Contemporary Oncology
eISSN: 1897-4309
ISSN: 1428-2526
Contemporary Oncology/Współczesna Onkologia
Current issue Archive Manuscripts accepted About the journal Supplements Addendum Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
4/2025
vol. 29
 
Share:
Share:
Review paper

Advancements and future directions of iron oxide nanoparticles in cancer therapy

Ahmed el-Mallul
1
,
Ryszard Tomasiuk
1
,
Piotr Jaworski
1
,
Sadia Mughali
2

  1. University of Radom, Faculty of Medical Sciences and Health Sciences, Radom, Poland
  2. Bahria University Medical and Dental College (BUMDC), Department of Medicine, Karachi, Pakistan
Contemp Oncol (Pozn) 2025; 29 (4): 323–332
Online publish date: 2025/11/05
Article file
Get citation
 
PlumX metrics:
 

Introduction

Despite significant advances in cancer diagnosis and treatment, current prevention, early detection, and therapeutic strategies have not been fully utilized or realized, and remain insufficient to effectively reduce cancer incidence and mortality worldwide [1]. This ongoing challenge drives scientists to continually improve and innovate existing methods for cancer diagnosis, treatment, and prognosis [2]. Cancer is characterized by the uncontrolled division of cells, where mutated cells proliferate rapidly and evade natural cell death mechanisms [3]. Classical methods, such as radiotherapy, surgery, and chemotherapy, are widely used for tumor detection and treatment; however, they often fail to accurately detect cancer in its early stages and are associated with various unwanted side effects. According to a survey, approximately 9.6 million people die from cancer each year, and this number is predicted to rise to 13.1 million by 2030 if current trends persist [4].

Magnetic nanoparticles (MNPs) are nanosized materials (typically 5–150 nm) exhibiting ferromagnetic, ferrimagnetic, or superparamagnetic properties. These enable precise control over their delivery to target organs via an external magnetic field. Among MNPs, iron oxide nanoparticles (IONPs) are the most widely used in biomedicine. Based on molecular structure and iron oxidation state, the three common forms of IONPs correspond to natural iron oxide minerals: magnetite (Fe3O4), maghemite (γ-Fe2O3), and hematite (α-Fe2O3). Fe3O4 and γ-Fe2O3 are particularly prevalent in diagnostics (e.g., as contrast agents) and therapeutic applications, including magnetic hyperthermia, targeted drug delivery, and tumor sensitization [5, 6].

Toxicity and clearance kinetics

The potential of superparamagnetic iron-oxide nanoparticles (SPIONs) is ultimately dictated by their biocompatibility and clearance kinetics, as rapid recognition by the mononuclear phagocyte system (MPS) can curtail circulation half-life and provoke dose-limiting toxicities, with uptake efficiency influenced by physicochemical determinants such as size (20–50 nm particles cleared faster than 100 nm analogs), shape (rods exhibiting higher macrophage association than spheres of equal volume), and surface chemistry (neutral or slightly negative ζ-potentials [–10 to 0 mV] reducing opsonization relative to cationic surfaces > +20 mV), while coating strategies such as PEGylation, dextran coronas, silica shells, or zwitterionic polymers enhance colloidal stability and suppress protein corona formation, thus extending systemic circulation [79].

Administration routes also impact biodistribution and toxicity – intravenous injection subjects SPIONs to serum proteins and MPS filtration, inhalation promotes pulmonary retention, and topical application limits systemic exposure while enhancing skin penetration [10].

Cytotoxicity mechanisms include oxidative stress via the Fenton reaction and NADPH oxidase activation (ROS, lipid peroxidation, DNA damage), membrane disruption by electrostatic interactions, mitochondrial depolarization ↓ATP, caspase-3 activation), and pro-inflammatory cytokine release (TNF-α, IL-6), with uncoated SPIONs producing 2–4 times higher ROS levels than polymer-coated analogues [11]. Long-term exposure may cause iron accumulation in liver Kupffer cells and splenic red-pulp macrophages, leading to ferritin upregulation and potential fibrosis beyond 30 days [12].

Mitigation strategies encompass surface engineering (PEG, chitosan, zwitterionic brushes reducing protein adsorption > 80%), use of biodegradable cores (e.g., Fe3O4@SiO2 with enzymatically cleavable linkers) to enable renal and hepatobiliary clearance, and active targeting (e.g., RGD peptides or anti-HER2 scFv) to enhance tumor accumulation while minimizing off-target effects, with standardized toxicity and performance assays (e.g., MTT/WST-1, DCF-DA ROS, γ-H2AX, hemocompatibility) recommended alongside comprehensive particle characterization including size, ζ-potential, and relaxivity (r2) [13] (Table 1).

Table 1

Factors influencing iron oxide nanoparticle cytotoxicity [14]

FactorBiological impactRef.
SizeSmaller sizes increase cellular uptake, potentially causing toxicity[15]
Surface chemistryUncoated surfaces trigger immune responses[16]
Dose/exposureHigh doses or prolonged exposure induce stress[17]
CompositionMetal cores (e.g., Fe, Co) may release toxic ions[18]
AggregationAggregates disrupt cellular function[19]

The scientific community acknowledges the urgent need for early-stage cancer diagnosis and treatment, which has led to a surge in the development of new drugs and innovative therapies. Nanotechnology is now being employed to create new materials with the potential for precise and accurate cancer detection and treatment, a field known as nano-oncology [20, 21]. Nanotechnology has been integrated with various other technologies, such as optical technology and molecular biology, to develop new toolkits for cancer imaging and treatment [22].

Nanotechnology, often described as the science of small, involves the study and manipulation of materials that have at least one dimension in the nanoscale range (1–100 nm) [23, 24]. Various approaches, including physical, biological, and chemical methods, are used to develop nanomedicines. However, significant efforts are being made to develop nanoparticles that are eco-friendly, biocompatible, and biodegradable. There have been reports of nanomaterial synthesis using bio-chemical methods, also known as green nanotechnology, which demonstrate low toxicity to blood cells and effectiveness against bacterial strains, parasites, and cancer cell lines [25].

The first nanoparticle-based drug approved by the USA Food and Drug Administration was Doxil (liposomal doxorubicin) [26]. This approval, granted about two decades ago, came approximately 30 years after the first publication on liposomes in the mid-1960s. In the past five decades, substantial progress has been made in research and clinical nanomedicines [27]. According to ClinicalTrials.gov, at the time of writing, there are around 700 nanomedicines under trial.

In the realm of fundamental research, the discovery of the enhanced permeability and retention (EPR) effect has been pivotal in the development of nanosystems [28, 29]. However, challenges such as heterogeneity and susceptibility of these systems remain, impacting their effectiveness [30]. Cancer heterogeneity, which promotes the development of the tumor microenvironment (TME), has provided insights into the effects of nanomedicine on tumors [31]. It has been established that active tumor targeting, leading to nanomedicine accumulation at the tumor site, is a more effective approach compared to traditional therapies [32, 33]. The size, shape, and physicochemical properties of nanoparticles significantly influence their therapeutic properties, laying the foundation for nano-theranostics – the use of nanoparticles as both diagnostic and therapeutic agents [34]. The stimulation of the immune system and nano-bio-catalysis are also highlighted as promising strategies for cancer and tumor inhibition [3537]. Results from currently developed nano-theranostics have provided direction for the development of more effective nanomedicines in the future [28, 38, 39].

Despite the challenges associated with the interaction of nanomedicines with tumors, they are proving to be promising candidates for cancer treatment. Current studies indicate that the uptake rate of nanoparticles by tumors is only 0.7% of the injected dose, increasing to 0.9% when the nanoparticle surface is ligand-modified [40, 41]. Nevertheless, nanotechnology has made considerable breakthroughs in cancer treatment, showing promising results when used as chemotherapeutic agents and for hyperthermal applications [4244]. New discoveries are being made in the development of multifunctional nanomaterials that can be used for both diagnosis and treatment, including the introduction of smart nanomaterials that use various targeting approaches when introduced to tumors [4549]. Nanoparticles are also being tagged with antibodies to achieve target specificity, with minimal effects on normal or healthy blood cells [5052].

The aim of this review was to compile comprehensive data on the various methods used to develop nanomaterials, compare their sizes and anticancer efficacy, and explore the strategies employed by researchers to diagnose and treat cancer. This review will serve as a resource for scientists and researchers to generate new ideas for developing materials with better outcomes and lower toxicity.

Synthesis of iron oxide nanoparticles

The synthesis of IONPs is a critical factor that determines their physicochemical properties and, consequently, their efficacy in biomedical applications. Several synthesis methods have been developed, each with its advantages and limitations.

Chemical methods

The most commonly employed method for synthesizing IONPs is chemical co-precipitation, where ferrous and ferric salts are precipitated in an alkaline medium to form magnetite (Fe3O4) or maghemite (γ-Fe2O3) nanoparticles [23, 24]. This method is advantageous due to its simplicity and scalability, but it often results in a broad size distribution and requires careful control of reaction conditions to prevent oxidation and agglomeration of the nanoparticles [25]. Moreover, the environmental impact of the byproducts generated during chemical synthesis remains a concern [26] (Table 2).

Table 2

Chemically synthesized nanoparticles

Type of NPsChemical methodSize of NPs [nm]ApplicationRef.
FeOPolyol method13.8Magnetic particle imaging[53]
FeOMicroemulsions16Biomedical applications[54]
FeOThermal decomposition2–30Magnetic hyperthermia[55]
Au@CuElectro-deposition30–70Electrochemical applications[56]

[i] NPs – nanoparticles

Biological methods

To address the environmental issues associated with chemical synthesis, biological methods using microorganisms or plant extracts have been explored [27, 28]. These methods are considered more environmentally friendly and can produce nanoparticles with a narrower size distribution and higher biocompatibility. However, they often suffer from lower yields and longer reaction times, which may limit their practical application [29, 30] (Table 3).

Table 3

Biological synthesis of nanoparticles

Type of NPsPlant/microorganism nameApplicationSize of nanoparticles [nm]Ref.
TiO2Bacillus subtilisPhotocatalytic10–30[57]
TiO2Lactobacillus sp.Antibiotic50–100[58]
ZnOCochliobolus geniculatusAntibacterial2–6[59]
CuShewanella loihicaAntibacterial10–16[60]

[i] NPs – nanoparticles

Physical methods

Physical methods, such as thermal decomposition and microemulsion, offer greater control over particle size and shape, leading to more uniform and monodisperse nanoparticles [61]. However, these methods typically require high temperatures, organic solvents, and surfactants, which can be toxic and complicate the purification process [31] (Tables 4, 5).

Table 4

Synthesis of nanoparticles using physical methods

Type
of NPs
Ball mill type/timePowder size (initial) [µm]Final size [nm]ApplicationsCharacterizationRef.
ZnOHorizontal
oscillatory mill/50 h
≈ 0.6–130AntibacterialXRD, TEM, SEM, HEBM[62]
ZnFe2O4Ultrasonic
wave-assisted
ball mill/80 h
6020BiomedicalXRD, SEM[63]
Fe3O4Planetary
ball mill/30 h
4020.5BiomedicalXRD, SEM[64]

[i] NPs – nanoparticles

Table 5

Synthesis of nanoparticles using LASER ablation

Type of NPsLASER wavelength
[nm]
Ablation duration [min]SourceLASER sourceFinal SizeRef.
Au106230Pure Au plateNd-YAG30 nm[65]
ZnO35540Metallic Zn foilNd-YAG5–19 nm[66]
Ag53230Metallic Ag foilNd-YAG2–5 nm[67]
TiO2106410Ti plateYtterbium doped Fiber LASER5–25 nm[68]

[i] NPs – nanoparticles

Bio-chemical

The bio-chemical method is a hybrid technique, in which precursor salt is introduced to biological extracts, mostly plant-derived. Extracts are prepared using alcohols or water from various plant parts [69, 70], including bark, leaves, flowers, fruits, and/or roots. Alcohol-based extraction typically does not require heating, whereas water-based extraction involves heating to release phytochemicals. After obtaining the extract, the precursor salt is added, and mild heat is applied to produce uniformly distributed nanoparticles. Although this method is more time-consuming than chemical synthesis, it is considerably more environmentally friendly. Table 6 contains various examples of nanoparticles synthesized using the bio-chemical method, the part of plant used for their synthesis, their sizes, and their bio-medical applications.

Table 6

Bio-chemical method mediated synthesis of nanoparticles

Type of NPsPart of plantName of plantSize of NPs [nm]ApplicationsRef.
FeOFlowerCallistemon viminalis~21Biomedical[24]
CuOLeavesAcalypha indica26Antimicrobial[71]
NiOFlowerCallistemon viminalis16.5Biomedical[23]
ZnOLeavesSageretia thea15.2Biomedical[72]

[i] NPs – nanoparticles

Functionalization and targeting strategies

The effectiveness of IONPs in cancer therapy largely depends on their ability to specifically target tumor cells while minimizing off-target effects. To achieve this, IONPs are often functionalized with various ligands, including antibodies, peptides, and small molecules, that can recognize and bind to tumor-specific antigens or receptors [32].

Core-shell and hybrid magnetic nanoparticles

To improve the functionality of MNPs, core-shell structures have been developed, where a magnetic core is surrounded by a shell.

This design improves biocompatibility, dispersibility, and chemical stability while allowing for functionalization with targeted molecules (Figure 1, Table 7).

Figure 1

Core-shell and hybrid magnetic nanoparticles [14, 73]

/f/fulltexts/WO/57052/WO-29-57052-g001_min.jpg
Table 7

Investigational biomedical applications of various types of magnetic nanoparticles [14]

Magnetic nanoparticlesMethods
of synthesis
Specific diseases targetedMechanisms of actionClinical applicationsRef.
Gold-coated
MNPs (Fe3O4@Au)
Thermal decomposition,
co-precipitation
Glioblastoma,
breast cancer
Photothermal ablation via plasmonic heating, targeted drug releaseTheranostics targeted drug delivery, and photothermal therapy[74]
Silica-coated
MNPs (Fe3O4@SiO2)
Sol-gel methodLiver cancer, brain tumorsEnhanced MRI contrast, controlled drug release via porous structureBiosensors, drug delivery systems, and enhanced MRI imaging[75]
Polymer-coated MNPsCo-precipitation, emulsion polymerizationBreast cancer, pancreatic cancerMagnetic hyperthermia, biocompatible drug encapsulation and releaseControlled drug delivery, biocompatible imaging agents, and hyperthermia[76]
Magnetic nanoclusters
(e.g., Fe3O4 clusters)
Thermal
decomposition
Solid tumors
(e.g., melanoma)
Collective magnetic heating, enhanced MRI sensitivityEnhanced imaging and hyperthermia due to collective magnetic properties[77]
Cerium-doped iron oxide nanoparticles
(Ce-doped Fe3O4)
Hydrothermal
synthesis
Osteosarcoma,
lung cancer
ROS modulation, radiosensitization via cerium dopingRadiotherapy enhancement and reactive oxygen species (modulation for cancer therapy[78]
Magnetite-gold hybrid
nanoparticles (Fe3O4-Au)
Thermal
decomposition
Glioblastoma, colorectal cancerDual imaging (magnetic and X-ray absorption), photothermal cell destructionDual imaging (MRI and X-ray CT), photothermal therapy, and drug delivery[79]
Lanthanide-doped MNPs (Ln-doped Fe3O4)Solvothermal
synthesis
Brain tumors, lymphomasLuminescent signaling for imaging, magnetic targetingLuminescence-based bioimaging combined with MRI[80]
Mn-Zn ferrite nanocrystalsMicroemulsionHepatocellular carcinomaMagnetically induced hyperthermia targeting cancer cellsMagnetically induced cancer-targeted hyperthermia[81]
Mn-Zn ferrite
MNPs
Co-precipitationLung cancer, pancreatic
cancer
Synergistic hyperthermia and radiosensitizationEnhancing targeted cancer treatment by combining hyperthermia and radiotherapy[82]
Cobalt ferrite nanoparticles (CoFe2O4)Hydrothermal
synthesis
Diabetes, cardiovascular diseasesMagnetic signal amplification for biosensingMagnetic biosensors for detecting diseases such as diabetes or cardiovascular disorders[83]
Cubic-shaped
cobalt ferrite nanoparticles
(Co-Fe NCs)
Solvothermal
synthesis
Melanoma,
breast cancer
High-anisotropy magnetic heating for hyperthermiaServe as magnetic hyperthermia agents[84]

[i] MNPs – magnetic nanoparticles, MRI – magnetic resonance imaging

Recent scientific studies have shown promising results regarding the anti-cancer effects of various iron oxide-based nanomaterials

A nanocomposite was effective in targeting and suppressing cancer cell growth using Fe3O4 nanoparticles functionalized with 3-chloropropyl trimethoxy silane and conjugated with 1-((3-(4-chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)methylene)-2-(4-phenylthiazol-2-yl) hydrazine [85].

Iron oxide nanoparticles exhibited a strong potential for inducing apoptosis in HepG2 liver cancer cells, suggesting a promising therapeutic pathway using investigated IONPs coated with glucose and conjugated with Safranal (Fe3O4@Glu-Safranal NPs) [86].

Active targeting

In addition to passive targeting through the EPR effect, active targeting strategies involve conjugating IONPs with ligands that specifically bind to receptors overexpressed on cancer cells, such as folate receptors or HER2 receptors [37]. This approach can significantly improve the specificity of IONPs, leading to higher tumor accumulation and reduced systemic toxicity. Recent studies have demonstrated the potential of ligand-functionalized IONPs in enhancing the therapeutic efficacy of chemotherapeutic drugs, with promising results in preclinical models [38, 39].

Applications in cancer therapy

Iron oxide nanoparticles have been extensively studied for their potential applications in cancer therapy, particularly in the areas of magnetic hyperthermia, drug delivery, and imaging [40, 41].

Magnetic hyperthermia is a therapeutic approach that involves the generation of localized heat by IONPs when exposed to an alternating magnetic field [42]. The heat generated can induce cancer cell death through apoptosis or necrosis, while sparing surrounding healthy tissues [43]. Iron oxide nanoparticles are particularly well suited for this application due to their superparamagnetic properties, which enable them to generate heat efficiently [44, 45]. Several preclinical studies have shown the potential of magnetic hyperthermia to enhance the efficacy of conventional therapies, such as radiotherapy and chemotherapy [46, 47].

Iron oxide nanoparticles can also be used as carriers for chemotherapeutic drugs, enabling targeted delivery and controlled release at the tumor site [48, 49]. The magnetic properties of IONPs allow for their accumulation at specific sites using an external magnetic field, further enhancing their targeting capabilities [50]. This approach has been shown to reduce the systemic toxicity of chemotherapeutic agents and improve their therapeutic index [51]. In addition, the surface of IONPs can be functionalized with stimuli-responsive polymers that release the drug in response to specific triggers, such as pH or temperature changes in the TME [51]. This controlled release mechanism enhances drug efficacy and minimizes off-target effects, which is crucial for reducing the adverse side effects commonly associated with chemotherapy [52, 87].

Iron oxide nanoparticles have also been explored as contrast agents for magnetic resonance imaging (MRI), given their strong magnetic properties and ability to enhance image contrast [88]. The superparamagnetic nature of IONPs causes a reduction in the relaxation time of surrounding water protons, leading to enhanced contrast in T2-weighted MRI images [89]. This makes them highly effective in detecting tumors at an early stage, improving the accuracy of cancer diagnosis [90]. In addition to MRI, IONPs have been studied for their potential in other imaging modalities, such as magnetic particle imaging and photoacoustic imaging, which can provide complementary information about tumor location, size, and vascularization [91, 92].

The multifunctional nature of IONPs allows them to be used in combination therapies, where they can simultaneously serve as drug carriers, imaging agents, and hyperthermia inducers [53, 93]. This approach has the potential to improve the therapeutic outcome by targeting multiple pathways involved in tumor growth and resistance, thereby reducing the likelihood of recurrence [54]. For example, recent studies have demonstrated the use of IONPs in combined chemotherapy and hyperthermia, where the heat generated by the nanoparticles enhances the uptake of chemotherapeutic drugs by tumor cells, leading to synergistic effects [55, 56] (Table 8).

Table 8

Applications of magnetic nanoparticles in drug delivery and cancer therapy [14]

Drug nameNanoparticleTargeted cancerBiological pathwayRef.
AcyclovirFe3O4 MNPsBrain cancerInhibition of viral replication in tumor cells[94]
DoxorubicinGelatin/Fe3O4-alginateBreast cancerInduction of apoptosis via DNA intercalation[95]
DoxorubicinMagnetic iron oxide NPsLiver cancerROS-mediated oxidative stress and apoptosis[96]
DoxorubicinIron oxide nanoparticlesBreast cancerTumor cell killing via (magnetic hyperthermia and chemotherapy)[97,98]
ErlotinibMesoporous MNPs/folic acidLung cancerInhibition of EGFR signaling pathway[99]
MethotrexateChitosan-coated Fe3O4Ovarian cancerFolic acid receptor-mediated endocytosis[100]
GemcitabineFe3O4, metformin, and peptide pHLIPPancreatic cancerDisruption of tumor metabolism and apoptosis[101]
TelmisartanFe3O4/chitosanProstate cancerAngiotensin receptor blockade and cell cycle arrest[102]
Chemo/hyperthermia therapyTragacanth gum/polycrylic acid/Fe3O4 nanoparticlesColorectal cancerSynergistic effect of hyperthermia and chemotherapy[103]
Gene therapyFe3O4/polyethyleneimineLeukemiaEnhanced gene transfection and targeted therapy[104]
Radiation therapyAu/iron oxideVarious solid tumorsEnhancement of radiation sensitivity through localized hyperthermia[105]
ZidovudineNiFe2O4/poly (ethylene glycol)/lipid NPsLymphomaInhibition of viral replication and tumor progression[106]
DoxorubicinPorous carbon-coated Fe3O4 nanoparticlesMelanomaHeat-induced apoptosis and enhanced drug delivery[107]

[i] EGFR – epidermal growth factor receptor, MNPs – magnetic nanoparticles

Challenges and future directions

Despite the promising potential of IONPs in cancer therapy, several challenges remain that need to be addressed to facilitate their clinical translation.

Toxicity and biocompatibility

One of the primary concerns with the use of IONPs is their potential toxicity and long-term biocompatibility [57, 108]. While iron is an essential element in the body, excessive accumulation of iron oxide can lead to oxidative stress and inflammation, particularly in organs such as the liver and spleen [58]. Therefore, careful consideration must be given to the dosage, surface modification, and clearance mechanisms of IONPs to ensure their safety in clinical applications [59].

Targeting efficiency

Achieving efficient and selective targeting of tumor cells remains a significant challenge. Although various targeting ligands have been developed, the heterogeneity of tumor cells and the dynamic nature of the tumor microenvironment can limit the effectiveness of these strategies [60, 109]. Moreover, the formation of a protein corona around the nanoparticles in the bloodstream can hinder their ability to bind to target cells [110]. Ongoing research is focused on developing more sophisticated targeting strategies that can overcome these barriers and enhance the specificity and efficacy of IONPs [111, 112].

Regulatory and manufacturing challenges

The clinical translation of IONPs is also hindered by regulatory and manufacturing challenges [62, 63]. The complex synthesis and functionalization processes involved in the production of IONPs must be standardized and scaled up to meet the stringent requirements of regulatory agencies [64]. Additionally, the lack of established guidelines for the characterization and quality control of IONPs presents a significant hurdle for their approval as therapeutic agents [65].

Future directions

To overcome these challenges, future research should focus on the development of next-generation IONPs with enhanced targeting capabilities, reduced toxicity, and improved stability in biological environments [66, 67]. Advances in nanotechnology, such as the use of biodegradable and stimuli-responsive materials, could pave the way for safer and more effective IONP-based therapies [68, 113]. Furthermore, the integration of IONPs with emerging technologies, such as gene editing and immunotherapy, holds great promise for the development of personalized cancer treatments [114, 115].

Conclusions

Iron oxide nanoparticles represent a powerful tool in the fight against cancer, offering unique opportunities for targeted drug delivery, magnetic hyperthermia, and advanced imaging techniques. While significant progress has been made in the synthesis, functionalization, and application of IONPs, several challenges remain that must be addressed to fully realize their potential in clinical settings [71, 72]. Continued research and collaboration between scientists, clinicians, and regulatory bodies will be essential to overcome these obstacles and develop safe, effective, and widely accessible nanomedicine-based cancer therapies.

Disclosures

  1. Institutional review board statement: Not applicable.

  2. Assistance with the article: None.

  3. Financial support and sponsorship: None.

  4. Conflicts of interest: None.

References

1 

Nagai H, Kim YH. Cancer prevention from the perspective of global cancer burden patterns. J Thorac Dis 2017; 9: 448-451.

2 

Hassan D, Sani A, Medina DI. Limitations of nanocarriers such as cell and tissue toxicity, genotoxicity, scale-up of nanomaterials. In: Khan FA (ed.). Nano drug delivery for cancer therapy: principles and practices. Springer Nature Singapore, Singapore 2023, 149-171.

3 

Pourmadadi M, Ostovar S, Ruiz-Pulido G, Hassan D, Souri M, Manicum ALE, et al. Novel epirubicin-loaded nanoformulations: advancements in polymeric nanocarriers for efficient drug delivery. Front Pharmacol 2023; 14: 1132465.

4 

Ferrara F, Caputo D, Canale C, Xu Y, Chen Q, Wang Y, et al. Substantial impacts of engineered nano-delivery systems in cancer treatment: current status and future trends. Biomed Pharmacother 2023; 158: 114154.

5 

Fahim YA, El-Khawaga AM, Sallam RM, Elsayed MA, Assar MFA. Immobilized lipase enzyme on green synthesized magnetic nanoparticles using Psidium guava leaves for dye degradation and antimicrobial activities. Sci Rep. 2024; 14: 8820.

6 

Reddy LH, Arias JL, Nicolas J, Couvreur P. Magnetic nanoparticles: design and characterization, toxicity and biocompatibility, pharmaceutical and biomedical applications. Chem Rev 2012; 112: 5818-5878.

7 

Hilger I, Kaiser WA. Iron oxide-based nanostructures for MRI and magnetic hyperthermia. Nanomedicine 2012; 7: 1443-1459.

8 

Zanganeh S, Spitler R, Erfanzadeh M, Alkilany AM, Mahmoudi M. Protein corona: opportunities and challenges. Int J Biochem Cell Biol 2016; 75: 143-147.

9 

Zhang P, Sun F, Tsao C, Liu S, Jain P, Sinclair A, et al. Zwitterionic gel encapsulation promotes protein stability, enhances pharmacokinetics, and reduces immunogenicity. Proc Natl Acad Sci U S A 2015; 112: 12046-12051.

10 

Miller MR, Raftis JB, Langrish JP, McLean SG, Samutrtai P, Connell SP, et al. Inhaled nanoparticles accumulate at sites of vascular disease. ACS Nano 2017; 11: 4542-4552.

11 

Singh N, Jenkins GJS, Asadi R, Doak SH. Potential toxicity of superparamagnetic iron oxide nanoparticles (SPION). Nano Rev 2010; 1.

12 

Chrishtop VV, Mironov VA, Prilepskii AY, Nikonorova VG, Vinogradov VV. Organ-specific toxicity of magnetic iron oxide-based nanoparticles. Nanotoxicology 2020; 15: 167-204.

13 

OECD. Test Guidelines for Nanomaterial Safety. OECD Publishing, Paris 2023.

14 

Fahim YA, Hasani IW, Mahmoud Ragab W. Promising biomedical applications using superparamagnetic nanoparticles. Eur J Med Res 2025; 30: 441.

15 

Reddy LH, Arias JL, Nicolas J, Couvreur P. Magnetic nanoparticles: design and characterization, toxicity and biocompatibility, pharmaceutical and biomedical applications. Chem Rev 2012; 112: 5818-5878.

16 

Colombo M, Carregal-Romero S, Casula MF, Gutiérrez L, Mora-les MP, Böhm IB, et al. Biological applications of magnetic nanoparticles. Chem Soc Rev 2012; 41: 4306-4334.

17 

Kim JE, Shin JY, Cho MH. Magnetic nanoparticles: an update of application for drug delivery and possible toxic effects. Arch Toxicol 2012; 86: 685-700.

18 

Radu M, Dinu D, Sima C, Burlacu R, Hermenean A, Ardelean A, et al. Magnetite nanoparticles induced adaptive mechanisms counteract cell death in human pulmonary fibroblasts. Toxicol In Vitro 2015; 29: 1492-1502.

19 

Zhang J, Saltzman M. Engineering biodegradable nanoparticles for drug and gene delivery. Chem Eng Prog 2013; 109: 25-30.

20 

Tewari S, Sah JF, Sahoo A. Regulatory perspectives and challenges of nanoparticle research: advances in nanomedicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2023; e2079.

21 

Doane TL, Burda C. The unique role of nanoparticles in nanomedicine: imaging, drug delivery and therapy. Chem Soc Rev 2012; 41: 2885-2911.

22 

Ferrari M. Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer 2005; 5: 161-171.

23 

Singh N, Jenkins GJS, Asadi R, Doak SH. Potential toxicity of superparamagnetic iron oxide nanoparticles (SPION). Nano Rev 2010; 1: 5358.

24 

Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. Biomaterials 2005; 26: 3995-4021.

25 

Laurent S, Forge D, Port M, Roch A, Robic C, Elst EV, et al. Magnetic iron oxide nanoparticles: synthesis, stabilization, vectorization, physicochemical characterizations, and biological applications. Chem Rev 2008; 108: 2064-2110.

26 

Sun C, Lee JSH, Zhang M. Magnetic nanoparticles in MR imaging and drug delivery. Adv Drug Deliv Rev 2008; 60: 1252-1265.

27 

Veiseh O, Gunn JW, Zhang M. Design and fabrication of magnetic nanoparticles for targeted drug delivery and imaging. Adv Drug Deliv Rev 2010; 62: 284-304.

28 

Lu AH, Salabas EL, Schüth F. Magnetic nanoparticles: synthesis, protection, functionalization, and application. Angew Chem Int Ed 2007; 46: 1222-1244.

29 

Berry CC, Curtis ASG. Functionalisation of magnetic nanoparticles for applications in biomedicine. J Phys D Appl Phys 2003; 36: R198-R206.

30 

Mah C, Zolotukhin I, Fraites TJ, Song S, Flotte TR, Dobson J, et al. Improved method of recombinant AAV2 delivery for systemic targeted gene therapy. Mol Ther 2000; 1: S239.

31 

Levy M, Wilhelm C, Siaugue JM, Horner O, Bacri1 JC, Gazeau F. Magnetically induced hyperthermia: size-dependent heating power of γ-Fe2O3 nanoparticles. J Phys Condens Matter 2008; 20: 204133.

32 

Ito A, Shinkai M, Honda H, Kobayashi T. Medical application of functionalized magnetic nanoparticles. J Biosci Bioeng 2005; 100: 1-11.

33 

Zhang Z, Liu Y, Xie P, Li W, Dou S, Wang P. Studying the interaction between gyrase and DNA using magnetic tweezers. Chin Sci Bull 2012; 57: 3560-3566.

34 

Kohler N, Sun C, Wang J, Zhang M. Methotrexate-modified superparamagnetic nanoparticles and their intracellular uptake into human cancer cells. Langmuir 2005; 21: 8858-8864.

35 

Levy M, Gazeau F, Wilhelm C. Focused ultrasound: a promising experimental modality for magnetically induced hyperthermia. Eur Phys J B 2008; 65: 49-56.

36 

Fortin JP, Gazeau F, Wilhelm C. Intracellular heating of living cells through Néel relaxation of magnetic nanoparticles. Eur Biophys J 2008; 37: 223-228.

37 

Ho D, Wang CH, Chow EKH. Nanodiamonds: the intersection of nanotechnology, drug development, and personalized medicine. Sci Adv 2015; 1: e1500439.

38 

Torchilin VP. Multifunctional and stimuli-sensitive pharmaceutical nanocarriers. Eur J Pharm Biopharm 2009; 71: 431-444.

39 

Hansol K, Seokha J, Hyukjun C, MungSoo K, Seong Guk P, Heejin J, et al. Target-switchable Gd(III)-DOTA/protein cage nanoparticle conjugates with multiple targeting affibody molecules as target selective T1 contrast agents for high-field MRI. J Con Rel 2012; 335: 269-280.

40 

Mahmoudi M, Sant S, Wang B, Laurent S, Sen T. Superparamagnetic iron oxide nanoparticles (SPIONs): development, surface modification and applications in chemotherapy. Adv Drug Deliv Rev 2011; 63: 24-46.

41 

Jain TK, Reddy MK, Morales MA, Leslie-Pelecky DL, Labhasetwar V. Biodistribution, clearance, and biocompatibility of iron oxide magnetic nanoparticles in rats. Mol Pharm 2008; 5: 316-327.

42 

Zhao F, Zhao Y, Liu Y, Chang X, Chen C, Zhao Y. Cellular uptake, intracellular trafficking, and cytotoxicity of nanomaterials. Small 2011; 7: 1322-1337.

43 

Di Corato R, Bigall NC, Ragusa A, Dorfs D, Genovese A, Maro-tta R, et al. Multifunctional nanobeads based on quantum dots and magnetic nanoparticles: synthesis and cancer cell targeting and imaging. Nano Lett 2011; 11: 2632-2639.

44 

Ai H, Jones SA, de Villiers MM. Nano-encapsulation as a novel approach to improve solubility and stability of poorly soluble anticancer drugs. Int J Pharm 2011; 419: 194-204.

45 

Mornet S, Vasseur S, Grasset F, Veverka P, Goglio G, Demourgues A, et al. Magnetic nanoparticle design for medical applications. Prog Solid State Chem 2006; 34: 237-247.

46 

Pankhurst QA, Connolly J, Jones SK, Dobson J. Applications of magnetic nanoparticles in biomedicine. J Phys D Appl Phys 2003; 36: R167-R181.

47 

Chomoucka J, Drbohlavova J, Huska D, Adam V, Kizek R, Hubalek J. Magnetic nanoparticles and targeted drug delivering. Pharmacol Res 2010; 62: 144-149.

48 

Häfeli UO, Riffle JS, Harris-Shekhawat L, Carmichael-Baranauskas A, Mark F, Dailey JP, et al. Cell uptake and in vitro toxicity of magnetic nanoparticles suitable for drug delivery. Mol Pharm 2009; 6: 1417-1428.

49 

Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv Drug Deliv Rev 2012; 64: 206-212.

50 

Kim JS, Yoon TJ, Yu KN, Noh MS, Woo M, Kim BG, et al. Cellular uptake of magnetic nanoparticle is mediated through energy-dependent endocytosis in A549 cells. J Vet Sci 2006; 7: 321-326.

51 

Ma X, Huilin S, Tang Y, Qu F, Priestley RD. Core-shell structured Fe3O4 polydopamine nanoparticle for combined chemo-photothermal therapy. Colloids Surf B Biointerfaces 2013; 106: 205-212.

52 

Shevtsov M, Multhoff G. Recent developments of magnetic nanoparticles for theranostic applications. J Nanomater 2011; 2011: 165942.

53 

Djebbi K, Shi B, Weng T, Bahri M, Elaguech MA, Liu J, et al. Highly sensitive fluorescence assay for miRNA detection: investigation of the DNA spacer effect on the dsn enzyme activity toward magnetic-bead-tethered probes. ACS Omega 2022; 7: 2224-2233.

54 

Li J, He Y, Sun W, Luo Y, Cai H, Pan Y, et al. Hyaluronic acid-modified hydrothermally synthesized iron oxide nanoparticles for targeted tumor MR imaging. Biomaterials 2014; 35: 3666-3677.

55 

Choi MR, Stanton-Maxey KJ, Stanley JK, Levin CS, Bardhan R, Akin D, et al. A cellular Trojan horse for delivery of therapeutic nanoparticles into tumors. Nano Lett 2007; 7: 3759-3765.

56 

Dreaden EC, Alkilany AM, Huang X, Murphy CJ, El-Sayed MA. The golden age: gold nanoparticles for biomedicine. Chem Soc Rev 2012; 41: 2740-2779.

57 

Hergt R, Dutz S, Röder M. Effects of size distribution on hystere-sis losses of magnetic nanoparticles for hyperthermia. J Phys Condens Matter 2008; 20: 385214.

58 

Wilhelm C, Gazeau F. Universal cell labelling with anionic magnetic nanoparticles. Biomaterials 2008; 29: 3161-3174.

59 

Schenck JF. Safety of strong, static magnetic fields. J Magn Reson Imaging 2000; 12: 2-19.

60 

Kubo T, Sugita T, Shimose S, Ikuta Y, Murakami T. Targeted systemic chemotherapy using magnetic liposomes with incorporated adriamycin for osteosarcoma in hamsters. Int J Oncol 2000; 17: 309-315.

61 

McCarthy JR, Weissleder R. Multifunctional magnetic nanoparticles for targeted imaging and therapy. Adv Drug Deliv Rev 2008; 60: 1241-1251.

62 

Rapoport N. Physical stimuli-responsive polymeric micelles for anti-cancer drug delivery. Prog Polym Sci 2007; 32: 962-990.

63 

Kim J, Kim J, Bae JS. ROS-responsive nanoplatform for combined delivery of NO and H2S: potential applications in treating ischemic diseases. J Control Release 2020; 317: 26-34.

64 

Gallo J, Long NJ, Aboagye EO. Magnetic nanoparticles as contrast agents in the diagnosis and treatment of cancer. Chem Soc Rev 2013; 42: 7816-7833.

65 

Kievit FM, Zhang M. Surface engineering of iron oxide nanoparticles for targeted cancer therapy. Acc Chem Res 2011; 44: 853-862.

66 

Yildirim A, Bayindir M. Gas phase functionalization of mesoporous silica nanoparticles. ACS Appl Mater Interfaces 2010; 2: 3333-3341.

67 

Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent SMANCS. Cancer Res 1986; 46: 6387-6392.

68 

Sadhukha T, Wiedmann TS, Panyam J. Inhalable magnetic nanoparticles for targeted hyperthermia in lung cancer therapy. Biomaterials 2013; 34: 5163-5171.

69 

Hassan D, Bakhsh H, Khurram AM, Bhutto SA, Jalbani NS, Ghumro T, et al. Fluorescent nanotechnology: an evolution in optical sensors. Curr Anal Chem 2022; 18: 176-185.

70 

Valencia-Lazcano AA, Hassan D, Pourmadadi M, Shamsabadipour A, Behzadmehr R, Rahdar A, et al. 5-Fluorouracil nano-delivery systems as a cutting-edge for cancer therapy. Eur J Med Chem 2023; 246: 14995.

71 

De Jong WH, Borm PJ. Drug delivery and nanoparticles: applications and hazards. Int J Nanomedicine 2008; 3: 133-149.

72 

Berry CC. Intracellular delivery of nanoparticles via the HIV-1 tat peptide. Nanomedicine 2008; 3: 357-365.

73 

Das S, Pérez-Ramirez J, Gong J, Dewangan N, Hidajat K, Gates BC, et al. Core-shell structured catalysts for thermocatalytic, photocatalytic, and electrocatalytic conversion of CO2. Chem Soc Rev 2020; 49: 2937-3004.

74 

Jain S, Hirst DG, O’Sullivan JM. Gold nanoparticles as novel agents for cancer therapy. Br J Radiol 2012; 85: 101-113.

75 

Lu AH, Salabas EL, Schüth F. Magnetic nanoparticles: synthesis, protection, functionalization, and application. Angewandte Chem Int Ed 2007; 46: 1222-1244.

76 

Zhao X, Bai J, Yang W. Stimuli-responsive nanocarriers for therapeutic applications in cancer. Cancer Biol Med 2021; 18: 319.

77 

Park J, An K, Hwang Y, Park JG, Noh HJ, Kim JY, et al. Ultra-large-scale syntheses of monodisperse nanocrystals. Nat Mater 2004; 3: 891-895.

78 

Wei F, Neal CJ, Sakthivel TS, Seal S, Kean T, Razavi M, et al. Ce-rium oxide nanoparticles protect against irradiation-induced cellular damage while augmenting osteogenesis. Mater Sci Eng C 2021; 126: 112145.

79 

Van de Looij SM, Hebels ER, Viola M, Hembury M, Oliveira S, Vermonden T. Gold nanoclusters: imaging, therapy, and theranostic roles in biomedical applications. Bioconjug Chem 2021; 33: 4-23.

80 

Comby S, Surender EM, Kotova O, Truman LK, Molloy JK, Gunnlaugsson T. Lanthanide-functionalized nanoparticles as MRI and luminescent probes for sensing and/or imaging applications. Inorg Chem 2014; 53: 1867-1879.

81 

Xie J, Yan C, Yan Y, Chen L, Song L, Zang F, et al. Multi-modal Mn-Zn ferrite nanocrystals for magnetically-induced cancer targeted hyperthermia: a comparison of passive and active targeting effects. Nanoscale 2016; 8: 16902-16915.

82 

Wang Y, Zou L, Qiang Z, Jiang J, Zhu Z, Ren J. Enhancing targeted cancer treatment by combining hyperthermia and radiotherapy using Mn-Zn ferrite magnetic nanoparticles. ACS Biomater Sci Eng 2020; 6: 3550-3562.

83 

Cardoso VF, Francesko A, Ribeiro C, Bañobre-López M, Martins P, Lanceros-Mendez S. Advances in magnetic nanoparticles for biomedical applications. Adv Healthc Mater 2018; 7: 1700845.

84 

Balakrishnan PB, Silvestri N, Fernandez-Cabada T. Exploiting unique alignment of cobalt ferrite nanoparticles, mild hyperthermia, and controlled intrinsic cobalt toxicity for cancer therapy. Adv Mater 2020; 32: 2003712.

85 

Habibzadeh SZ, Salehzadeh A, Moradi-Shoeili Z, Shandiz SAS. A novel bioactive nanoparticle synthesized by conjugation of 3-chloropropyl trimethoxy silane functionalized Fe3O4 and 1-((3-(4-chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)methylene)-2-(4-phenylthiazol-2-yl) hydrazine: assessment on anti-cancer against gastric AGS cancer cells. Mol Biol Rep 2020; 47: 1637-1647.

86 

Mikaeili Ghezeljeh S, Salehzadeh A, Ataei-E Jaliseh S. Iron oxide nanoparticles coated with glucose and conjugated with safranal (Fe3O4 glu-safranal NPs) inducing apoptosis in liver cancer cell line (HepG2). BMC Chem 2024; 18: 33.

87 

Sosnovik DE, Nahrendorf M, Weissleder R. Magnetic nanoparticles for MR imaging: agents, techniques and cardiovascular applications. Basic Res Cardiol 2008; 103: 122-130.

88 

Arruebo M, Fernández-Pacheco R, Ibarra MR, Santamaría J. Magnetic nanoparticles for drug delivery. Nano Today 2007; 2: 22-32.

89 

Martins C, Rolo C, Cacho VRG, Pereira LCJ, Borges JP, Silva JC, et al. Enhancing the magnetic properties of superparamagnetic iron oxide nanoparticles using hydrothermal treatment for magnetic hyperthermia application. Mater Adv 2025; 6: 1726-1743.

90 

Bulte JWM, Kraitchman DL. Iron oxide MR contrast agents for molecular and cellular imaging. NMR Biomed 2004; 17: 484-499.

91 

Srinivasan B, Kolluru C, Mitragotri S. Nanoparticles for topical drug delivery: potential for skin cancer treatment. Adv Drug Deliv Rev 2011; 63: 478-489.

92 

Yu MK, Jeong YY, Park J, Park S, Kim JW, Min JJ, et al. Drug-loaded superparamagnetic iron oxide nanoparticles for combined cancer imaging and therapy in vivo. Angew Chem Int Ed 2008; 47: 5362-5365.

93 

Weissleder R, Nahrendorf M, Pittet MJ. Imaging macrophages with nanoparticles. Nat Mater 2014; 13: 125-138.

94 

Xie X, Zhang L, Zhang W, Tayebee R, Hoseininasr A, Vatan-pour HH, et al. Fabrication of temperature and pH sensitive decorated magnetic nanoparticles as effective biosensors for targeted delivery of acyclovir anti-cancer drug. J Mol Liq 2020; 309: 113024.

95 

Huang CH, Huang TJ, Ke CJ, Yao CH. Doxorubicin-gelatin/Fe3O4-alginate dual-layer magnetic nanoparticles as targeted anticancer drug delivery vehicles. Polymers 2020; 12: 1747.

96 

Augustin E, Czubek B, Nowicka AM, Kowalczyk A, Stojek Z, Mazerska Z. Improved cytotoxicity and preserved level of cell death induced in colon cancer cells by doxorubicin after its conjugation with iron-oxide magnetic nanoparticles. Toxicol In Vitro 2016; 33: 45-53.

97 

Kossatz S, Grandke J, Couleaud P, Latorre A, Aires A, Crosbie-Staunton K, et al. Efficient treatment of breast cancer xenografts with multifunctionalized iron oxide nanoparticles combining magnetic hyperthermia and anti-cancer drug delivery. Breast Cancer Res 2015; 17: 1-17.

98 

Piehler S, Dähring H, Grandke J, Göring J, Couleaud P, Aires A, et al. Iron oxide nanoparticles as carriers for DOX and magnetic hyperthermia after intratumoral application into breast cancer in mice: impact and future perspectives. Nanomaterials 2020; 10: 1016.

99 

Avedian N, Zaaeri F, Daryasari MP, Javar HA, Khoobi M. pH-sensitive biocompatible mesoporous magnetic nanoparticles labeled with folic acid as an efficient carrier for controlled anticancer drug delivery. J Drug Deliv Sci Technol 2018; 44: 323-332.

100 

Fathi M, Barar J, Erfan-Niya H, Omidi Y. Methotrexate-conjugated chitosan-grafted pH-and thermo-responsive magnetic nanoparticles for targeted therapy of ovarian cancer. Int J Biol Macromol 2020; 154: 1175-1184.

101 

Han H, Hou Y, Chen X, Zhang P, Kang M, Jin Q, et al. Metformin-induced stromal depletion to enhance the penetration of gemcitabine-loaded magnetic nanoparticles for pancreatic cancer targeted therapy. J Am Chem Soc 2020; 142: 4944-4954.

102 

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; 148: 109749.

103 

Sayadnia S, Arkan E, Jahanban-Esfahlan R, Sayadnia S, Jay-mand M. Tragacanth gum-based pH-responsive magnetic hydrogels for “smart” chemo/hyperthermia therapy of solid tumors. Polym Adv Technol 2021; 32: 262-271.

104 

Grabowska M, Grześkowiak BF, Szutkowski K, Wawrzyniak D, Głodowicz P, Barciszewski J, et al. Nano-mediated delivery of double-stranded RNA for gene therapy of glioblastoma multiforme. PLoS One 2019; 14: e0213852.

105 

Sun L, Joh DY, Al-Zaki A, Stangl M, Murty S, Davis JJ, et al. Theranostic application of mixed gold and superparamagnetic iron oxide nanoparticle micelles in glioblastoma multiforme. J Biomed Nanotechnol 2016; 12: 347-356.

106 

Joshy KS, Augustine R, Mayeen A, Alex SM, Hasan A, Thomas S, et al. NiFe2O4/poly (ethylene glycol)/lipid-polymer hybrid nano-particles for anti-cancer drug delivery. New J Chem 2020; 44: 18162-18172.

107 

Wu F, Sun B, Chu X, Zhang Q, She Z, Song S, et al. Hyaluronic acid-modified porous carbon-coated Fe3O4 nanoparticles for magnetic resonance imaging-guided photothermal/chemotherapy of tumors. Langmuir 2019; 35: 13135-13144.

108 

Laurent S, Dutz S, Häfeli UO, Mahmoudi M. Magnetic fluid hyperthermia: focus on superparamagnetic iron oxide nanoparticles. Adv Colloid Interface Sci 2011; 166: 8-23.

109 

Natarajan A, Gruettner C, Ivkov R, DeNardo GL, Mirick G, Yuan A, et al. NanoFerrite particle based radiofrequency thermal ablation treatment: potential for treatment of colorectal cancer. Int J Hyperthermia 2008; 24: 497-503.

110 

Jain RK. Vascular and interstitial barriers to delivery of therapeutic agents in tumors. Cancer Metastasis Rev 1990; 9: 253-266.

111 

Jordan A, Scholz R, Wust P, Fähling H, Felix R. Magnetic fluid hyperthermia (MFH): cancer treatment with AC magnetic field induced excitation of biocompatible superparamagnetic nanoparticles. J Magn Magn Mater 1999; 201: 413-419.

112 

Herd H, Daum N, Jones AT, Huwer H, Ghandehari H, Lehr CM. Nanoparticle geometry and surface orientation influence mode of cellular uptake. ACS Nano 2013; 7: 1961-1973.

113 

Davis ME, Chen ZG, Shin DM. Nanoparticle therapeutics: an emerging treatment modality for cancer. Nat Rev Drug Discov 2008; 7: 771-782.

114 

Gonzales M, Krishnan KM. Phase transfer of highly monodisperse iron oxide nanocrystals with pluronic P123 for biomedical applications. J Magn Magn Mater 2005; 293: 265-270.

115 

Hernot S, Klibanov AL. Microbubbles in ultrasound-triggered drug and gene delivery. Adv Drug Deliv Rev 2008; 60: 1153-1166.

Copyright: © 2025 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2025 Termedia Sp. z o.o.
Developed by Bentus.