Magnetic Hydrogels as a Treatment for Oncological Pathologies
Abstract
1. Introduction
2. Synthesis Methods for Magnetic Hydrogels
2.1. Crosslinking Strategies
2.2. Preparation Strategies for Magnetic-Based Hydrogels
| Preparation Method of Magnetic Hydrogel | Polymer Type | Hydrogel Matrix | MNPs or Other Magnetic Structures/Preparation Method/MNPs’ Concentration | Magnetic Field-Related Devices, Set-Ups, and Properties | Remarks | Ref. |
|---|---|---|---|---|---|---|
| In situ blending method | Synthetic | Polyethylene glycol (PEG) | Ferromagnetic vortex domain iron oxide/Co-precipitation/0.6 mg/mL | 10 min exposure to an alternating magnetic field (AMF) (220 Oe (17.5 kA/m), 495 kHz)/SAR of about 371 W/g (150 Oe (11.9 kA/m)) and 856 W/g (220 Oe) | Compared to control samples, which were manufactured with superparamagnetic iron oxide MNPs, almost double values of SAR were achieved | Gao et al. [63] |
| Poly(N-isopropylacrylamide-co-acrylic acid) (poy(NIPAAm-co-AA)) | Fe3O4 MNPs/Co-precipitation method/Fe3O4 magnetic fluid 1% molar ratio of the other components | VSM characterization/MG saturation magnetization Ms of 15 emu/g (15 Am2/kg) compared to 45.8 emu/g (48.5 Am2/kg) for pure MNPs | The prepared magnetic hydrogel exhibited superparamagnetic properties | Fan et al. [67] | ||
| Natural/Synthetic | Alginate/polyacrylamide (PAAm) crosslinked to Fe3+ ions | Alginate-coated Fe3O4/Co-precipitation method/Weight percent of MNPs in ferrofluid was 1, 2, 3, 4, 5, 10, and 20 wt.% with respect to the total weight of water and polymers | VSM (735 VSM Controller, LakeShore, Westerville, OH, USA) with a maximum applied field of 9000 Oe (71.6 kA/m) at 23 °C/Ms between 18 emu/g (18 Am2/kg) for 20 wt.% MNPs and 1 emu/g (1 Am2/kg) for 1 wt.% for 1 wt.% MNPs | The prepared magnetic hydrogels exhibited outstanding mechanical properties with an increased toughness point and adequate magnetic superparamagnetic behavior | Haider et al. [62] | |
| Dual-network sodium alginate/poly(vinyl alcohol) PVA physically crosslinked with Ca2+ and freezing–thawing cycles | Magnetic laponite nanodiscs/Fe3+/Fe2+ co-precipitation in laponite/magnetic beads with 16.7 and 28.5 wt.% magnetic laponite | VSM (model 7400, LakeShore, Westerville, OH, USA)/measurements performed at ±9 kOe (71.6 kA/m) at 298 K/Ms of 2.9 emu/g (2.9 Am2/kg) and 13.3 emu/g (13.3 Am2/kg) | By adding magnetic laponite, the swelling capacity of the hydrogel decreased, but a magnetic response was achieved | Mahdavinia et al. [65] | ||
| Chitosan/PEG | Fe3O4 MNPs/Co-precipitation/0 ÷ 40 wt.% | 20 A at magnetic field strength (H) of 12 kA/m for 2.5 min and 15.5 A at H of 9.6 kA/m for another 15 min/A temperature of about 46.5 °C was achieved for 20A, while 43 °C was obtained for 15.5 A (both measurements were made for a concentration of about 30 wt.% MH) | The MH with 30 wt.% MNPs concentration was selected as an optimal platform for MHT application | Cao et al. [68] | ||
| Natural | Collagen | Fe3O4 MNPs/-/0.5 mg/mL | By applying a constant magnetic field generated by a 255 G permanent magnet, a normal electrical activity was established in the aligned collagen fibers of MG | A new method based on gel orientation external control inside the patient’s body was proposed by directly injecting MNPs at the defect site and manipulating them based on a permanent magnet | Antman-Passig and Shefi [69] | |
| Agarose/collagen | Streptavidin-coated MNPs and Nano-HA-coated—γ-Fe2O3 MNPs/-/10 v/v% | By using a 2 mT cylindrical magnet (Supermagnete, Gottmadingen, Germany), a similar structure with natural multilayered tissues was obtained | The MNPs were used to align the collagen fibers in a given direction to obtain tissue-engineered structures | Betsch et al. [70] | ||
| Type I Collagen | Magnetically oriented silica SiO2@Fe3O4 rods/Synthesized in the high temperature range through thermal decomposition of iron-tris(acetylacetonate) in silica rods and aligned parallel with a magnetic field based on a house-made device with two plate magnets (Nd 70 × 50 × 5 mm Ni-Cu-Ni N45)/30 mg/mL | SQUID magnetometer (Quantum Design MPMS-XL equipped with 7 T direct current (DC) magnet)/magnetic measurements performed at 2 K and zero field curves (ZFC) for a temperature range 2 K to 250 K at 100 Oe (7.9 kA/m) with temperature rate of 2 K/min/Ms of 4.5 emu/g (4.5 Am2/kg) obtained at 2 K from hysteresis cycle and 1.1 emu/g (1.1 Am2/kg) for ZFC investigation | Orientation of the magnetic nanorods exhibited an important influence on the rheological properties of MG and a moderate effect on magnetic properties | Shi et al. [66] | ||
| Agarose | Commercial dextran-coated Micromod® (Micromod, Rostock, Germany, product code 84-00-102) nanoparticles/20 wt.% (surface), 7 wt.% (middle), and 10 wt.% (deep) [71] | The magnetocompressive response of MG under the action of an external magnetic field was investigated. Magnetic field source: NdFeB magnets (0.4, 0.5, or 0.75 T; Grade N42, E-Magnets®, Berkhamsted, UK). By varying the MNPs’ concentration an almost linear dependence of engineering strain was achieved for agarose 1 wt.% gel, while a more constant trend was noticed in the case of 2 wt.% agarose gel | The MG behavior under the effect of an external magnetic field showed biochemical gradients and depth-dependent strain after 14 days of immersion in cell culture | Brady et al. [72] | ||
| In situ precipitation method | Synthetic | Polyethylene glycol modified with factor XIIa (PEG) | PEG-functionalized MNPs (Fe3O4 and Fe2O3)/Co-precipitation/1 mg/mL | A static magnetic field (SMF) was made by a Nd-Fe-B cylindrical magnet (thickness of 1 mm, diameter of 20 mm, magnetic induction 50 mT, Supermagnete, Uster, Switzerland). The magnetic analysis was performed in the presence of 12-well stromal vascular fraction cell plates, by placing the SMF (direction according to the north pole) below the wells. In vitro Magnetic Resonance Imaging (MRI) equipped with a Bruker Avance 300 Spectrometer (Billerica, MA, USA) were used. In the case of the SMF application, the MRI revealed different signal loss and various relaxation times | The developed magneto-mechanical actuation hydrogel proved to have a very good in vitro cell preconditioning ability | Filippi et al. [77] |
| Acrylamide | Polydopamine-chelated carbon Fe3O4 nanotubes/Co-precipitation/0 ÷ 15 wt.% | Low static magnetic field (30 mT) application generated nanohybrids orientation along the SMF | A mussel-inspired approach to manufacturing a magnetic field-controlled high-conductivity hydrogel for anisotropic medical applications | Liu et al. [78] | ||
| Six-arm star-PEG-acrylate | Superparamagnetic anionic-coated iron oxide nanoparticles (SPIONs—EMG700, Ferrotec, Unterensingen, Germany [99])/Co-precipitation/0.0046 vol% | Alignment of MG was achieved with permanent magnets with various magnetic inductions (100, 130, 300 mT) | The hierarchically designed MG exhibited the property to guide cell and nerve growth | Rose et al. [80] | ||
| Natural/Synthetic | Alginate/PAAm | Fe3O4/Co-precipitation/concentration of Fe2+ and Fe3+ was set 0.1 M and 0.2 M, respectively | Magnetic induction heating was performed using an AMF setup (oscilloscope, water-cooling system, and induction heating system [100]). The solenoid for MHT has a diameter of 40 mm and two turns. AMF was applied for 3 min | A SAR value of 1.30 ± 0.12 W/g was achieved for 5.93 kA/m. It was concluded that the magnetic induction heating character of MG can be controlled as a function of magnetic field strength (5.93 kA/m ÷ 11.86 kA/m) | Tang et al. [73] | |
| Poly(N, N-dimethylacrylamide) (PDMA)/nanoclay composite | Fe3O4/Co-precipitation/14 v/v% | An induction heating system (Easyheat 224, Cheltenham Induction Heating) with a solenoid of 52 mm diameter and two turns. Saturation magnetization Ms = 10.34 emu/g (10.34 Am2/kg), remanent magnetization Mr = 1.33 emu/g (1.33 Am2/kg), coercive field Hc = 32.67 Oe (2.6 kA/m) | An intense heating process was put in evidence in combination with excellent mechanical toughness | Tang et al. [100] | ||
| Natural | RGD peptides changed alginate | MNPs (Fe2O3)/Co-precipitation/7 wt.% | SMF application for 5 min every 12 h based on a magnet (6510 Gs (0.651 T), 1 Hz). The magnetic stimulation of the biphasic ferrogel provides adequate fatigue resistance, while the elasticity modulus and toughness were modified only to a small amount in the first 2 weeks of analysis | The biological-free MG was considered a promising candidate for muscle regeneration | Cezar et al. [79] | |
| Chitosan | Fe3O4/Co-precipitation/magnetite precursor containing 7 mmol Fe3+ and 3.5 mmol Fe2+ | The drug release effect under a low-frequency alternating magnetic field was investigated. The drugs were loaded into 100 mL of simulated medium (phosphate-buffered solution, PBS) under the influence of a 0.4 T magnetic field generated by a rotating magnet at a frequency of 2 Hz. A total of 40% of the active drug (Adriamycin) was released in the first 2 h and was still sustained after 4 h | An improvement in the drug release phenomenon by 67.2% from the MG was obtained under magnetic field application | Wang et al. [76] | ||
| Bacterial cellulose | Dextran-coated Fe3O4/Co-precipitation/25 ÷ 100 mM | VSM (Quantum Design, San Diego, CA, USA). Magnetization was measured at 300 K under the action of an applied magnetic field strength of ± 11,000 Oe (875.3 A/m). Ms was about 10 emu/g (10 Am2/kg) with no hysteresis cycle | The developed MG was dedicated to being a retention platform and cell capture for vascular tissue regeneration in the case of an intracranial aneurysm | Arias et al. [81] | ||
| Ex situ crosslinking | Synthetic | Thermosensitive P(NIPAm-co-Am) chelerythrine (CHE) drug loading | Fe3O4@SiO2 modified with vinyl groups/Solvothermal method and modified with silica by the TEOS/300 mg | VSM (Quantum Design PPMS-9, Beijing, China). The MG was characterized by zero values for coercivity and remanence with no hysteresis cycle. The saturation magnetization was estimated at 3.59 emu/g (3.59 Am2/kg). An AMF (5 kA/m, 100 kHz) led to about 42 °C within 30 min | The main conclusion of this study was that composite MNPs included in the hydrogel matrix resulted in an MG that could be considered for MHT applications and drug delivery | Wang et al. [87] |
| Polyethylene glycol | Thiol-functionalized magnetic microparticles/−/− | Chronic magnetomechanical stimulation (4 days for 30 min/day (day 1: 0.145 μN, day 2: 0.244 μN, day 3: 0.457 μN, day 4: 1.00 μN)) was applied to modulate the expression of the human gene PIEZO2 | The model presented in the paper can be used to stimulate neural types and excitable cells magnetomechanically | Tay et al. [89] | ||
| Natural/Synthetic | Chitosan-polyolefin double network | Acid etched Fe3O4/Solvothermal method/0 ÷ 16 mg/mL | Magnetothermal properties of hydrogel were investigated under the action of AMF (282 kHz, 180 Oe (14.3 kA/m)) and showed a dose dependence variation | The developed hydrogel exhibited good mechanical properties, increased magnetothermal effects, self-healing capabilities, and high biocompatibility | Gang et al. [91] | |
| Natural | Dextran | Dextran-coated Fe3O4/Co-precipitation/6 wt.% | MPMS3 Quantum Design SQUID Magnetometer (Quantum Design Inc., San Diego, CA, USA). At 300 K, the magnetic hydrogel Ms was about 41.2 emu/g Fe (41.2 Am2/kg) | The MG exhibited good properties for using it as an MRI agent | Su et al. [85] | |
| Chitosan-alginate | 5-Fluorouracil (Fu) loaded magnetic gelatin microspheres containing Fe3O4 MNPs/−/0.2 g | Superconducting quantum interference device magnetometer (Lake Shore 7400, Westerville, OH, USA). Hysteresis curves were measured at 300 K with a magnetic field strength of—60 ÷ 60 kOe (4.7 MA/m). Saturation was achieved at 4.95 emu/g (4.95 Am2/kg) | A superparamagnetic behavior for MG was evidenced in combination with good compressive and rheological properties, proving also self-healing property. The MG was considered adequate for drug delivery in cancer therapy | Chen et al. [84] | ||
| Grafting onto method | Synthetic | Polyacrylamide | Saline modified carbonyl iron (CI) particles/Silane chemistry for surface modification/- | An external magnetic field with variable magnetic flux density between 0 T and 1 T was applied, and an increase in Young’s modulus, from 104 to 105 Pa was achieved | The phenomenon of elastic hysteresis was observed. It was concluded that by modifying the hydrogel stiffness, various differentiations of stem cells could be achieved | Abdeen et al. [94] |
| Natural/Synthetic | Hybrid polymeric material (hyaluronic acid (HA), collagen type II, and polyethylene glycol (PEG) | Poly(vinyl alcohol) modified Fe3O4 | N42 neodymium magnet (D3X0, K&J Magnetics, Inc., Pipersville, PA, USA), with a height of 25 mm and a diameter of 22 mm | The MG exhibited a good biodegradation time and an increased stiffness | Zhang et al. [97] | |
| Methacrylate chondroitin sulfate (MA-CS) enriched with platelet lysate (PL) | MA-CS coated iron oxide MNPs/Co-precipitation/200 and 400 μg/L | Superconducting quantum interference magnetometer (SQUID-VSM, Quantum Design, San Diego, CA, USA) applied magnetic field between—20.0 and 20.0 kOe (1.6 MA/m) at room temperature. Magnetic stimulation was performed with an oscillating magnet array (2 Hz and 0.2 mm of displacement, 0.35 T/per magnet) | The magnetic field application exhibited a control effect on the hydrogel swelling. In addition, the modulation of growth factors released from platelet lysate | Silva et al. [96] | ||
| Natural | Cellulose nanocrystal/dextran | Kartogenin (KGN) ultrasmall superparamagnetic iron oxide grafted Fe3O4@SiO2-KGN/Co-precipitation/0.06 ÷ 0.3 wt.% | VSM analysis. By comparing the saturation magnetization for Fe3O4 and Fe3O4@SiO2 -KGN, it was found in the last case that a decreased value was obtained. In addition, a superparamagnetic character was evidenced, with zero remanence and coercivity | The developed hydrogel was utilized as an MRI agent, producing high-quality images with no artifacts both in vivo and in vitro. Additionally, a long-term KGN release was evidenced | Yang et al. [93] | |
| Bisphosphonate (BP)—modified hyaluronic acid (HA-BP) | Fe3O4/Co-precipitation/3% (w/v) | VSM (Lake Shore 7400, Westerville, OH, USA). The MG exhibited a superparamagnetic behavior with a saturation magnetization of about 20 emu/g (1.6 MA/m) measured at 10 kOe (0.8 MA/m) applied field. To investigate the heat generation properties of MG, 1 mL of MG was subjected to the effect of AMF (528 Oe (4.2 kA/m), 258 kHz). A temperature increase of about 50 °C was reached after about 100 s of field application with a SAR value of 54.5 W/g | The novel MG that is based on dynamic coordination bonds developed between BP and MNPs used as crosslinkers resulted in a smooth, injectable, shear-thinning, and self-healing hydrogel. It was concluded that it can be successfully applied in MHT treatments | Shi et al. [90] | ||
| Chitosan crosslinked with doxorubicin (DOX) and docetaxel (DTX) drug loading | Telechelic difunctional poly(ethylene glycol) (DF-PEG-DF) modified Fe3O4/Hydrothermal method/10 mL | SQUID MPMS XL-7 magnetometer. A saturation magnetization value of 80 emu/g (80 Am2/kg) was achieved at 10 kGs (1 T) magnetic flux density. The induced heating capability was analyzed under AMF (19.99 kA/m, 282 kHz) influence, and a temperature rise of 28 °C was achieved | The developed dual-drug-loaded MG could be successfully used as synergistic chemotherapy for triple-negative breast cancer | Xie et al. [88] | ||
| Agarose | Glycosylated MNPs/−/1011 surface modified MNPs in 100 μL 1 wt.% agarose hydrogel | SQUID magnetometer (MPMS-7, Quantum Design, Surrey, UK). A saturation magnetization Ms of 53.6 ± 0.4 emu/g (53.6 ± 0.4 Am2/kg) and a magnetic susceptibility of 0.54 emu/(g·Oe) (147 m3/g) was obtained for glycosylated MNPs | The magnetic field influence was used for alignment of hydrogen to generate smooth biochemical gradients for cartilage tissue regeneration | Li et al. [95] |
3. Magnetic Hydrogel Attributes for Oncological Use
| Property | MG Hydrogel Characteristics | Type of Possible Application in Cancer Therapies | In Vitro/In Vivo Models | Magnetic Measurements/Devices for Magnetic Field Generation | Magnetic Field-Guided or Other Physical Mechanisms | Important Study Properties | Ref. |
|---|---|---|---|---|---|---|---|
| Injectability | Supramolecular MG self-assembled by PEGylated Fe3O4 MNPs and α-cyclodextrins (CD) | Thermo-chemotherapy (Magnetocaloric effect + MHT) for locoregional recurrence in breast cancer/MNPs were loaded with paclitaxel (PTX) or doxorubicin (DOX) | In vivo: breast mouse model (female BALB/c mice—4 weeks, 20–25 g) | VSM (LakeShore VSM 7407, Westerville, OH, USA): Ms—93.9 emu/g (93.9 Am2/kg) at 300 K, SAR—1334 W/g Fe (heating test time 120 s) Water-cooled copper coil powered by AMF (410 kHz, 1.8 kA/m) | The drug release was accelerated due to magnetically generated heat of MNPs under AMF irradiation | Rheological parameters of MG: maximum values at 14 min and at 37 °C for storage modulus (G’) and loss modulus (G”): G’ = 6000 Pa, G” = 1000 Pa, viscosity value at 37 °C: 400 mPa·s; minimum value of viscosity at 37 °C and a shear rate of 100 (s−1) 2000 mPa·s | Wu et al. [103] |
| Crosslinked oxidized hydroxypropyl cellulose with carboxymethyl chitosan preloaded with artesunate (ART), ferroferric oxide (Fe3O4) MNPs, black phosphorous nanosheets (BPs) | Drug delivery system with magnetic targeting, chemodynamic therapy, pH sensitivity, and photothermal therapy for eliminating hepatocellular carcinoma (HepG2) tumor | In vitro: Human HCC cell line (HepG2) and normal liver cell L02 In vivo: male BALB/c (4 weeks old) | VSM (LakeShore 7400, Westerville, OH, USA): Ms—0.6 emu/g (0.6 Am2/kg) at 300 K Mouse anatomy-adapted 3D printed magnetic device for control of MG to the target location and tumor treatment | When the action of the magnetic field (MF) was added, it improved the targeting effect. When MF was combined with near-infrared laser irradiation (1 W/cm2, 5 min) tumor inhibition was achieved | Rheological parameters of MG: minimum values at 100% strain: G’ = 50 Pa, G” = 400 Pa and maximum values at a frequency of 100 Hz: G’ = 2450 Pa, G” = 200 Pa (a shear-thinning effect was noticed); minimal value of viscosity at a shear rate of 100 (Hz) was 50 Pa·s | Ma et al. [104] | |
| Silk fibroin and iron oxide nanocubes coated with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG2000) | Remote hyperthermia performance combined with thermal ablation under AMF for hepatocellular carcinoma treatment | In vitro: enzymatic degradation of MG, 4T1 cell survival analysis In vivo: MHT test on BALB/c female mice (6–8 weeks old), ultrasound-guided interventional MHT on VX-2 xenografted tumor-bearing New Zealand rabbits | SQUID (Quantum Design MPMS XL): Ms—1.9 emu/g (1.9 Am2/kg) at 300 K and 30 kOe (2.4 MA/m). MHT test: AMF (312 kHz, 30 kA/m) Ultrasound-guided MHT: AMF (312 kHz, 30 kA/m) for 15 min | For MHT session, a magnetic field strength of 30 kA/m was applied and a temperature of 46.1 °C was reached after 3 min. AMF application, and 49.6 °C at 6 min. AMF action was achieved. For the deeper located transplantation liver tumor, when AMF with a strength of 30 kA/m was applied, a satisfactory magnetocaloric effect was considered | Rheological parameters of MG: G’ decreased with increasing shear strain (at a shear strain of 200% a value of 2.8 GPa), while the loss firstly increased to 20 Pa and then decreased to 15 Pa in the same testing conditions. It was noticed that the MG began to liquify at a shear strain of 25% | Qian et al. [105] | |
| Polyethylene glycol, oil-in-water, and oleic acid (OA)-coated Zn ferrite MNPs | Multimodal imaging-guided thermoablative cancer therapy (MHT) | In vivo: 4T1 xenografted tumor mice | VSM measurements at 300 K evidenced a Ms of 98.7 emu/g Fe (98.7 Am2/kg), higher than 56 emu/g Fe (56 Am2/kg) obtained for Ferumoxytol Water-cooled magnetic induction copper coil (diameter of 3 cm, length of 1.5 cm) | For in vitro magnetic heating performance analysis, an AMF (410 kHz, 1.8 kA/m) was applied for 15 min. A SAR value of 872 W/g Fe was achieved For in vivo multimodal-imaging-guided MHT, a temperature of 60 °C after 120 s AMF irradiation was noticed. After 300 s, the entire tumor was heated, and only a few degrees of increase were obtained in the neighborhood tissue | Rheological parameters of MG: maximum values at 50 °C of G’ = 22 Pa and G” = 19 Pa; viscosity at 50 °C and at 5 Hz equal to 10 Pa·s | Wu et al. [107] | |
| Pluronic F-127 matrix with γ-Fe2O3 MNPs loaded with DOX | Combined MHT and localized drug delivery for progressive adenocarcinoma of the ovary treatment | In vitro: OVCAR3 cell line | VSM (MDKB, Iran): Ms = 20 emu/g (20 Am2/kg) at room temperature and 20 kOe (1.6 MA/m) | Based on in vitro MHT tests, a temperature of about 45 °C was achieved. At the application of AMF (400 kHz, 5 min.) 75% of the DOX drug was released | Rheological parameters of MG: viscosity at 100 Hz and 10 °C (sol) was about 200 Pa·s, and at 35 °C (gel) was equal to 2700 Pa·s | Farzaneh et al. [108] | |
| Carboxymethylcellulose (CMC) polymer and CoFe2O3 functionalized with aminopropyl silane (NP) | Potential applications for a targeted drug delivery system | - | - | - | Rheological parameters of CMC-NP hydrogel varied after they were squeezed through a syringe from 1050 ± 140 Pa to 190 ± 20 Pa for G’, and from 80 ± 15 Pa to 20 ± 3 Pa in the case of G” | Barbucci et al. [109] | |
| Carboxymethylcellulose (CMC) polymer and CoFe2O3 MNPs (NP) with a concentration of 50% and 70% in relation to the polymer quantity | Drug delivery under alternating and static magnetic fields | - | - | - | Rheological parameters were determined to be a function of MNPs percent: G’—CMP-NP-50 (50% MNPs): 3300 ± 300 Pa, CMP-NP-70 (70% MNPs): 2000 ± 200 Pa; G”—CMP-NP-50: 90 ± 20 Pa, CMP-NP-70: 110 ± 20 | Uva et al. [110] | |
| Carboxymethylcellulose (CMC) polymer and Fe3O4 MNPs functionalized with aminopropyl silane | Drug delivery | In vitro: MG-63 cell line | SQUID (Quantum Design MPMS): Ms 16 Am2/kg at 2.5 K and 14 Am2/kg at 300 K | The release of doxorubicin was investigated in the absence of a magnetic field, under the effect of an AMF (40 kHz, 2 mT) or a static field (SMF) (0.5 T). The best drug release kinetics was evidenced under the AMF condition | Rheological parameters: G’ = 3250 ± 120 Pa, G” = 258 ± 30 Pa | Uva et al. [111] | |
| Shear-thinning | Hyaluronan (HA) filled with alumina (Al2O3) and multicore MNPs (MCPs). MCPs were characterized by clusters of superparamagnetic FeOx nanoparticles | Possible application for MHT treatment and bioprinting | In vitro: mouse embryonic fibroblast cell line (ATCC CRL-1658 NIH/3T3, USA) for cytotoxicity investigation and bioprinting with BALB/3T3 mouse fibroblast cell line | - | Laboratory-made AMF generator system (signal generator Agilent 33512 (Keysight Technologies, Santa Rosa, CA, USA), RF broadband amplifier AR RF/Microwave Instrumentation 800A3A, induction coil (90 mm diameter), magnetic field sensor, interchangeable capacitors) was used. MHT experiments were performed at 525 kHz and a variable magnetic field of 5.4 ÷ 9.4 mT, or at 1050 kHz and induction amplitude of 5.4 ÷ 7.4 mT. Temperature was measured with the monitoring system ReFlex 4, Neoptix (Qualitrol, Quebec, Canada). A SAR value of 25 W/g at 1050 kHz and an amplitude of 7.4 mT was obtained | Rheological parameters: A distinct shear thinning behavior was evidenced with viscosity modifications in the range of 102 Pa·s with a shear rate range of 102 Hz. A storage modulus of about 300 Pa was measured | Vitkova et al. [112] |
| Polyacrylamide (PAM) matrix with Fe3O4 MNPs bonded to the crosslinked PAM network based on hydrogen bonding | Possible application for MHT | - | VSM (Lakeshore 8604, Westerville, OH, USA). Measurements were performed at room temperature and at a maximum magnetic field strength of 8 kOe. Ms for 5 wt%, 10 wt%, and 15 wt% was 3.41 emu/g (3.41 Am2/kg), 5.15 emu/g (3.15 Am2/kg), and 9.05 emu/g (9.05 Am2/kg), respectively. All the MGs exhibited a reduced coercive field | The magnetic induction heating was made based on an AMF set-up (power supply, water-cooling system, induction heating system (copper tube resonant frequency: 76 kHz, DC voltage: 53.5 V, maximum current: 28 A), and a solenoid with 8 turns and a diameter of 50 mm. A temperature of 44.4 °C was achieved in 600 s | Rheological parameters: The storage modulus G’ increases directly proportionally with the MNPs percent. The viscosity decreases from 103 Pa·s to 100 Pa·s for a frequency variation between 10−2 to 102 Hz shear rate | Xuan et al. [113] | |
| Hydroxypropyl methyl cellulose (HPMC) and Fe3O4 loaded with doxorubicin (DOX) | pH and magnetic dual-response hydrogel for synergistic chemo-magnetic hyperthermia tumor therapy | In vitro: human umbilical vein endothelial cells (HUVEC) for biosafety evaluation, In vivo: nude mouse 4T1 mouse breast cancer xenograft model—40 female SPF nude mice (weight 20 ± 0.3 g) injected with 0.1 mL of 4T1 cells | - | A custom-built MHT machine (frequency: 400 kHz, output power: 7.2 kW, coil diameter: 10 cm) and an infrared thermal imaging instrument Fortric225 (Fortric Technology, Santa Clara, CA, USA). A temperature higher than 41 °C was obtained for 10% MNPs concentration. To analyze the in vitro drug release, the physiological media PBS exhibiting different pH values (7.4 and 5.5) were chosen. AMF was also applied, and an increase in DOX release of about 57.6% was determined for the pH of 7.4 in PBS at 24 h, while for the pH of 5.5 the cumulative DOX release was estimated at 78.8% in the same conditions | Rheological parameters: The shear-thinning property of the MG proved that during the injection procedure, when the stress strengthened (0 s ÷ 1000 s) the hydrogel becomes thinner (30 ÷ 5 Pa·s shear viscosity). A good injectability of the MG was established | Zhou et al. [114] | |
| Montmorillonite colloidal gel with MNPs loaded with DOX | Postoperative treatment of hepatocellular carcinoma based on minimally invasive MHT | In vitro: NIH3T3 fibroblast cell line derived from mouse embryos In vivo: ICR HepG2 tumor-bearing mice (6 ÷ 8 weeks old), New Zealand rabbit with VX2 liver cancer | - | For MHT the rabbit animal model was exposed to AMF (312 kHz, 30 kA/m) for 15 min based on a heating equipment (Shuangping SPG, Shenzhen, China). The MG exhibited a Ms value of about 50 emu/g. The combined targeted therapy (MHT + chemotherapy) proved to be a very good solution for recurrence control | Rheological parameters: G’ decrease from 1400 Pa to 200 Pa at a temperature increase from 25 °C to 60 °C; the G” value was kept constant at about 100 Pa for the same temperature range | Chen et al. [115] | |
| Xanthan gum with a molecular weight of 2.5 × 106 g/mol with superparamagnetic Fe3O4 MNPs and terbinafine (antifungal drug) incorporated | Thermal-induced control of drug release and T2-MRI contrast increase for possible treatment based on MHT and MRI imaging of skin carcinoma | In vitro cytocompatibility: human dermal fibroblast (HDF), human skin carcinoma (A431) cell lines. Antifungal performance: Candida alibicans (C. albicans) (ATCC 24433) | SQUID-VSM (Quantum Design, San Diego, CA, USA). Hysteresis cycles were measured at 300 K with a magnetic field variation ranging from −20 kOe to +20 kOe (1.6 MA/m). The temperature-dependent magnetization was measured under a magnetic field of 100 Oe in the 1.8 ÷ 300 K temperature range and zero-field-cooled (ZFC) and field-cooled (FC) conditions. Ms was about 2.6 emu/g (2.6 Am2/kg), remanence Mr of 0.12 emu/g (0.12 Am2/kg), coercivity Hc of 0.04 kOe (3.2 kA/m). ZFC and FC measurements confirm a superparamagnetic behavior with a blocking temperature (TB) of 98 K. MHT devices: DM1 calorimeter module from NanoScale Biomagnetics for AMF (890 kHz, 20 mT), temperature measurement sensor made of optical fiber for 15 min MRI measurements: MR Solutions 3.0 Tesla benchtop system | When the AMF (869 kHz, 20 mT) was applied, a SAR value of 100 W/g and an increase in temperature from 10 to 30 °C were determined. The release profile of terbinafine was analyzed under the action of an applied AMF (262.1 kHz, 23 mT) and AMF (174.5 kHz, 23 mT). Due to the local temperature increase around the MNPs under the AMF effect, it forces the relaxation capacity of the polymeric chains, leading to an improved diffusion of the drug. In addition, a drug release behavior depends on the applied field frequency. A higher release was associated with an increased frequency. Regarding the T2-MRI enhancement, it was noticed when the MNPs concentration was increased under the effect of a 3.0 T MRI scanner device | Rheological parameters: after drug incorporation G’ was higher than G” with an almost constant variation independent of frequency (rad/s). The same observation was maintained during the strain variation, which evidenced the solid (gel)-liquid (viscous fluid) transition at 100% strain. The MG viscosity decreased from 100,000 Pa·s (0.001 Hz) to about 1 Pa·s at 1000 Hz, proving a good shear-thinning character | Ribeiro et al. [116] | |
| Self-healing | Chitosan and telechelic difunctional poly(ethylene glycol) (DF-PEG) loaded with Fe3O4 MNPs | Application in drug delivery and image enhancement in cervical cancer therapy | In vitro test: HeLa cervical cancer cels | - | - | Rheological parameters were analyzed to prove the self-healing capacity. The elastic response of MG was analyzed under a strain amplitude sweep test. The critical strain region of G’ and G” was determined at γ = 200% and 100%, respectively. G’ decreases above this critical strain zone, evidencing the collapse of the gel network. After that, the MG was subsequently subjected to a large amplitude oscillatory force (γ = 200%, f = 1.0 Hz), and the G’ value decreased from 400 to 300 Pa, indicating a loose network (tan δ = G’/G” = 0.5 ÷ 0.6). After decreasing the amplitude (γ= 1%, f =1.0 Hz), the G’ recovered quickly to the initial value, and the MG exhibits the original state (tan δ = 0.15 to 0.2). Based on an NdFeB magnet, it was demonstrated that under a static magnetic field, broken MG pieces can be glued together due to MNPs’ intrinsic magnetism, and after the field removal, an integral self-healed gel was obtained. High biocompatibility and cell viability were reported | Zhang et al. [128] |
| Biopolymer containing xanthan gum (XG) crosslinked with chitosan (CS) with modified magnetic Fe3O4@SiO2 | Possible application in cancer therapy | - | - | - | The self-healing property consisted of hydrogel property to withstand heavy counterweights (11.78 g), and after they were applied, a full shape recovery was achieved | Sanoh et al. [125] | |
| Alginate hydrogel platform loaded with Fe3O4 MNPs | Photothermal therapy for colorectal cancer | In vitro: CT26 murine colon cell carcinoma | - | The photothermal properties of MG (200 μg/mL Fe concentration) were investigated. Based on an 808 nm laser at various power densities (0.5, 1.0, and 1.5 W/cm2), MG irradiation was performed for 5 min. In addition, Fe concentration was set at 100, 200, 350, 500 μg/mL and irradiated with an 808 nm laser (1 W/cm2) for 5 min. The laser was turned on/off at every 5 min for 50 min. For the in vitro analysis, cells were seeded together with MG extracts for 12 h. Then they were irradiated with the NIR laser for 5 min. A good photothermal effect was obtained | As future investigation self-healing properties of the proposed MG will be further developed | Ji et al. [129] | |
| Mechanical properties | Fe3O4@Fe-alginate/polyacrylamide (PAAm) | Possible guided catheter used in different magnetic navigation systems | VSM (735 VSM Controller, LakeShore, Westerville, OH, USA) with a maximum magnetic field strength of 9 kOe (716.2 kA/m) at room temperature. By modifying the MNPs percentage between 1 wt% and 20 wt%, the Ms varied from 2 emu/g (2 Am2/kg) to 17 emu/g (17 Am2/kg). A NdFeB permanent magnet was used to control the hydrogel movements | To investigate the MG behavior in external applied fields a sensor with a length of 100 mm and a diameter of 6 mm was moved by a NdFeB magnet. Under constant magnetic field influence, the MG navigated due to the strong magnetic attraction forces and low friction coefficient | Mechanical properties: for the MG with 1.0 and, respectively, 20.0 wt% MNPs were: 11.4 ± 1.5 rupture tensile strain and 0.915 ± 0.053 MPa tensile strength, 2.7 ± 0.4 and 0.201 ± 0.009 MPa, respectively. The MG can recover to its original shape without breaking after 90% compression with a compressive strength of 3.1 ± 0.2 − 5.6 ± 0.6 MPa and ~200 kPa compressive modulus from the same MNPs concentration. The fracture- energy decreases with increasing the MNPs percent 1550.5 ± 194.9 ÷ 2814.0 ± 69.6 Jm2 | Haider et al. [62] | |
| Polyacrylamide with carbonyl iron | Secretions of proangionenic molecules and dynamic control of osteogenesis. Possible application in cancer therapy after tumor surgery to regenerate the operatory site | In vitro: Mesenchymal stem cells (MSCs) | - | Shear rheometry was involved in analyzing the MG’s mechanical properties under magnetic field action | Mechanical properties: 1 min. cycle testing condition: 30 s at 0 T and 30 s at 0.75 T. G′ at 0 T ranged between 0.1 and 0.14 kPa, and at 0.75 T reached 60 ÷ 90 kPa. The gel recovered its elastic modulus at 0 T with each cycle | Abdeen et al. [94] | |
| Poly(N-isopropylacrylamide) (PNIPAm) embedding Fe3O4 MNPs | Application in hyperthermia cancer therapy for melanoma | In vitro: A375 human malignant melanoma cells | Alternating gradient magnetometer (MicroMag TM-2900, Princeton Instruments), commercial induction heating system (Easyheat 224, Cheltenham Induction Heating), IR camera (Fortric 285#L21) | The temperature increases from 11.4 °C to 86.5 °C at 100 s, when the MNPs content in MG varied between 3 wt% ÷ 20 wt%. The saturated magnetization increases from 31 emu/g (31 Am2/kg) to 120 emu/g (120 Am2/kg), for the same concentration of MNPs. The MG can wrap the tumor during mechanical deformation, resulting in a uniform tumor heating process | The shape shifter structure can simultaneously encase and kill cancer cells (through magnetic hyperthermia. About 50% of cancer cells can be destroyed by applying MHT based on MG behavior under mechanical deformation. The link between mechanical deformation and MG shape under AMF action could exhibit an important discovery in cancer therapy | Tang et al. [119] | |
| Porosity | Terephthaloyl thiourea crosslinked chitosan with Fe3O4 MNPs | Application dedicated to MHT | - | VSM analysis. Ms was reported to be about 66.92 emu/g (66.92 Am2/kg). The hyperthermia possible application was performed based on NATSYCO, Iran device | AMF with a frequency of 200 kHz applied for 1 mg/mL sample generated the highest SAR of about 78.43 W/g. The minimum SAR was achieved for a 10 mg/mL sample at a frequency of 350 kHz, corresponding to a value of approximately 7.51 W/g | A porous hydrogel was developed. Fe3O4 MNPs have covered the porous structure of MG. The localization and distribution of MNPs on the surface of MG within the channels are well observed based on scanning electron microscopy | Eivazzadeh-Keihan et al. [120] |
| Silk fibroin loaded with doxorubicin hydrochloride (DOX) and loaded with Fe3O4 MNPs | The ability to release drug ability under different static or alternating magnetic fields for kidney cancer treatment in children | In vitro: Human fibroblast cells isolated from the foreskin by enzymatic digestion and Wilms’ tumor (nephroblastoma cells) | - | At about 35 days, the DOX release from MG was more than twice faster (47.33%) than in the absence of MNPs (21.12%). In addition, the DOX release increased in the presence of AMF | The average size of the pore and the overall porosity have an important influence in addition of MNPs. The total porosity was estimated at 23.59 ± 8.123% with an average pore size of about 188.299 μm2 | Haghighattalab et al. [121] | |
| Macro porous alginate ferrogel microbeads | Drug release possible application for breast cancer treatment | In vitro: 4T1 cells (murine breast cancer cell line used for triple-negative breast cancer analyses) | - | A solid NdFeB magnet with a magnetic induction of 500 mT maintained for 1 min every 10 min showed a burst release of mitoxantrone | The manufacturing of macroporous hydrogels exhibiting an increased porosity and mechanical properties that are maintained during injection process is considered today an important challenge | Shin et al. [122] | |
| Biodegradability/Biocompatibility | Poly(vinyl alcohol) (PVA)/water-soluble tricarboxy cellulose (CO)/Fe3O4 MNPs stabilized with a hydrophilic double layer of oleic acid (OA) molecules and dispersed in distilled water | Potential application for cancer therapy | - | VSM (ADE Technologies VSM880). Measurements were performed at room temperature with an applied field variation of +/−1000 kA/m. Ms was comprised between 0.0539 emu/g (0.0539 Am2/kg) and 0.9147 emu/g (0.9147 Am2/kg) as a function of MNPs quantity. Magnetic permeability determined at the saturation point μsat was between 0.345 memu (0.345 μA/m2) and 7.226 memu (7.226 μA/m2) | - | The biodegradability was evidenced through thermogravimetric analysis. Firstly, the first weight loss level was noticed at 50 ÷ 120 °C and corresponded to water evaporation. Then the second stage characterized by a major weight loss was observed between 220 ÷ 320 °C and corresponded to the structural degradation of PVA and CO/- | Baron et al. [126] |
| Poly (L-lactic acid) (PLLA) microspheres combined with magnetostrictive cobalt ferrites (CoFe2O4) introduced in oil-water emulsion | Analysis of magnetoelectric/magneto-mechanical effect on cell development | In vitro: MC3T3-E1 cells | VSM (MicroSense EZ7, Lowell, MA, USA). Magnetic hysteresis loops and saturation magnetization were determined at room temperature for a field variation between +/−6000 Oe (477.4 kA/m). Ms was estimated at about 3 emu/g (3 Am2/kg) | Cell magnetic stimulation in a special bioreactor (1 Hz frequency and 1 mm amplitude for 16 h followed by non-active time of 8 h) | -/Biocompatibility was investigated by preparing cell culture under magnetic stimulation Magnetic stimulation was characterized by two effects: magnetostrictive variations of the MNPs propagated through the PLLA matrix and directly linked to surface charge variations through the magnetoelectric effect. In this way, it mimics the mechanical stress variations, which occur at the natural human body movements | Carvalho et al. [127] | |
| Iron (III)-alginate MG | Emission of Fe3+ ions under different ionic conditions and with important contribution in triggering the ferroptosis process in cancer therapy | - | VSM (LakeShore 8604, Westerville, OH, USA) with a measurement precision of 10 ÷ 7 emu. The maximum magnetic flux density was chosen to ±2 T. The maximum Ms was about 1.5 emu/g | After placing the MG into an acetic acid solution (pH = 3), the saturation magnetization decreased to 0.08 emu/g (0.08 Am2/kg) | It was concluded that after insertion in an acid, the superparamagnetic behavior is maintained, but the magnetization is greatly decreased in the absence of an applied magnetic field. This application is useful for the analysis of iron ion release with high impact on ROS generation | Chen et al. [130] |
4. Biomedical Applications of Magnetic Hydrogels in Oncology
4.1. Magnetic Hyperthermia
4.2. Drug Delivery
4.3. Immunotherapy
| Cancer Type | Application | Hydrogel/Chemotherapeutic Drug | MNPs/NPs | In Vitro | In Vivo | Properties | Clinical Relevance | Ref. |
|---|---|---|---|---|---|---|---|---|
| Breast cancer | MHT + Chemotherapy | Chitosan/Doxorubicin (DOX) | Ferromagnetic vortex domain iron oxide (FVIOs) (0.6 mg/mL) | L929 and MCF-7 (ER+, HER2+, EGFR+) cell lines | Mouse animal model | In vitro: 10 min. exposure to AMF (495 kHz, 220 Oe (17.507 kA/m)), SAR values 371 (150 Oe (11.936 kA/m)) and 856 (220 Oe (17.507 kA/m)) In vivo: DOX release under AMF effect: The absence of high step release of the drug proved the negligible influence of the AMF irradiation | H × f = 8 × 109 A/(ms) | Gao et al. [63] |
| Two gelatin-based pH- and thermal-responsive (PDMAEMA) or PDMAEMA/PNIPAAm hydrogels/DOX | Fe3O4 | NIH3T3 and MCF-7 cell lines | - | In vitro: At an acidic pH of 4.2, both MGs exhibited a property of “smart” chemotherapeutic effect. When combined with MHT in an acidic medium, an increased drug release was noticed at 42 °C | pH-responsive (4.2) meets the condition of drug delivery at an acidic pH | Derakhshankhah et al. [140] | ||
| Contractible HPMC/DOX | Fe3O4 | HUVEC cell line | Nude mouse 4T1 mouse breast xenograft model | In vitro: AMF (400 kHz, 7.2 kW, coil diameter 10 cm) for 120 s determined an increase in temperature from 25 °C (0% MNPs) to 100 °C (40% MNPs) In vivo: temperature on the surface of the tumor center placed above the injection reached 48 ± 1.2 °C in 1 min. Then MHT expanded to the whole tumor surface, with a temperature value of 68 °C. The AMF enhanced the DOX release with no recurrence of cancer | pH-responsive (5.5) meets the existing recommendations | Zhou et al. [114] | ||
| Poly (N-vinylcaprolactam) (PNVCL)/DOX | Iron oxide MNPs | L929, NIH3T3 mouse embryonic fibroblasts cell lines and MCF-7, MDAMB (ER-, PR-, HER2-) human breast cancer cell lines | - | In vitro: Radio frequency (RF) generator (250 kHz, 450 A) powered a coil of 6 cm diameter for 10 min. The MHT temperature of 43 °C was reached in 600 s in combination with a SAR value of 204 W/g. The cell death occurred in the presence of magnetic nanogel in a percent of 83% (MCF7), 87% (MDAMB232), 62%, and 68% for the healthy fibroblast cell lines, respectively. Under AMF action, the DOX release increased due to the synergistic effect of magnetic field oscillations that disrupted the cell membrane and permitted the DOX release | H × f = 0.11 × 109 A/s per meter unit, pH-responsive (4.5) meets the existing recommendations | Indulekha et al. [142] | ||
| β-cyclodextrin-graft-poly(Nisopropylacrylamide) (β-CD-g-PNIPAAm)/DOX | Fe3O4 | MCF7 | - | In vitro: Ms of MG was about 8.2 emu/g (8.2 Am2/kg). In physiological conditions, it was observed that a low drug release value due to strong hydrogen bonds between the drug molecules and the hydrogel network. By increasing the temperature up to 40 ◦C, the release amount was increased due to collapse of PNIPAAm chains. On the other hand, the release of DOX was slow, and within 24 h, only 33.8% of the total DOX quantity was released | The release rate kinetic meets the existing recommendations | Eskandani et al. [166] | ||
| Poly N-isopropylacrylamide (PNIPAAm) nanosized MG/DOX | Oleic acid and oleyl amine-coated Fe3O4 MNPs | MDAMB231 cell line | - | In vitro: RF (300 kHz, 7 kW, 5 kA/m) absence: 50% cell viability was found when DOX was used, compared to 90% viability in the DOX absence RF applied for 15 min: cell viability was decreased up to 38% | H × f = 1.5 × 109 A/(ms) meets the existing recommendations | Nandwana et al. [165] | ||
| Magnetic supramolecular hydrogel from gellan gum self-assembled by PEGylated MNPs and α—cyclodextrin (α-CDs)/DOX + Paclitaxel (PTX) | PEGylated Fe3O4 | RAW264.4, 4T1 (ER-, PR-, HER2-) cell lines | Female BALB/c mice (4 weeks, 20–25 g) with 4T1 xenografted tumor (2 × 106, in 150 μL serum-free RPMI-1640 medium) | In vitro: the release rate of PTX was low at 2% per day over 15 days, and the MGs released sustainedly only ~30% of PTX for half a month. The release percentage of DOX from the hydrogel was about 60% in the first 2 days. The release was slow after that, and the total release percentage was more than 90% in a month. In AMF (400 kHz, 1.8 kA/m): DOX and PTX release varied between 49 ÷ 63% and 3 ÷ 13% in the first day, respectively; MHT (AMF applied for 80 s), a SAR value of 1334 W/g Fe was achieved In vivo: the cancer cells were constantly killed, and the inflammatory reaction disappeared after 4 weeks. No recurrence was noticed in the main organs of the animals. MG released steadily DOX and PTX for about 2 weeks after implantation. Survival rate at 20 days post-surgery was about 85% | H × f = 0.73 × 109 A/(ms) meets the existing recommendations | Wu et al. [103] | ||
| MHT + Controlled Drug Delivery (Methotrexate (MTX)) | Poly (NIPAM-co-DEAEMA)/Anti-metabolite drug Methotrexate (MTX) | Bare surface Fe3O4 and TMSPMA surface modification Fe3O4 | MCF-7 cell line | - | In vitro: AMF (375 kHz, 4 kA/m) exposure led to 42 °C in 220 s. A SAR value of 83.7 W/g was achieved. Up to 40 wt % of MTX was released at pH 5.5 and 37 °C | H × f = 1.5 × 109 A/(ms), pH-responsive (5.5) meets the existing recommendations | Najafipour et al. [141] | |
| MHT | Nanobiocomposite hydrogel (chitosan, silk fibroin, polyvinyl alcohol) | Fe3O4 | BT549 cell line (Erβ2+, Y5R+, TRPC4+, AR+), HEK293T cell line | - | In vitro: Oscillating magnetic field (100 kHz, 200 kHz, 300 kHz, 400 kHz). SAR values: 21 W/g (100 kHz), 35.72 W/g, 44.65 W/g, and 53.16 W/g (400 kHz) | The maximum SAR value at 400 kHz meets the existing recommendations | Eivazzadeh-Keihan et al. [138] | |
| NIR Hyperthermia + Gene therapy + Drug targeting (Folic acid) | Folate/polyethylenimine-conjugated poly(organophosphazene) polymer, which encapsulates small interfering RNA (siRNA) | Au-Fe3O4 | MDAMB231 cell line | Female BALB/c nude mice (5 weeks old) with xenografted tumor (1 × 107 MDA-MB231 cells) | In vitro: An increased weight loss was noticed within 7 days, and 32% of the hydrogel weight remained at 18 days after injection. The complete dissolution of the hydrogels occurred at 30 days post-injection. The different release rates could be linked to electronic and hydrophobic interactions that bind siRNA and Au-Fe3O4 MNPs In vivo: NIR irradiations (laser power 1.2 W/cm2) were applied in 2 cycles performed at one- and two-weeks post-surgery, and an increase in temperature of about 44 °C in about 5 min was achieved. The best tumor inhibition was reached in the case of two cycles of hyperthermia, which facilitates the release of drugs | Laser power meets the existing recommendations | Zhang et al. [164] | |
| Colon cancer | MHT + Chemotherapy | Medical chitosan/DOX@Ferumoxytol (FMT) | Ferumoxytol commercial MNPs | HT-29 cell line | Male BALB/c nude mice (18 ÷ 27 g weight, 6 weeks old) with a colon xenograft tumor | In vitro: a temperature’s increase up to 42 °C was achieved. In PBS solution (pH of 5.2), a much slower DOX release was noticed at 43 °C compared to the MG behavior at 37 °C In vivo: MHT temperature of about 42 ÷ 43 °C was achieved after 20 min starting from 34 °C (380 kHz, 15 A). A good heating property was noticed only in the tumor region, with no harmful effects on healthy tissue | H × f = 0.05 × 109 A/s per meter unit | Chen et al. [143] |
| NIR + magnet+ enzyme response + Chemotherapy | Thermosensitive poly(N-acryloyl glycinamide) (PNAGA)/DOX and polyester (PE) capped mesoporous silica nanocarriers (MSNs) (PNAGA-DMP-Fe3O4@GO) | Fe3O4 nanoparticles grafted graphene oxide (GO) | LoVo cell line | Tumor-bearing nude mice | In vitro: the magnetothermal efficiency of MG was maintained even after five cycles of turning on and off the magnetic field. A temperature of 55 °C was achieved. NIR light permitted the right manipulation of nanocarriers’ release by changing the laser power. The esterase’s use triggered DOX release over 60% after 24 h incubation in the esterase group In vivo: the mice treated by PNAGA-DMP-Fe3O4@GO MG with NIR irradiation showed the best tumor inhibition with complete remission after 3 weeks | pH-responsive (5) meets the existing recommendations | Yuan et al. [167] | |
| Hepatocellular carcinoma (liver cancer) | MHT + Chemotherapy | Gelatin/DOX | Magnetic zeolite-based imidazolyl skeleton (Fe@ZIF-8) | - | Female BALB/c mice (6 ÷ 8 weeks) with H22 tumor cells subcutaneously injected | In vitro: under AMF (312 kHz, 25 kA/m) influence, the temperature of MG with various Fe@ZIF-8 increased by approximately 24.1, 34.9, and 46.6 °C after 5 min of field irradiation. The DOX@Fe@ZIF-8 was characterized by magnetic heating- accelerated drug release. The ratio of released DOX increased with AMF exposure, up to 42.1% and 33.4%, under pH 6.5 and pH 7.4 In vivo: the temperature at the tumor site reached 42 °C within 10 min of AMF irradiation and gradually degraded in the tumor environment, concomitantly releasing DOX | H × f = 7.8 × 109 A/(ms) | Du et al. [148] |
| Gelatin/DOX | Clinically available montmorillonites and amphoteric gelatin nanoparticles | NIH3T3 cell line | VX2 tumor rabbits | In vitro: Ms of MG loaded with DOX was 55 emu/g (55 Am2/kg). The temperature increased to about 53 °C in 10 min. MG loaded with DOX released a high amount of drug (maximum value of 40%) under AMF, which was considered a double release of drug in the absence of AMF (maximum value of 18%) In vivo: after MHT application for 15 min., tumor tissue was collected and exhibited an important cell apoptosis | AMF (312 kHz, 30 kA/m) applied for 10 min H × f = 9.3 × 109 A/(ms) | Chen et al. [115] | ||
| MHT | Triblock copolymers of poly-((N-isopropylacrylamide-co-dopamine)-b-poly(ethylene-glycol)-b-poly(N-isopropylacrylamide-co-dopamine)) (Poly-(NIPAM-co-DOPA)-PEG-poly(NIPAM-co-DOPA), NDP) | Fe3O4 | - | Subcutaneous liver cancer recurrence mouse model | In vivo: investigations under AMF (312 kHz, 25 kA/m) influence. The temperature of tumor sites rose from 30 to 46 °C within 15 min. This MHT result was considered enough and inhibited tumor growth. In the case of surgically induced recurrence, a reduction to 20% in 14 days was observed under AMF action | H × f = 7.8 × 109 A/(ms) | Yan et al. [144] | |
| Cervical cancer | Targeted drug release chemotherapy | Trangacanth gum (TG), poly(acrylic acid) (PAA) pH-responsive/DOX | Fe3O4 (MH1: 53.26% C, 42.28% O, 4.46% Fe; MH2: 48.57% C, 44.65% O, 6.78% Fe) | HeLa cell line | - | In vitro: negligible drug release (22.3% -MH2 and 19.7%–MH1) in normal physiological conditions (pH 7.4, T 37 °C); in cancerous conditions (pH 5.0), higher drug (64.3%-MH2 and 58.3%-MH1) release was noticed due to interactions between protonated carboxyl groups, PAA segments and TG and protonated DOX Saturation magnetizations of MGs were about 32.5 emu/g (32.5 Am2/kg) for MH2 and 23.9 emu/g (23.9 Am2/kg) for MH1, respectively | pH-responsive (5) meets the existing recommendations | Sayadnia et al. [168] |
| Skin cancer | Targeted drug release chemotherapy under magnetic stimulation | Alginate/DOX | Iron oxide superparamagnetic nanoparticles, gelatin particles | SCC7 cell line | Mice (6 weeks, 20 g) with SCC7 xenografted tumor (5 × 105 cells/mouse) | In vitro: drug release under magnetic stimulation (0.3 T, 0.5 Hz for 120 s, every 30 min): the cumulative DOX release was about 60% within 120 min; the cell reduction was achieved for chemotherapy applied under magnetic field simulation In vivo: tumor growth was highly reduced in mice treated with MG under magnetic stimulation (0.3 T, 0.5 Hz for 2 min, one application per day). No important weight loss was observed | Magnetic stimulation’s importance | Kim et al. [169] |
| Immunotherapy (PD-1) + MHT | Gelatinic–tannic acid-based gel/checkpoint inhibitor PD-1 | Bimetallic magnetic nanoarchitectonics (Zn0.35Fe0.65) Fe2O4 | B16F10 cell line | Female C57 mice with a B16F10 xenografted tumor (5 × 106 cells/mouse) injected into the flank region | In vitro: absence of AMF (309 kHz, 40 kA/m): 90% viability, under AMF action: 60% viability. Analysis performed based on the CCK-8 assay In vivo: the inhibitory effect of the MG was analyzed on distant tumors and pulmonary metastases. A negative Ki-67 staining was achieved under AMF application, which proved the treatment efficiency obtained through the synergistic effect of pyrolysis and immunotherapy compared to the mice group that was treated only with MG, or MG loaded with PD-1 in the absence of AMF | H × f = 1.2 × 1010 A/(ms) | Wang et al. [182] | |
| Immunomodulator effect | Hyaluronic acid with borosilicate nanoparticles | Fe3O4 | B16F10 cell line | Mouse animal model with B16F10 xeno-grafted tumor | In vitro: at the maximum concentration of MNPs (250 μg/mL), a reduced cell viability of less than 10% occurred. MG were incubated with interleukin 4 (IL-4)-pretreated M2 macrophages. An enhancement of M1 macrophages number concomitantly with an important decrease in M2 macrophages population was noticed In vivo: the MG group exhibited the lowest tumor fluorescence intensity and the smallest tumor volume. The mice’s body weight was maintained 10 days after surgery, indicating no signs of toxicity. The histopathological analysis revealed an increase in M1 macrophages compared to the M2 state | Macrophage state modulation | Quan et al. [183] | |
| Lung cancer | Magnetic field-driven chemotherapy | Salecan-g-poly(vinylacetic acid-co-2-hydroxyethyl acrylate) [poly(VA-co-HEA)] copolymer/DOX | Fe3O4@Agarose | COS-7 and A549 cell lines | - | In vitro: DOX release was investigated under different pH values (pH of 7.4, 6.8, and 4.5 with/without a magnetic field (2200 G (0.22 T))) for endosomal and lysosomal microenvironment analysis. Ms of MG was about 19.8 emu/g (19.8 Am2/kg) at 15 kOe (1.2 MA/m). The DOX release was higher at pH 4.5 than at the others. The cumulative release of DOX could reach 76% after 64 h at acidic pH compared to normal tissue pH (pH 7.4—16.9%; pH 6.8–10.5%) | pH-responsive (4.5) meets the existing recommendations | Hu et al. [170] |
| Bone cancer | Remotely stimulated drug release under a low-frequency alternating magnetic field (LAMF) | Chitosan loaded with DOX (commercial name Adriamycin) and rifampicin (RFP) | Iron oxide MNPs | MG-63 cell line | - | In vitro: absence of LAMF: about 40% percent of DOX was released within 2 h, and after 4 h a continuous drug release was noticed. Regarding RFP, almost 60% percent of RFP was released within 60 h and the RFP emission was observed even after 100 h LAMF (60 Hz, 15 min ON, 15 min OFF, 1 h): LAMF for 1 h, the cumulative release percentage of DOX increased by 67.2% (from 37.5% to 62.6%); LAMF accelerated the RFP release | LAMF application importance | Wang et al. [76] |
| Head and neck cancer | Targeting delivery and multi-stimuli release of chemotherapeutic drug | DNA nanogel made via rolling circle amplification, an enzymatic polymerization of the DNA chain, and in situ coating of the MNPs based on physical crosslinking procedure/DOX | Amino-modified Fe3O4 nanoparticle (Fe3O4@SiO2-NH2) used as a core with a shell of DNA nanogel | U87MG cell line | - | In vitro: DOX, as a cationic molecule, can be linked to negatively charged DNA molecules. Firstly, the drug release of the magnetic DNA@DOX nanogel was tested at various temperatures and physiological pH levels. The release rate increased directly proportional to temperature and was 13% (25 °C), 25% (37 °C), and 59%(50 °C). Secondly, the pH influence was investigated. The release amount of DOX at 37 °C within 5 h was 25% (pH of 7.4), 31% (pH of 6.0), 38% (pH of 5.0), respectively. Thirdly, the presence of the nuclease was analyzed. The release amount of DOX at 37 °C (5 h) increased directly proportionally with DNase I concentration, up to 68% at 560 U/mL DNase I. The U87MG cells internalized the developed magnetic DNA nanogel | pH-responsive (5) meets the existing recommendations | Yao et al. [171] |
| Kidney cancer (nephroblastoma) | Targeted drug release | Silk fibroin/DOX | Fe3O4 (25 μg of powder) | Laboratory-developed human fibroblast cells (isolated from the foreskin) and Wilms’ tumor cells | - | In vitro: absence of electromagnetic field application: the DOX release profile was in a direct relationship with viscosity and porosity. At lower MG viscosity, the drug moved faster. Regarding the porosity, adding the MNPs increased the porosity, which facilitated the DOX release Electromagnetic field application: AMF (5 mT), static magnetic fields (SMF1 0.18 T and SMF2 0.28 T): reduced DOX release compared to EMF absence: the release of DOX from MG decreased with increasing the SMF magnetic flux density value. DOX release was diminished in the presence of AMF compared with SMF1 and SMF2 applications | Comparison between the effects of AMF and SMF application | Haghighattalab et al. [121] |
| Potential large B-cell lymphoma | MHT + Chemodynamic therapy (Pt) + Immunomodulation | Poly(lactic-co-glycolic) PLGA-polyethylene glycol (PEG)-PLGA | Fe3O4@Dimercaptosuccinic acid (DMSA)@Platinum (Pt) | A10 cell line | - | In vitro: Flow cytometry revealed an initial increase in ROS levels at 12 h, followed by a reduction at 48 h. This phenomenon was accompanied by a low cell viability and a high cell apoptosis | ROS importance, H × f = 3.5 × 109 A/(ms) meets the existing recommendations | Yan et al. [184] |
4.4. Magnetic Hydrogel Toxicity
5. Conclusions, Challenges, Future Trends, and Limitations
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Global Cancer Facts & Figures. Available online: https://www.cancer.org/research/cancer-facts-statistics/global-cancer-facts-and-figures.html (accessed on 12 July 2025).
- Global Cancer Burden Growing, Amidst Mounting Need for Services. Available online: https://www.who.int/news/item/01-02-2024-global-cancer-burden-growing--amidst-mounting-need-for-services (accessed on 12 July 2025).
- Cojocaru, F.D.; Balan, V.; Popa, I.M.; Munteanu, A.; Anghelache, A.; Verestiuc, L. Magnetic Composite Scaffolds for Potential Applications in Radiochemotherapy of Malignant Bone Tumors. Medicina 2019, 55, 153. [Google Scholar] [CrossRef]
- Taciuc, I.-A.; Dumitru, M.; Marinescu, A.; Serboiu, C.; Musat, G.; Gherghe, M.; Costache, A.; Vrinceanu, D. Enhancing Malignant Lymph Node Detection in Ultrasound Imaging: A Comparison Between the Artificial Intelligence Accuracy, Dice Similarity Coefficient and Intersection over Union. J. Mind Med. Sci. 2025, 12, 29. [Google Scholar] [CrossRef]
- Ionescu, S.; Marincas, M.; Madge, O.L.; Dicu-Andreescu, I.G.; Chitoran, E.; Rotaru, V.; Cirimbei, C.; Gherghe, M.; Ene, A.; Rosca, R.; et al. Ovarian Causes of Pseudomyxoma Peritonei (PMP)—A Literature Review. Cancers 2024, 16, 1446. [Google Scholar] [CrossRef]
- Ciongariu, A.-M.; Țăpoi, D.A.; Dumitru, A.-V.; Enache, V.; Marin, A.; Creangă Snr, C.A.; Costache, M. Enhancing Liposarcoma Prognosis—A New Predictive Scoring System Integrating Histopathological Insights. Cancer Manag. Res. 2025, 17, 331–348. [Google Scholar] [CrossRef]
- Lișcu, H.-D.; Verga, N.; Atasiei, D.-I.; Badiu, D.-C.; Dumitru, A.V.; Ultimescu, F.; Pavel, C.; Stefan, R.-E.; Manole, D.-C.; Ionescu, A.-I. Biomarkers in Colorectal Cancer: Actual and Future Perspectives. Int. J. Mol. Sci. 2024, 25, 11535. [Google Scholar] [CrossRef] [PubMed]
- Onisâi, M.; Vlădăreanu, A.-M.; Iordan, I.; Bumbea, H.; Aliuș, C.; Grădinaru, S.; Bohiltea, R.; Carstoiu, M. Pregnancy-associated gastric cancer: Real-life diagnosis and management. Pharmacophore 2020, 11, 89–92. [Google Scholar]
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
- Șerban, B.; Cursaru, A.; Iordache, S.; Costache, M.; Cretu, B.; Dumitru, A.; Cirstoiu, C. Soft-Tissue Sarcomas—A Correlation Among Tumor Margin Infiltration, Immunological Markers, and Survival Rate. Int. J. Mol. Sci. 2025, 26, 4363. [Google Scholar] [CrossRef] [PubMed]
- Phung, D.C.; Nguyen, H.T.; Phuong Tran, T.T.; Jin, S.G.; Yong, C.S.; Truong, D.H.; Tran, T.H.; Kim, J.O. Combined hyperthermia and chemotherapy as a synergistic anticancer treatment. J. Pharm. Investig. 2019, 49, 519–526. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
- Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef]
- Mihăilescu, A.-A.; Onisâi, M.; Alexandru, A.; Teodorescu, M.; Aliuș, C.; Blendea, C.-D.; Neagu, Ș.-I.; Șerban, D.; Grădinaru, S. A Comparative Analysis between Enhanced Recovery after Surgery and Traditional Care in the Management of Obstructive Colorectal Cancer. Medicina 2024, 60, 1319. [Google Scholar] [CrossRef] [PubMed]
- Shevchenko, I.; Grigorescu, C.C.; Serban, D.; Cristea, B.M.; Simion, L.; Gherghiceanu, F.; Costea, A.C.; Dumitrescu, D.; Alius, C.; Tudor, C.; et al. The Value of Systemic Inflammatory Indices for Predicting Early Postoperative Complications in Colorectal Cancer. Medicina 2024, 60, 1481. [Google Scholar] [CrossRef]
- Zhi, D.; Yang, T.; Yang, J.; Fu, S.; Zhang, S. Targeting strategies for superparamagnetic iron oxide nanoparticles in cancer therapy. Acta Biomater. 2020, 102, 13–34. [Google Scholar] [CrossRef] [PubMed]
- Manescu Paltanea, V.; Paltanea, G.; Antoniac, I.; Vasilescu, M. Magnetic Nanoparticles Used in Oncology. Materials 2021, 14, 5948. [Google Scholar] [CrossRef]
- Paltanea, G.; Manescu (Paltanea), V.; Antoniac, I.; Antoniac, A.; Nemoianu, I.V.; Robu, A.; Dura, H. A Review of Biomimetic and Biodegradable Magnetic Scaffolds for Bone Tissue Engineering and Oncology. Int. J. Mol. Sci. 2023, 24, 4312. [Google Scholar] [CrossRef]
- Xiong, L.; Liang, B.; Yu, K. Magnetic hyperthermia in oncology: Nanomaterials-driven combinatorial strategies for synergistic therapeutic gains. Mater. Today Bio 2025, 33, 102070. [Google Scholar] [CrossRef]
- Toader, C.; Dumitru, A.V.; Eva, L.; Serban, M.; Covache-Busuioc, R.-A.; Ciurea, A.V. Nanoparticle Strategies for Treating CNS Disorders: A Comprehensive Review of Drug Delivery and Theranostic Applications. Int. J. Mol. Sci. 2024, 25, 13302. [Google Scholar] [CrossRef] [PubMed]
- Du, Y.; Liu, X.; Liang, Q.; Liang, X.-J.; Tian, J. Optimization and Design of Magnetic Ferrite Nanoparticles with Uniform Tumor Distribution for Highly Sensitive MRI/MPI Performance and Improved Magnetic Hyperthermia Therapy. Nano Lett. 2019, 19, 3618–3626. [Google Scholar] [CrossRef]
- Şerboiu, C.S.; Aliuș, C.; Dumitru, A.; Țăpoi, D.; Costache, M.; Nica, A.E.; Mihăilescu, A.-A.; Antoniac, I.; Grădinaru, S. Gallbladder Pancreatic Heterotopia—The Importance of Diagnostic Imaging in Managing Intraoperative Findings. Medicina 2023, 59, 1407. [Google Scholar] [CrossRef]
- Beik, J.; Asadi, M.; Khoei, S.; Laurent, S.; Abed, Z.; Mirrahimi, M.; Farashahi, A.; Hashemian, R.; Ghaznavi, H.; Shakeri-Zadeh, A. Simulation-guided photothermal therapy using MRI-traceable iron oxide-gold nanoparticle. J. Photochem. Photobiol. B Biol. 2019, 199, 111599. [Google Scholar] [CrossRef]
- Cheng, H.-W.; Tsao, H.-Y.; Chiang, C.-S.; Chen, S.-Y. Advances in Magnetic Nanoparticle-Mediated Cancer Immune-Theranostics. Adv. Healthc. Mater. 2021, 10, 2001451. [Google Scholar] [CrossRef]
- Manescu Paltanea, V.; Antoniac, I.; Paltanea, G.; Nemoianu, I.V.; Mohan, A.G.; Antoniac, A.; Rau, J.V.; Laptoiu, S.A.; Mihai, P.; Gavrila, H.; et al. Magnetic Hyperthermia in Glioblastoma Multiforme Treatment. Int. J. Mol. Sci. 2024, 25, 10065. [Google Scholar] [CrossRef]
- Gavilán, H.; Avugadda, S.K.; Fernández-Cabada, T.; Soni, N.; Cassani, M.; Mai, B.T.; Chantrell, R.; Pellegrino, T. Magnetic nanoparticles and clusters for magnetic hyperthermia: Optimizing their heat performance and developing combinatorial therapies to tackle cancer. Chem. Soc. Rev. 2021, 50, 11614–11667. [Google Scholar] [CrossRef] [PubMed]
- Gavilán, H.; Simeonidis, K.; Myrovali, E.; Mazarío, E.; Chubykalo-Fesenko, O.; Chantrell, R.; Balcells, L.; Angelakeris, M.; Morales, M.P.; Serantes, D. How size, shape and assembly of magnetic nanoparticles give rise to different hyperthermia scenarios. Nanoscale 2021, 13, 15631–15646. [Google Scholar] [CrossRef]
- Soetaert, F.; Korangath, P.; Serantes, D.; Fiering, S.; Ivkov, R. Cancer therapy with iron oxide nanoparticles: Agents of thermal and immune therapies. Adv. Drug Deliv. Rev. 2020, 163–164, 65–83. [Google Scholar] [CrossRef] [PubMed]
- Oprea, M.; Pandele, A.M.; Nechifor, A.C.; Nicoara, A.I.; Antoniac, I.V.; Semenescu, A.; Voicu, S.I.; Enachescu, C.I.; Fratila, A.M. Improved Biomineralization Using Cellulose Acetate/Magnetic Nanoparticles Composite Membranes. Polymers 2025, 17, 209. [Google Scholar] [CrossRef]
- Yanase, M.; Shinkai, M.; Honda, H.; Wakabayashi, T.; Yoshida, J.; Kobayashi, T. Intracellular Hyperthermia for Cancer Using Magnetite Cationic Liposomes: An in vivo Study. Jpn. J. Cancer Res. 1998, 89, 463–470. [Google Scholar] [CrossRef]
- Alphandéry, E.; Idbaih, A.; Adam, C.; Delattre, J.-Y.; Schmitt, C.; Guyot, F.; Chebbi, I. Chains of magnetosomes with controlled endotoxin release and partial tumor occupation induce full destruction of intracranial U87-Luc glioma in mice under the application of an alternating magnetic field. J. Control. Release 2017, 262, 259–272. [Google Scholar] [CrossRef]
- Mamiya, H. Recent Advances in Understanding Magnetic Nanoparticles in AC Magnetic Fields and Optimal Design for Targeted Hyperthermia. J. Nanomater. 2013, 2013, 752973. [Google Scholar] [CrossRef]
- Hergt, R.; Dutz, S.; Müller, R.; Zeisberger, M. Magnetic particle hyperthermia: Nanoparticle magnetism and materials development for cancer therapy. J. Phys. Condens. Matter 2006, 18, S2919. [Google Scholar] [CrossRef]
- Thiesen, B.; Jordan, A. Clinical applications of magnetic nanoparticles for hyperthermia. Int. J. Hyperth. 2008, 24, 467–474. [Google Scholar] [CrossRef] [PubMed]
- Frachini, E.; Petri, D. Magneto-Responsive Hydrogels: Preparation, Characterization, Biotechnological and Environmental Applications. J. Braz. Chem. Soc. 2019, 30, 2010–2028. [Google Scholar] [CrossRef]
- Xue, L.; Sun, J. Magnetic hydrogels with ordered structure for biomedical applications. Front. Chem. 2022, 10, 1040492. [Google Scholar] [CrossRef]
- Abou-Okeil, A.; Refaei, R.; Moustafa, S.E.; Ibrahim, H.M. Magnetic hydrogel scaffold based on hyaluronic acid/chitosan and gelatin natural polymers. Sci. Rep. 2024, 14, 28206. [Google Scholar] [CrossRef]
- Londhe, P.V.; Londhe, M.V.; Salunkhe, A.B.; Laha, S.S.; Mefford, O.T.; Thorat, N.D.; Khot, V.M. Magnetic hydrogel (MagGel): An evolutionary pedestal for anticancer therapy. Coord. Chem. Rev. 2025, 522, 216228. [Google Scholar] [CrossRef]
- Cao, H.; Duan, L.; Zhang, Y.; Cao, J.; Zhang, K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct. Target. Ther. 2021, 6, 426. [Google Scholar] [CrossRef] [PubMed]
- Correa, S.; Grosskopf, A.K.; Lopez Hernandez, H.; Chan, D.; Yu, A.C.; Stapleton, L.M.; Appel, E.A. Translational Applications of Hydrogels. Chem. Rev. 2021, 121, 11385–11457. [Google Scholar] [CrossRef]
- Yu, F.; Yang, P.; Yang, Z.; Zhang, X.; Ma, J. Double-network hydrogel adsorbents for environmental applications. Chem. Eng. J. 2021, 426, 131900. [Google Scholar] [CrossRef]
- Iulian, A.; Dan, L.; Camelia, T.; Claudia, M.; Sebastian, G. Synthetic Materials for Osteochondral Tissue Engineering. Adv. Exp. Med. Biol. 2018, 1058, 31–52. [Google Scholar] [CrossRef]
- Huang, S.; Hong, X.; Zhao, M.; Liu, N.; Liu, H.; Zhao, J.; Shao, L.; Xue, W.; Zhang, H.; Zhu, P.; et al. Nanocomposite hydrogels for biomedical applications. Bioeng. Transl. Med. 2022, 7, e10315. [Google Scholar] [CrossRef]
- Messing, R.; Frickel, N.; Belkoura, L.; Strey, R.; Rahn, H.; Odenbach, S.; Schmidt, A.M. Cobalt Ferrite Nanoparticles as Multifunctional Cross-Linkers in PAAm Ferrohydrogels. Macromolecules 2011, 44, 2990–2999. [Google Scholar] [CrossRef]
- Hermenegildo, B.; Ribeiro, C.; Pérez-Álvarez, L.; Vilas, J.L.; Learmonth, D.A.; Sousa, R.A.; Martins, P.; Lanceros-Méndez, S. Hydrogel-based magnetoelectric microenvironments for tissue stimulation. Colloids Surf. B Biointerfaces 2019, 181, 1041–1047. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Wang, C.; Gao, Q.; Liu, X.; Tong, Z. Magnetic hydrogels with supracolloidal structures prepared by suspension polymerization stabilized by Fe2O3 nanoparticles. Acta Biomater. 2010, 6, 275–281. [Google Scholar] [CrossRef]
- Sapir, Y.; Cohen, S.; Friedman, G.; Polyak, B. The promotion of in vitro vessel-like organization of endothelial cells in magnetically responsive alginate scaffolds. Biomaterials 2012, 33, 4100–4109. [Google Scholar] [CrossRef]
- Thoniyot, P.; Tan, M.J.; Karim, A.A.; Young, D.J.; Loh, X.J. Nanoparticle–Hydrogel Composites: Concept, Design, and Applications of These Promising, Multi-Functional Materials. Adv. Sci. 2015, 2, 1400010. [Google Scholar] [CrossRef]
- Gil, S.; Mano, J.F. Magnetic composite biomaterials for tissue engineering. Biomater. Sci. 2014, 2, 812–818. [Google Scholar] [CrossRef]
- Hu, W.; Wang, Z.; Xiao, Y.; Zhang, S.; Wang, J. Advances in crosslinking strategies of biomedical hydrogels. Biomater. Sci. 2019, 7, 843–855. [Google Scholar] [CrossRef] [PubMed]
- Saini, K. Preparation method, Properties and Crosslinking of hydrogel: A review. Pharma Tutor 2017, 5, 27–36. [Google Scholar]
- Li, Y.; Huang, G.; Zhang, X.; Li, B.; Chen, Y.; Lu, T.; Lu, T.J.; Xu, F. Magnetic Hydrogels and Their Potential Biomedical Applications. Adv. Funct. Mater. 2013, 23, 660–672. [Google Scholar] [CrossRef]
- Liu, Z.; Liu, J.; Cui, X.; Wang, X.; Zhang, L.; Tang, P. Recent Advances on Magnetic Sensitive Hydrogels in Tissue Engineering. Front. Chem. 2020, 8, 124. [Google Scholar] [CrossRef]
- Xue, X.; Hu, Y.; Wang, S.; Chen, X.; Jiang, Y.; Su, J. Fabrication of physical and chemical crosslinked hydrogels for bone tissue engineering. Bioact. Mater. 2022, 12, 327–339. [Google Scholar] [CrossRef]
- Rodkate, N.; Rutnakornpituk, M. Multi-responsive magnetic microsphere of poly(N-isopropylacrylamide)/carboxymethylchitosan hydrogel for drug controlled release. Carbohydr. Polym. 2016, 151, 251–259. [Google Scholar] [CrossRef]
- Wang, W.; Zhang, Y.; Liu, W. Bioinspired fabrication of high strength hydrogels from non-covalent interactions. Prog. Polym. Sci. 2017, 71, 1–25. [Google Scholar] [CrossRef]
- Wang, W.; Zhang, H.; Shen, J.; Ye, M. Facile preparation of magnetic chitosan/poly (vinyl alcohol) hydrogel beads with excellent adsorption ability via freezing-thawing method. Colloids Surf. A Physicochem. Eng. Asp. 2018, 553, 672–680. [Google Scholar] [CrossRef]
- Huang, J.; Liang, Y.; Jia, Z.; Chen, J.; Duan, L.; Liu, W.; Zhu, F.; Liang, Q.; Zhu, W.; You, W.; et al. Development of Magnetic Nanocomposite Hydrogel with Potential Cartilage Tissue Engineering. ACS Omega 2018, 3, 6182–6189. [Google Scholar] [CrossRef]
- Wang, Y.; Zhu, Y.; Xue, Y.; Wang, J.; Li, X.; Wu, X.; Qin, Y.; Chen, W. Sequential in-situ route to synthesize novel composite hydrogels with excellent mechanical, conductive, and magnetic responsive properties. Mater. Des. 2020, 193, 108759. [Google Scholar] [CrossRef]
- Stancu, I.-C.; Dragusin, D.M.; Vasile, E.; Trusca, R.; Antoniac, I.; Vasilescu, D.S. Porous calcium alginate-gelatin interpenetrated matrix and its biomineralization potential. J. Mater. Sci. Mater. Med. 2011, 22, 451–460. [Google Scholar] [CrossRef] [PubMed]
- Zhao, W.; Odelius, K.; Edlund, U.; Zhao, C.; Albertsson, A.-C. In Situ Synthesis of Magnetic Field-Responsive Hemicellulose Hydrogels for Drug Delivery. Biomacromolecules 2015, 16, 2522–2528. [Google Scholar] [CrossRef]
- Haider, H.; Yang, C.H.; Zheng, W.J.; Yang, J.H.; Wang, M.X.; Yang, S.; Zrínyi, M.; Osada, Y.; Suo, Z.; Zhang, Q.; et al. Exceptionally tough and notch-insensitive magnetic hydrogels. Soft Matter 2015, 11, 8253–8261. [Google Scholar] [CrossRef] [PubMed]
- Gao, F.; Xie, W.; Miao, Y.; Wang, D.; Guo, Z.; Ghosal, A.; Li, Y.; Wei, Y.; Feng, S.-S.; Zhao, L.; et al. Magnetic Hydrogel with Optimally Adaptive Functions for Breast Cancer Recurrence Prevention. Adv. Healthc. Mater. 2019, 8, 1900203. [Google Scholar] [CrossRef]
- Li, Z.; Li, Y.; Chen, C.; Cheng, Y. Magnetic-responsive hydrogels: From strategic design to biomedical applications. J. Control. Release 2021, 335, 541–556. [Google Scholar] [CrossRef]
- Mahdavinia, G.R.; Mousanezhad, S.; Hosseinzadeh, H.; Darvishi, F.; Sabzi, M. Magnetic hydrogel beads based on PVA/sodium alginate/laponite RD and studying their BSA adsorption. Carbohydr. Polym. 2016, 147, 379–391. [Google Scholar] [CrossRef]
- Shi, Y.; Li, Y.; Coradin, T. Magnetically-oriented type I collagen-SiO2@Fe3O4 rods composite hydrogels tuning skin cell growth. Colloids Surf. B Biointerfaces 2020, 185, 110597. [Google Scholar] [CrossRef]
- Fan, T.; Li, M.; Wu, X.; Li, M.; Wu, Y. Preparation of thermoresponsive and pH-sensitivity polymer magnetic hydrogel nanospheres as anticancer drug carriers. Colloids Surf. B Biointerfaces 2011, 88, 593–600. [Google Scholar] [CrossRef]
- Cao, Z.; Wang, D.; Li, Y.; Xie, W.; Wang, X.; Tao, L.; Wei, Y.; Wang, X.; Zhao, L. Effect of nanoheat stimulation mediated by magnetic nanocomposite hydrogel on the osteogenic differentiation of mesenchymal stem cells. Sci. China Life Sci. 2018, 61, 448–456. [Google Scholar] [CrossRef] [PubMed]
- Antman-Passig, M.; Shefi, O. Remote Magnetic Orientation of 3D Collagen Hydrogels for Directed Neuronal Regeneration. Nano Lett. 2016, 16, 2567–2573. [Google Scholar] [CrossRef]
- Betsch, M.; Cristian, C.; Lin, Y.-Y.; Blaeser, A.; Schöneberg, J.; Vogt, M.; Buhl, E.M.; Fischer, H.; Duarte Campos, D.F. Incorporating 4D into Bioprinting: Real-Time Magnetically Directed Collagen Fiber Alignment for Generating Complex Multilayered Tissues. Adv. Healthc. Mater. 2018, 7, 1800894. [Google Scholar] [CrossRef] [PubMed]
- Micromod Partikeltechnologie GmbH. Available online: https://micromod.de/en/ (accessed on 8 September 2025).
- Brady, M.A.; Talvard, L.; Vella, A.; Ethier, C.R. Bio-inspired design of a magnetically active trilayered scaffold for cartilage tissue engineering. J. Tissue Eng. Regen. Med. 2017, 11, 1298–1302. [Google Scholar] [CrossRef]
- Tang, J.; Qiao, Y.; Chu, Y.; Tong, Z.; Zhou, Y.; Zhang, W.; Xie, S.; Hu, J.; Wang, T. Magnetic double-network hydrogels for tissue hyperthermia and drug release. J. Mater. Chem. B 2019, 7, 1311–1321. [Google Scholar] [CrossRef] [PubMed]
- Gao, Y.; Wei, Z.; Li, F.; Yang, Z.M.; Chen, Y.M.; Zrinyi, M.; Osada, Y. Synthesis of a morphology controllable Fe3O4 nanoparticle/hydrogel magnetic nanocomposite inspired by magnetotactic bacteria and its application in H2O2 detection. Green Chem. 2014, 16, 1255–1261. [Google Scholar] [CrossRef]
- Gao, Y.; Hu, C.; Zheng, W.J.; Yang, S.; Li, F.; Sun, S.D.; Zrínyi, M.; Osada, Y.; Yang, Z.M.; Chen, Y.M. Fe3O4 Anisotropic Nanostructures in Hydrogels: Efficient Catalysts for the Rapid Removal of Organic Dyes from Wastewater. ChemPhysChem 2016, 17, 1999–2007. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Li, B.; Xu, F.; Han, Z.; Wei, D.; Jia, D.; Zhou, Y. Tough Magnetic Chitosan Hydrogel Nanocomposites for Remotely Stimulated Drug Release. Biomacromolecules 2018, 19, 3351–3360. [Google Scholar] [CrossRef]
- Filippi, M.; Dasen, B.; Guerrero, J.; Garello, F.; Isu, G.; Born, G.; Ehrbar, M.; Martin, I.; Scherberich, A. Magnetic nanocomposite hydrogels and static magnetic field stimulate the osteoblastic and vasculogenic profile of adipose-derived cells. Biomaterials 2019, 223, 119468. [Google Scholar] [CrossRef]
- Liu, K.; Han, L.; Tang, P.; Yang, K.; Gan, D.; Wang, X.; Wang, K.; Ren, F.; Fang, L.; Xu, Y.; et al. An Anisotropic Hydrogel Based on Mussel-Inspired Conductive Ferrofluid Composed of Electromagnetic Nanohybrids. Nano Lett. 2019, 19, 8343–8356. [Google Scholar] [CrossRef]
- Cezar, C.A.; Roche, E.T.; Vandenburgh, H.H.; Duda, G.N.; Walsh, C.J.; Mooney, D.J. Biologic-free mechanically induced muscle regeneration. Proc. Natl. Acad. Sci. USA 2016, 113, 1534–1539. [Google Scholar] [CrossRef]
- Rose, J.C.; Cámara-Torres, M.; Rahimi, K.; Köhler, J.; Möller, M.; De Laporte, L. Nerve Cells Decide to Orient inside an Injectable Hydrogel with Minimal Structural Guidance. Nano Lett. 2017, 17, 3782–3791. [Google Scholar] [CrossRef]
- Arias, S.L.; Shetty, A.; Devorkin, J.; Allain, J.-P. Magnetic targeting of smooth muscle cells in vitro using a magnetic bacterial cellulose to improve cell retention in tissue-engineering vascular grafts. Acta Biomater. 2018, 77, 172–181. [Google Scholar] [CrossRef]
- Bimendra Gunatilake, U.; Venkatesan, M.; Basabe-Desmonts, L.; Benito-Lopez, F. Ex situ and in situ Magnetic Phase Synthesised Magneto-Driven Alginate Beads. J. Colloid Interface Sci. 2022, 610, 741–750. [Google Scholar] [CrossRef] [PubMed]
- Ahmadi, M.; Monji, D.; Taromi, F.A. Bio-inspired surface modification of iron oxide nanoparticles for active stabilization in hydrogels. Soft Matter 2021, 17, 955–964. [Google Scholar] [CrossRef]
- Chen, X.; Fan, M.; Tan, H.; Ren, B.; Yuan, G.; Jia, Y.; Li, J.; Xiong, D.; Xing, X.; Niu, X.; et al. Magnetic and self-healing chitosan-alginate hydrogel encapsulated gelatin microspheres via covalent cross-linking for drug delivery. Mater. Sci. Eng. C 2019, 101, 619–629. [Google Scholar] [CrossRef]
- Su, H.; Han, X.; He, L.; Deng, L.; Yu, K.; Jiang, H.; Wu, C.; Jia, Q.; Shan, S. Synthesis and characterization of magnetic dextran nanogel doped with iron oxide nanoparticles as magnetic resonance imaging probe. Int. J. Biol. Macromol. 2019, 128, 768–774. [Google Scholar] [CrossRef]
- Hu, X.; Nian, G.; Liang, X.; Wu, L.; Yin, T.; Lu, H.; Qu, S.; Yang, W. Adhesive Tough Magnetic Hydrogels with High Fe3O4 Content. ACS Appl. Mater. Interfaces 2019, 11, 10292–10300. [Google Scholar] [CrossRef]
- Wang, Y.; Zhong, M.; Wang, L.; Liu, Y.; Wang, B.; Li, Y. Chelerythrine loaded composite magnetic thermosensitive hydrogels as a novel anticancer drug-delivery system. J. Drug Deliv. Sci. Technol. 2019, 54, 101293. [Google Scholar] [CrossRef]
- Xie, W.; Gao, Q.; Guo, Z.; Wang, D.; Gao, F.; Wang, X.; Wei, Y.; Zhao, L. Injectable and Self-Healing Thermosensitive Magnetic Hydrogel for Asynchronous Control Release of Doxorubicin and Docetaxel to Treat Triple-Negative Breast Cancer. ACS Appl. Mater. Interfaces 2017, 9, 33660–33673. [Google Scholar] [CrossRef] [PubMed]
- Tay, A.; Sohrabi, A.; Poole, K.; Seidlits, S.; Di Carlo, D. A 3D Magnetic Hyaluronic Acid Hydrogel for Magnetomechanical Neuromodulation of Primary Dorsal Root Ganglion Neurons. Adv. Mater. 2018, 30, e1800927. [Google Scholar] [CrossRef] [PubMed]
- Shi, L.; Zeng, Y.; Zhao, Y.; Yang, B.; Ossipov, D.; Tai, C.-W.; Dai, J.; Xu, C. Biocompatible Injectable Magnetic Hydrogel Formed by Dynamic Coordination Network. ACS Appl. Mater. Interfaces 2019, 11, 46233–46240. [Google Scholar] [CrossRef]
- Gang, F.; Yan, H.; Ma, C.; Jiang, L.; Gu, Y.; Liu, Z.; Zhao, L.; Wang, X.; Zhang, J.; Sun, X. Robust magnetic double-network hydrogels with self-healing, MR imaging, cytocompatibility and 3D printability. Chem. Commun. 2019, 55, 9801–9804. [Google Scholar] [CrossRef]
- Ghazy, O.; Hamed, M.G.; Breky, M.; Borai, E.H. Synthesis of magnetic nanoparticles-containing nanocomposite hydrogel and its potential application for simulated radioactive wastewater treatment. Colloids Surf. A Physicochem. Eng. Asp. 2021, 621, 126613. [Google Scholar] [CrossRef]
- Yang, W.; Zhu, P.; Huang, H.; Zheng, Y.; Liu, J.; Feng, L.; Guo, H.; Tang, S.; Guo, R. Functionalization of Novel Theranostic Hydrogels with Kartogenin-Grafted USPIO Nanoparticles To Enhance Cartilage Regeneration. ACS Appl. Mater. Interfaces 2019, 11, 34744–34754. [Google Scholar] [CrossRef]
- Abdeen, A.A.; Lee, J.; Bharadwaj, N.A.; Ewoldt, R.H.; Kilian, K.A. Temporal Modulation of Stem Cell Activity Using Magnetoactive Hydrogels. Adv. Healthc. Mater. 2016, 5, 2536–2544. [Google Scholar] [CrossRef]
- Li, C.; Armstrong, J.P.K.; Pence, I.J.; Kit-Anan, W.; Puetzer, J.L.; Correia Carreira, S.; Moore, A.C.; Stevens, M.M. Glycosylated superparamagnetic nanoparticle gradients for osteochondral tissue engineering. Biomaterials 2018, 176, 24–33. [Google Scholar] [CrossRef] [PubMed]
- Silva, E.D.; Babo, P.S.; Costa-Almeida, R.; Domingues, R.M.A.; Mendes, B.B.; Paz, E.; Freitas, P.; Rodrigues, M.T.; Granja, P.L.; Gomes, M.E. Multifunctional magnetic-responsive hydrogels to engineer tendon-to-bone interface. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 2375–2385. [Google Scholar] [CrossRef] [PubMed]
- Zhang, N.; Lock, J.; Sallee, A.; Liu, H. Magnetic Nanocomposite Hydrogel for Potential Cartilage Tissue Engineering: Synthesis, Characterization, and Cytocompatibility with Bone Marrow Derived Mesenchymal Stem Cells. ACS Appl. Mater. Interfaces 2015, 7, 20987–20998. [Google Scholar] [CrossRef]
- Chen, W.; Zhang, Y.; Kumari, J.; Engelkamp, H.; Kouwer, P.H.J. Magnetic Stiffening in 3D Cell Culture Matrices. Nano Lett. 2021, 21, 6740–6747. [Google Scholar] [CrossRef]
- Ferrotec. Manufacturing Advanced Material, Component, and System Solutions for Precision Processes. Available online: https://www.ferrotec.com/ (accessed on 20 July 2025).
- Tang, J.; Tong, Z.; Xia, Y.; Liu, M.; Lv, Z.; Gao, Y.; Lu, T.; Xie, S.; Pei, Y.; Fang, D.; et al. Super tough magnetic hydrogels for remotely triggered shape morphing. J. Mater. Chem. B 2018, 6, 2713–2722. [Google Scholar] [CrossRef]
- Piantanida, E.; Alonci, G.; Bertucci, A.; De Cola, L. Design of Nanocomposite Injectable Hydrogels for Minimally Invasive Surgery. Acc. Chem. Res. 2019, 52, 2101–2112. [Google Scholar] [CrossRef] [PubMed]
- Du, W.; Zong, Q.; Guo, R.; Ling, G.; Zhang, P. Injectable Nanocomposite Hydrogels for Cancer Therapy. Macromol. Biosci. 2021, 21, 2100186. [Google Scholar] [CrossRef]
- Wu, H.; Song, L.; Chen, L.; Zhang, W.; Chen, Y.; Zang, F.; Chen, H.; Ma, M.; Gu, N.; Zhang, Y. Injectable magnetic supramolecular hydrogel with magnetocaloric liquid-conformal property prevents post-operative recurrence in a breast cancer model. Acta Biomater. 2018, 74, 302–311. [Google Scholar] [CrossRef]
- Ma, H.; Yu, G.; Cheng, J.; Song, L.; Zhou, Z.; Zhao, Y.; Zhao, Q.; Liu, L.; Wei, X.; Yang, M. Design of an Injectable Magnetic Hydrogel Based on the Tumor Microenvironment for Multimodal Synergistic Cancer Therapy. Biomacromolecules 2023, 24, 868–885. [Google Scholar] [CrossRef]
- Qian, K.-Y.; Song, Y.; Yan, X.; Dong, L.; Xue, J.; Xu, Y.; Wang, B.; Cao, B.; Hou, Q.; Peng, W.; et al. Injectable ferrimagnetic silk fibroin hydrogel for magnetic hyperthermia ablation of deep tumor. Biomaterials 2020, 259, 120299. [Google Scholar] [CrossRef]
- Masanam, H.B.; Muthuraman, J.; Chandra, B.; Kottapalli, V.N.S.M.; Chandra, S.S.; Gupta, P.K.; Narasimhan, A.K. Investigation on the heating effects of intra-tumoral injectable magnetic hydrogels (IT-MG) for cancer hyperthermia. Biomed. Phys. Eng. Express 2025, 11, 025036. [Google Scholar] [CrossRef]
- Wu, H.; Song, L.; Chen, L.; Huang, Y.; Wu, Y.; Zang, F.; An, Y.; Lyu, H.; Ma, M.; Chen, J.; et al. Injectable thermosensitive magnetic nanoemulsion hydrogel for multimodal-imaging-guided accurate thermoablative cancer therapy. Nanoscale 2017, 9, 16175–16182. [Google Scholar] [CrossRef]
- Farzaneh, M.; Hassanzadeh-Tabrizi, S.A.; Mokhtarian, N. Preparation of an Injectable Thermosensitive Pluronic F127/ γ-Fe2O3 Hydrogel for Combined Hyperthermia and Localized Drug Delivery for Cancer Treatment. Polym. Adv. Technol. 2024, 35, e6616. [Google Scholar] [CrossRef]
- Barbucci, R.; Pasqui, D.; Giani, G.; De Cagna, M.; Fini, M.; Giardino, R.; Atrei, A. A novel strategy for engineering hydrogels with ferromagnetic nanoparticles as crosslinkers of the polymer chains. Potential applications as a targeted drug delivery system. Soft Matter 2011, 7, 5558. [Google Scholar] [CrossRef]
- Uva, M.; Pasqui, D.; Mencuccini, L.; Fedi, S.; Barbucci, R. Influence of Alternating and Static Magnetic Fields on Drug Release from Hybrid Hydrogels Containing Magnetic Nanoparticles. J. Biomater. Nanobiotechnol. 2014, 5, 116–127. [Google Scholar] [CrossRef]
- Uva, M.; Mencuccini, L.; Atrei, A.; Innocenti, C.; Fantechi, E.; Sangregorio, C.; Maglio, M.; Fini, M.; Barbucci, R. On the Mechanism of Drug Release from Polysaccharide Hydrogels Cross-Linked with Magnetite Nanoparticles by Applying Alternating Magnetic Fields: The Case of DOXO Delivery. Gels 2015, 1, 24–43. [Google Scholar] [CrossRef]
- Vítková, L.; Kazantseva, N.; Musilová, L.; Smolka, P.; Valášková, K.; Kocourková, K.; Humeník, M.; Minařík, A.; Humpolíček, P.; Mráček, A.; et al. Magneto-responsive hyaluronan hydrogel for hyperthermia and bioprinting: Magnetic, rheological properties and biocompatibility. APL Bioeng. 2023, 7, 036113. [Google Scholar] [CrossRef]
- Xuan, X.; Li, Y.; Xu, X.; Pan, Z.; Li, Y.; Luo, Y.; Sun, L. Three-Dimensional Printable Magnetic Hydrogels with Adjustable Stiffness and Adhesion for Magnetic Actuation and Magnetic Hyperthermia Applications. Gels 2025, 11, 67. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.; Wang, L.; Xu, Y.; Du, W.; Cai, X.; Wang, F.; Ling, Y.; Chen, H.; Wang, Z.; Hu, B.; et al. A pH and magnetic dual-response hydrogel for synergistic chemo-magnetic hyperthermia tumor therapy. RSC Adv. 2018, 8, 9812–9821. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.; Song, Y.; Yan, X.; Dong, L.; Xu, Y.; Xuan, S.; Shu, Q.; Cao, B.; Hu, J.; Xing, H.; et al. Injectable magnetic montmorillonite colloidal gel for the postoperative treatment of hepatocellular carcinoma. J. Nanobiotechnol 2022, 20, 381. [Google Scholar] [CrossRef] [PubMed]
- Ribeiro, M.; Boudoukhani, M.; Belmonte-Reche, E.; Genicio, N.; Sillankorva, S.; Gallo, J.; Rodríguez-Abreu, C.; Moulai-Mostefa, N.; Bañobre-López, M. Xanthan-Fe3O4 Nanoparticle Composite Hydrogels for Non-Invasive Magnetic Resonance Imaging and Magnetically Assisted Drug Delivery. ACS Appl. Nano Mater. 2021, 4, 7712–7729. [Google Scholar] [CrossRef]
- Bertsch, P.; Diba, M.; Mooney, D.J.; Leeuwenburgh, S.C.G. Self-Healing Injectable Hydrogels for Tissue Regeneration. Chem. Rev. 2023, 123, 834–873. [Google Scholar] [CrossRef]
- Chen, M.; Gong, G.; Zhou, L.; Zhang, F. Facile fabrication of a magnetic self-healing poly(vinyl alcohol) composite hydrogel. RSC Adv. 2017, 7, 21476–21483. [Google Scholar] [CrossRef]
- Tang, J.; Yin, Q.; Shi, M.; Yang, M.; Yang, H.; Sun, B.; Guo, B.; Wang, T. Programmable shape transformation of 3D printed magnetic hydrogel composite for hyperthermia cancer therapy. Extrem. Mech. Lett. 2021, 46, 101305. [Google Scholar] [CrossRef]
- Eivazzadeh-Keihan, R.; Radinekiyan, F.; Maleki, A.; Salimi Bani, M.; Hajizadeh, Z.; Asgharnasl, S. A novel biocompatible core-shell magnetic nanocomposite based on cross-linked chitosan hydrogels for in vitro hyperthermia of cancer therapy. Int. J. Biol. Macromol. 2019, 140, 407–414. [Google Scholar] [CrossRef] [PubMed]
- Haghighattalab, M.; Kajbafzadeh, A.; Baghani, M.; Gharehnazifam, Z.; Jobani, B.M.; Baniassadi, M. Silk Fibroin Hydrogel Reinforced With Magnetic Nanoparticles as an Intelligent Drug Delivery System for Sustained Drug Release. Front. Bioeng. Biotechnol. 2022, 10, 891166. [Google Scholar] [CrossRef]
- Shin, B.Y.; Cha, B.G.; Jeong, J.H.; Kim, J. Injectable Macroporous Ferrogel Microbeads with a High Structural Stability for Magnetically Actuated Drug Delivery. ACS Appl. Mater. Interfaces 2017, 9, 31372–31380. [Google Scholar] [CrossRef] [PubMed]
- Hu, Q.; Li, H.; Archibong, E.; Chen, Q.; Ruan, H.; Ahn, S.; Dukhovlinova, E.; Kang, Y.; Wen, D.; Dotti, G.; et al. Inhibition of post-surgery tumour recurrence via a hydrogel releasing CAR-T cells and anti-PDL1-conjugated platelets. Nat. Biomed. Eng. 2021, 5, 1038–1047. [Google Scholar] [CrossRef]
- Shirsat, S.D.; Londhe, P.V.; Gaikwad, A.P.; Rizwan, M.; Laha, S.S.; Khot, V.M.; Achal, V.; Tabish, T.A.; Thorat, N.D. Endosomal escape in magnetic nanostructures: Recent advances and future perspectives. Mater. Today Adv. 2024, 22, 100484. [Google Scholar] [CrossRef]
- Sanoh, N.; Salazar, G.; Penaloza, D.P., Jr. Magnetic Biopolymeric Hydrogel Composite Material with Self-healing Attribute. Biointerface Res. Appl. Chem. 2021, 11, 14881–14888. [Google Scholar] [CrossRef]
- Baron, R.I.; Biliuta, G.; Socoliuc, V.; Coseri, S. Affordable Magnetic Hydrogels Prepared from Biocompatible and Biodegradable Sources. Polymers 2021, 13, 1693. [Google Scholar] [CrossRef]
- Carvalho, E.O.; Ribeiro, C.; Correia, D.M.; Botelho, G.; Lanceros-Mendez, S. Biodegradable Hydrogels Loaded with Magnetically Responsive Microspheres as 2D and 3D Scaffolds. Nanomaterials 2020, 10, 2421. [Google Scholar] [CrossRef]
- Zhang, Y.; Yang, B.; Zhang, X.; Xu, L.; Tao, L.; Li, S.; Wei, Y. A magnetic self-healing hydrogel. Chem. Commun. 2012, 48, 9305. [Google Scholar] [CrossRef]
- Ji, Y.; Wang, C. Magnetic iron oxide nanoparticle-loaded hydrogels for photothermal therapy of cancer cells. Front. Bioeng. Biotechnol. 2023, 11, 1130523. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Yu, R.-Y.; Wang, K.-Q.; Zhang, Z.-Y.; Ardekani, A.; Hu, Y.-D. Spherical Magnetic Fe-Alginate Microgels Fabricated by Droplet-Microfluidics Combining with an External Crosslinking Approach and the Study of Their pH Dependent Fe3+ Release Behaviors. Chin. J. Polym. Sci. 2025, 43, 289–302. [Google Scholar] [CrossRef]
- Bairoch, A. The Cellosaurus, a Cell-Line Knowledge Resource. J. Biomol. Tech. 2018, 29, 25–38. [Google Scholar] [CrossRef] [PubMed]
- Dash, P.; Panda, P.K.; Su, C.; Lin, Y.-C.; Sakthivel, R.; Chen, S.-L.; Chung, R.-J. Near-infrared-driven upconversion nanoparticles with photocatalysts through water-splitting towards cancer treatment. J. Mater. Chem. B 2024, 12, 3881–3907. [Google Scholar] [CrossRef]
- Mahmoudi, K.; Bouras, A.; Bozec, D.; Ivkov, R.; Hadjipanayis, C. Magnetic hyperthermia therapy for the treatment of glioblastoma: A review of the therapy’s history, efficacy and application in humans. Int. J. Hyperth. 2018, 34, 1316–1328. [Google Scholar] [CrossRef] [PubMed]
- Ganguly, S.; Margel, S. Design of Magnetic Hydrogels for Hyperthermia and Drug Delivery. Polymers 2021, 13, 4259. [Google Scholar] [CrossRef]
- Gherghe, M.; Lazar, A.M.; Mutuleanu, M.-D.; Bordea, C.I.; Ionescu, S.; Mihaila, R.I.; Petroiu, C.; Stanciu, A.E. Evaluating Cardiotoxicity in Breast Cancer Patients Treated with HER2 Inhibitors: Could a Combination of Radionuclide Ventriculography and Cardiac Biomarkers Predict the Cardiac Impact? Cancers 2023, 15, 207. [Google Scholar] [CrossRef]
- Meenach, S.A.; Hilt, J.Z.; Anderson, K.W. Poly(ethylene glycol)-based magnetic hydrogel nanocomposites for hyperthermia cancer therapy. Acta Biomater. 2010, 6, 1039–1046. [Google Scholar] [CrossRef]
- Meenach, S.A.; Otu, C.G.; Anderson, K.W.; Hilt, J.Z. Controlled synergistic delivery of paclitaxel and heat from poly(β-amino ester)/iron oxide-based hydrogel nanocomposites. Int. J. Pharm. 2012, 427, 177–184. [Google Scholar] [CrossRef]
- Eivazzadeh-Keihan, R.; Pajoum, Z.; Aliabadi, H.A.M.; Mohammadi, A.; Kashtiaray, A.; Bani, M.S.; Pishva, B.; Maleki, A.; Heravi, M.M.; Mahdavi, M.; et al. Magnetized chitosan hydrogel and silk fibroin, reinforced with PVA: A novel nanobiocomposite for biomedical and hyperthermia applications. RSC Adv. 2023, 13, 8540–8550. [Google Scholar] [CrossRef]
- Le Renard, P.-E.; Jordan, O.; Faes, A.; Petri-Fink, A.; Hofmann, H.; Rüfenacht, D.; Bosman, F.; Buchegger, F.; Doelker, E. The in vivo performance of magnetic particle-loaded injectable, in situ gelling, carriers for the delivery of local hyperthermia. Biomaterials 2010, 31, 691–705. [Google Scholar] [CrossRef]
- Derakhshankhah, H.; Jahanban-Esfahlan, R.; Vandghanooni, S.; Akbari-Nakhjavani, S.; Massoumi, B.; Haghshenas, B.; Rezaei, A.; Farnudiyan-Habibi, A.; Samadian, H.; Jaymand, M. A bio-inspired gelatin-based pH- and thermal-sensitive magnetic hydrogel for in vitro chemo/hyperthermia treatment of breast cancer cells. J. Appl. Polym. Sci. 2021, 138, 50578. [Google Scholar] [CrossRef]
- Najafipour, A.; Gharieh, A.; Fassihi, A.; Sadeghi-Aliabadi, H.; Mahdavian, A.R. MTX-Loaded Dual Thermoresponsive and pH-Responsive Magnetic Hydrogel Nanocomposite Particles for Combined Controlled Drug Delivery and Hyperthermia Therapy of Cancer. Mol. Pharm. 2021, 18, 275–284. [Google Scholar] [CrossRef] [PubMed]
- Indulekha, S.; Arunkumar, P.; Bahadur, D.; Srivastava, R. Dual responsive magnetic composite nanogels for thermo-chemotherapy. Colloids Surf. B Biointerfaces 2017, 155, 304–313. [Google Scholar] [CrossRef] [PubMed]
- Chen, B.; Xing, J.; Li, M.; Liu, Y.; Ji, M. DOX@Ferumoxytol-Medical Chitosan as magnetic hydrogel therapeutic system for effective magnetic hyperthermia and chemotherapy in vitro. Colloids Surf. B Biointerfaces 2020, 190, 110896. [Google Scholar] [CrossRef]
- Yan, X.; Sun, T.; Song, Y.; Peng, W.; Xu, Y.; Luo, G.; Li, M.; Chen, S.; Fang, W.-W.; Dong, L.; et al. In situ Thermal-Responsive Magnetic Hydrogel for Multidisciplinary Therapy of Hepatocellular Carcinoma. Nano Lett. 2022, 22, 2251–2260. [Google Scholar] [CrossRef]
- Onisâi, M.; Dumitru, A.; Iordan, I.; Aliuș, C.; Teodor, O.; Alexandru, A.; Gheorghiță, D.; Antoniac, I.; Nica, A.; Mihăilescu, A.-A.; et al. Synchronous Multiple Breast Cancers—Do We Need to Reshape Staging? Medicina 2020, 56, 230. [Google Scholar] [CrossRef]
- Grădinaru, E.S.; Stoicea, M.-C.; Mocanu, L.; Antoniac, I.; Gheorghiță, D.; Grigore, A.G.M. Rare Breast Carcinoma with Paradoxical Plasma Cell Immunoprofile: A Case Report. Medicina 2020, 56, 62. [Google Scholar] [CrossRef] [PubMed]
- Lazar, A.M.; Mutuleanu, M.-D.; Spiridon, P.M.; Bordea, C.I.; Suta, T.L.; Blidaru, A.; Gherghe, M. Feasibility of Sentinel Lymph Node Biopsy in Breast Cancer Patients with Axillary Conversion after Neoadjuvant Chemotherapy—A Single-Tertiary Centre Experience and Review of the Literature. Diagnostics 2023, 13, 3000. [Google Scholar] [CrossRef] [PubMed]
- Du, Y.; Yan, X.; Wu, W.; Cao, B.; Hu, J.; Xue, J.; Lu, Y. Magnetic and pH dual responsive injectable magnetic zeolitic imidazole framework based colloidal gel for interventional treatment on hepatocellular carcinoma. Chem. Eng. J. 2024, 497, 154180. [Google Scholar] [CrossRef]
- Carter, T.J.; Agliardi, G.; Lin, F.; Ellis, M.; Jones, C.; Robson, M.; Richard-Londt, A.; Southern, P.; Lythgoe, M.; Zaw Thin, M.; et al. Potential of Magnetic Hyperthermia to Stimulate Localized Immune Activation. Small 2021, 17, 2005241. [Google Scholar] [CrossRef]
- Doktorovova, S.; Souto, E.B. Nanostructured lipid carrier-based hydrogel formulations for drug delivery: A comprehensive review. Expert Opin. Drug Deliv. 2009, 6, 165–176. [Google Scholar] [CrossRef]
- Ganguly, S.; Das, N.C. Synthesis of Mussel Inspired Polydopamine Coated Halloysite Nanotubes Based Semi-IPN: An Approach to Fine Tuning in Drug Release and Mechanical Toughening. Macromol. Symp. 2018, 382, 1800076. [Google Scholar] [CrossRef]
- Ganguly, S.; Maity, P.P.; Mondal, S.; Das, P.; Bhawal, P.; Dhara, S.; Das, N.C. Polysaccharide and poly(methacrylic acid) based biodegradable elastomeric biocompatible semi-IPN hydrogel for controlled drug delivery. Mater. Sci. Eng. C 2018, 92, 34–51. [Google Scholar] [CrossRef]
- Ganguly, S. 2—Preparation/processing of polymer-graphene composites by different techniques. In Polymer Nanocomposites Containing Graphene; Rahaman, M., Nayak, L., Hussein, I.A., Das, N.C., Eds.; Woodhead Publishing Series in Composites Science and Engineering; Woodhead Publishing: Cambridge, UK, 2022; pp. 45–74. ISBN 978-0-12-821639-2. [Google Scholar]
- Falk, B.; Garramone, S.; Shivkumar, S. Diffusion coefficient of paracetamol in a chitosan hydrogel. Mater. Lett. 2004, 58, 3261–3265. [Google Scholar] [CrossRef]
- Mohamed, M.A.; Fallahi, A.; El-Sokkary, A.M.A.; Salehi, S.; Akl, M.A.; Jafari, A.; Tamayol, A.; Fenniri, H.; Khademhosseini, A.; Andreadis, S.T.; et al. Stimuli-responsive hydrogels for manipulation of cell microenvironment: From chemistry to biofabrication technology. Prog. Polym. Sci. 2019, 98, 101147. [Google Scholar] [CrossRef]
- Prabaharan, M.; Mano, J.F. Stimuli-Responsive Hydrogels Based on Polysaccharides Incorporated with Thermo-Responsive Polymers as Novel Biomaterials. Macromol. Biosci. 2006, 6, 991–1008. [Google Scholar] [CrossRef]
- Ganguly, S.; Das, N.C. Water Uptake Kinetics and Control Release of Agrochemical Fertilizers from Nanoclay-Assisted Semi-interpenetrating Sodium Acrylate-Based Hydrogel. Polym.-Plast. Technol. Eng. 2017, 56, 744–761. [Google Scholar] [CrossRef]
- Lee, P.I. Kinetics of drug release from hydrogel matrices. J. Control. Release 1985, 2, 277–288. [Google Scholar] [CrossRef]
- Liu, T.-Y.; Hu, S.-H.; Liu, K.-H.; Liu, D.-M.; Chen, S.-Y. Preparation and characterization of smart magnetic hydrogels and its use for drug release. J. Magn. Magn. Mater. 2006, 304, e397–e399. [Google Scholar] [CrossRef]
- Huang, Y.; Liu, M.; Chen, J.; Gao, C.; Gong, Q. A novel magnetic triple-responsive composite semi-IPN hydrogels for targeted and controlled drug delivery. Eur. Polym. J. 2012, 48, 1734–1744. [Google Scholar] [CrossRef]
- Dong, J.; Du, X.; Zhang, Y.; Zhuang, T.; Cui, X.; Li, Z. Thermo/glutathione-sensitive release kinetics of heterogeneous magnetic micro-organogel prepared by sono-catalysis. Colloids Surf. B Biointerfaces 2021, 208, 112109. [Google Scholar] [CrossRef] [PubMed]
- Hamidian, H.; Tavakoli, T. Preparation of a new Fe3O4/starch-g-polyester nanocomposite hydrogel and a study on swelling and drug delivery properties. Carbohydr. Polym. 2016, 144, 140–148. [Google Scholar] [CrossRef] [PubMed]
- Lin, F.; Zheng, J.; Guo, W.; Zhu, Z.; Wang, Z.; Dong, B.; Lin, C.; Huang, B.; Lu, B. Smart cellulose-derived magnetic hydrogel with rapid swelling and deswelling properties for remotely controlled drug release. Cellulose 2019, 26, 6861–6877. [Google Scholar] [CrossRef]
- Zhang, Z.-Q.; Kim, Y.-M.; Song, S.-C. Injectable and Quadruple-Functional Hydrogel as an Alternative to Intravenous Delivery for Enhanced Tumor Targeting. ACS Appl. Mater. Interfaces 2019, 11, 34634–34644. [Google Scholar] [CrossRef]
- Nandwana, V.; Ryoo, S.-R.; Zheng, T.; You, M.M.; Dravid, V.P. Magnetic Nanostructure-Coated Thermoresponsive Hydrogel Nanoconstruct as a Smart Multimodal Theranostic Platform. ACS Biomater. Sci. Eng. 2019, 5, 3049–3059. [Google Scholar] [CrossRef]
- Eskandani, M.; Jahanban-Esfahlan, R.; Sadughi, M.M.; Jaymand, M. Thermal-responsive β-cyclodextrin-based magnetic hydrogel as a de novo nanomedicine for chemo/hyperthermia treatment of cancerous cells. Heliyon 2024, 10, e32183. [Google Scholar] [CrossRef]
- Yuan, P.; Yang, T.; Liu, T.; Yu, X.; Bai, Y.; Zhang, Y.; Chen, X. Nanocomposite hydrogel with NIR/magnet/enzyme multiple responsiveness to accurately manipulate local drugs for on-demand tumor therapy. Biomaterials 2020, 262, 120357. [Google Scholar] [CrossRef] [PubMed]
- Sayadnia, S.; Arkan, E.; Jahanban-Esfahlan, R.; Sayadnia, S.; Jaymand, M. Tragacanth gum-based pH-responsive magnetic hydrogels for “smart” chemo/hyperthermia therapy of solid tumors. Polym. Adv. Technol. 2021, 32, 262–271. [Google Scholar] [CrossRef]
- Kim, C.; Kim, H.; Park, H.; Lee, K.Y. Controlling the porous structure of alginate ferrogel for anticancer drug delivery under magnetic stimulation. Carbohydr. Polym. 2019, 223, 115045. [Google Scholar] [CrossRef]
- Hu, X.; Wang, Y.; Zhang, L.; Xu, M.; Zhang, J.; Dong, W. Magnetic field-driven drug release from modified iron oxide-integrated polysaccharide hydrogel. Int. J. Biol. Macromol. 2018, 108, 558–567. [Google Scholar] [CrossRef]
- Yao, C.; Yuan, Y.; Yang, D. Magnetic DNA Nanogels for Targeting Delivery and Multistimuli-Triggered Release of Anticancer Drugs. ACS Appl. Bio Mater. 2018, 1, 2012–2020. [Google Scholar] [CrossRef]
- Sui, X.; Jin, T.; Liu, T.; Wu, S.; Wu, Y.; Tang, Z.; Ren, Y.; Ni, D.; Yao, Z.; Zhang, H. Tumor Immune Microenvironments (TIMEs): Responsive Nanoplatforms for Antitumor Immunotherapy. Front. Chem. 2020, 8, 804. [Google Scholar] [CrossRef]
- Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
- Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [PubMed]
- Sarfaty, M.; Hall, P.S.; Chan, K.K.W.; Virik, K.; Leshno, M.; Gordon, N.; Moore, A.; Neiman, V.; Rosenbaum, E.; Goldstein, D.A. Cost-effectiveness of Pembrolizumab in Second-line Advanced Bladder Cancer. Eur. Urol. 2018, 74, 57–62. [Google Scholar] [CrossRef]
- Verma, V.; Sprave, T.; Haque, W.; Simone, C.B.; Chang, J.Y.; Welsh, J.W.; Thomas, C.R. A systematic review of the cost and cost-effectiveness studies of immune checkpoint inhibitors. J. Immunother. Cancer 2018, 6, 128. [Google Scholar] [CrossRef]
- Tartari, F.; Santoni, M.; Burattini, L.; Mazzanti, P.; Onofri, A.; Berardi, R. Economic sustainability of anti-PD-1 agents nivolumab and pembrolizumab in cancer patients: Recent insights and future challenges. Cancer Treat. Rev. 2016, 48, 20–24. [Google Scholar] [CrossRef]
- Remon, J.; Besse, B.; Soria, J.-C. Successes and failures: What did we learn from recent first-line treatment immunotherapy trials in non-small cell lung cancer? BMC Med. 2017, 15, 55. [Google Scholar] [CrossRef]
- Wang, S.; Sun, Z.; Hou, Y. Engineering Nanoparticles toward the Modulation of Emerging Cancer Immunotherapy. Adv. Healthc. Mater. 2021, 10, e2000845. [Google Scholar] [CrossRef]
- El-Sawy, H.S.; Al-Abd, A.M.; Ahmed, T.A.; El-Say, K.M.; Torchilin, V.P. Stimuli-Responsive Nano-Architecture Drug-Delivery Systems to Solid Tumor Micromilieu: Past, Present, and Future Perspectives. ACS Nano 2018, 12, 10636–10664. [Google Scholar] [CrossRef]
- Day, N.B.; Wixson, W.C.; Shields, C.W. Magnetic systems for cancer immunotherapy. Acta Pharm. Sin. B 2021, 11, 2172–2196. [Google Scholar] [CrossRef]
- Wang, S.; Jing, H.; Yang, R.; Heger, Z.; Krizkova, S.; Zhou, Y.; Liang, X.; Adam, V.; Li, N. Magnetocaloric-Responsive Hydrogel Nanoarchitectonics for Pyroptosis-Relay-Immunotherapy to Suppress Post-Operation Tumor Recurrence and Metastasis. Adv. Funct. Mater. 2024, 34, 2314194. [Google Scholar] [CrossRef]
- Quan, Z.; Chen, L.; Chen, B.; Guo, K.; Tian, P.; Pan, H.; Li, Y.; Zhao, N.; Xu, F.-J. Immunomodulatory hydrogel neutralizes tumor acidity to augment immune responses for the prevention of post-surgical recurrence of melanoma. Sci. China Mater. 2025, 68, 270–279. [Google Scholar] [CrossRef]
- Yan, M.; Peng, J.; Wu, H.; Ma, M.; Zhang, Y. Injectable Magnetic-Nanozyme Based Thermosensitive Hydrogel for Multimodal DLBCL Therapy. Gels 2025, 11, 218. [Google Scholar] [CrossRef] [PubMed]
- Massana Roquero, D.; Smutok, O.; Othman, A.; Melman, A.; Katz, E. “Smart” Delivery of Monoclonal Antibodies from a Magnetic Responsive Microgel Nanocomposite. ACS Appl. Bio Mater. 2021, 4, 8487–8497. [Google Scholar] [CrossRef] [PubMed]
- Nowak-Jary, J.; Machnicka, B. Comprehensive Analysis of the Potential Toxicity of Magnetic Iron Oxide Nanoparticles for Medical Applications: Cellular Mechanisms and Systemic Effects. Int. J. Mol. Sci. 2024, 25, 12013. [Google Scholar] [CrossRef] [PubMed]
- Wu, L.; Wen, W.; Wang, X.; Huang, D.; Cao, J.; Qi, X.; Shen, S. Ultrasmall iron oxide nanoparticles cause significant toxicity by specifically inducing acute oxidative stress to multiple organs. Part. Fibre Toxicol. 2022, 19, 24. [Google Scholar] [CrossRef] [PubMed]
- Ying, H.; Ruan, Y.; Zeng, Z.; Bai, Y.; Xu, J.; Chen, S. Iron oxide nanoparticles size-dependently activate mouse primary macrophages via oxidative stress and endoplasmic reticulum stress. Int. Immunopharmacol. 2022, 105, 108533. [Google Scholar] [CrossRef]
- Ansari, M.O.; Parveen, N.; Ahmad, M.F.; Wani, A.L.; Afrin, S.; Rahman, Y.; Jameel, S.; Khan, Y.A.; Siddique, H.R.; Tabish, M.; et al. Evaluation of DNA interaction, genotoxicity and oxidative stress induced by iron oxide nanoparticles both in vitro and in vivo: Attenuation by thymoquinone. Sci. Rep. 2019, 9, 6912. [Google Scholar] [CrossRef]
- Kenzaoui, B.H.; Bernasconi, C.C.; Hofmann, H.; Juillerat-Jeanneret, L. Evaluation of uptake and transport of ultrasmall superparamagnetic iron oxide nanoparticles by human brain-derived endothelial cells. Nanomedicine 2012, 7, 39–53. [Google Scholar] [CrossRef] [PubMed]
- Zhang, T.; Qian, L.; Tang, M.; Xue, Y.; Kong, L.; Zhang, S.; Pu, Y. Evaluation on Cytotoxicity and Genotoxicity of the L-Glutamic Acid Coated Iron Oxide Nanoparticles. J. Nanosci. Nanotechnol. 2012, 12, 2866–2873. [Google Scholar] [CrossRef]
- Watanabe, M.; Yoneda, M.; Morohashi, A.; Hori, Y.; Okamoto, D.; Sato, A.; Kurioka, D.; Nittami, T.; Hirokawa, Y.; Shiraishi, T.; et al. Effects of Fe3O4 Magnetic Nanoparticles on A549 Cells. Int. J. Mol. Sci. 2013, 14, 15546–15560. [Google Scholar] [CrossRef]
- Bourrinet, P.; Bengele, H.H.; Bonnemain, B.; Dencausse, A.; Idee, J.-M.; Jacobs, P.M.; Lewis, J.M. Preclinical safety and pharmacokinetic profile of ferumoxtran-10, an ultrasmall superparamagnetic iron oxide magnetic resonance contrast agent. Investig. Radiol. 2006, 41, 313–324. [Google Scholar] [CrossRef]
- Zou, J.; Wang, X.; Zhang, L.; Wang, J. Iron nanoparticles significantly affect the in vitro and in vivo expression of Id genes. Chem. Res. Toxicol. 2015, 28, 373–383. [Google Scholar] [CrossRef]
- Askri, D.; Cunin, V.; Béal, D.; Berthier, S.; Chovelon, B.; Arnaud, J.; Rachidi, W.; Sakly, M.; Amara, S.; Sève, M.; et al. Investigating the toxic effects induced by iron oxide nanoparticles on neuroblastoma cell line: An integrative study combining cytotoxic, genotoxic and proteomic tools. Nanotoxicology 2019, 13, 1021–1040. [Google Scholar] [CrossRef]
- Pongrac, I.M.; Dobrivojević, M.; Ahmed, L.B.; Babič, M.; Šlouf, M.; Horák, D.; Gajović, S. Improved biocompatibility and efficient labeling of neural stem cells with poly(L-lysine)-coated maghemite nanoparticles. Beilstein J. Nanotechnol. 2016, 7, 926–936. [Google Scholar] [CrossRef]
- Majeed, S.; Mohd Rozi, N.A.B.; Danish, M.; Mohamad Ibrahim, M.N.; Joel, E.L. In vitro apoptosis and molecular response of engineered green iron oxide nanoparticles with l-arginine in MDA-MB-231 breast cancer cells. J. Drug Deliv. Sci. Technol. 2023, 80, 104185. [Google Scholar] [CrossRef]
- Fahmy, H.M.; Aly, E.M.; Mohamed, F.F.; Noor, N.A.; Elsayed, A.A. Neurotoxicity of green- synthesized magnetic iron oxide nanoparticles in different brain areas of wistar rats. Neurotoxicology 2020, 77, 80–93. [Google Scholar] [CrossRef]
- Kim, Y.; Kong, S.D.; Chen, L.-H.; Pisanic, T.R.; Jin, S.; Shubayev, V.I. In vivo nanoneurotoxicity screening using oxidative stress and neuroinflammation paradigms. Nanomed. Nanotechnol. Biol. Med. 2013, 9, 1057–1066. [Google Scholar] [CrossRef]
- Kim, J.S.; Yoon, T.-J.; Yu, K.N.; Kim, B.G.; Park, S.J.; Kim, H.W.; Lee, K.H.; Park, S.B.; Lee, J.-K.; Cho, M.H. Toxicity and Tissue Distribution of Magnetic Nanoparticles in Mice. Toxicol. Sci. 2006, 89, 338–347. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim Fouad, G.; El-Sayed, S.A.M.; Mabrouk, M.; Ahmed, K.A.; Beherei, H.H. Neuroprotective Potential of Intranasally Delivered Sulforaphane-Loaded Iron Oxide Nanoparticles Against Cisplatin-Induced Neurotoxicity. Neurotox. Res. 2022, 40, 1479–1498. [Google Scholar] [CrossRef]
- Paulini, F.; Marangon, A.R.M.; Azevedo, C.L.; Brito, J.L.M.; Lemos, M.S.; Sousa, M.H.; Veiga-Souza, F.H.; Souza, P.E.N.; Lucci, C.M.; Azevedo, R.B. In Vivo Evaluation of DMSA-Coated Magnetic Nanoparticle Toxicity and Biodistribution in Rats: A Long-Term Follow-Up. Nanomaterials 2022, 12, 3513. [Google Scholar] [CrossRef] [PubMed]
- Askri, D.; Ouni, S.; Galai, S.; Arnaud, J.; Chovelon, B.; Lehmann, S.G.; Sturm, N.; Sakly, M.; Sève, M.; Amara, S. Intranasal instillation of iron oxide nanoparticles induces inflammation and perturbation of trace elements and neurotransmitters, but not behavioral impairment in rats. Environ. Sci. Pollut. Res. Int. 2018, 25, 16922–16932. [Google Scholar] [CrossRef]
- Vu-Quang, H.; Yoo, M.-K.; Jeong, H.-J.; Lee, H.-J.; Muthiah, M.; Rhee, J.H.; Lee, J.-H.; Cho, C.-S.; Jeong, Y.Y.; Park, I.-K. Targeted delivery of mannan-coated superparamagnetic iron oxide nanoparticles to antigen-presenting cells for magnetic resonance-based diagnosis of metastatic lymph nodes in vivo. Acta Biomater. 2011, 7, 3935–3945. [Google Scholar] [CrossRef] [PubMed]
- Sekino, M.; Kuwahata, A.; Ookubo, T.; Shiozawa, M.; Ohashi, K.; Kaneko, M.; Saito, I.; Inoue, Y.; Ohsaki, H.; Takei, H.; et al. Handheld magnetic probe with permanent magnet and Hall sensor for identifying sentinel lymph nodes in breast cancer patients. Sci. Rep. 2018, 8, 1195. [Google Scholar] [CrossRef]
- Maier-Hauff, K.; Ulrich, F.; Nestler, D.; Niehoff, H.; Wust, P.; Thiesen, B.; Orawa, H.; Budach, V.; Jordan, A. Efficacy and safety of intratumoral thermotherapy using magnetic iron-oxide nanoparticles combined with external beam radiotherapy on patients with recurrent glioblastoma multiforme. J. Neuro-Oncol. 2011, 103, 317–324. [Google Scholar] [CrossRef]
- Johannsen, M.; Gneveckow, U.; Taymoorian, K.; Thiesen, B.; Waldöfner, N.; Scholz, R.; Jung, K.; Jordan, A.; Wust, P.; Loening, S.A. Morbidity and quality of life during thermotherapy using magnetic nanoparticles in locally recurrent prostate cancer: Results of a prospective phase I trial. Int. J. Hyperth. 2007, 23, 315–323. [Google Scholar] [CrossRef]
- Wust, P.; Gneveckow, U.; Johannsen, M.; Böhmer, D.; Henkel, T.; Kahmann, F.; Sehouli, J.; Felix, R.; Ricke, J.; Jordan, A. Magnetic nanoparticles for interstitial thermotherapy—Feasibility, tolerance and achieved temperatures. Int. J. Hyperth. 2006, 22, 673–685. [Google Scholar] [CrossRef]
- Maier-Hauff, K.; Rothe, R.; Scholz, R.; Gneveckow, U.; Wust, P.; Thiesen, B.; Feussner, A.; von Deimling, A.; Waldoefner, N.; Felix, R.; et al. Intracranial thermotherapy using magnetic nanoparticles combined with external beam radiotherapy: Results of a feasibility study on patients with glioblastoma multiforme. J. Neuro-Oncol. 2007, 81, 53–60. [Google Scholar] [CrossRef]
- Johannsen, M.; Gneveckow, U.; Thiesen, B.; Taymoorian, K.; Cho, C.H.; Waldöfner, N.; Scholz, R.; Jordan, A.; Loening, S.A.; Wust, P. Thermotherapy of prostate cancer using magnetic nanoparticles: Feasibility, imaging, and three-dimensional temperature distribution. Eur. Urol. 2007, 52, 1653–1661. [Google Scholar] [CrossRef]
- van Landeghem, F.K.H.; Maier-Hauff, K.; Jordan, A.; Hoffmann, K.-T.; Gneveckow, U.; Scholz, R.; Thiesen, B.; Brück, W.; von Deimling, A. Post-mortem studies in glioblastoma patients treated with thermotherapy using magnetic nanoparticles. Biomaterials 2009, 30, 52–57. [Google Scholar] [CrossRef]
- Ogita, M.; Yamashita, H.; Sawayanagi, S.; Takahashi, W.; Nakagawa, K. Efficacy of a hydrogel spacer in three-dimensional conformal radiation therapy for prostate cancer. Jpn. J. Clin. Oncol. 2020, 50, 303–309. [Google Scholar] [CrossRef] [PubMed]
- Karsh, L.I.; Gross, E.T.; Pieczonka, C.M.; Aliotta, P.J.; Skomra, C.J.; Ponsky, L.E.; Nieh, P.T.; Han, M.; Hamstra, D.A.; Shore, N.D. Absorbable Hydrogel Spacer Use in Prostate Radiotherapy: A Comprehensive Review of Phase 3 Clinical Trial Published Data. Urology 2018, 115, 39–44. [Google Scholar] [CrossRef]
- Chao, M.; Lim Joon, D.; Khoo, V.; Lawrentschuk, N.; Ho, H.; Spencer, S.; Chan, Y.; Tan, A.; Pham, T.; Sengupta, S.; et al. The use of hydrogel spacer in men undergoing high-dose prostate cancer radiotherapy: Results of a prospective phase 2 clinical trial. World J. Urol. 2019, 37, 1111–1116. [Google Scholar] [CrossRef] [PubMed]
- Mahmoudi, M.; Simchi, A.; Vali, H.; Imani, M.; Shokrgozar, M.A.; Azadmanesh, K.; Azari, F. Cytotoxicity and Cell Cycle Effects of Bare and Poly(vinyl alcohol)-Coated Iron Oxide Nanoparticles in Mouse Fibroblasts. Adv. Eng. Mater. 2009, 11, B243–B250. [Google Scholar] [CrossRef]
- Mahmoudi, M.; Simchi, A.; Imani, M. Cytotoxicity of Uncoated and Polyvinyl Alcohol Coated Superparamagnetic Iron Oxide Nanoparticles. J. Phys. Chem. C 2009, 113, 9573–9580. [Google Scholar] [CrossRef]
- Cavalu, S.; Antoniac, I.V.; Mohan, A.; Bodog, F.; Doicin, C.; Mates, I.; Ulmeanu, M.; Murzac, R.; Semenescu, A. Nanoparticles and Nanostructured Surface Fabrication for Innovative Cranial and Maxillofacial Surgery. Materials 2020, 13, 5391. [Google Scholar] [CrossRef] [PubMed]
- Cavalu, S.; Antoniac, I.V.; Fritea, L.; Mates, I.M.; Milea, C.; Laslo, V.; Vicas, S.; Mohan, A. Surface modifications of the titanium mesh for cranioplasty using selenium nanoparticles coating. J. Adhes. Sci. Technol. 2018, 32, 2509–2522. [Google Scholar] [CrossRef]
- Iulian, A.; Cosmin, S.; Aurora, A. Adhesion aspects in biomaterials and medical devices. J. Adhes. Sci. Technol. 2016, 30, 1711–1715. [Google Scholar] [CrossRef]
- Karlsson, H.L.; Cronholm, P.; Gustafsson, J.; Möller, L. Copper Oxide Nanoparticles Are Highly Toxic: A Comparison between Metal Oxide Nanoparticles and Carbon Nanotubes. Chem. Res. Toxicol. 2008, 21, 1726–1732. [Google Scholar] [CrossRef]
- Wada, S.; Yue, L.; Tazawa, K.; Furuta, I.; Nagae, H.; Takemori, S.; Minamimura, T. New local hyperthermia using dextran magnetite complex (DM) for oral cavity: Experimental study in normal hamster tongue. Oral Dis. 2001, 7, 192–195. [Google Scholar] [CrossRef] [PubMed]








| MNPs Core | Hydrodynamic Size | Coating | Dose Metric | Exposure to Outcomes | Remarks in Relation with Oncology | Ref. |
|---|---|---|---|---|---|---|
| Fe3O4 | 2.3, 4.2, and 9.3 nm | - | 100 mg/kg | Decreased MNPs toxicity; an increase in ROS in serum and internal organs in the 2.3 nm MNPs group, (2.16, 1.61, 4.03, and 1.9 times higher than in the control group). ROS level in the 2.3 nm Fe3O4 group was the highest compared to other hydrodynamic size particles | The ultrasmall Fe3O4 with sizes of 2.3 and 4.2 nm was highly toxic, compared to the larger MNPs with little toxic effects (9.3 nm) | Wu et al. [187] |
| Iron oxide MNPs | 10 and 30 nm | Polyethylene glycol (PEG) | 10 mg/kg body weight MNPs; control group (PBS solution) | MNPs induced the production of IL-6 and tumor necrosis factor-α (TNF-α). The inflammatory reactions were size-dependent, demonstrating that 30 nm MNPs induced greater oxidative stress | Exposure to MNPs has enhanced Neutrophil migration and infiltration | Ying et al. [188] |
| Fe3O4 | 60 nm | Coated with biocompatible polymers (non-specified in the study) | 15, 50, or 100 mg/kg body weight | The decrease in cell viability was linked to an increase in ROS generation as a function of MNPs dose | Used in conjunction with thymoquinone, the oxidative stress and genetic toxicity were attenuated | Ansari et al. [189] |
| γ-Fe2O3 | 15 nm | L-Glutamic acid | 128 μg/mL gel, cell density 5 × 105 cells/mL | A slight increase in DNA damage was noticed compared to the control group. ROS apparition was evidenced in in vitro study, and a maximum effect was achieved at the highest MNPs concentration | No genotoxicity effects were noticed in combination with reduced toxicity. Acellular and intracellular ROS were present with no DNA damage | Zhang et al. [191] |
| Fe3O4 | 12 nm | DMSA-coated MNPs | 50 and 100 µg/mL | The transcription of the Id3 gene was significantly downregulated in different cell lines treated with two doses (50 and 100 µg/mL) of coated MNPs | The MNPs exerted a significant effect on the expression of Id genes | Zou et al. [194] |
| Fe2O3 | 14, 22, and 30 nm | - | 12.5, 25, 50, 100, and 200 lg/mL gel | Genotoxicity and cytotoxicity tests have shown that MNPs are more toxic to cells compared to free ferric ions | The proteomic investigation proved cytoskeleton disruption and an important influence on cell division when MNPs are used as a potential tumor treatment | Askari et al. [195] |
| Carob-leaf synthesized iron oxide nanoparticles | 15.63 ± 2.38 nm | - | 10 mg/kg | The hippocampus and striatum of Wistar rats exhibited increased toxicity related to MNPs | A good potential for the carob-synthesized MNPs for the treatment of cerebral cancer was foreseen | Fahmy et al. [198] |
| Fe3O4 | 8 nm ÷ 10 nm | DMSA, PEG, PEG-Au | 15 mM | ROS existence was evidenced in T cells in nerve concomitantly with activation of the inflammation (MMP-9, IL-1β) signaling pathways | The intraneural MNPs injection into the sciatic nerve in rats proved an enhanced toxicity with applicability in cancer therapy | Kim et al. [199] |
| Section | Pros | Cons | Recommendation for Clinical Translation |
|---|---|---|---|
| MG preparation methods and main attributes | Blending: fast and simple method (attribute—superparamagnetism); In situ: uniform distribution (attribute—biocompatibility); Ex situ: scalability and ease of processing (attribute—tunable properties); Grafting onto: no MNPs leakage, higher stability of magnetic properties (attribute—magnetic property stability) | Blending: MNPs aggregation and poor distribution; In situ: complex process requires a large number of precursors; Ex situ: MNPs non-uniform diffusion and aggregation; Grafting onto: time-consuming and challenging procedure | One should choose the synthesis method by considering the particularity of the oncological application |
| In vivo tests | MHT: good hyperthermia effects and possible chemoembolization; Drug delivery: improved drug delivery process and controlled release; Immunotherapy: good potential even in the absence of an external AMF | MHT: potential toxicity; Drug delivery: difficulty in drug release control; Immunotherapy: increased costs and unexpected immune reactions | The in vivo tests for MHT showed promising potential for cancer cell death, but the risks of some cellular damage must be considered in relation to MNP toxicity. When localized drug delivery is involved, reduced systemic side effects are achieved, but precise drug release control could be hard to conduct. Regarding immunotherapy, unexpected immune reactions may occur alongside MNPs’ toxicity. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Manescu, V.; Dumitru, A.-V.; Antoniac, A.; Antoniac, I.; Paltanea, G.; Zeca, E.-C.; Gherghe, M.; Nemoianu, I.V.; Streza, A.; Paun, C.; et al. Magnetic Hydrogels as a Treatment for Oncological Pathologies. J. Funct. Biomater. 2025, 16, 414. https://doi.org/10.3390/jfb16110414
Manescu V, Dumitru A-V, Antoniac A, Antoniac I, Paltanea G, Zeca E-C, Gherghe M, Nemoianu IV, Streza A, Paun C, et al. Magnetic Hydrogels as a Treatment for Oncological Pathologies. Journal of Functional Biomaterials. 2025; 16(11):414. https://doi.org/10.3390/jfb16110414
Chicago/Turabian StyleManescu (Paltanea), Veronica, Adrian-Vasile Dumitru, Aurora Antoniac, Iulian Antoniac, Gheorghe Paltanea, Elena-Cristina Zeca (Berbecar), Mirela Gherghe, Iosif Vasile Nemoianu, Alexandru Streza, Costel Paun, and et al. 2025. "Magnetic Hydrogels as a Treatment for Oncological Pathologies" Journal of Functional Biomaterials 16, no. 11: 414. https://doi.org/10.3390/jfb16110414
APA StyleManescu, V., Dumitru, A.-V., Antoniac, A., Antoniac, I., Paltanea, G., Zeca, E.-C., Gherghe, M., Nemoianu, I. V., Streza, A., Paun, C., & Gradinaru, S. (2025). Magnetic Hydrogels as a Treatment for Oncological Pathologies. Journal of Functional Biomaterials, 16(11), 414. https://doi.org/10.3390/jfb16110414

