1
|
Chow JCL. Monte Carlo Simulations in Nanomedicine: Advancing Cancer Imaging and Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:117. [PMID: 39852732 PMCID: PMC11767847 DOI: 10.3390/nano15020117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025]
Abstract
Monte Carlo (MC) simulations have become important in advancing nanoparticle (NP)-based applications for cancer imaging and therapy. This review explores the critical role of MC simulations in modeling complex biological interactions, optimizing NP designs, and enhancing the precision of therapeutic and diagnostic strategies. Key findings highlight the ability of MC simulations to predict NP bio-distribution, radiation dosimetry, and treatment efficacy, providing a robust framework for addressing the stochastic nature of biological systems. Despite their contributions, MC simulations face challenges such as modeling biological complexity, computational demands, and the scarcity of reliable nanoscale data. However, emerging technologies, including hybrid modeling approaches, high-performance computing, and quantum simulation, are poised to overcome these limitations. Furthermore, novel advancements such as FLASH radiotherapy, multifunctional NPs, and patient-specific data integration are expanding the capabilities and clinical relevance of MC simulations. This topical review underscores the transformative potential of MC simulations in bridging fundamental research and clinical translation. By facilitating personalized nanomedicine and streamlining regulatory and clinical trial processes, MC simulations offer a pathway toward more effective, tailored, and accessible cancer treatments. The continued evolution of simulation techniques, driven by interdisciplinary collaboration and technological innovation, ensures that MC simulations will remain at the forefront of nanomedicine's progress.
Collapse
Affiliation(s)
- James C. L. Chow
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1X6, Canada; ; Tel.: +1-416-946-4501
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
2
|
Hey G, Mehkri I, Mehkri Y, Maqbool H, Tahirkheli M, Woodford S, Lucke-Wold B. Nanoparticle-Based Therapies for Management of Subarachnoid Hemorrhage, Neurotrauma, and Stroke. Biomedicines 2024; 13:16. [PMID: 39857600 PMCID: PMC11760890 DOI: 10.3390/biomedicines13010016] [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: 12/02/2024] [Revised: 12/21/2024] [Accepted: 12/22/2024] [Indexed: 01/27/2025] Open
Abstract
Neurotrauma, stroke, and subarachnoid hemorrhage (SAH) are symptomatically diverse and etiologically complex central nervous system pathologies. Despite numerous therapeutic modalities that are available to minimize neurologic damage and secondary injury, the prognosis can still be dismal and unpredictable. Nanoparticle (NP) technology allows for deliberate, modular, and minimally invasive drug delivery. This literature review encompasses pertinent information on the impact and versatility of nanoparticle therapeutics when treating neurotrauma, stroke, and SAH. Currently, notable treatments such as Perfluorooctyl-Bromide (PFOB), PLGA nanoparticles, and ischemic relief-based NPs are promising new techniques for the management of these complex pathologies.
Collapse
Affiliation(s)
- Grace Hey
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Ilyas Mehkri
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Yusuf Mehkri
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Hasan Maqbool
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Mubariz Tahirkheli
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Samuel Woodford
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Brandon Lucke-Wold
- Lillian S. Wells Department of Neurosurgery, College of Medicine, University of Florida, 1600 SW Archer Road, Gainesville, FL 32610, USA
| |
Collapse
|
3
|
Shan T, Chen X, Zhou X, Wang N, Ren B, Cheng L. Stimulus-responsive biomaterials for Helicobacter pylori eradication. J Adv Res 2024; 66:209-222. [PMID: 38160707 PMCID: PMC11675045 DOI: 10.1016/j.jare.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/27/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori), the only bacterium classified as a type I (definite) carcinogen, is strongly associated with the development of gastric inflammation and adenocarcinoma. It infects the stomach of approximately half of the global population, equivalent to nearly 4.4 billion people. However, due to physiological barriers in the stomach, microbial barriers and increased antibiotic resistance, the therapeutic efficiency of standard antibiotic therapy is limited and cannot meet the clinical needs in some areas. Combining stimulus-responsive biomaterials with certain stimuli is an emerging antibacterial strategy. Stimulus-responsive biomaterials can respond to chemical, biological or physical cues in the environment with corresponding changes in their own properties and functions, highlighting a more intelligent, targeting and efficient aspect for H. pylori therapy. AIM OF REVIEW This review describes the critical obstacles in the current treatment of H. pylori, summarizes the recent advances in stimulus-responsive biomaterials against H. pylori by elucidating their working mechanisms and antibacterial performances under different types of stimuli (pH, enzymes, light, magnetic and ultrasound irradiations), and attempts to analyze the future prospects of such smart biomaterial for H. pylori eradication. Key Scientific Concepts of Review: Any characteristic property or change in the biomilieu at the H. pylori infected site (endogenous stimuli) or specific iatrogenic conditions in vitro (exogenous stimuli) can act as cues to activate or potentiate the antibacterial activity of responsive biomaterials. The responsiveness of these materials to endogenous stimuli enhances antimicrobial targeting, and makes physiological barriers that would otherwise hinder conventional H. pylori therapies a key factor in facilitating antibacterial effects. The responsiveness to exogenous stimuli greatly prolongs the action time of antimicrobial materials and pinpoints the site of infection, thereby reducing toxic side effects. These findings pave the way for the development of more precise and effective anti-H. pylori treatment.
Collapse
Affiliation(s)
- Tiantian Shan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xi Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Nanxi Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Najjar R. Clinical applications, safety profiles, and future developments of contrast agents in modern radiology: A comprehensive review. IRADIOLOGY 2024; 2:430-468. [DOI: 10.1002/ird3.95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 07/30/2024] [Indexed: 01/06/2025]
Abstract
AbstractContrast agents have transformed the field of medical imaging, significantly enhancing the visualisation of internal structures and improving diagnostic accuracy across X‐rays, computed tomography, magnetic resonance imaging (MRI), and ultrasound. This review explores the historical development, physicochemical properties, and mechanisms of action of iodinated, gadolinium‐based, barium sulfate, microbubble, and nanoparticle contrast agents. It highlights key advancements, including the transition from high‐osmolar to low‐ and iso‐osmolar iodinated agents, the integration of gadolinium in MRI, and the innovative use of microbubbles and nanoparticles. The review critically examines the safety profiles and adverse reactions of these contrast agents, categorising them into hypersensitivity and physiological reactions. It outlines risk factors, common misconceptions, and management strategies for adverse reactions, emphasising the importance of personalised approaches in clinical practice. Additionally, it delves into broader implications, including ethical considerations, environmental impact, and global accessibility of contrast media. The review also discusses technological advancements such as targeted contrast agents and the integration of artificial intelligence to optimise contrast dosage. By synthesising current knowledge and emerging trends, this review underscores the pivotal role of contrast agents in advancing medical imaging. It aims to equip clinicians, researchers, and policymakers with a thorough understanding to enhance diagnostic efficacy, ensure patient safety, and address ethical and environmental challenges, thereby informing future innovations and regulatory frameworks to promote equitable access to advanced imaging technologies globally.
Collapse
Affiliation(s)
- Reabal Najjar
- The Canberra Hospital Canberra Health Services Canberra Australian Capital Territory Australia
- Australian National University College of Health and Medicine Acton Australian Capital Territory Australia
| |
Collapse
|
5
|
Reviansyah FH, Putra DRD, Supriatna JA, Takarini V, Komariah M. Green Dentistry in Oral Cancer Treatment Using Biosynthesis Superparamagnetic Iron Oxide Nanoparticles: A Systematic Review. Cancer Manag Res 2024; 16:1231-1245. [PMID: 39282609 PMCID: PMC11402364 DOI: 10.2147/cmar.s477791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Oral cancer is a worldwide health issue with high incidence and mortality, demands an effective treatment to improve patient prognosis. Superparamagnetic iron oxide nanoparticles (SPIONs) emerged as a candidate for oral cancer treatment due to their unique attributes, enabling a synergistic combination with its drug-delivery capabilities and hyperthermia when exposed to magnetic fields. SPIONs can be synthesized using biopolymers from agricultural waste like lignin from paddy, which produce biogenic nano iron oxide with superparamagnetic and antioxidant effects. In addition, lignin also acts as a stabilizing agent in creating SPIONs. This study aimed to explore how agricultural waste could be used to prepare SPIONs using the green synthesis method and to evaluate its potential for oral cancer specifically focusing on its effectiveness, side effects, biocompatibility, and toxicity. A systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses protocol. PubMed, EBSCO, and Scopus databases were exploited in the selection of articles published within the last decade. This study quality assessment uses OHAT for critical appraisal tools. Only 10 studies met the inclusion criteria. The findings suggest that the use of agricultural waste in the preparation of SPIONs not only holds potency for oral cancer treatment through drug delivery and hyperthermia but also aligns with the concept of green dentistry. SPIONs as a treatment modality for oral cancer have demonstrated notable effectiveness and versatility. This study provides robust evidence supporting green dentistry by using agricultural waste in the preparation and formulation of SPIONs for managing oral cancer. Its multifunctional nature and ability to enhance treatment efficacy while minimizing adverse effects on healthy tissues highlights the potency of SPION-based oral cancer treatments.
Collapse
Affiliation(s)
| | | | | | - Veni Takarini
- Department of Dental Materials and Technology, Faculty of Dentistry, Padjadjaran University, Bandung, 40132, Indonesia
- Oral Biomaterials Research Centre, Faculty of Dentistry, Padjadjaran University, Bandung, 40132, Indonesia
| | - Maria Komariah
- Department of Fundamental Nursing, Faculty of Nursing, Padjadjaran University, Bandung, 40132, Indonesia
| |
Collapse
|
6
|
ten Hove M, Smyris A, Booijink R, Wachsmuth L, Hansen U, Alic L, Faber C, Hӧltke C, Bansal R. Engineered SPIONs functionalized with endothelin a receptor antagonist ameliorate liver fibrosis by inhibiting hepatic stellate cell activation. Bioact Mater 2024; 39:406-426. [PMID: 38855059 PMCID: PMC11157122 DOI: 10.1016/j.bioactmat.2024.05.034] [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: 11/16/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024] Open
Abstract
Endothelin-1/endothelin A receptor (ET-1/ETAR) pathway plays an important role in the progression of liver fibrosis by activating hepatic stellate cells (HSCs) - a key cell type involved in the pathogenesis of liver fibrosis. Inactivating HSCs by blocking the ET-1/ETAR pathway using a selective ETAR antagonist (ERA) represents a promising therapeutic approach for liver fibrosis. Unfortunately, small-molecule ERAs possess limited clinical potential due to poor bioavailability, short half-life, and rapid renal clearance. To improve the clinical applicability, we conjugated ERA to superparamagnetic iron-oxide nanoparticles (SPIONs) and investigated the therapeutic efficacy of ERA and ERA-SPIONs in vitro and in vivo and analyzed liver uptake by in vivo and ex vivo magnetic resonance imaging (MRI), HSCs-specific localization, and ET-1/ETAR-pathway antagonism in vivo. In murine and human liver fibrosis/cirrhosis, we observed overexpression of ET-1 and ETAR that correlated with HSC activation, and HSC-specific localization of ETAR. ERA and successfully synthesized ERA-SPIONs demonstrated significant attenuation in TGFβ-induced HSC activation, ECM production, migration, and contractility. In an acute CCl4-induced liver fibrosis mouse model, ERA-SPIONs exhibited higher liver uptake, HSC-specific localization, and ET-1/ETAR pathway antagonism. This resulted in significantly reduced liver-to-body weight ratio, plasma ALT levels, and α-SMA and collagen-I expression, indicating attenuation of liver fibrosis. In conclusion, our study demonstrates that the delivery of ERA using SPIONs enhances the therapeutic efficacy of ERA in vivo. This approach holds promise as a theranostic strategy for the MRI-based diagnosis and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Marit ten Hove
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Andreas Smyris
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Richell Booijink
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Lydia Wachsmuth
- Clinic of Radiology, University Hospital Muenster, Muenster, Germany
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital Muenster, Muenster, Germany
| | - Lejla Alic
- Department of Magnetic Detection and Imaging, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Cornelius Faber
- Clinic of Radiology, University Hospital Muenster, Muenster, Germany
| | - Carsten Hӧltke
- Clinic of Radiology, University Hospital Muenster, Muenster, Germany
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| |
Collapse
|
7
|
Hong J, Wang L, Zheng Q, Cai C, Yang X, Liao Z. The Recent Applications of Magnetic Nanoparticles in Biomedical Fields. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2870. [PMID: 38930238 PMCID: PMC11204782 DOI: 10.3390/ma17122870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
Magnetic nanoparticles (MNPs) have found extensive application in the biomedical domain due to their enhanced biocompatibility, minimal toxicity, and strong magnetic responsiveness. MNPs exhibit great potential as nanomaterials in various biomedical applications, including disease detection and cancer therapy. Typically, MNPs consist of a magnetic core surrounded by surface modification coatings, such as inorganic materials, organic molecules, and polymers, forming a nucleoshell structure that mitigates nanoparticle agglomeration and enhances targeting capabilities. Consequently, MNPs exhibit magnetic responsiveness in vivo for transportation and therapeutic effects, such as enhancing medical imaging resolution and localized heating at the site of injury. MNPs are utilized for specimen purification through targeted binding and magnetic separation in vitro, thereby optimizing efficiency and expediting the process. This review delves into the distinctive functional characteristics of MNPs as well as the diverse bioactive molecules employed in their surface coatings and their corresponding functionalities. Additionally, the advancement of MNPs in various applications is outlined. Additionally, we discuss the advancements of magnetic nanoparticles in medical imaging, disease treatment, and in vitro assays, and we anticipate the future development prospects and obstacles in this field. The objective is to furnish readers with a thorough comprehension of the recent practical utilization of MNPs in biomedical disciplines.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhenlin Liao
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (L.W.); (Q.Z.); (C.C.); (X.Y.)
| |
Collapse
|
8
|
Bian X, Guo T, Chen G, Nie D, Yue M, Zhu Y, Lin M. The therapeutic effect and MR molecular imaging of FA-PEG-FePt/DDP nanoliposomes in AMF on ovarian cancer. Int J Nanomedicine 2024; 19:5227-5243. [PMID: 38855734 PMCID: PMC11162642 DOI: 10.2147/ijn.s453601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/17/2024] [Indexed: 06/11/2024] Open
Abstract
Purpose This study aimed to construct targeting drug-loading nanocomposites (FA-FePt/DDP nanoliposomes) to explore their potential in ovarian cancer therapy and molecular magnetic resonance imaging (MMRI). Methods FA-FePt-NPs were prepared by coupling folate (FA) with polyethylene-glycol (PEG)-coated ferroplatinum nanoparticles and characterized. Then cisplatin (DDP) was encapsulated in FA-FePt-NPs to synthesize FA-PEG-FePt/DDP nanoliposomes by thin film-ultrasonic method and high-speed stirring, of which MMRI potential, magnetothermal effect, and the other involved performance were analyzed. The therapeutic effect of FA-FePt/DDP nanoliposomes combined with magnetic fluid hyperthermia (MFH) on ovarian cancer in vitro and in vivo was evaluated. The expression levels of Bax and epithelial-mesenchymal transition related proteins were detected. The biosafety was also preliminarily observed. Results The average diameter of FA-FePt-NPs was about 30 nm, FA-FePt/DDP nanoliposomes were about 70 nm in hydrated particle size, with drug slow-release and good cell-specific targeted uptake. In an alternating magnetic field (AMF), FA-FePt/DDP nanoliposomes could rapidly reach the ideal tumor hyperthermia temperature (42~44 °C). MRI scan showed that FA-FePt-NPs and FA-FePt/DDP nanoliposomes both could suppress the T2 signal, indicating a good potential for MMRI. The in vitro and in vivo experiments showed that FA-FePt/DDP-NPs in AMF could effectively inhibit the growth of ovarian cancer by inhibiting cancer cell proliferation, invasion, and migration, and inducing cancer cell apoptosis, much better than that of the other individual therapies; molecularly, E-cadherin and Bax proteins in ovarian cancer cells and tissues were significantly increased, while N-cadherin, Vimentin, and Bcl-2 proteins were inhibited, effectively inhibiting the malignant progression of ovarian cancer. In addition, no significant pathological injury and dysfunction was observed in major visceras. Conclusion We successfully synthesized FA-FePt/DDP nanoliposomes and confirmed their good thermochemotherapeutic effect in AMF and MMRI potential on ovarian cancer, with no obvious side effects, providing a favorable strategy of integrated targeting therapy and diagnosis for ovarian cancer.
Collapse
Affiliation(s)
- Xuefeng Bian
- Imaging Department, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Ting Guo
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Guojie Chen
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Dengyun Nie
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Miao Yue
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Yinxing Zhu
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Mei Lin
- Clinical Medical Laboratory, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| |
Collapse
|
9
|
Ansari SR, Mahajan J, Teleki A. Iron oxide nanoparticles for treatment and diagnosis of chronic inflammatory diseases: A systematic review. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1963. [PMID: 38725229 DOI: 10.1002/wnan.1963] [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: 12/30/2023] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 05/15/2024]
Abstract
Chronic inflammatory conditions are among the most prevalent diseases worldwide. Several debilitating diseases such as atherosclerosis, inflammatory bowel disease, rheumatoid arthritis, and Alzheimer's are linked to chronic inflammation. These conditions often develop into complex and fatal conditions, making early detection and treatment of chronic inflammation crucial. Current diagnostic methods show high variability and do not account for disease heterogeneity and disease-specific proinflammatory markers, often delaying the disease detection until later stages. Furthermore, existing treatment strategies, including high-dose anti-inflammatory and immunosuppressive drugs, have significant side effects and an increased risk of infections. In recent years, superparamagnetic iron oxide nanoparticles (SPIONs) have shown tremendous biomedical potential. SPIONs can function as imaging modalities for magnetic resonance imaging, and as therapeutic agents due to their magnetic hyperthermia capability. Furthermore, the surface functionalization of SPIONs allows the detection of specific disease biomarkers and targeted drug delivery. This systematic review explores the utility of SPIONs against chronic inflammatory disorders, focusing on their dual role as diagnostic and therapeutic agents. We extracted studies indexed in the Web of Science database from the last 10 years (2013-2023), and applied systematic inclusion criteria. This resulted in a final selection of 38 articles, which were analyzed for nanoparticle characteristics, targeted diseases, in vivo and in vitro models used, and the efficacy of the therapeutic or diagnostic modalities. The results revealed that ultrasmall SPIONs are excellent for imaging arterial and neuronal inflammation. Furthermore, novel therapies using SPIONs loaded with chemotherapeutic drugs show promise in the treatment of inflammatory diseases. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Shaquib Rahman Ansari
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jessica Mahajan
- School of Applied Sciences, Abertay University, Dundee, Scotland, UK
| | - Alexandra Teleki
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Rais A, Sharma S, Mishra P, Khan LA, Prasad T. Biocompatible carbon quantum dots as versatile imaging nanotrackers of fungal pathogen - Candida albicans. Nanomedicine (Lond) 2024; 19:671-688. [PMID: 38426561 DOI: 10.2217/nnm-2023-0292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
Aim: The development of carbon quantum dots (C-QDs) as nanotrackers to understand drug-pathogen interactions, virulence and multidrug resistance. Methods: Microwave synthesis of C-QDs was performed using citric acid and polyethylene glycol. Further, in vitro toxicity was evaluated and imaging applications were demonstrated in Candida albicans isolates. Results: Well-dispersed, ultra small C-QDs exhibited no cyto/microbial/reactive oxygen species-mediated toxicity and internalized effectively in Candida yeast and hyphal cells. C-QDs were employed for confocal imaging of drug-sensitive and -resistant cells, and a study of the yeast-to-hyphal transition using atomic force microscopy in Candida was conducted for the first time. Conclusion: These biocompatible C-QDs have promising potential as next-generation nanotrackers for in vitro and in vivo targeted cellular and live imaging, after functionalization with biomolecules and drugs.
Collapse
Affiliation(s)
- Anam Rais
- Special Centre for Nano Science & AIRF, Jawaharlal Nehru University, New Delhi, 110067, India
- Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Shubham Sharma
- Department of Biochemical Engineering & Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Prashant Mishra
- Department of Biochemical Engineering & Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Luqman Ahmad Khan
- Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Tulika Prasad
- Special Centre for Nano Science & AIRF, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
11
|
Moisa SM, Burlacu A, Butnariu LI, Vasile CM, Brinza C, Spoiala EL, Maștaleru A, Leon MM, Rosu ST, Vatasescu R, Cinteză EE. Nanotechnology Innovations in Pediatric Cardiology and Cardiovascular Medicine: A Comprehensive Review. Biomedicines 2024; 12:185. [PMID: 38255290 PMCID: PMC10813221 DOI: 10.3390/biomedicines12010185] [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: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
(1) Background: Nanomedicine, incorporating various nanoparticles and nanomaterials, offers significant potential in medical practice. Its clinical adoption, however, faces challenges like safety concerns, regulatory hurdles, and biocompatibility issues. Despite these, recent advancements have led to the approval of many nanotechnology-based products, including those for pediatric use. (2) Methods: Our approach included reviewing clinical, preclinical, and animal studies, as well as literature reviews from the past two decades and ongoing trials. (3) Results: Nanotechnology has introduced innovative solutions in cardiovascular care, particularly in managing myocardial ischemia. Key developments include drug-eluting stents, nitric oxide-releasing coatings, and the use of magnetic nanoparticles in cardiomyocyte transplantation. These advancements are pivotal for early detection and treatment. In cardiovascular imaging, nanotechnology enables noninvasive assessments. In pediatric cardiology, it holds promise in assisting the development of biological conduits, synthetic valves, and bioartificial grafts for congenital heart defects, and offers new treatments for conditions like dilated cardiomyopathy and pulmonary hypertension. (4) Conclusions: Nanomedicine presents groundbreaking solutions for cardiovascular diseases in both adults and children. It has the potential to transform cardiac care, from enhancing myocardial ischemia treatment and imaging techniques to addressing congenital heart issues. Further research and guideline development are crucial for optimizing its clinical application and revolutionizing patient care.
Collapse
Affiliation(s)
- Stefana Maria Moisa
- Department of Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (E.L.S.)
- “Sfanta Maria” Clinical Emergency Hospital for Children, 700309 Iasi, Romania (S.T.R.)
| | - Alexandru Burlacu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iasi, Romania
| | - Lacramioara Ionela Butnariu
- “Sfanta Maria” Clinical Emergency Hospital for Children, 700309 Iasi, Romania (S.T.R.)
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Corina Maria Vasile
- Pediatric and Adult Congenital Cardiology Department, Centre Hospitalier Universitaire de Bordeaux, 33000 Bordeaux, France;
| | - Crischentian Brinza
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iasi, Romania
| | - Elena Lia Spoiala
- Department of Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (E.L.S.)
| | - Alexandra Maștaleru
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Maria Magdalena Leon
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Solange Tamara Rosu
- “Sfanta Maria” Clinical Emergency Hospital for Children, 700309 Iasi, Romania (S.T.R.)
- Department of Nursing, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Radu Vatasescu
- Cardio-Thoracic Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Clinical Emergency Hospital, 050098 Bucharest, Romania
| | - Eliza Elena Cinteză
- Department of Pediatrics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pediatric Cardiology, “Marie Curie” Emergency Children’s Hospital, 041451 Bucharest, Romania
| |
Collapse
|
12
|
Carrelo H, Escoval AR, Vieira T, Jiménez-Rosado M, Silva JC, Romero A, Soares PIP, Borges JP. Injectable Thermoresponsive Microparticle/Hydrogel System with Superparamagnetic Nanoparticles for Drug Release and Magnetic Hyperthermia Applications. Gels 2023; 9:982. [PMID: 38131968 PMCID: PMC10742759 DOI: 10.3390/gels9120982] [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: 11/10/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Cancer is a disease that continues to greatly impact our society. Developing new and more personalized treatment options is crucial to decreasing the cancer burden. In this study, we combined magnetic polysaccharide microparticles with a Pluronic thermoresponsive hydrogel to develop a multifunctional, injectable drug delivery system (DDS) for magnetic hyperthermia applications. Gellan gum and alginate microparticles were loaded with superparamagnetic iron oxide nanoparticles (SPIONs) with and without coating. The magnetic microparticles' registered temperature increases up to 4 °C upon the application of an alternating magnetic field. These magnetic microparticles were mixed with drug-loaded microparticles, and, subsequently, this mixture was embedded within a Pluronic thermoresponsive hydrogel that is capable of being in the gel state at 37 °C. The proposed DDS was capable of slowly releasing methylene blue, used as a model drug, for up to 9 days. The developed hydrogel/microparticle system had a smaller rate of drug release compared with microparticles alone. This system proved to be a potential thermoresponsive DDS suitable for magnetic hyperthermia applications, thus enabling a synergistic treatment for cancer.
Collapse
Affiliation(s)
- Henrique Carrelo
- CENIMAT/i3N, Department of Materials Science, NOVA School of Science and Technology (FCT NOVA), Campus de Caparica, 2829-516 Caparica, Portugal; (H.C.)
| | - André R. Escoval
- CENIMAT/i3N, Department of Materials Science, NOVA School of Science and Technology (FCT NOVA), Campus de Caparica, 2829-516 Caparica, Portugal; (H.C.)
| | - Tânia Vieira
- CENIMAT/i3N, Department of Physics, NOVA School of Science and Technology (FCT NOVA), Campus de Caparica, 2829-516 Caparica, Portugal
| | | | - Jorge Carvalho Silva
- CENIMAT/i3N, Department of Physics, NOVA School of Science and Technology (FCT NOVA), Campus de Caparica, 2829-516 Caparica, Portugal
| | - Alberto Romero
- Department of Chemical Engineering, Facultad de Química, Universidad de Sevilla, 41012 Sevilla, Spain;
| | - Paula Isabel P. Soares
- CENIMAT/i3N, Department of Materials Science, NOVA School of Science and Technology (FCT NOVA), Campus de Caparica, 2829-516 Caparica, Portugal; (H.C.)
| | - João Paulo Borges
- CENIMAT/i3N, Department of Materials Science, NOVA School of Science and Technology (FCT NOVA), Campus de Caparica, 2829-516 Caparica, Portugal; (H.C.)
| |
Collapse
|
13
|
Yun WS, Cho H, Jeon SI, Lim DK, Kim K. Fluorescence-Based Mono- and Multimodal Imaging for In Vivo Tracking of Mesenchymal Stem Cells. Biomolecules 2023; 13:1787. [PMID: 38136656 PMCID: PMC10742164 DOI: 10.3390/biom13121787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
The advancement of stem cell therapy has offered transformative therapeutic outcomes for a wide array of diseases over the past decades. Consequently, stem cell tracking has become significant in revealing the mechanisms of action and ensuring safe and effective treatments. Fluorescence stands out as a promising choice for stem cell tracking due to its myriad advantages, including high resolution, real-time monitoring, and multi-fluorescence detection. Furthermore, combining fluorescence with other tracking modalities-such as bioluminescence imaging (BLI), positron emission tomography (PET), photoacoustic (PA), computed tomography (CT), and magnetic resonance (MR)-can address the limitations of single fluorescence detection. This review initially introduces stem cell tracking using fluorescence imaging, detailing various labeling strategies such as green fluorescence protein (GFP) tagging, fluorescence dye labeling, and nanoparticle uptake. Subsequently, we present several combinations of strategies for efficient and precise detection.
Collapse
Affiliation(s)
- Wan Su Yun
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; (W.S.Y.); (D.-K.L.)
| | - Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman’s University, Seoul 03760, Republic of Korea; (H.C.); (S.I.J.)
| | - Seong Ik Jeon
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman’s University, Seoul 03760, Republic of Korea; (H.C.); (S.I.J.)
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; (W.S.Y.); (D.-K.L.)
| | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman’s University, Seoul 03760, Republic of Korea; (H.C.); (S.I.J.)
| |
Collapse
|
14
|
Jordan VC, Sojoodi M, Shroff S, Pagan PG, Barrett SC, Wellen J, Tanabe KK, Chung RT, Caravan P, Gale EM. Molecular magnetic resonance imaging of liver inflammation using an oxidatively activated probe. JHEP Rep 2023; 5:100850. [PMID: 37818152 PMCID: PMC10561122 DOI: 10.1016/j.jhepr.2023.100850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 10/12/2023] Open
Abstract
Background & Aims Many liver diseases are driven by inflammation, but imaging to non-invasively diagnose and quantify liver inflammation has been underdeveloped. The inflammatory liver microenvironment is aberrantly oxidising owing in part to reactive oxygen species generated by myeloid leucocytes. We hypothesised that magnetic resonance imaging using the oxidatively activated probe Fe-PyC3A will provide a non-invasive biomarker of liver inflammation. Methods A mouse model of drug-induced liver injury was generated through intraperitoneal injection of a hepatoxic dose of acetaminophen. A mouse model of steatohepatitis was generated via a choline-deficient, l-amino acid defined high-fat diet (CDAHFD). Images were acquired dynamically before and after intravenous injection of Fe-PyC3A. The contrast agent gadoterate meglumine was used as a non-oxidatively activated negative control probe in mice fed CDAHFD. The (post-pre) Fe-PyC3A injection change in liver vs. muscle contrast-to-noise ratio (ΔCNR) recorded 2 min post-injection was correlated with liver function test values, histologic scoring assigned using the NASH Clinical Research Network criteria, and intrahepatic myeloid leucocyte composition determined by flow cytometry. Results For mice receiving i.p. injections of acetaminophen, intrahepatic neutrophil composition correlated poorly with liver test values but positively and significantly with ΔCNR (r = 0.64, p <0.0001). For mice fed CDAHFD, ΔCNR generated by Fe-PyC3A in the left lobe was significantly greater in mice meeting histologic criteria strongly associated with a diagnosis NASH compared to mice where histology was consistent with likely non-NASH (p = 0.0001), whereas no differential effect was observed using gadoterate meglumine. In mice fed CDAHFD, ΔCNR did not correlate strongly with fractional composition of any specific myeloid cell subpopulation as determined by flow cytometry. Conclusions Magnetic resonance imaging using Fe-PyC3A merits further evaluation as a non-invasive biomarker for liver inflammation. Impact and implications Non-invasive tests to diagnose and measure liver inflammation are underdeveloped. Inflammatory cells such as neutrophils release reactive oxygen species which creates an inflammatory liver microenvironment that can drive chemical oxidation. We recently invented a new class of magnetic resonance imaging probe that is made visible to the scanner only after chemical oxidation. Here, we demonstrate how this imaging technology could be applied as a non-invasive biomarker for liver inflammation.
Collapse
Affiliation(s)
- Veronica Clavijo Jordan
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mozhdeh Sojoodi
- Harvard Medical School, Boston, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Stuti Shroff
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Patricia Gonzalez Pagan
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Stephen Cole Barrett
- Harvard Medical School, Boston, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | | | - Kenneth K. Tanabe
- Harvard Medical School, Boston, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Raymond T. Chung
- Harvard Medical School, Boston, MA, USA
- Gastroenterology Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Eric M. Gale
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Tomitaka A, Vashist A, Kolishetti N, Nair M. Machine learning assisted-nanomedicine using magnetic nanoparticles for central nervous system diseases. NANOSCALE ADVANCES 2023; 5:4354-4367. [PMID: 37638161 PMCID: PMC10448356 DOI: 10.1039/d3na00180f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023]
Abstract
Magnetic nanoparticles possess unique properties distinct from other types of nanoparticles developed for biomedical applications. Their unique magnetic properties and multifunctionalities are especially beneficial for central nervous system (CNS) disease therapy and diagnostics, as well as targeted and personalized applications using image-guided therapy and theranostics. This review discusses the recent development of magnetic nanoparticles for CNS applications, including Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, and drug addiction. Machine learning (ML) methods are increasingly applied towards the processing, optimization and development of nanomaterials. By using data-driven approach, ML has the potential to bridge the gap between basic research and clinical research. We review ML approaches used within the various stages of nanomedicine development, from nanoparticle synthesis and characterization to performance prediction and disease diagnosis.
Collapse
Affiliation(s)
- Asahi Tomitaka
- Department of Computer and Information Sciences, College of Natural and Applied Science, University of Houston-Victoria Texas 77901 USA
| | - Arti Vashist
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
- Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
| | - Nagesh Kolishetti
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
- Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
- Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
| |
Collapse
|
16
|
Li Z, Bai R, Yi J, Zhou H, Xian J, Chen C. Designing Smart Iron Oxide Nanoparticles for MR Imaging of Tumors. CHEMICAL & BIOMEDICAL IMAGING 2023; 1:315-339. [PMID: 37501794 PMCID: PMC10369497 DOI: 10.1021/cbmi.3c00026] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 07/29/2023]
Abstract
Iron oxide nanoparticles (IONPs) possess unique magnetism and good biocompatibility, and they have been widely applied as contrast agents (CAs) for magnetic resonance imaging (MRI). Traditional CAs typically show a fixed enhanced signal, thus exhibiting the limitations of low sensitivity and a lack of specificity. Nowadays, the progress of stimulus-responsive IONPs allows alteration of the relaxation signal in response to internal stimuli of the tumor, or external stimuli, thus providing an opportunity to overcome those limitations. This review summarizes the current status of smart IONPs as tumor imaging MRI CAs that exhibit responsiveness to endogenous stimuli, such as pH, hypoxia, glutathione, and enzymes, or exogenous stimuli, such as magnets, light, and so on. We discuss the challenges and future opportunities for IONPs as MRI CAs and comprehensively illustrate the applications of these stimuli-responsive IONPs. This review will help provide guidance for designing IONPs as MRI CAs and further promote the reasonable design of magnetic nanoparticles and achieve early and accurate tumor detection.
Collapse
Affiliation(s)
- Zhenzhen Li
- CAS
Key Laboratory for Biomedical Effects of Nanoparticles and Nanosafety
& CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Department
of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Ru Bai
- CAS
Key Laboratory for Biomedical Effects of Nanoparticles and Nanosafety
& CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- Research
Unit of Nanoscience and Technology, Chinese
Academy of Medical Sciences, Beijing 100021, China
| | - Jia Yi
- Guangdong
Provincial Development and Reform Commission, Guangzhou 510031, China
| | - Huige Zhou
- CAS
Key Laboratory for Biomedical Effects of Nanoparticles and Nanosafety
& CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- Research
Unit of Nanoscience and Technology, Chinese
Academy of Medical Sciences, Beijing 100021, China
| | - Junfang Xian
- Department
of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Chunying Chen
- CAS
Key Laboratory for Biomedical Effects of Nanoparticles and Nanosafety
& CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- Research
Unit of Nanoscience and Technology, Chinese
Academy of Medical Sciences, Beijing 100021, China
- The
GBA National Institute for Nanotechnology Innovation, Guangzhou 510700, China
| |
Collapse
|
17
|
Radu AF, Bungau SG. Nanomedical approaches in the realm of rheumatoid arthritis. Ageing Res Rev 2023; 87:101927. [PMID: 37031724 DOI: 10.1016/j.arr.2023.101927] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Rheumatoid arthritis (RA) is a heterogeneous autoimmune inflammatory disorder defined by the damage to the bone and cartilage in the synovium, which causes joint impairment and an increase in the mortality rate. It is associated with an incompletely elucidated pathophysiological mechanism. Even though disease-modifying antirheumatic drugs have contributed to recent improvements in the standard of care for RA, only a small fraction of patients is able to attain and maintain clinical remission without the necessity for ongoing immunosuppressive drugs. The evolution of tolerance over time as well as patients' inability to respond to currently available therapy can alter the overall management of RA. A significant increase in the research of RA nano therapies due to the possible improvements they may provide over traditional systemic treatments has been observed. New approaches to getting beyond the drawbacks of existing treatments are presented by advancements in the research of nanotherapeutic techniques, particularly drug delivery nano systems. Via passive or active targeting of systemic delivery, therapeutic drugs can be precisely transported to and concentrated in the affected sites. As a result, nanoscale drug delivery systems improve the solubility and bioavailability of certain drugs and reduce dose escalation. In the present paper, we provide a thorough overview of the possible biomedical applications of various nanostructures in the diagnostic and therapeutic management of RA, derived from the shortcomings of conventional therapies. Moreover, the paper suggests the need for improvement on the basis of research directions and properly designed clinical studies.
Collapse
Affiliation(s)
- Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania.
| | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| |
Collapse
|
18
|
Smith L, Kuncic Z, Byrne HL, Waddington D. Nanoparticles for MRI-guided radiation therapy: a review. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00145-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AbstractThe development of nanoparticle agents for MRI-guided radiotherapy is growing at an increasing pace, with clinical trials now underway and many pre-clinical evaluation studies ongoing. Gadolinium and iron-oxide-based nanoparticles remain the most clinically advanced nanoparticles to date, although several promising candidates are currently under varying stages of development. Goals of current and future generation nanoparticle-based contrast agents for MRI-guided radiotherapy include achieving positive signal contrast on T1-weighted MRI scans, local radiation enhancement at clinically relevant concentrations and, where applicable, avoidance of uptake by the reticuloendothelial system. Exploiting the enhanced permeability and retention effect or the use of active targeting ligands on nanoparticle surfaces is utilised to promote tumour uptake. This review outlines the current status of promising nanoparticle agents for MRI-guided radiation therapy, including several platforms currently undergoing clinical evaluation or at various stages of the pre-clinical development process. Challenges facing nanoparticle agents and possible avenues for current and future development are discussed.
Collapse
|
19
|
Dias AMM, Courteau A, Bellaye PS, Kohli E, Oudot A, Doulain PE, Petitot C, Walker PM, Decréau R, Collin B. Superparamagnetic Iron Oxide Nanoparticles for Immunotherapy of Cancers through Macrophages and Magnetic Hyperthermia. Pharmaceutics 2022; 14:2388. [PMID: 36365207 PMCID: PMC9694944 DOI: 10.3390/pharmaceutics14112388] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 07/30/2023] Open
Abstract
Cancer immunotherapy has tremendous promise, but it has yet to be clinically applied in a wider variety of tumor situations. Many therapeutic combinations are envisaged to improve their effectiveness. In this way, strategies capable of inducing immunogenic cell death (e.g., doxorubicin, radiotherapy, hyperthermia) and the reprogramming of the immunosuppressive tumor microenvironment (TME) (e.g., M2-to-M1-like macrophages repolarization of tumor-associated macrophages (TAMs)) are particularly appealing to enhance the efficacy of approved immunotherapies (e.g., immune checkpoint inhibitors, ICIs). Due to their modular construction and versatility, iron oxide-based nanomedicines such as superparamagnetic iron oxide nanoparticles (SPIONs) can combine these different approaches in a single agent. SPIONs have already shown their safety and biocompatibility and possess both drug-delivery (e.g., chemotherapy, ICIs) and magnetic capabilities (e.g., magnetic hyperthermia (MHT), magnetic resonance imaging). In this review, we will discuss the multiple applications of SPIONs in cancer immunotherapy, focusing on their theranostic properties to target TAMs and to generate MHT. The first section of this review will briefly describe immune targets for NPs. The following sections will deal with the overall properties of SPIONs (including MHT). The last section is dedicated to the SPION-induced immune response through its effects on TAMs and MHT.
Collapse
Affiliation(s)
- Alexandre M. M. Dias
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
| | - Alan Courteau
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
- ImViA Laboratory, EA 7535, University of Burgundy, 21000 Dijon, France
| | - Pierre-Simon Bellaye
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079 Dijon, France
| | - Evelyne Kohli
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079 Dijon, France
- University Hospital Centre François Mitterrand, 21000 Dijon, France
| | - Alexandra Oudot
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
| | | | - Camille Petitot
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
| | - Paul-Michael Walker
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
- ImViA Laboratory, EA 7535, University of Burgundy, 21000 Dijon, France
- University Hospital Centre François Mitterrand, 21000 Dijon, France
| | - Richard Decréau
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS/uB 6302, Université de Bourgogne Franche-Comté, 21079 Dijon, France
| | - Bertrand Collin
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS/uB 6302, Université de Bourgogne Franche-Comté, 21079 Dijon, France
| |
Collapse
|
20
|
Masanam HB, Perumal G, Krishnan S, Singh SK, Jha NK, Chellappan DK, Dua K, Gupta PK, Narasimhan AK. Advances and opportunities in nanoimaging agents for the diagnosis of inflammatory lung diseases. Nanomedicine (Lond) 2022; 17:1981-2005. [PMID: 36695290 DOI: 10.2217/nnm-2021-0427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The development of rapid, noninvasive diagnostics to detect lung diseases is a great need after the COVID-2019 outbreak. The nanotechnology-based approach has improved imaging and facilitates the early diagnosis of inflammatory lung diseases. The multifunctional properties of nanoprobes enable better spatial-temporal resolution and a high signal-to-noise ratio in imaging. Targeted nanoimaging agents have been used to bind specific tissues in inflammatory lungs for early-stage diagnosis. However, nanobased imaging approaches for inflammatory lung diseases are still in their infancy. This review provides a solution-focused approach to exploring medical imaging technologies and nanoprobes for the detection of inflammatory lung diseases. Prospects for the development of contrast agents for lung disease detection are also discussed.
Collapse
Affiliation(s)
- Hema Brindha Masanam
- Advanced Nano-Theranostics (ANTs), Biomaterials Lab, Department of Biomedical Engineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, 603 203, India
| | - Govindaraj Perumal
- Department of Conservative Dentistry & Endodontics, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Velappanchavadi, Chennai, 600 077, India.,Department of Biomedical Engineering, Rajalakshmi Engineering College, Thandalam, Chennai, 602 105, India
| | | | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201310, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences & Research (SBSR), Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201310, India.,Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248002, India.,Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Ashwin Kumar Narasimhan
- Advanced Nano-Theranostics (ANTs), Biomaterials Lab, Department of Biomedical Engineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, 603 203, India
| |
Collapse
|
21
|
Evaluation of different 89Zr-labeled synthons for direct labeling and tracking of white blood cells and stem cells in healthy athymic mice. Sci Rep 2022; 12:15646. [PMID: 36123386 PMCID: PMC9485227 DOI: 10.1038/s41598-022-19953-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/07/2022] [Indexed: 11/11/2022] Open
Abstract
Cell based therapies are evolving as an effective new approach to treat various diseases. To understand the safety, efficacy, and mechanism of action of cell-based therapies, it is imperative to follow their biodistribution noninvasively. Positron-emission-tomography (PET)-based non-invasive imaging of cell trafficking offers such a potential. Herein, we evaluated and compared three different ready-to-use direct cell radiolabeling synthons, [89Zr]Zr-DFO-Bn-NCS, [89Zr]Zr-Hy3ADA5-NCS, and [89Zr]Zr-Hy3ADA5-SA for PET imaging-based trafficking of white blood cells (WBCs) and stem cells (SCs) up to 7 days in athymic nude mice. We compared the degree of 89Zr complexation and percentage of cell radiolabeling efficiencies with each. All three synthons, [89Zr]Zr-DFO-Bn-NCS, [89Zr]Zr-Hy3ADA5-NCS, and [89Zr]Zr-Hy3ADA5-SA, were successfully prepared, and used for radiolabeling of WBCs and SCs. The highest cell radiolabeling yield was found for [89Zr]Zr-DFO-Bn-NCS, followed by [89Zr]Zr-Hy3ADA5-NCS, and [89Zr]Zr-Hy3ADA5-SA. In terms of biodistribution, WBCs radiolabeled with [89Zr]Zr-DFO-Bn-NCS or [89Zr]Zr-Hy3ADA5-NCS, were primarily accumulated in liver and spleen, whereas SCs radiolabeled with [89Zr]Zr-DFO-Bn-NCS or [89Zr]Zr-Hy3ADA5-NCS were found in lung, liver and spleen. A high bone uptake was observed for both WBCs and SCs radiolabeled with [89Zr]Zr-Hy3ADA5-SA, suggesting in-vivo instability of [89Zr]Zr-Hy3ADA5-SA synthon. This study offers an appropriate selection of ready-to-use radiolabeling synthons for noninvasive trafficking of WBCs, SCs and other cell-based therapies.
Collapse
|
22
|
Theus AS, Ning L, Kabboul G, Hwang B, Tomov ML, LaRock CN, Bauser-Heaton H, Mahmoudi M, Serpooshan V. 3D bioprinting of nanoparticle-laden hydrogel scaffolds with enhanced antibacterial and imaging properties. iScience 2022; 25:104947. [PMID: 36065192 PMCID: PMC9440295 DOI: 10.1016/j.isci.2022.104947] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/13/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
Biomaterial-associated microbial contaminations in biologically conducive three-dimensional (3D) tissue-engineered constructs have significantly limited the clinical applications of scaffold systems. To prevent such infections, antimicrobial biomaterials are rapidly evolving. Yet, the use of such materials in bioprinting-based approaches of scaffold fabrication has not been examined. This study introduces a new generation of bacteriostatic gelatin methacryloyl (GelMA)-based bioinks, incorporated with varying doses of antibacterial superparamagnetic iron oxide nanoparticles (SPIONs). The SPION-laden GelMA scaffolds showed significant resistance against the Staphylococcus aureus growth, while providing a contrast in magnetic resonance imaging. We simulated the bacterial contamination of cellular 3D GelMA scaffolds in vitro and demonstrated the significant effect of functionalized scaffolds in inhibiting bacterial growth, while maintaining cell viability and growth. Together, these results present a new promising class of functionalized bioinks to 3D bioprint tissue-engineered scaffold with markedly enhanced properties for the use in a variety of in vitro and clinical applications.
Collapse
Affiliation(s)
- Andrea S. Theus
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Gabriella Kabboul
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Boeun Hwang
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Martin L. Tomov
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Christopher N. LaRock
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Holly Bauser-Heaton
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Sibley Heart Center at Children’s Healthcare of Atlanta, Atlanta, GA 30342, USA
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI 48842, USA
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
23
|
Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol 2022; 15:132. [PMID: 36096856 PMCID: PMC9469622 DOI: 10.1186/s13045-022-01320-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Additionally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we comprehensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute to further improvements in clinical practice.
Collapse
Affiliation(s)
- Hailong Tian
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Tingting Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiayan Shi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Edouard C Nice
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China
| | - Na Xie
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China.
| | - Canhua Huang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
24
|
Emerging trends in the nanomedicine applications of functionalized magnetic nanoparticles as novel therapies for acute and chronic diseases. J Nanobiotechnology 2022; 20:393. [PMID: 36045375 PMCID: PMC9428876 DOI: 10.1186/s12951-022-01595-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/13/2022] [Indexed: 11/10/2022] Open
Abstract
High-quality point-of-care is critical for timely decision of disease diagnosis and healthcare management. In this regard, biosensors have revolutionized the field of rapid testing and screening, however, are confounded by several technical challenges including material cost, half-life, stability, site-specific targeting, analytes specificity, and detection sensitivity that affect the overall diagnostic potential and therapeutic profile. Despite their advances in point-of-care testing, very few classical biosensors have proven effective and commercially viable in situations of healthcare emergency including the recent COVID-19 pandemic. To overcome these challenges functionalized magnetic nanoparticles (MNPs) have emerged as key players in advancing the biomedical and healthcare sector with promising applications during the ongoing healthcare crises. This critical review focus on understanding recent developments in theranostic applications of functionalized magnetic nanoparticles (MNPs). Given the profound global economic and health burden, we discuss the therapeutic impact of functionalized MNPs in acute and chronic diseases like small RNA therapeutics, vascular diseases, neurological disorders, and cancer, as well as for COVID-19 testing. Lastly, we culminate with a futuristic perspective on the scope of this field and provide an insight into the emerging opportunities whose impact is anticipated to disrupt the healthcare industry.
Collapse
|
25
|
Nanotechnology in the Diagnosis and Treatment of Osteomyelitis. Pharmaceutics 2022; 14:pharmaceutics14081563. [PMID: 36015188 PMCID: PMC9412360 DOI: 10.3390/pharmaceutics14081563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Infection remains one of the largest threats to global health. Among those infections that are especially troublesome, osteomyelitis, or inflammation of the bone, typically due to infection, is a particularly difficult condition to diagnose and treat. This difficulty stems not only from the biological complexities of opportunistic infections designed to avoid the onslaught of both the host immune system as well as exogenous antibiotics, but also from changes in the host vasculature and the heterogeneity of infectious presentations. While several groups have attempted to classify and stage osteomyelitis, controversy remains, often delaying diagnosis and treatment. Despite a host of preclinical treatment advances being incubated in academic and company research and development labs worldwide, clinical treatment strategies remain relatively stagnant, including surgical debridement and lengthy courses of intravenous antibiotics, both of which may compromise the overall health of the bone and the patient. This manuscript reviews the current methods for diagnosing and treating osteomyelitis and then contemplates the role that nanotechnology might play in the advancement of osteomyelitis treatment.
Collapse
|
26
|
Iravani K, Mehrabani D, Doostkam A, Azarpira N, Iranpour P, Bahador M, Mehravar S. Use of MRI to Assess the Regenerative Effects of Adipose Tissue Derived Mesenchymal Stem Cells in a Rabbit Cartilaginous Laryngeal Defect Model. Curr Ther Res Clin Exp 2022; 97:100682. [PMID: 35959231 PMCID: PMC9361331 DOI: 10.1016/j.curtheres.2022.100682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
Background Stenosis and scar formation after repair of laryngeal tissue defects are serious problems that can significantly influence a patient's quality of life. Objective In this study, we evaluated the use of magnetic resonance imaging to assess the efficacy of adipose tissue-derived mesenchymal stem cells (ASCs) on cartilaginous regeneration in an experimental rabbit model. Methods Ten male white Dutch rabbits each had a 5 mm cartilaginous defect created surgically in the right and left thyroid lamina. On the right side, ASCs labeled with iron oxide particles were infused. As a control, the left side was left untreated. Repair of the defects were then evaluated by direct observation, histological evaluation, and magnetic resonance imaging monitoring done on days 1, 7, 14, and 28. Results Histological examination revealed that compared with control, transplanted ASCs significantly increased cartilage regeneration (P ˂ 0.001), reduced inflammation (P ˂ 0.001), and fibrosis (P = 0.050). Magnetic resonance imaging tracking showed accurate placement and viability of the infused ASCs, as evidenced by low signal intensity onT2 weighted images at the level of the right thyroid cartilage. Conclusions Infusion of ASCs improved laryngeal regeneration of surgically induced cartilaginous defects while decreasing fibrous tissue formation in this in vivo rabbit model. Furthermore, magnetic resonance imaging was shown to be a useful, noninvasive method to track correct ASCs placement and viability in cartilage regeneration in this animal model.
Collapse
Affiliation(s)
- Kamyar Iravani
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
- Address correspondence to: Kamyar Iravani, MD, Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Khalili Hospital, Khalili St, Shiraz, Iran, Postal code: 71936-16641.
| | - Davood Mehrabani
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Doostkam
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz Institute of Stem Cell and Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooya Iranpour
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Bahador
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Mehravar
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
27
|
Papachristou M, Priftakis D, Xanthopoulos S, Datseris I, Bouziotis P. Biodistribution of intravenous [ 99mTc]Tc-phytate in mouse models of chemically and foreign-body induced sterile inflammation. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2022; 12:91-98. [PMID: 35874295 PMCID: PMC9301090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 06/05/2022] [Indexed: 06/15/2023]
Abstract
When injected intravenously, [99mTc]Tc-phytate forms particles in the nanometer range. This size can favor its extravasation into tumor and inflammation through pores of the vasculature. The aim of this work is the evaluation of the use of [99mTc]Tc-phytate to assess sterile inflammation in mouse models. Biodistribution studies of [99mTc]Tc-phytate were performed in two groups of male Swiss Albino mice. Sterile inflammation was induced after intramuscular injection of turpentine in the first group (chemically induced sterile inflammation model) and after implantation of sterile metal bolts in the second group (foreign-body induced sterile inflammation model). [99mTc]Tc-phytate was intravenously injected after the development of inflammation in both groups and ex vivo biodistribution of the radiolabelled complex followed at different time-points. Biodistribution was expressed as percent injected dose per gram (%ID/g). Target-to-background ratios were also recorded. For the chemically induced sterile inflammation model, ex vivo biodistribution evaluation measurements revealed a pronounced uptake in the inflamed muscle when compared to uptake in the control/non-inflamed muscle. Moreover, as expected, there is a high uptake in the liver and spleen. For the foreign-body induced sterile inflammation model, a significantly higher uptake was observed in the inflamed muscle post [99mTc]Tc-phytate injection, both for the 24 hours post-bolt implantation and for the 7 days post-bolt implantation groups. The nanoparticle properties of [99mTc]Tc-phytate are potentially useful in the imaging of different types of sterile inflammation with translational potential clinical SPECT (single photon emission computed tomography) imaging applications in humans.
Collapse
Affiliation(s)
- Maria Papachristou
- Nuclear Medicine and PET/CT Department, General Hospital of Athens “Evaggelismos”Athens, Greece
| | - Dimitrios Priftakis
- Institute of Nuclear Medicine, University College London HospitalLondon, United Kingdom
| | - Stavros Xanthopoulos
- Radiochemical Studies Laboratory, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”Athens, Greece
| | - Ioannis Datseris
- Nuclear Medicine and PET/CT Department, General Hospital of Athens “Evaggelismos”Athens, Greece
| | - Penelope Bouziotis
- Radiochemical Studies Laboratory, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”Athens, Greece
| |
Collapse
|
28
|
Pucci C, Degl'Innocenti A, Belenli Gümüş M, Ciofani G. Superparamagnetic iron oxide nanoparticles for magnetic hyperthermia: Recent advancements, molecular effects, and future directions in the omics era. Biomater Sci 2022; 10:2103-2121. [DOI: 10.1039/d1bm01963e] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Superparamagnetic iron oxide nanoparticles have attracted attention in the biomedical field thanks to their ability to prompt hyperthermia in response to an alternated magnetic field. Hyperthermia is well-known for inducing...
Collapse
|
29
|
Wang J, Yang J, Kopeček J. Nanomedicines in B cell-targeting therapies. Acta Biomater 2022; 137:1-19. [PMID: 34687954 PMCID: PMC8678319 DOI: 10.1016/j.actbio.2021.10.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
B cells play multiple roles in immune responses related to autoimmune diseases as well as different types of cancers. As such, strategies focused on B cell targeting attracted wide interest and developed intensively. There are several common mechanisms various B cell targeting therapies have relied on, including direct B cell depletion, modulation of B cell antigen receptor (BCR) signaling, targeting B cell survival factors, targeting the B cell and T cell costimulation, and immune checkpoint blockade. Nanocarriers, used as drug delivery vehicles, possess numerous advantages to low molecular weight drugs, reducing drug toxicity, enhancing blood circulation time, as well as augmenting targeting efficacy and improving therapeutic effect. Herein, we review the commonly used targets involved in B cell targeting approaches and the utilization of various nanocarriers as B cell-targeted delivery vehicles. STATEMENT OF SIGNIFICANCE: As B cells are engaged significantly in the development of many kinds of diseases, utilization of nanomedicines in B cell depletion therapies have been rapidly developed. Although numerous studies focused on B cell targeting have already been done, there are still various potential receptors awaiting further investigation. This review summarizes the most relevant studies that utilized nanotechnologies associated with different B cell depletion approaches, providing a useful tool for selection of receptors, agents and/or nanocarriers matching specific diseases. Along with uncovering new targets in the function map of B cells, there will be a growing number of candidates that can benefit from nanoscale drug delivery.
Collapse
Affiliation(s)
- Jiawei Wang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
30
|
Zhang Z, Wells CJR, Davies GL, Williams GR. The effect of formulation morphology on stimuli-triggered co-delivery of chemotherapeutic and MRI contrast agents. Int J Pharm 2021; 609:121155. [PMID: 34624442 DOI: 10.1016/j.ijpharm.2021.121155] [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: 07/20/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 10/20/2022]
Abstract
Most conventional chemotherapeutics have narrow therapeutic windows, and thus their delivery remains challenging and often raises safety and efficacy concerns. Theranostic platforms, with simultaneous encapsulation of therapeutic and diagnostic agents, have been proposed as next-generation formulations which can overcome this issue. In this work, we used electrohydrodynamic approaches to fabricate core@shell formulations comprising a pH responsive Eudragit L100 shell embedded with superparamagnetic iron oxide nanoparticles (SPIONs), and a thermo-responsive poly(N-isopropylacrylamide) (PNIPAM)/ethyl cellulose core loaded with the model drug carmofur. By varying the weight ratio of core polymer to shell polymer, the morphology of PNIPAM/ethyl cellulose@Eudragit L100 microparticles could be changed from concave to spherical. Smooth cylindrical fibres could also be generated. All the formulations exist as amorphous solid dispersions of drug-in-polymer, with distinct core@shell architectures. The fibres have clear thermo-responsive drug release profiles, while no thermo-responsive properties can be seen with the particles. All the formulations can protect SPIONs from degradation in gastric fluids (pH ∼ 1.5), and around the physiological pH range the materials offer effective and pH-responsive relaxivity. The r2 values also display clear linear relationships with drug release data, suggesting the potential of using MRI signals to track drug release in vivo. Mathematical equations were established to track drug release in vitro, with very similar experimental and predicted release profiles obtained.
Collapse
Affiliation(s)
- Ziwei Zhang
- UCL School of Pharmacy, University College London, 29 - 39 Brunswick Square, London WC1N 1AX, UK; UCL Department of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, UK
| | - Connor J R Wells
- UCL Department of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, UK
| | - Gemma-Louise Davies
- UCL Department of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, UK.
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29 - 39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
31
|
Etemadi H, Buchanan JK, Kandile NG, Plieger PG. Iron Oxide Nanoparticles: Physicochemical Characteristics and Historical Developments to Commercialization for Potential Technological Applications. ACS Biomater Sci Eng 2021; 7:5432-5450. [PMID: 34786932 DOI: 10.1021/acsbiomaterials.1c00938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Iron oxide nanoparticles (IONPs) have gained increasing attention in various biomedical and industrial sectors due to their physicochemical and magnetic properties. In the biomedical field, IONPs are being developed for enzyme/protein immobilization, magnetofection, cell labeling, DNA detection, and tissue engineering. However, in some established areas, such as magnetic resonance imaging (MRI), magnetic drug targeting (MDT), magnetic fluid hyperthermia (MFH), immunomagnetic separation (IMS), and magnetic particle imaging (MPI), IONPs have crossed from the research bench, received clinical approval, and have been commercialized. Additionally, in industrial sectors IONP-based fluids (ferrofluids) have been marketed in electronic and mechanical devices for some time. This review explores the historical evolution of IONPs to their current state in biomedical and industrial applications.
Collapse
Affiliation(s)
- Hossein Etemadi
- School of Fundamental Sciences, Massey University, Private Bag 11 222, Palmerston North 4410, New Zealand
| | - Jenna K Buchanan
- School of Fundamental Sciences, Massey University, Private Bag 11 222, Palmerston North 4410, New Zealand
| | - Nadia G Kandile
- Department of Chemistry, Faculty of Women, Ain Shams University, Heliopolis 11757, Cairo, Egypt
| | - Paul G Plieger
- School of Fundamental Sciences, Massey University, Private Bag 11 222, Palmerston North 4410, New Zealand
| |
Collapse
|
32
|
Montague SJ, Patel P, Martin EM, Slater A, Quintanilla LG, Perrella G, Kardeby C, Nagy M, Mezzano D, Mendes PM, Watson SP. Platelet activation by charged ligands and nanoparticles: platelet glycoprotein receptors as pattern recognition receptors. Platelets 2021; 32:1018-1030. [PMID: 34266346 DOI: 10.1080/09537104.2021.1945571] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 02/08/2023]
Abstract
Charge interactions play a critical role in the activation of the innate immune system by damage- and pathogen-associated molecular pattern receptors. The ability of these receptors to recognize a wide spectrum of ligands through a common mechanism is critical in host defense. In this article, we argue that platelet glycoprotein receptors that signal through conserved tyrosine-based motifs function as pattern recognition receptors (PRRs) for charged endogenous and exogenous ligands, including sulfated polysaccharides, charged proteins and nanoparticles. This is exemplified by GPVI, CLEC-2 and PEAR1 which are activated by a wide spectrum of endogenous and exogenous ligands, including diesel exhaust particles, sulfated polysaccharides and charged surfaces. We propose that this mechanism has evolved to drive rapid activation of platelets at sites of injury, but that under some conditions it can drive occlusive thrombosis, for example, when blood comes into contact with infectious agents or toxins. In this Opinion Article, we discuss mechanisms behind charge-mediated platelet activation and opportunities for designing nanoparticles and related agents such as dendrimers as novel antithrombotics.
Collapse
Affiliation(s)
- Samantha J Montague
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Pushpa Patel
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, UK
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Lourdes Garcia Quintanilla
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Caroline Kardeby
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Magdolna Nagy
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Diego Mezzano
- Laboratorio de Trombosis y Hemostasia, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Paula M Mendes
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, UK
| |
Collapse
|
33
|
MacRitchie N, Di Francesco V, Ferreira MFMM, Guzik TJ, Decuzzi P, Maffia P. Nanoparticle theranostics in cardiovascular inflammation. Semin Immunol 2021; 56:101536. [PMID: 34862118 PMCID: PMC8811479 DOI: 10.1016/j.smim.2021.101536] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/30/2022]
Abstract
Theranostics, literally derived from the combination of the words diagnostics and therapy, is an emerging field of clinical and preclinical research, where contrast agents, drugs and diagnostic techniques are combined to simultaneously diagnose and treat pathologies. Nanoparticles are extensively employed in theranostics due to their potential to target specific organs and their multifunctional capacity. In this review, we will discuss the current state of theranostic nanomedicine, providing key examples of its application in the imaging and treatment of cardiovascular inflammation.
Collapse
Affiliation(s)
- Neil MacRitchie
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
| | - Valentina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | | | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
34
|
Canese R, Vurro F, Marzola P. Iron Oxide Nanoparticles as Theranostic Agents in Cancer Immunotherapy. NANOMATERIALS 2021; 11:nano11081950. [PMID: 34443781 PMCID: PMC8399455 DOI: 10.3390/nano11081950] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/13/2021] [Accepted: 07/22/2021] [Indexed: 11/16/2022]
Abstract
Starting from the mid-1990s, several iron oxide nanoparticles (NPs) were developed as MRI contrast agents. Since their sizes fall in the tenths of a nanometer range, after i.v. injection these NPs are preferentially captured by the reticuloendothelial system of the liver. They have therefore been proposed as liver-specific contrast agents. Even though their unfavorable cost/benefit ratio has led to their withdrawal from the market, innovative applications have recently prompted a renewal of interest in these NPs. One important and innovative application is as diagnostic agents in cancer immunotherapy, thanks to their ability to track tumor-associated macrophages (TAMs) in vivo. It is worth noting that iron oxide NPs may also have a therapeutic role, given their ability to alter macrophage polarization. This review is devoted to the most recent advances in applications of iron oxide NPs in tumor diagnosis and therapy. The intrinsic therapeutic effect of these NPs on tumor growth, their capability to alter macrophage polarization and their diagnostic potential are examined. Innovative strategies for NP-based drug delivery in tumors (e.g., magnetic resonance targeting) will also be described. Finally, the review looks at their role as tracers for innovative, and very promising, imaging techniques (magnetic particle imaging-MPI).
Collapse
Affiliation(s)
- Rossella Canese
- MRI Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy
- Correspondence: (R.C.); (P.M.)
| | - Federica Vurro
- Department of Computer Science, University of Verona, 37134 Verona, Italy;
| | - Pasquina Marzola
- Department of Computer Science, University of Verona, 37134 Verona, Italy;
- Correspondence: (R.C.); (P.M.)
| |
Collapse
|
35
|
Sillerud LO, Neuwelt AJ, Staquicini FI, Arap W, Pasqualini R. Repurposing Ferumoxytol as a Breast Cancer-Associated Macrophage Tracer with Five-Dimensional Quantitative [Fe]MRI of SPION Dynamics. Cancers (Basel) 2021; 13:cancers13153802. [PMID: 34359704 PMCID: PMC8345165 DOI: 10.3390/cancers13153802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/11/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary With the incorporation of immune-modulating therapies into the standard management of triple-negative breast cancer, there is increased interest in the non-invasive imaging of the tumor immune microenvironment. Ferumoxytol is FDA-approved as an iron replacement therapy for iron-deficiency anemia and is also a superparamagnetic iron oxide nanoparticle (SPION) resulting in negative enhancement on T2-weighted MR imaging. It has previously been established that ferumoxytol is taken up by macrophages. In the current study, we used ferumoxytol-contrasted MRI to quantitatively image the iron concentration, and, by extension, the tumor-associated macrophage infiltration within the tumor microenvironment of a highly inflammatory model of triple-negative breast cancer. Abstract Tumor-associated macrophages (TAMs) in breast cancer regulate inflammation, immunosuppression, angiogenesis, and metastasis. However, TAM imaging remains a clinical challenge. Ferumoxytol has long been an FDA-approved superparamagnetic iron oxide nanoparticle (SPION) preparation used as an intravenous (IV) treatment for iron-deficiency anemia. Given its high transverse relaxivity, ferumoxytol produces a negative image contrast upon cellular uptake in T2-weighted magnetic resonance imaging (MRI) studies. Here we evaluated ferumoxytol as a contrast agent to image/quantify TAMs in an aggressive mouse model of breast cancer: We developed [Fe]MRI to measure the 5-dimensional function c(x,y,z,t), where c is the concentration of nanoparticle iron and {x,y,z,t} is the 4-dimensional set of tumor space-time coordinates. Ferumoxytol SPIONs are readily phagocytosed (~104/cell) by the F4/80+CD11b+ TAMs within breast tumors. Quantitative [Fe]MRIs served to determine both the spatial and the temporal distribution of the SPION iron, and hence to measure [Fe] = c(x,y,z,t), a surrogate for TAM density. In single-dose pharmacokinetic studies, after an IV dose of 5 mg/Kg iron, [Fe]MRI measurements showed that c(x,y,z,t) within breast tumors peaked around [Fe] = 70 μM at 42 h post-administration, and decayed below the [Fe]MRI detection limit (~2 μM) by day 7. There was no SPION uptake in control organs (muscle and adipose tissue). Optical microscopy of tissue sections confirmed that F4/80+CD11b+ TAMs infiltrated the tumors and accumulated SPION iron. Our methodology and findings have translational applications for breast cancer patients.
Collapse
Affiliation(s)
- Laurel O. Sillerud
- Department of Neurology, UNM BRaIN Center, University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
- Correspondence: (L.O.S.); (R.P.)
| | - Alexander J. Neuwelt
- Division of Hematology, Oncology and Palliative Care, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA;
- Department of Medical Oncology, Veterans Affairs Medical Center, Richmond, VA 23249, USA
| | - Fernanda I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07103, USA; (F.I.S.); (W.A.)
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07103, USA; (F.I.S.); (W.A.)
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07103, USA; (F.I.S.); (W.A.)
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
- Correspondence: (L.O.S.); (R.P.)
| |
Collapse
|
36
|
Tang C, Wang Q, Li K, Li X, Wang C, Xue L, Ju C, Zhang C. A neutrophil-mimetic magnetic nanoprobe for molecular magnetic resonance imaging of stroke-induced neuroinflammation. Biomater Sci 2021; 9:5247-5258. [PMID: 34137404 DOI: 10.1039/d1bm00566a] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Neuroinflammation plays a key role in the progression of brain injury induced by stroke, and has become a promising target for therapeutic intervention for stroke. Monitoring this pivotal process of neuroinflammation is highly desirable to guide specific therapy. However, there is still a lack of a satisfactory nanoprobe to selectively monitor neuroinflammation. As endothelial cell activation is a hallmark of neuroinflammation, it would be clinically relevant to develop a non-invasive in vivo imaging technique to detect the endothelial activation process. Herein, inspired by the specific neutrophil-endothelium interaction, we designed neutrophil-camouflaged magnetic nanoprobes (NMNPs) that can be used to target activated endothelial cells for improved neuroinflammation imaging. NMNPs are composed of an inner core of superparamagnetic iron oxide (SPIO)-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles and a biomimetic outer shell of a neutrophil membrane, which maintained the biocompatibility and targeting ability of neutrophils and the excellent contrast effects of SPIO. Moreover, we demonstrated that NMNPs can successfully bind to inflamed cerebral vasculature using the intravital imaging of live cerebral microvessels in transient middle cerebral artery occlusion (tMCAO) mice. After that, NMNPs could further accumulate in the brain vasculature and exhibit excellent contrast effects for stroke-induced neuroinflammation and biosafety. We believe that the neutrophil-camouflaged magnetic nanoprobe could serve as a highly safe and selective nanoprobe for neuroinflammation imaging and has alluring prospects for clinical application.
Collapse
Affiliation(s)
- Chunming Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, P.R. China.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Nguyen SM, Wiepz GJ, Schotzko M, Simmons HA, Mejia A, Ludwig KD, Zhu A, Brunner K, Hernando D, Reeder SB, Wieben O, Johnson K, Shah D, Golos TG. Impact of ferumoxytol magnetic resonance imaging on the rhesus macaque maternal-fetal interface†. Biol Reprod 2021; 102:434-444. [PMID: 31511859 DOI: 10.1093/biolre/ioz181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/05/2019] [Accepted: 09/03/2019] [Indexed: 01/26/2023] Open
Abstract
Ferumoxytol is a superparamagnetic iron oxide nanoparticle used off-label as an intravascular magnetic resonance imaging (MRI) contrast agent. Additionally, ferumoxytol-uptake by macrophages facilitates detection of inflammatory sites by MRI through ferumoxytol-induced image contrast changes. Therefore, ferumoxytol-enhanced MRI holds great potential for assessing vascular function and inflammatory response, critical to determine placental health in pregnancy. This study sought to assess the fetoplacental unit and selected maternal tissues, pregnancy outcomes, and fetal well-being after ferumoxytol administration. In initial developmental studies, seven pregnant rhesus macaques were imaged with or without ferumoxytol administration. Pregnancies went to term with vaginal delivery and infants showed normal growth rates compared to control animals born the same year that did not undergo MRI. To determine the impact of ferumoxytol on the maternal-fetal interface (MFI), fetal well-being, and pregnancy outcome, four pregnant rhesus macaques at ~100 gestational day underwent MRI before and after ferumoxytol administration. Collection of the fetoplacental unit and selected maternal tissues was performed 2-3 days following ferumoxytol administration. A control group that did not receive ferumoxytol or MRI was used for comparison. Iron levels in fetal and MFI tissues did not differ between groups, and there was no significant difference in tissue histopathology with or without exposure to ferumoxytol, and no effect on placental hormone secretion. Together, these results suggest that the use of ferumoxytol and MRI in pregnant rhesus macaques does not negatively impact the MFI and can be a valuable experimental tool in research with this important animal model.
Collapse
Affiliation(s)
- Sydney M Nguyen
- Wisconsin National Primate Research Center (WNPRC), Madison, Wisconsin, USA.,Obstetrics & Gynecology, University of Wisconsin Madison School of Medicine, Madison, Wisconsin, USA
| | - Gregory J Wiepz
- Wisconsin National Primate Research Center (WNPRC), Madison, Wisconsin, USA
| | - Michele Schotzko
- Wisconsin National Primate Research Center (WNPRC), Madison, Wisconsin, USA
| | - Heather A Simmons
- Wisconsin National Primate Research Center (WNPRC), Madison, Wisconsin, USA
| | - Andres Mejia
- Wisconsin National Primate Research Center (WNPRC), Madison, Wisconsin, USA
| | - Kai D Ludwig
- Medical Physics, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Ante Zhu
- Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA.,Radiology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Kevin Brunner
- Wisconsin National Primate Research Center (WNPRC), Madison, Wisconsin, USA.,Emergency Medicine, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Diego Hernando
- Medical Physics, University of Wisconsin Madison, Madison, Wisconsin, USA.,Radiology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Scott B Reeder
- Medical Physics, University of Wisconsin Madison, Madison, Wisconsin, USA.,Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA.,Radiology, University of Wisconsin Madison, Madison, Wisconsin, USA.,Emergency Medicine, University of Wisconsin Madison, Madison, Wisconsin, USA.,Medicine, University of Wisconsin Madison, Madison, Wisconsin, USA, and
| | - Oliver Wieben
- Medical Physics, University of Wisconsin Madison, Madison, Wisconsin, USA.,Radiology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Kevin Johnson
- Medical Physics, University of Wisconsin Madison, Madison, Wisconsin, USA.,Radiology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Dinesh Shah
- Obstetrics & Gynecology, University of Wisconsin Madison School of Medicine, Madison, Wisconsin, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center (WNPRC), Madison, Wisconsin, USA.,Obstetrics & Gynecology, University of Wisconsin Madison School of Medicine, Madison, Wisconsin, USA.,Comparative Biosciences, University of Wisconsin Madison, Madison, Wisconsin, USA
| |
Collapse
|
38
|
Zhu FD, Hu YJ, Yu L, Zhou XG, Wu JM, Tang Y, Qin DL, Fan QZ, Wu AG. Nanoparticles: A Hope for the Treatment of Inflammation in CNS. Front Pharmacol 2021; 12:683935. [PMID: 34122112 PMCID: PMC8187807 DOI: 10.3389/fphar.2021.683935] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, an inflammatory response within the central nervous system (CNS), is a main hallmark of common neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), among others. The over-activated microglia release pro-inflammatory cytokines, which induces neuronal death and accelerates neurodegeneration. Therefore, inhibition of microglia over-activation and microglia-mediated neuroinflammation has been a promising strategy for the treatment of neurodegenerative diseases. Many drugs have shown promising therapeutic effects on microglia and inflammation. However, the blood–brain barrier (BBB)—a natural barrier preventing brain tissue from contact with harmful plasma components—seriously hinders drug delivery to the microglial cells in CNS. As an emerging useful therapeutic tool in CNS-related diseases, nanoparticles (NPs) have been widely applied in biomedical fields for use in diagnosis, biosensing and drug delivery. Recently, many NPs have been reported to be useful vehicles for anti-inflammatory drugs across the BBB to inhibit the over-activation of microglia and neuroinflammation. Therefore, NPs with good biodegradability and biocompatibility have the potential to be developed as an effective and minimally invasive carrier to help other drugs cross the BBB or as a therapeutic agent for the treatment of neuroinflammation-mediated neurodegenerative diseases. In this review, we summarized various nanoparticles applied in CNS, and their mechanisms and effects in the modulation of inflammation responses in neurodegenerative diseases, providing insights and suggestions for the use of NPs in the treatment of neuroinflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yu-Jiao Hu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Anesthesia, Southwest Medical University, Luzhou, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qing-Ze Fan
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
39
|
Zhang Z, Wells CJR, King AM, Bear JC, Davies GL, Williams GR. pH-Responsive nanocomposite fibres allowing MRI monitoring of drug release. J Mater Chem B 2021; 8:7264-7274. [PMID: 32642748 DOI: 10.1039/d0tb01033b] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Magnetic resonance imaging (MRI) is one of the most widely-used non-invasive clinical imaging tools, producing detailed anatomical images whilst avoiding side effects such as trauma or X-ray radiation exposure. In this article, a new approach to non-invasive monitoring of drug release from a delivery vehicle via MRI was developed, using pH-responsive Eudragit L100 and S100 fibres encapsulating superparamagnetic iron oxide nanoparticles (SPIONs) and carmofur (a drug used in the treatment of colon cancer). Fibres were prepared by electrospinning, and found to be smooth and cylindrical with diameters of 645 ± 225 nm for L100 and 454 ± 133 nm for S100. The fibres exhibited pH responsive dissolution behaviour. Around the physiological pH range, clear pH-responsive proton relaxation rate changes due to matrix swelling/dissolution can be observed: r2 values of L100 fibres increase from 29.3 ± 8.3 to 69.8 ± 2.5 mM-1 s-1 over 3 h immersion in a pH 7.4 medium, and from 13.5 ± 2.0 mM-1 s-1 to 42.1 ± 3.0 mM-1 s-1 at pH 6.5. The r2 values of S100 fibres grow from 30.4 ± 4.4 to 64.7 ± 1.0 mM-1 s-1 at pH 7.4, but at pH 6.5, where the S100 fibres are not soluble, r2 remains very low (< 4 mM-1 s-1). These dramatic changes in relaxivity demonstrate that pH-responsive dissolution results in SPION release. In vitro drug release studies showed the formulations gave rapid release of carmofur at physiological pH values (pH 6.5 and 7.4), and acid stability studies revealed that they can protect the SPIONs from digestion in acid environments, giving the fibres potential for oral administration. Exploration of the relationship between relaxivity and carmofur release suggests a linear correlation (R2 > 0.94) between the two. Mathematical equations were developed to predict carmofur release in vitro, with very similar experimental and predicted release profiles obtained. Therefore, the formulations developed herein have the potential to be used for non-invasive monitoring of drug release in vivo, and could ultimately result in dramatic reductions to off-target side effects from interventions such as chemotherapy.
Collapse
Affiliation(s)
- Ziwei Zhang
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK. and UCL Department of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, UK.
| | - Connor J R Wells
- UCL Department of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, UK.
| | - Aaron M King
- UCL Department of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, UK.
| | - Joseph C Bear
- Department of Chemical and Pharmaceutical Sciences, Kingston University, Penrhyn Rd, Kingston upon Thames, KT1 2EE, UK
| | - Gemma-Louise Davies
- UCL Department of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, UK.
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
40
|
Bednarczyk M, Medina-Montano C, Fittler FJ, Stege H, Roskamp M, Kuske M, Langer C, Vahldieck M, Montermann E, Tubbe I, Röhrig N, Dzionek A, Grabbe S, Bros M. Complement-Opsonized Nano-Carriers Are Bound by Dendritic Cells (DC) via Complement Receptor (CR)3, and by B Cell Subpopulations via CR-1/2, and Affect the Activation of DC and B-1 Cells. Int J Mol Sci 2021; 22:2869. [PMID: 33799879 PMCID: PMC8001596 DOI: 10.3390/ijms22062869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
The development of nanocarriers (NC) for biomedical applications has gained large interest due to their potential to co-deliver drugs in a cell-type-targeting manner. However, depending on their surface characteristics, NC accumulate serum factors, termed protein corona, which may affect their cellular binding. We have previously shown that NC coated with carbohydrates to enable biocompatibility triggered the lectin-dependent complement pathway, resulting in enhanced binding to B cells via complement receptor (CR)1/2. Here we show that such NC also engaged all types of splenic leukocytes known to express CR3 at a high rate when NC were pre-incubated with native mouse serum resulting in complement opsonization. By focusing on dendritic cells (DC) as an important antigen-presenting cell type, we show that CR3 was essential for binding/uptake of complement-opsonized NC, whereas CR4, which in mouse is specifically expressed by DC, played no role. Further, a minor B cell subpopulation (B-1), which is important for first-line pathogen responses, and co-expressed CR1/2 and CR3, in general, engaged NC to a much higher extent than normal B cells. Here, we identified CR-1/2 as necessary for binding of complement-opsonized NC, whereas CR3 was dispensable. Interestingly, the binding of complement-opsonized NC to both DC and B-1 cells affected the expression of activation markers. Our findings may have important implications for the design of nano-vaccines against infectious diseases, which codeliver pathogen-specific protein antigen and adjuvant, aimed to induce a broad adaptive cellular and humoral immune response by inducing cytotoxic T lymphocytes that kill infected cells and pathogen-neutralizing antibodies, respectively. Decoration of nano-vaccines either with carbohydrates to trigger complement activation in vivo or with active complement may result in concomitant targeting of DC and B cells and thereby may strongly enhance the extent of dual cellular/humoral immune responses.
Collapse
Affiliation(s)
- Monika Bednarczyk
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Frederic Julien Fittler
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Meike Roskamp
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Michael Kuske
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Christian Langer
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Marco Vahldieck
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Evelyn Montermann
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Ingrid Tubbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Nadine Röhrig
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Andrzej Dzionek
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| |
Collapse
|
41
|
Zhou H, Qin F, Chen C. Designing Hypoxia-Responsive Nanotheranostic Agents for Tumor Imaging and Therapy. Adv Healthc Mater 2021; 10:e2001277. [PMID: 32985141 DOI: 10.1002/adhm.202001277] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/06/2020] [Indexed: 12/15/2022]
Abstract
Hypoxia, a common feature of most solid tumors, plays an important role in tumor proliferation, metastasis, and invasion, leading to drug, radiation, and photodynamic therapy resistance, and resulting in a sharp reduction in the disease-free survival rate of tumor patients. The lack of sufficient blood supply to the interior regions of tumors hinders the delivery of traditional drugs and contrast agents, interfering with their accumulation in the hypoxic region, and preventing efficient theranostics. Thus, there is a need for the fabrication of novel tumor theranostic agents that overcome these obstacles. Reports, in recent years, of hypoxia-responsive nanomaterials may provide with such means. In this review, a comprehensive description of the physicochemical and biological characteristics of hypoxic tumor tissues is provided, the principles of designing the hypoxia-responsive tumor theranostic agents are discussed, and the recent research into hypoxia-triggered nanomaterials is examined. Additionally, other hypoxia-associated responsive strategies, the current limitations, and future prospects for hypoxia-responsive nanotheranostic agents in tumor treatment are discussed.
Collapse
Affiliation(s)
- Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
- College of Materials Sciences and Opto‐Electronic Technology University of Chinese Academy of Sciences Beijing 100049 China
- Research Unit of Nanoscience and Technology Chinese Academy of Medical Sciences Beijing 100190 China
| | - Fenglan Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
- College of Materials Sciences and Opto‐Electronic Technology University of Chinese Academy of Sciences Beijing 100049 China
- Research Unit of Nanoscience and Technology Chinese Academy of Medical Sciences Beijing 100190 China
| |
Collapse
|
42
|
Klontzas ME, Kakkos GA, Papadakis GZ, Marias K, Karantanas AH. Advanced clinical imaging for the evaluation of stem cell based therapies. Expert Opin Biol Ther 2021; 21:1253-1264. [PMID: 33576278 DOI: 10.1080/14712598.2021.1890711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: As stem cell treatments reach closer to the clinic, the need for appropriate noninvasive imaging for accurate disease diagnosis, treatment planning, follow-up, and early detection of complications, is constantly rising. Clinical radiology affords an extensive arsenal of advanced imaging techniques, to provide anatomical and functional information on the whole spectrum of stem cell treatments from diagnosis to follow-up.Areas covered: This manuscript aims at providing a critical review of major published studies on the utilization of advanced imaging for stem cell treatments. Uses of magnetic resonance imaging (MRI), computed tomography (CT), ultrasound, and positron emission tomography (PET) are reviewed and interrogated for their applicability to stem cell imaging.Expert opinion: A wide spectrum of imaging methods have been utilized for the evaluation of stem cell therapies. The majority of published techniques are not clinically applicable, using methods exclusively applicable to animals or technology irrelevant to current clinical practice. Harmonization of preclinical methods with clinical reality is necessary for the timely translation of stem cell therapies to the clinic. Methods such as diffusion weighted MRI, hybrid imaging, and contrast-enhanced ultrasound hold great promise and should be routinely incorporated in the evaluation of patients receiving stem cell treatments.
Collapse
Affiliation(s)
- Michail E Klontzas
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece.,Advanced Hybrid Imaging Systems, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece
| | - George A Kakkos
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece
| | - Georgios Z Papadakis
- Advanced Hybrid Imaging Systems, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece.,Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), Heraklion, Crete, Greece.,Department of Radiology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Kostas Marias
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), Heraklion, Crete, Greece.,Department of Electrical and Computer Engineering, Hellenic Mediterranean University, Heraklion, Crete, Greece
| | - Apostolos H Karantanas
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece.,Advanced Hybrid Imaging Systems, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece.,Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), Heraklion, Crete, Greece.,Department of Radiology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| |
Collapse
|
43
|
Jones MA, MacCuaig WM, Frickenstein AN, Camalan S, Gurcan MN, Holter-Chakrabarty J, Morris KT, McNally MW, Booth KK, Carter S, Grizzle WE, McNally LR. Molecular Imaging of Inflammatory Disease. Biomedicines 2021; 9:152. [PMID: 33557374 PMCID: PMC7914540 DOI: 10.3390/biomedicines9020152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 01/31/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammatory diseases include a wide variety of highly prevalent conditions with high mortality rates in severe cases ranging from cardiovascular disease, to rheumatoid arthritis, to chronic obstructive pulmonary disease, to graft vs. host disease, to a number of gastrointestinal disorders. Many diseases that are not considered inflammatory per se are associated with varying levels of inflammation. Imaging of the immune system and inflammatory response is of interest as it can give insight into disease progression and severity. Clinical imaging technologies such as computed tomography (CT) and magnetic resonance imaging (MRI) are traditionally limited to the visualization of anatomical information; then, the presence or absence of an inflammatory state must be inferred from the structural abnormalities. Improvement in available contrast agents has made it possible to obtain functional information as well as anatomical. In vivo imaging of inflammation ultimately facilitates an improved accuracy of diagnostics and monitoring of patients to allow for better patient care. Highly specific molecular imaging of inflammatory biomarkers allows for earlier diagnosis to prevent irreversible damage. Advancements in imaging instruments, targeted tracers, and contrast agents represent a rapidly growing area of preclinical research with the hopes of quick translation to the clinic.
Collapse
Affiliation(s)
- Meredith A. Jones
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA; (M.A.J.); (W.M.M.); (A.N.F.)
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
| | - William M. MacCuaig
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA; (M.A.J.); (W.M.M.); (A.N.F.)
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
| | - Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA; (M.A.J.); (W.M.M.); (A.N.F.)
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
| | - Seda Camalan
- Department of Internal Medicine, Wake Forest Baptist Health, Winston-Salem, NC 27157, USA; (S.C.); (M.N.G.)
| | - Metin N. Gurcan
- Department of Internal Medicine, Wake Forest Baptist Health, Winston-Salem, NC 27157, USA; (S.C.); (M.N.G.)
| | - Jennifer Holter-Chakrabarty
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Medicine, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Katherine T. Morris
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Molly W. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
| | - Kristina K. Booth
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Steven Carter
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Lacey R. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA
| |
Collapse
|
44
|
Siafaka PI, Okur NÜ, Karantas ID, Okur ME, Gündoğdu EA. Current update on nanoplatforms as therapeutic and diagnostic tools: A review for the materials used as nanotheranostics and imaging modalities. Asian J Pharm Sci 2021; 16:24-46. [PMID: 33613728 PMCID: PMC7878458 DOI: 10.1016/j.ajps.2020.03.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/21/2020] [Accepted: 03/10/2020] [Indexed: 12/13/2022] Open
Abstract
In the last decade, the use of nanotheranostics as emerging diagnostic and therapeutic tools for various diseases, especially cancer, is held great attention. Up to date, several approaches have been employed in order to develop smart nanotheranostics, which combine bioactive targeting on specific tissues as well as diagnostic properties. The nanotheranostics can deliver therapeutic agents by concomitantly monitor the therapy response in real-time. Consequently, the possibility of over- or under-dosing is decreased. Various non-invasive imaging techniques have been used to quantitatively monitor the drug delivery processes. Radiolabeling of nanomaterials is widely used as powerful diagnostic approach on nuclear medicine imaging. In fact, various radiolabeled nanomaterials have been designed and developed for imaging tumors and other lesions due to their efficient characteristics. Inorganic nanoparticles as gold, silver, silica based nanomaterials or organic nanoparticles as polymers, carbon based nanomaterials, liposomes have been reported as multifunctional nanotheranostics. In this review, the imaging modalities according to their use in various diseases are summarized, providing special details for radiolabeling. In further, the most current nanotheranostics categorized via the used nanomaterials are also summed up. To conclude, this review can be beneficial for medical and pharmaceutical society as well as material scientists who work in the field of nanotheranostics since they can use this research as guide for producing newer and more efficient nanotheranostics.
Collapse
Affiliation(s)
- Panoraia I. Siafaka
- Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Neslihan Üstündağ Okur
- Faculty of Pharmacy, Department of Pharmaceutical Technology, University of Health Sciences, Istanbul, Turkey
| | - Ioannis D. Karantas
- 2nd Clinic of Internal Medicine, Hippokration General Hospital, Thessaloniki, Greece
| | - Mehmet Evren Okur
- Faculty of Pharmacy, Department of Pharmacology, University of Health Sciences, Istanbul, Turkey
| | | |
Collapse
|
45
|
Chowdhury S, Ghosh S. Nanoparticles and Stem Cells. Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
46
|
Sillerud LO, Yang Y, Yang LY, Duval KB, Thompson J, Yang Y. Longitudinal monitoring of microglial/macrophage activation in ischemic rat brain using Iba-1-specific nanoparticle-enhanced magnetic resonance imaging. J Cereb Blood Flow Metab 2020; 40:S117-S133. [PMID: 32960690 PMCID: PMC7687035 DOI: 10.1177/0271678x20953913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Microglial/macrophage activation plays a dual role in response to brain injury after a stroke, promoting early neuroinflammation and benefit for neurovascular recovery. Therefore, the dynamics of stroke-induced cerebral microglial/macrophage activation are of substantial interest. This study used novel anti-Iba-1-targeted superparamagnetic iron-platinum (FePt) nanoparticles in conjunction with magnetic resonance imaging (MRI) to measure the spatiotemporal changes of the microglial/macrophage activation in living rat brain for four weeks post-stroke. Ischemic lesion areas were identified and measured using T2-weighted MR images. After injection of the FePt-nanoparticles, T2*-weighted MR images showed that the nanoparticles were seen solely in brain regions that coincided with areas of active microglia/macrophages detected by post-mortem immunohistochemistry. Good agreement in morphological and distributive dynamic changes was also observed between the Fe+-cells and the Iba-1+-microglia/macrophages. The spatiotemporal changes of nanoparticle detected by T2*-weighted images paralleled the changes of microglial/macrophage activation and phenotypes measured by post-mortem immunohistochemistry over the four weeks post-stroke. Maximum microglial/macrophage activation occurred seven days post-stroke for both measures, and the diminished activation found after two weeks continued to four weeks. Our results suggest that nanoparticle-enhanced MRI may constitute a novel approach for monitoring the dynamic development of neuroinflammation in living animals during the progression and treatment of stroke.
Collapse
Affiliation(s)
- Laurel O Sillerud
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- BRaIN Imaging Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yirong Yang
- BRaIN Imaging Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Lisa Y Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Kelsey B Duval
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Jeffrey Thompson
- Center for Memory and Aging, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Yi Yang, Department of Neurology, University of New Mexico, MSC11 6035, Albuquerque, NM 87131-0001, USA.
| |
Collapse
|
47
|
Modak M, Frey MA, Yi S, Liu Y, Scott EA. Employment of targeted nanoparticles for imaging of cellular processes in cardiovascular disease. Curr Opin Biotechnol 2020; 66:59-68. [PMID: 32682272 PMCID: PMC7744313 DOI: 10.1016/j.copbio.2020.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/13/2020] [Accepted: 06/07/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD) is a leading cause of global mortality, accounting for pathologies that are primarily of atherosclerotic origin and driven by specific cell populations. A need exists for effective, non-invasive methods to assess the risk of potentially fatal major adverse cardiovascular events (MACE) before occurrence and to monitor post-interventional outcomes such as tissue regeneration. Molecular imaging has widespread applications in CVD diagnostic assessment, through modalities including magnetic resonance imaging (MRI), positron emission tomography (PET), and acoustic imaging methods. However, current gold-standard small molecule contrast agents are not cell-specific, relying on non-specific uptake to facilitate imaging of biologic processes. Nanomaterials can be engineered for targeted delivery to specific cell populations, and several nanomaterial systems have been developed for pre-clinical molecular imaging. Here, we review recent advances in nanoparticle-mediated approaches for imaging of cellular processes in cardiovascular disease, focusing on efforts to detect inflammation, assess lipid accumulation, and monitor tissue regeneration.
Collapse
Affiliation(s)
- Mallika Modak
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Molly A Frey
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Sijia Yi
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Yugang Liu
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; Department of Microbiology-Immunology, Northwestern University, Chicago, IL 60611, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
48
|
Yue H, Park JA, Ho SL, Ahmad MY, Cha H, Liu S, Tegafaw T, Marasini S, Ghazanfari A, Kim S, Chae KS, Chang Y, Lee GH. New Class of Efficient T 2 Magnetic Resonance Imaging Contrast Agent: Carbon-Coated Paramagnetic Dysprosium Oxide Nanoparticles. Pharmaceuticals (Basel) 2020; 13:ph13100312. [PMID: 33076332 PMCID: PMC7602642 DOI: 10.3390/ph13100312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 11/16/2022] Open
Abstract
Nanoparticles are considered potential candidates for a new class of magnetic resonance imaging (MRI) contrast agents. Negative MRI contrast agents require high magnetic moments. However, if nanoparticles can exclusively induce transverse water proton spin relaxation with negligible induction of longitudinal water proton spin relaxation, they may provide negative contrast MR images despite having low magnetic moments, thus acting as an efficient T2 MRI contrast agent. In this study, carbon-coated paramagnetic dysprosium oxide (DYO@C) nanoparticles (core = DYO = DyxOy; shell = carbon) were synthesized to explore their potential as an efficient T2 MRI contrast agent at 3.0 T MR field. Since the core DYO nanoparticles have an appreciable (but not high) magnetic moment that arises from fast 4f-electrons of Dy(III) (6H15/2), the DYO@C nanoparticles exhibited an appreciable transverse water proton spin relaxivity (r2) with a negligible longitudinal water proton spin relaxivity (r1). Consequently, they acted as a very efficient T2 MRI contrast agent, as proven from negative contrast enhancements seen in the in vivo T2 MR images.
Collapse
Affiliation(s)
- Huan Yue
- Department of Chemistry, Department of Nanoscience and Nanotechnology (DNN), College of Natural Sciences, Kyungpook National University (KNU), Taegu 41566, Korea; (H.Y.); (S.L.H.); (M.Y.A.); (S.L.); (T.T.); (S.M.); (A.G.)
| | - Ji Ae Park
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul 01817, Korea;
| | - Son Long Ho
- Department of Chemistry, Department of Nanoscience and Nanotechnology (DNN), College of Natural Sciences, Kyungpook National University (KNU), Taegu 41566, Korea; (H.Y.); (S.L.H.); (M.Y.A.); (S.L.); (T.T.); (S.M.); (A.G.)
| | - Mohammad Yaseen Ahmad
- Department of Chemistry, Department of Nanoscience and Nanotechnology (DNN), College of Natural Sciences, Kyungpook National University (KNU), Taegu 41566, Korea; (H.Y.); (S.L.H.); (M.Y.A.); (S.L.); (T.T.); (S.M.); (A.G.)
| | - Hyunsil Cha
- Department of Molecular Medicine and Medical & Biological Engineering, DNN, School of Medicine, KNU and Hospital, Taegu 41566, Korea; (H.C.); (S.K.)
| | - Shuwen Liu
- Department of Chemistry, Department of Nanoscience and Nanotechnology (DNN), College of Natural Sciences, Kyungpook National University (KNU), Taegu 41566, Korea; (H.Y.); (S.L.H.); (M.Y.A.); (S.L.); (T.T.); (S.M.); (A.G.)
| | - Tirusew Tegafaw
- Department of Chemistry, Department of Nanoscience and Nanotechnology (DNN), College of Natural Sciences, Kyungpook National University (KNU), Taegu 41566, Korea; (H.Y.); (S.L.H.); (M.Y.A.); (S.L.); (T.T.); (S.M.); (A.G.)
| | - Shanti Marasini
- Department of Chemistry, Department of Nanoscience and Nanotechnology (DNN), College of Natural Sciences, Kyungpook National University (KNU), Taegu 41566, Korea; (H.Y.); (S.L.H.); (M.Y.A.); (S.L.); (T.T.); (S.M.); (A.G.)
| | - Adibehalsadat Ghazanfari
- Department of Chemistry, Department of Nanoscience and Nanotechnology (DNN), College of Natural Sciences, Kyungpook National University (KNU), Taegu 41566, Korea; (H.Y.); (S.L.H.); (M.Y.A.); (S.L.); (T.T.); (S.M.); (A.G.)
| | - Soyeon Kim
- Department of Molecular Medicine and Medical & Biological Engineering, DNN, School of Medicine, KNU and Hospital, Taegu 41566, Korea; (H.C.); (S.K.)
| | - Kwon Seok Chae
- Department of Biology Education, DNN, Teachers’ College, KNU, Taegu 41566, Korea;
| | - Yongmin Chang
- Department of Molecular Medicine and Medical & Biological Engineering, DNN, School of Medicine, KNU and Hospital, Taegu 41566, Korea; (H.C.); (S.K.)
- Correspondence: (Y.C.); (G.H.L.)
| | - Gang Ho Lee
- Department of Chemistry, Department of Nanoscience and Nanotechnology (DNN), College of Natural Sciences, Kyungpook National University (KNU), Taegu 41566, Korea; (H.Y.); (S.L.H.); (M.Y.A.); (S.L.); (T.T.); (S.M.); (A.G.)
- Correspondence: (Y.C.); (G.H.L.)
| |
Collapse
|
49
|
Zeng Y, Li Z, Zhu H, Gu Z, Zhang H, Luo K. Recent Advances in Nanomedicines for Multiple Sclerosis Therapy. ACS APPLIED BIO MATERIALS 2020; 3:6571-6597. [PMID: 35019387 DOI: 10.1021/acsabm.0c00953] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yujun Zeng
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqian Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyan Zhu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, California 91711, United States
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
50
|
Affiliation(s)
- Xinping Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University Nanjing China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University Nanjing China
| | - Yuxin Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University Nanjing China
| | - Fu‐Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University Nanjing China
| |
Collapse
|