1
|
Jacinto C, Javed Y, Lavorato G, Tarraga WA, Conde BIC, Orozco JM, Picco AS, Garcia J, Dias CSB, Malik S, Sharma SK. Biotransformation and biological fate of magnetic iron oxide nanoparticles for biomedical research and clinical applications. NANOSCALE ADVANCES 2025:d5na00195a. [PMID: 40255989 PMCID: PMC12004083 DOI: 10.1039/d5na00195a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/15/2025] [Indexed: 04/22/2025]
Abstract
Safe implementation of nanotechnology-based products in biomedical applications necessitates an extensive understanding of the (bio)transformations that nanoparticles undergo in living organisms. The long-term fate in the body is a crucial consideration because it governs potential risks for human health. To accurately predict the life cycle of nanoparticles, their fate after administration into the body-including their (bio)transformations, persistence, and biodegradation-needs to be thoroughly evaluated. Magnetic iron oxide nanoparticles (MIONPs) can enter the body through various routes, including inhalation, ingestion, dermal absorption, and injection. Microscale and nanoscale studies are performed to observe nanomaterial biotransformations and their effect on clinically relevant properties. Researchers are utilizing high-resolution TEM for nanoscale monitoring of the nanoparticles while microscale follow-up approaches comprise quantification tools at the whole organism level and the molecular level. Nanoparticle-cell interactions, including cellular uptake and intracellular trafficking, are key to understanding nanoparticle accumulation in cells and organs. Prolonged accumulation may induce cell stress and nanoparticle toxicity, often mediated through oxidative stress and inflammation. In this review article, the journey of nanoparticles in the body is depicted and their biotransformations and final fate are discussed. Immunohistochemical techniques are particularly valuable in tracking nanoparticle distribution within tissues and assessing their impact at the cellular level. A thorough description of a wide range of characterization techniques is provided to unveil the fate and biotransformations of clinically relevant nanoparticles and to assist in their design for successful biomedical applications.
Collapse
Affiliation(s)
- Carlos Jacinto
- Nano-Photonics and Imaging Group, Institute of Physics, Universidade Federal de Alagoas 57072-900 Maceió AL Brazil
| | - Yasir Javed
- Department of Physics, University of Agriculture Faisalabad Pakistan
| | - Gabriel Lavorato
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | - Wilson A Tarraga
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | | | - Juan Manuel Orozco
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | - Agustin S Picco
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | - Joel Garcia
- Department of Chemistry, De La Salle University Manila Philippines
| | - Carlos Sato Baraldi Dias
- Institute for Photon Science and Synchrotron Radiation (IPS), Karlsruhe Institute of Technology (KIT) Hermann-von-Helmholtz-Platz 1 Eggenstein-Leopoldshafen 76344 Germany
| | - Sonia Malik
- Physiology, Ecology & Environmental Laboratory (P2e), University of Orléans 45067 France
- Department of Biotechnology, Baba Farid College Bathinda 151001 India
| | - Surender Kumar Sharma
- Department of Physics, Central University of Punjab Bathinda 151401 India
- Department of Physics, Federal University of Maranhão São Luís 65080-805 Brazil
| |
Collapse
|
2
|
Calsolaro F, Garello F, Cavallari E, Magnacca G, Trukhan MV, Valsania MC, Cravotto G, Terreno E, Martina K. Amphoteric β-cyclodextrin coated iron oxide magnetic nanoparticles: new insights into synthesis and application in MRI. NANOSCALE ADVANCES 2024; 7:155-168. [PMID: 39569331 PMCID: PMC11575534 DOI: 10.1039/d4na00692e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024]
Abstract
This work presents a group of high-quality hydrophilic and negatively charged coated, iron oxide magnetic nanoparticles (MNPs) that have been prepared using a microwave-ultrasound-assisted protocol, and demonstrates the great impact that the synthetic strategy has on the resulting MNPs. The different coatings tested, including citric acid, carboxymethyl dextran and β-cyclodextrin (βCD)/citric acid have been compared and have shown good dispersibility and stability. The ability of βCD to maintain the inclusive properties of the coated MNPs has been proven as well as their cytocompatibility. An amino citrate-modified βCD is proposed and its capabilities as a flexible amphoteric adsorbing device have been studied. The NMR relaxometric properties of the coated MNPs have been investigated using field-cycling nuclear magnetic relaxation dispersion profiles. For the amino citrate-modified βCD system, the order of magnitude of the Néel relaxation time is in the typical range for superparamagnetic systems' reversal times, i.e., 10-10-10-7 s. The r d value corresponds to the physical radius of the magnetic core, suggesting that, in this particular case, the coating does not prevent the diffusive motion of water molecules, which provide the basis for potential future magnetic resonance imaging (MRI) applications.
Collapse
Affiliation(s)
- Federica Calsolaro
- Department of Drug Science and Technology, University of Turin Via P. Giuria 9 10125 Turin Italy
| | - Francesca Garello
- Department of Molecular Biotechnology and Health Sciences, University of Turin Piazza Nizza 44/bis 10126 Turin Italy
| | - Eleonora Cavallari
- Department of Molecular Biotechnology and Health Sciences, University of Turin Piazza Nizza 44/bis 10126 Turin Italy
| | - Giuliana Magnacca
- Department of Chemistry and NIS Interdepartmental Centre, University of Turin Via Pietro Giuria 7 10125 Turin Italy
| | - Mikhail V Trukhan
- Department of Drug Science and Technology, University of Turin Via P. Giuria 9 10125 Turin Italy
| | - Maria Carmen Valsania
- Department of Chemistry and NIS Interdepartmental Centre, University of Turin Via Pietro Giuria 7 10125 Turin Italy
| | - Giancarlo Cravotto
- Department of Drug Science and Technology, University of Turin Via P. Giuria 9 10125 Turin Italy
| | - Enzo Terreno
- Department of Molecular Biotechnology and Health Sciences, University of Turin Piazza Nizza 44/bis 10126 Turin Italy
| | - Katia Martina
- Department of Drug Science and Technology, University of Turin Via P. Giuria 9 10125 Turin Italy
| |
Collapse
|
3
|
Doi N, Yamauchi Y, Sasai Y, Suzuki K, Kuzuya M, Kondo SI. Dextran-based nanoparticles with 5-FU-conjugated polymethacrylate segments for drug delivery: Synthesis of amphiphilic graft copolymers by mechanochemical solid-state polymerization and characterization. Int J Biol Macromol 2024; 274:132950. [PMID: 38848849 DOI: 10.1016/j.ijbiomac.2024.132950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Dextran (Dx) is a biodegradable and biocompatible polysaccharide, thus promising as a drug delivery carrier for tumor therapy. Herein, we applied mechanical energy to a high molecular weight Dx to control its molecular weight and simultaneously generate mechanoradicals. The solid-state polymerization of methacrylate- or methacrylamide derivatives initiated with Dx mechanoradicals showed polymer conversion of >95%, yielding Dx-based graft copolymers with molecular weights of approximately 30,000 g mol-1. The Dx-based graft copolymers with hydrophobic segments formed nanoparticles with a particle size of 25-35 nm in an aqueous solution. The anti-pancreatic tumor drug 5-fluorouracil (5-FU) was covalently conjugated onto the hydrophobic segments of the amphiphilic Dx, and the nanoparticles were also prepared. The drug release profile from 5-FU-conjugated nanoparticles corresponded well to the Korsmeyer-Peppas model applied to drug release from matrix substrates, and was also immensely predicted by the Logistic and Gompertz curves. The 5-FU-conjugated nanoparticles showed cytotoxicity against the pancreatic adenocarcinoma cell lines (BxPC-3) that were not significantly inferior to the 5-FU positive group. Furthermore, the fluorescein-labeled nanoparticles internalized into BxPC-3 within 6 h and actively migrated into the cytosol. These results suggest that Dx-based graft copolymers with hydrophobic segments might be used to enhance therapeutic activity.
Collapse
Affiliation(s)
- Naoki Doi
- Laboratory of Pharmaceutical Physical Chemistry, Gifu Pharmaceutical University, 501-1196, 1-25-4, Daigaku-nishi, Gifu, Japan
| | - Yukinori Yamauchi
- Department of Pharmaceutical Physical Chemistry, College of Pharmaceutical Sciences, Matsuyama University, 790-8578, 4-2 Bunkyo-cho, Matsuyama, Ehime, Japan
| | - Yasushi Sasai
- Faculty of Pharmacy, Gifu University of Medical Science, 509-0293, 4-3-3 Nijigaoka, Kani, Gifu, Japan
| | - Kaho Suzuki
- Laboratory of Pharmaceutical Physical Chemistry, Gifu Pharmaceutical University, 501-1196, 1-25-4, Daigaku-nishi, Gifu, Japan
| | - Masayuki Kuzuya
- Laboratory of Pharmaceutical Physical Chemistry, Gifu Pharmaceutical University, 501-1196, 1-25-4, Daigaku-nishi, Gifu, Japan
| | - Shin-Ichi Kondo
- Laboratory of Pharmaceutical Physical Chemistry, Gifu Pharmaceutical University, 501-1196, 1-25-4, Daigaku-nishi, Gifu, Japan.
| |
Collapse
|
4
|
Petcov TE, Straticiuc M, Iancu D, Mirea DA, Trușcă R, Mereuță PE, Savu DI, Mogoșanu GD, Mogoantă L, Popescu RC, Kopatz V, Jinga SI. Unveiling Nanoparticles: Recent Approaches in Studying the Internalization Pattern of Iron Oxide Nanoparticles in Mono- and Multicellular Biological Structures. J Funct Biomater 2024; 15:169. [PMID: 38921542 PMCID: PMC11204647 DOI: 10.3390/jfb15060169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/15/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Nanoparticle (NP)-based solutions for oncotherapy promise an improved efficiency of the anticancer response, as well as higher comfort for the patient. The current advancements in cancer treatment based on nanotechnology exploit the ability of these systems to pass biological barriers to target the tumor cell, as well as tumor cell organelles. In particular, iron oxide NPs are being clinically employed in oncological management due to this ability. When designing an efficient anti-cancer therapy based on NPs, it is important to know and to modulate the phenomena which take place during the interaction of the NPs with the tumor cells, as well as the normal tissues. In this regard, our review is focused on highlighting different approaches to studying the internalization patterns of iron oxide NPs in simple and complex 2D and 3D in vitro cell models, as well as in living tissues, in order to investigate the functionality of an NP-based treatment.
Collapse
Affiliation(s)
- Teodora Eliana Petcov
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, 1–7 Gheorghe Polizu Street, 011061 Bucharest, Romania; (T.E.P.); (S.I.J.)
| | - Mihai Straticiuc
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Decebal Iancu
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Dragoș Alexandru Mirea
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Roxana Trușcă
- National Research Center for Micro and Nanomaterials, National University for Science and Technology Politehnica of Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania;
| | - Paul Emil Mereuță
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Diana Iulia Savu
- Department of Life and Environmental Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania
| | - George Dan Mogoșanu
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareș Street, 200349 Craiova, Romania;
| | - Laurențiu Mogoantă
- Research Center for Microscopic Morphology and Immunology, University of Medicine and Pharmacy of Craiova, 2 Petru Rareș Street, 200349 Craiova, Romania;
| | - Roxana Cristina Popescu
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, 1–7 Gheorghe Polizu Street, 011061 Bucharest, Romania; (T.E.P.); (S.I.J.)
- Department of Life and Environmental Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania
| | - Verena Kopatz
- Department of Radiation Oncology, Medical University of Vienna, 18–20 Waehringer Guertel Street, 1090 Vienna, Austria;
| | - Sorin Ion Jinga
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, 1–7 Gheorghe Polizu Street, 011061 Bucharest, Romania; (T.E.P.); (S.I.J.)
| |
Collapse
|
5
|
Natarajan P, Horak K, Rowe J, Yoon S, Lingo J, Tomich JM, Fleming SD. Biodistribution Analysis of Peptide-Coated Magnetic Iron Nanoparticles: A Simple and Quantitative Method. Mol Pharm 2024; 21:970-981. [PMID: 38206824 PMCID: PMC10918533 DOI: 10.1021/acs.molpharmaceut.3c01080] [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: 01/13/2024]
Abstract
Biodistribution tracks compounds or molecules of interest in vivo to understand a compound's anticipated efficacy and safety. Nanoparticles deliver nucleic acid and drug payloads and enhance tumor permeability due to multiple properties such as high surface area to volume ratio, surface functionalization, and modifications. Studying the in vivo biodistribution of nanoparticles documents the effectiveness and safety of nanoparticles and facilitates a more application-driven approach for nanoparticle development that allows for more successful translation into clinical use. In this study, we present a relatively simple method to determine the biodistribution of magnetic iron nanoparticles in mice. In vitro, cells take up branched amphiphilic peptide-coated magnetic nanobeads (BAPc-MNBs) like their counterparts, i.e., branched amphiphilic peptide capsules (BAPCs) with a hollow water-filled core. Both BAPc-MNBs and BAPCs have widespread applications as a nanodelivery system. We evaluated the BAPc-MNBs tissue distribution in wild-type mice injected intravenously (i.v.), intraperitoneally (i.p.), or orally gavaged to understand the biological interactions and to further the development of branched amphiphilic peptide-based nanoparticles. The magnetic nanoparticles allowed collection of the BAPc-MNBs from multiple organs by magnetic bead sorting, followed by a high-throughput screening for iron content. When injected i.v., nanoparticles were distributed widely to various organs before elimination from the system via the intestines in feces. The spleen accumulated the highest amount of BAPc-MNBs in mice administered NPs via i.v. and i.p. but not via oral gavage. Taken together, these data demonstrate that the magnetic sorting not only allowed quantification of the BAPc-MNBs but also identified the distribution of BAPc-MNBs after distinct administration methods.
Collapse
Affiliation(s)
- Pavithra Natarajan
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA 66506
| | - Katherine Horak
- National Wildlife Research Center, USDA APHIS WS, Fort Collins, CO, USA 80521
| | - Jennifer Rowe
- Division of Biology, Kansas State University, Manhattan, KS, USA 66506
| | - Sungmin Yoon
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA 66506
| | - Joshua Lingo
- Division of Biology, Kansas State University, Manhattan, KS, USA 66506
| | - John M Tomich
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA 66506
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, USA 66506
| |
Collapse
|
6
|
Hannon G, Bogdanska A, Keogh A, Finn SP, Gobbo OL, Prina-Mello A. Biodistribution and histological analysis of iron oxide-dextran nanoparticles in wistar rats. Nanotoxicology 2023; 17:562-580. [PMID: 37982374 DOI: 10.1080/17435390.2023.2276413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/11/2023] [Indexed: 11/21/2023]
Abstract
Iron oxide nanoparticles (IONP) are showing promise in many biomedical applications. One of these- magnetic hyperthermia- utilizes externally applied alternating magnetic fields and tumor-residing magnetic nanoparticles to generate localized therapeutic temperature elevations. Magnetic hyperthermia is approved in Europe to treat glioblastoma and is undergoing clinical assessment in the United States to treat prostate cancer. In this study, we performed biodistribution and histological analysis of a new IONP (RCL-01) in Wistar rats. These nanoparticles are currently undergoing clinical assessment in locally advanced pancreatic ductal adenocarcinoma to determine the feasibility of magnetic hyperthermia treatment in this disease. The study presented here aimed to determine the fate of these nanoparticles in vivo and whether this results in organ damage. Wistar rats were injected intravenously with relatively high doses of IONP (30 mgFe/kg, 45 mgFe/kg and 60 mgFe/kg) and compared to a vehicle control to determine the accumulation of iron in organs and whether this resulted in histological changes in these tissues. Dose-dependent increases of iron were observed in the liver, spleen and lungs of IONP-treated animals at 7 days postinjection; however, this did not result in significant histological changes in these tissues. Immunofluorescent imaging determined these nanoparticles are internalized by macrophages in tissue, suggesting they are readily phagocytosed by the reticuloendothelial system for eventual recycling. Notably, no changes in iron or dextran staining were found in the kidneys across all treatment groups, providing evidence for potential renal clearance.
Collapse
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Anna Bogdanska
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Anna Keogh
- Department of Histopathology, Trinity College Dublin, Ireland
| | - Stephen P Finn
- Department of Histopathology, Trinity College Dublin, Ireland
| | - Oliviero L Gobbo
- School of Pharmacy and Pharmaceutical Sciences, Ireland
- Trinity St James's Cancer Institute, Ireland
| | - Adriele Prina-Mello
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, Trinity College Dublin, Ireland
| |
Collapse
|
7
|
Ajith S, Almomani F, Elhissi A, Husseini GA. Nanoparticle-based materials in anticancer drug delivery: Current and future prospects. Heliyon 2023; 9:e21227. [PMID: 37954330 PMCID: PMC10637937 DOI: 10.1016/j.heliyon.2023.e21227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 09/18/2023] [Accepted: 10/18/2023] [Indexed: 11/14/2023] Open
Abstract
The past decade has witnessed a breakthrough in novel strategies to treat cancer. One of the most common cancer treatment modalities is chemotherapy which involves administering anti-cancer drugs to the body. However, these drugs can lead to undesirable side effects on healthy cells. To overcome this challenge and improve cancer cell targeting, many novel nanocarriers have been developed to deliver drugs directly to the cancerous cells and minimize effects on the healthy tissues. The majority of the research studies conclude that using drugs encapsulated in nanocarriers is a much safer and more effective alternative than delivering the drug alone in its free form. This review provides a summary of the types of nanocarriers mainly studied for cancer drug delivery, namely: liposomes, polymeric micelles, dendrimers, magnetic nanoparticles, mesoporous nanoparticles, gold nanoparticles, carbon nanotubes and quantum dots. In this review, the synthesis, applications, advantages, disadvantages, and previous studies of these nanomaterials are discussed in detail. Furthermore, the future opportunities and possible challenges of translating these materials into clinical applications are also reported.
Collapse
Affiliation(s)
- Saniha Ajith
- Department of Chemical Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Fares Almomani
- Department of Chemical Engineering, College of Engineering, Qatar University, Doha, Qatar
| | | | - Ghaleb A. Husseini
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates
| |
Collapse
|
8
|
Kozlov D, Rodimova S, Kuznetsova D. The Role of MicroRNAs in Liver Functioning: from Biogenesis to Therapeutic Approaches (Review). Sovrem Tekhnologii Med 2023; 15:54-79. [PMID: 39967915 PMCID: PMC11832066 DOI: 10.17691/stm2023.15.5.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Indexed: 01/03/2025] Open
Abstract
Molecular diagnostics based on small non-coding RNA molecules (in particular microRNA) is a new direction in modern biomedicine and is considered a promising method for identification of a wide range of pathologies at an early stage, clinical phenotype assessment, as well as monitoring the course of the disease, evaluation of therapy efficacy and the risk of the disease recurrence. Currently, the role of microRNAs as the most important epigenetic regulator in cancer development has been proven within the studies of normal and pathogenic processes. However, currently, there are insignificant studies devoted to studying the role of microRNAs in functioning of other organs and tissues, as well as to development of possible therapeutic approaches based on microRNAs. A huge number of metabolic processes in the liver are controlled by microRNAs, which creates enormous potential for the use of microRNAs as a diagnostic marker and makes it a target for therapeutic intervention in metabolic, oncological, and even viral diseases of this organ. This review examines various aspects of biological functions of microRNAs in different types of liver cells. Both canonical and non-canonical pathways of biogenesis, epigenetic regulation mediated by microRNAs, as well as the microRNAs role in intercellular communication and the course of viral diseases are shown. The potential of microRNAs as a diagnostic marker for various liver pathologies is described, as well as therapeutic approaches and medicines based on microRNAs, which are approved for clinical use and currently being developed.
Collapse
Affiliation(s)
- D.S. Kozlov
- Laboratory Assistant, Scientific Laboratory of Molecular Biotechnologies, I Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Student, Institute of Biology and Biomedicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| | - S.A. Rodimova
- Junior Researcher, Laboratory of Regenerative Medicine; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Junior Researcher, Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - D.S. Kuznetsova
- PhD, Head of the Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Head of the Research Laboratory for Molecular Genetic Researches, Institute of Clinical Medicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| |
Collapse
|
9
|
Natarajan P, Horak K, Rowe J, Lingo J, Tomich JM, Fleming SD. Biodistribution Analysis of Peptide-Coated Magnetic Iron Nanoparticles: A Simple and Quantitative Method. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.11.561862. [PMID: 37873129 PMCID: PMC10592714 DOI: 10.1101/2023.10.11.561862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Biodistribution is the tracking of compounds or molecules of interest in the subject which is integral to understanding their anticipated efficacy and safety. Nanoparticles are highly desirable delivery systems which have the ability to deliver higher nucleic acid and drug payloads and they have enhanced tumor permeability due to their unique properties such as high surface area to volume ratio. Studying the biodistribution of nanoparticles is crucial to understand their effectiveness and safety in vivo, facilitate a more application driven approach for nanoparticle development which will lead to their successful translation into clinical use. In this study, we present a relatively simple method to determine the biodistribution of magnetic iron nanoparticles in mice. Branched Amphiphilic Peptide coated Magnetic Nanobeads BAPc-MNBs like their counterpart i.e., Branched Amphiphilic Peptide capsules (BAPCs) with a hollow water-filled core, are readily taken up by cells in vitro and have widespread application as a nanodelivery systems. We evaluated the BAPc-MNBs tissue distribution in wildtype mice injected intravenously (i.v.), intraperitoneally (i.p.) or orally gavaged to understand the biological interactions of the peptide nanoparticles and to further the development of branched amphiphilic peptides-based nanoparticles. BAPc-MNBs were distributed widely to various organs when injected i.v. and were eliminated from the system via the intestines in feces. The spleen was found to accumulate the highest amount of BAPc-MNBs in mice administered the NPs i.v. and i.p. while they were not absorbed into the system via oral gavage. This study not only presents a relatively simple quantification method to determine in vivo biodistribution of magnetic iron nanoparticles that can be widely applied but also demonstrates the potential of Branched Amphiphilic Peptides in the form of BAPCs or BAPc-MNBs as a delivery system.
Collapse
|
10
|
Zhang Y, Wu X, Zhu H, Cong Y. Development and in functional study of a bi-specific sustained release drug-loaded nano-liposomes for hepatocellular carcinoma. J Biomater Appl 2023:8853282231179313. [PMID: 37243614 DOI: 10.1177/08853282231179313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
BACKGROUND Lenvatinib (LEN) is a first-line therapy for patients with hepatocellular carcinoma (HCC), but has a larger adverse effect profile. In this study, we developed a liposome with drug-carrying function and magnetic resonance imaging (MRI) imaging function to investigate the targeted drug-carrying function and MRI tracing ability of liposome for HCC. METHODS Magnetic nano-liposomes (MNL) with dual targeting function of epithelial cell adhesion molecule (EpCAM) and vimentin and capable of encapsulating LEN drugs were prepared. The characterization performance, drug loading efficiency and cytotoxicity of EpCAM/vimentin-LEN-MNL were tested, and the dual-targeting slow release drug loading function and MRI tracing ability were investigated in cellular and animal models. RESULTS EpCAM/vimentin-LEN-MNL has a mean particle size of 218.37 ± 5.13 nm and a mean potential of 32.86 ± 4.62 mV, and is spherical in shape and can be uniformly dispersed in solution. The encapsulation rate was 92.66 ± 0.73% and the drug loading rate was 9.35 ± 0.16%. It has low cytotoxicity, can effectively inhibit HCC cell proliferation and promote HCC cell apoptosis, and has specific targeting function and MRI tracing ability for HCC cells. CONCLUSIONS In this study, an HCC-specific dual-targeted sustained-release drug delivery liposome with dual-targeted recognition and sensitive MRI tracer was successfully prepared, which provides an important scientific basis for maximizing the multiple effects of nano-carriers in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Yufei Zhang
- Shanghai Seventh People's Hospital, Shanghai, China
| | - Xiaoxiong Wu
- Shanghai Seventh People's Hospital, Shanghai, China
| | - Hongfan Zhu
- Shanghai Seventh People's Hospital, Shanghai, China
| | - Yun Cong
- Shanghai Seventh People's Hospital, Shanghai, China
| |
Collapse
|
11
|
Ciont C, Mesaroș A, Pop OL, Vodnar DC. Iron oxide nanoparticles carried by probiotics for iron absorption: a systematic review. J Nanobiotechnology 2023; 21:124. [PMID: 37038224 PMCID: PMC10088223 DOI: 10.1186/s12951-023-01880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/30/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND One-third of the world's population has anemia, contributing to higher morbidity and death and impaired neurological development. Conventional anemia treatment raises concerns about iron bioavailability and gastrointestinal (GI) adverse effects. This research aims to establish how iron oxide nanoparticles (IONPs) interact with probiotic cells and how they affect iron absorption, bioavailability, and microbiota variation. METHODS Pointing to the study of the literature and developing a review and critical synthesis, a robust search methodology was utilized by the authors. The literature search was performed in the PubMed, Scopus, and Web of Science databases. Information was collected between January 2017 and June 2022 using the PRISMA (Preferred Reporting Items for Systematic Review and Meta-Analysis) protocols for systematic reviews and meta-analyses. We identified 122 compatible research articles. RESULTS The research profile of the selected scientific articles revealed the efficacy of IONPs treatment carried by probiotics versus conventional treatment. Therefore, the authors employed content assessment on four topics to synthesize previous studies. The key subjects of the reviewed reports are the characteristics of the IONPs synthesis method, the evaluation of cell absorption and cytotoxicity of IONPs, and the transport of IONPs with probiotics in treating anemia. CONCLUSIONS To ensure a sufficient iron level in the enterocyte, probiotics with the capacity to attach to the gut wall transport IONPs into the enterocyte, where the maghemite nanoparticles are released.
Collapse
Affiliation(s)
- Călina Ciont
- Department of Food Science, University of Agricultural Sciences and Veterinary Medicine, 400372, Cluj-Napoca, Romania
- Molecular Nutrition and Proteomics Laboratory, Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, 400372, Cluj-Napoca, Romania
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3-5, 400372, Cluj-Napoca, Romania
| | - Amalia Mesaroș
- Physics and Chemistry Department, C4S Centre, Technical University of Cluj-Napoca, 28 Memorandumului Street, 400114, Cluj-Napoca, Romania
- Molecular Nutrition and Proteomics Laboratory, Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Oana Lelia Pop
- Department of Food Science, University of Agricultural Sciences and Veterinary Medicine, 400372, Cluj-Napoca, Romania.
- Molecular Nutrition and Proteomics Laboratory, Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, 400372, Cluj-Napoca, Romania.
| | - Dan Cristian Vodnar
- Department of Food Science, University of Agricultural Sciences and Veterinary Medicine, 400372, Cluj-Napoca, Romania.
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3-5, 400372, Cluj-Napoca, Romania.
| |
Collapse
|
12
|
Arul MR, Zhang C, Alahmadi I, Moss IL, Banasavadi-Siddegowda YK, Abdulmalik S, Illien-Junger S, Kumbar SG. Novel Injectable Fluorescent Polymeric Nanocarriers for Intervertebral Disc Application. J Funct Biomater 2023; 14:52. [PMID: 36826851 PMCID: PMC9961171 DOI: 10.3390/jfb14020052] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Damage to intervertebral discs (IVD) can lead to chronic pain and disability, and no current treatments can fully restore their function. Some non-surgical treatments have shown promise; however, these approaches are generally limited by burst release and poor localization of diverse molecules. In this proof-of-concept study, we developed a nanoparticle (NP) delivery system to efficiently deliver high- and low-solubility drug molecules. Nanoparticles of cellulose acetate and polycaprolactone-polyethylene glycol conjugated with 1-oxo-1H-pyrido [2,1-b][1,3]benzoxazole-3-carboxylic acid (PBC), a novel fluorescent dye, were prepared by the oil-in-water emulsion. Two drugs, a water insoluble indomethacin (IND) and a water soluble 4-aminopyridine (4-AP), were used to study their release patterns. Electron microscopy confirmed the spherical nature and rough surface of nanoparticles. The particle size analysis revealed a hydrodynamic radius ranging ~150-162 nm based on dynamic light scattering. Zeta potential increased with PBC conjugation implying their enhanced stability. IND encapsulation efficiency was almost 3-fold higher than 4-AP, with release lasting up to 4 days, signifying enhanced solubility, while the release of 4-AP continued for up to 7 days. Nanoparticles and their drug formulations did not show any apparent cytotoxicity and were taken up by human IVD nucleus pulposus cells. When injected into coccygeal mouse IVDs in vivo, the nanoparticles remained within the nucleus pulposus cells and the injection site of the nucleus pulposus and annulus fibrosus of the IVD. These fluorescent nano-formulations may serve as a platform technology to deliver therapeutic agents to IVDs and other tissues that require localized drug injections.
Collapse
Affiliation(s)
- Michael R. Arul
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
| | - Changli Zhang
- Department of Orthopedic Surgery, Emory University, Atlanta, GA 30308, USA
| | - Ibtihal Alahmadi
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Isaac L. Moss
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
| | | | - Sama Abdulmalik
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
| | | | - Sangamesh G. Kumbar
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
13
|
In Vitro and In Vivo Biological Assays of Dextran Coated Iron Oxide Aqueous Magnetic Fluids. Pharmaceutics 2023; 15:pharmaceutics15010177. [PMID: 36678806 PMCID: PMC9865434 DOI: 10.3390/pharmaceutics15010177] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
The iron oxide nanoparticles coated with different surface coatings were studied and characterized by multiple physicochemical and biological methods. The present paper aims at estimating the toxicity in vitro and in vivo of dextran coated iron oxide aqueous magnetic fluids. The in vitro studies were conducted by quantifying the viability of HeLa cells after their incubation with the samples (concentrations of 62.5−125−250−500 μg/mL at different time intervals). The estimation of the toxicity in vivo of administering dextran coated iron oxide aqueous magnetic fluids (DIO-AMF) with hydrodynamic diameter of 25.73 ± 4 nm to Male Brown Norway rats has been made. Different concentrations (62.5−125−250−500 μg/mL) of dextran coated iron oxide aqueous magnetic fluids were administered for 7 consecutive days. Hematology and biochemistry of the Male Brown Norway rats assessment was performed at various time intervals (24−72 h and 21−28 days) after intra-peritoneal injection. The results showed that high concentrations of DIO-AMF (250 and 500 μg/mL) significantly increased white blood cells, red blood cells, hemoglobin and hematocrit compared to the values obtained for the control group (p < 0.05). Moreover, following the administration of DIO-AMF, the levels of alkaline phosphatase and aspartate aminotransferase increased compared to the control group (p < 0.05). After DIO-AMF administration, no significant difference was observed in the levels of alanine aminotransferase, gamma-glutamyl transpeptidase, urea and creatinine compared to the control group (p < 0.05). The results of the present study showed that dextran coated iron oxide aqueous magnetic fluids in concentrations lower than 250 μg/mL are reliable for medical and pharmaceutical applications.
Collapse
|
14
|
Thai VP, Nguyen HD, Saito N, Takahashi K, Sasaki T, Kikuchi T. Precise size-control and functionalization of gold nanoparticles synthesized by plasma-liquid interactions: using carboxylic, amino, and thiol ligands. NANOSCALE ADVANCES 2022; 4:4490-4501. [PMID: 36341298 PMCID: PMC9595108 DOI: 10.1039/d2na00542e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 06/16/2023]
Abstract
Using gold nanoparticles (GNPs) in high-standard applications requires GNPs to be fabricated with high-quality size and surface properties. Plasma-liquid interactions (PLIs) have the unique ability to synthesize GNPs without using any reducing agents, and the GNP surface is free of stabilizing agents. It is an extreme advantage that ensures success for the subsequent functionalization processes for GNPs. However, fabricating GNPs via PLIs at the desired size has still been a challenge. Here, we present a simple approach to achieving the precise size-control of GNPs synthesized by PLIs. By adding suitable ligands to the precursor solution, the ligands wrap GNPs which interrupts and slows down the rapid growth of GNPs under PLIs. This way, the size of the GNPs can be precisely controlled by adjusting the ligand concentration. Our results showed that the size of the GNPs in the range of 10-60 nm can be fitted to reciprocal functions of the ligand concentration. The potency of the size-control depends on the type of ligands in the order of thiol > amine > carboxylate. The size-control has been well investigated with four common ligands: l-cysteine, glucosamine, salicylic acid, and terephthalic acid. XPS, FTIR, and zeta potential techniques confirmed the presence of these ligands on GNPs. The results indicated that functionalized ligands could be utilized to control the size and functionalize the GNP surface. Hence our approach could simultaneously achieve two goals: precise size-control and functionalization of GNPs without the ligand-exchange step.
Collapse
Affiliation(s)
- Van-Phuoc Thai
- Faculty of Mechanical Engineering, HCMC University of Technology and Education Ho Chi Minh City 71307 Vietnam
- Department of Electrical, Electronics and Information Engineering, Nagaoka University of Technology Nagaoka 940-2188 Japan
| | - Hieu Duy Nguyen
- Research Center for Advanced Measurement and Characterization, National Institute for Materials Science 1-1 Namiki Tsukuba Ibaraki 305-0044 Japan
| | - Nobuo Saito
- Department of Materials Science and Bioengineering, Nagaoka University of Technology Nagaoka 940-2188 Japan
| | - Kazumasa Takahashi
- Department of Electrical, Electronics and Information Engineering, Nagaoka University of Technology Nagaoka 940-2188 Japan
| | - Toru Sasaki
- Department of Electrical, Electronics and Information Engineering, Nagaoka University of Technology Nagaoka 940-2188 Japan
- Department of Science of Technology Innovation, Nagaoka University of Technology Nagaoka 940-2188 Japan
| | - Takashi Kikuchi
- Department of Electrical, Electronics and Information Engineering, Nagaoka University of Technology Nagaoka 940-2188 Japan
- Department of Nuclear Technology, Nagaoka University of Technology Nagaoka 940-2188 Japan
- Extreme Energy-Density Research Institute, Nagaoka University of Technology Nagaoka 940-2188 Japan
| |
Collapse
|
15
|
Applegate CC, Deng H, Kleszynski BL, Cross TWL, Konopka CJ, Dobrucki LW, Nelson ER, Wallig MA, Smith AM, Swanson KS. Impact of administration route on nanocarrier biodistribution in a murine colitis model. JOURNAL OF EXPERIMENTAL NANOSCIENCE 2022; 17:599-616. [PMID: 36968097 PMCID: PMC10038121 DOI: 10.1080/17458080.2022.2134563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 06/18/2023]
Abstract
The incidence of inflammatory bowel disease (IBD) is increasing worldwide. Although current diagnostic and disease monitoring tests for IBD sensitively detect gut inflammation, they lack the molecular and cellular specificity of positron emission tomography (PET). In this proof-of-concept study, we use a radiolabeled macrophage-targeted nanocarrier probe (64Cu-NOTA-D500) administered by oral, enema, and intraperitoneal routes to evaluate the delivery route dependence of biodistribution across healthy and diseased tissues in a murine model of dextran sodium sulfate (DSS)-induced colitis. High inter-subject variability of probe uptake in intestinal tissue was reduced by normalization to uptake in liver or total intestines. Differences in normalized uptake between healthy and DSS colitis animal intestines were highest for oral and IP routes. Differences in absolute liver uptake reflected a possible secondary diagnostic metric of IBD pathology. These results should inform the preclinical development of inflammation-targeted contrast agents for IBD and related gut disorders to improve diagnostic accuracy.
Collapse
Affiliation(s)
- Catherine C. Applegate
- Division of Nutritional Sciences, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
- Department of Animal Sciences, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
| | - Hongping Deng
- Department of Bioengineering, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
| | - Brittany L. Kleszynski
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
| | - Tzu-Wen L. Cross
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
| | | | - L. Wawrzyniec Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
| | - Erik R. Nelson
- Division of Nutritional Sciences, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
| | - Matthew A. Wallig
- Division of Nutritional Sciences, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
| | - Andrew M. Smith
- Department of Bioengineering, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
- Carle Illinois College of Medicine, Urbana, Illinois, USA
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
| | - Kelly S. Swanson
- Division of Nutritional Sciences, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
- Department of Animal Sciences, University of Illinois at Urbana – Champaign, Urbana, Illinois, USA
| |
Collapse
|
16
|
Huang Z, Li F, Zhang J, Shi X, Xu Y, Huang X. Research on the Construction of Bispecific-Targeted Sustained-Release Drug-Delivery Microspheres and Their Function in Treatment of Hepatocellular Carcinoma. ACS OMEGA 2022; 7:22003-22014. [PMID: 35785307 PMCID: PMC9244910 DOI: 10.1021/acsomega.2c02584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 06/01/2023]
Abstract
Lenvatinib (LEN) is approved as one of the commonly used drugs in the treatment of hepatocellular carcinoma (HCC). It is recognized to be a novel therapeutic choice for the direct and targeted delivery of effective drugs to HCC tumor sites. The key to the proposed method lies in the requirement for efficient targeted drug delivery carriers with targeting performance to deliver effective drugs directly and safely to tumor lesions. Methods: Here, magnetic liposomes (MLs) were modified by phosphatidylinositol proteoglycan 3 (GPC3) and epithelial cell adhesion molecules (EpCAMs). Subsequently, bispecific-targeted sustained-release drug-loaded microspheres containing LEN (GPC3/EpCAM-LEN-MLs) were constructed. In addition, both cytotoxicity and magnetic resonance imaging (MRI) analyses were performed to establish a mouse model and further perform corresponding performance assessments. Results: The corresponding results showed that GPC3/EpCAM-LEN-MLs were spherical-shaped and evenly dispersed. The encapsulation and drug-loading efficiencies were 91.08% ± 1.83% and 8.22% ± 1.24%, respectively. Meanwhile, GPC3/EpCAM-LEN-MLs showed a high inhibition rate on the proliferation of HCC cells and significantly increased their apoptosis. Furthermore, MRI revealed that the system possessed the function of tracking and localizing tumor cells, and animal experiments verified that it could exert the function of disease diagnosis. Conclusions: Our experiments successfully constructed a safe and efficient bispecific-targeted sustained-release drug delivery system for HCC tumor cells. It provides a useful diagnostic and therapeutic scheme for the clinical diagnosis and targeted therapy of HCC. Moreover, it can be used as a potential tumor-specific MRI contrast agent for the localization and diagnosis of malignant tumors.
Collapse
Affiliation(s)
- Zi−Li Huang
- Department
of General Surgery, Shanghai Jiaotong University
Affiliated Sixth People’s Hospital, No. 600, Yishan RD., Shanghai 200233, PR China
- Department
of Radiology, Xuhui District Central Hospital of Zhongshan Hospital, Fudan University, No. 966, Huaihai Middle RD., Shanghai 200031, PR China
| | - Feng Li
- School
of Materials of Science and Engineering, Shanghai Jiao Tong University, No. 800, Dongchuan RD., Shanghai 200240, PR China
| | - Jun−Tao Zhang
- Institute
of Microsurgery on Extremities, Shanghai
Jiao Tong University Affiliated Sixth People’s Hospital, No. 600, Yishan RD., Shanghai 200233, PR China
| | - Xiang−Jun Shi
- Department
of General Surgery, Shanghai Jiaotong University
Affiliated Sixth People’s Hospital, No. 600, Yishan RD., Shanghai 200233, PR China
| | - Yong−Hua Xu
- Department
of Radiology, Xuhui District Central Hospital of Zhongshan Hospital, Fudan University, No. 966, Huaihai Middle RD., Shanghai 200031, PR China
| | - Xiu−Yan Huang
- Department
of General Surgery, Shanghai Jiaotong University
Affiliated Sixth People’s Hospital, No. 600, Yishan RD., Shanghai 200233, PR China
| |
Collapse
|
17
|
Pasek-Allen JL, Wilharm RK, Gao Z, Pierre VC, Bischof JC. Phosphonate coating of commercial iron oxide nanoparticles for nanowarming cryopreserved samples. J Mater Chem B 2022; 10:3734-3746. [PMID: 35466332 PMCID: PMC9116443 DOI: 10.1039/d1tb02483c] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/13/2022] [Indexed: 01/02/2023]
Abstract
New preservation technologies may allow for organ banking similar to blood and biomaterial banking approaches. Using cryoprotective agents (CPAs), aqueous solutions with organic components such as DMSO, propylene glycol, and added salts and sugars, organs can be used to vitrify and store organs at -140 °C. When needed, these organs can be rewarmed in a rapid and uniform manner if CPAs are supplemented with iron oxide nanoparticles (IONPs) in an applied radiofrequency field. Speed and uniformity of warming are both IONP concentration and CPA suspension dependent. Here we present a coating method of small molecule phosphonate linker (PLink) and biocompatible polymer (i.e. polyethylene glycol PEG) that tunes stability and increases the maximum allowable concentration of IONPs in CPA suspension. PLink contains a phosphonate 'anchor' for high irreversible binding to iron oxide and a carboxylic acid 'handle' for ligand attachment. PLink-PEG removes and replaces the initial coating layer of commercially available IONPs (EMG1200 (hydrophobic) and EMG308 (hydrophilic) Ferrotec, Inc., increasing colloidal stability and decreasing aggregation in both water and CPAs, (verified with dynamic light scattering) from minutes (uncoated) to up to 6 days. Heating properties of EMG1200, specific absorption rate (SAR), measured using an applied field of 360 kHz and 20 kA m-1, increased from 20 to 180 W per g Fe with increasing PLink-PEG5000. PEG replacing the initially hydrophobic coating decreased aggregation in water and CPA, consistent with earlier studies on heating performance. Furthermore, although the size is minimized at 0.20 mol PEG per g Fe, heating is not maximized until concentrations above 0.43 mol PEG per g Fe on EMG1200. SAR on hydrophilic EMG308 was preserved at 400 W per g Fe regardless of the amount of PLink added to the core. Herein concentrations of IONP in VS55 (common CPA) significantly above our previous capabilities, sIONP at 10 mg Fe per mL, was reached, 25 mg Fe per mL of 308-PEG5000 and 60 mg Fe per mL of 1200-PEG5000, approaching stock EMG308 in water, 60 mg Fe per mL. Furthermore, at these concentrations cryopreserved Human dermal fibroblast cells were successfully nanowarmed (at applied fields described above), with higher viability as compared to convective rewarming in a water bath and heating rate close to 200 °C min-1, 2.5 times faster than our current system. Using PLink as the coating method allowed for higher concentrations of IONPs to be successfully suspended in CPA without affecting the heating ability. Additionally, the model ligand, PEG, allowed for increased stability over time in nanowarming experiments.
Collapse
Affiliation(s)
- Jacqueline L Pasek-Allen
- Department of Biomedical Engineering, University of Minnesota, 312 Church St. SE, Minneapolis, MN 55455, USA.
| | - Randall K Wilharm
- Department of Chemistry, University of Minnesota, 207 Pleasant St SE, Minneapolis, MN 55455, USA.
| | - Zhe Gao
- Mechanical Engineering, University of Minnesota, 111 Church Street Se, Minneapolis, MN 55455, USA.
| | - Valerie C Pierre
- Department of Chemistry, University of Minnesota, 207 Pleasant St SE, Minneapolis, MN 55455, USA.
| | - John C Bischof
- Department of Biomedical Engineering, University of Minnesota, 312 Church St. SE, Minneapolis, MN 55455, USA.
- Mechanical Engineering, University of Minnesota, 111 Church Street Se, Minneapolis, MN 55455, USA.
| |
Collapse
|
18
|
Liporagi Lopes LC, Korangath P, dos Santos SR, Gabrielson KL, Ivkov R, Casadevall A. Bionized Nanoferrite Particles Alter the Course of Experimental Cryptococcus neoformans Pneumonia. Antimicrob Agents Chemother 2022; 66:e0239921. [PMID: 35293784 PMCID: PMC9017294 DOI: 10.1128/aac.02399-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/12/2022] [Indexed: 01/09/2023] Open
Abstract
Cryptococcosis is a devastating fungal disease associated with high morbidity and mortality even when treated with antifungal drugs. Bionized nanoferrite (BNF) nanoparticles are powerful immunomodulators, but their efficacy for infectious diseases has not been investigated. Administration of BNF nanoparticles to mice with experimental cryptococcal pneumonia altered the outcome of infection in a dose response manner as measured by CFU and survival. The protective effects were higher at lower doses, with reductions in IL-2, IL-4, and TNF-α, consistent with immune modulation whereby reductions in inflammation translate into reduced host damage, clearance of infection, and longer survival.
Collapse
Affiliation(s)
- Livia C. Liporagi Lopes
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Preethi Korangath
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samuel R. dos Santos
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Kathleen L. Gabrielson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
19
|
El-Boubbou K, Lemine OM, Ali R, Huwaizi SM, Al-Humaid S, AlKushi A. Evaluating magnetic and thermal effects of various Polymerylated magnetic iron oxide nanoparticles for combined chemo-hyperthermia. NEW J CHEM 2022. [DOI: 10.1039/d1nj05791j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Killing cancer cells with heat: Assessing the heat-generation from Polymerylated magnetic iron oxide nanoparticles (PMNPs) for synergistic chemo-hyperthermia therapy.
Collapse
Affiliation(s)
- Kheireddine El-Boubbou
- Department of Basic Sciences, College of Science & Health Professions (COSHP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - O. M. Lemine
- Department of Physics, College of Sciences, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Rizwan Ali
- King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Sarah M. Huwaizi
- King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Sulaiman Al-Humaid
- Department of Basic Sciences, College of Science & Health Professions (COSHP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11481, Saudi Arabia
| | - Abdulmohsen AlKushi
- Department of Basic Sciences, College of Science & Health Professions (COSHP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11481, Saudi Arabia
| |
Collapse
|
20
|
Ammassam Veettil R, Marcano DC, Yuan X, Zaheer M, Adumbumkulath A, Lee R, Isenhart LC, Soriano N, Mhatre K, Joseph R, Mani SA, Shin CS, Acharya G. Dextran Sulfate Polymer Wafer Promotes Corneal Wound Healing. Pharmaceutics 2021; 13:pharmaceutics13101628. [PMID: 34683921 PMCID: PMC8539456 DOI: 10.3390/pharmaceutics13101628] [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] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/11/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
Eye injuries due to corneal abrasions, chemical spills, penetrating wounds, and microbial infections cause corneal scarring and opacification that result in impaired vision or blindness. However, presently available eye drop formulations of anti-inflammatory and antibiotic drugs are not effective due to their rapid clearance from the ocular surface or due to drug-related side effects such as cataract formation or increased intraocular pressure. In this article, we presented the development of a dextran sulfate-based polymer wafer (DS-wafer) for the effective modulation of inflammation and fibrosis and demonstrated its efficacy in two corneal injury models: corneal abrasion mouse model and alkali induced ocular burn mouse model. The DS-wafers were fabricated by the electrospinning method. We assessed the efficacy of the DS-wafer by light microscopy, qPCR, confocal fluorescence imaging, and histopathological analysis. These studies demonstrated that the DS-wafer treatment is significantly effective in modulating corneal inflammation and fibrosis and inhibited corneal scarring and opacification compared to the unsulfated dextran-wafer treated and untreated corneas. Furthermore, these studies have demonstrated the efficacy of dextran sulfate as an anti-inflammatory and antifibrotic polymer therapeutic.
Collapse
Affiliation(s)
- Remya Ammassam Veettil
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (R.A.V.); (M.Z.); (A.A.); (R.L.); (N.S.); (K.M.)
| | - Daniela C. Marcano
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA; (D.C.M.); (X.Y.); (L.C.I.)
| | - Xiaoyong Yuan
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA; (D.C.M.); (X.Y.); (L.C.I.)
| | - Mahira Zaheer
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (R.A.V.); (M.Z.); (A.A.); (R.L.); (N.S.); (K.M.)
| | - Aparna Adumbumkulath
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (R.A.V.); (M.Z.); (A.A.); (R.L.); (N.S.); (K.M.)
| | - Richard Lee
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (R.A.V.); (M.Z.); (A.A.); (R.L.); (N.S.); (K.M.)
| | - Lucas C. Isenhart
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA; (D.C.M.); (X.Y.); (L.C.I.)
| | - Nicole Soriano
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (R.A.V.); (M.Z.); (A.A.); (R.L.); (N.S.); (K.M.)
| | - Kirti Mhatre
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (R.A.V.); (M.Z.); (A.A.); (R.L.); (N.S.); (K.M.)
| | - Robiya Joseph
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.J.); (S.A.M.)
| | - Sendurai A. Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.J.); (S.A.M.)
| | - Crystal S. Shin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (R.A.V.); (M.Z.); (A.A.); (R.L.); (N.S.); (K.M.)
- Correspondence: (C.S.S.); (G.A.)
| | - Ghanashyam Acharya
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (R.A.V.); (M.Z.); (A.A.); (R.L.); (N.S.); (K.M.)
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA; (D.C.M.); (X.Y.); (L.C.I.)
- Correspondence: (C.S.S.); (G.A.)
| |
Collapse
|
21
|
Luengo Morato Y, Ovejero Paredes K, Lozano Chamizo L, Marciello M, Filice M. Recent Advances in Multimodal Molecular Imaging of Cancer Mediated by Hybrid Magnetic Nanoparticles. Polymers (Basel) 2021; 13:2989. [PMID: 34503029 PMCID: PMC8434540 DOI: 10.3390/polym13172989] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is the second leading cause of death in the world, which is why it is so important to make an early and very precise diagnosis to obtain a good prognosis. Thanks to the combination of several imaging modalities in the form of the multimodal molecular imaging (MI) strategy, a great advance has been made in early diagnosis, in more targeted and personalized therapy, and in the prediction of the results that will be obtained once the anticancer treatment is applied. In this context, magnetic nanoparticles have been positioned as strong candidates for diagnostic agents as they provide very good imaging performance. Furthermore, thanks to their high versatility, when combined with other molecular agents (for example, fluorescent molecules or radioisotopes), they highlight the advantages of several imaging techniques at the same time. These hybrid nanosystems can be also used as multifunctional and/or theranostic systems as they can provide images of the tumor area while they administer drugs and act as therapeutic agents. Therefore, in this review, we selected and identified more than 160 recent articles and reviews and offer a broad overview of the most important concepts that support the synthesis and application of multifunctional magnetic nanoparticles as molecular agents in advanced cancer detection based on the multimodal molecular imaging approach.
Collapse
Affiliation(s)
- Yurena Luengo Morato
- Nanobiotechnology for Life Sciences Lab, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal, 28040 Madrid, Spain; (Y.L.M.); (K.O.P.); (L.L.C.)
| | - Karina Ovejero Paredes
- Nanobiotechnology for Life Sciences Lab, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal, 28040 Madrid, Spain; (Y.L.M.); (K.O.P.); (L.L.C.)
- Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC F.S.P.), Calle Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Laura Lozano Chamizo
- Nanobiotechnology for Life Sciences Lab, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal, 28040 Madrid, Spain; (Y.L.M.); (K.O.P.); (L.L.C.)
| | - Marzia Marciello
- Nanobiotechnology for Life Sciences Lab, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal, 28040 Madrid, Spain; (Y.L.M.); (K.O.P.); (L.L.C.)
| | - Marco Filice
- Nanobiotechnology for Life Sciences Lab, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal, 28040 Madrid, Spain; (Y.L.M.); (K.O.P.); (L.L.C.)
- Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC F.S.P.), Calle Melchor Fernández Almagro 3, 28029 Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
22
|
Le Fur M, Ross A, Pantazopoulos P, Rotile N, Zhou I, Caravan P, Medarova Z, Yoo B. Radiolabeling and PET-MRI microdosing of the experimental cancer therapeutic, MN-anti-miR10b, demonstrates delivery to metastatic lesions in a murine model of metastatic breast cancer. Cancer Nanotechnol 2021; 12:16. [PMID: 34531932 PMCID: PMC8442631 DOI: 10.1186/s12645-021-00089-5] [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] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/01/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND In our earlier work, we identified microRNA-10b (miR10b) as a master regulator of the viability of metastatic tumor cells. This knowledge allowed us to design a miR10b-targeted therapeutic consisting of anti-miR10b and ultrasmall iron oxide magnetic nanoparticles (MN), termed MN-anti-miR10b. In mouse models of breast cancer, we demonstrated that MN-anti-miR10b caused durable regressions of established metastases with no evidence of systemic toxicity. As a first step towards translating MN-anti-miR10b for the treatment of metastatic breast cancer, we needed to determine if MN-anti-miR10b, which is so effective in mice, will also accumulate in human metastases. RESULTS In this study, we devised a method to efficiently radiolabel MN-anti-miR10b with Cu-64 (64Cu) and evaluated the pharmacokinetics and biodistribution of the radiolabeled product at two different doses: a therapeutic dose, referred to as macrodose, corresponding to 64Cu-MN-anti-miR10b co-injected with non-labeled MN-anti-miR10b, and a tracer level dose of 64Cu-MN-anti-miR10b, referred to as microdose. In addition, we evaluated the uptake of 64Cu-MN-anti-miR10b by metastatic lesions using both in vivo and ex vivo positron emission tomography-magnetic resonance imaging (PET-MRI). A comparable distribution of the therapeutic was observed after administration of a microdose or macrodose. Uptake of the therapeutic by metastatic lymph nodes, lungs, and bone was also demonstrated by PET-MRI with a significantly higher PET signal than in the same organs devoid of metastatic lesions. CONCLUSION Our results demonstrate that PET-MRI following a microdose injection of the agent will accurately reflect the innate biodistribution of the therapeutic. The tools developed in the present study lay the groundwork for the clinical testing of MN-anti-miR10b and other similar therapeutics in patients with cancer.
Collapse
Affiliation(s)
- Mariane Le Fur
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
- Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, MA 02129 USA
| | - Alana Ross
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Pamela Pantazopoulos
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Nicholas Rotile
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
- Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, MA 02129 USA
| | - Iris Zhou
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
- Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, MA 02129 USA
| | - Peter Caravan
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
- Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, MA 02129 USA
| | - Zdravka Medarova
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Byunghee Yoo
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| |
Collapse
|
23
|
Kok HP, Cressman ENK, Ceelen W, Brace CL, Ivkov R, Grüll H, Ter Haar G, Wust P, Crezee J. Heating technology for malignant tumors: a review. Int J Hyperthermia 2021; 37:711-741. [PMID: 32579419 DOI: 10.1080/02656736.2020.1779357] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The therapeutic application of heat is very effective in cancer treatment. Both hyperthermia, i.e., heating to 39-45 °C to induce sensitization to radiotherapy and chemotherapy, and thermal ablation, where temperatures beyond 50 °C destroy tumor cells directly are frequently applied in the clinic. Achievement of an effective treatment requires high quality heating equipment, precise thermal dosimetry, and adequate quality assurance. Several types of devices, antennas and heating or power delivery systems have been proposed and developed in recent decades. These vary considerably in technique, heating depth, ability to focus, and in the size of the heating focus. Clinically used heating techniques involve electromagnetic and ultrasonic heating, hyperthermic perfusion and conductive heating. Depending on clinical objectives and available technology, thermal therapies can be subdivided into three broad categories: local, locoregional, or whole body heating. Clinically used local heating techniques include interstitial hyperthermia and ablation, high intensity focused ultrasound (HIFU), scanned focused ultrasound (SFUS), electroporation, nanoparticle heating, intraluminal heating and superficial heating. Locoregional heating techniques include phased array systems, capacitive systems and isolated perfusion. Whole body techniques focus on prevention of heat loss supplemented with energy deposition in the body, e.g., by infrared radiation. This review presents an overview of clinical hyperthermia and ablation devices used for local, locoregional, and whole body therapy. Proven and experimental clinical applications of thermal ablation and hyperthermia are listed. Methods for temperature measurement and the role of treatment planning to control treatments are discussed briefly, as well as future perspectives for heating technology for the treatment of tumors.
Collapse
Affiliation(s)
- H Petra Kok
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik N K Cressman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wim Ceelen
- Department of GI Surgery, Ghent University Hospital, Ghent, Belgium
| | - Christopher L Brace
- Department of Radiology and Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Holger Grüll
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Gail Ter Haar
- Department of Physics, The Institute of Cancer Research, London, UK
| | - Peter Wust
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Geppert M, Himly M. Iron Oxide Nanoparticles in Bioimaging - An Immune Perspective. Front Immunol 2021; 12:688927. [PMID: 34211476 PMCID: PMC8239972 DOI: 10.3389/fimmu.2021.688927] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/01/2021] [Indexed: 01/22/2023] Open
Abstract
Iron oxide nanoparticles (IONPs) bear big hopes in nanomedicine due to their (potential) applications in tumor therapy, drug delivery or bioimaging. However, as foreign entities, such particles may be recognized by the immune system and, thus, lead to inflammation, hypersensitivity or anaphylactic shock. In addition, an overload with iron is known to cause oxidative stress. In this short review, we summarize the biological effects of such particles with a major focus on IONP-formulations used for bioimaging purposes and their effects on the human immune system. We conclude that especially the characteristics of the particles (size, shape, surface charge, coating, etc.) as well as the presence of bystander substances, such as bacterial endotoxin are important factors determining the resulting biological and immunological effects of IONPs. Further studies are needed in order to establish clear structure-activity relationships.
Collapse
Affiliation(s)
- Mark Geppert
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | | |
Collapse
|
25
|
One-pot synthesis of carboxymethyl-dextran coated iron oxide nanoparticles (CION) for preclinical fMRI and MRA applications. Neuroimage 2021; 238:118213. [PMID: 34116153 PMCID: PMC8418149 DOI: 10.1016/j.neuroimage.2021.118213] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/15/2021] [Accepted: 05/25/2021] [Indexed: 11/21/2022] Open
Abstract
Superparamagnetic iron-oxide nanoparticles are robust contrast agents for magnetic resonance imaging (MRI) used for sensitive structural and functional mapping of the cerebral blood volume (CBV) when administered intravenously. To date, many CBV-MRI studies are conducted with Feraheme, manufactured for the clinical treatment of iron-deficiency. Unfortunately, Feraheme is currently not available outside the United States due to commercial and regulatory constraints, making CBV-MRI methods either inaccessible or very costly to achieve. To address this barrier, we developed a simple, one-pot recipe to synthesize Carboxymethyl-dextran coated Iron Oxide Nanoparticles, namely, “CION”, suitable for preclinical CBV-MRI applications. Here we disseminate a step-by-step instruction of our one-pot synthesis protocol, which allows CION to be produced in laboratories with minimal cost. We also characterized different CION-conjugations by manipulating polymer to metal stoichiometric ratio in terms of their size, surface chemistry, and chemical composition, and shifts in MR relaxivity and pharmacokinetics. We performed several proof-of-concept experiments in vivo, demonstrating the utility of CION for functional and structural MRI applications, including hypercapnic CO2 challenge, visual stimulation, targeted optogenetic stimulation, and microangiography. We also present evidence that CION can serve as a cross-modality research platform by showing concurrent in vivo optical and MRI measurement of CBV using fluorescent-labeled CION. The simplicity and cost-effectiveness of our one-pot synthesis method should allow researchers to reproduce CION and tailor the relaxivity and pharmacokinetics according to their imaging needs. It is our hope that this work makes CBV-MRI more openly available and affordable for a variety of research applications.
Collapse
|
26
|
Ramya S, Thiruvenkataswamy S, Kavithaa K, Preethi S, Winster H, Balachander V, Paulpandi M, Narayanasamy A. pH Dependent Drug Release of Silibinin, a Polyphenol Conjugated with Magnetic Nanoparticle Against the Human Colon Cancer Cell. J CLUST SCI 2021. [DOI: 10.1007/s10876-020-01789-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
27
|
Natarajan P, Tomich JM. Understanding the influence of experimental factors on bio-interactions of nanoparticles: Towards improving correlation between in vitro and in vivo studies. Arch Biochem Biophys 2020; 694:108592. [PMID: 32971033 PMCID: PMC7503072 DOI: 10.1016/j.abb.2020.108592] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022]
Abstract
Bionanotechnology has developed rapidly over the past two decades, owing to the extensive and versatile, functionalities and applicability of nanoparticles (NPs). Fifty-one nanomedicines have been approved by FDA since 1995, out of the many NPs based formulations developed to date. The general conformation of NPs consists of a core with ligands coating their surface, that stabilizes them and provides them with added functionalities. The physicochemical properties, especially the surface composition of NPs influence their bio-interactions to a large extent. This review discusses recent studies that help understand the nano-bio interactions of iron oxide and gold NPs with different surface compositions. We discuss the influence of the experimental factors on the outcome of the studies and, thus, the importance of standardization in the field of nanotechnology. Recent studies suggest that with careful selection of experimental parameters, it is possible to improve the positive correlation between in vitro and in vivo studies. This provides a fundamental understanding of the NPs which helps in assessing their potential toxic side effects and may aid in manipulating them further to improve their biocompatibility and biosafety.
Collapse
|
28
|
Zhu S, Zhu K, Li J, Lai H, Wang C. Nano-Biomaterials for the Delivery of Therapeutic and Monitoring Cues for Aortic Diseases. Front Bioeng Biotechnol 2020; 8:583879. [PMID: 33224934 PMCID: PMC7674648 DOI: 10.3389/fbioe.2020.583879] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/07/2020] [Indexed: 01/09/2023] Open
Abstract
The aorta is the largest artery in the body, so any diseases or conditions which could cause damage to the aorta would put patients at considerable and life-threatening risk. In the management of aortic diseases, the major treatments include drug therapy, endovascular treatment, and surgical treatment, which are of great danger or with a poor prognosis. The delivery of nano-biomaterials provides a potential development trend and an emerging field where we could monitor patients’ conditions and responses to the nanotherapeutics. One of the putative applications of nanotechnology is ultrasensitive monitoring of cardiovascular markers by detecting and identifying aneurysms. Moreover, the use of nanosystems for targeted drug delivery can minimize the systemic side effects and enhance drug positioning and efficacy compared to conventional drug therapies. This review shows some examples of utilizing nano-biomaterials in in vitro organ and cell culture experiments and explains some developing technologies in delivering and monitoring regenerative therapeutics.
Collapse
Affiliation(s)
- Shichao Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Jun Li
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| |
Collapse
|
29
|
Sancho-Albero M, Sebastián V, Sesé J, Pazo-Cid R, Mendoza G, Arruebo M, Martín-Duque P, Santamaría J. Isolation of exosomes from whole blood by a new microfluidic device: proof of concept application in the diagnosis and monitoring of pancreatic cancer. J Nanobiotechnology 2020; 18:150. [PMID: 33092584 PMCID: PMC7579907 DOI: 10.1186/s12951-020-00701-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Exosomes are endocytic-extracellular vesicles with a diameter around 100 nm that play an essential role on the communication between cells. In fact, they have been proposed as candidates for the diagnosis and the monitoring of different pathologies (such as Parkinson, Alzheimer, diabetes, cardiac damage, infection diseases or cancer). RESULTS In this study, magnetic nanoparticles (Fe3O4NPs) were successfully functionalized with an exosome-binding antibody (anti-CD9) to mediate the magnetic capture in a microdevice. This was carried out under flow in a 1.6 mm (outer diameter) microchannel whose wall was in contact with a set of NdFeB permanent magnets, giving a high magnetic field across the channel diameter that allowed exosome separation with a high yield. To show the usefulness of the method, the direct capture of exosomes from whole blood of patients with pancreatic cancer (PC) was performed, as a proof of concept. The captured exosomes were then subjected to analysis of CA19-9, a protein often used to monitor PC patients. CONCLUSIONS Here, we describe a new microfluidic device and the procedure for the isolation of exosomes from whole blood, without any need of previous isolation steps, thereby facilitating translation to the clinic. The results show that, for the cases analyzed, the evaluation of CA19-9 in exosomes was highly sensitive, compared to serum samples.
Collapse
Affiliation(s)
- María Sancho-Albero
- Department of Chemical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009, Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029, Madrid, Spain
| | - Víctor Sebastián
- Department of Chemical Engineering, University of Zaragoza, 50018, Zaragoza, Spain.
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009, Zaragoza, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029, Madrid, Spain.
| | - Javier Sesé
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009, Zaragoza, Spain
- Department of Condensed Matter Physics, University of Zaragoza, 50009, Zaragoza, Spain
| | - Roberto Pazo-Cid
- Medical Oncology Service, Miguel Servet Hospital, 50009, Zaragoza, Spain
| | - Gracia Mendoza
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029, Madrid, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS-Aragón), 50009, Zaragoza, Spain
| | - Manuel Arruebo
- Department of Chemical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009, Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029, Madrid, Spain
| | - Pilar Martín-Duque
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029, Madrid, Spain.
- Instituto de Investigación Sanitaria de Aragón (IIS-Aragón), 50009, Zaragoza, Spain.
- Health Sciences Institute of Aragón (IACS), 50009, Zaragoza, Spain.
- Fundación Araid, 50018, Zaragoza, Spain.
- Universidad San Jorge, 50830, Zaragoza, Spain.
| | - Jesús Santamaría
- Department of Chemical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009, Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029, Madrid, Spain
| |
Collapse
|
30
|
Song C, Gao C, Zhao J, Wang Z. Construction of long-circulation EpCAM targeted drug delivery system and its application in the diagnosis and treatment of breast cancer. J Biomater Appl 2020; 35:947-957. [PMID: 33081605 DOI: 10.1177/0885328220965135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Docetaxel (DOX) is usually one of drugs used for breast cancer treatment. The key of targeted drug delivery therapy is to deliver effective drugs directly and safely to the tumor focus via an efficient targeting drug carrier with immunogenicity. In this study, Long-circulating targeted drug carrying microspheres (DOX-PEG-EpCAM-MNs) entrapping DOX were constructed. In addition, both cytotoxicity and magnetic resonance imaging (MRI) analyses were performed to establish a mouse model and further complete corresponding performance analysis.The results showed that the average particle size of DOX-PEG-EpCAM-MNs was 139.3 ± 1.6 nm. Morphological analysis proves that they are spherical and uniformly dispersed. The Corresponding entrapment rate and drug carrying capacity are 82.43% and 7.16% respectively. Additionally, MRI shows that they have the capability to track tumor cells within 5 days. This study established a safe and efficient breast cancer cells targeted long-circulating drug delivery system.
Collapse
Affiliation(s)
- Chao Song
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chao Gao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jing Zhao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhenxin Wang
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
31
|
Ahmad A, Ansari MM, Kumar A, Vyawahare A, Mishra RK, Jayamurugan G, Raza SS, Khan R. Comparative acute intravenous toxicity study of triple polymer-layered magnetic nanoparticles with bare magnetic nanoparticles in Swiss albino mice. Nanotoxicology 2020; 14:1362-1380. [DOI: 10.1080/17435390.2020.1829144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Anas Ahmad
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| | - Md. Meraj Ansari
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| | - Ajay Kumar
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| | - Akshay Vyawahare
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| | | | | | - Syed Shadab Raza
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era’s Lucknow Medical College Hospital, Lucknow, India
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Lucknow, India
| | - Rehan Khan
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| |
Collapse
|
32
|
Badman RP, Moore SL, Killian JL, Feng T, Cleland TA, Hu F, Wang MD. Dextran-coated iron oxide nanoparticle-induced nanotoxicity in neuron cultures. Sci Rep 2020; 10:11239. [PMID: 32641693 PMCID: PMC7343881 DOI: 10.1038/s41598-020-67724-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 02/27/2020] [Indexed: 11/09/2022] Open
Abstract
Recent technological advances have introduced diverse engineered nanoparticles (ENPs) into our air, water, medicine, cosmetics, clothing, and food. However, the health and environmental effects of these increasingly common ENPs are still not well understood. In particular, potential neurological effects are one of the most poorly understood areas of nanoparticle toxicology (nanotoxicology), in that low-to-moderate neurotoxicity can be subtle and difficult to measure. Culturing primary neuron explants on planar microelectrode arrays (MEAs) has emerged as one of the most promising in vitro techniques with which to study neuro-nanotoxicology, as MEAs enable the fluorescent tracking of nanoparticles together with neuronal electrical activity recording at the submillisecond time scale, enabling the resolution of individual action potentials. Here we examine the dose-dependent neurotoxicity of dextran-coated iron oxide nanoparticles (dIONPs), a common type of functionalized ENP used in biomedical applications, on cultured primary neurons harvested from postnatal day 0-1 mouse brains. A range of dIONP concentrations (5-40 µg/ml) were added to neuron cultures, and cells were plated either onto well plates for live cell, fluorescent reactive oxidative species (ROS) and viability observations, or onto planar microelectrode arrays (MEAs) for electrophysiological measurements. Below 10 µg/ml, there were no dose-dependent cellular ROS increases or effects in MEA bursting behavior at sub-lethal dosages. However, above 20 µg/ml, cell death was obvious and widespread. Our findings demonstrate a significant dIONP toxicity in cultured neurons at concentrations previously reported to be safe for stem cells and other non-neuronal cell types.
Collapse
Affiliation(s)
- Ryan P Badman
- Department of Physics and LASSP, Cornell University, Ithaca, NY, 14853, USA.,Center for Brain Science, RIKEN, Saitama, 351-0198, Japan
| | - Shanna L Moore
- Department of Physics and LASSP, Cornell University, Ithaca, NY, 14853, USA.,Howard Hughes Medical Institute, Cornell University, Ithaca, NY, 14853, USA
| | - Jessica L Killian
- Department of Physics and LASSP, Cornell University, Ithaca, NY, 14853, USA.,Howard Hughes Medical Institute, Cornell University, Ithaca, NY, 14853, USA.,Quantum Biosystems, Menlo Park, CA, 94025, USA
| | - Tuancheng Feng
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Thomas A Cleland
- Department of Psychology, Cornell University, Ithaca, NY, 14853, USA
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Michelle D Wang
- Department of Physics and LASSP, Cornell University, Ithaca, NY, 14853, USA. .,Howard Hughes Medical Institute, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
33
|
Oei AL, Korangath P, Mulka K, Helenius M, Coulter JB, Stewart J, Velarde E, Crezee J, Simons B, Stalpers LJA, Kok HP, Gabrielson K, Franken NAP, Ivkov R. Enhancing the abscopal effect of radiation and immune checkpoint inhibitor therapies with magnetic nanoparticle hyperthermia in a model of metastatic breast cancer. Int J Hyperthermia 2020; 36:47-63. [PMID: 31795835 PMCID: PMC7017719 DOI: 10.1080/02656736.2019.1685686] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Purpose: Enhancing immune responses in triple negative breast cancers (TNBCs) remains a challenge. Our study aimed to determine whether magnetic iron oxide nanoparticle (MION) hyperthermia (HT) can enhance abscopal effects with radiotherapy (RT) and immune checkpoint inhibitors (IT) in a metastatic TNBC model.Methods: One week after implanting 4T1-luc cells into the mammary glands of BALB/c mice, tumors were treated with RT (3 × 8 Gy)±local HT, mild (HTM, 43 °C/20 min) or partially ablative (HTAbl, 45 °C/5 min plus 43 °C/15 min),±IT with anti-PD-1 and anti-CTLA-4 antibodies (both 4 × 10 mg/kg, i.p.). Tumor growth was measured daily. Two weeks after treatment, lungs and livers were harvested for histopathology evaluation of metastases.Results: Compared to untreated controls, all treatment groups demonstrated a decreased tumor volume; however, when compared against surgical resection, only RT + HTM+IT, RT + HTAbl+IT and RT + HTAbl had similar or smaller tumors. These cohorts showed more infiltration of CD3+ T-lymphocytes into the primary tumor. Tumor growth effects were partially reversed with T-cell depletion. Combinations that proved most effective for primary tumors generated modest reductions in numbers of lung metastases. Conversely, numbers of lung metastases showed potential to increase following HT + IT treatment, particularly when compared to RT. Compared to untreated controls, there was no improvement in survival with any treatment.Conclusions: Single-fraction MION HT added to RT + IT improved local tumor control and recruitment of CD3+ T-lymphocytes, with only a modest effect to reduce lung metastases and no improvement in overall survival. HT + IT showed potential to increase metastatic dissemination to lungs.
Collapse
Affiliation(s)
- Arlene L Oei
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands.,Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Preethi Korangath
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathleen Mulka
- Department of Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mikko Helenius
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan B Coulter
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacqueline Stewart
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Esteban Velarde
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Johannes Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Brian Simons
- Department of Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lukas J A Stalpers
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands.,Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H Petra Kok
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Kathleen Gabrielson
- Department of Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicolaas A P Franken
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands.,Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
34
|
Korangath P, Barnett JD, Sharma A, Henderson ET, Stewart J, Yu SH, Kandala SK, Yang CT, Caserto JS, Hedayati M, Armstrong TD, Jaffee E, Gruettner C, Zhou XC, Fu W, Hu C, Sukumar S, Simons BW, Ivkov R. Nanoparticle interactions with immune cells dominate tumor retention and induce T cell-mediated tumor suppression in models of breast cancer. SCIENCE ADVANCES 2020; 6:eaay1601. [PMID: 32232146 PMCID: PMC7096167 DOI: 10.1126/sciadv.aay1601] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 01/06/2020] [Indexed: 05/02/2023]
Abstract
The factors that influence nanoparticle fate in vivo following systemic delivery remain an area of intense interest. Of particular interest is whether labeling with a cancer-specific antibody ligand ("active targeting") is superior to its unlabeled counterpart ("passive targeting"). Using models of breast cancer in three immune variants of mice, we demonstrate that intratumor retention of antibody-labeled nanoparticles was determined by tumor-associated dendritic cells, neutrophils, monocytes, and macrophages and not by antibody-antigen interactions. Systemic exposure to either nanoparticle type induced an immune response leading to CD8+ T cell infiltration and tumor growth delay that was independent of antibody therapeutic activity. These results suggest that antitumor immune responses can be induced by systemic exposure to nanoparticles without requiring a therapeutic payload. We conclude that immune status of the host and microenvironment of solid tumors are critical variables for studies in cancer nanomedicine and that nanoparticle technology may harbor potential for cancer immunotherapy.
Collapse
Affiliation(s)
- Preethi Korangath
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - James D. Barnett
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Anirudh Sharma
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Elizabeth T. Henderson
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Jacqueline Stewart
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Shu-Han Yu
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Sri Kamal Kandala
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore 21218, USA
| | - Chun-Ting Yang
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- National Taiwan University, Taipei 10617, Taiwan
| | - Julia S. Caserto
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Mohammad Hedayati
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Todd D. Armstrong
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Elizabeth Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | - Xian C. Zhou
- Department of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Wei Fu
- Department of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Chen Hu
- Department of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Saraswati Sukumar
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Brian W. Simons
- Department of Urology, James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore 21218, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore 21218, USA
- Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore 21218, USA
| |
Collapse
|
35
|
Bolandparvaz A, Vapniarsky N, Harriman R, Alvarez K, Saini J, Zang Z, Van De Water J, Lewis JS. Biodistribution and toxicity of epitope-functionalized dextran iron oxide nanoparticles in a pregnant murine model. J Biomed Mater Res A 2020; 108:1186-1202. [PMID: 32031743 DOI: 10.1002/jbm.a.36893] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
Abstract
In pursuit of a preventive therapeutic for maternal autoantibody-related (MAR) autism, we assessed the toxicity, biodistribution, and clearance of a MAR specific peptide-functionalized dextran iron oxide nanoparticle system in pregnant murine dams. We previously synthesized ~15 nm citrate-coated dextran iron oxide nanoparticles (DIONPs), surface-modified with polyethylene glycol and MAR peptides to produce systems for nanoparticle-based autoantibody reception and entrapments (SNAREs). First, we investigated their immunogenicity and MAR lactate dehydrogenase B antibody uptake in murine serum in vitro. To assess biodistribution and toxicity, as well as systemic effects, we performed in vivo clinical and post mortem pathological evaluations. We observed minimal production of inflammatory cytokines-interleukin 10 (IL-10) and IL-12 following in vitro exposure of macrophages to SNAREs. We established the maximum tolerated dose of SNAREs to be 150 mg/kg at which deposition of iron was evident in the liver and lungs by histology and magnetic resonance imaging but no concurrent evidence of liver toxicity or lung infarction was detected. Further, SNAREs exhibited slower clearance from the maternal blood in pregnant dams compared to DIONPs based on serum total iron concentration. These findings demonstrated that the SNAREs have a prolonged presence in the blood and are safe for use in pregnant mice as evidenced by no associated organ damage, failure, inflammation, and fetal mortality. Determination of the MTD dose sets the basis for future studies investigating the efficacy of our nanoparticle formulation in a MAR autism mouse model.
Collapse
Affiliation(s)
- Amir Bolandparvaz
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Natalia Vapniarsky
- Department of Pathology Microbiology and Immunology, University of California Davis, Davis, California, USA
| | - Rian Harriman
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Kenneth Alvarez
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Jasmeen Saini
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Zexi Zang
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Judy Van De Water
- M.I.N.D. (Medical Investigation of Neurodevelopmental Disorders), University of California Davis, Davis, California, USA.,Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, California, USA
| | - Jamal S Lewis
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| |
Collapse
|
36
|
Baboci L, Capolla S, Di Cintio F, Colombo F, Mauro P, Dal Bo M, Argenziano M, Cavalli R, Toffoli G, Macor P. The Dual Role of the Liver in Nanomedicine as an Actor in the Elimination of Nanostructures or a Therapeutic Target. JOURNAL OF ONCOLOGY 2020; 2020:4638192. [PMID: 32184825 PMCID: PMC7060440 DOI: 10.1155/2020/4638192] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/16/2020] [Indexed: 02/06/2023]
Abstract
The development of nanostructures for therapeutic purpose is rapidly growing, following the results obtained in vivo in animal models and in the clinical trials. Unfortunately, the potential therapeutic efficacy is not completely exploited, yet. This is mainly due to the fast clearance of the nanostructures in the body. Nanoparticles and the liver have a unique interaction because the liver represents one of the major barriers for drug delivery. This interaction becomes even more relevant and complex when the drug delivery strategies employing nanostructures are proposed for the therapy of liver diseases, such as hepatocellular carcinoma (HCC). In this case, the selective delivery of therapeutic nanoparticles to the tumor microenvironment collides with the tendency of nanostructures to be quickly eliminated by the organ. The design of a new therapeutic approach based on nanoparticles to treat HCC has to particularly take into consideration passive and active mechanisms to avoid or delay liver elimination and to specifically address cancer cells or the cancer microenvironment. This review will analyze the different aspects concerning the dual role of the liver, both as an organ carrying out a clearance activity for the nanostructures and as target for therapeutic strategies for HCC treatment.
Collapse
Affiliation(s)
- Lorena Baboci
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO) di Aviano IRCCS, Aviano, Italy
| | - Sara Capolla
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO) di Aviano IRCCS, Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Federica Di Cintio
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO) di Aviano IRCCS, Aviano, Italy
| | - Federico Colombo
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Prisca Mauro
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO) di Aviano IRCCS, Aviano, Italy
| | - Monica Argenziano
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO) di Aviano IRCCS, Aviano, Italy
| | - Paolo Macor
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO) di Aviano IRCCS, Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
37
|
Magnetic Materials and Systems: Domain Structure Visualization and Other Characterization Techniques for the Application in the Materials Science and Biomedicine. INORGANICS 2020. [DOI: 10.3390/inorganics8010006] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Magnetic structures have attracted a great interest due to their multiple applications, from physics to biomedicine. Several techniques are currently employed to investigate magnetic characteristics and other physicochemical properties of magnetic structures. The major objective of this review is to summarize the current knowledge on the usage, advances, advantages, and disadvantages of a large number of techniques that are currently available to characterize magnetic systems. The present review, aiming at helping in the choice of the most suitable method as appropriate, is divided into three sections dedicated to characterization techniques. Firstly, the magnetism and magnetization (hysteresis) techniques are introduced. Secondly, the visualization methods of the domain structures by means of different probes are illustrated. Lastly, the characterization of magnetic nanosystems in view of possible biomedical applications is discussed, including the exploitation of magnetism in imaging for cell tracking/visualization of pathological alterations in living systems (mainly by magnetic resonance imaging, MRI).
Collapse
|
38
|
Xin X, Du X, Xiao Q, Azevedo HS, He W, Yin L. Drug Nanorod-Mediated Intracellular Delivery of microRNA-101 for Self-sensitization via Autophagy Inhibition. NANO-MICRO LETTERS 2019; 11:82. [PMID: 34138035 PMCID: PMC7770860 DOI: 10.1007/s40820-019-0310-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/30/2019] [Indexed: 05/20/2023]
Abstract
Autophagy is closely related to the drug resistance and metastasis in cancer therapy. Nanoparticle-mediated co-delivery of combinatorial therapy with small-molecular drugs and nucleic acids is promising to address drug resistance. Here, a drug-delivering-drug (DDD) platform consisting of anti-tumor-drug nanorods as a vehicle for cytosol delivery of nucleic acid (miR-101) with potent autophagic-inhibition activity is reported for combinatorial therapy. The developed 180-nm nanoplatform, with total drug loading of up to 66%, delivers miR-101 to cancer cells, with threefold increase in intracellular level compared to conventional gene carriers and inhibits the autophagy significantly, along with above twofold reduction in LC3II mRNA and approximately fivefold increase in p62 mRNA over the control demonstrated in the results in vivo. And in turn, the delivery of miR-101 potentiates the drug's ability to kill cancer cells, with a threefold increase in apoptosis over that of chemotherapy alone. The anti-tumor study in vivo indicates the combined therapy that enables a reduction of 80% in tumor volume and > twofold increase in apoptosis than of the single-drug strategy. In summary, via the carrier-free strategy of DDD, this work provides a delivery platform that can be easily customized to overcome drug resistance and facilitates the delivery of combined therapy of small-molecular drugs and nucleic acids.
Collapse
Affiliation(s)
- Xiaofei Xin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xiaoqing Du
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Qingqing Xiao
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Helena S Azevedo
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary, University of London, London, E1 4NS, UK
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| | - Lifang Yin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
39
|
Hu Y, Chen X, Xu Y, Han X, Wang M, Gong T, Zhang ZR, John Kao W, Fu Y. Hierarchical assembly of hyaluronan coated albumin nanoparticles for pancreatic cancer chemoimmunotherapy. NANOSCALE 2019; 11:16476-16487. [PMID: 31453622 DOI: 10.1039/c9nr03684a] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Pancreatic cancer is a highly malignant carcinoma with limited effective treatment options, resulting in a poor patient survival rate of less than 5%. In this study, cationic albumin nanoparticles were assembled with negatively charged hyaluronic acid (HA) to achieve a hierarchical nanostructure and efficient delivery of small molecule drugs to the tumor site in the pancreas. A combination of chemotherapy with indoleamine-2,3-dioxygenase (IDO) inhibition was explored to enhance the chemotherapeutic efficacy in vivo. Hydrophobic celastrol (CLT) and hydrophilic 1-methyltryptophan (MT) were concurrently loaded in HA coated cationic albumin nanoparticles (HNPs) with an average size of ∼300 nm. The size of HNPs was reduced in the presence of hyaluronidase to facilitate penetration into deep tumor tissues. Also, the biodistribution study in the C57BL/6 mice xenograft model showed enhanced tumor accumulation and prolonged circulation of HNPs. Compared with CLT solution, the combination of CLT with MT showed significantly enhanced tumor inhibition in both xenograft and orthotopic pancreatic cancer mice models via downregulating the immunosuppressive tumor microenvironment. Taken together, the combination of CLT with MT administered via HNPs represents a highly promising strategy for targeted pancreatic cancer therapy.
Collapse
Affiliation(s)
- Ying Hu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Xue Chen
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Yingying Xu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Xianru Han
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Mou Wang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Zhi-Rong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - W John Kao
- Department of Industrial and Manufacturing Systems Engineering, Biomedical Engineering, and Chemical Biology Centre, The University of Hong Kong, Pokfulam, HKSAR, China
| | - Yao Fu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
40
|
Zinicovscaia I, Grozdov D, Yushin N, Ivlieva A, Petritskaya E, Rogatkin D. Neutron activation analysis as a tool for tracing the accumulation of silver nanoparticles in tissues of female mice and their offspring. J Radioanal Nucl Chem 2019. [DOI: 10.1007/s10967-019-06746-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
41
|
Kevadiya BD, Ottemann BM, Thomas MB, Mukadam I, Nigam S, McMillan J, Gorantla S, Bronich TK, Edagwa B, Gendelman HE. Neurotheranostics as personalized medicines. Adv Drug Deliv Rev 2019; 148:252-289. [PMID: 30421721 PMCID: PMC6486471 DOI: 10.1016/j.addr.2018.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022]
Abstract
The discipline of neurotheranostics was forged to improve diagnostic and therapeutic clinical outcomes for neurological disorders. Research was facilitated, in largest measure, by the creation of pharmacologically effective multimodal pharmaceutical formulations. Deployment of neurotheranostic agents could revolutionize staging and improve nervous system disease therapeutic outcomes. However, obstacles in formulation design, drug loading and payload delivery still remain. These will certainly be aided by multidisciplinary basic research and clinical teams with pharmacology, nanotechnology, neuroscience and pharmaceutic expertise. When successful the end results will provide "optimal" therapeutic delivery platforms. The current report reviews an extensive body of knowledge of the natural history, epidemiology, pathogenesis and therapeutics of neurologic disease with an eye on how, when and under what circumstances neurotheranostics will soon be used as personalized medicines for a broad range of neurodegenerative, neuroinflammatory and neuroinfectious diseases.
Collapse
Affiliation(s)
- Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brendan M Ottemann
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Midhun Ben Thomas
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saumya Nigam
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - JoEllyn McMillan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
42
|
Vallabani NVS, Singh S, Karakoti AS. Magnetic Nanoparticles: Current Trends and Future Aspects in Diagnostics and Nanomedicine. Curr Drug Metab 2019; 20:457-472. [DOI: 10.2174/1389200220666181122124458] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/23/2018] [Accepted: 10/10/2018] [Indexed: 12/13/2022]
Abstract
Background:
Biomedical applications of Magnetic Nanoparticles (MNPs) are creating a major impact on
disease diagnosis and nanomedicine or a combined platform called theranostics. A significant progress has been
made to engineer novel and hybrid MNPs for their multifunctional modalities such as imaging, biosensors, chemotherapeutic
or photothermal and antimicrobial agents. MNPs are successfully applied in biomedical applications
due to their unique and tunable properties such as superparamagnetism, stability, and biocompatibility. Approval of
ferumoxytol (feraheme) for MRI and the fact that several Superparamagnetic Iron Oxide Nanoparticles (SPIONs) are
currently undergoing clinical trials have paved a path for future MNPs formulations. Intensive research is being
carried out in designing and developing novel nanohybrids for multiple applications in nanomedicine.
Objective:
The objective of the present review is to summarize recent developments of MNPs in imaging modalities
like MRI, CT, PET and PA, biosensors and nanomedicine including their role in targeting and drug delivery. Relevant
theory and examples of the use of MNPs in these applications have been cited and discussed to create a thorough
understanding of the developments in this field.
Conclusion:
MNPs have found widespread use as contrast agents in imaging modalities, as tools for bio-sensing, and
as therapeutic and theranostics agents. Multiple formulations of MNPs are in clinical testing and may be accepted in
clinical settings in near future.
Collapse
Affiliation(s)
- Naga Veera Srikanth Vallabani
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Sanjay Singh
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Ajay Singh Karakoti
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
43
|
Andra S, Balu SK, Jeevanandham J, Muthalagu M, Vidyavathy M, Chan YS, Danquah MK. Phytosynthesized metal oxide nanoparticles for pharmaceutical applications. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:755-771. [PMID: 31098696 DOI: 10.1007/s00210-019-01666-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/06/2019] [Indexed: 01/19/2023]
Abstract
Developments in nanotechnology field, specifically, metal oxide nanoparticles have attracted the attention of researchers due to their unique sensing, electronic, drug delivery, catalysis, optoelectronics, cosmetics, and space applications. Physicochemical methods are used to fabricate nanosized metal oxides; however, drawbacks such as high cost and toxic chemical involvement prevail. Recent researches focus on synthesizing metal oxide nanoparticles through green chemistry which helps in avoiding the involvement of toxic chemicals in the synthesis process. Bacteria, fungi, and plants are the biological sources that are utilized for the green nanoparticle synthesis. Due to drawbacks such as tedious maintenance and the time needed for the nanoparticle formation, plant extracts are widely used in nanoparticle production. In addition, plants are available all over the world and phytosynthesized nanoparticles show comparatively less toxicity towards mammalian cells. Secondary metabolites including flavonoids, terpenoids, and saponins are present in plant extracts, and these are highly responsible for nanoparticle formation and reduction of toxicity. Hence, this article gives an overview of recent developments in the phytosynthesis of metal oxide nanoparticles and their toxic analysis in various cells and animal models. Also, their possible mechanism in normal and cancer cells, pharmaceutical applications, and their efficiency in disease treatment are also discussed.
Collapse
Affiliation(s)
- Swetha Andra
- Department of Textile Technology, Anna University, Chennai, Tamil Nadu, 600025, India
| | - Satheesh Kumar Balu
- Department of Ceramic Technology, Anna University, Chennai, Tamil Nadu, 600025, India
| | - Jaison Jeevanandham
- Department of Chemical Engineering, Faculty of Engineering and Science, Curtin University, CDT 250, 98009, Miri, Sarawak, Malaysia
| | - Murugesan Muthalagu
- Department of Textile Technology, Anna University, Chennai, Tamil Nadu, 600025, India
| | - Manisha Vidyavathy
- Department of Ceramic Technology, Anna University, Chennai, Tamil Nadu, 600025, India
| | - Yen San Chan
- Department of Chemical Engineering, Faculty of Engineering and Science, Curtin University, CDT 250, 98009, Miri, Sarawak, Malaysia
| | | |
Collapse
|
44
|
Pinheiro WO, Fascineli ML, Farias GR, Horst FH, de Andrade LR, Corrêa LH, Magalhães KG, Sousa MH, de Almeida MC, Azevedo RB, Lacava ZGM. The influence of female mice age on biodistribution and biocompatibility of citrate-coated magnetic nanoparticles. Int J Nanomedicine 2019; 14:3375-3388. [PMID: 31123402 PMCID: PMC6511116 DOI: 10.2147/ijn.s197888] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Magnetic nanoparticles (MNPs) have been successfully tested for several purposes in medical applications. However, knowledge concerning the effects of nanostructures on elderly organisms is remarkably scarce. PURPOSE To fill part of this gap, this work aimed to investigate biocompatibility and bio-distribution aspects of magnetic nanoparticles coated with citrate (NpCit) in both elderly and young healthy mice. METHODS NpCit (2.4 mg iron) was administered intraperitoneally, and its toxicity was evaluated for 28 days through clinical, biochemical, hematological, and histopathological examinations. In addition, its biodistribution was evaluated by spectrometric (inductively coupled plasma optical emission spectrometry) and histological methods. RESULTS NpCit presented age-dependent effects, inducing very slight and temporary biochemical and hematological changes in young animals. These changes were even weaker than the effects of the aging process, especially those related to the hematological data, tumor necrosis factor alpha, and nitric oxide levels. On the other hand, NpCit showed a distinct set of results in the elderly group, sometimes reinforcing (decrease of lymphocytes and increase of monocytes) and sometimes opposing (erythrocyte parameters and cytokine levels) the aging changes. Leukocyte changes were still observed on the 28th day after treatment in the elderly group. Slight evidence of a decrease in liver and immune functions was detected in elderly mice treated or not treated with NpCit. It was noted that tissue damage or clinical changes related to aging or to the NpCit treatment were not observed. As detected for aging, the pattern of iron biodistribution was significantly different after NpCit administration: extra iron was detected until the 28th day, but in different organs of elderly (liver and kidneys) and young (spleen, liver, and lungs) mice. CONCLUSION Taken together, the data show NpCit to be a stable and reasonably biocompatible sample, especially for young mice, and thus appropriate for biomedical applications. The data showed important differences after NpCit treatment related to the animals' age, and this emphasizes the need for further studies in older animals to appropriately extend the benefits of nanotechnology to the elderly population.
Collapse
Affiliation(s)
- Willie O Pinheiro
- Department of Genetics and Morphology, Institute of Biological Sciences, CNANO, University of Brasilia, Brasilia, DF 70910-900, Brazil,
- Post-graduation Program in Molecular Pathology, Faculty of Medicine, University of Brasilia, Brasília, DF 70910-900, Brazil,
| | - Maria L Fascineli
- Department of Genetics and Morphology, Institute of Biological Sciences, CNANO, University of Brasilia, Brasilia, DF 70910-900, Brazil,
| | - Gabriel R Farias
- Department of Genetics and Morphology, Institute of Biological Sciences, CNANO, University of Brasilia, Brasilia, DF 70910-900, Brazil,
| | - Frederico H Horst
- Department of Genetics and Morphology, Institute of Biological Sciences, CNANO, University of Brasilia, Brasilia, DF 70910-900, Brazil,
| | - Laise Rodrigues de Andrade
- Department of Genetics and Morphology, Institute of Biological Sciences, CNANO, University of Brasilia, Brasilia, DF 70910-900, Brazil,
| | - Luis Henrique Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, DF 70910-900, Brazil
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, DF 70910-900, Brazil
| | - Marcelo Henrique Sousa
- Green Nanotechnology Group, Faculty of Ceilandia, University of Brasilia, Brasília, DF 72220-900, Brazil
| | - Marcos C de Almeida
- Department of Genetics and Morphology, Institute of Biological Sciences, CNANO, University of Brasilia, Brasilia, DF 70910-900, Brazil,
| | - Ricardo B Azevedo
- Department of Genetics and Morphology, Institute of Biological Sciences, CNANO, University of Brasilia, Brasilia, DF 70910-900, Brazil,
| | - Zulmira G M Lacava
- Department of Genetics and Morphology, Institute of Biological Sciences, CNANO, University of Brasilia, Brasilia, DF 70910-900, Brazil,
- Post-graduation Program in Molecular Pathology, Faculty of Medicine, University of Brasilia, Brasília, DF 70910-900, Brazil,
| |
Collapse
|
45
|
Giannousi K, Koutroumpis E, Georgiadou V, Karagkounis V, Dendrinou-Samara C. Nanoplatforms of Manganese Ferrite Nanoparticles Functionalized with Anti-Inflammatory Drugs. Eur J Inorg Chem 2019. [DOI: 10.1002/ejic.201801539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Kleoniki Giannousi
- Laboratory of Inorganic Chemistry; Department of Chemistry; Aristotle University of Thessaloniki; 54124 Thessaloniki Greece
| | - Emmanouil Koutroumpis
- Laboratory of Inorganic Chemistry; Department of Chemistry; Aristotle University of Thessaloniki; 54124 Thessaloniki Greece
| | - Violetta Georgiadou
- Laboratory of Inorganic Chemistry; Department of Chemistry; Aristotle University of Thessaloniki; 54124 Thessaloniki Greece
| | - Vasilis Karagkounis
- Laboratory of Inorganic Chemistry; Department of Chemistry; Aristotle University of Thessaloniki; 54124 Thessaloniki Greece
| | - Catherine Dendrinou-Samara
- Laboratory of Inorganic Chemistry; Department of Chemistry; Aristotle University of Thessaloniki; 54124 Thessaloniki Greece
| |
Collapse
|
46
|
Mai BT, Balakrishnan PB, Barthel MJ, Piccardi F, Niculaes D, Marinaro F, Fernandes S, Curcio A, Kakwere H, Autret G, Cingolani R, Gazeau F, Pellegrino T. Thermoresponsive Iron Oxide Nanocubes for an Effective Clinical Translation of Magnetic Hyperthermia and Heat-Mediated Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:5727-5739. [PMID: 30624889 PMCID: PMC6376448 DOI: 10.1021/acsami.8b16226] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/09/2019] [Indexed: 05/22/2023]
Abstract
The use of magnetic nanoparticles in oncothermia has been investigated for decades, but an effective combination of magnetic nanoparticles and localized chemotherapy under clinical magnetic hyperthermia (MH) conditions calls for novel platforms. In this study, we have engineered magnetic thermoresponsive iron oxide nanocubes (TR-cubes) to merge MH treatment with heat-mediated drug delivery, having in mind the clinical translation of the nanoplatform. We have chosen iron oxide based nanoparticles with a cubic shape because of their outstanding heat performance under MH clinical conditions, which makes them benchmark agents for MH. Accomplishing a surface-initiated polymerization of strongly interactive nanoparticles such as our iron oxide nanocubes, however, remains the main challenge to overcome. Here, we demonstrate that it is possible to accelerate the growth of a polymer shell on each nanocube by simple irradiation of a copper-mediated polymerization with a ultraviolet light (UV) light, which both speeds up the polymerization and prevents nanocube aggregation. Moreover, we demonstrate herein that these TR-cubes can carry chemotherapeutic doxorubicin (DOXO-loaded-TR-cubes) without compromising their thermoresponsiveness both in vitro and in vivo. In vivo efficacy studies showed complete tumor suppression and the highest survival rate for animals that had been treated with DOXO-loaded-TR-cubes, only when they were exposed to MH. The biodistribution of intravenously injected TR-cubes showed signs of renal clearance within 1 week and complete clearance after 5 months. This biomedical platform works under clinical MH conditions and at a low iron dosage, which will enable the translation of dual MH/heat-mediated chemotherapy, thus overcoming the clinical limitation of MH: i.e., being able to monitor tumor progression post-MH-treatment by magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Binh T. Mai
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy
- Dipartimento
di Chimica e Chimica Industriale, Università
di Genova, Via Dodecaneso,
31, 16146 Genova, Italy
| | - Preethi B. Balakrishnan
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy
- Dipartimento
di Chimica e Chimica Industriale, Università
di Genova, Via Dodecaneso,
31, 16146 Genova, Italy
| | | | | | - Dina Niculaes
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy
- Dipartimento
di Chimica e Chimica Industriale, Università
di Genova, Via Dodecaneso,
31, 16146 Genova, Italy
| | | | - Soraia Fernandes
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy
| | - Alberto Curcio
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy
| | - Hamilton Kakwere
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy
| | - Gwennhael Autret
- Centre
de Recherche Cardiovasculaire de Paris 56, rue Leblanc, 75737 Paris Cedex 15, France
| | | | - Florence Gazeau
- Laboratoire
Matière et Systèmes Complexes, UMR 7057, CNRS and University Paris Diderot, 75205 Paris Cedex 13, France
| | | |
Collapse
|
47
|
Farouk F, Shamma R. Chemical structure modifications and nano-technology applications for improving ADME-Tox properties, a review. Arch Pharm (Weinheim) 2019; 352:e1800213. [DOI: 10.1002/ardp.201800213] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/02/2018] [Accepted: 11/11/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Faten Farouk
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry; Ahram Canadian University; Giza Egypt
| | - Rehab Shamma
- Faculty of Pharmacy, Department of Pharmaceutics and Industrial Pharmacy; Cairo University; Cairo Egypt
| |
Collapse
|