Next Article in Journal
Operation Optimization of the Sea Container Fleet Based on the Double-Level Planning Model
Next Article in Special Issue
The Concept of Using 2D Self-Assembly of Magnetic Nanoparticles for Bioassays
Previous Article in Journal
Do Continuous Rotating Endodontic Instruments Extrude Fewer Apical Debris Than Reciprocating Instruments in Non-Surgical Endodontic Retreatments? A Systematic Review
Previous Article in Special Issue
DNA Damage of Iron-Gold Nanoparticle Heterojunction Irradiated by kV Photon Beams: A Monte Carlo Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Magnetic Nanoparticles: Synthesis, Characterization, and Their Use in Biomedical Field

by
Gabriela Fabiola Stiufiuc
1 and
Rares Ionut Stiufiuc
2,3,4,*
1
Faculty of Physics, “Babes Bolyai” University, 400084 Cluj-Napoca, Romania
2
MedFuture—Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
3
Department of Pharmaceutical Physics & Biophysics, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
4
Nanotechnology Laboratory, TRANSCEND Research Center, Regional Institute of Oncology, 700483 Iasi, Romania
*
Author to whom correspondence should be addressed.
Appl. Sci. 2024, 14(4), 1623; https://doi.org/10.3390/app14041623
Submission received: 15 January 2024 / Revised: 9 February 2024 / Accepted: 12 February 2024 / Published: 17 February 2024

Abstract

:
In recent years, the use of magnetic nanoparticles (MNPs) in biomedical applications has gained more and more attention. Their unusual properties make them ideal candidates for the advancement of diagnosis, therapy, and imaging applications. This review addresses the use of MNPs in the field of biomedicine encompassing their synthesis, biofunctionalization, and unique physicochemical properties that make them ideal candidates for such applications. The synthesis of magnetic nanoparticles involves a range of techniques that allow for control over particle size, shape, and surface modifications. The most commonly used synthesis techniques that play a crucial role in tailoring the magnetic properties of nanoparticles are summarized in this review. Nevertheless, the main characterization techniques that can be employed after a successful synthesis procedure are also included together with a short description of their biomedical applications. As the field of magnetic nanoparticles in biomedical applications is rapidly evolving, this review aims to serve as a valuable resource, especially for young researchers and medical professionals, offering basic but very useful insights into recent advancements and future prospects in this highly interdisciplinary research topic.

1. Introduction

From a dimensional point of view, nanomaterials can be divided in three main classes: two-dimensional nanomaterials (2D), such as thin films; one dimensional materials (1D), exemplified by nanowires and nanotubes; and zero-dimensional systems (0D), represented by nanoparticles (NPs) and quantum dots (QDs). In this review we will focus on a special class of nanoparticles (NPs): magnetic nanoparticles (MNPs).
MNPs are materials with dimensions in the nanometer range, typically between 1 and 100 nanometers. At this scale, unique magnetic properties emerge, distinguishing them from their bulk counterparts. MNPs represent a special class of NPs that can interact with an external magnetic field as a direct consequence of their superparamagnetic, ferrimagnetic, and/or ferromagnetic properties.
Their synthesis involves various methods designed to control the size, shape, and properties of these nanoscale materials. Commonly used methods are chemical methods, especially co-precipitation, sol–gel synthesis, and thermal decomposition [1]. Additionally, biological methods, such as biomimetic synthesis using microorganisms or plant extracts, offer eco-friendly alternatives [2], while physical methods offer precise control over nanoparticle size, but may require specialized equipment and controlled environments [3].
A “typical” architecture of the MNPs consists of a core and a coating shell, respectively. The core commonly consists of magnetic elements (Fe, Ni, Co, etc.) as well as their corresponding oxides, while the coating shell is responsible for limiting the interaction of the NPs with the medium, thus preserving their structure and properties. The high surface area of MNPs allows for easy surface modifications, making them versatile for various applications. In view of their use in biomedical applications, the coating plays a crucial role in providing the enhanced biocompatibility of MNPs, without affecting their unique magnetic properties. However, the characterization of such nanostructures is of major importance when it comes to biomedical applications. In this regard, many different techniques were developed that can be used to evaluate the physical and chemical properties of MNPs.
Since MNPs simultaneously possess magnetic properties and experience confinement effects in all three dimensions, a wider range of novel properties that can be harnessed for practical biomedical applications and the exploration of fundamental scientific phenomena can be envisaged.
This paper explores the remarkable properties of MNPs, offering insights into their synthesis and characterization methodologies alongside their applications in the biomedical field. In biomedicine, MNPs show promise for targeted drug delivery, magnetic hyperthermia for cancer treatment, and improved contrast in magnetic resonance imaging. Furthermore, MNPs provide a platform for interdisciplinary research with broad implications for practical applications and scientific exploration. The comprehensive and complete review of relevant aspects related to the synthesis, characterization, and potential applications of MNPs is reflected in the abundance of high-quality bibliographic resources within this scientific work. The novelty lies in its friendly approach to the subject matter and the clear delivery of concepts. This article has the potential to introduce even the most inexperienced readers to the captivating world of magnetic nanoparticles.

2. Synthesis Methods

Significant progress has been made in the synthesis of magnetic nanoparticles (MNPs) in the past decade. The emphasis has been on creating different techniques capable of generating MNPs with certain attributes such as size, shape, magnetic properties, and stability. Compatibility with living organisms is a very important issue that needs to be addressed especially in the case of their use in biological applications. A wide range of synthesis methods were developed over time. These synthesis methods can be divided into physical, chemical, and biological processes (Figure 1). The methods encompassed in this study are ball milling [4], laser ablation, and other physical techniques, coprecipitation, thermal decomposition, hydrothermal synthesis, microemulsion, and the sol–gel technique. Other non-thermal and biological synthesis methods are also mentioned. Each of these methods possesses distinct mechanisms and circumstances for generating MNPs, customized to meet certain requirements and applications [5].

2.1. Physical Methods

Both top–down and bottom–up strategies are utilized in the process of producing magnetic nanoparticles (MNPs) by means of physical methods of synthesis. They represent the two basic physical procedures that are carried out. Top–down techniques involve the process of breaking down bulk materials into nanosized particles. Bottom–up approaches, on the other hand, are used to construct nanoparticles by assembling them from atomic or molecular components. Both methods are essential in the field of nanoparticle synthesis; each has its own set of advantages and disadvantages, and the decision between the two is frequently determined by the particular requirements of the application that the MNPs are meant to be used for.
The physical synthesis methods of magnetic nanoparticles (MNPs) involve a range of procedures that mostly rely on mechanical or physical processes to generate nanoparticles.

2.1.1. Lithography

This versatile technique has been employed for the synthesis of a wide range of nanoparticles including MNPs. Lithography is part of the micropatterning technique being based on a controlled transfer of patterns from a mask or template to a substrate, allowing for the precise positioning and arrangement of nano-sized features. Over time, the technique has been optimized and new methods have been implemented: electron beam, scanning, nanosphere, and colloidal lithography, together with soft nanoimprinting [6].

2.1.2. Mechanical Milling

The ball milling technique is a popular physical process that involves the mechanical grinding of coarse-textured particles into fine nanoparticles, using a top–down approach. This technique, initially pioneered in the 1970s, is straightforward and practical. It entails utilizing a cylindrical jar with a hollow center (containing steel balls) that crushes the solid substance into a powder of nano/micrometric dimensions. Although ball milling is successful, it is limited by the possibility of product contamination and the generation of particles with a broad range of sizes [4].

2.1.3. Laser Evaporation

Laser evaporation, also referred to as laser ablation or vapor deposition, is an additional physical technique that follows a bottom–up strategy. This method involves the formation of nanoparticles by the condensation process from either a liquid or gaseous phase. High-energy lasers are utilized to vaporize raw materials when concentrated by a laser beam. The vaporous substance is subsequently cooled in a gaseous state, causing fast condensation and nucleation, ultimately leading to the creation of nanoparticles. This process is cost-effective, efficient, and does not generate any hazardous waste, unlike certain wet chemistry procedures [7,8,9,10,11].

2.1.4. Wire Explosion

The wire explosion method is a novel physical methodology used to synthesize MNPs. The procedure is both safe and clean, and it is highly efficient without the need for additional stages like separating nanoparticles from the solution or treating leftovers. This technique has been employed to synthesize iron oxide magnetic nanoparticles (MNPs) specifically for applications involving arsenic removal from water. Although the nanoparticles created using this process are ecologically friendly and energy-efficient, they are not uniformly dispersed, which may restrict their use in some applications [12,13,14].

2.1.5. Gas-Phase Synthesis

The gas-phase synthesis method is part of the bottom–up methods and was implemented for developing multifunctional nanoparticles [15]. The procedure involves the formation of nanoparticles directly from gaseous precursors providing control over size, composition, and crystallinity. There are two common gas-phase synthesis methods: chemical vapor condensation (CVC) and inert gas condensation (IGV).
CVC is based on the reduction of the metal precursors in the gas phase. In this regard, volatile metal precursors such as metal halides or carbonyls are used as chemical precursors. They are reduced at high temperatures, leading to the formation of MNPs that are cooled down and collected.
During the IGV process, the nanoparticles are produced as a condensation result after the inert gas-based cooling of the vapor produced by the vacuum evaporation of metal atoms or clusters.
The main advantage of the gas-phase synthesis procedures refers to their good control over particle size [16].

2.2. Chemical Methods

Chemical methods represent one of the major directions in this vast, dynamic, and rapidly evolving field of nanoparticles’ synthesis. The chemical synthesis of magnetic nanoparticles involves several methods, each with its own set of advantages and limitations. In the following paragraphs we will describe the main procedures belonging to this class that are reported in the scientific literature for the synthesis of MNPs.

2.2.1. Precipitation and Coprecipitation

Precipitation and coprecipitation stand out as the prevailing techniques for the fabrication of MNPs consisting of magnetic metal oxide nuclei from corresponding salts [17,18]. These MNPs possess a regulated size and magnetic properties, owing to the straightforward, adaptable, and less detrimental procedures employed for their synthesis [17,18,19,20]. Moreover, these approaches are secure, economical, and necessitate a lower synthesis temperature compared to alternative conventional methods [21,22]. Precipitation and coprecipitation are particularly suitable when there is a demand for the production of large quantities of nanocrystals [19].
In a typical scenario, metal ions are employed to dissolve in a solvent and to form a solution containing these metal ions, that will be further used for MNP production [19]. A common illustration of MNPs synthesized through these methods is Fe3O4 or γ-Fe2O3 NPs, obtained from ferrous (Fe2+) and ferric (Fe3+) salts dissolved in an alkaline solution [17,20]. The typical composition involves a 2:1 ratio of Fe2+ and Fe3+ salts, either at room temperature or an elevated one (80–85 °C) [20]. Once the reaction concludes, a precipitate of Fe3O4 forms at reactor’s base. It can be recovered through either centrifugation or magnetic isolation [20]. The expected pH range for the resulting precipitate is between 7.5 and 14 [20]. It’s noteworthy that the synthesis of this MNPs type heavily relies on the sources of metal ions and factors such as the temperature, pH, and ionic strength of the reaction medium [17]. In fact, a majority of commercially available iron oxide nanoparticles (IONPs) are synthesized using the aforementioned methods [23,24].
Other researchers obtained manganese ferrite (MnFe2O4) NPs using ferric chloride (FeCl3) and manganese (II) chloride (MnCl2) as metal ions sources, and sodium hydroxide (NaOH) as a precipitant [25]. MgFe2SO4 nanocrystals can be generated through the coprecipitation of Fe3+ and Mg2+ ions, facilitated by the addition of NaOH [26].

2.2.2. Thermal Decomposition Method

The thermal decomposition method, also referred to as pyrolysis, was developed to overcome the limitations of the co-precipitation technique by utilizing non-aqueous solvents with elevated boiling points [27]. This method is employed to produce highly crystalline and uniformly sized MNPs [20]. Primarily, it involves the elevated temperature-induced decomposition of organometallic compounds in organic solvents, conducted in the presence of stabilizing agents, such as surfactants [19,28,29].
Various organometallic precursors of NPs include those synthesized with acetylketonates, N-nitrosophenylhydroxylamine, and carbonyls possessing various metallic centers such as Fe2+,3+, Mn2+,3+, Co2+,3+, Ni2+,3+. Stabilizing agents like fatty acids, oleic acids, and hexadecylamine are frequently utilized [30]. The type of the NPs depends on the chosen organometallic compound used as a source [17].
The temperature requirement varies based on the precursor type [19]. To achieve a high degree of uniformity and a size ranging from 4 to 30 nm, the optimal temperature falls within the 100–350 °C range [31,32]. Some authors consider that the temperature, reaction time, type of surfactants and solvents, and aging period should be adjusted according to the desired shape and size [33,34]. For instance, NP size was controlled by adapting the reaction time, resulting in sizes of 4, 12, and 60 nm for times of 1, 10, and 24 h, respectively [17]. The reaction time also influenced the morphology of the NPs, a transition from spherical NPs (at an early stage) to cubic ones (over longer times) being observed [17].
Significant findings include the synthesis of monodispersed iron oxide MNPs ranging from 6 to 20 nm through the polymer-catalyzed decomposition of Fe(CO)5 [35,36]. Iron oxide MNPs (IOMNPS) were synthesized from metal fatty acid salts, yielding nearly monodispersed NPs with adjustable sizes between 6 and 50 nm and shapes evolving from points to cubes with increasing reaction time [37].
It is noted that any remaining surfactants may impact surface modification efficiency [20]. Oleic acid, hexadecylamine, and fatty acids are the most commonly used surfactants in this method [20].
MNPs synthesized using this method exhibit promising potential as magnetic resonance imaging (MRI) contrast agents for diagnosing cancerous tissue [17]. Other advantages of this method include high crystallinity, dispersibility, uniformity, homogeneous shape, and a fine particle size distribution [38,39].
Despite these substantial advantages, this method requires extensive purification steps of the synthesized MNPs (especially for biomedical applications) [40] due to the production of toxic organic-soluble solvents [41]. Additionally, this method is time-consuming and has low product yields [20].

2.2.3. Hydrothermal Method

The hydrothermal method, also known as solvothermal, is a “bottom–up” synthesis technique for IOMNPs, conducted in an aqueous solution under elevated pressure and temperature, typically using autoclaves or reactors [42,43]. Primarily, it involves the rapid nucleation and growth of newly synthesized MNPs, resulting in pure particles with controlled morphologies [44].
During the hydrothermal process, hydrolysis and oxidation reactions occur in order to generate MNPs [45]. Some authors emphasize that the morphology and crystallinity of the as-synthesized MNPs depend on the proper mixing of solvent, duration, pressure, and temperature [19]. It’s crucial to note that crystal formation relies on the solubility of minerals in water [19].
Impressive results achieved using this method include synthesis of Fe3O4 NPs with a size of 15 nm and a spherical shape, successfully employed in tumor MRI applications [46]. Additionally, chitosan coated Fe3O4 NPs, sized at 25 nm, were prepared and utilized for magnetic based enzyme immobilization [47].
However, this method has drawbacks such as: the inability to synthesize particles smaller than 10 nm and slow reaction kinetics at high temperatures [48]. It requires careful execution and specialized equipment, making the protocol laborious [49]. Despite these challenges, there are noteworthy advantages, including the production of NPs with a desirable shape, size, high crystallinity, and consistent composition, offering magnetic tuning and greater control over dispersion [48,49].

2.2.4. Polyol Method

The polyol method stands out as one of the simplest approaches for nanoparticle synthesis. This technique facilitates the synthesis of nanoparticles from inorganic compounds, including alloys, sulfides, oxides, fluorides, etc., [50]. It presents significant advantages by enabling the cost-effective single-step production of high-crystalline hydrophilic MNPs that can be easily scaled up [51].
In this method, polyols serve multiple roles, acting as a polar organic solvent for metal precursors, reducing agent, and, in certain instances, complexation agent for metal cations [20]. The polyol process has demonstrated its ability to control MNPs’ morphologies, ranging from single-core spherical MNPs to more intricate multicore nanostructures, including hollow spheres and nanoflowers [51]. Additionally, adjusting the reaction temperature can increase NP size [52].
This technique possesses several advantages, including the elimination of separate calcination steps and the use of polyol as the sole solvent [53]. Furthermore, polyols are recognized as cost-effective and environmentally friendly solvents, widely employed in various industries [53].

2.2.5. Sol–Gel Method

The sol–gel method is a wet chemistry procedure involving the formation of a gel at room temperature through hydrolysis and polycondensation reactions of metal alkoxides [19,42]. Metallic salts are dissolved in water or other solvents, forming a homogeneously dispersed sol or colloidal solution [48]. Van der Waals forces between the particles are induced. The interaction between the particles intensifies through stirring and temperature increase [19]. The mixture undergoes heating until the solvent is evaporated, resulting in the solution drying and the ultimate formation of a gel [19,54].
This method is particularly effective for the synthesis of iron oxide MNPs and silica coated MNPs. It enables the production of MNPs in significant quantities with controlled sizes and well-defined shapes [19].

2.2.6. Microemulsion Method

Microemulsion is a system of water, oil, and an amphiphilic surfactant that forms a stable, thermodynamically stable dispersion. The process involves the incorporation of precursor materials into different phases of the microemulsion, leading to NP formation. Chemical reactions occur within the confined spaces of the microemulsion droplets, leading to the formation of nanoparticles [55].
Different variations of microemulsion methods exist:
  • reverse microemulsions, where water is dispersed in an oil phase with a surfactant;
  • direct microemulsions, where oil is dispersed in a water phase with a surfactant;
  • where both oil and water are present in a comparable amount [56].
The advantage of using the microemulsion method is precise control over NP size, shape, and composition [55]. Several parameters (water-to-surfactant ratio, concentration of the reactants, and surfactant film flexibility) influence the size of MNP microdroplets [57]. This method’s drawbacks are polydispersity, a low yield of NPs, large solvent volumes, and poor versatility in metal precursors compared to coprecipitation and thermal decomposition [17].

2.2.7. Pyrolysis Methods

Spray pyrolysis is a technique commonly used for the synthesis of various materials, including MNPs. This method involves the formation of fine droplets of precursor solutions that are sprayed into a high-temperature environment. The solvent evaporates, and the remaining solute undergoes chemical reactions to form the desired material, collected on a substrate [58].
In precise context of MNP synthesis, spray pyrolysis offers several advantages: controlled particle size, homogeneous coating, scalability, versatility, and tunable magnetic properties [59].
Laser pyrolysis is a method which involves the use of CO2 laser irradiation to induce chemical reactions in a precursor material, leading to the formation of NPs. The principle consists of focusing a high-intensity laser beam on a precursor material in the gas or aerosol phase. The laser energy is absorbed by the precursor, leading to its rapid heating and decomposition. The resulting chemical reactions lead to the formation of NPs, which are then collected on a substrate [3]. This technique offers both advantages (rapid heating, high purity, fine tuning) and characteristics that need to be improved (scalability and cost) [60].

2.2.8. Non-Thermal Methods

Non-thermal methods for the synthesis of MNPs refer to techniques that do not involve high temperatures during the production process. These methods are often preferred for their ability to produce NPs with unique properties and for avoiding issues related to thermal decomposition or aggregation [61].
Common non-thermal methods used for MNP synthesis are:
  • Chemical Reduction
Magnetic nanoparticles are formed through the reduction of metal ions by chemical agents without the need for high temperatures. This method is simple, cost-effective, and can be used for a variety of metals. It is also commonly used for the synthesis of metal nanoparticles, such as gold, silver, and platinum nanoparticles [62].
  • Electrochemical Synthesis
MNPs are produced through electrochemical reactions at the electrodes. Using this method, size and morphology control, simplicity and scale-up ability can be achieved. Also, it is worth mentioning that it is suitable for various materials, including metals, metal oxides, and conducting polymers [63]. Such methods have gained confidence in relation to conventional ones by allowing the synthesis of superparamagnetic nanoparticles in surfactant or stabilizing agent-free conditions [64]. Moreover, new methods showed increased efficiency in obtaining multimetallic nanoparticles [65].
  • Microwave-Assisted Synthesis
Microwave radiation is used to heat reaction mixtures, facilitating rapid and uniform heating. This method possesses faster reaction rates, higher yields, and reduced energy consumption compared to conventional heating methods. It is applied to a wide range of materials, including metals, metal oxides, and carbon-based nanoparticles [66].
  • Ultrasound-Assisted Synthesis
Ultrasonic waves are applied to a reaction mixture, leading to acoustic cavitation and promoting particle formation. This technique has rapid reaction rates, control over particle size, and improved homogeneity. It is used for metals, metal oxides, and organic nanoparticles [67].

2.3. Biological Methods

The field of nanotechnology has witnessed the rise of a significant field known as biological synthesis of magnetic nanoparticles (MNPs). This approach offers an environmentally friendly and sustainable alternative to conventional physical and chemical manufacturing techniques. This method employs biological entities, such as microorganisms, plant extracts, and enzymes, to synthesize MNPs. Each of these biological entities holds distinct advantages and is suited for a range of applications, as follows:
  • Microorganism-based synthesis involves the utilization of microorganisms, including bacteria, fungi, viruses, and actinomycetes, to generate MNPs. The presence of microorganisms greatly enhances the efficiency of metal ion reduction, hence facilitating NP synthesis. Magnetotactic bacteria are renowned for their ability to synthesize MNPs intracellularly. The nanoparticles are employed for orienting themselves in geomagnetic fields [68,69]. Furthermore, the use of fungi in the production of MNPs has been extensively recorded. Various fungal species have been recognized for their ability to generate iron oxide nanoparticles, which have significant promise in the domains of remediation and biomedicine [70,71].
  • Plant extracts, including phytochemicals, are utilized in the manufacture of nanoparticles. They serve as both reducing and stabilizing agents. This is achieved through the utilization of botanical extracts. The method’s simplicity, cost-effectiveness, and scalability are driving its increasing worldwide popularity. Numerous plants have been employed for this objective. Diverse parts of plants such as leaves, stems, and roots were employed to provide the necessary bioactive constituents. For instance, the utilization of plant extracts, such as aloe vera and green tea, has proven to be successful in the production of IONPs. The nanoparticles showed promise in the areas of drug delivery and magnetic resonance imaging (MRI) applications [34,72].
  • Enzymatic synthesis process employs specific enzymes to catalyze the reduction of metal ions in order to produce metal nanoparticles. Enzymes have significant control over the nucleation and growth processes, leading to the formation of NPs that exhibit a uniform size and shape. The enzymatic approach is extremely intriguing for use in medical and pharmaceutical areas due to its exceptional biocompatibility and precision. Several enzymes are currently under investigation [72,73] as a direct consequence of growing research efforts in this area. Moreover, some protein assisted synthesis models were implemented for biosensing applications [74].
Since these methods apply biological principles for nanoparticle formation, they are also called biomimetic synthesis procedures [75]. These techniques are not only environmentally benign, but they yield products that are highly suitable for delicate applications such as drug administration, bioimaging, and environmental remediation. Due to their high biocompatibility and minimal environmental impact, they represent a very promising research topic in the field of nanotechnology.

2.4. Coating of Magnetic NPs

In order to improve the functionality, stability, and biocompatibility of MNPs, the coating process is an essential step. This is especially true for applications in biomedicine and environmental research. MNPs can be coated with a variety of different molecules, including:
Polymeric Coatings: polymers, both natural and synthetic, are frequently utilized for MNP coating. Due to their biocompatibility and capacity to inhibit particle aggregation, synthetic polymers such as poly(ethylene glycol) (PEG) [76], poly(vinyl alcohol) (PVA) [77], and poly(lactic acid) (PLA) [78] are among the most preferred options.
Coatings made of Silica: silica coatings are applied to MNPs in order to increase their reactivity and their potential for functionalization. The surface of an MNP consisting of maghemite or magnetite, for example, can be coated with silica. The silica shell can be further modified with a variety of functional groups by the formation of covalent bonds between organo-silane molecules and the silica shell. This approach is especially useful for attaching fluorescent dyes and/or other functional molecules to the outer surface of MNPs, which represent a particularly advantageous use [79,80,81].
MNPs can be coated either during the synthesis process (in situ) with stabilizers such as surfactants or polymers, or after the synthesis process (post-synthesis) with materials such as monolayer ligands, mixtures of polymers, biomolecules, and inorganic compounds. In situ coatings are more common than post-synthesis procedures [82]. When it comes to functionalizing MNPs for specific purposes, such as medical applications, where it is important to bind biological ligands to NP’ surfaces, post-synthesis procedures offer a wider range of options [83,84,85,86].
When it comes to medical diagnostics, drug delivery, or environmental cleanup, each form of coating confers particular features which align with the application that is they are specifically meant for. The functioning, biocompatibility, and stability requirements of MNPs need to be taken into consideration before deciding the coating selection.

3. NP Properties and Characterization Methods

The physico-chemical characterization of MNPs is an essential process that requires the use of a wide range of advanced techniques in order to determine the magnetic, chemical, and physical characteristics of these particles. When it comes to understanding MNPs behavior and ensuring that they are suitable for a specific application, these methodologies are absolutely necessary. This is particularly relevant in the fields of biomedicine, environmental remediation, and nanotechnology. In the next subchapters some of the techniques that offer comprehensive insights into the size, composition, structure, and magnetic properties of this class of NPs will be presented. To improve the development of MNPs and to maximize their potential use in a variety of scientific and industrial applications, the precise selection of these characterization methods is crucial.

3.1. Physical Properties and Their Characterization

Precisely determining the properties of MNPs is extremely important. The physical properties of MNPs are highly dependent on their size, bearing in mind that as the size decreases to nanoscale, various physical properties can change significantly [87].
The size and shape of MNPs has a direct impact on their behavior and functionality, influencing features such as magnetic response, biocompatibility, and cellular uptake. As MNPs decrease in size, they may transition from multi-domain to superparamagnetic behavior, exhibiting negligible magnetic remanence and coercivity. Smaller MNPs often reach magnetic saturation more quickly under an external magnetic field as compared to larger particles. This property is crucial for applications such as magnetic storage and sensing [88].
For example, nanoparticles with a size less than 25 nm, such as magnetite, display a superparamagnetic behavior [89], which is an essential characteristic for applications like targeted medication administration and MRI.
Size can also influence the blocking temperature at which MNPs become magnetically blocked. Smaller particles typically have lower blocking temperatures [90].
Moreover, smaller MNPs have a higher surface area-to-volume ratio, making them more reactive and suitable for drug delivery or catalysis applications. This particularity allows for more effective functionalization by attaching targeting ligands or other molecules to achieve higher biocompatibility [88].
On the other hand, smaller nanoparticles are more prone to agglomeration due to increased surface energy [91].
The characterization of magnetic nanoparticles (MNPs), particularly for biomedical applications, encompasses a range of procedures that primarily examine MNPs’ inorganic crystalline core and organic shell, which is required to inhibit the formation of MNPs aggregates and minimize the likelihood of an immune system reaction [92,93]. The main techniques capable of providing crucial insights into the physical characteristics of these NPs are transmission electron microscopy, dynamic light scattering and magnetometry methods.

3.1.1. Transmission Electron Microscopy (TEM)

TEM is essential for capturing high-resolution images of MNPs. It provides comprehensive information regarding their dimensions, form, and interior composition. This technique is crucial for observing the inorganic core and any organic shell present on the surface of the nanoparticles, offering crucial insights into their microstructure or ultrastructure. In contrast to other methods of size assessment, it accurately measures the true radius of samples. TEM, which requires desiccation before measurement, gives information about magnetic nanoparticles in their dry state . However, a significant limitation of TEM is its restriction to small sample sizes and the considerable time required for measurements. Furthermore, as operator bias can impact the selection of photos, researchers are recommended to refrain from introducing subjectivity. To obtain a statistically precise measurement of NPs’ diameters, it is necessary to collect many images at an appropriate level of magnification. This will ensure that there is sufficient contrast between the particles and the background, while also allowing for the inclusion of multiple particles (a minimum of 300 in order to obtain adequate statistical data) in each image. The photos can thereafter be examined with a software application like ImageJ [94].
High-resolution transmission electron microscopy-electron energy loss spectroscopy (HRTEM-EELS) provides a comprehensive investigation of MNPs’ composition. It is highly valuable for determining the elemental and electrical structures, including the characteristics of multifunctional NPs such as core–shell Fe3O4/ZnxCo1−xFe2O4 [95].

3.1.2. Scanning Electron Microscopy (SEM)

SEM is a powerful imaging method that uses electrons to examine the surface morphology of a sample at high resolution, enabling the detailed examination of individual nanoparticles [96].
In the case of MNPs, SEM provides quantitative information regarding their size, shape, and surface. SEM allows the visualization of surface modifications and functionalization, providing insights into their stability and potential applications [97]. SEM can reveal the agglomeration state of magnetic nanoparticles, which is important for understanding their behavior in different environments [98,99]. However, this technique does not directly provide information about the magnetic properties of MNPs which can be evaluated by means of magnetometry methods.
The SEM of MNPs is crucial for understanding their physical characteristics, which is vital for applications in drug delivery, magnetic hyperthermia, magnetic resonance imaging, and other biomedical and technological fields [97,98,99].

3.1.3. Atomic Force Microscopy (AFM)

AFM provides high-resolution topographical images of MNPs by allowing the visualization of surface morphology and size distribution. Moreover, this technique can be employed to map the magnetic properties of nanoparticles across a surface. This includes measurements of magnetic force, coercivity, and remanence, providing insights into the magnetic behavior of the nanoparticles.
When it comes to functionalizing MNPs, AFM can be engaged to assess the surface modifications involved in the interaction between nanoparticles and various molecules. The method can be used in conjunction with other imaging techniques for a rigorous characterization [100]. It must be mentioned that sample preparation is crucial to ensure accurate and reliable results.

3.1.4. Dynamic Light Scattering (DLS)

DLS is a technique employed to determine the hydrodynamic size of MNPs present in a liquid solution. It examines the patterns of light scattered by particles, which is crucial for understanding their behavior in colloidal suspensions.

3.1.5. Magnetometry

Comprehending and enhancing the magnetic properties of MNPs is essential for their effective application in diverse domains, particularly in biomedicine. The magnetic characteristics, including magnetic moment, coercivity, and responsiveness to external magnetic fields, determine the operational capabilities of MNPs in many applications such as MRI, targeted drug administration, hyperthermia, and magnetic data storage [66]. The optimization of these attributes guarantees the effectiveness, precision, and security of MNPs, rendering their evaluation through state-of-the-art methods such as Vibrating Sample Magnetometry (VSM) and/or Superconducting Quantum Interference Device (SQUID) Magnetometry essential [67,68].
VSM is a cost-effective and straightforward technique that offers accurate information about the value of the magnetic moment of the analyzed substance. The sample is positioned within a copper coil and subjected to an applied magnetic field. Subsequently, it is oscillated in a direction perpendicular to the field. The magnetic moment is quantified by detecting a variation in the electrical potential. This method is valuable for evaluating the magnetic moment in relation to temperature, magnetic field, and crystal orientation [101]. Nevertheless, VSM has a relatively lower sensitivity in comparison to more sophisticated techniques such as SQUID magnetometry [102].
SQUID magnetometry is renowned for its exceptional sensitivity and is employed to analyze superparamagnetic substances, which can be recognized by the absence of a hysteresis loop. Hysteresis loops, which depict the magnetization characteristics of a material, play a vital role in comprehending magnetic features such as remnant magnetization and coercive field. Superparamagnetic nanoparticles, which have a single magnetic domain, do not display a hysteresis loop like ferromagnetic ones [101,103]. This method also aids in determining intrinsic characteristics, such as the Curie temperature for ferromagnetic substances and the Neel temperature for antiferromagnetic materials [102,103].

3.2. Chemical Properties and Their Characterization of MNPs

From a chemical point of view, MNP characterization involves determining their composition, structure and surface properties. To achieve such information, different analytical techniques can be employed. In the next paragraph many of them are briefly presented.
Usually, MNPs contain magnetic materials such as iron oxide, magnetite, maghemite, but can contain other materials like cobalt, nickel, or alloys. Such compositions are typically coated with silica, surfactants, or biocompatible materials (e.g., polymers). These coatings provide biocompatibility, functionality, and stability [104].
The surface properties, also called reactivity, are highly dependent on the surface area. This property controls the surface charge, interactions, oxidation state and heating efficiency of MNPs [104,105].
Chemical characterization ensures the quality and the consistency of the MNPs in achieving the desired specifications and properties required for particular applications. As for biomedical applications, safety and biocompatibility are parameters of crucial importance. Understanding reactivity and interactions between NPs and other entities is essential for such applications.

3.2.1. UV–Vis Spectrophotometry

UV–Vis spectrophotometry is a widely used technique for characterizing nanoparticles, particularly metal nanoparticles. It is used for quantitative analysis of nanoparticle concentration based on the Beer–Lambert Law, which relates absorbance to concentration.
This method relies on an NP–light interaction, leading to changes in the absorption spectra. When metal nanoparticles, especially noble ones like gold and silver, are exposed to light, they interact with the electromagnetic field, leading to phenomena such as surface plasmon resonance (SPR).
The absorption of light by nanoparticles leads to the occurrence of characteristic peaks in the UV–Vis spectrum. The position of the SPR peak is sensitive to factors such as the NPs’ size, shape, and surrounding medium. This is particularly useful for monitoring the progress of nanoparticle synthesis reactions [106].

3.2.2. Nuclear Magnetic Resonance (NMR)

Nuclear Magnetic Resonance (NMR) spectroscopy is a powerful analytical technique commonly used for the characterization of magnetic nanoparticles. While NMR has some limitations when it comes to studying nanoscale materials directly, it can provide valuable information about the structure, composition, and interactions involving nanoparticles.
NMR can be used for quantitative analysis, allowing the determination of NPs concentration in a sample. This is particularly useful for assessing the synthesis yield.
Surface functionalization can be analyzed by observing changes in chemical shifts and peak intensities. Ligands or surface coatings on nanoparticles can be characterized by NMR, providing insights into their composition and arrangement.
Nevertheless, MNPs’ size and dispersion can be studied through NMR relaxometry.
Techniques such as diffusion-ordered spectroscopy (DOSY) can be employed to study the translational diffusion of nanoparticles [106].

3.2.3. Fourier Transform Infra-Red (FT-IR) Spectroscopy

It provides information about the molecular composition, functional groups, and chemical bonds present on the surface of MNPs.
FT-IR spectroscopy measures the absorption of infrared light by the sample. Different functional groups absorb specific wavelengths of infrared light, resulting in characteristic peaks in the spectrum. The Fourier Transform technique is used to convert the raw data into a Fourier Transform Infrared spectrum.
This technique is particularly useful for studying the surface functionalization of nanoparticles. Peaks in the spectrum can be assigned to specific functional groups, providing information about the ligands or surfactants attached to the NPs’ surface. It can also identify chemical bonds present in the system, allowing researchers to characterize the composition of the material.
Other applications are materialized through quantitative analysis by enabling the determination of the concentration of functional groups on the nanoparticle surface, particle size evaluation by monitoring the chemical reactions and, last but not least, the NPs’ agglomeration state quantification [106].

3.2.4. Raman Spectroscopy (RS)

RS represents a powerful set of methods that can be used to evaluate the structural and chemical properties of MNPs. The techniques are based on the inelastic scattering of the light that interacts with the sample due to molecular vibrations. Therefore, the resulting spectrum relies on the peaks corresponding to the vibrational modes of the MNPs. Modifications in peak positions or intensities, or the occurrence of new peaks, may provide valuable insights into the chemical composition, crystallinity, and structure [107,108,109].

3.2.5. Mass Spectrometry (MSp)

MSp methods can provide valuable information regarding nanoparticles structure, composition, and surface chemistry. MSp coupled with other techniques is useful for various analyses concerning the NPs’ properties [110].
MSp coupled with liquid chromatography is suitable for studying ligands’ nature and distribution. MSp combined with matrix-assisted laser desorption can be used to analyze the ligands or the capping agents attached at the surface of MNPs and to identify MNPs’ size distribution [111].
Inductively coupled plasma MSp can support the elemental composition in order to understand their chemistry and to detect the presence of certain impurities [112].
MSp can ensure both quantitative and qualitative assays and can provide insights into structural characterization. However, it is important to use other complementary techniques for a better understanding of the NPs’ properties [113].

3.2.6. Energy Dispersive X-ray Spectroscopy (EDS)

EDS is used in conjunction with Transmission Electron Microscopy (TEM) to perform elemental analysis. This approach is crucial for the identification of specific elements that have been absorbed onto the surface of MNPs, hence facilitating the study of surface coatings and alterations.

3.2.7. X-ray Diffraction (XRD)

XRD is employed to analyze the arrangement of atoms in the magnetic core, providing insights into their crystalline structure. It offers information regarding crystal arrangement, the distance between their lattices, and the dimensions of the crystallites. These factors are crucial in determining the magnetic behavior of these substances.

3.2.8. Small Angle X-ray Scattering (SAXS)

SAXS is a crucial technique for nanoscale analysis. It provides valuable information about the arrangement and self-organization of dispersed nanoscale colloids in their natural solutions. It possesses a distinct capability to investigate the averaged structural information of the materials, including information about the crystals’ structure, lattice spacing, and crystallites size [114].

4. Biomedical Applications

Magnetic nanoparticles (MNPs) have become a crucial tool in the biomedical area, significantly transforming different diagnostic and therapeutic methods. Due to their distinctive magnetic properties, as well as their compatibility with biological systems and capacity to be modified on the surface, these materials are highly suitable for a wide range of applications, including magnetic resonance imaging (MRI) [115], targeted drug administration, and cancer therapy [116,117]. The adaptability of MNPs resides in their capacity to react to external magnetic fields, allowing for accurate regulation and manipulation inside biological systems. This characteristic is especially beneficial for targeted therapies, as it allows for the direct administration of medications or therapeutic agents to specific locations, hence reducing the occurrence of adverse effects throughout the entire body [118]. Moreover, their use in diagnostic imaging, namely in augmenting MRI contrast, facilitates enhanced and more intricate vision of internal body structures, resulting in enhanced diagnostic precision [20].
The potential of MNPs in biomedical applications is extensive, and current research is investigating novel and effective methods to employ these NPs. Future prospects involve the advancement of multifunctional MNPs with the ability to perform both diagnosis and treatment simultaneously, commonly known as ‘theranostics’ [119,120,121]. This integration holds the potential to improve personalized medicine, wherein treatment approaches can be customized to meet the specific needs of each patient. Furthermore, the investigation of MNPs in the field of regenerative medicine and tissue engineering offers a fresh and promising direction for scientific inquiry, with the potential to facilitate innovative approaches to tissue repair and regeneration [121].
MNPs have a significant benefit in their large surface area-to-volume ratio, enabling various functionalization and alterations that respond to specific biomedical requirements. The ability to adapt allows for the creation of magnetic nanoparticles (MNPs) with tailored characteristics, suitable for a range of uses including hyperthermia cancer treatment [122] and molecular detection. However, a notable disadvantage is the potential toxicity and biocompatibility. In this regard, the physical properties of NPs, such as the size, concentration, shape, and coatings, together with exposure time, the number of exposed cells, and chemical compositions are of major importance [93]. For example, iron oxide nanoparticles enter the cell usually through endocytoses. The internalization process is highly dependent on their size [92,102] and coating [95]. At high doses, MNPs can lead to the generation of reactive oxygen species, affecting the cells’ fate [96,97].
It is essential to ensure that MNPs do not cause negative immune reactions or gather in important organs in order to safely use them in medical treatments. Furthermore, the ongoing investigation of the enduring stability and deterioration of MNPs in biological systems is being conducted to address any possible health hazards.
Based on the unique magnetic properties of these nanoparticles, they are suitable for diverse applications as can be seen in Figure 2.

4.1. Magnetic Hyperthermia Therapy (MHT)

MHT takes advantage of MNPs’ capacity to interact with an alternating external magnetic field in order to induce heat generation for cancer treatment applications by means of hyperthermia. This non-invasive technique elevates the temperature of tumors to a range of 41–47 °C, causing cancer cell destruction through either apoptosis or necrosis. Magnetic nanoparticles (MNPs) are administered into the body, gather at the location of the tumor where they specifically aim, and raise the temperature of cancer cells without causing damage to nearby healthy tissues. This approach offers distinct advantages compared to chemotherapy, as it exhibits precise targeting and minimizes harm to healthy cells [123,124], being implemented in cancer treatment since the 1950s [125].
Many studies have been reported in the literature, but in the next rows we would like to highlight some of the recent and interesting findings in the field of MHT applications.
It has been shown that the effectiveness of various molecules (chemotherapeutics, nucleic acids, proteins) has been enhanced when they were used in conjunction with MHT [126,127,128]. In other cases, MNPs harmed the cells even in the absence of MHT [91].
Vilas-Boas et al. have proposed a combination between superparamagnetic and magnetic immuno-conjugated nanoparticles. The strategy was to treat the cancer cells with immuno-conjugated magnetic nanoparticles and then with superparamagnetic iron oxide nanoparticles (SPIONs) and, by applying an external alternating magnetic field, to enhance the efficacy of the treatment [129]. The study showed promising results offering new perspectives for the development of new nanostructures for cancer applications.
Developing inhalable composites is a very challenging aspect when it comes to lung cancer treatment. Magnetic nanocomposite microparticles via MHT can serve as an effective thermal therapy platform inducing targeted damage at the tumoral site [130].
Exosome-based research has been influenced by the advances of hybrid nanostructures in tumor targeting by means of MHT. For example, by attaching magnetic nanoparticles at the surface of the exosomes previously loaded with targeting and therapeutic molecules, and by applying an external magnetic field, it has been developed an anti-cancer hybrid drug delivery system [131].
Many drug delivery nanostructures take advantage of MHT in delivering their cargo at the tumor site. Such applications together with other valuable examples of MHT applications can be found in Table 1.

4.2. Targeted Drug Delivery (TDD)

As a direct consequence of their magnetic properties, MNPs can be employed as carriers for precise drug delivery, as they may be directed to certain tissues with the aid of an external magnetic field. This method enhances the bioavailability of medications and enables their targeted administration to affected tissue, while minimizing systemic effects. The focused administration of medication improves its therapeutic efficacy and minimizes the likelihood of adverse reactions [124,132,133,134].
TDD efficiency is highly influenced by the applied magnetic field, NP magnetism, size, shape, and surface coating, but also by extracellular and intracellular barriers [135]. Beside drugs, other molecules that can be loaded in MNPs involves nucleic acids, cells, proteins, or antibodies [135,136].
SPIONs coated with paclitaxel–chitosan and folate–polyethyleneglycol proved to be an efficient strategy for targeting fibrosarcoma. The nanosystem induced cancer cells’ apoptosis and decreased tumor size [137].
An external magnetic field combined with ultrasound led to the development of a TDD system composed of magnetic nanoparticle coated liposomes and microbubbles as a therapy for atherosclerosis plaques. This strategy was based on MNPs’ capacity to penetrate and migrate into inaccessible tissue during MHT. Nevertheless, the ultrasound field can increase the therapeutic compounds delivery and thus the targeting of the desired tissue [138].
Such approaches have been used in MNP research in theranostics, a new and interesting topic that recently gained a lot of attention [139,140,141,142,143,144].
MNPs loaded with nanoparticles containing growth factors were used both as delivery and labeling systems with a great potential in regenerative medicine [145].
Multifunctional magnetic nanoparticles, also known as smart drug delivery systems, can update already developed cancer therapies [143]. Such examples can be found in Table 1.

4.3. Photothermal Therapy (PTT)

PTT using magnetic nanoparticles involves the use of nanoparticles that possess both photothermal and magnetic properties. These nanoparticles can be designed to absorb light and convert it into heat, as well as respond to an external magnetic field. This combination allows for a targeted and controlled approach for treating diseases, particularly in the field of cancer therapy. PTT is a minimally invasive treatment that utilizes MNPs to effectively address cancer. This technique involves subjecting diseased tissues containing nanoparticles to near-infrared radiation (NIR), resulting in the production of heat and subsequent eradication of malignant cells. This method of localized heating is highly efficient in specifically targeting and treating cancers [146,147,148].
The success of PTT is highly influenced by the absorbance, radiative emission, stability, crystallinity, and size of the nanoparticle [149].
The synergic action of plasmonic gold nanoparticles in combination with MNPs has proved successful in targeting and treating cancer [150].
Iron oxide functionalized with alumina has shown therapeutic potential in treating nosocomial and antibiotic resistant bacterial infections [151]. The strategy was based on the magnetic properties of MNPs to form aggregates under an applied magnetic field and, with the help of alumina molecules attached at their surface, to be able to target bacterial species and then damage the formed bacterial population by means of NIR laser irradiation.
Radiolabeled molecule-coated MNP multifunctional hybrid nanostructures have been also reported as efficient PTT agents [152].
In light of the ideas presented above, PTT combined with MHT has offered promising results and a few more examples can be found in Table 1.

4.4. Magnetofection (MF)

Magnetofection is a technique that combines the use of magnetic nanoparticles with conventional transfection methods to improve the delivery of genetic material (e.g., DNA, RNA, or siRNA) into target cells. The goal is to enhance the efficiency and specificity of gene transfer by leveraging the magnetic properties of nanoparticles [153]. MF was employed to transport nucleic acids into cells for gene delivery purposes [153]. Magnetic nanoparticles (MNPs) that are attached to genetic material are inserted into the body and directed towards specific cells or tissues by using an external magnetic field capable of interacting with the MNPs. This technique improves the effectiveness of gene transfection, offering a potent tool for gene treatment and research purposes. Nucleic acid molecules can be indirectly attached to the MNPs surface using linkers [154] or directly through electrostatic interactions, especially in the case of polyethylenimine coatings [155,156], as presented in Table 1.
MF is very efficient due to its swift targeting and decreased cytotoxicity, providing notable benefits compared to conventional chemical transfection techniques [157,158,159].
Besides these applications, MNPs can be employed in efficient DNA isolation directly from biological samples. A higher separation capacity as compared to commercially available kits has been reported [160].

4.5. Magnetic Resonance Imaging (MRI)

Magnetic resonance imaging, or MRI, is a noninvasive medical imaging test that produces detailed images of almost every internal structure in the human body, including organs, bones, muscles, and blood vessels. In this regard MNPs were investigated as negative contrast agents suitable for tumor detection by means of MRI.
For example, iron oxide nanoparticles conjugated with various antibodies were used to image glioma [161,162], glioblastoma [163], breast [164], pancreatic [165], colorectal [166], prostate [167], and other cancers [168].
Multimodal cancer imaging involving MRI, computed tomography, positron emission tomography, single photon emission tomography, and near infrared fluorescence can be assessed by using MNPs [169].
Fluorescent-labelled MNPs offer versatile and powerful tools for visualizing biological structures and processes at molecular and cellular levels. Bioimaging using MNPs has shown promise in different preclinical studies for diagnosing diseases, monitoring disease progression, and evaluating treatment efficacy [170].
It is worth mentioning that SPIONs have already been used for human sentinel lymph node MRI to evaluate intermediate and high-risk prostate cancer [171,172]. An innovative method based on bimodal imaging using a new magnetic fluorescent hybrid tracer presented promising results in SLN detection as shown in the ongoing trial DRKS00032808 [173].
In addition, functionalizing MNPs with various radioisotopes has garnered interest for biomedical imaging, drug delivery, and targeted therapy. MNPs functionalized with radioisotopes serve as dual-modal imaging probes, combining the magnetic properties of MNPs with the radioactivity of the attached isotopes. This enables sensitive and quantitative imaging of biological processes, such as tumor targeting, inflammation detection, and organ-specific imaging [174]. Moreover, the addition of a targeting ligand (e.g., antibody or peptide) represents a promising strategy for targeted therapy and theranostic applications, especially in cancer [175].
Even if most of contrast agents approved by Food and Drug Administration are based on gadolinium, some MNPs are undergoing preclinical and clinical evaluation for safety and efficacy, with the goal of obtaining regulatory approval for clinical use. Plenty of commercial MNPs, used as agents involved in MRI applications, are available in the worldwide virtual market. However, a large portion of this category is still in the patenting stage. If this process will be successful, the range of MNPs employed in this field will increase considerably, since the use of MNPs has the potential to eliminate many of the current limitations (signal intensity, background signal, biocompatibility and toxicity, spatial resolution, biodistribution and clearance, magnetic field strength dependence, challenges in quantification) [176,177].
Table 1 contains other valuable examples of MNPs used as MRI agents and not only for cancer detection.

4.6. Magnetic Particle Imaging (MPI)

MPI technology was developed more than 10 years ago and is still evolving in the biomedical field [178]. The main relevant features of this dynamic domain are represented by harmless, cost-effective, and high-precision detection when combined with computed tomography [179]. MPI is based on the creation and superimposition of a static selection field and an oscillatory drive field by evaluating the non-linear magnetization properties of MNPs. For this specific type of application, the SPIONs represent the most suitable class of MNPs [180].
This technique acts as a true helper in diagnosing and treating various complex diseases, especially from the oncological and surgical field. One of the in vivo applications is represented by the quantitative analysis of MNPs by determining their concentration and distribution, which can improve imaging quality [181].
By combining MPI with MRI, one can investigate the molecular basis of some treatment mechanisms (Vivotrax from Magnetic Insight company, Alameda, CA, USA) [182]. Further examples can also be found in Table 1. The clinical translation of MNP-based imaging techniques is underway, with ongoing research focusing on safety, efficacy, and regulatory approval [183]. Continued research and development in this field holds great potential for advancing diagnostics, therapeutics, and personalized medicine.

4.7. Magnetic Separation (MS)

Among all these aforementioned methods, some authors reported the use of MNPs for cell separation. Their magnetic properties cause them to be recommended for use in various fields such as biotechnology, medicine, environmental sciences, and material sciences. MNPs can be used to remove contaminants from water or soil by selectively binding to pollutants, facilitating their easy removal under a magnetic field. In material science, MS using MNPs is used for material purification and separation, especially in nanotechnology. It can be applied in various industries for the separation of waste materials, making disposal and recycling processes more efficient.
MS is widely used in diagnostics, drug delivery, and bio separation processes. It can be employed to isolate specific cells, proteins, or nucleic acids.
For example, a tumor cell separation technique by means of dendrimer-assisted hydrophilic magnetic nanoparticles was implemented. These sensors were able to detect tumoral cells from blood samples, achieving a cell viability of 99% and a capture efficiency of nearly 80% [43].
MS by means of MNPs is very challenging because the properties of the nanoparticles should be carefully optimized, involving high synthesis and functionalization costs. However, the advantages offered by this method are of immeasurable value: high specificity and selectivity, a non-destructive and gentle separation process, and rapid and efficient separation [184].
Table 1 summarizes different remarkable examples of MNP use in biomedical applications found in the literature.
Table 1. A synthesis of biomedical applications of magnetic nanoparticles.
Table 1. A synthesis of biomedical applications of magnetic nanoparticles.
Application Magnetic Nanoparticles TypeDiameter (nm)Synthesis MethodAimFindings References
Magnetic Hyperthermia TherapyManganese and zinc ferrite 80 Polyol based methodInvestigation of NPs properties in vitroMelanoma cellular toxicity of Zn ferrites NPs[51]
Iron oxide19–50Polyol based methodInvestigation of NPs properties in vitroLung cancer cell lines toxicity [185]
SPIONs and magnetic-CXCR4 NPs20 & 250 Targeting CSCR4 receptor of LN229 cancer cellsMagnetic-CXCR4 were disposed around the cell membrane, magnetic hyperthermia has induced cytotoxic effects[91]
Targeted Drug DeliveryDoxorubicin and methotrexate dendritic chitosan PEG MNPs20–50Soft chemical methods and surface modificationsCo-delivery of doxorubicin and methotrexate to MCF-7 cellsHigh encapsulation efficiency of both drugs; biocompatibility with red blood cells; synergistic effects of the drug combination; reduced side effects; anti-cancer properties [186]
Polymeric nanoparticles loaded with curcumin, verapamil and oleylamine-coated superparamagnetic nanoparticles 280–287 Thermolysis method combined with emulsification/solvent evaporationMulti-modal cancer thermotherapy and chemotherapyHigh therapeutic efficacy in HepG2 cancer cells [187]
Doxorubicin and enzyme precursors covalently tethered on the silica-coated magnetite NPs50Chemical co-precipitation and surface modificationTumor drug delivery and imagingStable and non-toxic nanosystem; anti-cancer effect; real-time monitoring of intracellular drug release; tumor cell imaging [188]
Photothermal TherapyMNPs decorated with gold NPs5–20Co-precipitation and surface decorationTumor site targeting Apoptosis induction in cancer cells without damaging the healthy ones [151]
Maghemite nanoflowers40–200Microwave polyol processDeveloping an efficient MHT and PTT agentHigh crystallinity leads to increased magnetic properties [149]
Iron oxide NPs functionalized with 125I-c(RGDyK) radiolabeled peptide40Thermal decompositionTumor targeting by means of MRI and SPECTLow mononuclear phagocyte uptake; small amounts of MNPs accumulated in liver and spleen; therapeutic efficacy in glioblastoma[152]
MagnetofectionPolyethyleneimine MNPs loaded with siRNA10–15Chemical co-precipitation and electrostatic adsorption Intracellular gene silencing and imagingTherapeutic efficiency through apoptosis and autophagy induction in glioblastoma cells[189]
Polyethyleneimine iron oxide NPs loaded with gene vectors (pACTER-EGFP)10Co-precipitation and electrostatic adsorptionGene expression mediation Anti-tumor effect based on apoptosis on oral squamous cell carcinoma; high transfection efficiency[190]
Polyethyleneimine iron oxide NPs loaded with RNA sequences 100–156Thermal decompositionRNA deliveryEfficient protein knockdown in breast cancer[191]
Magnetic resonance imagingIron oxide6Non-toxic metal salts-based protocolReproductive system evaluation in vivoSertoli cells damage in blood testicular barrier, organ accumulation and cytotoxicity [192]
Iron oxide1–3Modified thermal decomposition methodTesting tissue permeable contrast agentsEffective brain imaging [193]
Anti-mesothelin antibody conjugated with silica-coated iron oxide NPs110–130Solvothermal reaction and incubation with antibodyPancreatic cancer in vitro and in vivo targeting and imagingEfficient targeting of SW1990 cells; High stability and nontoxicity both in vitro and in vivo[194]
Magnetic particle imagingIron oxide NPsUnknown Unknown Preclinical evaluation of chimeric antigen receptor T cells in glioblastoma Highly iron oxide NPs accumulation into tumor tissue[182]
SPIONs200Thermal decompositionPreclinical investigation of MNPs after intra-articular joints injectionNPs were retained at least for 42 days into the tissue making possible the imaging measurements due to their biocompatibility[195]
SPIONs5Unknown Ex vivo and preclinical evaluation of intraplaque hemorrhage in atherosclerosis Imaging data quality was improved in atherosclerosis analysis[196]
Magnetic separationMicrofluidic chip with MNPs200Soft lithographyTo develop a MNPs assisted microfluidic system for cells selection Low abundance cell capture and high recovery rate of the cells[197]
Dextran-, siloxane-, heparin- coated SPIONsUnknown Co-precipitation Labeling of islet cells with MNPs for separationHigh cellular viability after incubation with dextran—and heparin- coated MNPs; efficient method for islet cells purification [198]
Silica encapsulated iron oxide MNPs100–200Co-precipitationNon-selective separation of bacterial cellsHigh separation efficiency in acidic environment (pH 2.2); surface charge of the MNPs is dictating the microbial selectivity; efficient method for environment monitoring[199]
The progress in obtaining such nanostructures has led to the development of ones that nowadays are commercially available and serve various purposes, especially in biomedical field [200]. Many companies offer support in obtaining MNPs for drug delivery, diagnostics, and imaging applications. Merck KGaA (Darmastadt, Germany), Nano Research Elements Inc. (Dhanora Jattan, India), Fortis Life Sciences (Waltham, MA, USA), Nanografi Nano Technology (Istanbul, Turkey), Nanoshell LLC (North Salt Lake, UT, USA), Read International Corp. (Culver City, CA, USA), SkySpring Nanomaterials Inc. (Houston, TX, USA), Strem Chemicals Inc. (Newburyport, MA, USA), US Research Nanomaterials Inc. (Houston, TX, USA) are some of the companies that produce magnetic nanoparticles for the healthcare industry.

5. Conclusions and Future Perspectives

In recent years, there has been a substantial surge in research and development focused on magnetic nanoparticles intended for biomedical applications. The capacity to precisely control the size, shape, and surface characteristics of these nanoparticles has sparked widespread exploration of their potential uses. Magnetic nanoparticles offer distinct advantages, including their applicability in treatments like hyperthermia and the ability to be precisely directed to specific locations within the body using an alternating magnetic field. With a high surface-area-to-volume ratio, magnetic nanoparticles facilitate substantial substitution levels, making them well-suited for drug delivery. Furthermore, their incorporation into composites such as magnetic hydrogels or liposomes generating new classes of multifunctional hybrid nanostructures enhances their biocompatibility and broadens their potential in medical applications.
Several challenges persist with these systems. Firstly, the efficiency of magnetic nanoparticles (MNPs) is significantly influenced by their size and shape, necessitating the development of appropriate synthetic procedures and subsequent studies to ascertain optimal behavior of the resulting materials. Another concern is the aggregation of MNPs, which can impact their efficiency and pose potential toxicity issues. While toxicity studies must be conducted, it is worth noting that iron oxide nanoparticles, among those proposed for biomedical applications, are generally considered to be less toxic than many other nanoparticle types.
Continued research and innovation in these directions are likely to pave the way for more effective and personalized biomedical interventions using magnetic nanoparticles. Smart MNPs (SMNPs) possess additional functionalities beyond their basic magnetic properties. Such structures are designed to respond to specific stimuli or conditions, allowing for controlled and targeted TDD, MHT, MRI, MF applications. SMNPs can be designed to respond to specific local stimuli, such as changes in pH, temperature, or the presence of certain biomolecules. Moreover, external stimuli, such as magnetic fields, can allow the remote activation and manipulation of SMNPs within the body [201].
To establish the foundations for potentially clinically applicable results, it is imperative to conduct a literature review with a very strong interdisciplinary perspective. Therefore, the collaboration between nanomaterials and bioengineering can certainly provide synergistic effects in biomedical applications.

Author Contributions

Conceptualization, G.F.S. and R.I.S.; methodology, G.F.S.; software, R.I.S.; validation, G.F.S. and R.I.S.; formal analysis, G.F.S.; investigation, G.F.S.; resources, G.F.S.; data curation, R.I.S.; writing—original draft preparation, G.F.S. and R.I.S.; writing—review and editing, R.I.S.; visualization, G.F.S.; supervision, R.I.S.; project administration, R.I.S.; funding acquisition, R.I.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the following projects: WIDESPREAD-06-2020-ERA Chairs/H2020 ERA-Chair projects—grant agreement No. 952390/2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Shen, B.; Sun, S. Chemical Synthesis of Magnetic Nanoparticles for Permanent Magnet Applications. Chem. Eur. J. 2020, 26, 6757–6766. [Google Scholar] [CrossRef] [PubMed]
  2. Dave, S.; Dave, S.; Mathur, A.; Das, J. Biological Synthesis of Magnetic Nanoparticles. In Nanobiotechnology; Elsevier: Amsterdam, The Netherlands, 2021; pp. 225–234. [Google Scholar]
  3. Majidi, S.; Zeinali Sehrig, F.; Farkhani, S.M.; Soleymani Goloujeh, M.; Akbarzadeh, A. Current Methods for Synthesis of Magnetic Nanoparticles. Artif. Cells Nanomed. Biotechnol. 2016, 44, 722–734. [Google Scholar] [CrossRef] [PubMed]
  4. Fecht, H.J.; Hellstern, E.; Fu, Z.; Johnson, W.L. Nanocrystalline Metals Prepared by High-Energy Ball Milling. Metall. Trans. A 1990, 21, 2333–2337. [Google Scholar] [CrossRef]
  5. Vashist, S.K. Magnetic Nanoparticles-Based Biomedical and Bioanalytical Applications. J. Nanomed. Nanotechnol. 2013, 4, e130. [Google Scholar] [CrossRef]
  6. Habibullah, G.; Viktorova, J.; Ruml, T. Current Strategies for Noble Metal Nanoparticle Synthesis. Nanoscale Res. Lett. 2021, 16, 47. [Google Scholar] [CrossRef] [PubMed]
  7. Biehl, P.; von der Lühe, M.; Dutz, S.; Schacher, F. Synthesis, Characterization, and Applications of Magnetic Nanoparticles Featuring Polyzwitterionic Coatings. Polymers 2018, 10, 91. [Google Scholar] [CrossRef] [PubMed]
  8. Shin, D.N.; Matsuda, Y.; Bernstein, E.R. On the Iron Oxide Neutral Cluster Distribution in the Gas Phase. II. Detection through 118 Nm Single Photon Ionization. J. Chem. Phys. 2004, 120, 4157–4164. [Google Scholar] [CrossRef]
  9. Kurland, H.-D.; Grabow, J.; Staupendahl, G.; Andrä, W.; Dutz, S.; Bellemann, M.E. Magnetic Iron Oxide Nanopowders Produced by CO2 Laser Evaporation. J. Magn. Magn. Mater. 2007, 311, 73–77. [Google Scholar] [CrossRef]
  10. Yang, G.W. Laser Ablation in Liquids: Applications in the Synthesis of Nanocrystals. Prog. Mater. Sci. 2007, 52, 648–698. [Google Scholar] [CrossRef]
  11. Amendola, V.; Meneghetti, M. Laser Ablation Synthesis in Solution and Size Manipulation of Noble Metal Nanoparticles. Phys. Chem. Chem. Phys. 2009, 11, 3805. [Google Scholar] [CrossRef]
  12. Kawamura, G.; Alvarez, S.; Stewart, I.E.; Catenacci, M.; Chen, Z.; Ha, Y.-C. Production of Oxidation-Resistant Cu-Based Nanoparticles by Wire Explosion. Sci. Rep. 2015, 5, 18333. [Google Scholar] [CrossRef] [PubMed]
  13. Song, K.; Kim, W.; Suh, C.-Y.; Shin, D.; Ko, K.-S.; Ha, K. Magnetic Iron Oxide Nanoparticles Prepared by Electrical Wire Explosion for Arsenic Removal. Powder Technol. 2013, 246, 572–574. [Google Scholar] [CrossRef]
  14. Kotov, Y.A. Electric Explosion of Wires as a Method for Preparation of Nanopowders. J. Nanoparticle Res. 2003, 5, 539–550. [Google Scholar] [CrossRef]
  15. Grammatikopoulos, P.; Steinhauer, S.; Vernieres, J.; Singh, V.; Sowwan, M. Nanoparticle Design by Gas-Phase Synthesis. Adv. Phys. X 2016, 1, 81–100. [Google Scholar] [CrossRef]
  16. Hammad, M.; Hardt, S.; Mues, B.; Salamon, S.; Landers, J.; Slabu, I.; Wende, H.; Schulz, C.; Wiggers, H. Gas-Phase Synthesis of Iron Oxide Nanoparticles for Improved Magnetic Hyperthermia Performance. J. Alloys Compd. 2020, 824, 153814. [Google Scholar] [CrossRef]
  17. Flores-Rojas, G.G.; López-Saucedo, F.; Vera-Graziano, R.; Mendizabal, E.; Bucio, E. Magnetic Nanoparticles for Medical Applications: Updated Review. Macromol 2022, 2, 374–390. [Google Scholar] [CrossRef]
  18. Hariani, P.L.; Faizal, M.; Ridwan, R.; Marsi, M.; Setiabudidaya, D. Synthesis and Properties of Fe3O4 Nanoparticles by Co-Precipitation Method to Removal Procion Dye. Int. J. Environ. Sci. Dev. 2013, 4, 336. [Google Scholar] [CrossRef]
  19. Ali, A.; Shah, T.; Ullah, R.; Zhou, P.; Guo, M.; Ovais, M.; Tan, Z.; Rui, Y. Review on Recent Progress in Magnetic Nanoparticles: Synthesis, Characterization, and Diverse Applications. Front. Chem. 2021, 9, 629054. [Google Scholar] [CrossRef]
  20. Kritika; Roy, I. Therapeutic Applications of Magnetic Nanoparticles: Recent Advances. Mater. Adv. 2022, 3, 7425–7444. [Google Scholar] [CrossRef]
  21. Slimani, S.; Meneghini, C.; Abdolrahimi, M.; Talone, A.; Murillo, J.P.M.; Barucca, G.; Yaacoub, N.; Imperatori, P.; Illés, E.; Smari, M.; et al. Spinel Iron Oxide by the Co-Precipitation Method: Effect of the Reaction Atmosphere. Appl. Sci. 2021, 11, 5433. [Google Scholar] [CrossRef]
  22. Ajeesha, T.; Ashwini, A.; George, M.; Manikandan, A.; Mary, J.A.; Slimani, Y.; Almessiere, M.A.; Baykal, A. Nickel Substituted MgFe2O4 Nanoparticles via Co-Precipitation Method for Photocatalytic Applications. Phys. B Condens. Matter 2021, 606, 412660. [Google Scholar] [CrossRef]
  23. Dadfar, S.M.; Roemhild, K.; Drude, N.I.; von Stillfried, S.; Knüchel, R.; Kiessling, F.; Lammers, T. Iron Oxide Nanoparticles: Diagnostic, Therapeutic and Theranostic Applications. Adv. Drug Deliv. Rev. 2019, 138, 302–325. [Google Scholar] [CrossRef]
  24. Farinha, P.; Coelho, J.M.P.; Reis, C.P.; Gaspar, M.M. A Comprehensive Updated Review on Magnetic Nanoparticles in Diagnostics. Nanomaterials 2021, 11, 3432. [Google Scholar] [CrossRef]
  25. Chen, J.P.; Sorensen, C.M.; Klabunde, K.J.; Hadjipanayis, G.C.; Devlin, E.; Kostikas, A. Size-Dependent Magnetic Properties of MnFe2O4 fine particles synthesized by coprecipitation. Phys. Rev. B 1996, 54, 9288–9296. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, Q.; Rondinone, A.J.; Chakoumakos, B.C.; John Zhang, Z. Synthesis of Superparamagnetic MgFe2O4 Nanoparticles by Coprecipitation. J. Magn. Magn. Mater. 1999, 194, 1–7. [Google Scholar] [CrossRef]
  27. Niculescu, A.-G.; Chircov, C.; Grumezescu, A.M. Magnetite Nanoparticles: Synthesis Methods—A Comparative Review. Methods 2022, 199, 16–27. [Google Scholar] [CrossRef] [PubMed]
  28. Park, J.; An, K.; Hwang, Y.; Park, J.-G.; Noh, H.-J.; Kim, J.-Y.; Park, J.-H.; Hwang, N.-M.; Hyeon, T. Ultra-Large-Scale Syntheses of Monodisperse Nanocrystals. Nat. Mater. 2004, 3, 891–895. [Google Scholar] [CrossRef] [PubMed]
  29. Jeraal, M.I.; Roberts, K.J.; McRobbie, I.; Harbottle, D. Assessment of the Thermal Degradation of Sodium Lauroyl Isethionate Using Predictive Isoconversional Kinetics and a Temperature-Resolved Analysis of Evolved Gases. Ind. Eng. Chem. Res. 2019, 58, 8112–8122. [Google Scholar] [CrossRef]
  30. Stojanovic, B.D.; Dzunuzovic, A.S.; Ilic, N.I. Review of Methods for the Preparation of Magnetic Metal Oxides. In Magnetic, Ferroelectric, and Multiferroic Metal Oxides; Elsevier: Amsterdam, The Netherlands, 2018; pp. 333–359. [Google Scholar]
  31. Nicola (Crişan), R.; Costişor, O.; Ianăşi, C.; Lazău, R.; Săcărescu, L.; Nižňanský, D.; Ercuţa, A.; Putz, A.-M.; Savii, C. Fractal Surface Maghemite Nanoparticles Prepared by Co-Precipitation: The Influence of Iron Concentration and Base Nature. Stud. Univ. Babeș-Bolyai Chem. 2018, 63, 15–29. [Google Scholar] [CrossRef]
  32. Jović Orsini, N.; Babić-Stojić, B.; Spasojević, V.; Calatayud, M.P.; Cvjetićanin, N.; Goya, G.F. Magnetic and Power Absorption Measurements on Iron Oxide Nanoparticles Synthesized by Thermal Decomposition of Fe(Acac)3. J. Magn. Magn. Mater. 2018, 449, 286–296. [Google Scholar] [CrossRef]
  33. Patsula, V.; Kosinová, L.; Lovrić, M.; Ferhatovic Hamzić, L.; Rabyk, M.; Konefal, R.; Paruzel, A.; Šlouf, M.; Herynek, V.; Gajović, S.; et al. Superparamagnetic Fe3O4 Nanoparticles: Synthesis by Thermal Decomposition of Iron(III) Glucuronate and Application in Magnetic Resonance Imaging. ACS Appl. Mater. Interfaces 2016, 8, 7238–7247. [Google Scholar] [CrossRef] [PubMed]
  34. Lu, A.; Salabas, E.L.; Schüth, F. Magnetic Nanoparticles: Synthesis, Protection, Functionalization, and Application. Angew. Chem. Int. Ed. 2007, 46, 1222–1244. [Google Scholar] [CrossRef] [PubMed]
  35. Smith, T.W.; Wychick, D. Colloidal Iron Dispersions Prepared via the Polymer-Catalyzed Decomposition of Iron Pentacarbonyl. J. Phys. Chem. 1980, 84, 1621–1629. [Google Scholar] [CrossRef]
  36. Huber, D.L. Synthesis, Properties, and Applications of Iron Nanoparticles. Small 2005, 1, 482–501. [Google Scholar] [CrossRef] [PubMed]
  37. Jana, N.R.; Chen, Y.; Peng, X. Size- and Shape-Controlled Magnetic (Cr, Mn, Fe, Co, Ni) Oxide Nanocrystals via a Simple and General Approach. Chem. Mater. 2004, 16, 3931–3935. [Google Scholar] [CrossRef]
  38. Pérez-Yarza, E.G.; Badía, X.; Badiola, C.; Cobos, N.; Garde, J.; Ibero, M.; Villa, J.R. Development and Validation of a Questionnaire to Assess Asthma Control in Pediatrics. Pediatr. Pulmonol. 2009, 44, 54–63. [Google Scholar] [CrossRef] [PubMed]
  39. 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]
  40. Cheah, P.; Qu, J.; Li, Y.; Cao, D.; Zhu, X.; Zhao, Y. The Key Role of Reaction Temperature on a Polyol Synthesis of Water-Dispersible Iron Oxide Nanoparticles. J. Magn. Magn. Mater. 2021, 540, 168481. [Google Scholar] [CrossRef]
  41. Faraji, M.; Yamini, Y.; Rezaee, M. Magnetic Nanoparticles: Synthesis, Stabilization, Functionalization, Characterization, and Applications. J. Iran. Chem. Soc. 2010, 7, 1–37. [Google Scholar] [CrossRef]
  42. Materón, E.M.; Miyazaki, C.M.; Carr, O.; Joshi, N.; Picciani, P.H.S.; Dalmaschio, C.J.; Davis, F.; Shimizu, F.M. Magnetic Nanoparticles in Biomedical Applications: A Review. Appl. Surf. Sci. Adv. 2021, 6, 100163. [Google Scholar] [CrossRef]
  43. Zhang, P.; Zhang, Y.; Gao, M.; Zhang, X. Dendrimer-Assisted Hydrophilic Magnetic Nanoparticles as Sensitive Substrates for Rapid Recognition and Enhanced Isolation of Target Tumor Cells. Talanta 2016, 161, 925–931. [Google Scholar] [CrossRef] [PubMed]
  44. Gan, Y.X.; Jayatissa, A.H.; Yu, Z.; Chen, X.; Li, M. Hydrothermal Synthesis of Nanomaterials. J. Nanomater. 2020, 2020, 8917013. [Google Scholar] [CrossRef]
  45. Reddy, L.H.; Arias, J.L.; Nicolas, J.; Couvreur, P. Magnetic Nanoparticles: Design and Characterization, Toxicity and Biocompatibility, Pharmaceutical and Biomedical Applications. Chem. Rev. 2012, 112, 5818–5878. [Google Scholar] [CrossRef] [PubMed]
  46. Li, J.; Zheng, L.; Cai, H.; Sun, W.; Shen, M.; Zhang, G.; Shi, X. Polyethyleneimine-Mediated Synthesis of Folic Acid-Targeted Iron Oxide Nanoparticles for in Vivo Tumor MR Imaging. Biomaterials 2013, 34, 8382–8392. [Google Scholar] [CrossRef] [PubMed]
  47. Li, G.; Jiang, Y.; Huang, K.; Ding, P.; Chen, J. Preparation and Properties of Magnetic Fe3O4–Chitosan Nanoparticles. J. Alloys Compd. 2008, 466, 451–456. [Google Scholar] [CrossRef]
  48. Ansari, S.; Ficiarà, E.; Ruffinatti, F.; Stura, I.; Argenziano, M.; Abollino, O.; Cavalli, R.; Guiot, C.; D’Agata, F. Magnetic Iron Oxide Nanoparticles: Synthesis, Characterization and Functionalization for Biomedical Applications in the Central Nervous System. Materials 2019, 12, 465. [Google Scholar] [CrossRef] [PubMed]
  49. Zahid, M.; Nadeem, N.; Hanif, M.A.; Bhatti, I.A.; Bhatti, H.N.; Mustafa, G. Metal Ferrites and Their Graphene-Based Nanocomposites: Synthesis, Characterization, and Applications in Wastewater Treatment. In Magnetic Nanostructures; Springer: Berlin/Heidelberg, Germany, 2019; pp. 181–212. [Google Scholar]
  50. Ruz, P.; Sudarsan, V. Polyol Method for Synthesis of Nanomaterials. In Handbook on Synthesis Strategies for Advanced Materials; Springer: Berlin/Heidelberg, Germany, 2021; pp. 293–332. [Google Scholar]
  51. Iacovita, C.; Florea, A.; Scorus, L.; Pall, E.; Dudric, R.; Moldovan, A.I.; Stiufiuc, R.; Tetean, R.; Lucaciu, C.M. Hyperthermia, Cytotoxicity, and Cellular Uptake Properties of Manganese and Zinc Ferrite Magnetic Nanoparticles Synthesized by a Polyol-Mediated Process. Nanomaterials 2019, 9, 1489. [Google Scholar] [CrossRef]
  52. Tartaj, P.; González-Carreño, T.; Bomatí-Miguel, O.; Serna, C.J.; Bonville, P. Magnetic Behavior of Superparamagnetic Fe Nanocrystals Confined inside Submicron-Sized Spherical Silica Particles. Phys. Rev. B 2004, 69, 094401. [Google Scholar] [CrossRef]
  53. Oh, A.H.; Park, H.-Y.; Jung, Y.-G.; Choi, S.-C.; An, G.S. Synthesis of Fe3O4 Nanoparticles of Various Size via the Polyol Method. Ceram. Int. 2020, 46, 10723–10728. [Google Scholar] [CrossRef]
  54. Hasany, S.F.; Ahmed, I.; Rajan, J.; Rehman, A. Systematic Review of the Preparation Techniques of Iron Oxide Magnetic Nanoparticles. Nanosci. Nanotechnol. 2013, 2, 148–158. [Google Scholar] [CrossRef]
  55. Pineda, M.; Torres, S.; López, L.; Enríquez-Medrano, F.; de León, R.; Fernández, S.; Saade, H.; López, R. Chitosan-Coated Magnetic Nanoparticles Prepared in One-Step by Precipitation in a High-Aqueous Phase Content Reverse Microemulsion. Molecules 2014, 19, 9273–9287. [Google Scholar] [CrossRef]
  56. López Pérez, J.A.; López Quintela, M.A.; Mira, J.; Rivas, J.; Charles, S.W. Advances in the Preparation of Magnetic Nanoparticles by the Microemulsion Method. J. Phys. Chem. B 1997, 101, 8045–8047. [Google Scholar] [CrossRef]
  57. Salvador, M.; Gutiérrez, G.; Noriega, S.; Moyano, A.; Blanco-López, M.C.; Matos, M. Microemulsion Synthesis of Superparamagnetic Nanoparticles for Bioapplications. Int. J. Mol. Sci. 2021, 22, 427. [Google Scholar] [CrossRef]
  58. Bomatí-Miguel, O.; Mazeina, L.; Navrotsky, A.; Veintemillas-Verdaguer, S. Calorimetric Study of Maghemite Nanoparticles Synthesized by Laser-Induced Pyrolysis. Chem. Mater. 2008, 20, 591–598. [Google Scholar] [CrossRef]
  59. Tahir, M.B.; Rafique, M.; Rafique, M.S.; Nawaz, T.; Rizwan, M.; Tanveer, M. Photocatalytic Nanomaterials for Degradation of Organic Pollutants and Heavy Metals. In Nanotechnology and Photocatalysis for Environmental Applications; Elsevier: Amsterdam, The Netherlands, 2020; pp. 119–138. [Google Scholar]
  60. Wang, S.; Gao, L. Laser-Driven Nanomaterials and Laser-Enabled Nanofabrication for Industrial Applications. In Industrial Applications of Nanomaterials; Elsevier: Amsterdam, The Netherlands, 2019; pp. 181–203. [Google Scholar]
  61. Woodard, A.; Xu, L.; Barragan, A.A.; Nava, G.; Wong, B.M.; Mangolini, L. On the Non-thermal Plasma Synthesis of Nickel Nanoparticles. Plasma Process. Polym. 2018, 15, 1700104. [Google Scholar] [CrossRef]
  62. Kelgenbaeva, Z.; Murzubraimov, B.; Kozlovsky, A.; Adil Akai Tegin, R.; Turdubai Kyzy, A.; Murzabekova, E.; Aidaraliev, J.; Dyusheeva, B. Magnetic Nanoparticles Preparation by Chemical Reduction for Biomedical Applications. EPJ Web Conf. 2019, 201, 01002. [Google Scholar] [CrossRef]
  63. Pascal, C.; Pascal, J.L.; Favier, F.; Elidrissi Moubtassim, M.L.; Payen, C. Electrochemical Synthesis for the Control of γ-Fe2O3 Nanoparticle Size. Morphology, Microstructure, and Magnetic Behavior. Chem. Mater. 1999, 11, 141–147. [Google Scholar] [CrossRef]
  64. Gopi, D.; Thameem Ansari, M.; Kavitha, L. Electrochemical Synthesis and Characterization of Cubic Magnetite Nanoparticle in Aqueous Ferrous Perchlorate Medium. Arab. J. Chem. 2016, 9, S829–S834. [Google Scholar] [CrossRef]
  65. Park, J.H.; Ahn, H.S. Electrochemical Synthesis of Multimetallic Nanoparticles and Their Application in Alkaline Oxygen Reduction Catalysis. Appl. Surf. Sci. 2020, 504, 144517. [Google Scholar] [CrossRef]
  66. Riaz, S.; Ashraf, R.; Akbar, A.; Naseem, S. Microwave Assisted Iron Oxide Nanoparticles—Structural and Magnetic Properties. IEEE Trans. Magn. 2014, 50, 2201504. [Google Scholar] [CrossRef]
  67. da Silva Assis, M.B.; Werneck, I.H.S.R.; de Moraes, G.N.; Semaan, F.S.; Pacheco Pereira, R. Citrate-Capped Iron Oxide Nanoparticles: Ultrasound-Assisted Synthesis, Structure and Thermal Properties. Mater. Res. Express 2019, 6, 045064. [Google Scholar] [CrossRef]
  68. Verma, R.; Pathak, S.; Srivastava, A.K.; Prawer, S.; Tomljenovic-Hanic, S. ZnO Nanomaterials: Green Synthesis, Toxicity Evaluation and New Insights in Biomedical Applications. J. Alloys Compd. 2021, 876, 160175. [Google Scholar] [CrossRef]
  69. Komeili, A. Molecular Mechanisms of Compartmentalization and Biomineralization in Magnetotactic Bacteria. FEMS Microbiol. Rev. 2012, 36, 232–255. [Google Scholar] [CrossRef]
  70. Lenders, J.J.M.; Altan, C.L.; Bomans, P.H.H.; Arakaki, A.; Bucak, S.; de With, G.; Sommerdijk, N.A.J.M. A Bioinspired Coprecipitation Method for the Controlled Synthesis of Magnetite Nanoparticles. Cryst. Growth Des. 2014, 14, 5561–5568. [Google Scholar] [CrossRef]
  71. Gul, S.; Khan, S.B.; Rehman, I.U.; Khan, M.A.; Khan, M.I. A Comprehensive Review of Magnetic Nanomaterials Modern Day Theranostics. Front. Mater. 2019, 6, 179. [Google Scholar] [CrossRef]
  72. Zhang, Q.; Yang, X.; Guan, J. Applications of Magnetic Nanomaterials in Heterogeneous Catalysis. ACS Appl. Nano Mater. 2019, 2, 4681–4697. [Google Scholar] [CrossRef]
  73. Duan, M.; Shapter, J.G.; Qi, W.; Yang, S.; Gao, G. Recent Progress in Magnetic Nanoparticles: Synthesis, Properties, and Applications. Nanotechnology 2018, 29, 452001. [Google Scholar] [CrossRef] [PubMed]
  74. Mutharani, B.; Tsai, H.-C.; Lai, J.-Y.; Chen, S.-M. Protein-Assisted Biomimetic Synthesis of Nanoscale Gadolinium-Integrated Polypyrrole for Synergetic and Ultrasensitive Electrochemical Assays of Nicardipine in Biological Samples. Anal. Chim. Acta 2022, 1199, 339567. [Google Scholar] [CrossRef] [PubMed]
  75. Nadaf, S.; Jena, G.K.; Rarokar, N.; Gurav, N.; Ayyanar, M.; Prasad, S.; Gurav, S. Biogenic and Biomimetic Functionalized Magnetic Nanosystem: Synthesis, Properties, and Biomedical Applications. Hybrid Adv. 2023, 3, 100038. [Google Scholar] [CrossRef]
  76. Suzuki, M.; Shinkai, M.; Kamihira, M.; Kobayashi, T. Preparation and Characteristics of Magnetite-Labelled Antibody with the Use of Poly(Ethylene Glycol) Derivatives. Biotechnol. Appl. Biochem. 1995, 21, 335–345. [Google Scholar] [CrossRef] [PubMed]
  77. Lee, J.; Isobe, T.; Senna, M. Preparation of Ultrafine Fe3O4 Particles by Precipitation in the Presence of PVA at High PH. J. Colloid Interface Sci. 1996, 177, 490–494. [Google Scholar] [CrossRef]
  78. Gómez-Lopera, S.A.; Plaza, R.C.; Delgado, A.V. Synthesis and Characterization of Spherical Magnetite/Biodegradable Polymer Composite Particles. J. Colloid Interface Sci. 2001, 240, 40–47. [Google Scholar] [CrossRef]
  79. Liu, X.; Xing, J.; Guan, Y.; Shan, G.; Liu, H. Synthesis of Amino-Silane Modified Superparamagnetic Silica Supports and Their Use for Protein Immobilization. Colloids Surf. A Physicochem. Eng. Asp. 2004, 238, 127–131. [Google Scholar] [CrossRef]
  80. Liu, X.; Ma, Z.; Xing, J.; Liu, H. Preparation and Characterization of Amino–Silane Modified Superparamagnetic Silica Nanospheres. J. Magn. Magn. Mater. 2004, 270, 1–6. [Google Scholar] [CrossRef]
  81. Levy, L.; Sahoo, Y.; Kim, K.-S.; Bergey, E.J.; Prasad, P.N. Nanochemistry: Synthesis and Characterization of Multifunctional Nanoclinics for Biological Applications. Chem. Mater. 2002, 14, 3715–3721. [Google Scholar] [CrossRef]
  82. LaConte, L.; Nitin, N.; Bao, G. Magnetic Nanoparticle Probes. Mater. Today 2005, 8, 32–38. [Google Scholar] [CrossRef]
  83. Berry, C.C.; Charles, S.; Wells, S.; Dalby, M.J.; Curtis, A.S.G. The Influence of Transferrin Stabilised Magnetic Nanoparticles on Human Dermal Fibroblasts in Culture. Int. J. Pharm. 2004, 269, 211–225. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, Y.; Kohler, N.; Zhang, M. Surface Modification of Superparamagnetic Magnetite Nanoparticles and Their Intracellular Uptake. Biomaterials 2002, 23, 1553–1561. [Google Scholar] [CrossRef]
  85. Kinoshita, T.; Seino, S.; Okitsu, K.; Nakayama, T.; Nakagawa, T.; Yamamoto, T.A. Magnetic Evaluation of Nanostructure of Gold–Iron Composite Particles Synthesized by a Reverse Micelle Method. J. Alloys Compd. 2003, 359, 46–50. [Google Scholar] [CrossRef]
  86. Kobayashi, Y.; Horie, M.; Konno, M.; Rodríguez-González, B.; Liz-Marzán, L.M. Preparation and Properties of Silica-Coated Cobalt Nanoparticles. J. Phys. Chem. B 2003, 107, 7420–7425. [Google Scholar] [CrossRef]
  87. Malhotra, N.; Lee, J.-S.; Liman, R.A.D.; Ruallo, J.M.S.; Villaflores, O.B.; Ger, T.-R.; Hsiao, C.-D. Potential Toxicity of Iron Oxide Magnetic Nanoparticles: A Review. Molecules 2020, 25, 3159. [Google Scholar] [CrossRef] [PubMed]
  88. Wu, K.; Liu, J.; Chugh, V.K.; Liang, S.; Saha, R.; Krishna, V.D.; Cheeran, M.C.-J.; Wang, J.-P. Magnetic Nanoparticles and Magnetic Particle Spectroscopy-Based Bioassays: A 15 Year Recap. Nano Futures 2022, 6, 022001. [Google Scholar] [CrossRef] [PubMed]
  89. Krishnan, K.M.; Pakhomov, A.B.; Bao, Y.; Blomqvist, P.; Chun, Y.; Gonzales, M.; Griffin, K.; Ji, X.; Roberts, B.K. Nanomagnetism and Spin Electronics: Materials, Microstructure and Novel Properties. J. Mater. Sci. 2006, 41, 793–815. [Google Scholar] [CrossRef]
  90. Nkurikiyimfura, I.; Wang, Y.; Safari, B.; Nshingabigwi, E. Temperature-Dependent Magnetic Properties of Magnetite Nanoparticles Synthesized via Coprecipitation Method. J. Alloys Compd. 2020, 846, 156344. [Google Scholar] [CrossRef]
  91. Yeap, S.P.; Lim, J.; Ooi, B.S.; Ahmad, A.L. Agglomeration, Colloidal Stability, and Magnetic Separation of Magnetic Nanoparticles: Collective Influences on Environmental Engineering Applications. J. Nanoparticle Res. 2017, 19, 368. [Google Scholar] [CrossRef]
  92. Saville, S.L.; Stone, R.C.; Qi, B.; Mefford, O.T. Investigation of the Stability of Magnetite Nanoparticles Functionalized with Catechol Based Ligands in Biological Media. J. Mater. Chem. 2012, 22, 24909. [Google Scholar] [CrossRef]
  93. Zahraei, M.; Marciello, M.; Lazaro-Carrillo, A.; Villanueva, A.; Herranz, F.; Talelli, M.; Costo, R.; Monshi, A.; Shahbazi-Gahrouei, D.; Amirnasr, M.; et al. Versatile Theranostics Agents Designed by Coating Ferrite Nanoparticles with Biocompatible Polymers. Nanotechnology 2016, 27, 255702. [Google Scholar] [CrossRef]
  94. Available online: https://imagej.net/ij/ (accessed on 14 January 2024).
  95. Fabris, F.; Lima, E.; De Biasi, E.; Troiani, H.E.; Vásquez Mansilla, M.; Torres, T.E.; Fernández Pacheco, R.; Ibarra, M.R.; Goya, G.F.; Zysler, R.D.; et al. Controlling the Dominant Magnetic Relaxation Mechanisms for Magnetic Hyperthermia in Bimagnetic Core–Shell Nanoparticles. Nanoscale 2019, 11, 3164–3172. [Google Scholar] [CrossRef]
  96. Von Pushkar Singh, J. Experimental Investigations of Plasmon Induced Catalytic Reactions Using TERS. Ph.D. Thesis, University of Jena, Jena, Germany, 1976. [Google Scholar]
  97. Li, G.; Yang, Z.; Pei, Z.; Li, Y.; Yang, R.; Liang, Y.; Zhang, Q.; Jiang, G. Single-Particle Analysis of Micro/Nanoplastics by SEM-Raman Technique. Talanta 2022, 249, 123701. [Google Scholar] [CrossRef]
  98. Singh, R.K.; Patel, K.D.; Lee, J.H.; Lee, E.-J.; Kim, J.-H.; Kim, T.-H.; Kim, H.-W. Potential of Magnetic Nanofiber Scaffolds with Mechanical and Biological Properties Applicable for Bone Regeneration. PLoS ONE 2014, 9, e91584. [Google Scholar] [CrossRef]
  99. Kang, S.; Baskaran, R.; Ozlu, B.; Davaa, E.; Kim, J.J.; Shim, B.S.; Yang, S.-G. T1-Positive Mn2+-Doped Multi-Stimuli Responsive Poly(L-DOPA) Nanoparticles for Photothermal and Photodynamic Combination Cancer Therapy. Biomedicines 2020, 8, 417. [Google Scholar] [CrossRef]
  100. Silva, L.P.; Lacava, Z.G.M.; Buske, N.; Morais, P.C.; Azevedo, R.B. Atomic Force Microscopy and Transmission Electron Microscopy of Biocompatible Magnetic Fluids: A Comparative Analysis. J. Nanoparticle Res. 2004, 6, 209–213. [Google Scholar] [CrossRef]
  101. Foner, S. Versatile and Sensitive Vibrating-Sample Magnetometer. Rev. Sci. Instrum. 1959, 30, 548–557. [Google Scholar] [CrossRef]
  102. Sandler, S.E.; Fellows, B.; Mefford, O.T. Best Practices for Characterization of Magnetic Nanoparticles for Biomedical Applications. Anal. Chem. 2019, 91, 14159–14169. [Google Scholar] [CrossRef] [PubMed]
  103. Teja, A.S.; Koh, P.-Y. Synthesis, Properties, and Applications of Magnetic Iron Oxide Nanoparticles. Prog. Cryst. Growth Charact. Mater. 2009, 55, 22–45. [Google Scholar] [CrossRef]
  104. Jahan, S.; Alias, Y.B.; Abu Bakar, A.F.B.; Yusoff, I. Bin Ionic Release Behavior of Polymer-Coated and Uncoated Metal Nanoparticles (MNPs) in Various Conditions: Effects of Particle Shape, Size, and Natural Media Reactivity. Colloid Polym. Sci. 2017, 295, 1961–1971. [Google Scholar] [CrossRef]
  105. Mamontova, E.; Favier, I.; Pla, D.; Gómez, M. Organometallic Interactions between Metal Nanoparticles and Carbon-Based Molecules: A Surface Reactivity Rationale. In Advances in Organometallic Chemistry; Academic Press: Cambridge, MA, USA, 2022; pp. 43–103. [Google Scholar]
  106. Le Ru, E.C.; Etchegoin, P.G. Principles of Surface-Enhanced Raman Spectroscopy: And Related Plasmonic Effects; Elsevier: Amsterdam, The Netherlands, 2009; ISBN 9780444527790. [Google Scholar]
  107. Soler, M.A.G.; Qu, F. Raman Spectroscopy of Iron Oxide Nanoparticles. In Raman Spectroscopy for Nanomaterials Characterization; Springer: Berlin/Heidelberg, Germany, 2012; pp. 379–416. [Google Scholar]
  108. Testa-Anta, M.; Ramos-Docampo, M.A.; Comesaña-Hermo, M.; Rivas-Murias, B.; Salgueiriño, V. Raman Spectroscopy to Unravel the Magnetic Properties of Iron Oxide Nanocrystals for Bio-Related Applications. Nanoscale Adv. 2019, 1, 2086–2103. [Google Scholar] [CrossRef]
  109. Slavov, L.; Abrashev, M.V.; Merodiiska, T.; Gelev, C.; Vandenberghe, R.E.; Markova-Deneva, I.; Nedkov, I. Raman Spectroscopy Investigation of Magnetite Nanoparticles in Ferrofluids. J. Magn. Magn. Mater. 2010, 322, 1904–1911. [Google Scholar] [CrossRef]
  110. Bustos, A.R.M.; Encinar, J.R.; Sanz-Medel, A. Mass Spectrometry for the Characterisation of Nanoparticles. Anal. Bioanal. Chem. 2013, 405, 5637–5643. [Google Scholar] [CrossRef]
  111. Yan, B.; Jeong, Y.; Mercante, L.A.; Tonga, G.Y.; Kim, C.; Zhu, Z.-J.; Vachet, R.W.; Rotello, V.M. Characterization of Surface Ligands on Functionalized Magnetic Nanoparticles Using Laser Desorption/Ionization Mass Spectrometry (LDI-MS). Nanoscale 2013, 5, 5063. [Google Scholar] [CrossRef] [PubMed]
  112. Olesik, J.W. Elemental Analysis Using ICP-OES and ICP/MS. Anal. Chem. 1991, 63, 12A–21A. [Google Scholar] [CrossRef]
  113. Borowska, M.; Jankowski, K. Basic and Advanced Spectrometric Methods for Complete Nanoparticles Characterization in Bio/Eco Systems: Current Status and Future Prospects. Anal. Bioanal. Chem. 2023, 415, 4023–4038. [Google Scholar] [CrossRef] [PubMed]
  114. Mourdikoudis, S.; Pallares, R.M.; Thanh, N.T.K. Characterization Techniques for Nanoparticles: Comparison and Complementarity upon Studying Nanoparticle Properties. Nanoscale 2018, 10, 12871–12934. [Google Scholar] [CrossRef]
  115. Shasha, C.; Krishnan, K.M. Nonequilibrium Dynamics of Magnetic Nanoparticles with Applications in Biomedicine. Adv. Mater. 2021, 33, 1904131. [Google Scholar] [CrossRef] [PubMed]
  116. Soetaert, F.; Korangath, P.; Serantes, D.; Fiering, S.; Ivkov, R. Cancer Therapy with Iron Oxide Nanoparticles: Agents of Thermal and Immune Therapies. Adv. Drug Deliv. Rev. 2020, 163–164, 65–83. [Google Scholar] [CrossRef] [PubMed]
  117. Stueber, D.D.; Villanova, J.; Aponte, I.; Xiao, Z.; Colvin, V.L. Magnetic Nanoparticles in Biology and Medicine: Past, Present, and Future Trends. Pharmaceutics 2021, 13, 943. [Google Scholar] [CrossRef]
  118. Liu, J.; Su, D.; Wu, K.; Wang, J.-P. High-Moment Magnetic Nanoparticles. J. Nanoparticle Res. 2020, 22, 66. [Google Scholar] [CrossRef]
  119. Luo, K.; Zhao, J.; Jia, C.; Chen, Y.; Zhang, Z.; Zhang, J.; Huang, M.; Wang, S. Integration of Fe3O4 with Bi2S3 for Multi-Modality Tumor Theranostics. ACS Appl. Mater. Interfaces 2020, 12, 22650–22660. [Google Scholar] [CrossRef]
  120. Gobbo, O.L.; Sjaastad, K.; Radomski, M.W.; Volkov, Y.; Prina-Mello, A. Magnetic Nanoparticles in Cancer Theranostics. Theranostics 2015, 5, 1249–1263. [Google Scholar] [CrossRef]
  121. Belyanina, I.; Kolovskaya, O.; Zamay, S.; Gargaun, A.; Zamay, T.; Kichkailo, A. Targeted Magnetic Nanotheranostics of Cancer. Molecules 2017, 22, 975. [Google Scholar] [CrossRef]
  122. Peiravi, M.; Eslami, H.; Ansari, M.; Zare-Zardini, H. Magnetic Hyperthermia: Potentials and Limitations. J. Indian Chem. Soc. 2022, 99, 100269. [Google Scholar] [CrossRef]
  123. Freitas, M.; Nouws, H.P.A.; Keating, E.; Delerue-Matos, C. High-Performance Electrochemical Immunomagnetic Assay for Breast Cancer Analysis. Sens. Actuators B Chem. 2020, 308, 127667. [Google Scholar] [CrossRef]
  124. Kumar, C.S.S.R.; Mohammad, F. Magnetic Nanomaterials for Hyperthermia-Based Therapy and Controlled Drug Delivery. Adv. Drug Deliv. Rev. 2011, 63, 789–808. [Google Scholar] [CrossRef]
  125. Gilchrist, R.K.; Medal, R.; Shorey, W.D.; Hanselman, R.C.; Parrott, J.C.; Taylor, C.B. Selective Inductive Heating of Lymph Nodes. Ann. Surg. 1957, 146, 596–606. [Google Scholar] [CrossRef]
  126. Torres-Lugo, M.; Alvarez-Berrios, M.P.; Castillo, A.; Rinaldi, C. Magnetic Fluid Hyperthermia Enhances Cytotoxicity of Bortezomib in Sensitive and Resistant Cancer Cell Lines. Int. J. Nanomed. 2013, 9, 145–153. [Google Scholar] [CrossRef] [PubMed]
  127. Court, K.A.; Hatakeyama, H.; Wu, S.Y.; Lingegowda, M.S.; Rodríguez-Aguayo, C.; López-Berestein, G.; Ju-Seog, L.; Rinaldi, C.; Juan, E.J.; Sood, A.K.; et al. HSP70 Inhibition Synergistically Enhances the Effects of Magnetic Fluid Hyperthermia in Ovarian Cancer. Mol. Cancer Ther. 2017, 16, 966–976. [Google Scholar] [CrossRef]
  128. Jiang, P.; Zhang, Y.; Zhu, C.; Zhang, W.; Mao, Z.; Gao, C. Fe3O4/BSA Particles Induce Osteogenic Differentiation of Mesenchymal Stem Cells under Static Magnetic Field. Acta Biomater. 2016, 46, 141–150. [Google Scholar] [CrossRef]
  129. Vilas-Boas, V.; Espiña, B.; Kolen’ko, Y.V.; Bañobre-López, M.; Brito, M.; Martins, V.; Duarte, J.A.; Petrovykh, D.Y.; Freitas, P.; Carvalho, F. Effectiveness and Safety of a Nontargeted Boost for a CXCR4-Targeted Magnetic Hyperthermia Treatment of Cancer Cells. ACS Omega 2019, 4, 1931–1940. [Google Scholar] [CrossRef]
  130. Stocke, N.A.; Sethi, P.; Jyoti, A.; Chan, R.; Arnold, S.M.; Hilt, J.Z.; Upreti, M. Toxicity Evaluation of Magnetic Hyperthermia Induced by Remote Actuation of Magnetic Nanoparticles in 3D Micrometastasic Tumor Tissue Analogs for Triple Negative Breast Cancer. Biomaterials 2017, 120, 115–125. [Google Scholar] [CrossRef] [PubMed]
  131. Kwon, S.-H.; Al Faruque, H.; Kee, H.; Kim, E.; Park, S. Exosome-Based Hybrid Nanostructures for Enhanced Tumor Targeting and Hyperthermia Therapy. Colloids Surf. B Biointerfaces 2021, 205, 111915. [Google Scholar] [CrossRef]
  132. Aguilar, Z.P. Targeted Drug Delivery. In Nanomaterials for Medical Applications; Elsevier: Amsterdam, The Netherlands, 2013; pp. 181–234. [Google Scholar]
  133. Huang, J.; Li, Y.; Orza, A.; Lu, Q.; Guo, P.; Wang, L.; Yang, L.; Mao, H. Magnetic Nanoparticle Facilitated Drug Delivery for Cancer Therapy with Targeted and Image-Guided Approaches. Adv. Funct. Mater. 2016, 26, 3818–3836. [Google Scholar] [CrossRef] [PubMed]
  134. Huang, C.-H.; Chuang, T.-J.; Ke, C.-J.; Yao, C.-H. Doxorubicin–Gelatin/Fe3O4–Alginate Dual-Layer Magnetic Nanoparticles as Targeted Anticancer Drug Delivery Vehicles. Polymers 2020, 12, 1747. [Google Scholar] [CrossRef] [PubMed]
  135. Kianfar, E. Magnetic Nanoparticles in Targeted Drug Delivery: A Review. J. Supercond. Nov. Magn. 2021, 34, 1709–1735. [Google Scholar] [CrossRef]
  136. Almessiere, M.A.; Slimani, Y.; Korkmaz, A.D.; Baykal, A.; Güngüneş, H.; Sözeri, H.; Shirsath, S.E.; Güner, S.; Akhtar, S.; Manikandan, A. Impact of La3+ and Y3+ Ion Substitutions on Structural, Magnetic and Microwave Properties of Ni0.3Cu0.3Zn0.4Fe2O4 Nanospinel Ferrites Synthesized via Sonochemical Route. RSC Adv. 2019, 9, 30671–30684. [Google Scholar] [CrossRef]
  137. Al-Obaidy, R.; Haider, A.J.; Al-Musawi, S.; Arsad, N. Targeted Delivery of Paclitaxel Drug Using Polymer-Coated Magnetic Nanoparticles for Fibrosarcoma Therapy: In Vitro and in Vivo Studies. Sci. Rep. 2023, 13, 3180. [Google Scholar] [CrossRef] [PubMed]
  138. Alishiri, M.; Ebrahimi, S.; Shamloo, A.; Boroumand, A.; Mofrad, M.R.K. Drug Delivery and Adhesion of Magnetic Nanoparticles Coated Nanoliposomes and Microbubbles to Atherosclerotic Plaques under Magnetic and Ultrasound Fields. Eng. Appl. Comput. Fluid Mech. 2021, 15, 1703–1725. [Google Scholar] [CrossRef]
  139. Jiang, M.; Liu, Q.; Zhang, Y.; Wang, H.; Zhang, J.; Chen, M.; Yue, Z.; Wang, Z.; Wei, X.; Shi, S.; et al. Construction of Magnetic Drug Delivery System and Its Potential Application in Tumor Theranostics. Biomed. Pharmacother. 2022, 154, 113545. [Google Scholar] [CrossRef]
  140. Chertok, B.; Langer, R. Circulating Magnetic Microbubbles for Localized Real-Time Control of Drug Delivery by Ultrasonography-Guided Magnetic Targeting and Ultrasound. Theranostics 2018, 8, 341–357. [Google Scholar] [CrossRef]
  141. Lee, K.; David, A.E.; Zhang, J.; Shin, M.C.; Yang, V.C. Enhanced Accumulation of Theranostic Nanoparticles in Brain Tumor by External Magnetic Field Mediated in Situ Clustering of Magnetic Nanoparticles. J. Ind. Eng. Chem. 2017, 54, 389–397. [Google Scholar] [CrossRef]
  142. Tang, H.; Guo, Y.; Peng, L.; Fang, H.; Wang, Z.; Zheng, Y.; Ran, H.; Chen, Y. In Vivo Targeted, Responsive, and Synergistic Cancer Nanotheranostics by Magnetic Resonance Imaging-Guided Synergistic High-Intensity Focused Ultrasound Ablation and Chemotherapy. ACS Appl. Mater. Interfaces 2018, 10, 15428–15441. [Google Scholar] [CrossRef]
  143. Aram, E.; Moeni, M.; Abedizadeh, R.; Sabour, D.; Sadeghi-Abandansari, H.; Gardy, J.; Hassanpour, A. Smart and Multi-Functional Magnetic Nanoparticles for Cancer Treatment Applications: Clinical Challenges and Future Prospects. Nanomaterials 2022, 12, 3567. [Google Scholar] [CrossRef] [PubMed]
  144. Chaparro, C.I.P.; Simões, B.T.; Borges, J.P.; Castanho, M.A.R.B.; Soares, P.I.P.; Neves, V. A Promising Approach: Magnetic Nanosystems for Alzheimer’s Disease Theranostics. Pharmaceutics 2023, 15, 2316. [Google Scholar] [CrossRef]
  145. Zhang, W.; Yang, G.; Wang, X.; Jiang, L.; Jiang, F.; Li, G.; Zhang, Z.; Jiang, X. Magnetically Controlled Growth-Factor-Immobilized Multilayer Cell Sheets for Complex Tissue Regeneration. Adv. Mater. 2017, 29, 1703795. [Google Scholar] [CrossRef] [PubMed]
  146. Wu, F.; Sun, B.; Chu, X.; Zhang, Q.; She, Z.; Song, S.; Zhou, N.; Zhang, J.; Yi, X.; Wu, D.; et al. Hyaluronic Acid-Modified Porous Carbon-Coated Fe3O4 Nanoparticles for Magnetic Resonance Imaging-Guided Photothermal/Chemotherapy of Tumors. Langmuir 2019, 35, 13135–13144. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, J.; Zhang, Y.; Liu, L.; Cui, Z.; Liu, X.; Wang, L.; Li, Y.; Li, Q. Combined Chemo/Photothermal Therapy Based on Mesoporous Silica-Au Core-Shell Nanoparticles for Hepatocellular Carcinoma Treatment. Drug Dev. Ind. Pharm. 2019, 45, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
  148. Kaur, P.; Aliru, M.L.; Chadha, A.S.; Asea, A.; Krishnan, S. Hyperthermia Using Nanoparticles—Promises and Pitfalls. Int. J. Hyperth. 2016, 32, 76–88. [Google Scholar] [CrossRef] [PubMed]
  149. Shaw, S.K.; Kailashiya, J.; Gangwar, A.; Alla, S.K.; Gupta, S.K.; Prajapat, C.L.; Meena, S.S.; Dash, D.; Maiti, P.; Prasad, N.K. γ-Fe2O3 Nanoflowers as Efficient Magnetic Hyperthermia and Photothermal Agent. Appl. Surf. Sci. 2021, 560, 150025. [Google Scholar] [CrossRef]
  150. Multari, C.; Miola, M.; Laviano, F.; Gerbaldo, R.; Pezzotti, G.; Debellis, D.; Verné, E. Magnetoplasmonic Nanoparticles for Photothermal Therapy. Nanotechnology 2019, 30, 255705. [Google Scholar] [CrossRef]
  151. Yu, T.-J.; Li, P.-H.; Tseng, T.-W.; Chen, Y.-C. Multifunctional Fe3O4/Alumina Core/Shell MNPs as Photothermal Agents for Targeted Hyperthermia of Nosocomial and Antibiotic-Resistant Bacteria. Nanomedicine 2011, 6, 1353–1363. [Google Scholar] [CrossRef]
  152. Wang, J.; Zhao, H.; Zhou, Z.; Zhou, P.; Yan, Y.; Wang, M.; Yang, H.; Zhang, Y.; Yang, S. MR/SPECT Imaging Guided Photothermal Therapy of Tumor-Targeting Fe@Fe3O4 Nanoparticles in Vivo with Low Mononuclear Phagocyte Uptake. ACS Appl. Mater. Interfaces 2016, 8, 19872–19882. [Google Scholar] [CrossRef]
  153. Kami, D.; Takeda, S.; Itakura, Y.; Gojo, S.; Watanabe, M.; Toyoda, M. Application of Magnetic Nanoparticles to Gene Delivery. Int. J. Mol. Sci. 2011, 12, 3705–3722. [Google Scholar] [CrossRef]
  154. Mah, C.; Fraites, T.J.; Zolotukhin, I.; Song, S.; Flotte, T.R.; Dobson, J.; Batich, C.; Byrne, B.J. Improved Method of Recombinant AAV2 Delivery for Systemic Targeted Gene Therapy. Mol. Ther. 2002, 6, 106–112. [Google Scholar] [CrossRef]
  155. Akinc, A.; Thomas, M.; Klibanov, A.M.; Langer, R. Exploring Polyethylenimine-mediated DNA Transfection and the Proton Sponge Hypothesis. J. Gene Med. 2005, 7, 657–663. [Google Scholar] [CrossRef]
  156. McBain, S.C.; Yiu, H.H.P.; El Haj, A.; Dobson, J. Polyethyleneimine Functionalized Iron Oxide Nanoparticles as Agents for DNA Delivery and Transfection. J. Mater. Chem. 2007, 17, 2561. [Google Scholar] [CrossRef]
  157. Sizikov, A.A.; Nikitin, P.I.; Nikitin, M.P. Magnetofection In Vivo by Nanomagnetic Carriers Systemically Administered into the Bloodstream. Pharmaceutics 2021, 13, 1927. [Google Scholar] [CrossRef]
  158. Stein, R.; Pfister, F.; Friedrich, B.; Blersch, P.-R.; Unterweger, H.; Arkhypov, A.; Mokhir, A.; Kolot, M.; Alexiou, C.; Tietze, R. Plasmid-DNA Delivery by Covalently Functionalized PEI-SPIONs as a Potential ‘Magnetofection’ Agent. Molecules 2022, 27, 7416. [Google Scholar] [CrossRef]
  159. Farooq, N.; Ather, L.; Shafiq, M.; Nawaz-ul-Rehman, M.S.; Haseeb, M.; Anjum, T.; Abbas, Q.; Hussain, M.; Ali, N.; Asad Abbas, S.A.A.; et al. Magnetofection Approach for the Transformation of Okra Using Green Iron Nanoparticles. Sci. Rep. 2022, 12, 16568. [Google Scholar] [CrossRef] [PubMed]
  160. Danthanarayana, A.N.; Manatunga, D.C.; De Silva, R.M.; Chandrasekharan, N.V.; Nalin De Silva, K.M. Magnetofection and Isolation of DNA Using Polyethyleneimine Functionalized Magnetic Iron Oxide Nanoparticles. R. Soc. Open Sci. 2018, 5, 181369. [Google Scholar] [CrossRef] [PubMed]
  161. Shevtsov, M.A.; Nikolaev, B.P.; Ryzhov, V.A.; Yakovleva, L.Y.; Marchenko, Y.Y.; Parr, M.A.; Rolich, V.I.; Mikhrina, A.L.; Dobrodumov, A.V.; Pitkin, E.; et al. Ionizing Radiation Improves Glioma-Specific Targeting of Superparamagnetic Iron Oxide Nanoparticles Conjugated with CmHsp70.1 Monoclonal Antibodies (SPION–CmHsp70.1). Nanoscale 2015, 7, 20652–20664. [Google Scholar] [CrossRef] [PubMed]
  162. Shevtsov, M.A.; Nikolaev, B.P.; Yakovleva, L.Y.; Parr, M.A.; Marchenko, Y.Y.; Eliseev, I.; Yudenko, A.; Dobrodumov, A.V.; Zlobina, O.; Zhakhov, A.; et al. 70-KDa Heat Shock Protein Coated Magnetic Nanocarriers as a Nanovaccine for Induction of Anti-Tumor Immune Response in Experimental Glioma. J. Control. Release 2015, 220, 329–340. [Google Scholar] [CrossRef] [PubMed]
  163. Tomanek, B.; Iqbal, U.; Blasiak, B.; Abulrob, A.; Albaghdadi, H.; Matyas, J.R.; Ponjevic, D.; Sutherland, G.R. Evaluation of Brain Tumor Vessels Specific Contrast Agents for Glioblastoma Imaging. Neuro-Oncology 2012, 14, 53–63. [Google Scholar] [CrossRef]
  164. Salimi, M.; Sarkar, S.; Saber, R.; Delavari, H.; Alizadeh, A.M.; Mulder, H.T. Magnetic Hyperthermia of Breast Cancer Cells and MRI Relaxometry with Dendrimer-Coated Iron-Oxide Nanoparticles. Cancer Nanotechnol. 2018, 9, 7. [Google Scholar] [CrossRef]
  165. Chen, C.; Wu, C.Q.; Chen, T.W.; Tang, M.Y.; Zhang, X.M. Molecular Imaging with MRI: Potential Application in Pancreatic Cancer. Biomed. Res. Int. 2015, 2015, 624074. [Google Scholar] [CrossRef] [PubMed]
  166. Kiamohammadi, L.; Asadi, L.; Shirvalilou, S.; Khoei, S.; Khoee, S.; Soleymani, M.; Minaei, S.E. Physical and Biological Properties of 5-Fluorouracil Polymer-Coated Magnetite Nanographene Oxide as a New Thermosensitizer for Alternative Magnetic Hyperthermia and a Magnetic Resonance Imaging Contrast Agent: In Vitro and In Vivo Study. ACS Omega 2021, 6, 20192–20204. [Google Scholar] [CrossRef]
  167. Tse, B.W.-C.; Cowin, G.J.; Soekmadji, C.; Jovanovic, L.; Vasireddy, R.S.; Ling, M.-T.; Khatri, A.; Liu, T.; Thierry, B.; Russell, P.J. PSMA-Targeting Iron Oxide Magnetic Nanoparticles Enhance MRI of Preclinical Prostate Cancer. Nanomedicine 2015, 10, 375–386. [Google Scholar] [CrossRef]
  168. Wang, Y.; Yang, Y.; Zheng, X.; Shi, J.; Zhong, L.; Duan, X.; Zhu, Y. Application of Iron Oxide Nanoparticles in the Diagnosis and Treatment of Leukemia. Front. Pharmacol. 2023, 14, 1177068. [Google Scholar] [CrossRef]
  169. Thomas, R.; Park, I.-K.; Jeong, Y. Magnetic Iron Oxide Nanoparticles for Multimodal Imaging and Therapy of Cancer. Int. J. Mol. Sci. 2013, 14, 15910–15930. [Google Scholar] [CrossRef] [PubMed]
  170. Ganguly, S.; Margel, S. Bioimaging Probes Based on Magneto-Fluorescent Nanoparticles. Pharmaceutics 2023, 15, 686. [Google Scholar] [CrossRef]
  171. Winter, A.; Chavan, A.; Wawroschek, F. Magnetic Resonance Imaging of Sentinel Lymph Nodes Using Intraprostatic Injection of Superparamagnetic Iron Oxide Nanoparticles in Prostate Cancer Patients: First-in-Human Results. Eur. Urol. 2018, 73, 813–814. [Google Scholar] [CrossRef] [PubMed]
  172. Winter, A.; Woenkhaus, J.; Wawroschek, F. A Novel Method for Intraoperative Sentinel Lymph Node Detection in Prostate Cancer Patients Using Superparamagnetic Iron Oxide Nanoparticles and a Handheld Magnetometer: The Initial Clinical Experience. Ann. Surg. Oncol. 2014, 21, 4390–4396. [Google Scholar] [CrossRef]
  173. Michalik, B.; Engels, S.; Otterbach, M.C.; Frerichs, J.; Suhrhoff, P.E.; van Oosterom, M.N.; Maurer, M.H.; Wawroschek, F.; Winter, A. A New Bimodal Approach for Sentinel Lymph Node Imaging in Prostate Cancer Using a Magnetic and Fluorescent Hybrid Tracer. Eur. J. Nucl. Med. Mol. Imaging 2023. [Google Scholar] [CrossRef]
  174. Poletto, G.; Evangelista, L.; Venturini, F.; Gramegna, F.; Seno, F.; Moro, S.; Vettor, R.; Realdon, N.; Cecchin, D. Nanoparticles and Radioisotopes: A Long Story in a Nutshell. Pharmaceutics 2022, 14, 2024. [Google Scholar] [CrossRef] [PubMed]
  175. Onishi, T.; Mihara, K.; Matsuda, S.; Sakamoto, S.; Kuwahata, A.; Sekino, M.; Kusakabe, M.; Handa, H.; Kitagawa, Y. Application of Magnetic Nanoparticles for Rapid Detection and In Situ Diagnosis in Clinical Oncology. Cancers 2022, 14, 364. [Google Scholar] [CrossRef] [PubMed]
  176. Sánchez-Cabezas, S.; Montes-Robles, R.; Gallo, J.; Sancenón, F.; Martínez-Máñez, R. Combining Magnetic Hyperthermia and Dual T1/ T2 MR Imaging Using Highly Versatile Iron Oxide Nanoparticles. Dalton Trans. 2019, 48, 3883–3892. [Google Scholar] [CrossRef] [PubMed]
  177. Sharma, A.; Cressman, E.; Attaluri, A.; Kraitchman, D.L.; Ivkov, R. Current Challenges in Image-Guided Magnetic Hyperthermia Therapy for Liver Cancer. Nanomaterials 2022, 12, 2768. [Google Scholar] [CrossRef]
  178. Goodwill, P.W.; Conolly, S.M. The X-Space Formulation of the Magnetic Particle Imaging Process: 1-D Signal, Resolution, Bandwidth, SNR, SAR, and Magnetostimulation. IEEE Trans. Med. Imaging 2010, 29, 1851–1859. [Google Scholar] [CrossRef] [PubMed]
  179. Liu, S.; Heshmat, A.; Andrew, J.; Barreto, I.; Rinaldi-Ramos, C.M. Dual Imaging Agent for Magnetic Particle Imaging and Computed Tomography. Nanoscale Adv. 2023, 5, 3018–3032. [Google Scholar] [CrossRef] [PubMed]
  180. Buzug, T.M.; Bringout, G.; Erbe, M.; Gräfe, K.; Graeser, M.; Grüttner, M.; Halkola, A.; Sattel, T.F.; Tenner, W.; Wojtczyk, H.; et al. Magnetic Particle Imaging: Introduction to Imaging and Hardware Realization. Z. Med. Phys. 2012, 22, 323–334. [Google Scholar] [CrossRef]
  181. Wang, L.; Huang, Y.; Zhao, Y.; Tian, J.; Zhang, L.; Du, Y. Improved Quantitative Analysis Method for Magnetic Particle Imaging Based on Deblurring and Region Scalable Fitting. Mol. Imaging Biol. 2023, 25, 788–797. [Google Scholar] [CrossRef]
  182. Wu, W.E.; Chang, E.; Jin, L.; Liu, S.; Huang, C.-H.; Kamal, R.; Liang, T.; Aissaoui, N.M.; Theruvath, A.J.; Pisani, L.; et al. Multimodal In Vivo Tracking of Chimeric Antigen Receptor T Cells in Preclinical Glioblastoma Models. Investig. Radiol. 2023, 58, 388–395. [Google Scholar] [CrossRef]
  183. 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]
  184. Yildiz, I. Applications of Magnetic Nanoparticles in Biomedical Separation and Purification. Nanotechnol. Rev. 2016, 5, 331–340. [Google Scholar] [CrossRef]
  185. Iacovita, C.; Fizeșan, I.; Pop, A.; Scorus, L.; Dudric, R.; Stiufiuc, G.; Vedeanu, N.; Tetean, R.; Loghin, F.; Stiufiuc, R.; et al. In Vitro Intracellular Hyperthermia of Iron Oxide Magnetic Nanoparticles, Synthesized at High Temperature by a Polyol Process. Pharmaceutics 2020, 12, 424. [Google Scholar] [CrossRef] [PubMed]
  186. Rahimi, M.; Safa, K.D.; Salehi, R. Co-Delivery of Doxorubicin and Methotrexate by Dendritic Chitosan-g-MPEG as a Magnetic Nanocarrier for Multi-Drug Delivery in Combination Chemotherapy. Polym. Chem. 2017, 8, 7333–7350. [Google Scholar] [CrossRef]
  187. Kandasamy, G.; Sudame, A.; Maity, D.; Soni, S.; Sushmita, K.; Veerapu, N.S.; Bose, S.; Tomy, C.V. Multifunctional Magnetic-Polymeric Nanoparticles Based Ferrofluids for Multi-Modal in Vitro Cancer Treatment Using Thermotherapy and Chemotherapy. J. Mol. Liq. 2019, 293, 111549. [Google Scholar] [CrossRef]
  188. Yang, Y.; Aw, J.; Chen, K.; Liu, F.; Padmanabhan, P.; Hou, Y.; Cheng, Z.; Xing, B. Enzyme-Responsive Multifunctional Magnetic Nanoparticles for Tumor Intracellular Drug Delivery and Imaging. Chem. Asian J. 2011, 6, 1381–1389. [Google Scholar] [CrossRef]
  189. Wang, X.; Zhu, L.; Hou, X.; Wang, L.; Yin, S. Polyethylenimine Mediated Magnetic Nanoparticles for Combined Intracellular Imaging, SiRNA Delivery and Anti-Tumor Therapy. RSC Adv. 2015, 5, 101569–101581. [Google Scholar] [CrossRef]
  190. Miao, L.; Liu, C.; Ge, J.; Yang, W.; Liu, J.; Sun, W.; Yang, B.; Zheng, C.; Sun, H.; Hu, Q. Antitumor Effect of TRAIL on Oral Squamous Cell Carcinoma Using Magnetic Nanoparticle-Mediated Gene Expression. Cell Biochem. Biophys. 2014, 69, 663–672. [Google Scholar] [CrossRef]
  191. Cruz-Acuña, M.; Halman, J.R.; Afonin, K.A.; Dobson, J.; Rinaldi, C. Magnetic Nanoparticles Loaded with Functional RNA Nanoparticles. Nanoscale 2018, 10, 17761–17770. [Google Scholar] [CrossRef]
  192. Yang, H.; Wang, H.; Wen, C.; Bai, S.; Wei, P.; Xu, B.; Xu, Y.; Liang, C.; Zhang, Y.; Zhang, G.; et al. Effects of Iron Oxide Nanoparticles as T2-MRI Contrast Agents on Reproductive System in Male Mice. J. Nanobiotechnol. 2022, 20, 98. [Google Scholar] [CrossRef]
  193. Wei, H.; Wiśniowska, A.; Fan, J.; Harvey, P.; Li, Y.; Wu, V.; Hansen, E.C.; Zhang, J.; Kaul, M.G.; Frey, A.M.; et al. Single-Nanometer Iron Oxide Nanoparticles as Tissue-Permeable MRI Contrast Agents. Proc. Natl. Acad. Sci. USA 2021, 118, e2102340118. [Google Scholar] [CrossRef]
  194. Shao, C.; Liu, F.; Le, W.; Mei, T.; Wang, T.; Chen, L.; Lei, Y.; Cui, S.; Chen, B.; Cui, Z. In Vitro and in Vivo Targeting Imaging of Pancreatic Cancer Using a Fe3O4@SiO2 Nanoprobe Modified with Anti-Mesothelin Antibody. Int. J. Nanomed. 2016, 11, 2195–2207. [Google Scholar] [CrossRef] [PubMed]
  195. Ajayi, T.O.; Liu, S.; Rosen, C.; Rinaldi-Ramos, C.M.; Allen, K.D.; Sharma, B. Application of Magnetic Particle Imaging to Evaluate Nanoparticle Fate in Rodent Joints. J. Control. Release 2023, 356, 347–359. [Google Scholar] [CrossRef] [PubMed]
  196. Tong, W.; Zhang, Y.; Hui, H.; Feng, X.; Ning, B.; Yu, T.; Wang, W.; Shang, Y.; Zhang, G.; Zhang, S.; et al. Sensitive Magnetic Particle Imaging of Haemoglobin Degradation for the Detection and Monitoring of Intraplaque Haemorrhage in Atherosclerosis. EBioMedicine 2023, 90, 104509. [Google Scholar] [CrossRef] [PubMed]
  197. Sun, Y.; Li, H.; Cui, G.; Wu, X.; Yang, M.; Piao, Y.; Bai, Z.; Wang, L.; Kraft, M.; Zhao, W.; et al. A Magnetic Nanoparticle Assisted Microfluidic System for Low Abundance Cell Sorting with High Recovery. Micro Nano Eng. 2022, 15, 100136. [Google Scholar] [CrossRef]
  198. Mettler, E.; Trenkler, A.; Feilen, P.J.; Wiegand, F.; Fottner, C.; Ehrhart, F.; Zimmermann, H.; Hwang, Y.H.; Lee, D.Y.; Fischer, S.; et al. Magnetic Separation of Encapsulated Islet Cells Labeled with Superparamagnetic Iron Oxide Nano Particles. Xenotransplantation 2013, 20, 219–226. [Google Scholar] [CrossRef] [PubMed]
  199. Gao, X.-L.; Shao, M.-F.; Xu, Y.-S.; Luo, Y.; Zhang, K.; Ouyang, F.; Li, J. Non-Selective Separation of Bacterial Cells with Magnetic Nanoparticles Facilitated by Varying Surface Charge. Front. Microbiol. 2016, 7, 1891. [Google Scholar] [CrossRef] [PubMed]
  200. Wu, K.; Liu, J.; Saha, R.; Peng, C.; Su, D.; Wang, Y.A.; Wang, J.-P. Investigation of Commercial Iron Oxide Nanoparticles: Structural and Magnetic Property Characterization. ACS Omega 2021, 6, 6274–6283. [Google Scholar] [CrossRef]
  201. Torres-Herrero, B.; Armenia, I.; Alleva, M.; Asín, L.; Correa, S.; Ortiz, C.; Fernández-Afonso, Y.; Gutiérrez, L.; de la Fuente, J.M.; Betancor, L.; et al. Remote Activation of Enzyme Nanohybrids for Cancer Prodrug Therapy Controlled by Magnetic Heating. ACS Nano 2023, 17, 12358–12373. [Google Scholar] [CrossRef]
Figure 1. A brief representation of MNP synthesis methods. Created with BioRender.com, accessed on 7 February 2024.
Figure 1. A brief representation of MNP synthesis methods. Created with BioRender.com, accessed on 7 February 2024.
Applsci 14 01623 g001
Figure 2. Biomedical applications of MNPs. Created with BioRender.com, accessed on 6 February 2024.
Figure 2. Biomedical applications of MNPs. Created with BioRender.com, accessed on 6 February 2024.
Applsci 14 01623 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Stiufiuc, G.F.; Stiufiuc, R.I. Magnetic Nanoparticles: Synthesis, Characterization, and Their Use in Biomedical Field. Appl. Sci. 2024, 14, 1623. https://doi.org/10.3390/app14041623

AMA Style

Stiufiuc GF, Stiufiuc RI. Magnetic Nanoparticles: Synthesis, Characterization, and Their Use in Biomedical Field. Applied Sciences. 2024; 14(4):1623. https://doi.org/10.3390/app14041623

Chicago/Turabian Style

Stiufiuc, Gabriela Fabiola, and Rares Ionut Stiufiuc. 2024. "Magnetic Nanoparticles: Synthesis, Characterization, and Their Use in Biomedical Field" Applied Sciences 14, no. 4: 1623. https://doi.org/10.3390/app14041623

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop