Next Article in Journal
Application of Mobile Operators’ Data in Modern Geographical Research
Previous Article in Journal
Pathogenesis of Glaucoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Entry

Magnetite Nanoparticles for Biomedical Applications

by
Kirill D. Petrov
and
Alexey S. Chubarov
*
Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia
*
Author to whom correspondence should be addressed.
Encyclopedia 2022, 2(4), 1811-1828; https://doi.org/10.3390/encyclopedia2040125
Submission received: 30 September 2022 / Revised: 7 November 2022 / Accepted: 11 November 2022 / Published: 14 November 2022
(This article belongs to the Section Medicine & Pharmacology)

Definition

:
Magnetic nanoparticles (MNPs) have great potential in various areas such as medicine, cancer therapy and diagnostics, biosensing, and material science. In particular, magnetite (Fe3O4) nanoparticles are extensively used for numerous bioapplications due to their biocompatibility, high saturation magnetization, chemical stability, large surface area, and easy functionalization. This paper describes magnetic nanoparticle physical and biological properties, emphasizing synthesis approaches, toxicity, and various biomedical applications, focusing on the most recent advancements in the areas of therapy, diagnostics, theranostics, magnetic separation, and biosensing.

Graphical Abstract

1. Introduction

Nanotechnology combines various areas of science. The small sizes of nanomaterials possess unique chemical, physical, and biological properties. To date, many nanomaterial types have been described, and many more will be developed for various applications. Magnetic nanoparticles have great potential in biochemistry, nanomedicine, and bio-inspired material areas [1,2,3,4,5,6,7,8]. One of the most promising magnetic nanoparticles is iron oxide (II, III) due to their ferrimagnetism [1,9,10,11]. In particular, Fe3O4 magnetite nanoparticles (MNPs) have demonstrated a promising effect in numerous applications [1,2,3,4,5,6,7,8].
MNPs have become a vital tool for material science, biochemistry, diagnostics, magnetic drug and gene delivery, hyperthermia, magnetic resonance imaging (MRI), and theranostics [1,3,4,5,7,9,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30]. The manipulation of MNPs by an external magnetic field is essential for bioseparation and biosensing areas [1,6,17,30,31,32,33]. Moreover, magnetic transport of MNPs to the tissue allows targeted therapy and diagnostics (theranostics) applications [1,2,3,4,5,6,7,8,34]. A combination of possible local heating (hyperthermia), anticancer drug delivery, and monitoring by MRI or other imaging technology open the tremendous potential for cancer treatment [1,2,3,4,5,8,11,16,21,35].
Many papers about the synthesis, coating, and applications of MNPs have been reported [1,2,3,4,5,6,7,8,26,34,35]. The number of articles with the key term “magnetic nanoparticles” increases every year (Figure 1). The actual number of papers in the area is much higher, which can be calculated using other keywords. However, before 1996, less than 100 articles per year were published annually by the Scopus database. After the first successful clinical trial in 1996, the number of papers greatly increased. Since 2013, more than 5000 manuscripts have been published annually. Such results are associated with the increasing recognition of MNPs in achieving excellent results in various applications.
MNPs show high field irreversibility, high saturation field, and superparamagnetism, which are highly dependent on particle size and surface coating. The relationship between MNP size and magnetism (coercivity) has been extensively reported [36,37,38,39,40]. The coercivity gradually increases for bulk nanoparticles to a maximum value at a particular size. In this region, the magnetization is stable and nonuniform (Weiss domains, magnetic multi-domain state). The critical size of the magnetite nanoparticles, above which they become multi-domain, has been theoretically calculated and is 76 nm and 128 nm, respectively, for cubic and spherical nanoparticles [38]. However, the experimental data indicate that the critical size of transition between single- and multi-domain magnetic structure highly depends on the crystal structure and coating [37,38,41]. By reducing the size of the nanoparticles, the coercivity rapidly decreases to zero, reaching a superparamagnetic state [36,37]. Superparamagnetism is especially important in applications such as drug delivery and imaging. Particle sizes below 20 nm are required to achieve superparamagnetism for magnetite MNPs. Superparamagnetic MNPs provide a stronger response to external magnetic fields than simple MNPs. Frenkel J. and Doefman J. in 1930 predicted that, below a critical size, MNPs would consist of a single magnetic domain [42]. However, superparamagnetic MNP synthesis was achieved half a century later.
Research on nano-emulsion began in 1943 (Figure 2, historical timeline) [43]. However, the nanotechnology concept was first proposed in 1959 by Richard Feynman in the lecture entitled “Plenty of Room at the Bottom”. This was a historical event in nanoscience. The first synthesis of iron nanoparticles by gas condensation was achieved in 1981. The concept of using magnetic forces for enhanced therapeutic and imaging performance has evolved over the years. Two of the milestones were the development of MNPs for imaging purposes in 1990 and silica-coated MNPs in 1995. Since 2000, numerous studies have investigated the potential applications of MNPs and nanocomposites with magnetic cores. Another essential development was the successful magnetic hyperthermia clinical trials in 2010. Magnetic hyperthermia utilizes MNPs that are exposed to an alternating magnetic field to generate heat in local regions [23,44]. Magnetic hyperthermia therapy was first proposed much earlier, in 1957. However, about fifty years were required to synthesize stable and non-toxic MNPs with optimal physical properties. Colloidal stability, biocompatibility, and toxicity studies are crucial for in vivo application. Recently, numerous MNP-based “Smart” nanocomposites with pH-stimuli-responsive drug release, theranostics, and multimodal constructions have been developed [5,8,13,24,25,45,46,47,48,49]. Numerous research papers have focused on the possible procedures for MNPs synthesis, coating, drug-loading, toxicity studies, and clinical trials [1,2,8,20,44,50,51,52,53,54].

2. Synthesis of Magnetic Nanoparticles

Many research articles have focused on the synthesis and coating procedures of MNPs to obtain desired morphology, particle size, physico-chemical properties, and biocompatibility [1,7,9,10,11,28,50,51,55]. The basic techniques involved in MNP synthesis are physical, chemical, and biological in nature (Figure 3, classification of synthetic approaches). Each route produces MNPs with various properties, which highly depend on the synthesis conditions [1,8,35,56,57,58]. This section presents the most common syntheses, their advantages, and their effects over different properties of MNPs.
Co-precipitation is a widely used, simple, and cost-effective chemical synthesis, which proceeds in an aqueous solution with a high yield and purity (Table 1). However, the procedure often requires surfactant and clear reaction parameters (concentration, temperature, tube size, mixing speed, etc.) [1,56]. The method is conducted in a mixture of Fe (II) and Fe (III) salts with base (NH3 or NaOH solution) at ambient temperature or slight heating. To prevent Fe (II) oxidation in base conditions, the presence of inert gas flow is recommended. The changes of such simple reaction conditions result in not having the same size and shape of nanoparticles, such that reproduction of the synthesis method usually fails to produce MNPs with the same physico-chemical and biological properties. Moreover, “in one person’s hands”, the MNPs size usually varies from one synthesis to another. The only possibility is to thoroughly record all actions, equipment, and chemical manufacturers during synthesis.
A hydrothermal or solvothermal method is used to prepare MNPs under high pressure and temperature, generally carried out in an autoclave [1,8,35]. This method yields excellent shape-controlled, monodisperse ultrafine magnetite nanopowders. This route is chosen over other approaches to grow high-crystalline MNPs. However, the synthesis procedures usually require a large reaction time.
In the sonochemical or sonolysis method, acoustic cavitation produces bubbles via ultrasound [1]. The shape and size of MNPs can be easily controlled through the intensity of irradiation, irradiation time, and reaction time. Despite the synthesis requiring high-intensity ultrasound, the reaction occurs quickly in mild conditions.
Micro- or nano-emulsion method include a stable liquid mixture (usually water and ‘fatty’ solvent) and amphiphilic surfactant. The properties of MNPs are highly dependent on surfactant type. The method has a narrow working window to obtain stable emulsion, high solvent consumption, and low MNP yield [57]. These shortcomings make the emulsion method unprofitable.
Recently, some modern biosynthesis approaches and green technologies for MNP synthesis have been reported [1,56,58]. Of course, the first, and best, ‘researcher’ of this technology is the magnetostatic bacterium. Using intracellular biomineralization processes, magnetostatic bacteria can synthesize specific organelle magnetosomes [58]. At the moment, many researchers use plant extracts, DNA, and proteins as templates, microorganisms, viruses, and fungi for MNPs green/biosynthesis [1,56,58] (Table 1).
In addition to these synthetic routes, gas-phase deposition, ball milling, spattering, laser ablation, electron beam deposition, aerosol spray pyrolysis, electrochemical synthesis, and wet reduction microbial synthesis, among others, have been reported [1,8,35,56,57,58]. Table 1 summarizes some primary synthetic methods along with their conditions and characteristics.

3. Toxicity of Magnetic Nanoparticles

The toxicity of MNPs is an important factor for future healthcare applications [50]. The synthetic procedures of MNPs may look easy. However, MNPs toxicity depends on size, coating, surface modification, etc. MNPs with a size higher than 200 nm will be filtrated by the spleen. The nanoparticles less than 10 nm can be quickly removed through renal clearance. The surface chemistry of MNPs is also important. Having the wrong coating for MNPs leads to magnetic core oxidation, aggregation, instability in physiological liquids, and high reactive oxygen species (ROS) formation [3,13,50,51,54,59,60,61]. Various organic molecules (tannic, polyacrylic, lauric, myristic, hyaluronic, and oleic acids), inorganic coating (gold, silica, calcium carbonate), artificial (tween or polysorbate, polyethylene glycol, polyethylene imine, etc.), and natural polymers (e.g., dextran, chitosan, proteins such as albumin, casein, etc.) are used to improve MNP properties [3,7,11,22,24,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87]. However, no universal recipe exists. For example, PEG-coating enhances the colloidal stability of MNPs and decreases protein adsorption [24,79,88]. Oleic acid is the primary surfactant for improving magnetic properties and stabilize the MNPs [84,85]. Protein-coating provides greater biocompatibility and biodegradability and less immunogenicity than MNPs [7,62,89,90,91]. For the major human plasma protein, human serum albumin (HSA) [92,93,94], adsorption on the MNPs surface prevents nucleation and aggregation and increases colloidal stability in aqueous solution in a wide pH range [62,72,95,96,97,98,99,100,101,102,103]. Albumin coating prevents non-specific interactions with blood components and immune response [62,95,97,101,102,104]. HSA enhances biocompatibility, prolongs blood circulation of MNPs, and provides targeted delivery to tumors [7].
The cytotoxic effect of MNPs lies in Fe ion release, dysregulation of ion channels and gene expression, immune response, inflammation, ulceration, metabolic disorders, decrease in growth rate or changes of alterations, etc. [5,50,54]. The literature on the topic usually presents the simplest cytotoxicity assay (MTT test) on cancer cell lines [105,106]. The MTT assay usually shows only ‘acute toxic effects’. It does not provide interaction with blood proteins and cells, tissue-specific toxicity, chronic toxicity, etc. Moreover, cancer cells have activated “survival systems”. Recent research has moved significantly forward with in vivo toxicological assays [54]. The MNP dose, initial concentration, biodistribution, and circulation time greatly influence the trials. MNP accumulation in tissues may interfere with physiological Fe metabolism and activate inflammatory or immune responses [50,54,60]. MNP degradation in the cells may lead to ROS formation with cell or mitochondrial membrane damage, adverse cell proliferation, DNA oxidation with subsequent point mutation formation, and cell death. Extended toxicity experiments are hard-going work for the many researchers involved, which greatly slows down the progress in this area [54]. Despite the widespread introduction of MNPs, new and easier toxicity tests are required. Integration of nanotoxicology and nanomedicine into one element has proven to be a significant step toward the sustainable development of nanotechnology for biomedical applications [107,108].

4. Biomedical Applications of Magnetic Nanoparticles

MNPs have great potential in the nanomedicine field. Magnetic nanoparticles have been widely used for analytical purposes for biomolecule detection, disease diagnosis, and treatment. This section focuses on the therapeutic, diagnostic, theranostic, magnetic separation, and biosensing applications of MNPs.

4.1. Therapy

Nanoparticles have become extremely popular for cancer treatment. Their ability to pass cell membranes is a potential method to solve the problem of drug resistance [109]. MNPs may act as a carrier providing magnetically guided drug delivery (Figure 4, top right). Moving the MNPs by the influence of an external magnetic field is currently used for modern chemotherapy in several diseases, including cancers [109,110]. Solid tumors usually have an immature, highly permeable vasculature. For better growth, tumors produce a nutritional gap in which nanoconstructions accumulate. This effect is known as the enhanced permeation and retention effect (EPR effect). For cancer treatment, MNPs can act as nanocarriers for chemotherapeutic drugs. The whole nanoconstruction must be stable in blood and not prematurely release the drug. In the tumor media, the drug release should be effective. Controlled drug release is a stumbling stone for drug-loaded MNPs. A combination of siRNA and antisense oligonucleotides may be used instead of a chemotherapeutic drug [111,112,113,114]. Such cocktails can drastically influence the targeted gene activity, resulting in a therapeutic effect [115]. This technology is called magnetofection and is defined as the delivery of nucleic acids using MNPs. Magnetofection has been introduced as a powerful tool for nucleic acid delivery into cells [114]. The method shows good results in vitro and in vivo [113,114,116,117,118,119,120,121,122,123,124,125].
Several strategies could be used for such an approach. One is covalent drug conjugation, which possesses hard-synthesized smart construction [8,35,109]. A physiologically cleavable linker for drug binding may be used in this case. However, much work is also conducted on noncovalent drug-loading procedures on the MNP surface. For example, doxorubicin (DOX) is a primary chemotherapeutic drug for drug-loaded MNP synthesis. DOX has been FDA-approved for more than 50 years. However, multidrug resistance and many side effects are also associated with DOX [126,127]. DOX-loaded MNPs are much more popular [128,129,130,131,132,133,134,135,136,137,138]. Researchers have tried to obtain good drug capacity per MNP and high stability at plasma pH~7.4. At the same time, effective drug release is required in the tumor environment and endosomal compartment with pH~4–5. However, for such promising constructions, simple MNPs may not be used. The magnetic core may be protected by noble metals (e.g., gold [139]), calcium carbonate [65,140], various polymers, and proteins [7,8,35,125,141]. For example, albumin protein MNP bio-inspired coating results in low ROS production, excellent biocompatibility, and moderate particle uptake, which is shown in in vitro and in vivo experiments [7]. Moreover, albumin coating could be chemically modified by address groups for cancer-tissue-targeted delivery [142,143,144], magnetic resonance imaging, or fluorescence reporter groups [145,146,147,148]. The protein–nanoparticle corona is an important tool to prevent MNPs’ toxic side effects and enhance stability and biocompatibility.
Hyperthermia is a well-established method for cancer treatment [23]. MNP in an alternating magnetic field generates heat (Figure 4, bottom left). The high temperature in local regions damages tumor cells. However, the results of the heat release are highly dependent on the MNPs’ size, shape, geometry, and coating [23,149]. The recent progress in the chemical synthesis of MNPs with well-controlled composition and morphology has allowed us to go forward with hyperthermia in medicine. The other limitation is that MNPs should be successfully delivered to the desired tissue. Combined cancer therapy may be obtained for drug-loaded MNPs. A smart drug-loaded construction can use the hyperthermia effect to disrupt the tumor tissue. In this way, a cumulative effect in the cells can occur [150,151]. Hyperthermia temperature rise may be easily controlled, which could be exploited in multiple therapeutic approaches. For example, deep-brain stimulation (DBS) could be obtained by a magnetic hyperthermia procedure (Figure 4, top left). Under low heating, temperature-sensitive ion channels cause an influx of Ca2+-sensitive promoter and activation of individual neurons [152,153]. Moreover, shot MNP heating by an external radio-frequency magnetic field provides the local pH changes, which regulate the complex formation and cell processes [154].
MNPs can magnetically stimulate stem cells, which influences protein synthesis and gene expression. This effect may be used for damaged tissues or organ repair and regeneration (tissue engineering, Figure 4) [155]. MNPs have been used for bone, tendon, cartilage regeneration, and neuroregeneration [156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173].

4.2. Diagnostics and Theranostics

Over the past few decades, MNPs have become essential for various imaging techniques [1,20,28,97,174,175,176]. MNPs play an important role in magnetic resonance imaging (MRI) and magnetic particle imaging (MPI). MRI is a primary medical method that provides high resolution and excellent contrast capabilities without the use of damaging ionizing radiation [28]. It is often used for disease detection, diagnosis, and treatment monitoring. MRI is based on the nuclear magnetic resonance (NMR) effect of water protons, which depends on the concentration, relaxation times (T1 and T2), and mobility of the water molecules in tissues. Using MNPs, it is possible to change the signal from the water protons and see the localization of the paramagnetic tracer. MNP-based contrast agents are helpful for malignant tumor detection. Since 1990, numerous papers have been published on MNP applications for MRI [20,28,174,175,176] (Figure 5, application examples).
Using T1 contrast agents, the MRI image becomes brighter and T2 darker. MNPs are often known to shorten the T2 time, which leads to a dark zone on the MRI image. The MNP coating influences the relaxation time due to changes in water molecule availability near the magnetic core [28]. However, the universal recipe that obtains the best MRI agent is unknown [28]. In 1996, the FDA approved the Feridex® T2-contrast agent for liver lesion detection. Feridex is a dextran-coated aqueous colloid of superparamagnetic MNPs. In the same period, several dextran-coated MNPs (e.g., Endorem, Resovist) were also approved for clinics. However, Feridex and some other contrast agents were withdrawn in 2009 due to side effects and major safety concerns (see Section 3, possible toxicity effects) [20]. Notably, the novel biocompatible MNP-based MRI contrast agents are still receiving attention for clinical use. There are a number of successful cell experiments, extensive safety experiments, and undergoing pre-clinical animal studies [20]. However, clinical trials for use in humans are not successful or in the early stages of investigations [20].
MPI is a relatively novel non-invasive technique [5]. MPI was first conceived in 2001 and reported in 2005. It uses changing magnetic fields to generate a signal from superparamagnetic MNP tracers. The signal can be observed without background, providing information about the probe location with a high signal-to-noise ratio. Among the applications of MPI are cell tracking, tumor detection, and blood-pool, vascular, and perfusion imaging (Figure 5) [5]. The high sensitivity of MPI is suitable for cancer detection in the early stage.
Recently, various MNP-based constructions for multimodal imaging have been developed [20]. These include systems for simultaneous MRI and MPI detection and combinations with computed tomography (CT), single-photon emission computed tomography (SPECT), positron emission tomography (PET), optical and ultrasound imaging, and magneto-acoustic tomography [8,20,177,178]. Combining MNP-based imaging and therapeutic approaches provides possible theranostic construction production (Figure 6) [5,8,20]. Theranostic MNPs offer considerable potential for drug-resistant cancer treatment. The primary theranostic system combines MRI and chemotherapeutic drugs or hyperthermia treatment (Figure 6). The progress in this area is limited, but has increased over the last several years [5,7,8,20].

4.3. Magnetic Separation, MNP-Based Biosensors, and Magnetic Microreactors

The purity of materials plays a crucial role in further analysis, which is why fast and low-cost separation approaches have to be designed. Magnetic separation is the process by which various compound separations use a magnet to attract magnetic substances (e.g., MNPs). The process is an important method for the bioseparation and quantification of peptides, proteins, nucleic acids, and metabolites [6,22,32,33,179,180,181,182,183,184,185,186,187,188,189,190,191,192]. Moreover, magnetic separation may be used for more than simple biomolecule capture. Recently, magnetic cell separation has become a vital method for a wide range of applications [6]. Magnetic separation using MNPs is more efficient than traditional approaches, which require several stages, organic solvents, and chromatography systems [2,32,182,184,193,194,195,196]. The procedure of magnetic separation is presented in Figure 7.
Of course, these are not simple MNPs used for organic molecule and biopolymer separation. The surface of the MNPs could be modified by ligands, which can specifically bind the target molecule. Antibodies, aptamers, DNA/RNA, or proteins are used for such modification [6,16,32,180,191]. For example, avidin-coated MNPs and biotinylated oligonucleotides are utilized for specific nucleic acid separation. The biotinylated oligonucleotide forms a duplex with a targeted nucleic acid. Afterward, MNPs are added, and biotin residue interacts with avidin, forming a high-affinity complex. Such avidin-coated MNPs are commercially available elsewhere.
MNP-based biosensors could be widely used in the detection of antibiotics, toxins, antigens, proteins, nucleic acids, disease biomarkers, pathogenic bacteria, and viruses [17]. One of the MNP-based sensing strategies is presented in Figure 8. MNP may be used for target magnetic separation into the detector zone or magnetoresistive sensors [17,197,198,199]. The first approach enhances detection sensitivity, decreases analysis time, and provides a high signal-to-noise ratio and complex sample analysis [199]. Without a magnet, the MNP moves along with the sample in the solution, but when using a magnet, the MNP-target complex is focused on the signal generation zone. The detection of the target may be achieved with a wide range of methods: electrochemical (e.g., voltammetry, amperometry, potentiometry, electrochemiluminescence, and impedance), optical (e.g., surface plasmon resonance, and fluorescence spectroscopy), and piezoelectric [199,200]. Magnetoresistive sensors are based on the binding of MNPs to a sensor surface, which results in electrical current changes. These sensors demonstrate extremely high sensitivity. However, it is not possible to detect multiple analytes in the solution since the change in the magnetic field is restricted to only one parameter [199].
Magnetic microreactors have attracted wide attention in nanobiotechnology [201,202,203,204,205,206,207,208]. Enzyme immobilization on the MNPs with reliable magnetic properties facilitates solid-phase biocatalysis. This strategy offers high stability in enzymes during biomass processing and their easy separation by an external magnetic field. The process of enzyme recovery becomes an easy procedure for the protection of enzymes from inhibition, pH, and thermal denaturation in the continuous-mode microreactor. The MNP–enzyme system has major advantages in homo- and heterogeneous catalysis in high mass-transfer rates, selectivity, high yield, easy recovery, recyclability, and high activity [208]. Future research should be oriented toward magnetic enzyme microreactor configurations, optimization, and application extensions. As it stands, this technology may already represent a significant step in biotechnology.

5. Conclusions

MNPs are a promising platform for creating novel therapy, diagnostics, theranostics, and sensor constructions. Such systems may occupy a niche in the development of next-generation drugs for disease detection and treatment. Moreover, multifunctional MNPs with bio-inspired coatings could be a breakthrough in nanomedicine. Even after many years of research, there are still many challenges that must be taken into account for translating MNPs into clinics. Understanding the physical, chemical, and biological problems and principles of property manipulation for MNP may lead to a new era in nanomedicine.

Author Contributions

Conceptualization, A.S.C.; writing—original draft preparation, A.S.C. and K.D.P.; writing—review and editing, A.S.C.; supervision, A.S.C.; project administration, A.S.C.; funding acquisition, A.S.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Russian Science Foundation (grant no. 21-74-00120).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ganapathe, L.S.; Mohamed, M.A.; Yunus, R.M.; Berhanuddin, D.D. Magnetite (Fe3O4) nanoparticles in biomedical application: From synthesis to surface functionalisation. Magnetochemistry 2020, 6, 68. [Google Scholar] [CrossRef]
  2. Anik, M.I.; Hossain, M.K.; Hossain, I.; Mahfuz, A.M.U.B.; Rahman, M.T.; Ahmed, I. Recent progress of magnetic nanoparticles in biomedical applications: A review. Nano Sel. 2021, 2, 1146–1186. [Google Scholar] [CrossRef]
  3. Shabatina, T.I.; Vernaya, O.I.; Shabatin, V.P.; Melnikov, M.Y. Magnetic nanoparticles for biomedical purposes: Modern trends and prospects. Magnetochemistry 2020, 6, 30. [Google Scholar] [CrossRef]
  4. Socoliuc, V.; Peddis, D.; Petrenko, V.I.; Avdeev, M.V.; Susan-Resiga, D.; Szabó, T.; Turcu, R.; Tombácz, E.; Vékás, L. Magnetic nanoparticle systems for nanomedicine—A materials science perspective. Magnetochemistry 2020, 6, 2. [Google Scholar] [CrossRef] [Green Version]
  5. Hepel, M. Magnetic nanoparticles for nanomedicine. Magnetochemistry 2020, 6, 3. [Google Scholar] [CrossRef] [Green Version]
  6. Frenea-Robin, M.; Marchalot, J. Basic Principles and Recent Advances in Magnetic Cell Separation. Magnetochemistry 2022, 8, 11. [Google Scholar] [CrossRef]
  7. Chubarov, A.S. Serum Albumin for Magnetic Nanoparticles Coating. Magnetochemistry 2022, 8, 13. [Google Scholar] [CrossRef]
  8. Mittal, A.; Roy, I.; Gandhi, S. Magnetic Nanoparticles: An Overview for Biomedical Applications. Magnetochemistry 2022, 8, 107. [Google Scholar] [CrossRef]
  9. Katz, E. Synthesis, properties and applications of magnetic nanoparticles and nanowires—A brief introduction. Magnetochemistry 2019, 5, 61. [Google Scholar] [CrossRef] [Green Version]
  10. Antone, A.J.; Sun, Z.; Bao, Y. Preparation and application of iron oxide nanoclusters. Magnetochemistry 2019, 5, 45. [Google Scholar] [CrossRef]
  11. Kudr, J.; Haddad, Y.; Richtera, L.; Heger, Z.; Cernak, M.; Adam, V.; Zitka, O. Magnetic nanoparticles: From design and synthesis to real world applications. Nanomaterials 2017, 7, 243. [Google Scholar] [CrossRef] [PubMed]
  12. Anderson, S.D.; Gwenin, V.V.; Gwenin, C.D. Magnetic Functionalized Nanoparticles for Biomedical, Drug Delivery and Imaging Applications. Nanoscale Res. Lett. 2019, 14, 188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lamichhane, N.; Sharma, S.; Parul; Verma, A.K.; Roy, I.; Sen, T. Iron oxide-based magneto-optical nanocomposites for in vivo biomedical applications. Biomedicines 2021, 9, 288. [Google Scholar] [CrossRef] [PubMed]
  14. Chouhan, R.S.; Horvat, M.; Ahmed, J.; Alhokbany, N.; Alshehri, S.M.; Gandhi, S. Magnetic nanoparticles—A multifunctional potential agent for diagnosis and therapy. Cancers 2021, 13, 2213. [Google Scholar] [CrossRef]
  15. Dulińska-Litewka, J.; Łazarczyk, A.; Hałubiec, P.; Szafrański, O.; Karnas, K.; Karewicz, A. Superparamagnetic iron oxide nanoparticles-current and prospective medical applications. Materials 2019, 12, 617. [Google Scholar] [CrossRef] [Green Version]
  16. Stueber, D.D.; Villanova, J.; Aponte, I.; Xiao, Z. Magnetic Nanoparticles in Biology and Medicine: Past, Present, and Future Trends. Pharmaceutics 2021, 13, 943. [Google Scholar] [CrossRef]
  17. Krishnan; Goud Magnetic Particle Bioconjugates: A Versatile Sensor Approach. Magnetochemistry 2019, 5, 64. [CrossRef] [Green Version]
  18. Sharma, B.; Pervushin, K. Magnetic nanoparticles as in vivo tracers for alzheimer’s disease. Magnetochemistry 2020, 6, 13. [Google Scholar] [CrossRef] [Green Version]
  19. Bruschi, M.L.; de Toledo, L.d.A.S. Pharmaceutical applications of iron-oxide magnetic nanoparticles. Magnetochemistry 2019, 5, 50. [Google Scholar] [CrossRef] [Green Version]
  20. Creţu, B.E.B.; Dodi, G.; Shavandi, A.; Gardikiotis, I.; Şerban, I.L.; Balan, V. Imaging constructs: The rise of iron oxide nanoparticles. Molecules 2021, 26, 3437. [Google Scholar] [CrossRef]
  21. Ulbrich, K.; Holá, K.; Šubr, V.; Bakandritsos, A.; Tuček, J.; Zbořil, R. Targeted Drug Delivery with Polymers and Magnetic Nanoparticles: Covalent and Noncovalent Approaches, Release Control, and Clinical Studies. Chem. Rev. 2016, 116, 5338–5431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Bobrikova, E.; Chubarov, A.; Dmitrienko, E. The Effect of pH and Buffer on Oligonucleotide Affinity for Iron Oxide Nanoparticles. Magnetochemistry 2021, 7, 128. [Google Scholar] [CrossRef]
  23. Obaidat, I.M.; Narayanaswamy, V.; Alaabed, S.; Sambasivam, S.; Muralee Gopi, C.V.V. Principles of Magnetic Hyperthermia: A Focus on Using Multifunctional Hybrid Magnetic Nanoparticles. Magnetochemistry 2019, 5, 67. [Google Scholar] [CrossRef] [Green Version]
  24. Jiao, W.; Zhang, T.; Peng, M.; Yi, J.; He, Y.; Fan, H. Design of Magnetic Nanoplatforms for Cancer Theranostics. Biosensors 2022, 12, 38. [Google Scholar] [CrossRef]
  25. Schneider, M.G.M.; Martín, M.J.; Otarola, J.; Vakarelska, E.; Simeonov, V.; Lassalle, V.; Nedyalkova, M. Biomedical Applications of Iron Oxide Nanoparticles: Current Insights Progress and Perspectives. Pharmaceutics 2022, 14, 204. [Google Scholar] [CrossRef]
  26. Tran, H.; Ngo, N.M.; Medhi, R.; Srinoi, P.; Liu, T.; Rittikulsittichai, S.; Lee, T.R. Multifunctional Iron Oxide Magnetic Nanoparticles for Biomedical Applications: A Review. Materials 2022, 15, 503. [Google Scholar] [CrossRef]
  27. Caspani, S.; Magalhães, R.; Araújo, J.P.; Sousa, C.T. Magnetic nanomaterials as contrast agents for MRI. Materials 2020, 13, 2586. [Google Scholar] [CrossRef]
  28. Kostevšek, N. A review on the optimal design of magnetic nanoparticle-based t2 mri contrast agents. Magnetochemistry 2020, 6, 11. [Google Scholar] [CrossRef] [Green Version]
  29. Fernández-Barahona, I.; Muñoz-Hernando, M.; Ruiz-Cabello, J.; Herranz, F.; Pellico, J. Iron oxide nanoparticles: An alternative for positive contrast in magnetic resonance imaging. Inorganics 2020, 8, 28. [Google Scholar] [CrossRef] [Green Version]
  30. Katz, E. Magnetic Nanoparticles. Magnetochemistry 2020, 6, 6. [Google Scholar] [CrossRef]
  31. Berensmeier, S. Magnetic particles for the separation and purification of nucleic acids. Appl. Microbiol. Biotechnol. 2006, 73, 495–504. [Google Scholar] [CrossRef]
  32. Li, P.; Li, M.; Yue, D.; Chen, H. Solid-phase extraction methods for nucleic acid separation. A review. J. Sep. Sci. 2022, 45, 172–184. [Google Scholar] [CrossRef]
  33. Tang, C.; He, Z.; Liu, H.; Xu, Y.; Huang, H.; Yang, G.; Xiao, Z.; Li, S.; Liu, H.; Deng, Y.; et al. Application of magnetic nanoparticles in nucleic acid detection. J. Nanobiotechnol. 2020, 18, 62. [Google Scholar] [CrossRef] [Green Version]
  34. Xu, S.; Lee, T.R. Fe3O4 Nanoparticles: Structures, Synthesis, Magnetic Properties, Surface Functionalization, and Emerging Applications. Appl. Sci. 2021, 11, 11301. [Google Scholar]
  35. Hosu, O.; Tertis, M.; Cristea, C. Implication of magnetic nanoparticles in cancer detection, screening and treatment. Magnetochemistry 2019, 5, 55. [Google Scholar] [CrossRef] [Green Version]
  36. Akbarzadeh, A.; Samiei, M.; Davaran, S. Magnetic nanoparticles: Preparation, physical properties, and applications in biomedicine. Nanoscale Res. Lett. 2012, 7, 144. [Google Scholar] [CrossRef] [Green Version]
  37. Caizer, C. Nanoparticle Size Effect on Some Magnetic Properties. In Handbook of Nanoparticles; Springer: Cham, Switzerland, 2016; pp. 1–1426. ISBN 9783319153384. [Google Scholar]
  38. Li, Q.; Kartikowati, C.W.; Horie, S.; Ogi, T.; Iwaki, T.; Okuyama, K. Correlation between particle size/domain structure and magnetic properties of highly crystalline Fe3O4 nanoparticles. Sci. Rep. 2017, 7, 9894. [Google Scholar] [CrossRef]
  39. Kolhatkar, A.G.; Jamison, A.C.; Litvinov, D.; Willson, R.C.; Lee, T.R. Tuning the Magnetic Properties of Nanoparticles. Int. J. Mol. Sci. 2013, 14, 15977–16009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Koksharov, Y.A. Magnetism of Nanoparticles: Effects of Size, Shape, and Interactions. In Magnetic Nanoparticles; Wiley-VCH: Hoboken, NJ, USA, 2009; pp. 197–254. ISBN 9783527407903. [Google Scholar]
  41. Lee, J.S.; Cha, J.M.; Yoon, H.Y.; Lee, J.K.; Kim, Y.K. Magnetic multi-granule nanoclusters: A model system that exhibits universal size effect of magnetic coercivity. Sci. Rep. 2015, 5, 12135. [Google Scholar] [CrossRef] [Green Version]
  42. Frenkel, J.; Doefman, J. Spontaneous and Induced Magnetisation in Ferromagnetic Bodies. Nature 1930, 126, 274–275. [Google Scholar] [CrossRef]
  43. Xu, H.; Li, S.; Liu, Y. Nanoparticles in the diagnosis and treatment of vascular aging and related diseases. Signal Transduct. Target. Ther. 2022, 7, 231. [Google Scholar] [CrossRef] [PubMed]
  44. Pucci, C.; Degl’Innocenti, A.; Belenli Gümüş, M.; Ciofani, G. Superparamagnetic iron oxide nanoparticles for magnetic hyperthermia: Recent advancements, molecular effects, and future directions in the omics era. Biomater. Sci. 2022, 10, 2103–2121. [Google Scholar] [CrossRef]
  45. Ramin, N.A.; Ramachandran, M.R.; Saleh, N.M.; Mat Ali, Z.M.; Asman, S. Magnetic Nanoparticles Molecularly Imprinted Polymers: A Review. Curr. Nanosci. 2022, 18, 1–29. [Google Scholar] [CrossRef]
  46. Harish, V.; Tewari, D.; Gaur, M.; Yadav, A.B.; Swaroop, S.; Bechelany, M.; Barhoum, A. Review on Nanoparticles and Nanostructured Materials: Bioimaging, Biosensing, Drug Delivery, Tissue Engineering, Antimicrobial, and Agro-Food Applications. Nanomaterials 2022, 12, 457. [Google Scholar] [CrossRef]
  47. Footer, C. Tuneable Magnetic Nanocomposites for Remote self-healing. Sci. Rep. 2022, 12, 10180. [Google Scholar]
  48. Koksharov, Y.A.; Gubin, S.P.; Taranov, I.V.; Khomutov, G.B.; Gulyaev, Y.V. Magnetic Nanoparticles in Medicine: Progress, Problems, and Advances. J. Commun. Technol. Electron. 2022, 67, 101–116. [Google Scholar] [CrossRef]
  49. Geppert, M.; Himly, M. Iron Oxide Nanoparticles in Bioimaging—An Immune Perspective. Front. Immunol. 2021, 12, 688927. [Google Scholar] [CrossRef]
  50. Malhotra, N.; Lee, J.S.; Liman, R.A.D.; Ruallo, J.M.S.; Villaflore, O.B.; Ger, T.R.; Hsiao, C. Der Potential toxicity of iron oxide magnetic nanoparticles: A review. Molecules 2020, 25, 3159. [Google Scholar] [CrossRef]
  51. Abakumov, M.A.; Semkina, A.S.; Skorikov, A.S.; Vishnevskiy, D.A.; Ivanova, A.V.; Mironova, E.; Davydova, G.A.; Majouga, A.G.; Chekhonin, V.P. Toxicity of iron oxide nanoparticles: Size and coating effects. J. Biochem. Mol. Toxicol. 2018, 32, e22225. [Google Scholar] [CrossRef]
  52. Kim, J.E.; Shin, J.Y.; Cho, M.H. Magnetic nanoparticles: An update of application for drug delivery and possible toxic effects. Arch. Toxicol. 2012, 86, 685–700. [Google Scholar] [CrossRef]
  53. Seeney, C.E. The emerging applications of magnetic nanovectors in nanomedicine. Pharm. Pat. Anal. 2015, 4, 285–304. [Google Scholar] [CrossRef] [PubMed]
  54. Chrishtop, V.V.; Mironov, V.A.; Prilepskii, A.Y.; Nikonorova, V.G.; Vinogradov, V.V. Organ-specific toxicity of magnetic iron oxide-based nanoparticles. Nanotoxicology 2021, 15, 167–204. [Google Scholar] [CrossRef] [PubMed]
  55. Batlle, X.; Moya, C.; Escoda-Torroella, M.; Iglesias, Ò.; Fraile Rodríguez, A.; Labarta, A. Magnetic nanoparticles: From the nanostructure to the physical properties. J. Magn. Magn. Mater. 2022, 543, 168594. [Google Scholar] [CrossRef]
  56. Chircov, C.; Vasile, B.S. New Approaches in Synthesis and Characterization Methods of Iron Oxide Nanoparticles. In Iron Oxide Nanoparticles; IntechOpen: London, UK, 2022. [Google Scholar]
  57. Ali, A.; Shah, T.; Ullah, R.; Zhou, P.; Guo, M.; Ovais, M.; Tan, Z.; Rui, Y.K. Review on Recent Progress in Magnetic Nanoparticles: Synthesis, Characterization, and Diverse Applications. Front. Chem. 2021, 9, 629054. [Google Scholar] [CrossRef] [PubMed]
  58. Vargas, G.; Cypriano, J.; Correa, T.; Leao, P.; Bazylinski, D.; Abreu, F. Applications of Magnetotactic Bacteria, Magnetosomes and Magnetosome Crystals in Biotechnology and Nanotechnology: Mini-Review. Molecules 2018, 23, 2438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Canaparo, R.; Foglietta, F.; Limongi, T.; Serpe, L. Biomedical applications of reactive oxygen species generation by metal nanoparticles. Materials 2021, 14, 53. [Google Scholar] [CrossRef] [PubMed]
  60. Nelson, N.; Port, J.; Pandey, M. Use of Superparamagnetic Iron Oxide Nanoparticles (SPIONs) via Multiple Imaging Modalities and Modifications to Reduce Cytotoxicity: An Educational Review. J. Nanotheranostics 2020, 1, 105–135. [Google Scholar] [CrossRef]
  61. Zelepukin, I.V.; Yaremenko, A.V.; Ivanov, I.N.; Yuryev, M.V.; Cherkasov, V.R.; Deyev, S.M.; Nikitin, P.I.; Nikitin, M.P. Long-Term Fate of Magnetic Particles in Mice: A Comprehensive Study. ACS Nano 2021, 15, 11341–11357. [Google Scholar] [CrossRef]
  62. Khramtsov, P.; Barkina, I.; Kropaneva, M.; Bochkova, M.; Timganova, V.; Nechaev, A.; Byzov, I.; Zamorina, S.; Yermakov, A.; Rayev, M. Magnetic nanoclusters coated with albumin, casein, and gelatin: Size tuning, relaxivity, stability, protein corona, and application in nuclear magnetic resonance immunoassay. Nanomaterials 2019, 9, 1345. [Google Scholar] [CrossRef] [Green Version]
  63. Schwaminger, S.P.; Blank-Shim, S.A.; Scheifele, I.; Pipich, V.; Fraga-García, P.; Berensmeier, S. Design of Interactions Between Nanomaterials and Proteins: A Highly Affine Peptide Tag to Bare Iron Oxide Nanoparticles for Magnetic Protein Separation. Biotechnol. J. 2019, 14, 55. [Google Scholar] [CrossRef]
  64. Mylkie, K.; Nowak, P.; Rybczynski, P.; Ziegler-Borowska, M. Polymer-coated magnetite nanoparticles for protein immobilization. Materials 2021, 14, 248. [Google Scholar] [CrossRef] [PubMed]
  65. Xue, J.; Li, X.; Li, Q.; Lyu, J.; Wang, W.; Zhuang, L.; Xu, Y. Magnetic drug-loaded osteoinductive Fe3O4/CaCO3 hybrid microspheres system: Efficient for sustained release of antibiotics. J. Phys. D. Appl. Phys. 2020, 53, 245401. [Google Scholar] [CrossRef]
  66. Begines, B.; Ortiz, T.; Pérez-Aranda, M.; Martínez, G.; Merinero, M.; Argüelles-Arias, F.; Alcudia, A. Polymeric nanoparticles for drug delivery: Recent developments and future prospects. Nanomaterials 2020, 10, 1403. [Google Scholar] [CrossRef] [PubMed]
  67. Turrina, C.; Oppelt, A.; Mitzkus, M.; Berensmeier, S.; Schwaminger, S.P. Silica-coated superparamagnetic iron oxide nanoparticles: New insights into the influence of coating thickness on the particle properties and lasioglossin binding. MRS Commun. 2022, 12, 632–639. [Google Scholar] [CrossRef]
  68. Schwaminger, S.P.; Blank-Shim, S.A.; Scheifele, I.; Fraga-García, P.; Berensmeier, S. Peptide binding to metal oxide nanoparticles. Faraday Discuss. 2017, 204, 233–250. [Google Scholar] [CrossRef]
  69. Tarkistani, M.A.M.; Komalla, V.; Kayser, V. Recent advances in the use of iron–gold hybrid nanoparticles for biomedical applications. Nanomaterials 2021, 11, 1227. [Google Scholar] [CrossRef]
  70. Schwaminger, S.P.; Bauer, D.; Fraga-García, P. Gold-iron oxide nanohybrids: Insights into colloidal stability and surface-enhanced Raman detection. Nanoscale Adv. 2021, 3, 6438–6445. [Google Scholar] [CrossRef]
  71. Zaloga, J.; Feoktystov, A.; Garamus, V.M.; Karawacka, W.; Ioffe, A.; Brückel, T.; Tietze, R.; Alexiou, C.; Lyer, S. Studies on the Adsorption and Desorption of Mitoxantrone to Lauric Acid/Albumin Coated Iron Oxide Nanoparticles. Colloids Surf. B Biointerfaces 2018, 161, 18–26. [Google Scholar] [CrossRef]
  72. Vismara, E.; Bongio, C.; Coletti, A.; Edelman, R.; Serafini, A.; Mauri, M.; Simonutti, R.; Bertini, S.; Urso, E.; Assaraf, Y.G.; et al. Albumin and hyaluronic acid-coated superparamagnetic iron oxide nanoparticles loaded with paclitaxel for biomedical applications. Molecules 2017, 22, 1030. [Google Scholar] [CrossRef] [Green Version]
  73. Zaloga, J.; Pöttler, M.; Leitinger, G.; Friedrich, R.P.; Almer, G.; Lyer, S.; Baum, E.; Tietze, R.; Heimke-Brinck, R.; Mangge, H.; et al. Pharmaceutical formulation of HSA hybrid coated iron oxide nanoparticles for magnetic drug targeting. Eur. J. Pharm. Biopharm. 2016, 101, 152–162. [Google Scholar] [CrossRef]
  74. Zaloga, J.; Stapf, M.; Nowak, J.; Pöttler, M.; Friedrich, R.P.; Tietze, R.; Lyer, S.; Lee, G.; Odenbach, S.; Hilger, I.; et al. Tangential flow ultrafiltration allows purification and concentration of lauric acid-/albumin-coated particles for improved magnetic treatment. Int. J. Mol. Sci. 2015, 16, 19291–19307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Zaloga, J.; Janko, C.; Nowak, J.; Matuszak, J.; Knaup, S.; Eberbeck, D.; Tietze, R.; Unterweger, H.; Friedrich, R.P.; Duerr, S.; et al. Development of a lauric acid/albumin hybrid iron oxide nanoparticle system with improved biocompatibility. Int. J. Nanomed. 2014, 9, 4847–4866. [Google Scholar] [CrossRef] [PubMed]
  76. Corem-Salkmon, E.; Ram, Z.; Daniels, D.; Perlstein, B.; Last, D.; Salomon, S.; Tamar, G.; Shneor, R.; Guez, D.; Margel, S.; et al. Convection-enhanced delivery of methotrexate-loaded maghemite nanoparticles. Int. J. Nanomed. 2011, 6, 1595–1602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Zhou, L.; Ye, L.; Lu, Y. Flexible and Effective Preparation of Magnetic Nanoclusters via One-Step Flow Synthesis. Nanomaterials 2022, 12, 350. [Google Scholar] [CrossRef]
  78. Mukhopadhyay, A.; Joshi, N.; Chattopadhyay, K.; De, G. A facile synthesis of PEG-coated magnetite (Fe3O4) nanoparticles and their prevention of the reduction of cytochrome C. ACS Appl. Mater. Interfaces 2012, 4, 142–149. [Google Scholar] [CrossRef]
  79. Huang, Y.; Zhang, B.; Xie, S.; Yang, B.; Xu, Q.; Tan, J. Superparamagnetic Iron Oxide Nanoparticles Modified with Tween 80 Pass through the Intact Blood-Brain Barrier in Rats under Magnetic Field. ACS Appl. Mater. Interfaces 2016, 8, 11336–11341. [Google Scholar] [CrossRef]
  80. Yoon, H.M.; Kang, M.S.; Choi, G.E.; Kim, Y.J.; Bae, C.H.; Yu, Y.B.; Jeong, Y. Il Stimuli-responsive drug delivery of doxorubicin using magnetic nanoparticle conjugated poly(Ethylene glycol)-g-chitosan copolymer. Int. J. Mol. Sci. 2021, 22, 13169. [Google Scholar] [CrossRef]
  81. Shete, P.B.; Patil, R.M.; Tiwale, B.M.; Pawar, S.H. Water dispersible oleic acid-coated Fe3O4 nanoparticles for biomedical applications. J. Magn. Magn. Mater. 2015, 377, 406–410. [Google Scholar] [CrossRef]
  82. Junejo, Y.; Baykal, A.; Sözeri, H. Simple hydrothermal synthesis of Fe3O4-PEG nanocomposite. Cent. Eur. J. Chem. 2013, 11, 1527–1532. [Google Scholar] [CrossRef]
  83. Snoderly, H.T.; Freshwater, K.A.; Martinez de la Torre, C.; Panchal, D.M.; Vito, J.N.; Bennewitz, M.F. PEGylation of Metal Oxide Nanoparticles Modulates Neutrophil Extracellular Trap Formation. Biosensors 2022, 12, 123. [Google Scholar] [CrossRef]
  84. Yallapu, M.M.; Foy, S.P.; Jain, T.K.; Labhasetwar, V. PEG-functionalized magnetic nanoparticles for drug delivery and magnetic resonance imaging applications. Pharm. Res. 2010, 27, 2283–2295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Mahdavi, M.; Ahmad, M.B.; Haron, M.J.; Namvar, F.; Nadi, B.; Ab Rahman, M.Z.; Amin, J. Synthesis, surface modification and characterisation of biocompatible magnetic iron oxide nanoparticles for biomedical applications. Molecules 2013, 18, 7533–7548. [Google Scholar] [CrossRef] [PubMed]
  86. Vavaev, E.S.; Novoselova, M.; Shchelkunov, N.M.; German, S.; Aleksei, S.; Mokrousov, M.D.; Zelepukin, I.V.; Burov, A.M.; Khlebtsov, B.N.; Lyubin, E.V.; et al. CaCO3 Nanoparticles Coated with Alternating Layers of Poly-L-Arginine Hydrochloride and Fe3O4 Nanoparticles as Navigable Drug Carriers and Hyperthermia Agents. ACS Appl. Nano Mater. 2022, 5, 2994–3006. [Google Scholar] [CrossRef]
  87. Zelepukin, I.V.; Shipunova, V.O.; Mirkasymov, A.B.; Nikitin, P.I.; Nikitin, M.P. Synthesis and Characterization of Hybrid Core-Shell Fe3O4/SiO2 Nanoparticles for Biomedical Applications. Acta Naturae 2017, 9, 58–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Ayub, A.; Wettig, S. An Overview of Nanotechnologies for Drug Delivery to the Brain. Pharmaceutics 2022, 14, 224. [Google Scholar] [CrossRef] [PubMed]
  89. Samanta, B.; Yan, H.; Fischer, N.O.; Shi, J.; Jerry, D.J.; Rotello, V.M. Protein-passivated Fe3O4 nanoparticles: Low toxicity and rapid heating for thermal therapy. J. Mater. Chem. 2008, 18, 1204–1208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Bychkova, A.V.; Sorokina, O.N.; Pronkin, P.G.; Tatikolov, A.S.; Kovarski, A.L.; Rosenfeld, M.A. Protein-Coated Magnetic Nanoparticles: Creation and Investigation. Proc. Int. Conf. Nanomater. Appl. Prop. 2013, 2, 1–5. [Google Scholar]
  91. Sakulkhu, U.; Mahmoudi, M.; Maurizi, L.; Salaklang, J.; Hofmann, H. Protein corona composition of superparamagnetic iron oxide nanoparticles with various physico-Chemical properties and coatings. Sci. Rep. 2014, 4, 5020. [Google Scholar] [CrossRef] [Green Version]
  92. Chuang, V.T.G.; Maruyama, T.; Otagiri, M. Human Serum Albumin in Blood Detoxification Treatment. In Albumin in Medicine; Springer: Singapore, 2016; pp. 209–225. [Google Scholar]
  93. Kragh-hansen, U. Human Serum Albumin: A Multifunctional Protein. In Albumine in Medicine; Springer: Singapore, 2016; pp. 1–24. ISBN 978-981-10-2115-2. [Google Scholar]
  94. Fanali, G.; di Masi, A.; Trezza, V.; Marino, M.; Fasano, M.; Ascenzi, P. Human serum albumin: From bench to bedside. Mol. Aspects Med. 2012, 33, 209–290. [Google Scholar] [CrossRef]
  95. Li, H.; Wang, Y.; Tang, Q.; Yin, D.; Tang, C.; He, E.; Zou, L.; Peng, Q. The Protein Corona and its Effects on Nanoparticle-Based Drug Delivery Systems. Acta Biomater. 2021, 129, 57–72. [Google Scholar] [CrossRef]
  96. Mazario, E.; Forget, A.; Belkahla, H.; Lomas, J.S.; Decorse, P.; Chevillot-Biraud, A.; Verbeke, P.; Wilhelm, C.; Ammar, S.; El Hage Chahine, J.M.; et al. Functionalization of Iron Oxide Nanoparticles With HSA Protein for Thermal Therapy. IEEE Trans. Magn. 2017, 53, 1–5. [Google Scholar] [CrossRef]
  97. Baki, A.; Remmo, A.; Löwa, N.; Wiekhorst, F.; Bleul, R. Albumin-coated single-core iron oxide nanoparticles for enhanced molecular magnetic imaging (Mri/mpi). Int. J. Mol. Sci. 2021, 22, 6235. [Google Scholar] [CrossRef] [PubMed]
  98. Rahdar, S.; Rahdar, A.; Ahmadi, S.; Trant, J.F. Adsorption of bovine serum albumin (BSA) by bare magnetite nanoparticles with surface oxidative impurities that prevent aggregation. Can. J. Chem. 2019, 97, 577–583. [Google Scholar] [CrossRef]
  99. Aires, A.; Ocampo, S.M.; Cabrera, D.; La Cueva, L.D.; Salas, G.; Teran, F.J.; Cortajarena, A.L. BSA-coated magnetic nanoparticles for improved therapeutic properties. J. Mater. Chem. B 2015, 3, 6239–6247. [Google Scholar] [CrossRef]
  100. Ziegler-Borowska, M. Magnetic nanoparticles coated with aminated starch for HSA immobilization- simple and fast polymer surface functionalization. Int. J. Biol. Macromol. 2019, 136, 106–114. [Google Scholar] [CrossRef]
  101. Yu, S.M.; Laromaine, A.; Roig, A. Enhanced stability of superparamagnetic iron oxide nanoparticles in biological media using a pH adjusted-BSA adsorption protocol. J. Nanoparticle Res. 2014, 16, 2484. [Google Scholar] [CrossRef] [Green Version]
  102. Gonzalez-Moragas, L.; Yu, S.M.; Carenza, E.; Laromaine, A.; Roig, A. Protective Effects of Bovine Serum Albumin on Superparamagnetic Iron Oxide Nanoparticles Evaluated in the Nematode Caenorhabditis elegans. ACS Biomater. Sci. Eng. 2015, 1, 1129–1138. [Google Scholar] [CrossRef] [Green Version]
  103. Moya, C.; Escudero, R.; Malaspina, D.C.; De La Mata, M.; Hernández-Saz, J.; Faraudo, J.; Roig, A. Insights into Preformed Human Serum Albumin Corona on Iron Oxide Nanoparticles: Structure, Effect of Particle Size, Impact on MRI Efficiency, and Metabolization. ACS Appl. Bio Mater. 2019, 2, 3084–3094. [Google Scholar] [CrossRef]
  104. Mariam, J.; Sivakami, S.; Dongre, P.M. Albumin corona on nanoparticles–a strategic approach in drug delivery. Drug Deliv. 2016, 23, 2668–2676. [Google Scholar] [CrossRef] [Green Version]
  105. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
  106. Präbst, K.; Engelhardt, H.; Ringgeler, S.; Hübner, H. Basic Colorimetric Proliferation Assays: MTT, WST, and Resazurin. In Cell Viability Assays; Methods in Molecular Biology book series; Humana Press: New York, NY, USA, 2017; pp. 1–17. [Google Scholar]
  107. Bondarenko, O.; Mortimer, M.; Kahru, A.; Feliu, N.; Javed, I.; Kakinen, A.; Lin, S.; Xia, T.; Song, Y.; Davis, T.P.; et al. Nanotoxicology and nanomedicine: The Yin and Yang of nano-bio interactions for the new decade. Nano Today 2021, 39, 101184. [Google Scholar] [CrossRef]
  108. Lin, S.; Yu, T.; Yu, Z.; Hu, X.; Yin, D. Nanomaterials Safer-by-Design: An Environmental Safety Perspective. Adv. Mater. 2018, 30, 1705691. [Google Scholar] [CrossRef] [PubMed]
  109. Kenchegowda, M.; Rahamathulla, M.; Hani, U.; Begum, M.Y.; Guruswamy, S.; Osmani, R.A.M.; Gowrav, M.P.; Alshehri, S.; Ghoneim, M.M.; Alshlowi, A.; et al. Smart Nanocarriers as an Emerging Platform for Cancer Therapy: A Review. Molecules 2022, 27, 146. [Google Scholar] [CrossRef]
  110. Vangijzegem, T.; Stanicki, D.; Laurent, S.; Vangijzegem, T. Expert Opinion on Drug Delivery Magnetic iron oxide nanoparticles for drug delivery: Applications and characteristics Magnetic iron oxide nanoparticles for drug delivery: Applications and characteristics. Expert Opin. Drug Deliv. 2019, 16, 69–78. [Google Scholar] [CrossRef] [PubMed]
  111. Leach, J.C.; Wang, A.; Ye, K.; Jin, S. A RNA-DNA hybrid aptamer for nanoparticle-based prostate tumor targeted drug delivery. Int. J. Mol. Sci. 2016, 17, 380. [Google Scholar] [CrossRef] [Green Version]
  112. Taghavi Pourianazar, N.; Gunduz, U. CpG oligodeoxynucleotide-loaded PAMAM dendrimer-coated magnetic nanoparticles promote apoptosis in breast cancer cells. Biomed. Pharmacother. 2016, 78, 81–91. [Google Scholar] [CrossRef] [PubMed]
  113. Bassetto, M.; Sen, M.; Poulhes, F.; Arango-Gonzalez, B.; Bonvin, E.; Sapet, C.; Ueffing, M.; Zelphati, O. New Method for Efficient siRNA Delivery in Retina Explants: Reverse Magnetofection. Bioconjug. Chem. 2021, 32, 1078–1093. [Google Scholar] [CrossRef]
  114. Laurent, N.; Sapet, C.; Gourrierec, L.L.; Bertosio, E.; Zelphati, O. Nucleic acid delivery using magnetic nanoparticles: The Magnetofection TM technology. Ther. Deliv. 2011, 2, 471–482. [Google Scholar] [CrossRef]
  115. Gozuacik, D.; Akkoc, Y.; Kosar, A.; Dogan-ekici, A.I.; Ekici, S. Anticancer Use of Nanoparticles as Nucleic Acid Carriers. J. Biomed. Nanotechnol. 2014, 10, 1751–1783. [Google Scholar] [CrossRef]
  116. Cen, C.; Wu, J.; Zhang, Y.; Luo, C.; Xie, L.; Zhang, X.; Yang, X.; Li, M.; Bi, Y.; Li, T.; et al. Improving Magnetofection of Magnetic Polyethylenimine Nanoparticles into MG-63 Osteoblasts Using a Novel Uniform Magnetic Field. Nanoscale Res. Lett. 2019, 14, 90. [Google Scholar] [CrossRef]
  117. Zuvin, M.; Kuruoglu, E.; Kaya, V.O.; Unal, O.; Kutlu, O.; Yagci Acar, H.; Gozuacik, D.; Kosar, A. Magnetofection of green fluorescent protein encoding DNA-bearing polyethyleneimine-coated superparamagnetic iron oxide nanoparticles to human breast cancer cells. ACS Omega 2019, 4, 12366–12374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Wang, R.; Hu, Y.; Zhao, N.; Xu, F.J. Well-Defined Peapod-like Magnetic Nanoparticles and Their Controlled Modification for Effective Imaging Guided Gene Therapy. ACS Appl. Mater. Interfaces 2016, 8, 11298–11308. [Google Scholar] [CrossRef] [PubMed]
  119. Huang, R.Y.; Chiang, P.H.; Hsiao, W.C.; Chuang, C.C.; Chang, C.W. Redox-Sensitive Polymer/SPIO Nanocomplexes for Efficient Magnetofection and MR Imaging of Human Cancer Cells. Langmuir 2015, 31, 6523–6531. [Google Scholar] [CrossRef] [PubMed]
  120. Stephen, Z.R.; Dayringer, C.J.; Lim, J.J.; Revia, R.A.; Halbert, M.V.; Jeon, M.; Bakthavatsalam, A.; Ellenbogen, R.G.; Zhang, M. Approach to Rapid Synthesis and Functionalization of Iron Oxide Nanoparticles for High Gene Transfection. ACS Appl. Mater. Interfaces 2016, 8, 6320–6328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Cui, Y.; Li, X.; Zeljic, K.; Shan, S.; Qiu, Z.; Wang, Z. Effect of PEGylated Magnetic PLGA-PEI Nanoparticles on Primary Hippocampal Neurons: Reduced Nanoneurotoxicity and Enhanced Transfection Efficiency with Magnetofection. ACS Appl. Mater. Interfaces 2019, 11, 38190–38204. [Google Scholar] [CrossRef]
  122. Kievit, F.M.; Veiseh, O.; Fang, C.; Bhattarai, N.; Lee, D.; Ellenbogen, R.G.; Zhang, M. Chlorotoxin labeled magnetic nanovectors for targeted gene delivery to glioma. ACS Nano 2010, 4, 4587–4594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Lo, Y.L.; Chou, H.L.; Liao, Z.X.; Huang, S.J.; Ke, J.H.; Liu, Y.S.; Chiu, C.C.; Wang, L.F. Chondroitin sulfate-polyethylenimine copolymer-coated superparamagnetic iron oxide nanoparticles as an efficient magneto-gene carrier for microRNA-encoding plasmid DNA delivery. Nanoscale 2015, 7, 8554–8565. [Google Scholar] [CrossRef]
  124. Xie, L.; Jiang, Q.; He, Y.; Nie, Y.; Yue, D.; Gu, Z. Insight into the efficient transfection activity of a designed low aggregated magnetic polyethyleneimine/DNA complex in serum-containing medium and the application in vivo. Biomater. Sci. 2015, 3, 446–456. [Google Scholar] [CrossRef]
  125. Tian, G.; Zhang, X.; Gu, Z.; Zhao, Y. Recent Advances in Upconversion Nanoparticles-Based Multifunctional Nanocomposites for Combined Cancer Therapy. Adv. Mater. 2015, 27, 7692–7712. [Google Scholar] [CrossRef]
  126. Sritharan, S.; Sivalingam, N. A comprehensive review on time-tested anticancer drug doxorubicin. Life Sci. 2021, 278, 119527. [Google Scholar] [CrossRef]
  127. Christidi, E.; Brunham, L.R. Regulated cell death pathways in doxorubicin-induced cardiotoxicity. Cell Death Dis. 2021, 12, 339. [Google Scholar] [CrossRef] [PubMed]
  128. Popescu, R.C.; Savu, D.; Dorobantu, I.; Vasile, B.S.; Hosser, H.; Boldeiu, A.; Temelie, M.; Straticiuc, M.; Iancu, D.A.; Andronescu, E.; et al. Efficient uptake and retention of iron oxide-based nanoparticles in HeLa cells leads to an effective intracellular delivery of doxorubicin. Sci. Rep. 2020, 10, 10530. [Google Scholar] [CrossRef] [PubMed]
  129. Popescu, R.C.; Savu, D.I.; Bierbaum, M.; Grbenicek, A.; Schneider, F.; Hosser, H.; Vasile, B.Ș.; Andronescu, E.; Wenz, F.; Giordano, F.A.; et al. Intracellular delivery of doxorubicin by iron oxide-based nano-constructs increases clonogenic inactivation of ionizing radiation in hela cells. Int. J. Mol. Sci. 2021, 22, 6778. [Google Scholar] [CrossRef] [PubMed]
  130. Piehler, S.; Dähring, H.; Grandke, J.; Göring, J.; Couleaud, P.; Aires, A.; Cortajarena, A.L.; Courty, J.; Latorre, A.; Somoza, Á.; et al. Iron oxide nanoparticles as carriers for DOX and magnetic hyperthermia after intratumoral application into breast cancer in mice: Impact and future perspectives. Nanomaterials 2020, 10, 1016. [Google Scholar] [CrossRef]
  131. Norouzi, M.; Yathindranath, V.; Thliveris, J.A.; Kopec, B.M.; Siahaan, T.J.; Miller, D.W. Doxorubicin-loaded iron oxide nanoparticles for glioblastoma therapy: A combinational approach for enhanced delivery of nanoparticles. Sci. Rep. 2020, 10, 11292. [Google Scholar] [CrossRef]
  132. Khaledian, M.; Nourbakhsh, M.S.; Saber, R.; Hashemzadeh, H.; Darvishi, M.H. Preparation and evaluation of doxorubicin-loaded pla–peg–fa copolymer containing superparamagnetic iron oxide nanoparticles (Spions) for cancer treatment: Combination therapy with hyperthermia and chemotherapy. Int. J. Nanomed. 2020, 15, 6167–6182. [Google Scholar] [CrossRef]
  133. Shen, C.; Wang, X.; Zheng, Z.; Gao, C.; Chen, X.; Zhao, S.; Dai, Z. Doxorubicin and indocyanine green loaded superparamagnetic iron oxide nanoparticles with PEGylated phospholipid coating for magnetic resonance with fluorescence imaging and chemotherapy of glioma. Int. J. Nanomed. 2019, 14, 101–117. [Google Scholar] [CrossRef] [Green Version]
  134. Eslami, P.; Albino, M.; Scavone, F.; Chiellini, F.; Morelli, A.; Baldi, G.; Cappiello, L.; Doumett, S.; Lorenzi, G.; Ravagli, C.; et al. Smart Magnetic Nanocarriers for Multi-Stimuli On-Demand Drug Delivery. Nanomaterials 2022, 12, 303. [Google Scholar] [CrossRef]
  135. Nieciecka, D.; Celej, J.; Żuk, M.; Majkowska-pilip, A.; Żelechowska-Matysiak, K.; Lis, A.; Osial, M. Hybrid system for local drug delivery and magnetic hyperthermia based on spions loaded with doxorubicin and epirubicin. Pharmaceutics 2021, 13, 480. [Google Scholar] [CrossRef]
  136. Nogueira, J.; Soares, S.F.; Amorim, C.O.; Amaral, J.S.; Silva, C.; Martel, F.; Trindade, T.; Daniel-Da-Silva, A.L. Magnetic driven nanocarriers for pH-responsive doxorubicin release in cancer therapy. Molecules 2020, 25, 333. [Google Scholar] [CrossRef] [Green Version]
  137. Singh, N.; Sallem, F.; Mirjolet, C.; Nury, T.; Sahoo, S.K.; Millot, N.; Kumar, R. Polydopamine modified superparamagnetic iron oxide nanoparticles as multifunctional nanocarrier for targeted prostate cancer treatment. Nanomaterials 2019, 9, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Kovrigina, E.; Chubarov, A.; Dmitrienko, E. High Drug Capacity Doxorubicin-Loaded Iron Oxide Nanocomposites for Cancer Therapy. Magnetochemistry 2022, 8, 54. [Google Scholar] [CrossRef]
  139. Al-Musawi, S.; Albukhaty, S.; Al-Karagoly, H.; Almalki, F. Design and synthesis of multi-functional superparamagnetic core-gold shell coated with chitosan and folate nanoparticles for targeted antitumor therapy. Nanomaterials 2021, 11, 32. [Google Scholar] [CrossRef] [PubMed]
  140. Wang, P.; Xue, J.; Wu, S.; Pei, Y.; Xu, L.; Wang, Y. Cell-Friendly Isolation and pH-Sensitive Controllable Release of Circulating Tumor Cells by Fe3O4@CaCO3 Nanoplatform. Adv. Mater. Interfaces 2021, 8, 2101191. [Google Scholar] [CrossRef]
  141. Piñeiro, Y.; Gómez, M.G.; Alves, L.d.C.; Prieto, A.A.; Acevedo, P.G.; Gudiña, R.S.; Puig, J.; Teijeiro, C.; Vilar, S.Y.; Rivas, J. Hybrid nanostructured magnetite nanoparticles: From bio-detection and theragnostics to regenerative medicine. Magnetochemistry 2020, 6, 4. [Google Scholar] [CrossRef] [Green Version]
  142. Hassanin, I.; Elzoghby, A. Albumin-based nanoparticles: A promising strategy to overcome cancer drug resistance. Cancer Drug Resist. 2020, 3, 930–946. [Google Scholar] [CrossRef]
  143. Lamichhane, S.; Lee, S. Albumin nanoscience: Homing nanotechnology enabling targeted drug delivery and therapy. Arch. Pharm. Res. 2020, 43, 118–133. [Google Scholar] [CrossRef]
  144. Kudarha, R.R.; Sawant, K.K. Albumin based versatile multifunctional nanocarriers for cancer therapy: Fabrication, surface modification, multimodal therapeutics and imaging approaches. Mater. Sci. Eng. C 2017, 81, 607–626. [Google Scholar] [CrossRef]
  145. Chubarov, A.S.; Shakirov, M.M.; Koptyug, I.V.; Sagdeev, R.Z.; Knorre, D.G.; Godovikova, T.S. Synthesis and characterization of fluorinated homocysteine derivatives as potential molecular probes for 19F magnetic resonance spectroscopy and imaging. Bioorg. Med. Chem. Lett. 2011, 21, 4050–4053. [Google Scholar] [CrossRef]
  146. Chubarov, A.S.; Zakharova, O.D.; Koval, O.A.; Romaschenko, A.V.; Akulov, A.E.; Zavjalov, E.L.; Razumov, I.A.; Koptyug, I.V.; Knorre, D.G.; Godovikova, T.S. Design of protein homocystamides with enhanced tumor uptake properties for 19F magnetic resonance imaging. Bioorg. Med. Chem. 2015, 23, 6943–6954. [Google Scholar] [CrossRef]
  147. Dobrynin, S.; Kutseikin, S.; Morozov, D.; Krumkacheva, O.; Spitsyna, A.; Gatilov, Y.; Silnikov, V.; Angelovski, G.; Bowman, M.K.; Kirilyuk, I.; et al. Human Serum Albumin Labelled with Sterically-Hindered Nitroxides as Potential MRI Contrast Agents. Molecules 2020, 25, 1709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Lisitskiy, V.A.; Khan, H.; Popova, T.V.; Chubarov, A.S.; Zakharova, O.D.; Akulov, A.E.; Shevelev, O.B.; Zavjalov, E.L.; Koptyug, I.V.; Moshkin, M.P.; et al. Multifunctional human serum albumin-therapeutic nucleotide conjugate with redox and pH-sensitive drug release mechanism for cancer theranostics. Bioorganic Med. Chem. Lett. 2017, 27, 3925–3930. [Google Scholar] [CrossRef] [PubMed]
  149. Gavilán, H.; Simeonidis, K.; Myrovali, E.; Mazarío, E.; Chubykalo-Fesenko, O.; Chantrell, R.; Balcells, L.; Angelakeris, M.; Morales, M.P.; Serantes, D. How size, shape and assembly of magnetic nanoparticles give rise to different hyperthermia scenarios. Nanoscale 2021, 13, 15631–15646. [Google Scholar] [CrossRef] [PubMed]
  150. Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.Y.; Sood, A.K.; Hua, S. Advances and challenges of liposome assisted drug delivery. Front. Pharmacol. 2015, 6, 286. [Google Scholar] [CrossRef] [Green Version]
  151. Lu, Y.J.; Chuang, E.Y.; Cheng, Y.H.; Anilkumar, T.S.; Chen, H.A.; Chen, J.P. Thermosensitive magnetic liposomes for alternating magnetic field-inducible drug delivery in dual targeted brain tumor chemotherapy. Chem. Eng. J. 2019, 373, 720–733. [Google Scholar] [CrossRef]
  152. Le, T.A.; Bui, M.P.; Yoon, J. Theoretical analysis for wireless magnetothermal deep brain stimulation using commercial nanoparticles. Int. J. Mol. Sci. 2019, 20, 2873. [Google Scholar] [CrossRef] [Green Version]
  153. Munshi, R.; Qadri, S.M.; Zhang, Q.; Rubio, I.C.; del Pino, P.; Pralle, A. Magnetothermal genetic deep brain stimulation of motor behaviors in awake, freely moving mice. eLife 2017, 6, e27069. [Google Scholar] [CrossRef]
  154. Mleczko, J.; Defort, A.; Kozioł, J.J.; Nguyen, T.T.; Mirończyk, A.; Zapotoczny, B.; Nowak-Jary, J.; Gronczewska, E.; Marć, M.; Dudek, M.R. Limitation of tuning the antibody-antigen reaction by changing the value of pH and its consequence for hyperthermia. J. Biochem. 2016, 159, 421–427. [Google Scholar] [CrossRef] [Green Version]
  155. Liu, Z.; Liu, J.; Cui, X.; Wang, X.; Zhang, L.; Tang, P. Recent Advances on Magnetic Sensitive Hydrogels in Tissue Engineering. Front. Chem. 2020, 8, 124. [Google Scholar] [CrossRef]
  156. Peng, J.; Zhao, J.; Long, Y.; Xie, Y.; Nie, J.; Chen, L. Magnetic Materials in Promoting Bone Regeneration. Front. Mater. 2019, 6, 268. [Google Scholar] [CrossRef]
  157. Yun, H.M.; Ahn, S.J.; Park, K.R.; Kim, M.J.; Kim, J.J.; Jin, G.Z.; Kim, H.W.; Kim, E.C. Magnetic nanocomposite scaffolds combined with static magnetic field in the stimulation of osteoblastic differentiation and bone formation. Biomaterials 2016, 85, 88–98. [Google Scholar] [CrossRef] [PubMed]
  158. Huang, W.S.; Chu, I.M. Injectable polypeptide hydrogel/inorganic nanoparticle composites for bone tissue engineering. PLoS ONE 2019, 14, e0210285. [Google Scholar] [CrossRef] [PubMed]
  159. Shuai, C.; Yang, W.; He, C.; Peng, S.; Gao, C.; Yang, Y.; Qi, F.; Feng, P. A magnetic micro-environment in scaffolds for stimulating bone regeneration. Mater. Des. 2020, 185, 108275. [Google Scholar] [CrossRef]
  160. Pesqueira, T.; Costa-Almeida, R.; Mithieux, S.M.; Babo, P.S.; Franco, A.R.; Mendes, B.B.; Domingues, R.M.A.; Freitas, P.; Reis, R.L.; Gomes, M.E.; et al. Engineering magnetically responsive tropoelastin spongy-like hydrogels for soft tissue regeneration. J. Mater. Chem. B 2018, 6, 1066–1075. [Google Scholar] [CrossRef]
  161. Silva, E.D.; Babo, P.S.; Costa-Almeida, R.; Domingues, R.M.A.; Mendes, B.B.; Paz, E.; Freitas, P.; Rodrigues, M.T.; Granja, P.L.; Gomes, M.E. Multifunctional magnetic-responsive hydrogels to engineer tendon-to-bone interface. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 2375–2385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Santos, L. Magnetically Actuated Biomaterials and Prospects in Tendon Healing. Nanomedicine 2016, 11, 1107–1122. [Google Scholar] [CrossRef] [PubMed]
  163. Betsch, M.; Cristian, C.; Lin, Y.Y.; Blaeser, A.; Schöneberg, J.; Vogt, M.; Buhl, E.M.; Fischer, H.; Duarte Campos, D.F. Incorporating 4D into Bioprinting: Real-Time Magnetically Directed Collagen Fiber Alignment for Generating Complex Multilayered Tissues. Adv. Healthc. Mater. 2018, 7, 1800894. [Google Scholar] [CrossRef] [PubMed]
  164. Bonhome-Espinosa, A.B.; Campos, F.; Durand-Herrera, D.; Sánchez-López, J.D.; Schaub, S.; Durán, J.D.G.; Lopez-Lopez, M.T.; Carriel, V. In vitro characterization of a novel magnetic fibrin-agarose hydrogel for cartilage tissue engineering. J. Mech. Behav. Biomed. Mater. 2020, 104, 103619. [Google Scholar] [CrossRef]
  165. Bianchi, E.; Vigani, B.; Viseras, C.; Ferrari, F.; Rossi, S.; Sandri, G. Inorganic Nanomaterials in Tissue Engineering. Pharmaceutics 2022, 14, 1127. [Google Scholar] [CrossRef]
  166. Funnell, J.L.; Balouch, B.; Gilbert, R.J. Magnetic composite biomaterials for neural regeneration. Front. Bioeng. Biotechnol. 2019, 7, 179. [Google Scholar] [CrossRef]
  167. Johnson, C.D.L.; Ganguly, D.; Zuidema, J.M.; Cardinal, T.J.; Ziemba, A.M.; Kearns, K.R.; McCarthy, S.M.; Thompson, D.M.; Ramanath, G.; Borca-Tasciuc, D.A.; et al. Injectable, Magnetically Orienting Electrospun Fiber Conduits for Neuron Guidance. ACS Appl. Mater. Interfaces 2019, 11, 356–372. [Google Scholar] [CrossRef] [PubMed]
  168. Tay, A.; Sohrabi, A.; Poole, K.; Seidlits, S.; Di Carlo, D. A 3D Magnetic Hyaluronic Acid Hydrogel for Magnetomechanical Neuromodulation of Primary Dorsal Root Ganglion Neurons. Adv. Mater. 2018, 30, 1800927. [Google Scholar] [CrossRef]
  169. Rose, J.C.; Cámara-Torres, M.; Rahimi, K.; Köhler, J.; Möller, M.; De Laporte, L. Nerve Cells Decide to Orient inside an Injectable Hydrogel with Minimal Structural Guidance. Nano Lett. 2017, 17, 3782–3791. [Google Scholar] [CrossRef] [PubMed]
  170. Pavón, J.J.; Allain, J.P.; Verma, D.; Echeverry-Rendón, M.; Cooper, C.L.; Reece, L.M.; Shetty, A.R.; Tomar, V. In situ Study Unravels Bio-Nanomechanical Behavior in a Magnetic Bacterial Nano-cellulose (MBNC) Hydrogel for Neuro-Endovascular Reconstruction. Macromol. Biosci. 2019, 19, 1800225. [Google Scholar] [CrossRef] [PubMed]
  171. Yuan, M.; Wang, Y.; Qin, Y.X. Promoting neuroregeneration by applying dynamic magnetic fields to a novel nanomedicine: Superparamagnetic iron oxide (SPIO)-gold nanoparticles bounded with nerve growth factor (NGF). Nanomed. Nanotechnol. Biol. Med. 2018, 14, 1337–1347. [Google Scholar] [CrossRef]
  172. Pal, A.; Kumar, S.; Jain, S.; Nag, T.C.; Mathur, R. Neuroregenerative Effects of Electromagnetic Field and Magnetic Nanoparticles on Spinal Cord Injury in Rats. J. Nanosci. Nanotechnol. 2018, 18, 6756–6764. [Google Scholar] [CrossRef]
  173. Liu, T.; Wang, Y.; Lu, L.; Liu, Y. SPIONs mediated magnetic actuation promotes nerve regeneration by inducing and maintaining repair-supportive phenotypes in Schwann cells. J. Nanobiotechnol. 2022, 20, 159. [Google Scholar] [CrossRef]
  174. Wallyn, J.; Anton, N.; Vandamme, T.F. Synthesis, principles, and properties of magnetite nanoparticles for in vivo imaging applications—A review. Pharmaceutics 2019, 11, 601. [Google Scholar] [CrossRef] [Green Version]
  175. Ellis, C.M.; Pellico, J.; Davis, J.J. Magnetic Nanoparticles Supporting Bio-responsive T1/T2 Magnetic Resonance Imaging. Materials 2019, 12, 4096. [Google Scholar] [CrossRef] [Green Version]
  176. Bruno, F.; Granata, V.; Bellisari, F.C.; Sgalambro, F.; Tommasino, E.; Palumbo, P.; Arrigoni, F.; Cozzi, D.; Grassi, F.; Brunese, M.C.; et al. Advanced Magnetic Resonance Imaging (MRI) Techniques: Technical Principles and Applications in Nanomedicine. Cancers 2022, 14, 1626. [Google Scholar] [CrossRef]
  177. Xie, J.; Chen, K.; Huang, J.; Lee, S.; Wang, J.; Gao, J.; Li, X.; Chen, X. PET/NIRF/MRI triple functional iron oxide nanoparticles. Biomaterials 2010, 31, 3016–3022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Wang, X.; Tu, M.; Tian, B.; Yi, Y.; Wei, Z.Z.; Wei, F. Synthesis of tumor-targeted folate conjugated fluorescent magnetic albumin nanoparticles for enhanced intracellular dual-modal imaging into human brain tumor cells. Anal. Biochem. 2016, 512, 8–17. [Google Scholar] [CrossRef] [PubMed]
  179. Bowers, A.N.; Trujillo-Rodríguez, M.J.; Farooq, M.Q.; Anderson, J.L. Extraction of DNA with magnetic ionic liquids using in situ dispersive liquid–liquid microextraction. Anal. Bioanal. Chem. 2019, 411, 7375–7385. [Google Scholar] [CrossRef] [PubMed]
  180. Ma, Y.; Chen, T.; Iqbal, M.Z.; Yang, F.; Hampp, N.; Wu, A.; Luo, L. Applications of magnetic materials separation in biological nanomedicine. Electrophoresis 2019, 40, 2011–2028. [Google Scholar] [CrossRef]
  181. Marengo, A.; Cagliero, C.; Sgorbini, B.; Anderson, J.L.; Emaus, M.N.; Bicchi, C.; Bertea, C.M.; Rubiolo, P. Development of an innovative and sustainable one-step method for rapid plant DNA isolation for targeted PCR using magnetic ionic liquids. Plant Methods 2019, 15, 23. [Google Scholar] [CrossRef]
  182. Wang, L.; He, K.; Sadak, O.; Wang, X.; Wang, Q.; Xu, X. Visual detection of in vitro nucleic acid replication by submicro- and nano-sized materials. Biosens. Bioelectron. 2020, 169, 112602. [Google Scholar] [CrossRef]
  183. Sosa-Acosta, J.R.; Iriarte-Mesa, C.; Ortega, G.A.; Díaz-García, A.M. DNA–Iron Oxide Nanoparticles Conjugates: Functional Magnetic Nanoplatforms in Biomedical Applications. Top. Curr. Chem. 2020, 378, 13. [Google Scholar] [CrossRef]
  184. Gessner, I.; Fries, J.W.U.; Brune, V.; Mathur, S. Magnetic nanoparticle-based amplification of microRNA detection in body fluids for early disease diagnosis. J. Mater. Chem. B 2021, 9, 9–22. [Google Scholar] [CrossRef]
  185. Sosa-Acosta, J.R.; Silva, J.A.; Fernández-Izquierdo, L.; Díaz-Castañón, S.; Ortiz, M.; Zuaznabar-Gardona, J.C.; Díaz-García, A.M. Iron Oxide Nanoparticles (IONPs) with potential applications in plasmid DNA isolation. Colloids Surf. A Physicochem. Eng. Asp. 2018, 545, 167–178. [Google Scholar] [CrossRef]
  186. Vanyorek, L.; Ilosvai, Á.M.; Szőri-Dorogházi, E.; Váradi, C.; Kristály, F.; Prekob, Á.; Fiser, B.; Varga, T.; Kónya, Z.; Viskolcz, B. Synthesis of iron oxide nanoparticles for DNA purification. J. Dispers. Sci. Technol. 2021, 42, 693–700. [Google Scholar] [CrossRef]
  187. Wang, J.; Ali, Z.; Si, J.; Wang, N.; He, N.; Li, Z. Simultaneous extraction of DNA and RNA from hepatocellular carcinoma (Hep G2) based on silica-coated magnetic nanoparticles. J. Nanosci. Nanotechnol. 2017, 17, 802–806. [Google Scholar] [CrossRef] [PubMed]
  188. Danthanarayana, A.N.; Manatunga, D.C.; De Silva, R.M.; Chandrasekharan, N.V.; De Silva, K.M.N. Magnetofection and isolation of DNA using polyethyleneimine functionalized magnetic iron oxide nanoparticles. R. Soc. Open Sci. 2018, 5, 181369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Chacón-Torres, J.C.; Reinoso, C.; Navas-León, D.G.; Briceño, S.; González, G. Optimized and scalable synthesis of magnetic nanoparticles for RNA extraction in response to developing countries’ needs in the detection and control of SARS-CoV-2. Sci. Rep. 2020, 10, 19004. [Google Scholar] [CrossRef]
  190. Ali, T.H.; Mandal, A.M.; Heidelberg, T.; Hussen, R.S.D. Sugar based cationic magnetic core–shell silica nanoparticles for nucleic acid extraction. RSC Adv. 2022, 12, 13566–13579. [Google Scholar] [CrossRef]
  191. Bulgakova, A.; Chubarov, A.; Dmitrienko, E. Magnetic Nylon 6 Nanocomposites for the Microextraction of Nucleic Acids from Biological Samples. Magnetochemistry 2022, 8, 85. [Google Scholar] [CrossRef]
  192. Eivazzadeh-Keihan, R.; Bahreinizad, H.; Amiri, Z.; Aliabadi, H.A.M.; Salimi-Bani, M.; Nakisa, A.; Davoodi, F.; Tahmasebi, B.; Ahmadpour, F.; Radinekiyan, F.; et al. Functionalized magnetic nanoparticles for the separation and purification of proteins and peptides. TrAC-Trends Anal. Chem. 2021, 141, 116291. [Google Scholar] [CrossRef]
  193. Damavandi, F.; Wang, W.; Shen, W.Z.; Cetinel, S.; Jordan, T.; Jovel, J.; Montemagno, C.; Wong, G.K.S. Enrichment of low abundance DNA/RNA by oligonucleotide-clicked iron oxide nanoparticles. Sci. Rep. 2021, 11, 13053. [Google Scholar] [CrossRef]
  194. Jiang, S.; Hua, L.; Guo, Z.; Sun, L. One-pot green synthesis of doxorubicin loaded-silica nanoparticles for in vivo cancer therapy. Mater. Sci. Eng. C 2018, 90, 257–263. [Google Scholar] [CrossRef]
  195. Pinchon, E.; Leon, F.; Temurok, N.; Morvan, F.; Vasseur, J.J.; Clot, M.; Foulongne, V.; Cantaloube, J.F.; Perre, P.V.; Daynès, A.; et al. Rapid and specific DNA detection by magnetic field-enhanced agglutination assay. Talanta 2020, 219, 121344. [Google Scholar] [CrossRef]
  196. Yildiz, I. Applications of magnetic nanoparticles in biomedical separation and purification. Nanotechnol. Rev. 2016, 5, 331–340. [Google Scholar] [CrossRef]
  197. Haun, J.B.; Yoon, T.J.; Lee, H.; Weissleder, R. Magnetic nanoparticle biosensors. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2010, 2, 291–304. [Google Scholar] [CrossRef]
  198. Koh, I.; Josephson, L. Magnetic Nanoparticle Sensors. Sensors 2009, 9, 8130–8145. [Google Scholar] [CrossRef]
  199. Rocha-Santos, T.A.P. Sensors and biosensors based on magnetic nanoparticles. TrAC-Trends Anal. Chem. 2014, 62, 28–36. [Google Scholar] [CrossRef]
  200. Sayad, A.; Skafidas, E.; Kwan, P. Magneto-impedance biosensor sensitivity: Effect and enhancement. Sensors 2020, 20, 5213. [Google Scholar] [CrossRef]
  201. Magnetic, B.E.; Selective, N.E.; Morpholin--yl, A.; Evelin, S. A Convenient U-Shape Microreactor for Continuous Flow. Catalyst 2022, 12, 1065. [Google Scholar]
  202. Ender, F.; Weiser, D.; Poppe, L. Microfluidic Multiple Chamber Chip Reactor Filled with Enzyme-Coated Magnetic Nanoparticles. In Lab-on-a-Chip Fabrication and Application; IntechOpen: London, UK, 2016. [Google Scholar]
  203. Digigow, R.G.; Dechézelles, J.F.; Kaufmann, J.; Vanhecke, D.; Knapp, H.; Lattuada, M.; Rothen-Rutishauser, B.; Petri-Fink, A. Magnetic microreactors for efficient and reliable magnetic nanoparticle surface functionalization. Lab Chip 2014, 14, 2276–2286. [Google Scholar] [CrossRef] [Green Version]
  204. Gkantzou, E.; Patila, M.; Stamatis, H. Magnetic microreactors with immobilized enzymes-From assemblage to contemporary applications. Catalysts 2018, 8, 282. [Google Scholar] [CrossRef] [Green Version]
  205. Peñaranda, P.A.; Noguera, M.J.; Florez, S.L.; Husserl, J.; Ornelas-Soto, N.; Cruz, J.C.; Osma, J.F. Treatment of Wastewater, Phenols and Dyes Using Novel Magnetic Torus Microreactors and Laccase Immobilized on Magnetite Nanoparticles. Nanomaterials 2022, 12, 1688. [Google Scholar] [CrossRef]
  206. Baki, A.; Wiekhorst, F.; Bleul, R. Advances in magnetic nanoparticles engineering for biomedical applications—A review. Bioengineering 2021, 8, 134. [Google Scholar] [CrossRef]
  207. Abedini-nassab, R.; Pouryosef Miandoab, M.; Şaşmaz, M. Microfluidic synthesis, control, and sensing of magnetic nanoparticles: A review. Micromachines 2021, 12, 768. [Google Scholar] [CrossRef]
  208. Mariño, M.A.; Fulaz, S.; Tasic, L. Magnetic nanomaterials as biocatalyst carriers for biomass processing: Immobilization strategies, reusability, and applications. Magnetochemistry 2021, 7, 133. [Google Scholar] [CrossRef]
Figure 1. The number of articles published per year in PubMed (Medline) and Scopus databases under the search phrase “magnetic nanoparticle”. The lower number of papers in 2022 is because the literature search was conducted in September 2022. An upward trend is expected for 2022. PubMed comprises biomedical literature from the MEDLINE database and life science journals. Scopus database is the largest abstract and citation database, which covers much more than PubMed scientific journals, books, and conference proceedings.
Figure 1. The number of articles published per year in PubMed (Medline) and Scopus databases under the search phrase “magnetic nanoparticle”. The lower number of papers in 2022 is because the literature search was conducted in September 2022. An upward trend is expected for 2022. PubMed comprises biomedical literature from the MEDLINE database and life science journals. Scopus database is the largest abstract and citation database, which covers much more than PubMed scientific journals, books, and conference proceedings.
Encyclopedia 02 00125 g001
Figure 2. Historic timeline for development of MNPs.
Figure 2. Historic timeline for development of MNPs.
Encyclopedia 02 00125 g002
Figure 3. Primary MNP synthesis methods.
Figure 3. Primary MNP synthesis methods.
Encyclopedia 02 00125 g003
Figure 4. Therapeutic applications of MNPs. MNPs could be applied in various fields of therapy, such as drug/gene delivery, hyperthermia cancer treatment, deep-brain stimulation (DBS), and tissue engineering for regenerative medicine.
Figure 4. Therapeutic applications of MNPs. MNPs could be applied in various fields of therapy, such as drug/gene delivery, hyperthermia cancer treatment, deep-brain stimulation (DBS), and tissue engineering for regenerative medicine.
Encyclopedia 02 00125 g004
Figure 5. Diagnostics applications of MNPs via MRI and MPI techniques.
Figure 5. Diagnostics applications of MNPs via MRI and MPI techniques.
Encyclopedia 02 00125 g005
Figure 6. Therapy and diagnostics application combination (theranostics).
Figure 6. Therapy and diagnostics application combination (theranostics).
Encyclopedia 02 00125 g006
Figure 7. Basic principles of magnetic separation. Magnetically separated targets may be released and analyzed by various physical and biochemical methods. Hexagon, circle, and star are analytes in the solution. The star is a specific analyte, which can be bound for the MNPs surface.
Figure 7. Basic principles of magnetic separation. Magnetically separated targets may be released and analyzed by various physical and biochemical methods. Hexagon, circle, and star are analytes in the solution. The star is a specific analyte, which can be bound for the MNPs surface.
Encyclopedia 02 00125 g007
Figure 8. Schematic representation of the MNP sensing approach. MNPs form a stable complex with target and detector surfaces. The sorption on the surface is caused by a specific capture residue or a magnetic field. The red and yellow circle is the specific analyte.
Figure 8. Schematic representation of the MNP sensing approach. MNPs form a stable complex with target and detector surfaces. The sorption on the surface is caused by a specific capture residue or a magnetic field. The red and yellow circle is the specific analyte.
Encyclopedia 02 00125 g008
Table 1. MNP synthesis comparison.
Table 1. MNP synthesis comparison.
MethodsProcedureConditionsTemperature, TimeMNPs Size and Yield *
Chemical Co-precipitationVery simpleAmbient20–150 °C, minRelatively narrow, High
Hydro/SolvothermalSimpleHigh pressure150–250 °C, h/dayVery narrow, High
SonochemichalVery simpleAmbient20–50 °C, minNarrow, Medium
EmulsionComplicatedAmbient20–80 °C, hNarrow, Low
Thermal decompositionVery simpleHigh temperature250–400 °C, hVery narrow, High
Sol-helSimpleHigh temperature300–500 °C, 3–4 hVery narrow, High
Wet ReductionVery simpleAmbient20–150 °C, minRelatively narrow, High
ElectrochemicalComplicatedAmbient25 °C, min/hNarrow, High
Polyol SynthesisSimpleHigh temperature200–350 °C, 7–10 hRelatively narrow, High
PhysicalGas-phase depositionSimpleHigh temperature150–250 °C, hNarrow, Medium
Ball millingVery simplePower ball/Ambient25 °C, h/dayHighly broad, Medium
SpatteringVery simpleAmbient25 °C, min/hBroad, High
Laser ablationSimpleAmbient25 °C, min/hBroad, High
Electron beam depositionSimpleAmbient25 °C, min/hBroad, Medium
Aerosol spray pyrolysisSimpleHigh temperature300–500 °C, hBroad, Medium
BiologicalMicroorganism and virus mediatedComplicatedAmbient25 °C, h/dayBroad, Medium
Template-mediatedSimpleAmbient25 °C, min/hRelatively narrow, High
Plant-mediatedComplicatedAmbient25 °C, h/dayBroad, Low
* Yield: High = > 90%, Medium = 60–90%, Low lower = 60%.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Petrov, K.D.; Chubarov, A.S. Magnetite Nanoparticles for Biomedical Applications. Encyclopedia 2022, 2, 1811-1828. https://doi.org/10.3390/encyclopedia2040125

AMA Style

Petrov KD, Chubarov AS. Magnetite Nanoparticles for Biomedical Applications. Encyclopedia. 2022; 2(4):1811-1828. https://doi.org/10.3390/encyclopedia2040125

Chicago/Turabian Style

Petrov, Kirill D., and Alexey S. Chubarov. 2022. "Magnetite Nanoparticles for Biomedical Applications" Encyclopedia 2, no. 4: 1811-1828. https://doi.org/10.3390/encyclopedia2040125

Article Metrics

Back to TopTop