Open access peer-reviewed chapter

Nanoferrites-Based Drug Delivery Systems as Adjuvant Therapy for Cancer Treatments. Current Challenges and Future Perspectives

Written By

Felipe Ocampo Osorio, Jhon Augusto Jativa Herrera, Oscar Moscoso Londoño and César Leandro Londoño Calderón

Submitted: 09 April 2021 Reviewed: 31 August 2021 Published: 17 November 2021

DOI: 10.5772/intechopen.100225

From the Edited Volume

Ferrites - Synthesis and Applications

Edited by Maaz Khan

Chapter metrics overview

435 Chapter Downloads

View Full Metrics

Abstract

Cancer is the second cause of death worldwide, whose treatment often involves chemotherapy. In a conventional therapy, drug is transported (and usually absorbed) across biological membranes through diffusion and systemic transport. The pathway that medicine must travel before reaching the desired location, can bring adverse or unwanted effects, which are mainly the result of: low bioavailability, low solubility and toxicity. To avoiding risks, nanoparticles coated with the drug could be used as a therapeutic substance to selectively reach an area of interest to act without affecting non-target cells, organs, or tissues (drug delivery). Here, the goal is to enhance the concentration of the chemotherapeutic drug in the disease parts of the body. Among all nanostructured systems, ferrites attract worldwide attention in drug delivery applications. It is due to their versatile magnetic and physicochemical properties. Here, it is reviewed and analyzed recent advances in synthesis, morphology, size, magnetic properties, functionalization with a focus in drug delivery applications of nanoferrites.

Keywords

  • Ferrites
  • Nanostructures
  • Functionalization
  • Drug-loading
  • Drug delivery
  • Cancer

1. Introduction

Cancer is a disease originating from unregulated cell growth. Those cells can spread throughout the body, causing erroneous behaviors in organs or tissues [1]. Cancer is one of the principal problems in public health and currently is the second leading cause of death worldwide. According to the American Joint Committee on Cancer (AJCC) and the International Union for Cancer Control (UICC), there are many types of cancer treatments. Treatment or therapy depends on the cancer, as well as its stage of progress. Most individuals with cancer receive a combination of treatments, such as surgery with chemotherapy and/or radiation therapy [2]. Chemotherapy is crucial in the prevention of tumor recurrence and progression. Some patients have been treated with chemotherapeutic agents (e.g., Doxorubicin, Cyclodextrin, Cisplatin, Taxanes, Gemcitabine, among others) for long-term survival. Despite recent advances in treatments for various types of cancer, the recurrence rate and severe side effects still are a problem. To improve the life quality of cancer patients, more efficient and accurate targeting treatment is an urgent need.

A nanotechnology-based drug delivery system may provide a feasible means to solve the previous challenges. This kind of technology can be a formulation or device that enables a therapeutic substance to selectively reach an area of interest to act without affecting non-target cells, organs, or tissues [3]. One of the most studied, promising, and simplest ways to transport pharmaceutical compounds in the body is using nano entities as delivery vehicles [4]. Moreover, some nanoparticulated systems exhibit sensitivity to external stimuli, such as visible light, near-infrared light, ultrasound, AC or DC magnetic fields, among others. These stimuli could be made use as a tool to flexible control of dose magnitude and timing from the responsiveness (triggered remotely) [5].

Specifically, nanoferrites have been attracted worldwide scientific community attention for applications against cancer due to [6]:

  • Their great potential for hyperthermia treatments.

  • The possibility to guide nanoparticles to specific regions using an external magnetic field.

  • The chance to remotely activate the drug release in a controlled way (alternating magnetic fields).

Ferrites are compounds derived from iron oxides, whose composition allows tuning the magnetic properties. According to the magnetic atoms disposition and its chemical environment, material can manifest hard of soft magnetic properties. Nanostructure ferrites have received the most attention for drug delivery applications due to their versatile magnetic and chemical properties [7].

Previous reports have pay attention to the use of nanoferrites in biomedical applications for:

  • Improving magnetic resonance imaging sensitivity [8].

  • Effective targeted treatment of lung cancer [7].

  • Magnetite nanoparticles as an advanced platform for cancer theranostics [9].

  • Hydrogel beads-based nanoferrites in novel drug delivery platforms [10].

  • Magnetic and superparamagnetic ferrites for cancer therapy applications [11].

  • Iron oxide and substitute ferrite nanoparticles in drug delivery [12].

  • The toxicity of spinel ferrite nanoparticles [13].

  • Biosensing platform on ferrite nanoparticles [14].

Thus, the chapter aims to correlate the morphology, size, ferrite type, magnetic properties, functionalization, and pharmacokinetics. These correlations allow obtaining a perspective to the physical targeting precision for cancer drug delivery applications. Furthermore, we also discussed the current challenges and future perspectives of nanoferrites in the field of oncology.

Advertisement

2. Size, morphology, and magnetic properties of nanoferrites for drug delivery in cancer

The size, morphology, and magnetic properties of nanoparticles in drug delivery applications, have been identified as keys parameters in the literature [1, 2]. The easy way to tune these properties is from the synthetic routes [15]. The growth mechanism involved in the final morphology and structure is not completely clear. The conditions synthesis and their correlation with the physicochemical properties have been discussed in the literature [16]. Here, we focus on the recent advances in morphology, size, magnetic properties, and their relationship with the synthetic routes of nanoferrites used in drug delivery for cancer. Table A1 shows a summary of these properties recently reported in the literature. For there, it is clear that the synthetic routes more employed for nanoferrites synthesis are:

  • Chemical coprecipitation: it is a straightforward and inexpensive method. In this case, the precursor salts solutions containing the cationic metals are mix into an alkaline medium in a stoichiometric proportion.

  • Hydrothermal: here, the chemical reactions take place in aqueous solutions at pressure and temperature higher than the room conditions.

  • Sol–gel: the chemical reactions of hydrolysis and condensation are carried out of precursors in solution.

  • Solvothermal: it is like the hydrothermal technique. The difference is the use of nonaqueous solutions.

  • Less popular synthesis techniques for nanoferrites obtention are thermal decomposition, sonochemical, thermal treatment, and thermolysis.

Tripathy et al. [14] reported a comparison among the different techniques for ferrite nanoparticles obtention.

Based on some scientific reports, nanoferrites used in cancer drug delivery applications range from 5.2 nm to 300 nm (Table A1). Calcium ferrite (CaFe2O4) obtained by coprecipitation is the smallest nanostructure system. Magnetite (Fe3O4) fabricate from the solvothermal method is the larger one (Figure 1). However, particles larger than 200 nm segregate by mechanical filtering and eventually get removed by phagocytic cells. Nanoparticles with sizes smaller than 10 nm lead to renal filtration and accumulation into the fenestration of the kidneys’ glomerular endothelium. Therefore, the most effective drug delivery agents possess sizes ranging between 10 and 100 nm [68]. However, Sivaraj et al. [69] suggest that the nuclear membrane pores allow entry of nanoparticles with a size below 9 nm. Nanoparticles penetration into the cells may be maximized by surface functionalization with small molecules (e.g. folate, proteins, peptides, antibodies, and aptamers). This penetration induces receptor-mediated endocytosis, caveolae-mediated endocytosis, lipid raft mediated endocytosis, and/or micropinocytosis. After endocytosis in cancer therapy, nanomaterial releases maximum drug to inhibit the DNA/RNA synthesis and mitochondria damage.

Figure 1.

Transmission Electron microscopy (TEM) images for a) calcium ferrite nanoparticles with a size of 5 nm, reproduced from Ref. [17] with permission of the editors, b) magnetite hollow spheres of diameter ∼350 nm, reproduced from Ref. [65] with permission of the editors, c) magnetite nanorods, reproduced from Ref. [48] with permission of the editors, and d) magnetite hexagonal nanoparticles, reproduced from Ref. [56] with permission of the editors. Copyright 2019 MDPI, 2016 Nature, 2020 Elsevier, and 2021 the Royal Society of Chemistry.

The most common ferrite nanoparticles use for cancer drug delivery systems ranging from 20 nm to 30 nm (Table A1). Moreover, the most popular morphology obtained from the synthetic routes is spherical particles (Table 1). Nanorods and particles with hexagonal shapes are the less common nanostructures used for drug delivery in cancer applications (Figure 1).

FTMethodSMSHCMRReference
Fe3O4Coprecipitation11590[20]
CoFe2O4Thermal decomposition1370.730.2[22]
MnFe2O4Sonochemical1334.900[23]
Mn-Zn (Fe2O4)Coprecipitation1556.000[25]
NiFe2O4Solvothermal177000[29]
Fe3O4Thermal treatment237.1143.82.2[32]
Fe3O4Coprecipitation3047.603.8[39]
Fe3O4Coprecipitation3536.30[40]
Fe3O4Coprecipitation401.5769.10.15[41]
CoFe2O4Coprecipitation4336.0200[42]
GdFe2O4Coprecipitation904700[57]
CoFe2O4Solvothermal10451.800[59]
CaFe2O4Sol–gel11214.90.38[61]
MnFe2O4Coprecipitation14056.142.65.2[62]
CoFe2O4Thermolysis20051.100[64]
CoFe2O4Coprecipitation250401.7[65]

Table 1.

Summary of spherical nanoparticles ferrite type (FT) obtained by different methods with their sizes (S in nm), saturation magnetization (MS in emu/g), coercivity (HC in Oe), and remanence (MR in emu/g), reported in the literature. All the magnetic properties were reported at room temperature.

A complete understanding of magnetic properties is essential for a proper implementation of nanoferrites in drug delivery applications [6]. The saturation magnetization (Ms), coercive force (HC), and remanence (MR) are the most popular magnetic parameters reported for nanoferrites to cancer drug delivery applications (Table A1). Nanoferrites with the highest magnetic response (Ms) are cobalt ferrite (CoFe2O4) with a size of 15 nm obtained by sonochemical technique [28]. The smallest saturation magnetization was reported to zinc ferrite (ZnFe2O4) nanostructures (75 nm), which were synthesized by the sol–gel method [53]. Usually, nanoferrites used in cancer drug delivery applications show superparamagnetic behavior. Superparamagnetic nanoparticles evidence zero coercivity and remanence at temperatures above the blocking one (Table 1). In other cases, ferrite nanostructures with coercivity as high as 3409 Oe are used in cancer drug delivery applications (CoFe2O4 nanofibers with a diameter of 50 nm [45]. Moreover, cobalt ferrite nanoparticles show the highest magnetic remanence of 30.2 emu/g with a size of 30 nm obtained by thermal decomposition (Table 1).

Advertisement

3. Nanoferrites functionalized and functional groups for drug delivery in cancer

Non-functionalized nanoferrites (non-coating material on their surface) seems to be not optimal for drug delivery application. Surface energy minimization processes can promote agglomeration, percolation as well as other unwanted effects. Some of the most common problems whit this kind of nanosystems are [12]:

  1. Agglomeration due to the attractive forces leading to non-stability of the nanoparticle dispersion.

  2. Toxicity represents a problem in bare nanoferrites when they use without functionalization.

  3. Bare nanoparticles do not have a functional group on their surface. This makes it hard to link drugs molecules.

To deal with these problems, nanoparticles have been coating with organic or inorganic molecules (functionalization). The surface engineering of ferrites could be accomplished during nanoparticle synthesis (in-situ) or after this (ex-situ). A detailed review of the coating and functionalization strategies was reported for nanoparticles in drug delivery applications by Pinelli et al. [70]. The surface functionalization procedure and choice of appropriate solvent are crucial factors for obtaining nanoferrites. Here, the repulsive interactions among nanoparticles prevent agglomerations [71]. Moreover, functionalization promotes several advantages such as stable dispersions, biocompatibility, biodegradability, and reduced toxicity. Usually, functionalized nanoparticles loaded with drugs adopt covalent/noncovalent interaction methods. Conjugation of a drug to a carrier by non-biodegradable linkages results in: changing the drug chemical units, reducing drug efficacy, and displaying relevant side effects. The drug remains unharmed by using physical adsorption for drug conjugation, and no changes occur in the chemical units and the controlled drug release behavior. In this case, the idea deals with functionalized nanoparticles that have an opposite electrical charge to the cancer drug to promote the electrostatic interaction [42]. Moreover, surface functionality gives significant strength to bind and adsorb cancer drugs using specific functional groups. The characterization techniques for studying the functional groups attached to nanoparticles for drug delivery applications have been reported previously [12].

Some examples of functional groups commonly used to functionalized nanoferrites in drug delivery applications are:

  • The carboxyl functional group of the meso, 2–3-dimercaptosuccinic acid (DMSA), was used to functionalize cobalt ferrite nanoparticles [22].

  • Magnetite nanoparticles functionalized with mesoporous silica (SiO2) [58].

  • Zinc ferrite nanoparticles coated with hydroxyapatite as an intermediate of the cancer drug [72].

  • The carboxyl functional groups of citrate molecules use to functionalize manganese ferrite nanorods [24].

  • Calcium ferrite functionalized with biomolecules (casein). The hybrid molecule combines the merits of both inorganic and organic counterparts [61].

The functionalization can allows high drug encapsulation, stabilizes the nanocarrier, and reaches the cancer site-specific. Furthermore, the coating uses to reach the target cells without getting removed by the reticuloendothelial system of the body and to have a capable surface for keeping the drug unharmed until reaching the location of interest. The performance enhancement achieves through functionalization with suitable ligands that will bind to the aimed receptors of pathological tissues. The size of the nanocarrier has paramount importance for rendering it absorbable by tumor tissues [68]. The inclusion of active targeting functionalities results in drug accumulation within tumors, tumor cells, or immune cells and allows for reduced dosages due to specificity. Functionalized ferrite nanoparticles have been used for: a) imitate ligand binding to receptors, b) for initiation of cellular signaling, c) for increased stimulation of immune cells to better infiltrate and extinguish immunosuppressive tumors [73]. Commonly, the pH of cancer cells (tumor) is acidic ranging between 4 and 5. It is due to the presence of lactic acid, which starts due to inefficient consumption of glucose [74]. On the other hand, the pH in an extracellular matrix or bloodstream is natural (pH = 7) [75]. This difference in pH offers to fabricate functionalized nanoparticles as a pH-sensitive trigger for drug delivery applications.

The most popular drugs for cancer delivery applications, using ferrites as nanocarriers are: Doxorubicin [58], 5-Fluorouracil [21], Docetaxel [76], Hesperidin and Eugenol [60], Curcumin [77], Tamoxifen [55], Cisplatin [78], Nilotinib [79], Camptothecin [38], and Telmisartan [20]. Hydrophobicity of the orally administered drugs for cancer treatments has low systemic bioavailability [80]. It produces low water solubility and can cause serious adverse effects [62].

Among functionalized nanoferrites investigated to load cancer drugs, one can find:

  1. Zinc ferrite functionalized with Polyethylene Glycol (PEG) and chitosan loaded with Curcumin [80]. Chitosan takes cationic amine functional groups, at low pH, which would involve an ionic gelation process with polyanions to form nanoparticles. It is used as an effective drug carrier, where the reactive amine groups on the chitosan side chain are used for functional group modifications. The hydrophobically modified chitosan improves the encapsulation efficiency of the carrier towards the hydrophobic drugs [34].

  2. Cobalt ferrite nanoparticles functionalized with DMSA used the amine functional group of Doxorubicin molecules. Here, it is attached through electrostatic interaction and/or hydrogen bonding interactions with the carboxylic functional group of the DMSA [22].

  3. Magnetite nanoparticles functionalized with mesoporous silica used the amine group of Doxorubicin to attach [58].

  4. Zinc ferrite nanoparticles functionalized with hydroxyapatite had covalent bonds with the zoledronic acid drug. Amino or hydroxyl functional groups presented in hydroxyapatite are a strong chemical bond with the mineral material of bone phases [54].

  5. Magnetite nanoparticles functionalized with gelatin using the functional groups -NH3+-. It produces by partial hydrolysis of collagen to interact with Doxorubicin [81].

  6. Calcium ferrite nanoparticles functionalized with biomolecules (casein), which allows the conjugation of targeting ligands with functional groups. Actively bind with specific receptors that may be overexpressed on tumor cells, allowing improved biodistribution and delivery of the drugs at the cancer site [61].

  7. Manganese ferrite nanorods functionalized with citrate molecules to electrostatically attach Doxorubicin [24].

Proteins are promising carriers for drug delivery applications. The main advantages are the abundance of active sites, improved biocompatibility, easy availability, and pH-dependent swelling behavior. The last one allows the programmed release of the cytotoxic agent in response to the acidic cancer microenvironment [82].

DFT calculations demonstrated Cisplatin on graphene oxide can be adsorbed by the functionalized nanoferrites. Here, hydrogen bonds forming with hydroxyl and epoxy functional groups. It involves the formation of the amide bond between Cisplatin and the COOH functional group of graphene oxide. In the case of glutaraldehyde, the functional group is CHO, which formed the amide bond between Cisplatin and the CHO functional group [18].

Advertisement

4. Drugs loaded on functionalized nanoferrites for cancer treatments

Drug-loading of nanoparticles plays an essential role in drug delivery systems. There are several ways through which the drug can load with the functionalized nanoparticle:

  • Encapsulation. It can entrap inside the nanoparticle.

  • Functionalization. It can coat the surface of the nanoparticle.

  • Chemically linked. It can be bond with the functionalized particle itself.

The second key point in functionalized nanoparticles design is the necessity to provide the nanoparticles with specific properties. The interaction with the external environment in the human body increases the targeting action towards determined sites [70].

Drug-loading involves several variables such as the solvent type and amount of it, the temperature, time of loading, and the drug-loading capacity. The most popular solvent for drug-loading is water (see Table A2). Less popular solvents involved in drug-loading are ethanol, dichloromethane, and saline solution. Usually, the solvent quantity varies from 1 ml to 200 ml. The drug-loading capacity represents the amount of drug loader per unit weight of the nanoparticle. Drug-loading represents the percentage of the nanoparticle mass that is due to the encapsulated drug. Loading capacity can calculate by the amount of total entrapped drug divided by the total nanoparticle weight. The drug-loading values reported for nanoferrites ranging from 0.016 [64] to 3.3 [63]. These values correspond to cobalt ferrite loaded with Doxorubicin and Docetaxel, respectively. The loading-drug temperature ranges from 4°C [54] to 55°C [33].

From Table A2 many reports did not include the drug-loading solvent, the solvent quantity, and the drug loading capacity. The efficiency of drug-loading measure by a high-performance liquid chromatography system (HPLC) [30] or ultraviolet–visible spectroscopy (UV–Vis) [18]:

Drugloading%=total amount of drugfree amount of drugtotal amount of drugx100E1

The free amount of the drug is measure by the absorbance of the supernatant in a UV–Vis spectrophotometer at the maximum wavelength of the dissolved drug. The nanoferrites can magnetically remove from the solution instead of the centrifugation process. The maximum wavelength for anticancer drugs are: Doxorubicin at 479 nm [23], Curcumin at 425 nm [34], Camptothecin at 480 nm, [68], 5-Fluorouracil at 266 nm [21], Cisplatin at 300 nm [18], Imanitib at 260 nm [31], Telmisartan at 296 nm [20], and Tamoxifen at 250 nm [55].

The time of loading is one of the essential factors in drug-loading. Figure 2 shows a summary of the drug-loading efficiency results reported in the literature. The highest efficiency for drug-loading (98,3%) is reporting for calcium ferrite loaded with Curcumin in ethanol solvent at 100 mL with a drug-loading capacity of 0.4, at room temperature for 3 h [34]. The lowest efficiency for drug-loading (8,4%) is reporting for cobalt ferrite. It is loaded with Docetaxel in 10 mL of dichloromethane at room temperature for 1 h.

Figure 2.

Summary of the drug-loading percentage as a function of the time reported in the literature. All the data plotted are shown in Table A2.

Other alternatives for drug-loading of nanoferrites composites include:

  1. Anchored nanoferrites of cobalt [28] and manganese [23] on graphene oxide were developed for controlled drug delivery nanocomposites.

  2. Doxorubicin and nickel ferrite nanoparticles were incorporate into N-carboxymethyl chitosan/poly(ε-caprolactone) nanofibers for drug delivery applications [51].

Advertisement

5. Drug delivery of functionalized nanoferrites for cancer treatments

Conventional drug delivery methods rely on the absorption of drugs and transport across biological membranes through diffusion and systemic transport. The targeted drug delivery, on the contrary, focuses on enhancing the concentration of the chemotherapeutic drug in the disease parts of the body [87]. The drug release studies, usually, are realized in simulated physiological conditions and measured by HPLC [30] or UV–Vis spectra [88]. For UV–vis spectrophotometer, the percentage of release drug is given by [49]:

Drug release%=amount of release drugamount of loaded drugx100E2

The drug release mechanisms evaluate with different models, such as zero-order, first-order, Higuchi, Korsmeyer-Peppas, and Hixson-Crowell. A detailed explanation of these five mathematical models to investigate drug release kinetic on in-vitro release data was reported by Jafari et al. [75]. The best mathematical model with a high correlation coefficient determines the suitable mathematical model and confirms drug release kinetics. Some results reported in the literature are summarized next:

  • Curcumin drug loading on vanillin-chitosan coated with calcium ferrite hybrid nanoparticles as a carrier [34]. In most cases, the release mechanism follows non-Fickian diffusion, which may be due to the porous nature of the material, swelling ability, or the presence of an excess amount of surface adsorbed drug on the nanoparticles.

  • Doxorubicin hydrochloride and Methotrexate drugs load on magnetite nanoparticles based on polyurethane matrices. The best fitting for the drug’s release was the Higuchi kinetic model [75].

  • Hesperidin drug loaded on magnetic casein-CaFe2O4 nanohybrid carrier conjugated with progesterone. Here, the release profile exhibiting the best fit towards the Higuchi model. Fickiand diffusion was validated as the release mechanism, which is a concentration gradient process [61].

  • Doxorubicin drug loaded on carboxymethyl chitosan/poly(ɛ-caprolactone)/doxorubicin/nickel ferrite core-shell fibers. Here, the Korsmeyer-Peppas model showed the best pharmacokinetic fit [51].

  • Hesperidin and Eugenol drugs loaded on folic acid functionalized BSA-CaFe2O4 nanohybrid carrier. The Korsmeyer-Peppas model showed the best fit for releasing the drug. The release mechanism at pH 1.2 is by anomalous diffusion. It is a combination of Fickian diffusion and the gradual erosion of the polymer. The release data for pH 5.8 and pH 7.4 fits well with the Higuchi model indicating a surface diffusion mechanism, in other words, the diffusion of the surface-bound [60].

  • Cisplatin drug loaded on magnetite nanocomposite. The results of the kinetic studies suggest that the most proper model to interpret the release of the drug in pH 5.5 is the Korsmeyer-Peppas equation. The value of the release exponent in this model suggests that the prime mechanism of drug release is diffusion and Fickian [78].

  • Doxorubicin drug loaded on pure and lanthanum doped bismuth ferrite nanostructures. The kinetic studies and adsorption isotherms revealed that the adsorption of the drug fitted well to the pseudo-second-order and Freundlich isotherm models. The adsorption of doxorubicin followed the multi-layered heterogeneous adsorption. The probable loading mechanism was electrostatic interaction [89].

The efficiency in-vitro tumor-targeted drug delivery of the nanoferrites loaded with anticancer drugs is evaluated by fluorescence microscopy imaging [58]. Here, the authors used human cancer cell lines as: MCF-7, A-549 [81], A431 [37], SKOV-3, MDA-MB-231 [61], SK-BR3 [33], MDA-MB-231, MCF-10A [90] in a culture media which is incubate in presence of the nanoferrites. Moreover, cytotoxicity determines the efficiency of the formulation [51]. The cytotoxic effect of the nanoformulation tests the cell viability (MTT) assay. It evaluated the ability of viable cells to reduce MTT to formazan crystals. The following equation used to calculate the % of cell viability is [91]:

%Cell viability=average sample readaverage control readx100E3

In-vivo antitumor therapy came tests in mice. Here, hepatoma cell lines (H22) inoculate into the back of the hind leg through subcutaneous injection. When the size of the tumor was grown to about 40 mm3 were treated with the drug-loading nanoferrites. All the formulations were injected intravenously through the tail of mice. The tumor inhibition rates could be determined by fluorescence microscopy [67]:

Tumor inhibition rates=1tumor volume with drug grouptumor volume in control groupx100E4

Nanoferrites have got importance in terms of biological applications due to their physicochemical properties. To enhance their cancer therapeutic effect stimuli-responsive combine treatments have been developed:

  • Magnetic hyperthermia therapy. Here, drug release may be activated applying an external alternating magnetic field which transforms electromagnetic energy into heat and induces the drug carrier to release its contents into the target site [34, 36, 57]. The combined techniques can enhance the therapeutic effect by increasing the blood flow and improving the oxygen supply to the tumor sites when increasing the temperature of the tumor sites from 37°C to hyperthermia temperature 42–45°C. This phenomenon can also enhance the drug delivery efficacy and increase the drug dosage to the target tumor sites. To determine the intracellular drug delivery of ferrite nanoparticles load with anticancer drugs, which were placed in dialysis membrane tubes and dialyzed at 37°C with different pHs [22].

  • Chemo-sonodynamic therapy. It is another strategy for cancer treatment because the low-intensity ultrasound caused the activation of drug-loaded magnetic nanosonosensitizers. With synthesized ultrasound-sensitive nanocarriers, chemo-sonodynamic therapy is a generator of cellular reactive oxygen species, mitochondrial damage, and inducer effect through the release of the loaded drug in magnetic nanoferrite [53].

  • Photodynamic therapy. Photodynamics is a method to treat cancer via light and photosensitizing chemical material. Here, small bandgap energy of the nanocarrier is desired to excite the electrons by light. The electrons transferred from the conduction band to the valence band produce an electron–hole pair. These pairs react with H2O and O2 and produce reactive oxygen species (ROS) [77].

  • Microwave irradiation. Microwave irradiation added to magnetite nanocomposite increases the drug release [19].

Usually, drug delivery is dramatically pH-dependent. Most of the papers reported in the literature studied the influence of pH on the release behavior of the carrier. pH variations at different physiological situations trigger a controlled delivery of drugs at different sites. The pH-responsive drug release under three conditions of simulated gastric fluid (pH 1.2), cancer microenvironment (pH 5.4), and simulated body fluid (pH 7.4) during a determined time [61]. Table A3 shows the influence of the pH on the release efficiency of the carrier. From there, in all cases, the acidic pH stimuli the rate of drug delivery. Table 2 shows the drug release percentage at cancer microenvironment conditions for nanoferrite formulations. The time of drug release is one of the essential factors in drug delivery. Figure 3 shows a summary of the drug delivery efficiency results reported in the literature. The highest efficiency for drug delivery (97%) is reporting for magnesium-cobalt ferrite loaded with 5-fluorouracil for 48 h [21]. The lowest efficiency for drug delivery (8,9%) was reported for magnetite load with Curcumin for 37 h.

SystempHt (h)T (°C)DR (%)Reference
Fe3O4 Doxorubicin5.5803760[58]
Mg0.5Co0.5
Fe2O4-5-fluorouracil
4.5483797[21]
CoFe2O4 -Doxorubicin5.4753742[83]
CoFe2O4 -Doxorubicin5.41203752[47]
CoFe2O4-Hesperidin and eugenol5.8243573.7[60]
MnFe2O4 -curcumin5.51203790[60]
CoFe2O4 - Doxorubicin4.0243760[43]
MnFe2O4- Doxorubicin5,5103717.16[24]
Fe3O4 - Curcumin5.01203740[92]
Fe3O4 -Telmisartan5.5523782[20]
ZnFe2O4 - Curcumin5.5963764.71[53]
Fe3O4- Cisplatin5,5483796[78]
Fe3O4-Doxorubicin5.8722570[26]
CoFe2O4-Doxorubicin5.4723780[42]

Table 2.

Summary of drug delivery conditions and results reported in the literature for ferrite nanoparticles loaded with anticancer drugs (system). The main conditions are the cancer microenvironments (pH), the time (t), and the temperature (T) of release. The drug release (DR) percentage measures the efficiency of the process.

Figure 3.

Summary of the drug delivery efficiency as a function of the time reported in the literature. All the data plotted are shown in Table 2 and A3.

Advertisement

6. Conclusions

Recent advances reviewed on synthetic routes for the obtention of nanoferrites for drug delivery applications. The most popular ferrite is magnetite obtained by chemical coprecipitation method with sizes ranging from 20 nm to 30 nm, and spherical shape. Moreover, it reviews the magnetic properties of ferrite nanoparticles. Often, the nanoferrites are superparamagnetic. Coated the nanoparticle’s surface with organic or inorganic molecules makes the nanostructures optimal for drug delivery applications. Functionalization reduces the agglomeration and toxicity of the nanoferrites. Physical adsorption among the functional groups of the cancer drugs and the coated molecules on the nanoparticles preserve the chemical structure of the medicament. Oncology drugs were detailed for drug delivery applications. The most popular solvent for drug-loading is water. It discussed the influence of parameters such as: pH, temperature, and time on drug-loading.

It reviewed the main drug release mechanisms for investigating pharmacokinetics. The release mechanism is highly dependent on the pH, the type of drug, and the nanocarrier. It discussed the stimuli-responsive combine treatments for cancer drug delivery applications. Some challenges persist:

  • The nonspecific accumulation of the drug and the lack of real-time monitoring of the delivery.

  • Develop novel multifunctional theranostic platforms with the abilities of intelligent controlled released and in-vivo site targeting delivery and treatment of illnesses.

  • Improve the delivery effectiveness of a drug by maintaining the concentration of the drug between the effective and toxic levels.

  • Inhibiting the dilution of the drug in the body fluids, and allowing targeting and localization of a drug at a specific site

  • Determine the optimal temperature and concentration of drug required to promote effective apoptosis.

Advertisement

Acknowledgments

C.L.L.C and J.A.J.H acknowledge Ministerio de Ciencia Tecnología e Innovación - Colombia (MINCIENCIAS, Call # 848). O.M.L and C.L.L.C acknowledge Unidad de Investigación from Universidad Autónoma de Manizales (Colombia).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

FTMethodS (nm)MoMS (emu/g)HC (Oe)MR (emu/g)R
CaCoprecipitation5Spherical12.83.40.06[17]
FeCoprecipitation7Spherical[18]
MnCoprecipitation10Semi- spherical1312.90.15[19]
FeCoprecipitation11Spherical590[20]
MnCoGlycol-Thermal12Spherical[21]
CoThermal decomposition13Spherical70.7*30.2[22]
MnSonochemical13Spherical34.900[23]
MnHydrolysis15Nanorod69.3116[24]
MnZnCoprecipitation15Spherical5600[25]
FeCoprecipitation15Spherical[26]
CoCoprecipitation15Spherical[27]
CoSonochemical15Spherical9400[28]
NiSolvothermal17Spherical7000[29]
HoHydrothermal21Spherical47.8[30]
ZnHydrothermal21Coral20[31]
FeThermal treatment23Spherical7.11143.92.21[32]
NiSol–Gel24Spherical[33]
CaCombustion of solutions25Spherical9.110.02[34]
CoCoprecipitation26Spherical49549[35]
CoMicrowave heating27Spherical90.5830[36]
CaCoprecipitation30Semi- spherical58.3[37]
FeHydrothermal30Hexagonal1.21750.91[38]
FePrecipitation30Spherical47.603.81[39]
FeCoprecipitation35Semi- spherical36.30[40]
FeCoprecipitation40Spherical1.5769.10.15[41]
CoCoprecipitation42Spherical36.000[42]
CoSolvothermal43Spherical47.7[43]
CoCoprecipitation43Spherical62.4[44]
CoPrecipitation50Nanofiber3.934092.1[45]
CoCoprecipitation50Spherical59.9[46]
CoSol–Gel50Semi- spherical64[47]
FeHydrothermal50Nanorods22.20[48]
FeCoprecipitation inverse60Spherical63[49]
CaCombustion of solutions60Spherical9.10.02[50]
NiSol–Gel70Spherical50.5[51]
HoHydrothermal74Semi- spherical43[52]
ZnSol–Gel75Spherical0.665.63.8[53]
ZnCoprecipitation80Semi- spherical31100[54]
FeCoprecipitation80Semi- spherical80.1[55]
FeHydrothermal90Hexagonal34714[56]
GdCoprecipitation90.1Spherical4700[57]
FeSolvothermal95Spherical59[58]
CoSolvothermal104Semi- spherical51.800[59]
CaSol–Gel112Spherical150.160.38[60]
CaSol–Gel112Spherical150.38[61]
MnCoprecipitation140Spherical56.142.65.22[62]
CoThermolysis157Spherical13.70[30]
FeHydrothermal200Spherical71.900[63]
CoThermolysis200Spherical51.100[64]
CoCoprecipitation250Spherical401.7[65]
MnCoprecipitation300Nanorods18[66]
FeSolvothermal300Spherical57.457.5[67]

Table A1.

Summary of nanoparticles ferrite type (FT) obtained by different methods with their sizes (S), morphology (Mo), saturation magnetization (MS), coercivity (HC) and remanence (MR), reported in the literature (R). The magnetic properties were reported at room temperature.

FTDrugSolQSLDT(°C)t(h)%LR
CoDD10RT18,4[30]
FeTE50.2301212[55]
CoDW303.3RT212,2[63]
NiCERT12[29]
CoDoW51[65]
CoDoW250.042524[83]
Mn-ZnDoW10RT2417.3[25]
MnCW25RT321,3[84]
FeC0.2RT0,523,7[85]
GdDoS50.2RT2424[57]
FeCW100.2RT2442[48]
CoDoRT2443[59]
CoMW0.005RT147[35]
FeCW250.3RT52,3[32]
CoDo0.67252453,3[42]
FeIW30RT2461[82]
CaHERT462,9[40]
ZnCE1363[60]
NiZW2055963.2[80]
CoDoRT2464[66]
CoDoW1252764.3[33]
CoDoW50.016302465[43]
FeDo0.5RT2465[27]
ZnZo42466,7[64]
Mn-Co5-FRT4867[58]
CoDoW529.85668,8[54]
FeCW50RT2472,4[21]
ZnCW500.016RT76,4[71]
CoDoW5RT178[41]
CaCi2586.5[53]
FeCisRT2487[86]
Mn5-F0.2RT1288[82]
CoDoRT2489[18]
CoDo0.2RT690[62]
ZnI0.42450,594,0[47]
CoDoS200372495[22]
MnDoW100.5RT2497[31]
CaE372497.5[37]
CaCE1000.4RT398,3[34]

Table A2.

Summary of nanoparticles ferrite type (FT) loaded (Fe: Magnetite) with different anticancer drugs (D: Docetaxel, T: Tamoxifen, C: Curcumin; do: Doxorubicin, M: Methotrexate, I: Imatinib, H: Hesperidin, Z:Zidovudine, zo:Zoledronic acid, 5-F: 5-fluorouracil, ci: Cinnamaldehyde, Cis: Cisplatin, and E: Epirubicin,), the solvent (sol) used for loading (D: Dichloromethane, E: Ethanol, W: Water, and S: Saline solution) and the quantity (QS, in mL), the loading capacity (LD), the temperature (T), time (t, in hours), and percentage (%L) of loading, with their references (R).

SystempHt (h)T (°C)DR (%)Reference
Fe3O4 - Doxorubicin7.4803720[58]
Mg0.5Co0.5
Fe2O4–5-fluorouracil
6.5
7.4
483773
73
[21]
Fe3O4- Docetaxel7.4483782.43[76]
CoFe2O4 -Doxorubicin7.41203722[47]
CoFe2O4- Hesperidin and Eugenol1.2
7.4
243587.44
54.29
[60]
MnFe2O4 - curcumin7.41203741[84]
CoFe2O4 - Doxorubicin7.45037.542.6[43]
CoFe2O4 - Doxorubicin7.4243730[59]
MnFe2O4 - Doxorubicin7.4103711.93[24]
CoFe2O4 - Curcumin7.4723757,1[77]
Fe3O4- Curcumin7.4120378,9[92]
CoFe2O4 - Docetaxel7.44083781[30]
Fe3O4 -Telmisartan7.4523725[20]
ZnFe2O4 - Curcumin7.4963776.45[53]
Fe3O4 - Tamoxifen7.41203775[55]
Ag(1-x)NiFe2O4 - Curcumin653174[29]
ZnFe2O4 - Curcumin61538[93]
NiFe2O4 -, CoFe2O4 - and Fe3O4 -Doxorubicin7.40.00693713.3[71]
Fe3O4 - Cisplatin7.4483793[78]
Fe3O4 - Doxorubicin6,862535[39]
Fe3O4 - Doxorubicin7722535[26]
CoGdxFe2-xO4 - Curcumin7.4243095[94]
CoFe2O4 - Doxorubicin7.4723780[27]
ErFe3O4- Camptothecin7.4453772[38]

Table A3.

Summary of drug delivery conditions and results, reported in the literature for ferrite nanoparticles loaded with anticancer drugs (system). The main conditions are the pH, the time (t) and the temperature (T) of release. The drug release (DR) percentage measure the efficiency of the process.

References

  1. 1. Aisida SO, Akpa PA, Ahmad I, Zhao T, Maaza M, Ezema FI. Bio-inspired encapsulation and functionalization of iron oxide nanoparticles for biomedical applications. Eur Polym J. 2020 Jan;122:109371.
  2. 2. Shende P, Shah P. Carbohydrate-based magnetic nanocomposites for effective cancer treatment. Int J Biol Macromol. 2021 Apr;175:281–93.
  3. 3. Pandey A. Role of Cyclodextrins in Nanoparticle-Based Systems for Drug Delivery. 2020;305–43.
  4. 4. Maghsoudnia N, Eftekhari RB, Sohi AN, Zamzami A, Dorkoosh FA. Application of nano-based systems for drug delivery and targeting: a review. J Nanoparticle Res. 2020 Aug;22(8):245.
  5. 5. Li F, Qin Y, Lee J, Liao H, Wang N, Davis TP, et al. Stimuli-responsive nano-assemblies for remotely controlled drug delivery. J Control Release. 2020 Jun;322:566–92.
  6. 6. Soares PIP, Romão J, Matos R, Silva JC, Borges JP. Design and engineering of magneto-responsive devices for cancer theranostics: Nano to macro perspective. Prog Mater Sci. 2021 Feb;116:100742.
  7. 7. Sadhasivam J, Sugumaran A. Magnetic nanocarriers: Emerging tool for the effective targeted treatment of lung cancer. J Drug Deliv Sci Technol. 2020 Feb;55:101493.
  8. 8. Wang Y, Miao Y, Li G, Su M, Chen X, Zhang H, et al. Engineering ferrite nanoparticles with enhanced magnetic response for advanced biomedical applications. Mater Today Adv. 2020 Dec;8:100119.
  9. 9. Zhao S, Yu X, Qian Y, Chen W, Shen J. Multifunctional magnetic iron oxide nanoparticles: an advanced platform for cancer theranostics. Theranostics. 2020;10(14):6278–309.
  10. 10. Amiri M, Khazaeli P, Salehabadi A, Salavati-Niasari M. Hydrogel beads-based nanocomposites in novel drug delivery platforms: Recent trends and developments. Adv Colloid Interface Sci. 2021 Feb;288:102316.
  11. 11. Avval ZM, Malekpour L, Raeisi F, Babapoor A, Mousavi SM, Hashemi SA, et al. Introduction of magnetic and supermagnetic nanoparticles in new approach of targeting drug delivery and cancer therapy application. Drug Metab Rev. 2020 Jan;52(1):157–84.
  12. 12. Al-Rawi NN, Anwer BA, Al-Rawi NH, Uthman AT, Ahmed IS. Magnetism in drug delivery: The marvels of iron oxides and substituted ferrites nanoparticles. Saudi Pharm J. 2020 Jul;28(7):876–87.
  13. 13. Kefeni KK, Msagati TAM, Nkambule TT, Mamba BB. Spinel ferrite nanoparticles and nanocomposites for biomedical applications and their toxicity. Mater Sci Eng C. 2020 Feb;107:110314.
  14. 14. Tripathy A, Nine MJ, Silva FS. Biosensing platform on ferrite magnetic nanoparticles: Synthesis, functionalization, mechanism and applications. Adv Colloid Interface Sci. 2021 Apr;290:102380.
  15. 15. Ghosh N, Pant P, Bhuvaneswari S. Chemical Methodologies for Preparation of Micron and Nanometer Scale Ferrites - A Mini Review of Patents. Recent Pat Nanotechnol. 2008 Jan;2(1):8–18.
  16. 16. Kharisov BI, Dias HVR, Kharissova O V. Mini-review: Ferrite nanoparticles in the catalysis. Arab J Chem. 2019 Nov;12(7):1234–46.
  17. 17. Pereira, Cardoso, Rodrigues, Amorim, Amaral, Almeida, et al. Magnetoliposomes Containing Calcium Ferrite Nanoparticles for Applications in Breast Cancer Therapy. Pharmaceutics. 2019 Sep;11(9):477.
  18. 18. Abdel-Bary AS, Tolan DA, Nassar MY, Taketsugu T, El-Nahas AM. Chitosan, magnetite, silicon dioxide, and graphene oxide nanocomposites: Synthesis, characterization, efficiency as cisplatin drug delivery, and DFT calculations. Int J Biol Macromol. 2020 Jul;154:621–33.
  19. 19. Yang Z, Wang L, Liu Y, Liu S, Tang D, Meng L, et al. ZnO capped flower-like porous carbon-Fe3O4 composite as carrier for bi-triggered drug delivery. Mater Sci Eng C. 2020 Feb;107:110256.
  20. 20. Dhavale RP, Dhavale RP, Sahoo SC, Kollu P, Jadhav SU, Patil PS, et al. Chitosan coated magnetic nanoparticles as carriers of anticancer drug Telmisartan: pH-responsive controlled drug release and cytotoxicity studies. J Phys Chem Solids. 2021 Jan;148:109749.
  21. 21. Mngadi S, Mokhosi S, Singh M, Mdlalose W. Chitosan-Functionalized Mg0.5Co0.5Fe2O4 Magnetic Nanoparticles Enhance Delivery of 5-Fluorouracil In Vitro. Coatings [Internet]. 2020 May 2;10(5):446. Available from: https://www.mdpi.com/2079-6412/10/5/446
  22. 22. Oh Y, Moorthy MS, Manivasagan P, Bharathiraja S, Oh J. Magnetic hyperthermia and pH-responsive effective drug delivery to the sub-cellular level of human breast cancer cells by modified CoFe2O4 nanoparticles. Biochimie. 2017 Feb;133:7–19.
  23. 23. Wang G, Ma Y, Zhang L, Mu J, Zhang Z, Zhang X, et al. Facile synthesis of manganese ferrite/graphene oxide nanocomposites for controlled targeted drug delivery. J Magn Magn Mater. 2016 Mar;401:647–50.
  24. 24. R.K. C, Rajagopalan V, Sahu NK. Synthesis of manganese doped β-FeOOH and MnFe 2 O 4 nanorods for enhanced drug delivery and hyperthermia application. IET Nanobiotechnology. 2020 Dec;14(9):823–9.
  25. 25. Montha W, Maneeprakorn W, Tang I-M, Pon-On W. Hyperthermia evaluation and drug/protein-controlled release using alternating magnetic field stimuli-responsive Mn–Zn ferrite composite particles. RSC Adv. 2020;10(66):40206–14.
  26. 26. Nieciecka D, Celej J, Żuk M, Majkowska-Pilip A, Żelechowska-Matysiak K, Lis A, et al. Hybrid System for Local Drug Delivery and Magnetic Hyperthermia Based on SPIONs Loaded with Doxorubicin and Epirubicin. Pharmaceutics. 2021 Apr;13(4):480.
  27. 27. Beagan AM, Alghamdi AA, Lahmadi SS, Halwani MA, Almeataq MS, Alhazaa AN, et al. Folic Acid-Terminated Poly(2-Diethyl Amino Ethyl Methacrylate) Brush-Gated Magnetic Mesoporous Nanoparticles as a Smart Drug Delivery System. Polymers (Basel). 2020 Dec;13(1):59.
  28. 28. Wang G, Ma Y, Wei Z, Qi M. Development of multifunctional cobalt ferrite/graphene oxide nanocomposites for magnetic resonance imaging and controlled drug delivery. Chem Eng J [Internet]. 2016 Apr;289:150–60. Available from: http://dx.doi.org/10.1016/j.cej.2015.12.072
  29. 29. Jose R, J R, Jothi NSN. The synthesis and characterisation of curcumin loaded Ag (1-X) Ni X Fe 2 O 4 for drug delivery. Mater Technol. 2021 May;36(6):339–46.
  30. 30. Panda J, Satapathy BS, Mandal B, Sen R, Mukherjee B, Sarkar R, et al. Anticancer potential of docetaxel-loaded cobalt ferrite nanocarrier: an in vitro study on MCF-7 and MDA-MB-231 cell lines. J Microencapsul. 2021 Jan;38(1):36–46.
  31. 31. Amiri M, Gholami T, Amiri O, Pardakhti A, Ahmadi M, Akbari A, et al. The magnetic inorganic-organic nanocomposite based on ZnFe2O4-Imatinib-liposome for biomedical applications, in vivo and in vitro study. J Alloys Compd [Internet]. 2020 Dec;849:156604. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0925838820329686
  32. 32. Ehsan N, Magmoud N, Heidar TR, Reza ZE, Gholamreza F, Ramezan AT. In vivo and In vitro Biocompatibility Study of Fe3O4@ZnO and Fe3O4@SiO2 as Photosensitizer for Targeted Breast Cancer Drug Delivery. J Sci. 2020;31(4):12.
  33. 33. Joshy KS, Augustine R, Mayeen A, Alex SM, Hasan A, Thomas S, et al. NiFe 2 O 4 /poly(ethylene glycol)/lipid–polymer hybrid nanoparticles for anti-cancer drug delivery. New J Chem. 2020;44(42):18162–72.
  34. 34. Kamaraj S, Palanisamy UM, Kadhar Mohamed MSB, Gangasalam A, Maria GA, Kandasamy R. Curcumin drug delivery by vanillin-chitosan coated with calcium ferrite hybrid nanoparticles as carrier. Eur J Pharm Sci [Internet]. 2018 Apr;116:48–60. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0928098718300307
  35. 35. Shahzad K, Mushtaq S, Rizwan M, Khalid W, Atif M, Din FU, et al. Field-controlled magnetoelectric core-shell CoFe2O4@BaTiO3 nanoparticles as effective drug carriers and drug release in vitro. Mater Sci Eng C. 2021 Feb;119:111444.
  36. 36. Radmansouri M, Bahmani E, Sarikhani E, Rahmani K, Sharifianjazi F, Irani M. Doxorubicin hydrochloride - Loaded electrospun chitosan/cobalt ferrite/titanium oxide nanofibers for hyperthermic tumor cell treatment and controlled drug release. Int J Biol Macromol. 2018 Sep;116:378–84.
  37. 37. Shahabadi N, Razlansari M, Khorshidi A, Zhaleh H. Investigation of controlled release properties and anticancer effect of folic acid conjugated magnetic core–shell nanoparticles as a dual responsive drug delivery system on A-549 and A-431 cancer cell lines. Res Chem Intermed. 2020 Sep;46(9):4257–78.
  38. 38. Kaliyamoorthi K, Sumohan Pillai A, Alexander A, Ramasamy S, Arivarasu A, Enoch IVM V. Designed poly(ethylene glycol) conjugate-erbium-doped magnetic nanoparticle hybrid carrier: enhanced activity of anticancer drug. J Mater Sci. 2021 Feb;56(5):3925–34.
  39. 39. Ren M-X, Wang Y-Q, Lei B-Y, Yang X-X, Hou Y-L, Meng W-J, et al. Magnetite nanoparticles anchored on graphene oxide loaded with doxorubicin hydrochloride for magnetic hyperthermia therapy. Ceram Int. 2021 Jul;47(14):20686–92.
  40. 40. Karimi Ghezeli Z, Hekmati M, Veisi H. Synthesis of Imatinib-loaded chitosan-modified magnetic nanoparticles as an anti-cancer agent for pH responsive targeted drug delivery. Appl Organomet Chem. 2019 Apr;33(4):e4833.
  41. 41. Rostami M, Aghajanzadeh M, Zamani M, Manjili HK, Danafar H. Sono-chemical synthesis and characterization of Fe3O4@mTiO2-GO nanocarriers for dual-targeted colon drug delivery. Res Chem Intermed. 2018 Mar;44(3):1889–904.
  42. 42. Arkaban H, Khajeh Ebrahimi A, Yarahmadi A, Zarrintaj P, Barani M. Development of a multifunctional system based on CoFe 2 O 4 @polyacrylic acid NPs conjugated to folic acid and loaded with doxorubicin for cancer theranostics. Nanotechnology. 2021 Jul;32(30):305101.
  43. 43. Zhang H, Wang J, Zeng Y, Wang G, Han S, Yang Z, et al. Leucine-coated cobalt ferrite nanoparticles: Synthesis, characterization and potential biomedical applications for drug delivery. Phys Lett A. 2020 Aug;384(24):126600.
  44. 44. Amiri M, Akbari A, Ahmadi M, Pardakhti A, Salavati-Niasari M. Synthesis and in vitro evaluation of a novel magnetic drug delivery system; proecological method for the preparation of CoFe2O4 nanostructures. J Mol Liq. 2018 Jan;249:1151–60.
  45. 45. Sangeetha K, Vidhya G, Girija EK, Ashok M. Fabrications of magnetic responsive hydroxyapatite platform: In vitro release of chemo drug for cancer therapy. Mater Today Proc. 2020;26:3579–82.
  46. 46. Hassanzadeh-Tabrizi SA, Norbakhsh H, Pournajaf R, Tayebi M. Synthesis of mesoporous cobalt ferrite/hydroxyapatite core-shell nanocomposite for magnetic hyperthermia and drug release applications. Ceram Int. 2021 Jul;47(13):18167–76.
  47. 47. Ghanbari M, Davar F, Shalan AE. Effect of rosemary extract on the microstructure, phase evolution, and magnetic behavior of cobalt ferrite nanoparticles and its application on anti-cancer drug delivery. Ceram Int. 2021 Apr;47(7):9409–17.
  48. 48. Kermanian M, Naghibi M, Sadighian S. One-pot hydrothermal synthesis of a magnetic hydroxyapatite nanocomposite for MR imaging and pH-Sensitive drug delivery applications. Heliyon. 2020 Sep;6(9):e04928.
  49. 49. Pooresmaeil M, Namazi H. β-Cyclodextrin grafted magnetic graphene oxide applicable as cancer drug delivery agent: Synthesis and characterization. Mater Chem Phys. 2018 Oct;218:62–9.
  50. 50. Bilas R, Sriram K, Maheswari PU, Sheriffa Begum KMM. Highly biocompatible chitosan with super paramagnetic calcium ferrite (CaFe2O4) nanoparticle for the release of ampicillin. Int J Biol Macromol. 2017 Apr;97:513–25.
  51. 51. Abasalta M, Asefnejad A, Khorasani MT, Saadatabadi AR. Fabrication of carboxymethyl chitosan/poly(ε-caprolactone)/doxorubicin/nickel ferrite core-shell fibers for controlled release of doxorubicin against breast cancer. Carbohydr Polym. 2021 Apr;257:117631.
  52. 52. Kaliyamoorthi K, Ramasamy S, Pillai AS, Alexander A, Arivarasu A, Enoch IVMV. Camptothecin-loaded holmium ferrite nanocarrier. Expanded activity on breast cancer cells. Mater Lett. 2021 Feb;285:129164.
  53. 53. Hafezi M, Rostami M, Hosseini A, Rahimi-Nasrabadi M, Fasihi-Ramandi M, Badiei A, et al. Cur-loaded ZnFe2O4@mZnO@N-GQDs biocompatible nano-carriers for smart and controlled targeted drug delivery with pH-triggered and ultrasound irradiation. J Mol Liq. 2021 Jan;322:114875.
  54. 54. Seyfoori A, Ebrahimi SAS, Omidian S, Naghib SM. Multifunctional magnetic ZnFe2O4-hydroxyapatite nanocomposite particles for local anti-cancer drug delivery and bacterial infection inhibition: An in vitro study. J Taiwan Inst Chem Eng. 2019 Mar;96:503–8.
  55. 55. Theragnostic Magnetic Core-Shell Nanoparticle as Versatile Nanoplatform for Magnetic Resonance Imaging and Drug Delivery. Biointerface Res Appl Chem [Internet]. 2021 Feb 8;11(5):13276–89. Available from: https://biointerfaceresearch.com/wp-content/uploads/2021/02/20695837115.1327613289.pdf
  56. 56. Ramasamy S, Dhamecha D, Kaliyamoorthi K, Pillai AS, Alexander A, Dhanaraj P, et al. Magnetic hydroxyapatite nanomaterial–cyclodextrin tethered polymer hybrids as anticancer drug carriers. Mater Adv. 2021;2(10):3315–27.
  57. 57. Mekonnen TW, Birhan YS, Andrgie AT, Hanurry EY, Darge HF, Chou H-Y, et al. Encapsulation of gadolinium ferrite nanoparticle in generation 4.5 poly(amidoamine) dendrimer for cancer theranostics applications using low frequency alternating magnetic field. Colloids Surfaces B Biointerfaces. 2019 Dec;184:110531.
  58. 58. Fang Z, Li X, Xu Z, Du F, Wang W, Shi R, et al. Hyaluronic acid-modified mesoporous silica-coated superparamagnetic Fe3O4 nanoparticles for targeted drug delivery. Int J Nanomedicine. 2019 Jul;Volume 14:5785–97.
  59. 59. Shi Z, Zeng Y, Chen X, Zhou F, Zheng L, Wang G, et al. Mesoporous superparamagnetic cobalt ferrite nanoclusters: Synthesis, characterization and application in drug delivery. J Magn Magn Mater. 2020 Mar;498:166222.
  60. 60. Uma Maheswari P, Muthappa R, Bindhya KP, Meera Sheriffa Begum KM. Evaluation of folic acid functionalized BSA-CaFe2O4 nanohybrid carrier for the controlled delivery of natural cytotoxic drugs hesperidin and eugenol. J Drug Deliv Sci Technol. 2021 Feb;61:102105.
  61. 61. K. Purushothaman B, P UM, K. M. MSB. Magnetic casein-CaFe2O4 nanohybrid carrier conjugated with progesterone for enhanced cytotoxicity of citrus peel derived hesperidin drug towards breast and ovarian cancer. Int J Biol Macromol. 2020 May;151:293–304.
  62. 62. Karimi Z, Abbasi S, Shokrollahi H, Yousefi G, Fahham M, Karimi L, et al. Pegylated and amphiphilic Chitosan coated manganese ferrite nanoparticles for pH-sensitive delivery of methotrexate: Synthesis and characterization. Mater Sci Eng C. 2017 Feb;71:504–11.
  63. 63. Panda J, Satapathy BS, Majumder S, Sarkar R, Mukherjee B, Tudu B. Engineered polymeric iron oxide nanoparticles as potential drug carrier for targeted delivery of docetaxel to breast cancer cells. J Magn Magn Mater. 2019 Sep;485:165–73.
  64. 64. Fan H, Li B, Shi Z, Zhao L, Wang K, Qiu D. A fibrous morphology silica-CoFe2O4 nanocarrier for anti-cancer drug delivery. Ceram Int. 2018 Feb;44(2):2345–50.
  65. 65. Mandal Goswami M. Synthesis of Micelles Guided Magnetite (Fe3O4) Hollow Spheres and their application for AC Magnetic Field Responsive Drug Release. Sci Rep. 2016 Dec;6(1):35721.
  66. 66. Abbasi Pour S, Shaterian HR, Afradi M, Yazdani-Elah-Abadi A. Carboxymethyl cellulose (CMC)-loaded Co-Cu doped manganese ferrite nanorods as a new dual-modal simultaneous contrast agent for magnetic resonance imaging and nanocarrier for drug delivery system. J Magn Magn Mater. 2017 Sep;438:85–94.
  67. 67. Liu X, Wang C, Wang X, Tian C, Shen Y, Zhu M. A dual-targeting Fe3O4@C/ZnO-DOX-FA nanoplatform with pH-responsive drug release and synergetic chemo-photothermal antitumor in vitro and in vivo. Mater Sci Eng C. 2021 Jan;118:111455.
  68. 68. Kaliyamoorthy K, Pillai AS, Alexander A, Arivarasu A, Enoch IVMV, Ramasamy S. β-Cyclodextrin-folate functionalized poly(lactic-co-glycolide)–superparamagnetic ytterbium ferrite hybrid nanocarrier for targeted delivery of camptothecin. Mater Sci Eng C. 2021 Mar;122:111796.
  69. 69. Mehnath S, Das AK, Verma SK, Jeyaraj M. Biosynthesized/green-synthesized nanomaterials as potential vehicles for delivery of antibiotics/drugs. In 2021. p. 363–432.
  70. 70. Pinelli F, Perale G, Rossi F. Coating and Functionalization Strategies for Nanogels and Nanoparticles for Selective Drug Delivery. Gels. 2020 Feb;6(1):6.
  71. 71. Mannu R, Karthikeyan V, Velu N, Arumugam C, Roy VAL, Gopalan A-I, et al. Polyethylene Glycol Coated Magnetic Nanoparticles: Hybrid Nanofluid Formulation, Properties and Drug Delivery Prospects. Nanomaterials. 2021 Feb;11(2):440.
  72. 72. Seyfoori A, Ebrahimi SAS, Omidian S, Naghib SM. Multifunctional magnetic ZnFe2O4-hydroxyapatite nanocomposite particles for local anti-cancer drug delivery and bacterial infection inhibition: An in vitro study. J Taiwan Inst Chem Eng [Internet]. 2019 Mar;96:503–8. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1876107018305777
  73. 73. Day NB, Wixson WC, Shields CW. Magnetic systems for cancer immunotherapy. Acta Pharm Sin B. 2021 Apr;
  74. 74. Mandal P, Panja S, Banerjee SL, Ghorai SK, Maji S, Maiti TK, et al. Magnetic particle anchored reduction and pH responsive nanogel for enhanced intracellular drug delivery. Eur Polym J. 2020 Apr;129:109638.
  75. 75. Jafari S, Soleimani M, Badinezhad M. Application of different mathematical models for further investigation of in vitro drug release mechanisms based on magnetic nano-composite. Polym Bull. 2021 Jan;
  76. 76. Song C, Gao C, Zhao J, Wang Z. Construction of long-circulation EpCAM targeted drug delivery system and its application in the diagnosis and treatment of breast cancer. J Biomater Appl. 2021 Mar;35(8):947–57.
  77. 77. Naderi E, Aghajanzadeh M, Zamani M, Sharafi A, Naseri M, Danafar H. The Effect of Calcination Temperature on the Anticancer Activity of CaFe2O4@PVA Nanocarriers: Photodynamic Therapy and Drug Delivery Study. J Inorg Organomet Polym Mater. 2020 Dec;30(12):5261–9.
  78. 78. Siavashy S, Soltani M, Ghorbani-Bidkorbeh F, Fallah N, Farnam G, Mortazavi SA, et al. Microfluidic platform for synthesis and optimization of chitosan-coated magnetic nanoparticles in cisplatin delivery. Carbohydr Polym. 2021 Aug;265:118027.
  79. 79. Zhalechin M, Dehaghi SM, Najafi M, Moghimi A. Magnetic polymeric core-shell as a carrier for gradual release in-vitro test drug delivery. Heliyon. 2021 May;7(5):e06652.
  80. 80. Sawant VJ, Bamane SR, Shejwal RV, Patil SB. Comparison of drug delivery potentials of surface functionalized cobalt and zinc ferrite nanohybrids for curcumin in to MCF-7 breast cancer cells. J Magn Magn Mater. 2016 Nov;417:222–9.
  81. 81. 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 (Basel). 2020 Aug;12(8):1747.
  82. 82. Purushothaman BK, Maheswari P U, Sheriffa Begum K M M. <scp>pH</scp> and magnetic field responsive protein-inorganic nanohybrid conjugated with biotin: A biocompatible carrier system targeting lung cancer cells. J Appl Polym Sci. 2021 Mar;138(10):49949.
  83. 83. Cheraghi A, Davar F, Homayoonfal M, Hojjati-Najafabadi A. Effect of lemon juice on microstructure, phase changes, and magnetic performance of CoFe2O4 nanoparticles and their use on release of anti-cancer drugs. Ceram Int. 2021 Jul;47(14):20210–9.
  84. 84. Aghajanzadeh M, Naderi E, Zamani M, Sharafi A, Naseri M, Danafar H. In vivo and in vitro biocompatibility study of MnFe 2 O 4 and Cr 2 Fe 6 O 12 as photosensitizer for photodynamic therapy and drug delivery of anti-cancer drugs. Drug Dev Ind Pharm. 2020 May;46(5):846–51.
  85. 85. Ayyanaar S, Kesavan MP, Sivaraman G, Maddiboyina B, Annaraj J, Rajesh J, et al. A novel curcumin-loaded PLGA micromagnetic composite system for controlled and pH-responsive drug delivery. Colloids Surfaces A Physicochem Eng Asp. 2019 Jul;573:188–95.
  86. 86. Dey C, Baishya K, Ghosh A, Goswami MM, Ghosh A, Mandal K. Improvement of drug delivery by hyperthermia treatment using magnetic cubic cobalt ferrite nanoparticles. J Magn Magn Mater. 2017 Apr;427:168–74.
  87. 87. Kaliyamoorthy K, Pillai AS, Alexander A, Arivarasu A, Enoch IVMV, Ramasamy S. β-Cyclodextrin-folate functionalized poly(lactic-co-glycolide)–superparamagnetic ytterbium ferrite hybrid nanocarrier for targeted delivery of camptothecin. Mater Sci Eng C [Internet]. 2021 Mar;122:111796. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0928493120337152
  88. 88. Ehi-Eromosele CO, Ita BI, Iweala EEJ. Silica coated LSMO magnetic nanoparticles for the pH-Responsive delivery of 5-Fluorouracil anticancer drug. Colloids Surfaces A Physicochem Eng Asp. 2017 Oct;530:164–71.
  89. 89. Abbasi MA, Ali Z, Qamar Z, Shahzad K, Siddiqui HK, Atif M, et al. Phase pure synthesis of lanthanum doped bismuth ferrite nanostructures for the adsorption of doxorubicin. Ceram Int. 2021 May;47(10):14390–8.
  90. 90. Klein S, Distel LVR, Neuhuber W, Kryschi C. Caffeic Acid, Quercetin and 5-Fluorocytidine-Functionalized Au-Fe3O4 Nanoheterodimers for X-ray-Triggered Drug Delivery in Breast Tumor Spheroids. Nanomaterials. 2021 Apr;11(5):1167.
  91. 91. Jermy R, Ravinayagam V, Alamoudi W, Almohazey D, Elanthikkal S, Dafalla H, et al. Tuning pH sensitive chitosan and cisplatin over spinel ferrite/silica nanocomposite for anticancer activity in MCF-7 cell line. J Drug Deliv Sci Technol. 2020 Jun;57:101711.
  92. 92. Asgari M, Miri T, Soleymani M, Barati A. A novel method for in situ encapsulation of curcumin in magnetite-silica core-shell nanocomposites: A multifunctional platform for controlled drug delivery and magnetic hyperthermia therapy. J Mol Liq. 2021 Feb;324:114731.
  93. 93. Jose R, J R, Jothi NSN. Synthesis and characterisation of stimuli-responsive drug delivery system using ZnFe 2 O 4 and Ag 1-X Zn x Fe 2 O 4 nanoparticles. Mater Technol. 2021 May;36(6):347–55.
  94. 94. Javed F, Abbas MA, Asad MI, Ahmed N, Naseer N, Saleem H, et al. Gd3+ Doped CoFe2O4 Nanoparticles for Targeted Drug Delivery and Magnetic Resonance Imaging. Magnetochemistry. 2021 Mar;7(4):47.

Written By

Felipe Ocampo Osorio, Jhon Augusto Jativa Herrera, Oscar Moscoso Londoño and César Leandro Londoño Calderón

Submitted: 09 April 2021 Reviewed: 31 August 2021 Published: 17 November 2021