsky AS, Zhernakov MA, Khritokhin NA, Azarapin NO, Glukhova PO, Shelpakova NA, Müller-Buschbaum K, Denisenko YG. Thermochemistry of Solid-State Formation, Structure, Optical, and Luminescent Properties of Complex Oxides Eu 2MeO 6 (Me-Mo, W), Eu 2W 2O 9: A Combined Experimental and DFT Study. Chemistry 2024; 30:e202402084. [PMID: 38975664 DOI: 10.1002/chem.202402084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Complex oxides Eu2MeO6 (Me-Mo, W), Eu2W2O9 were obtained by a solid-phase reaction between binary oxides. The thermodynamic and kinetic mechanisms of the reaction processes were established using a variety of physical-chemical methods. All compounds obtained in this work crystallize in the low-symmetry monoclinic system, forming complex framework structures, which determine a set of very valuable physical-chemical properties. Comparison of experimental Kubelka-Munk functions and DFT- calculated absorption spectra shows adequate agreement and reveals the origin of the fundamental absorption. In addition, the deficiency in DFT calculations in the part of mutual contribution of CTBs of Mo-O and W-O, from one side, and Eu-O contributions, from the other side, is reported. Calculations of absorption spectra are shown to be superior to band structure analysis in the determination of optical band gaps. Additionally, luminescent properties of Eu2MeO6 and Eu2W2O9 compounds were investigated. These studies provide a better understanding of the electronic and optical properties of the compounds Eu2MeO6 and Eu2W2O9, along with their potential applications in various areas.
Collapse
Affiliation(s)
- Yulia V Seredina
- School of Natural Sciences, University of Tyumen, Tyumen, 625003, Russia
| | - Aleksandr S Oreshonkov
- Laboratory of Molecular Spectroscopy, Kirensky Institute of Physics Federal Research Center KSC SB RAS, Krasnoyarsk, 660036, Russia
- School of Engineering and Construction, Siberian Federal University, Krasnoyarsk, 660041, Russia
| | - Maxim S Molokeev
- Laboratory of Crystal Physics, Kirensky Institute of Physics, Federal Research Center KSC SB RASK, Krasnoyarsk, 660036, Russia
- Laboratory of Theory and Optimization of Chemical and Technological Processes, Tyumen State University, Tyumen, 625003, Russia
| | - Alexander E Sedykh
- Institute of Inorganic and Analytical Chemistry, Justus-Liebig-University Giessen, Giessen, 35392, Germany
| | - Aleksandr S Aleksandrovsky
- Laboratory of Coherent Optics, Kirensky Institute of Physics Federal Research Center KSC SB RAS, Krasnoyarsk, 660036, Russia
- Institute of Nanotechnology, Spectroscopy and Quantum Chemistry, Siberian Federal University, Krasnoyarsk, 660041, Russia
| | - Maksim A Zhernakov
- Institute of Inorganic and Analytical Chemistry, Justus-Liebig-University Giessen, Giessen, 35392, Germany
- A.M. Butlerov Chemistry Institute, Kazan Federal University, Kazan, 420008, Russia
| | | | - Nikita O Azarapin
- School of Natural Sciences, University of Tyumen, Tyumen, 625003, Russia
| | - Polina O Glukhova
- School of Natural Sciences, University of Tyumen, Tyumen, 625003, Russia
| | | | - Klaus Müller-Buschbaum
- Institute of Inorganic and Analytical Chemistry, Justus-Liebig-University Giessen, Giessen, 35392, Germany
- Center for Materials Research (LaMa), Justus-Liebig-University Giessen, Giessen, 35392, Germany
| | - Yuriy G Denisenko
- School of Natural Sciences, University of Tyumen, Tyumen, 625003, Russia
- Institute of Inorganic and Analytical Chemistry, Justus-Liebig-University Giessen, Giessen, 35392, Germany
- Department of Construction Materials, Industrial University of Tyumen, Tyumen, 625000, Russia
| |
Collapse
|
47
|
Lu J, Yu C, Du K, Chen S, Huang S. Targeted delivery of cisplatin magnetic nanoparticles for diagnosis and treatment of nasopharyngeal carcinoma. Colloids Surf B Biointerfaces 2024; 245:114252. [PMID: 39317040 DOI: 10.1016/j.colsurfb.2024.114252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Rapid advances in nanotechnology are paving the way for innovative breakthroughs in overcoming the current limitations in the clinical treatment of cancer and other prevalent diseases plaguing mankind. Magnetic nanoparticles composed of iron oxide (Fe3O4) are a novel class of nanoparticles that are receiving increasing attention in the field of cancer therapy. To address the inherent limitations, bare Fe3O4 can be functionalized, polymerized, assembled, or combined with other functional materials to produce a range of smart nanoplatforms suitable for tumor therapy. In this paper, we present a unique multifunctional therapeutic nanoplatform centered on aldehyde-oxidized sodium alginate-stabilized iron oxide nanoparticles (NPs) designed for T2-weighted magnetic resonance (MR) imaging. Sodium alginate oxide and ferric oxide nanoparticles were prepared respectively, and the two particles were mixed in a certain molar ratio to form a complex, which was coupled to target polypeptide GE11 by Schiff base reaction, and finally supported by cisplatin through coordination complexation. The prepared magnetic nanoparticles (hereinafter referred to as GE11-CDDP-ASA@Fe3O4) have an average diameter of 152.9 nm, and have good colloidal stability and cytocompatibility. The distinctive structure and composition of GE11-CDDP-ASA@Fe3O4 contribute to its excellent MRI imaging performance, positioning it as a nano platform suitable for enhancing the efficacy of combination therapy in tumor treatment. This is of great significance for translational nanomedicine applications.
Collapse
Affiliation(s)
- Jing Lu
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Chaosheng Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Kun Du
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Shuaijun Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | | |
Collapse
|
48
|
Hou Y, Kong F, Tang Z, Zhang R, Li D, Ge J, Yu Z, Wahab A, Zhang Y, Iqbal MZ, Kong X. Nitroxide radical conjugated ovalbumin theranostic nanosystem for enhanced dendritic cell-based immunotherapy and T 1 magnetic resonance imaging. J Control Release 2024; 373:547-563. [PMID: 39059501 DOI: 10.1016/j.jconrel.2024.07.050] [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: 04/20/2024] [Revised: 07/20/2024] [Accepted: 07/21/2024] [Indexed: 07/28/2024]
Abstract
Melanoma, known for its aggressive metastatic nature, presents a formidable challenge in cancer treatment, where conventional therapies often fall short. This study introduces a pioneering approach utilizing metal-free nanosystem as tumor vaccines, spotlighting their potential in revolutionizing melanoma treatment. This work employed organic nitroxides, specifically 4-carboxy-TEMPO, in combination with chitosan (CS), to create a novel nanocomposite material - the CS-TEMPO-OVA nanovaccines. This composition not only improves biocompatibility and extends blood circulation time of TEMPO but also marks a significant departure from traditional gadolinium-based contrast agents in MRI technology, addressing safety concerns. CS-TEMPO-OVA nanovaccines demonstrate excellent biocompatibility at both the cellular and organoid level. They effectively stimulate bone marrow-derived dendritic cells (BMDCs), which in turn promote the maturation and activation of T cells. This ultimately leads to a strong production of essential cytokines. These nanovaccines serve a dual purpose as both therapeutic and preventive. By inducing an immune response, activating cytotoxic T cells, and promoting macrophage M1 polarization, they effectively inhibit melanoma growth and enhance survival in mouse models. When combined with αPD-1, the CS-TEMPO-OVA nanovaccines significantly bolster the infiltration of cytotoxic T lymphocytes (CTLs) within tumors, sparking a powerful systemic antitumor response that effectively curbs tumor metastasis. The ability of these nanovaccines to control both primary (subcutaneous) and metastatic B16-OVA tumors highlights their remarkable efficacy. Furthermore, the CS-TEMPO-OVA nanovaccine can be administered in vivo via both intravenous and intramuscular routes, both of which effectively enhance the T1 contrast of magnetic resonance imaging in tumor tissue. This study offers invaluable insights into the integrated application of these nanovaccines in both clinical diagnostics and treatment, marking a significant stride in cancer research and patient care.
Collapse
Affiliation(s)
- Yike Hou
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Fei Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Zhe Tang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, PR China
| | - Rui Zhang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Dan Li
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Jian Ge
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone, Hangzhou 310018, Zhejiang, PR China
| | - Zhangsen Yu
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province 312000, PR China
| | - Abdul Wahab
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Yunyang Zhang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - M Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| |
Collapse
|
49
|
Albukhaty S, Sulaiman GM, Al-Karagoly H, Mohammed HA, Hassan AS, Alshammari AAA, Ahmad AM, Madhi R, Almalki FA, Khashan KS, Jabir MS, Yusuf M, Al-aqbi ZT, Sasikumar P, Khan RA. Iron oxide nanoparticles: The versatility of the magnetic and functionalized nanomaterials in targeting drugs, and gene deliveries with effectual magnetofection. J Drug Deliv Sci Technol 2024; 99:105838. [DOI: 10.1016/j.jddst.2024.105838] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
50
|
Wang S, Li J, Chen L, Zeng J, Gao M. Fe 2+-Dominated Relaxometric Properties of Iron Oxide Nanoparticles as MRI Contrast Agents. J Phys Chem Lett 2024; 15:8861-8866. [PMID: 39169277 DOI: 10.1021/acs.jpclett.4c01876] [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: 08/23/2024]
Abstract
Iron oxide nanoparticles (IONPs) have garnered significant interest as magnetic resonance imaging (MRI) contrast agents due to their exceptional magnetic properties and biocompatibility. Toward more precise diagnosis of diseases, the relaxometric properties of IONPs have become a key research focus. Despite extensive studies on structural factors such as size, morphology, surface modification, crystalline phase, and aggregation state, the correlation between the intrinsic structure and relaxometric behavior remains unclear, particularly for ultrasmall IONPs. To address this issue, we carefully compared IONPs with identical size, shape, and surface modification and found out strong correlations among the content of Fe2+ ions, oxygen vacancies, and the relaxometric properties. By optimizing the reaction system, ultrasmall IONPs showing outstanding relaxometric performance, with longitudinal relaxivity up to 9.0 mM-1 s-1 and transverse relaxivity up to 28.5 mM-1 s-1, were successfully obtained. These results underscore the pivotal role of Fe2+ in the relaxometric properties of IONP-based MRI contrast agents.
Collapse
Affiliation(s)
- Sixia Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Junyan Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Lei Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Mingyuan Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|