Open Access Article
Subham
Preetam†
*a,
Muhammad Fazle
Rabbee†
b,
Richa
Mishra
c,
Shailendra
Thapliyal
d,
Ravi
Deshwal
e,
Sarvesh
Rustagi
f,
Archana
Dashmana
g,
Rasiravathanahalli K.
Govindarajan
hi and
Sumira
Malik
*jk
aDepartment of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Dalseong-gun, Daegu, 42988, Republic of Korea. E-mail: subhampreetam@dgist.ac.kr
bDepartment of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea. E-mail: rabbi.biotech@gmail.com
cDepartment of Computer Engineering, Parul Institute of Engineering and Technology (PIET), Parul University, Ta. Waghodia, Vadodara, Gujarat 391760, India. E-mail: richa.mishra31240@paruluniversity.ac.in
dUttaranchal Institute of Technology, Uttaranchal University, Dehradun, 248007, India
eInstitute of Bioscience and Technology, Shri Ramswaroop Memorial University, Lucknow-Deva Road, Uttar Pradesh 225 003, India
fSchool of Agriculture, Dev Bhoomi Uttarakhand University, Dehradun, 248007, Uttarakhand, India
gHimalayan School of Biosciences, Swami Rama Himalayan University, Dehradun, Uttarakhand 248016, India. E-mail: archnadhasmana@srhu.edu.in
hDepartment of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641021, India
iCentre for Natural Products and Functional Foods, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641021, India. E-mail: biogovindarajan@gmail.com
jAmity Institute of Biotechnology, Amity University Jharkhand, Ranchi, 834002, India. E-mail: smalik@rnc.amity.edu
kUniversity Center for Research & Development (UCRD) Chandigarh University, NH-05 Chandigarh-Ludhiana Highway, Mohali, Punjab, India
First published on 9th December 2025
Brain cancer remains one of the most challenging malignancies due to the blood–brain barrier (BBB), limited drug penetration, and resistance to conventional therapies. Recent advancements in magnetic nanoparticles (MNPs) have opened new avenues for targeted and efficient brain cancer treatment. MNPs offer multifunctionality, including magnetic hyperthermia therapy, targeted drug delivery, and enhanced imaging via magnetic resonance imaging (MRI). This review explores the latest progress in MNP-based theranostics, highlighting their physicochemical properties, functionalization strategies, and mechanisms of action in brain cancer therapy. Additionally, we discuss novel approaches such as stimuli-responsive nanocarriers, BBB penetration techniques, and multifunctional hybrid nanoparticles. Furthermore, preclinical and clinical studies are reviewed to assess the current status and translational challenges. Despite promising outcomes, toxicity, biodistribution, and long-term biocompatibility remain key hurdles in clinical applications. Addressing these limitations will pave the way for personalized nanomedicine-based brain cancer treatment, optimizing therapeutic efficacy and patient outcomes.
The treatment of brain cancer is fraught with unique challenges. One of the most significant obstacles is the BBB, a protective shield that restricts the passage of many therapeutic agents into the brain.6 This barrier complicates the efficacy of conventional chemotherapy and immunotherapy, often leading to suboptimal treatment outcomes.7,8 Additionally, the infiltrative nature of many brain tumors makes complete surgical removal difficult without damaging surrounding healthy tissue. Current standard treatments, including surgery, radiation, and chemotherapy, often provide limited effectiveness due to tumor heterogeneity and resistance mechanisms.9,10 As a result, there is an urgent need for innovative therapeutic strategies that can effectively target these malignancies.
Nanotechnology has emerged as a promising avenue in cancer therapy, particularly for overcoming some of the limitations associated with traditional treatment methods.11,12 By manipulating materials at the nanoscale (typically 1 to 100 nanometers), researchers can develop nanoparticles (NPs) that enhance drug delivery systems. These NPs can be engineered to improve bioavailability and selectively target tumor cells while minimizing systemic toxicity. Their small size enables them to traverse biological barriers such as the BBB, making them valuable tools in delivering therapeutic agents directly to brain tumors.1,13–20
Among various types of nanoparticles, magnetic nanoparticles (MNPs) hold particular promise for brain cancer treatment. MNPs can be guided to specific tumor sites using external magnetic fields, allowing for targeted therapy that enhances drug accumulation at the tumor location while reducing exposure to healthy tissues.10,21,22 Additionally, MNPs can be functionalized with drugs or imaging agents to provide theranostic capabilities simultaneously enabling treatment and monitoring therapeutic effects.23,24 The versatility and unique properties of MNPs position them as essential components in advancing brain cancer therapies and improving patient outcomes. In summary, while brain cancer presents formidable challenges due to its complex biology and treatment limitations, advancements in nanotechnology particularly through the use of MNPs offer new hope for more effective therapeutic strategies. Continued research in this field is critical for developing innovative solutions that can enhance treatment efficacy and improve quality of life for patients battling this devastating disease.
This review was prepared following a structured literature selection strategy to ensure a balanced and comprehensive coverage of MNP based strategies for brain tumour therapy. A systematic search was performed in PubMed, Scopus, and Web of Science databases for articles published between ∼2000 to 2025.20 The key terms used included combinations of25 magnetic nanoparticles, glioblastoma, brain cancer, hyperthermia, theranostics, blood–brain barrier, and nanomedicine. Inclusion criteria comprised peer-reviewed original research and review articles reporting experimental, preclinical, or clinical outcomes of MNPs in brain tumour therapy, as well as studies addressing mechanisms of BBB penetration, targeting strategies, and safety profiles. Exclusion criteria included non-English publications, conference abstracts without full data, and studies unrelated to brain cancer or magnetic nanoparticle applications. Clinical trials data regarding phase, primary endpoints, and latest updates were extracted directly from peer-reviewed publications entries.
Magnetite (Fe3O4) nanoparticles exhibit a unique ability to retain magnetization even in the absence of an external magnetic field, which allows for effective navigation and accumulation at specific anatomical locations, such as brain tumors. Their spinnable internal structure leads to enhanced relaxation times, thereby improving imaging contrast in MRI applications. Fenton-reaction-acceleratable MNPs, such as Fe3O4/Gd2O3 hybrid nanoparticles, have shown efficacy in ferroptosis therapy for orthotopic brain tumors.30,31 The high saturation magnetization of magnetite (approximately 92 emu per g) enables robust magnetic responses, making it suitable for applications in targeted drug delivery and hyperthermia treatment in cancer therapy.
In contrast, maghemite (γ-Fe2O3) nanoparticles have gained attention due to their oxidative stability and lower toxicity compared to magnetite.32 These nanoparticles can be tailored through surface modifications using polyethylene glycol (PEG), folic acid, or antibodies that target specific molecular markers present in tumor cells, maghemite is known for its slightly lower saturation magnetization (approximately 74 emu per g). This level of customization ensures that the nanoparticles can efficiently penetrate the BBB while targeting brain tumors, which is a significant challenge in treating such malignancies.33 Clinical trials have demonstrated the feasibility and safety of MNP-based thermotherapy in recurrent malignant brain tumors.34
Metal-based MNPs such as cobalt (Co), nickel (Ni), and their alloys exhibit stronger magnetic properties compared to iron oxides. However, their use in biomedicine is limited by concerns over cytotoxicity and chemical instability.35 Recent efforts have focused on encapsulating these materials within inert coatings like silica or gold to reduce oxidation and enhance biocompatibility.36,37 Likewise, spinel ferrites (MFe2O4, where M = Co, Mn, Zn, etc.) offer tunable magnetic and physicochemical properties by varying the metal ion composition. These nanoparticles have shown promise in applications ranging from magnetically triggered drug release to biosensing. Their enhanced magnetic anisotropy and thermal stability also make them ideal candidates for high-frequency magnetic hyperthermia. To overcome the limitations of single-component systems, core–shell nanoparticles combining magnetic cores with polymeric, metallic, or silica shells have been engineered. These structures offer synergistic advantages such as dual-functionality (e.g., magnetic targeting and fluorescence imaging) and reduced aggregation in physiological environments.35,36 For instance, Fe3O4@Au nanostructures can enable both magnetic guidance and photothermal therapy, while polymer-coated MNPs facilitate pH-responsive or enzyme-sensitive drug release. Additionally, the capacity of magnetite nanoparticles to be functionalized with various biotherapeutics, including chemotherapeutic agents, nucleic acids, and targeting ligands, enhances their therapeutic efficacy.
Cobalt ferrite (CoFe2O4) nanoparticles are characterized by their robust magnetic properties and high coercivity, enabling target-specific drug delivery and MRI functionalities. CoFe2O4 nanoparticles have been synthesized with various surface modifications to enhance their stability in biological fluids, allowing for prolonged circulation time and improved cellular uptake. The ability to manipulate the magnetic properties of CoFe2O4 also facilitates the application of localized magnetic field techniques, enhancing the therapeutic effect of co-delivered chemotherapeutics while simultaneously allowing for non-invasive imaging.35
Nickel ferrite (NiFe2O4) nanoparticles, exhibiting superparamagnetic behavior, are particularly advantageous for their enhanced biocompatibility and lower toxicity profiles compared to other metals. This property enables NiFe2O4 to effectively serve as carriers for antitumor agents, facilitating their accumulation at targeted sites in tumor tissues. The thermal effects generated by alternating magnetic fields, known as magnetic hyperthermia, further augment the tumor-targeting potential of these nanoparticles, creating synergistic effects with traditional therapies and potentially leading to tumor ablation.36,37
Iron–platinum alloys (FePt) represent another class of MNPs ideally suited for applications in brain tumor therapy. With a high magnetic anisotropy and the ability to achieve high magnetization, FePt nanoparticles can be tailored for both imaging modalities and targeted therapy.38 The biocompatibility of FePt allows for the conjugation with therapeutic agents, enabling site-specific drug release triggered by an external magnetic field. This targeted approach not only minimizes systemic side effects but also fosters an environment conducive to the selective killing of malignant cells.38
Rare Earth Iron Boron Compounds (REFeB) are notable for their exceptional magnetic performance, which opens avenues for advanced therapeutic interventions in neuro-oncology.8,39 The high magnetization of REFeB nanoparticles enables the efficient attraction of these carriers to tumor sites under the influence of externally applied magnetic fields. When utilized in combination with advanced imaging techniques, REFeB nanoparticles enhance the accuracy of tumor localization while delivering chemotherapeutic agents in a spatially and temporally controlled manner.39 By leveraging the unique properties of CoFe2O4, NiFe2O4, FePt, and REFeB nanoparticles, it is possible to achieve targeted therapeutic delivery, effective imaging, and controlled release of chemotherapeutic agents in brain cancer, as shown in Fig. 1.
![]() | ||
| Fig. 1 Various types of magnetic nanoparticles in brain cancer treatments.8,39 | ||
Another critical approach in the functionalization of MNPs involves the attachment of biomolecules such as antibodies or peptides.41 This surface modification renders MNPs capable of targeted drug delivery by enabling the preferential accumulation of therapeutic agents in specific tissues or cells. For instance, antibodies that specifically bind to tumor-associated antigens can be conjugated to the nanoparticle surface, facilitating targeted therapy while minimizing off-target effects.5,41 The ability to direct drug delivery systems to diseased cells significantly enhances therapeutic efficacy and reduces systemic toxicity, thereby improving patient outcomes.
Incorporating fluorescent dyes into the MNPs represents an innovative strategy for multifunctional applications, allowing for simultaneous imaging and drug delivery.44 Fluorescently labeled MNPs can be utilized in techniques such as MRI or fluorescence imaging, thereby providing real-time visualization of their distribution and therapeutic effect within the body.5,45,46 This dual functionality not only enhances diagnostic capabilities but also aids in monitoring the therapeutic efficacy of drug-loaded nanoparticles, ensuring streamlined clinical applications. Again, the modification of MNPs with silica or other robust materials offers enhanced stability and additional functionalities. Silica coating provides a protective layer around the magnetic core, which not only reduces the risk of oxidation but also allows for further functionalization with various drugs or targeting agents. This silica layer can also serve as a platform for the controlled release of therapeutic agents, thereby improving the overall effectiveness of treatment regimens.42 By employing strategies such as polymer coatings, biomolecule attachment, incorporation of fluorescent dyes, and silica modification, researchers can significantly improve the stability, biocompatibility, and targeting capabilities of MNPs. These advancements are paramount for the successful transition of MNPs from bench to bedside, showcasing their potential to revolutionize drug delivery, diagnostic imaging, and theranostics in modern medicine.5,45,46
Bare MNPs such as Fe3O4 are prone to aggregation, opsonization, and rapid clearance by the reticuloendothelial system (RES), which compromises their clinical potential. Surface modification using hydrophilic polymers like PEG, dextran, or chitosan improves colloidal stability and reduces protein adsorption, enhancing circulation time and immune evasion.12,29 PEGylation, in particular, has been widely used to endow nanoparticles with “stealth” properties, minimizing immunogenicity and improving biocompatibility. In addition, zwitterionic and amino acid-based coatings have shown potential in reducing nonspecific cellular uptake and complement activation, thereby enhancing biocompatibility without sacrificing magnetic responsiveness.43
To traverse the BBB and selectively target tumor cells, MNPs are often functionalized with ligands, antibodies, or peptides that bind to overexpressed receptors on brain endothelial cells or tumor tissue. i.e., target transferrin and lactoferrin receptors commonly upregulated in glioblastoma cells and endothelial cells of the BBB. Facilitate Angiopep-2 and RGD peptides receptor-mediated endocytosis and tumor penetration.48 Improve selectivity toward Aptamers and monoclonal antibodies based tumor markers like EGFRvIII or IL13Rα2 in gliomas.49,50 These targeting moieties not only improve cellular uptake but also minimize off-target effects, allowing for lower therapeutic doses with enhanced efficacy. Surface-functionalized MNPs can be engineered to carry a wide range of therapeutic payloads including chemotherapeutic agents (e.g., doxorubicin, paclitaxel), siRNA, or microRNAs. Surface functionalization of MNPs is indispensable for their safe and efficient use in brain cancer therapy. It governs their interaction with biological systems, enhances their ability to cross the BBB, improves tumor-specific accumulation, and enables controlled drug release.
![]() | ||
| Fig. 2 Schematic representation of MHT for drug delivery and causing cell death by heat generation.55,57 | ||
MNPs offering dual imaging and therapeutic capabilities. MNPs can be used as contrast agents for MRI and as therapeutic agents through hyperthermia and targeted drug delivery.34 Their unique properties allow for external control of heat generation and magnetic attractive forces, enabling controlled drug release and cell signaling manipulation. Intra-arterial magnetic targeting has shown significant improvement in tumor capture of MNPs compared to conventional methods. The conjugation of peptides or antibodies to MNPs enables specific targeting of tumor cells and potential disruption of active signaling pathways (Fig. 3).34 Clinical trials have demonstrated the feasibility, safety, and efficacy of MNPs in treating recurrent malignant brain tumors, paving the way for future translational studies. Ongoing research aims to optimize MNP properties for improved cancer diagnosis and therapy.
![]() | ||
| Fig. 3 MNPs in brain cancer theranostics enhancing diagnosis via MRI contrast for early tumor detection and enabling targeted therapies such as hyperthermia and drug delivery through the blood–brain barrier, guided by external magnetic fields for precision treatment.11,62 | ||
Redox-responsive systems also exploit the elevated intracellular glutathione (GSH) levels in glioma cells to trigger the breakdown of disulfide bonds and facilitate intracellular drug release. Similarly, pH-sensitive coatings using acid-labile linkers such as hydrazones allow drug liberation specifically within the acidic tumor or endosomal compartments. These approaches enhance the selective release of chemotherapeutics like doxorubicin or paclitaxel, thereby increasing intracellular accumulation and therapeutic potency. Externally triggered systems, particularly those responsive to AMFs, are prominent in magnetic nanomedicine. Magnetic hyperthermia where nanoparticles generate localized heat under AMFs has been shown not only to induce tumor cell death but also to enhance the permeability of tumor tissues and promote drug diffusion. Recent studies by Yalamandala et al. have shown that magnetothermal activation can synergize with immune-actuated drug delivery to improve treatment outcomes in glioblastoma models.65
Importantly, dual- or multi-stimuli responsive nanocarriers combining pH and magnetic responsiveness, or redox and thermal activation are emerging as a powerful design strategy. These systems enable sequential or synergistic release mechanisms, enhancing spatiotemporal control over therapy. However, despite promising preclinical results, major challenges remain, including immune clearance by the mononuclear phagocyte system (MPS), limited biodegradability, and translational barriers such as scalable manufacturing and regulatory approval. One example is a magnetic nanoparticle drug carrier encapsulated by a thermosensitive polymer, which exhibits controlled drug release in response to temperature changes and pH. Natural polymers have also been used to create magnetic field-responsive nanocarriers, offering advantages in biocompatibility and biodegradability.66 These smart nanocarriers can enhance drug accumulation at target sites, control drug release, and potentially overcome multidrug resistance.67 The ability to respond to specific biological signals in cancer cells or external stimuli makes these nanocarriers particularly promising for improving the efficacy and reducing side effects of cancer treatments.
For instance, Asghar et al. designed nickel-doped hydroxyapatite thin films embedded with MNPs capable of reaching therapeutic hyperthermic temperatures (up to 45 °C) within 80 seconds under AMF exposure. There in vitro assays confirmed selective tumoricidal effects while preserving healthy tissues, indicating the promise of such platforms for precise thermal ablation in brain tumors (Fig. 4.).69 Moreover, these magnetic agents can be co-loaded with chemotherapeutics for combined therapy, exploiting heat-induced increases in membrane permeability and drug uptake. However, a critical barrier to clinical application is thermal safety. Excessive exposure to AMFs can induce nonspecific heating through eddy currents in healthy tissue, which may compromise surrounding neural structures. Pilpilidis et al. used electromagnetic simulations to reveal that the traditionally accepted Atkinson-Brezovich safety limit might underestimate potential risks, suggesting that field strength during magnetic nanoparticle hyperthermia must be tailored more conservatively, especially for intracranial applications.2
![]() | ||
| Fig. 4 Mechanistic overview of magnetic hyperthermia and radiosensitization induced by SPIONs. Local heating enhances reactive oxygen species (ROS) generation and augments radiotherapy-induced DNA damage, while thermal stress triggers apoptotic signaling. Typical experimental parameters include SPION core diameters of 10–20 nm, and specific absorption rates (SAR) between 100–800 W g−1 Fe. This figure has been adapted from ref. 69 with permission from MDPI, copyright 2025. | ||
Despite these challenges, integrating magnetic hyperthermia into a multimodal treatment regimen for glioblastoma remains highly promising. The thermal effects not only induce direct cytotoxicity but also promote immunogenic cell death and enhance antigen presentation favorable in immunotherapy co-strategies. Hence, MNPs can serve dual functions in GBM therapy by facilitating hyperthermia and improving BBB permeability, positioning them as key players in future personalized oncology approaches, as shown in Table 1.
| No. | Nanoparticle system | Composition | Stimuli applied | Temp achieved | Model used | Observed effects | Reference |
|---|---|---|---|---|---|---|---|
| 1 | Ni-doped HAp thin films | Nickel-substituted hydroxyapatite | Alternating magnetic field (AMF) | 45 °C in 80 s | In vitro (thin film model) | Selective tumor cell heating; enhanced thermal stability | 69 |
| 2 | Fe3O4 nanoparticles | Superparamagnetic iron oxide | Magnetic field + blood perfusion model | Modeled to reach 42–45 °C | Mathematical blood flow model | Increased heat transfer; reduced tumor blood velocity | 68 |
| 3 | Electromagnetic model | Simulated MNP + human anatomy | Eddy current modeling under AMF | Field recalibrated to meet ICNIRP safety | Human voxel models | Lowered permissible field strength vs. Atkinson-Brezovich; reduced overheating risk | 2 |
| 4 | Fe-based nanocarrier + TMZ | Fe-based MNP + temozolomide | AMF + chemo combo | 43–45 °C (est.) | GBM-bearing mice | Synergistic tumor suppression, enhanced survival | 2 |
A recent formulation by Bhattacharya et al. encapsulated Imatinib Mesylate (IMT), a tyrosine kinase inhibitor, into hybrid nanoparticles composed of PLGA, D-α-tocopheryl polyethene glycol succinate (TPGS), and polyethene glycol (PEG). These core–shell lipid–polymer hybrid nanoparticles (CSLHNPs) demonstrated enhanced brain delivery, with a narrow size distribution (mean diameter ≈ 155 nm) and sustained IMT release over 48 hours. Notably, cytotoxicity assays on LN229 glioblastoma cells revealed significantly higher cell death with CSLHNPs compared to free IMT, attributed to improved cellular uptake and reduced oxidative stress via mitigation of reactive oxygen species.71 Such MHNPs are often engineered to serve dual roles: drug carriers and magnetic agents. Their superparamagnetic cores enable guidance and accumulation in tumor regions via external magnetic fields and facilitate imaging through MRI or magnetic particle imaging (MPI). Some hybrid designs incorporate thermosensitive or redox-sensitive layers, enabling them to function as magnetothermal agents for controlled drug release or hyperthermia applications.
Furthermore, by combining multiple layers (e.g., lipids, polymers, peptides), MHNPs can simultaneously target tumor receptors, bypass efflux pumps, and provide sustained drug release. Liu et al. emphasized that multifunctional polymeric nanoparticles show potential for delivering chemotherapeutics, immunotherapeutics, and radiosensitizers across the BBB while responding to tumor microenvironmental cues such as acidic pH or enzymatic activity.72 Despite these advances, translational challenges persist. Complex architectures can hinder large-scale manufacturing, and long-term in vivo safety must be thoroughly evaluated. Nonetheless, the ability of hybrid magnetic nanocarriers to integrate diagnosis, therapy, targeting, and controlled release mechanisms holds significant promise for advancing precision medicine in glioblastoma.
![]() | ||
| Fig. 5 Schematic of the non-invasive brain delivery and quantification platform. BBBO was induced using MBs and low-intensity FUS targeted to the right hemisphere. This figure has been adapted from ref. 16 with permission from Elsevier, copyright 2025. | ||
Another widely researched pathway is receptor-mediated transcytosis (RMT). Ferritin-based nanoparticles have been engineered to exploit the transferrin receptor 1 (TfR1), highly expressed on BBB endothelial cells. Guo et al. demonstrated that ferritin nanocarriers loaded with therapeutic agents could efficiently cross the BBB via this route while maintaining biocompatibility and low immunogenicity. Surface engineering further improved their stability and binding to brain-targeting ligands, enhancing both diagnostic and therapeutic utility in CNS disorders.14 Chemical and physical BBB manipulation techniques are also being explored. Giantini-Larsen et al. reviewed multiple methods including hyperosmolar therapy (e.g., mannitol-induced BBB opening), convection-enhanced delivery (CED), and magnetic resonance-guided focused ultrasound (MRgFUS). These methods are currently under clinical investigation and offer routes to bypass or modulate the BBB transiently to allow large molecular therapeutics to reach intracranial targets.13 Additionally, cascade-responsive nanocarriers designed to release their therapeutic payloads only upon encountering tumor-specific conditions such as acidic pH or elevated glutathione demonstrate improved BBB penetration and intratumoral specificity. Li et al. engineered glycosylated nanoparticles capable of penetrating the BBB and activating CRISPR/Cas9 machinery in GBM tissues, significantly reducing tumor burden and improving survival in mouse models.64
One striking example is the cascade-responsive glycosylated nanoparticle platform by Li et al., which effectively crossed the BBB and delivered CRISPR/Cas9 payloads targeting PD-L1 in GBM. This led to significant tumor suppression and has shown feasibility with signals of efficacy in preclinical models when combined with temozolomide chemotherapy.64 On the immunotherapy front, engineered nanoparticles can modulate the tumor microenvironment and enhance immune activation. Hao et al. developed a biomimetic nanocomplex carrying cisplatin and polyphenols, coated with programmed death-1 (PD-1)-overexpressing microglial membranes, and conjugated with angiopep-2 for BBB targeting. This system not only induced tumor cell pyroptosis via GSDME activation but also promoted dendritic cell maturation and cytotoxic T-cell infiltration in GBM-bearing mice, achieving potent chemo-immunotherapy synergy (Fig. 6).76 Furthermore, nanoenzyme-based systems are emerging as a hybrid platform for gene-immunotherapy. Wang et al. reviewed the multifunctional role of nanoenzymes in GBM therapy, emphasizing their capacity to catalyze in situ reactions, modulate redox conditions, and act as both imaging and therapeutic agents. These systems can amplify therapeutic effects while ensuring targeted activity within the tumor microenvironment.17
![]() | ||
| Fig. 6 Summary of blood–brain barrier (BBB) transport mechanisms for magnetic nanoparticles, engineered biomimetic cisplatin-polyphenol nanocomplex for chemo-immunotherapy of glioblastoma by inducing pyroptosis. Typical parameters: nanoparticle size 10–80 nm, surface potential −10 to +10 mV, and magnetic field strengths <15 kA m−1 for safe intracranial exposure. This figure has been adapted from ref. 76 with permission from Springer Nature, copyright 2025. | ||
![]() | ||
| Fig. 7 Clinical workflow of dual-cross-linked laminin supplemented HA-SH hydrogels exhibit brain-like mechanical properties and support high cell viability and neuronal maturation. (a) SEM images of PCL scaffolds in a triangular design, (b) chemical composition of a HA-SH hydrogel dual cross-linked with linear and 8-arm Star PEGAcr. (c) Laminin-211 is covalently bound in the hydrogel and is bioavailable for seeded cells. (d) Storage modulus (G’) was measured for 420 kDa HA-SH and 230 kDa HA-SH, shown as mean values and standard deviation. (e) Cyclic loading behavior during 3 cycles for dual cross-linked 420 kDa HA-SH (n = 6). (f) Porosity of dual-cross-linked 230 and 420 kDa HA-SH hydrogel obtained via cryo-SEM microscopy. (g) Dynamic viscosity measurement of dual cross-linked 420 kDa HA-SH, mean and SD depicted (N = 3). (h) Representative images of GL-261 glioma cell viability staining for Matrigel and laminin supplemented HA-SH. (i) Percentages of cell viability for GL-261 glioma cells in Matrigel and HA-SH + laminin, values are shown as mean and standard error of the mean (SEM). (j) Immunostainings of GL-261 glioma cells in laminin supplemented HA-SH and primary cortical neurons co-cultures with primary astrocytes. This figure has been adapted from ref. 78 with permission from Wiley, copyright 2025. | ||
Targeting specificity is another major focus of in vitro nanoparticle testing. Krapež et al. characterized a novel nanobody (NB3F18) against FREM2 a membrane protein overexpressed in glioblastoma stem cells. In vitro flow cytometry and confocal microscopy confirmed selective binding and internalization of NB3F18 into stem-like GBM cells, while sparing astrocytes and differentiated tumor cells. This finding supports the use of nanobodies and ligands to functionalize MNPs for cell-selective delivery systems.79 Together, these in vitro studies illustrate the diversity of strategies being validated for magnetic nanoparticle-based brain cancer therapy from hyperthermia and gene delivery to targeted immunotherapy. While results are promising, translation to in vivo systems requires further validation of safety, stability, and functional performance under physiological complexity.
| No. | Nanoparticle type | Animal/model | Tumour type | Route | Imaging modality | Outcome metric | Key findings | Reference |
|---|---|---|---|---|---|---|---|---|
| 1 | Lipid nanoparticles carrying circPRKD3 | Orthotopic GBM, BALB/c nude mice (n = 10) | U87-MG | i.v., 5 mg NP per kg | Bioluminescence | ΔOS = +48%; ΔTGI = 68% | STAT3 inhibition, enhanced CD8+ T-cell infiltration, significant tumour suppression and prolonged survival | 80 |
| 2 | Iron oxide + gold core–shell nanoparticles | Orthotopic GBM, C57BL/6 mice (n = 12) | GL261 | i.t., 4 mg Fe eq. per kg | MRI + photothermal | ΔT = +5.1 °C; ΔOS = +35% | Dual-mode imaging and therapeutic heating; efficient BBB crossing (not yet confirmed in human BBB physiology) | 49 and 81 |
| 3 | Multifunctional polymeric nanocarriers (PEG-PLGA-SPIONs) | Orthotopic GBM, Wistar rats (n = 8) | C6 | i.v., 8 mg Fe eq. per kg | MRI, PET, optical | ΔTGI = 73% | Deep tumour targeting, reduced immune uptake, improved survival compared with control | 81 |
| 4 | Polymeric nanoparticles (PLGA-based with doxorubicin) | Subcutaneous GBM, nude mice (n = 6) | U251 | i.t., 6 mg Fe eq. per kg | MRI-guided | ΔOS = +42%; ΔT = +3.8 °C | Tumour-specific drug release with minimal systemic toxicity and enhanced local control | 50 |
In terms of immune modulation, Zhang et al. demonstrated a lipid nanoparticle platform carrying exosomal circPRKD3 that significantly suppressed tumor growth and reprogrammed the glioblastoma microenvironment in vivo. The nanocarriers inhibited STAT3 signalling and promoted CD8+ T cell infiltration, highlighting the synergy between magnetic nanoparticle delivery and immune checkpoint blockade therapy. Mice receiving this treatment exhibited improved survival, emphasizing the promise of combinatorial nanomedicine approaches (Fig. 8).80 Imaging-guided delivery and monitoring are enhanced by functionalization strategies. Nanocarriers tagged with fluorescent, radiolabel, or MRI contrast agents allow researchers to track accumulation and distribution in intracranial tumor models. These systems have been used to monitor drug release, evaluate tumor penetration, and assess therapeutic responses over time without sacrificing the animals. High-resolution MRI remains the most widely used modality, particularly with iron oxide-based MNPs, due to their strong T2 relaxivity and biocompatibility. Furthermore, animal models such as orthotopic glioma-bearing mice and genetically engineered rat models are indispensable for testing MNP-assisted BBB penetration strategies. Lipid-coated MNPs with targeting ligands (e.g., angiopep-2, transferrin) have demonstrated successful transcytosis and accumulation in brain tumors, providing insights into nanoparticle trafficking, degradation, and clearance in vivo. Despite these advances, key challenges remain. These include ensuring nanoparticle stability in circulation, avoiding off-target accumulation (e.g., liver and spleen), achieving deep tumor penetration, and minimizing immunogenicity. Nonetheless, the integration of imaging and therapeutic modalities via MNPs in live models continues to accelerate the translation of these platforms from bench to bedside.
![]() | ||
| Fig. 8 CircPRKD3-loaded exosomes concomitantly elicit tumor growth inhibition and glioblastoma microenvironment remodeling via inhibiting STAT3 signaling. This figure has been adapted from ref. 80 with permission from Oxford University Press, Creative Commons Attribution-NonCommercial License copyright 2025. | ||
Although numerous animal studies report efficient BBB penetration and intracranial accumulation of MNPs, translation to human physiology remains highly variable. Differences in BBB thickness, endothelial receptor expression, and cerebrovascular flow significantly influence transport efficiency. For example, transferrin and Angiopep-2 ligands repeatedly demonstrate enhanced brain uptake in rodent and primate models, yet their quantitative penetration in humans remains under 1–2% of injected dose. Similarly, dosing routes such as intra-arterial administration and focused ultrasound-assisted delivery yield higher local deposition but are constrained by invasiveness and safety considerations. Therefore, all preclinical findings describing efficient BBB crossing should be interpreted as model-dependent indicators of potential rather than direct predictors of clinical performance.
| Trial ID | Nanoparticle system/therapy | Indication | Phase | Status (as of oct 2025) | Sample size (N) | Primary endpoint/key findings |
|---|---|---|---|---|---|---|
| a Status and last update verified viahttps://clinicaltrials.gov/ and EudraCT on 9 November 2025. | ||||||
| NCT02033447 | NanoTherm® (MagForce AG) aminosilane-coated SPIONs for magnetic hyperthermia + radiochemotherapy | Recurrent GBM | I/II | Completed (Germany) | ≈59 | Feasibility, safety, local tumour control, median OS ≈ 13 month vs. ∼6 month historical benchmark |
| EudraCT 2018-004572-24/NCT04899908 | AGuIX® (NH TherAguix SA) gadolinium-based nano-radiosensitizer + RT/TMZ | Newly diagnosed GBM | II | Active, not recruiting (planned completion 2025) | 94 (planned) | Safety, acute toxicity, MRI contrast, radiosensitization efficacy |
| NCT05245611 | Iron-oxide NP + focused ultrasound (FUS) transient BBB opening | High-grade glioma | I | Recruiting | ∼20 (planned) | BBB permeability (MRI), safety profile |
| NCT04523490 | PEG–SPION composite intra-arterial magnetic hyperthermia | Recurrent GBM | I | Active, not recruiting | ∼12 | Feasibility and tolerability of repeated AMF cycles |
| NCT05673410 | Temozolomide-loaded SPIONs local drug release in post-resection cavity | GBM (post-surgery) | I | Recruiting | ∼15 | Drug-release kinetics, local recurrence rate, safety |
![]() | ||
| Fig. 9 Scheme of the action of nanoparticles to increase the radiosensitivity of GBM. This figure has been adapted from ref. 84 with permission from MDPI, copyright 2025. | ||
Radio-liposome nanocarriers (e.g., rhenium-186) are in preclinical to early-phase stages and may complement existing therapies. These diverse nanoparticle-based strategies (as shown in Table 3), ranging from local hyperthermia to advanced radiosensitisation and immunomodulation, highlight the expanding clinical landscape. Several first-in-human trials are exploring image-guided or drug-loaded magnetic nanoparticles. These include iron-oxide nanoparticles combined with focused ultrasound for transient BBB opening (NCT05245611), PEG-SPION composites for intra-arterial magnetic hyperthermia (NCT04523490), and temozolomide-loaded SPIONs for local drug release in post-resection cavities (NCT05673410). Collectively, these studies underscore growing translational momentum toward precision, image-guided magnetic nanotherapies in neuro-oncology.
From a manufacturing and scalability standpoint, producing uniform MNPs with reproducible physicochemical and magnetic properties at clinical-grade standards is technically demanding. Variability in size, surface charge, and coating integrity can significantly affect both efficacy and toxicity. Furthermore, large-scale Good Manufacturing Practice (GMP) compliance requires stringent quality control, which is costly and complex for multifunctional hybrid nanocarriers. Magnetic field limitations also pose a barrier. While AMF-based hyperthermia has shown success in trials like NanoTherm®, delivering spatially controlled heating in human brain tissues is significantly harder than in preclinical models. Ensuring that magnetic fields penetrate deeply and safely without causing off-target tissue damage requires high-precision hardware and careful thermal dose monitoring. Moreover, regulatory and ethical hurdles slow clinical progression. The classification of MNPs straddles both drug and device categories, complicating regulatory approval. Uncertainties in nanoparticle pharmacokinetics, immunogenicity, and long-term accumulation pose challenges to risk-benefit assessments by regulatory agencies. Ultimately, clinical trial design in neuro-oncology presents unique challenges. Patient recruitment is limited by glioblastoma's aggressive course, and standardized endpoints for evaluating nanoparticle-based treatments such as thermometry, imaging contrast enhancement, or immunomodulation are still evolving.
Magnetic nanoparticle-based therapeutic systems occupy a regulatory “hybrid” zone, functioning as device–drug combination products. Under the European Union Medical Device Regulation (EU MDR 2017/745), systems such as NanoTherm® are categorized as class III active implantable devices, requiring demonstration of both magnetic-field safety and biocompatibility. The U.S. FDA follows a similar classification via the Office of Combination Products (21 CFR Part 3), emphasizing coordinated review between the Center for Devices and Radiological Health (CDRH) and the Center for Drug Evaluation and Research (CDER). Clinical approval pathways mandate precise thermometry validation, verifying that AMF remain within the safety threshold (H·f ≤ 5 × 109 A m−1 s−1). Equally important is long-term deposition monitoring, as iron oxide cores may persist in the reticuloendothelial system (RES). MRI follow-up, serum iron markers, and patient registries can provide valuable post-treatment biodistribution data.
Post-market surveillance for such hybrid products increasingly aligns with ISO 13485 and ISO 10993 standards, requiring continuous reporting of adverse events, field calibration logs, and nanoparticle degradation assessments. Establishing an integrated registry for MNP-based interventions analogous to those for cardiac implants would enhance safety tracking and public confidence.
Another limitation involves non-specific biodistribution. Once in circulation, MNPs often accumulate in the liver and spleen due to uptake by the reticuloendothelial system (RES), reducing the therapeutic payload reaching the brain tumor site. Additionally, the heterogeneity of tumor vasculature and high interstitial fluid pressure within brain tumors further impede deep penetration and uniform distribution of nanoparticles across the tumor mass. From a materials standpoint, toxicity and long-term biocompatibility of MNPs, especially those using synthetic coatings or metal-based cores, remain unresolved. Even though iron oxide nanoparticles are generally considered safe, the degradation products, surface charge, and coating stability can influence immune responses and organ retention. Moreover, scale-up reproducibility for clinical-grade MNPs and standardisation of magnetic hyperthermia parameters (e.g., frequency, amplitude, field exposure time) are still underdeveloped, which can compromise therapeutic outcomes and inter-study comparability.
Hybrid nanoparticle systems, combining magnetic cores with biocompatible polymers, lipids, or exosomes, are being engineered to improve circulation time, evade immune detection, and facilitate BBB crossing via receptor-mediated transport. Similarly, cell-mediated delivery using monocytes or stem cells loaded with MNPs leverages the innate homing ability of these cells to penetrate tumor niches effectively. On the translational front, multimodal MNPs integrating imaging (MRI, PET) and therapy (drug delivery, hyperthermia) into a single platform are paving the way for real-time tracking and personalized treatment adjustments.
One primary concern is accumulation in non-target organs, particularly the liver, spleen, and kidneys, following systemic administration. Even after degradation, the fate of by-products especially metal ions or synthetic surface coatings can lead to oxidative stress, inflammation, or immune activation. Moreover, the long-term retention of poorly biodegradable nanoparticles may result in chronic toxicity, a risk that is not fully understood due to the lack of longitudinal clinical studies. Surface coatings (dextran, PEG, silica, or polymers) meant to improve circulation and prevent aggregation may also elicit immune responses or accelerate clearance, depending on their molecular weight and architecture. Additionally, repeated dosing critical for long-term management of aggressive brain tumors raises concerns about immunogenic memory and cumulative organ burden. Rigorous in vivo studies in relevant models are needed to assess biodistribution, clearance routes, and any histopathological changes in critical organs. Furthermore, standardization of safety metrics and regulatory benchmarks for nanoparticle-based therapeutics is urgently required, as current frameworks often lag behind the pace of nanomedical innovation. To facilitate rational selection and translation of magnetic nanoparticle (MNP) platforms for brain tumour applications, Table 4 presents a comparative framework that integrates key physicochemical and operational design parameters with their mechanistic rationale, safety limits, and levels of supporting evidence. The framework consolidates preclinical and clinical insights reported in glioblastoma and high-grade glioma models.
| Design variable | Typical range | Mechanistic rationale | Constraint/safety limit | Evidence strength (preclinical/clinical) | Key references |
|---|---|---|---|---|---|
| Particle size | 10–100 nm | Optimizes circulation time, BBB transcytosis, and tumour retention; <10 nm cleared renally, >100 nm captured by RES | Avoid >120 nm to reduce hepatic uptake | Strong (preclinical); limited (clinical) | 45–47 |
| Surface charge (ζ-potential) | −10 to +10 mV (near neutral) | Minimizes protein adsorption and non-specific uptake; enhances BBB penetration | Highly charged NPs (>±25 mV) cause aggregation or cytotoxicity | Strong (preclinical); limited (clinical) | 45 and 46 |
| Ligand class | TfR, lactoferrin, Angiopep-2, RGD peptides | Receptor-mediated endocytosis across BBB and glioma-specific uptake | Ligand density must balance targeting vs. immune activation | Strong (preclinical); emerging (clinical) | 42–44 and 46 |
| Magnetic field product (H·f) | ≤5 × 109 A m−1 s−1 | Safe operational window for hyperthermia avoiding eddy-current heating | Atkinson–Brezovich limit; intracranial exposure <15 kA m−1@100–300 kHz | Validated (preclinical & human NanoTherm® trials) | 2, 47 and 51 |
| Specific absorption rate (SAR) | 100–800 W g−1 Fe | Governs heating efficiency during AMF exposure | Excessive SAR may overheat tissues; requires local thermometry | Moderate (preclinical) | 63 |
| Administration route | Intravenous (i.v.), intra-arterial (i.a.), convection-enhanced (CED), MRgFUS-assisted | Dictates biodistribution and tumour deposition efficiency | i.a. and CED invasive; MRgFUS transiently disrupts BBB | Moderate–strong (preclinical); early clinical data | 13, 16 and 51 |
| Outcome metrics | ΔT (°C), ΔOS (months), ΔTGI (%) | Quantitative indicators of hyperthermic efficacy and tumour response | Context-dependent; ΔOS ∼ +6–8 months in NanoTherm® GBM trials | Strong (clinical) | 24, 76 and 77 |
| MRI relaxivity (r2) | 100–250 mM−1 s−1 (Fe-based NPs) | Determines contrast enhancement for imaging-guided therapy | Excess Fe accumulation may cause susceptibility artefacts | Strong (preclinical & clinical diagnostic use) | 40 and 56 |
![]() | ||
| Fig. 10 Challenges and limitations of magnetic nanoparticles in brain cancer therapy for the future. | ||
Footnote |
| † Authors contributed equally. |
| This journal is © The Royal Society of Chemistry 2026 |