Cancer IX
Cancer IX
A R T I C L E I N F O A B S T R A C T
Keywords:                                                    Cancer cures remain limited with conventional treatments due to tumor microenvironment (TME)-driven
Cancer                                                       resilience and systemic toxicity. Magnetic hyperthermia therapy (MHT), which utilizes magnetic nanoparticles
Magnetic hyperthermia therapy                                (MNPs) to convert alternating magnetic field (AMF) energy into localized heat, has emerged as a minimally
History
                                                             invasive and spatially precise strategy for tumor ablation. This review critically examines the evolution of MHT
Mechanism
                                                             from its historical roots to cutting-edge combinatorial strategies. The unique advantages of MHT over other
Material
Combination                                                  hyperthermia modalities are firstly analyzed, highlighting its ability to synergize with the hypoxic and acidic
                                                             TME for enhanced tumor cell susceptibility. In addition, the pathophysiological mechanisms of cell death
                                                             induced by MHT were summarized in detail. Next, MHT-optimized nanomaterials are systematically classified
                                                             based on their heating efficiency, biodistribution, and functionalization strategies for tumor targeting. A key
                                                             focus lies in elucidating the multimodal therapeutic synergy between MHT and established oncology treatments:
                                                             (photothermal therapy [PTT], radiotherapy, chemotherapy, immunotherapy) and analyzed the clinical trans
                                                             lational bottlenecks of each combination. Finally, the review delineates the translational roadmap of MHT by
                                                             addressing key bottlenecks in clinical adoption, including AMF device standardization, long-term MNP biosafety,
                                                             and scalable nanomanufacturing. This work provides a nano-biomaterials perspective to guide the rational
                                                             design of next-generation MHT platforms for precision oncology.
 * Corresponding author. Department of Pathology from College of Basic Medicine, and Department of Clinical Pathology Laboratory of Pathology Diagnostic
Center, and Molecular Medicine Diagnostic & Testing Center, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong Distinct, Chongqing, 400016, PR China.
 ** Corresponding author. Department of Orthopedics, Chongqing Traditional Chinese Medicine Hospital, The First Affiliated Hospital of Chongqing College of
Traditional Chinese Medicine, No. 6 Panxi Seventh Branch Road, Jiangbei District, Chongqing 400021, PR China.
   E-mail addresses: doctorliang51@163.com (B. Liang), csyxk@126.com (K. Yu).
https://doi.org/10.1016/j.mtbio.2025.102070
Received 20 April 2025; Received in revised form 3 July 2025; Accepted 7 July 2025
Available online 9 July 2025
2590-0064/© 2025 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC license (http://creativecommons.org/licenses/by-
nc/4.0/).
L. Xiong et al.                                                                                                                   Materials Today Bio 33 (2025) 102070
their applicability, increases patient burden, and hampers treatment of              core limitations of both conventional therapies and traditional MHT,
deep-seated or multiple lesions. Furthermore, large implants can also                leveraging the unique superiority conferred by AMF and MNPs.
lead to issues like inhomogeneous heat distribution.                                     As an external energy source, AMF exhibits strong penetration
    In this context, nanotechnology-mediated thermal therapy, particu               capability with minimal attenuation in biological tissues, making MHT
larly nano-MHT, has emerged as a significant breakthrough, demon                    suitable for treating deep-seated tumors—a stark contrast to the limited
strating distinct advantages. By applying an external AMF, MNPs                      tissue penetration of laser and microwave thermotherapy [10,11].
localized within the tumor transform electromagnetic energy into                     Concurrently, the physical thermogenesis mechanism of MHT enables
thermal energy. This effectively elevates the intratumoral temperature               repeatable treatments, significantly reducing patient discomfort and
to cytotoxic levels (typically 42–46 ◦ C), selectively destroying cancer             facilitating potential future outpatient tumor management. Compared to
cells [9]. Among various nano-thermal techniques (laser, microwave,                  implanting large particles, NPs can be administered via intravenous or
radiofrequency, ultrasound), AMF-mediated nano-MHT has garnered                      local injection in a minimally invasive manner, accessing diverse tumor
considerable attention in recent years. It systematically addresses the              locations while generating heat more uniformly and efficiently.
Fig. 1. We introduce MHT from various aspects in this review. It includes the history, mechanisms and superiorities, and focuses on the latest advances in materials
and the advantages of combinational therapy for tumor killing.
                                                                                 2
L. Xiong et al.                                                                                                           Materials Today Bio 33 (2025) 102070
Functionalized MNPs can be engineered for active tumor targeting,               in a rat model [18], further demonstrated the potential of MHT in
concentrating heat production within the tumor, minimizing damage to            medical device applications, gradually expanding its scope from basic
surrounding healthy tissue, and thereby reducing systemic toxicity [12].        materials to practical medical uses. Furthermore, from 2003 to 2005,
Additionally, nanotechnology allows for precise control of heating by           glioblastoma clinical trial at the University of Berlin confirmed the
adjusting parameters such as NPs composition, size, shape, and the              safety of MF-mediated magnetic induction hyperthermia (MIH)
characteristics of the applied AMF, meeting diverse therapeutic needs           [19–21], also marking an acceleration of clinical translation.
[9,13]. Moreover, nano-MHT synergistically enhances the efficacy of                 By 2006, surface-modified SPIONs had been applied to drug delivery,
chemotherapy/radiotherapy/immunotherapy: thermal energy remotely                immunoassays, and transarterial chemoembolization of hepatocellular
activates drug release and amplifies chemotherapeutic cytotoxicity by           carcinoma (TACE) [22], and the material platform was further expanded
increasing tumor cell membrane permeability, promoting trans                   in 2008 with nanothermal therapy technology and Ferucarbotran-based
membrane transport, and improving intratumoral blood circulation.               arterial embolic hyperthermia (AEH) [23,24]. Current research on
Collectively, MHT demonstrates seven core characteristics: 1.                   MNPs for MHT focuses on optimizing the synthesis pathway, structural
Non-invasiveness; 2. Repeatability; 3. Localization; 4. Deep-tissue             morphology and surface modifications to enhance the rate of heat pro
penetration; 5. Effectiveness; 6. Personalization and 7. Combinatorial          duction, monodispersity, cyclic half-life, targeting accuracy and
synergy, positioning it as a promising adjunct to conventional cancer           biocompatibility [25,26].
therapies.                                                                          Currently, clinical trials of magnetothermal therapy are being con
    This review chronicles the evolution of MHT, spotlighting its tumor-        ducted primarily in prostate cancer and glioblastoma multiforme (GBM)
selective mechanisms driven by TME. We critically evaluate state-of-the-        patients (Table 1). For instance, three-stage therapy for prostate cancer:
art nanomaterials optimized for heating efficiency, biodistribution, and        (1) CT-based tumor assessment; (2) integrated tumor boundary and
functionalization. Furthermore, we dissect multimodal synergies be             vascular system for particle dose/deposition calculation planning; and
tween MHT and conventional therapies—PTT, radiotherapy, chemo                  (3) ultrasound-guided transurethral injection of 12.5 mL of iron oxide
therapy, and immunotherapy—revealing their TME-remodeling actions               solution (prostate volume of 35 mL), followed by radiofrequency MHT
and translational hurdles. We have summarized each section (history,            treatment [27]. Similarly, the MHT combined with radiotherapy
mechanism, superiorities, materials and combination) for the theme of           regimen for recurrent GBM achieved prolonged survival without side
MHT in Fig. 1. By bridging nanomaterial innovation with clinical im            effects through tumor resection, stereotactic SPION delivery, and
peratives, this work charts a roadmap for next-generation MHT plat             MRI/CT thermal border modeling [28]. These trials corroborate the
forms in precision oncology.                                                    synergistic potential of MHT, but technical bottlenecks remain.
                                                                                    High requirements for the development of magneto-thermal devices
2. History of MHT materials and clinical development of MHT                     (balancing safety and efficacy): the most used safety standard for AMF is
                                                                                the Brezovich limit (H0f ≤ 4.8 × 108 Am− 1s− 1) [29], but it is often
    Over the past seventy years, MHT has undergone significant devel           difficult to achieve the target thermal efficacy at this limit. Bellizzi’s
opment, mainly driven by the continuous optimization of magnetic                team proposes raising this threshold to 1.9 × 109 Am− 1s− 1 for the
fluids (MFs) and biomaterials (Fig. 2). In 1960, a crucial milestone was        treatment of brain tumors, or to halve the nanoparticle dose [30].
set when NASA first utilized iron oxide nanoparticles (IONPs) [14].                 NanoTherm® (aminosilane-coated Fe3O4 nanoparticles), approved
Their biocompatibility and subsequent FDA approval laid the founda             as an adjuvant for GBM radiotherapy, prolonged median survival by 4.2
tion for MHT, making them a cornerstone material in this field [15].            months but was limited by a low specific uptake rate (738-985 pH-
Building on this, in 1985, Lilly et al. developed nickel-copper thermal         m2⋅kg− 1), which required a high iron concentration of 112 mg mL− 1 to
seeds with a Curie temperature of 50 ◦ C for mesenchymal implant                trigger MRI artifacts and nanoparticle leakage. Tumor osmotic pressure
thermotherapy [16], expanding the range of MHT applications. Then, in           gradients exacerbate NPs redistribution, leading to off-target heating
2002, Rehman’s team made a breakthrough by creating ferromagnetic               [31–33].
self-regulating heat seeds [17]. This innovation improved the tempera              In addition, mild HT (39–42 ◦ C) induces overexpression of heat
ture control of MHT, enhancing its safety and effectiveness. In 2005,           shock proteins (HSPs), triggering transient thermoresistance [34,35].
Shoi’s team developed a magnetic stent for bile duct stenosis. Their            Although the minimally invasive and precise nature of MHT has clinical
research, which validated the safe heating of up to 45 ◦ C at 157 kHz AMF       advantages, the above limitations require mechanistic studies to
                                                                            3
L. Xiong et al.                                                                                                                      Materials Today Bio 33 (2025) 102070
Table 1
Summary of clinical trials of MHT for tumor treatment.
  Trial      Cancer type           Patient   Injection       Adjuvant        AMF           Maximum                  SAR     Treatment results        Year    Reference
  phase                            number    route           therapy         strengths     temperature &
                                                                                           Temperature
                                                                                           measurement method
  I          Glioblastoma          14        Intratumoral    Radio/          3.8 to 13.5   44.6 ◦ C(median)         –       MHT was well             2003    [249]
             multiforme                                      chemotherapy    kAm− 1        (Invasive                        tolerated.
                                                                             (median:      temperature probing              Only mild side effects
                                                                             8.5)          with thermal probes)             were observed
  I          Prostate carcinoma    10        Transperineal   –               H = 100       48.5 ◦ C (Invasive       288     The patients were able   2005    [250]
                                                                             kHz, f = 4    temperature probing      W       to tolerate an AMF
                                                                             kAm− 1        with thermal probes)     kg− 1   strength of 4–5 kAm-1.
                                                                                                                            Serum PSA levels
                                                                                                                            decreased in 8
                                                                                                                            patients after
                                                                                                                            treatment, and the
                                                                                                                            mean duration of PSA
                                                                                                                            control was 5 months
                                                                                                                            (3–8 months).
  I          Chondrosarcoma,       22        Transperineal   Radio/          –             39.5 ◦ C (Intratumoral   130     Tolerable AMF            2006    [19]
             sarcinoma cervical/                             chemotherapy                  temperature              W       strengths:
             prostate/ovarian/                                                             monitoring via fiber-    kg− 1   Pelvis:3.0–6.0 kAm− 1
             rectal carcinoma                                                              optic probes)                    Chest and neck
                                                                                                                            area:7.5 kAm− 1
                                                                                                                            Head:>10.0 kAm− 1
  II         Glioblastoma           3        Intratumoral    Radio/          H = 100       Patient 1:49.5 ◦ C/      –       Low uptake of            2009    [244]
             multiforme                                      chemotherapy    kHz,f =       Patient 2 dead before            aggregated particles
                                                                             2.5–18        MHT/Patient                      in glioblastoma cells.
                                                                             kAm-1         3:65.5 ◦ C                       High uptake of
                                                                                           (Intratumoral                    particles by
                                                                                           temperature                      macrophages.
                                                                                           monitoring via fiber-            No clinically adverse
                                                                                           optic probes)                    effects.
  II         Glioblastoma          66        Intratumoral    Radio/          H = 100       51.2 ◦ C (median)        –       The median overall       2011    [28]
             multiforme                                      chemotherapy    kHz,f =       (Intratumoral                    survival was 13.4
                                                                             2–15 kAm-     temperature                      months.
                                                                             1
                                                                                           monitoring via
                                                                                           thermal probes)
                                                                                 4
L. Xiong et al.                                                                                                           Materials Today Bio 33 (2025) 102070
                                                                            5
L. Xiong et al.                                                                                                             Materials Today Bio 33 (2025) 102070
apoptosis in macrophages. HT also induces ER stress by accumulating               3.5. HT promotes cuproptosis of tumor cells
misfolded proteins, activating the intrinsic ER apoptosis pathway
(Fig. 4). Collectively, HT engages multiple apoptotic cascades, under                Recent advances in cell death research have identified cuproptosis, a
scoring its therapeutic potential. By delineating these mechanisms, re           copper-dependent cell death mechanism distinct from apoptosis, nec
searchers can optimize HT-based strategies to selectively target cancer           roptosis, pyroptosis, and ferroptosis [61,62]. This process, driven by
cells while sparing healthy tissues [53].                                         intracellular copper accumulation, disrupts mitochondrial function by
                                                                                  inducing aggregation of tricarboxylic acid (TCA) cycle proteins and
                                                                                  destabilizing iron-sulfur (Fe-S) cluster proteins, culminating in proteo
3.3. HT increases oxidative stress in tumor cells
                                                                                  toxic stress and cell death [63]. Elevated copper levels, frequently
                                                                                  observed in tumor tissues and serum of cancer patients, position
    Heat stress is a well-documented inducer of oxidative stress in cells,
                                                                                  cuproptosis as a promising therapeutic target for malignancies such as
primarily through increased generation of reactive oxygen species
                                                                                  breast cancer, lung cancer, and melanoma.
(ROS). ROS, including superoxide anion (O2− ), hydrogen peroxide
                                                                                      Current strategies to induce cuproptosis focus on enhancing mito
(H2O2), hydroxyl radical (⋅OH), nitric oxide (⋅NO), peroxynitrite
                                                                                  chondrial copper bioavailability through: (1) direct delivery of copper
(ONOO− ), and lipid peroxides (LPO), are continuously produced during
                                                                                  ions or ionophores [64,65], (2) inhibition of copper efflux transporters
electron transfer reactions in aerobic metabolism. Under physiological
                                                                                  (ATP7A/ATP7B) [66,67], and (3) depletion of GSH, a key copper
conditions, ROS levels are tightly regulated by antioxidants such as su
                                                                                  chelator [68,69]. However, intrinsic barriers—including the Warburg
peroxide dismutase (SOD), catalase, glutathione (GSH), peroxidases,
                                                                                  effect-mediated suppression of mitochondrial respiration and hetero
and vitamins to maintain redox homeostasis. However, during heat
                                                                                  geneous expression of cuproptosis regulators like ferredoxin 1 (FDX1)
stress [54], intracellular oxygen levels rise due to xanthine oxidase
                                                                                  and lipoylated proteins—limit therapeutic efficacy across cancer types
activation and mitochondrial electron transport chain (ETC) activity
                                                                                  (Fig. 4) [70].
[55], leading to excessive ROS accumulation. This oxidative surge
                                                                                      The interplay between HT and cuproptosis remains underexplored.
damages cellular components and activates multiple cell death path
                                                                                  While HT modulates mitochondrial oxidative phosphorylation
ways. Notably, heat stress downregulates SOD1 (CuZnSOD) expression
                                                                                  (OXPHOS), a process linked to copper sensitivity, its effects vary by
and activity, amplifying ROS production [56]. Uncontrolled ROS and
                                                                                  cancer lineage. In ovarian and uterine cancers, HT reduces glycolysis
reactive nitrogen species (RNS) initiate free radical chain reactions that
                                                                                  while enhancing OXPHOS to sustain ATP production, whereas colorectal
oxidize proteins, lipids, polysaccharides, and DNA, with mitochondria
                                                                                  cancer exhibits increased glycolysis and spare respiratory capacity
being both the primary ROS source and target. Mitochondrial antioxi
                                                                                  under thermal stress [71]. Notably, HT-driven OXPHOS amplification
dant failure destabilizes free radical equilibrium, impairing ETC func
                                                                                  sensitizes tumors to cuproptosis, as demonstrated by Lin et al. [72], who
tion, ATP synthesis, respiration coupling, and structural integrity of
                                                                                  engineered a thermoresponsive nanomedicine (DIE) co-encapsulating
biomolecules. Consequently, mitochondrial dysfunction disrupts
                                                                                  indocyanine green (ICG) and elesclomol (ES) within disulfide-rich
cellular homeostasis, accelerating cell death (Fig. 4).
                                                                                  dextran copolymers. Near-infrared (NIR) laser activation of DIE
    While heat-induced oxidative damage affects both normal and ma
                                                                                  reduced GSH levels, elevated cytotoxic Cu2+ concentrations, and
lignant cells, cancer cells often resist apoptosis, driving therapeutic in
                                                                                  amplified FDX1-mediated lipoylated protein aggregation, thereby
terest in non-apoptotic programmed cell death (PCD) mechanisms.
                                                                                  potentiating cuproptosis in melanoma (Fig. 5). This approach synergized
Identified non-apoptotic PCD pathways include necroptosis, autophagy,
                                                                                  with PD-L1 checkpoint blockade to suppress tumor growth and metas
ferroptosis, pyroptosis, and cuproptosis. NPs further enhance thera
                                                                                  tasis via immunogenic cell death.
peutic synergy by augmenting chemo-/radio-/immune-/HT-mediated
                                                                                      Although MHT itself does not directly regulate the cuproptosis
PCD induction. NPs themselves can trigger apoptotic and non-apoptotic
                                                                                  pathway, as research clarifies the role of HT (a downstream effect of
death, diversifying treatment modalities. In summary, heat stress exac
                                                                                  MHT) in inducing cuproptosis, integrated strategies combining thermal
erbates oxidative damage, activating intrinsic apoptotic and alternative
                                                                                  modulation, copper biology, and immunotherapy are emerging as
PCD pathways. Elucidating ROS dynamics and their cellular impacts is
                                                                                  transformative tools in oncology.
pivotal for designing targeted therapies that selectively eliminate cancer
cells while preserving normal tissue [57].
                                                                                  3.6. HT promotes ferroptosis of tumor cells
3.4. HT promotes necroptosis of tumor cells                                           HT synergistically induces ferroptosis in tumor cells by regulating
                                                                                  iron metabolism and redox balance, with its underlying mechanisms
    Necroptosis is defined as a novel cell death that has some dis               closely tied to the microenvironment-responsive properties of iron-
tinguishing features compared to other cell death types. The primary              containing nanomaterials. This primarily involves iron ion release trig
causes of necroptosis are widely recognized to be the pharmacological             gered by acidic microenvironments and activation of the Fenton reac
blocking or genetic removal of caspases, which, when cells are unable to          tion [73].
initiate apoptosis, can serve as an alternative "safety net" within the               Iron-based nanomaterials (e.g., Fe3O4 mesocrystals, iron MOFs) un
spectrum of PCD [58]. And targeting necroptotic cell death in cancer              dergo structural dissociation and release free iron (Fe2+/Fe3+) under the
cells has potential therapeutic benefits. Necroptosis, distinct from              acidic conditions (pH 6.5–6.9) of the TME or the low-pH environment
apoptosis, constitutes an inflammatory type of PCD. The inflammation              (pH 4.5–5.5) of lysosomes. HT further accelerates this process: localized
triggered by necroptosis has the potential to enhance immune-mediated             temperature elevation enhances the degradation kinetics of nano
mechanisms against cancer [59]. There are different pathways by which             materials, promoting greater accumulation of iron ions in the cytoplasm.
NPs trigger necrosis in cells (Fig. 4). The predominant mechanism in              Released iron ions drive ferroptosis through two main pathways: 1.
necrotic cell death is the pro-oxidant pathway, which involves the                Amplified oxidative damage via the Fenton reaction: Fe2+ directly par
production of ROS. This can result in a decrease in mitochondrial                 ticipates in the classical Fenton reaction with H2O2 to generate ⋅OH:
membrane potential and DNA damage, ultimately steering the cell to               Fe2++H2O2→Fe3++OH− +⋅OH. ⋅OH is one of the most potent ROS spe
wards a necrotic demise [60]. Additionally, another pathway by which              cies (oxidation potential 2.8 V), capable of directly attacking poly
NPs induce necrosis involves the disruption of lysosomal structure due            unsaturated fatty acids in cell membrane phospholipids to initiate a lipid
to the rapid lysosomotropic degradation of NPs, leading to the release of         peroxidation chain reaction and form phospholipid hydroperoxides
toxic contents into the cytosol and culminating in necrotic cell death            (Fig. 4) [74]. Fe3+, the primary form of iron in circulation, binds to
[57].                                                                             transferrin (TF). It is transported into cells with the help of the
                                                                              6
L. Xiong et al.                                                                                                        Materials Today Bio 33 (2025) 102070
Fig. 5. Thermal stimulation boosts ES-induced cuproptosis via increased FDX1 expression and mitochondrial respiration. Reproduced with permission [72].
Copyright 2025, Wiley.
                                                                           7
L. Xiong et al.                                                                                                                   Materials Today Bio 33 (2025) 102070
Fig. 6. Schematic drawing of (A) a hysteresis loop of a ferromagnetic material and (B) typical plot of a superparamagnetic material. Reproduced with permission [7].
Copyright 2020, Wiley.
                                                                                 8
L. Xiong et al.                                                                                                                       Materials Today Bio 33 (2025) 102070
5. Advantages of MHT compared with other types of HT                                    these systems permits a unified theragnostic approach, blending di
                                                                                        agnostics and therapy [75,86]. Additionally, static AMF can trigger the
    The utility of MF is based on their unique responsive nature. This                  release of therapeutic agents, nucleic acids, and antioxidant enzymes
encompasses the use of magnetic guidance under a static AMF, the in                    [87]. MNPs in an oscillating or AMF generate heat due to hysteresis loss
crease in temperature under an oscillating AMF, or a combination of                     and/or Néel relaxation, which is widely used for targeted heating and
both in alternating applications. These methods can effectively treat                   controlled drug release within tumors [88,89]. The thermal bystander
deeply located diseases, penetrating biological barriers such as tissues,               effect ensures that the injected MF is evenly distributed for uniform
bones, or blood vessels without obstruction, thanks to the high pene                   heating and expanded coverage. In complex tumor tissues, precise
tration capability of AMF. Furthermore, the integration of MRI within                   control over the distribution and heating range of the MF is achievable
Table 2
This table summarizes the main advantages and disadvantages of different current hyperthermia treatments, tissue penetration depth, thresholds and clinical
application status.
  Type              Heat technology       Major advantages              Major disadvantages           Tissue           Threshold     Clinic application          Reference
                                                                                                      penetration      level
                                                                                                      depth
  Magnetic          Alternating           • Non-invasive                • Side effects and toxicity   Unlimited        7Tesla        Still in the pre-clinical   [251–253]
   hyperthermia     magnetic field        • Easy accumulation of          induced by particle                                        research stage
                                            magnetic particles            accumulation                                               Currently, only
                                          • Spatiotemporal control      • High cost                                                  Germany approved the
                                          • Insensitive to the          • Complexity                                                 use of MHT as an
                                            surrounding medium          • Large facilities                                           adjuvant therapy with
                                          • Hot bystander effect        • Limited information of                                     RT
                                          • Curie temperature             MIONP distribution in
                                          • Intracellular                 tissue
                                            hyperthermia                • Limited and invasive
                                          • No penetration depth          thermometry
                                            limit
                                          • Repeated treatment
                                          • Treatment of brain
                                            tumors through the
                                            blood-brain barrier
                                                                                                                                 2
  Photothermal      Near-infrared (NIR)   • Low cost                    • Limited invasive for the    3 cm             10J⋅cm−       PTT agents have not         [254,255]
    therapy(PTT)    laser                 • Easily tuned                  deep zone tissue                                           yet been tested in large
                    photoabsorbers、       • High specificity              penetration                                                clinical trials; laser
                    NIR                   • Microinvasive               • Harmful UV                                                 ablation without PTT
                                          • Accurate space-time         • Inconsistent responses                                     agents has been used
                                            selectivity                   to light                                                   clinically
                                          • Low adverse reactions to
                                            normal tissues
  Microwave         Probes that make      • The highest rate of bulk    • Frequent pain and           3–7 cm           915 MHz       Commonly used               [256]
   hyperthermia     energy from             tissue heating                discomfort                                                 clinical hyperthermia
                    microwaves            • Relatively unaffected by    • Heat dose and                                              technology
                                            tissue charring               positioning is difficult                                   Examples:superficial
                                          • The ability to create         to accurately                                              and deep tumors、
                                            ablations that can cover    • Heating of normal                                          chronic strain of soft
                                            most tumors up to 5 cm        tissue                                                     tissue bones and joints
                                            in diameterin the           • Blood perfusion and
                                            shortest amount of time       vascular heat sink
                                            (typically 4–10 min)        • Electromagnetic and
                                                                          thermal
                                                                          inhomogeneities in and
                                                                          around the tumor
  Radiofrequency    Radio wave            • Microinvasive               • Eschar or dry tissue        5 cm             2.45 GHz      The most commonly           [257]
   ablation                               • Partially curative effect     affects the current                                        used clinical
                                            on some diseases (such        conduction so that the                                     hyperthermia
                                            as liver tumors less than     scope of action is less                                    technology
                                            5 cm in diameter)             than microwave                                             Examples: early liver
                                          • Multiple treatment can        ablation                                                   cancer、early non-
                                            be repeated                 • Positioning means still                                    small cell lung cancer
                                                                          need to be optimized                                       benign thyroid tumors
                                                                        • Tissue damage risk
                                                                        • Uncomfortable for
                                                                          patients
  Ultrasound        Ultrasound            ⋅ High penetration depth      ⋅ Hard to achieve             10–15 cm         Exposure      The clinical                [258–263]
    hyperthermia                          ⋅ Easy to adjust                homogeneous exposure        Intensity: 100   time:         transformation of
                                          ⋅ Spatiotemporal control        within large zones          mW cm− 2         10min         ultrasound-mediated
                                          ⋅ Low cost                    • High reflection at air      (Diagnosis);                   nanomedicines
                                          ⋅ Capability of focused         and bone interface          3W⋅cm− 2                       remains to be fully
                                            heating                     • High absorption in bone     (Therapy)                      studied
                                          ⋅ Integrating diagnostic                                    Mechanical
                                            imaging with                                              index: 0.3
                                            therapeutic function
                                          • Non-invasiveness and
                                            safety
                                                                                    9
L. Xiong et al.                                                                                                             Materials Today Bio 33 (2025) 102070
through multi-point injection, selectively targeting tumors while sparing         through ROS generation, position ultrasound as a multifunctional
normal tissues. The MF’s Curie temperature property allows for auto              therapeutic platform.
matic temperature control and maintenance, which is crucial for deep
tumor thermotherapy. These features set magnetic thermotherapy apart              6. Magnetothermal related materials
from other thermal treatments. However, its application is typically
limited to accessible tumor nodules and not suitable for metastatic or                The magnetothermal performance of MNPs is critically determined
disseminated tumors. The use of magnetic guidance is complex,                     by their structural morphology (size, shape, crystallinity) and surface
requiring precisely focused and tissue-penetrating external AMF.                  modifications (coatings, functionalization). Smaller NPs (<50 nm)
Real-time accurate temperature measurement of the target HT tissue                exhibit enhanced tumor penetration and renal clearance efficiency,
remains a technical challenge [88]. Current research focuses on opti             while larger particles (>100 nm) can be more advantageous in targeting
mizing magnetic and irradiation technologies. We summarize the pros               the tumor associated macrophages (TAMs) in certain applications [98].
and cons of magnetic heating versus other thermal therapies and their             Regarding shape, non-spherical architectures (e.g., rods, cubes, plates,
clinical applications in Table 2.                                                 and needles) exhibit distinct magnetic properties compared to spherical
    PTT is an emerging cancer treatment modality that utilizes near-              particles due to anisotropic demagnetizing factors, which influence
infrared (NIR) laser-absorbing agents to selectively destroy malignant            magnetic moment relaxation dynamics and hysteresis losses. For
cells through localized hyperthermia upon NIR irradiation [90]. PTT               example, rod-shaped MNPs with high aspect ratios show enhanced
shows potential for eradicating primary tumors and locoregional lymph             magnetic anisotropy, leading to higher hysteresis losses and signifi
node metastases, effectively addressing early-stage metastatic progres           cantly increased SAR values. Cube-shaped particles, with their sharp
sion. Furthermore, it exhibits synergistic potential with systemic ther          edges and corners, can further modulate magnetic dipole-dipole in
apies for managing disseminated metastases [91]. However, critical                teractions between particles, potentially inducing collective magnetic
challenges persist, such as incomplete ablation of thermotolerant cells in        behaviors that enhance heat generation under AMF. Conversely,
deep-seated metastases and the necessity to maintain intratumoral                 spherical MNPs offer superior colloidal stability and reduced nonspecific
temperatures above 50 ◦ C for optimal efficacy [92]. Tissue attenuation           cell uptake due to their symmetrical geometry, making them favorable
of NIR wavelengths limits penetration depth, often requiring increased            for systemic circulation and tumor accumulation via the EPR effect.
laser intensity or prolonged exposure, which risks collateral thermal             Surface engineering with polymers (e.g., PEG, chitosan) or inorganic
damage to adjacent healthy tissues. Current research prioritizes nano            shells (e.g., silica, gold) improves colloidal stability, reduces immune
particle engineering and dosimetry optimization to enhance therapeutic            recognition, and prolongs circulation half-life. For instance, PEG can
specificity.                                                                      reduce the overall adsorption of plasma proteins and prevent the ag
    Thermal ablation techniques, particularly radiofrequency ablation             gregation of MNP, thereby helping MNP escape from mononuclear
(RFA) and microwave ablation (MWA), constitute mainstream clinical                phagocyte system (MPS). Moreover, studies have found that as the
interventions. These modalities achieve cytotoxic temperatures                    molecular weight of PEG increases from several thousand to several
exceeding 50 ◦ C, serving as either standalone therapies or adjuvants to          hundred thousand, the circulation time of MNP in the blood can be
conventional treatments. MWA operates at high power (typically 4–10               extended from 30 min to 24 h [98]. Crystalline anisotropy and defect
min), enabling rapid volumetric heating less impeded by tissue desic             density directly govern magnetic susceptibility and SAR, with
cation and capable of ablating lesions ≤5 cm with margins [138].                  single-domain superparamagnetic NPs demonstrating optimal heating
Clinical trials demonstrate MWA’s efficacy across diverse anatomical              efficiency under AMF [99].
sites, including bone, pancreas, and prostate. Nevertheless, technical                The metabolic pathways of MNPs depend on size and surface prop
limitations persist inconsistent thermal dosing due to antenna design             erties: renal clearance (<15 nm particles), hepatobiliary excretion, or
constraints, procedural discomfort, and heat sink effects from vascular           MPS uptake. Compared to earlier nanomaterials, advancements in
structures. Although early innovations advanced interstitial ablation             nanoengineering have significantly enhanced the metabolic profiles and
protocols, the complexity of achieving homogeneous tumor heating has              biosafety of MNPs. Traditional inorganic MNPs (e.g., SPIONs) faced
restricted MWA’s oncological applications.                                        challenges such as hepatic/splenic accumulation, iron ion leakage-
    RFA remains the gold standard for solid tumor ablation, employing             induced oxidative stress, and long-term retention risks. Modern sur
high-frequency alternating currents (350–500 kHz) to induce ionic                 face engineering strategies—including PEGylation, zwitterionic coat
friction and thermal coagulation (60–120 ◦ C) [93]. The ablated volume            ings, and biodegradable polymer encapsulation (e.g., PLGA, dextran)—
depends on both local joule heating and convective cooling from blood             enable controlled biodegradation and improved renal clearance.
flow. Following FDA approval for lung tumors (2007) and NCCN                          The nanosystems currently reported for MHT biomedicine can be
guideline inclusion (2009), RFA gained recognition as a minimally                 generally classified into three categories: inorganic, organic, and
invasive alternative to resection for lesions <5 cm, particularly in pa          organic-inorganic hybrid micro-/nanoplatforms. The disciplines of ma
tients with comorbidities contraindicating surgery [94]. Advantages               terials engineering and synthetic chemistry exhibit distinct differences,
include percutaneous applicability under local anesthesia, repeatability          which in turn lead to varied biomedical applications of these multi
for multifocal/recurrent tumors, and reduced iatrogenic seeding risk.             functional NPs (Fig. 7).
However, RFA efficacy diminishes in highly desiccated tissues compared
to MWA, and procedural pain remains a patient concern.
    Ultrasound-based therapies address penetration limitations of                 6.1. Inorganic nanomaterials
external beam modalities while avoiding ionizing radiation. Adjustable
parameters (frequency, duty cycle, duration) enable depth-specific en                MNPs are essential counterparts of magnetic nanocomposites. They
ergy deposition (up to tens of centimeters) with minimal off-target ef           can be classified into magnetic alloy NPs (eg iron, nickel, and cobalt) and
fects [95]. High-intensity focused ultrasound (HIFU) achieves thermal             magnetic metal oxide NPs (eg iron oxides, nickel oxide, and lanthanum
ablation, whereas pulsed protocols facilitate mechanical tissue frac             strontium manganite). Table 3 encapsulates a review of the primary pros
tionation. Low-intensity ultrasound combined with microbubbles en                and cons associated with various commonly employed inorganic mate
hances drug delivery via cavitation-mediated vascular permeability                rials in MHT [100].
[96]. Emerging strategies employ sonosensitizers activated by ultra
sound to induce localized sonochemical cytotoxicity, forming the basis            6.1.1. Monocomponent MNPs
of sonodynamic therapy (SDT) [97]. These multifunctional applications,
including diagnostics, targeted drug release, and tumor eradication               6.1.1.1. Fe, Ni, Co-based MNPs. Iron nanoparticles (FeNPs) exhibit
                                                                             10
L. Xiong et al.                                                                                                                Materials Today Bio 33 (2025) 102070
Fig. 7. Schematic diagram of several classic and latest magnetothermal related materials introduced in this article.
good physical and chemical stability, less expensive, and environmen                easy scale-up and high activity. However, although single-atom NPs
tally safe. In prior studies, iron carbonyl [Fe(CO)5] was decomposed in              have broad application prospects in many fields, there are still many
the presence of oleic acid to yield mono-dispersed FeNPs [101].                      challenges. One is that the methods and techniques for obtaining higher
Considering the susceptibility of iron NPs to oxidation, a straightforward           purity and controllable single-component NPs are not mature enough
aqueous-phase        synthesis     was     employed,      utilizing     poly         and need to be further improved. In addition, the stability of single-atom
(N-vinylpyrrolidone) (PVP) to confer antioxidation properties to the                 NPs is also an important issue. Due to the high surface energy and sol
metal surface [102]. Nickel (Ni) NPs with uniform size distribution were             ubility of single-atom particles, they are often prone to aggregation and
synthesized by reducing Ni(acac)3 in the presence of hexadecylamine                  loss of activity in biological applications. Therefore, researchers are
(HDA), resulting in an average particle size of 3.7 nm [103]. Cobalt (Co)            developing some methods to enhance the stability of single-atom NPs,
NPs with a size of approximately 26 nm were produced using a bulky                   such as surface modification and coating to reduce their interaction with
trialkyl phosphine as a reducing agent, whereas larger NPs, with sizes up            the external environment.
to 71-11 nm, were formed in the presence of a less bulky trialkyl
phosphine, demonstrating the role of trialkyl phosphine as a coordi                 6.1.1.2. Superparamagnetic gold-nanoparticles (SPAuNPs). Beyond the
nating surfactant interacting with the neutral metal surface sites [104].            aforementioned single-component magnetic nanostructures, there have
Including but not limited to the above single component, NPs have many               been reports in recent years of a novel cancer diagnostic agent that
unique properties and applications. Due to its small size and high spe              harnesses the gold-magnetic effect. SPAuNPs, have been developed by
cific surface area with organisms, it can be used as an efficient catalyst to        synthesizing them on a viral capsid particle engineered to target tumor
accelerate the chemical reaction rate. Compared with natural enzymes,                cell receptors (TCR). This innovation addresses the limitations
nanozymes have obvious advantages such as low cost, high stability,                  commonly associated with other NPs. For instance, SPIONs are hindered
                                                                                11
L. Xiong et al.                                                                                                                    Materials Today Bio 33 (2025) 102070
                                                                                  12
L. Xiong et al.                                                                                                                              Materials Today Bio 33 (2025) 102070
Table 4
Summary of some ferrite materials in recent years.
  NPs type          Size    Cancer cell    Injection               Injection        AMF             SAR       Maximum         Results                        Year    Reference
                    (nm)    line           dose                    route            strengths                 temperature
  Iron oxide NPs    18      4T1            50 μL of 0.5            Intratumoral     –               –         43 ◦ C          The improved targeting and     2019    [266]
    (IOs)                                  mg kg− 1                                                                           delivery uniformity enables
                                                                                                                              more effective MHT cancer
                                                                                                                              ablation than otherwise
                                                                                                                              identical, nontargeting IOs.
  Co–Fe NPs         17      A431           50 μL 14 mg             Intratumoral     105 kHz,20      400 Wg-   –               Complete tumor regression      2020    [96]
                    ±2                     mL− 1                                    kAm− 1          1
                                                                                                                              and improved overall
                                                                                                                              survival in an in vivo
                                                                                                                              murine xenograft model.
                                                       1
  Zn0.4Mg0.6Fe2O4   28      L929           6 mg mL−                In vitro cells   –               –         46 ◦ C          Ensuring the                   2021    [267]
                                                                                                                              implementation of OA-
                                                                                                                              coated Zn-Mg Ferrite
                                                                                                                              nanoparticles with
                                                                                                                              minimum dose rate.
  FVIO              73.9    H22            1 mg of                 Intratumoral     365             2300Wg-   44.1–44.3 ◦ C   FVIO-mediated MH and           2024    [268]
                                           Fe⋅cm− 3                                 kHz,300 Oe      1
                                                                                                                              Sorafenib offer sastrategy
                                                                                                                              for HCC treatment by
                                                                                                                              promoting accelerated
                                                                                                                              ferroptosis.
                                                               1
  CuFe2O4           200     4T1            1.5 mg mL−              Intratumoral     577             –         45 ◦ C          Magnetic mesoporous            2022    [269]
                                                                                    kHz,3700 W                                CuFe2O4 NPs with excellent
                                                                                                                              magnetothermal conversion
                                                                                                                              ability and large mesopores
                                                                                                                              enables them as an excellent
                                                                                                                              MHT agent and a good drug
                                                                                                                              carrier.
  CoFe2O4           12      CT26           106.2–3000              Intratumoral     261             400 Wg-   46–48 ◦ C       Provides more efficient        2021    [270]
                    ±4                     mg kg− 1                                 kHz,15–25       1
                                                                                                                              heating while achieving
                                                                                    mT                                        greater biosafety.
                                                           1
  Fe3O4 PLGA        172     CT26           25 mg kg−               Intratumoral     293 kHz,        –         44 ◦ C          MHT + chemotherapy             2018    [191]
    doxorubicin                                                                     12.57                                     presented marked tumor
                                                                                    kAm− 1                                    growth suppression
                                                                                                                              compared to MHT and
                                                                                                                              chemotherapy alone.
                                                           1
  Fe-Fe3O4 PECc     33      U87MC          40 mg kg−               Intravenous      13.56           –         38◦ C-water     On the day 15 post-MHT the     2018    [271]
    (RGDyK)                 glioblastoma                                            kHz,40                                    relative tumor size volume
    peptide                                                                         kAm− 1                                    reduced by half.
                                                                                                                              Hematoxylin-eosin staining
                                                                                                                              revealed nuclear
                                                                                                                              fragmentation and
                                                                                                                              shrinkage.
                                                       1
  Fe3O4 NGO-        36.8    C6 glioma      2 mg kg−                Intravenous      242.5           –         65◦ C-          MRI and Prussian blue          2018    [272]
    PLGA IUdR                                                                       kHz,21.8                  medium          staining revealed the higher
                                                                                    kAm− 1                                    localization of NPs in tumor
                                                                                                                              under AMF.
6.1.3.2. Surface-engineered metal oxides. Sood and his team have made                           common practice in the field of nanomedicine, particularly for HT ap
significant strides in the production of Fe3O4/Au core-shell nano                              plications. This method not only improves the stability and biocom
composites through a novel method involving the integration of gold                             patibility of the NPs but also serves as a versatile platform for further
chloride and trisodium citrate dihydrate into a suspension of existing                          modifications to enhance targeting specificity. The advantages of silica,
Fe3O4 NPs, which were then gently heated during their study [121]. The                          such as biocompatibility, stability, non-toxicity, and compatibility with
addition of the gold shell served a dual purpose by shielding the core                          various functional groups, have been widely recognized in the scientific
from corrosion while displaying excellent biocompatibility and binding                          community [124]. Silica coating plays a crucial role in forming a pro
capabilities through amine/thiol terminal functionalities [122]. These                          tective shield around the iron oxide core of the NPs, preventing direct
nanocomposites, derived from pre-synthesized magnetite NPs, demon                              contact with the external environment. This protective barrier helps
strated remarkable potential in biosensing applications, especially in                          prevent agglomeration and reduces the potential toxicity associated
protein detection and disease diagnosis. Moreover, the incorporation of                         with the NPs, making them safer for use in medical applications. Re
gold nanoshells was found to significantly enhance photodegradation                             searchers, like Madhappan Santha Moorthy and their team [125], have
efficiency under UV light, allowing for photodegradation even under                             successfully synthesized Fe3O4@SiO2 NPs for HT applications in cancer
natural sunlight due to gold’s responsiveness to visible light [123].                           treatment. In their study, they encapsulated FeNPs with silica to prevent
Tamer and his research team also made significant contributions to this                         aggregation in suspensions and to allow for the attachment of biological
area by developing anisotropic core-shell nanocomposites using                                  molecules for targeted therapy. This innovative approach combines
Fe3O4@Au NPs dispersed in sodium citrate before introducing them into                           chemotherapy and thermal therapy to effectively target cancer cells,
a growth solution comprising cetyl trimethylammonium bromide,                                   showcasing the promising potential of silica-coated IONPs in advancing
HAuCl4, AgNO3, and ascorbic acid at room temperature. The resulting                             medical treatments. The ability of silica coating to enhance the thera
high surface area of the product greatly improved its ability to detect                         peutic efficacy of IONPs while minimizing potential risks highlights the
Escherichia coli in large quantities, showcasing the potential of these                         significance of this technique in the field of nanomedicine.
nanocomposites in a wide range of applications.
    The use of silica-coated IONPs followed by functionalization is a                           6.1.3.3. Binary ferrite. Pure magnetic metals such as Cobalt, Iron, or
                                                                                          13
L. Xiong et al.                                                                                                             Materials Today Bio 33 (2025) 102070
Cobalt Iron have high saturation magnetization values but are prone to              polyol method in ethylene glycol, MnFe2O4 NPs achieve Ms of 90–95
high losses and toxicity. Additionally, they are sensitive to oxidation,            emu⋅g − 1 at 4 K, yielding SAR up to 1200 Wg-1 (Fe + Mn basis). Align
making them unsuitable for biomedical applications without a safe                   ment in a uniform static AMF followed by matrix immobilization en
biocompatible coating. An alternative option is the use of ferrites, which          hances heating efficiency by 40–60 % compared to dispersed NPs [128].
can be created by substituting various metal ions like Nickel, Cobalt, and          Notably, MnFe2O4 demonstrates lower cytotoxicity than zinc ferrite,
Manganese for Fe2+ ions in the magnetite lattice. Among these ferrites,             with 50 % cell viability observed at 0.2 mg mL− 1 after 24-h exposure
cobalt ferrite stands out with 90 % of the saturation magnetization of              [128].
magnetite but possesses significantly higher magnetic anisotropy and                    Particle size critically governs HT efficacy: NPs >15 nm exhibit
coercivity. This results in a larger area for magnetic hysteresis, indi            higher Ms (~80 emu⋅g− 1) and SAR values (H0 = 18 kAm− 1, f = 336 kHz),
cating that cobalt ferrite NPs require more intense AMF for effective               while sub-10 nm particles show diminished heating due to suppressed
utilization compared to other spinel NPs of similar size. These findings            Néel relaxation [129]. Incorporating ZnS into MnFe2O4 nanocomposites
highlight the potential of cobalt ferrite in various applications but also          (MnFe2O4/ZnS) elevates Ms by 25 % via cation redistribution. Zn2+
emphasize the need for careful consideration of their magnetic proper              substitution in tetrahedral (A) sites displaces Mn2+, forcing Fe3+
ties when designing devices or treatments utilizing these NPs [126].                migration to octahedral (B) sites. As a result, the MnFe2O4/ZnS nano
    Cobalt ferrite nanoparticles (CFNPs, Co0.65Fe2.35O4) with a 17 nm               composite exhibits an enhanced net saturation magnetization compared
average diameter were synthesized via thermal decomposition, followed               to MnFe2O4 NPs alone [129].
by hydrophilic coating to enhance water solubility and biocompatibility.                Nickel ferrite nanoparticles (NiFe2O4 NPs), despite intrinsic non-
These CFNPs demonstrated superior HT performance compared to cubic                  biocompatibility, exhibit biomedical utility owing to their high mag
IONPs, achieving a SAR of 400 Wg-1 under low-frequency AMF (H0 = 20                 netic moments (~90 emu⋅g− 1) when functionalized with biocompatible
kAm− 1, f = 105 kHz)—double the SAR of cubic IONPs (200 Wg-1)                       coatings such as oleic acid and tetramethyl ammonium hydroxide
(Fig. 8) [96]. In murine models, intratumorally CFNP administration                 (TMAH) [130]. For instance, Umut et al. synthesized superparamagnetic
and three consecutive 30-min AMF cycles (4 kAm− 1, 280 kHz) induced a               NiFe2O4 NPs (4.4 nm core, 15 nm hydrodynamic diameter) via
6 ◦ C temperature differential between tumor and distal tail tissue on day          co-precipitation, achieving SAR of 4–11 Wg-1 under AMF (H0 =
one, stabilizing at 3.5 ◦ C on days two and three. Despite this attenuation,        17.2–23.7 kAm− 1, f = 170 kHz). Residual hysteresis in magnetization
sustained intratumorally heating confirmed CFNP efficacy.                           curves arose from interparticle exchange/dipolar interactions within
    Manganese ferrite nanoparticles (MnFe2O4 NPs) exhibit negligible                aggregates [131]. Comparative studies of 20 nm ferrites reveal CoFe2O4
coercivity, high magnetic susceptibility, and minimal retentivity (<5               NPs yield the highest specific loss power (SLP = 315 Wg-1), out
emu⋅g − 1), distinguishing them from conventional superparamagnetic                 performing NiFe2O4 and MnFe2O4 (SLP = 295Wg-1). Notably, increasing
materials. Their biocompatibility, chemical stability, and high Néel               CoFe2O4 concentration enhances SLP by 20–30 %, whereas NiFe2O4 and
transition temperatures (>573 K) position them as promising candidates              MnFe2O4 exhibit 15–25 % SLP reduction at >0.4 g mL− 1 due to
for MHT and MRI contrast enhancement [127]. Synthesized via the                     aggregation-induced dipolar coupling losses [132]. These findings
Fig. 8. Characterization of cobalt ferrite nanocubes. (A) Schematic representation of aqueous solution of polymer-coated Co–Fe NCs compatible for biological
application. (B–C) TEM images of PMAO-coated Co–Fe NCs with mean sizes of 17 ± 2 nm. (D) Higher SAR value for Co–Fe NCs at lower clinically relevant AMF
compared to IONCs. (E) No toxicity for IONCs and significant intrinsic toxicity of Co–Fe NCs on cancer cells. Reproduced with permission [96]. Copyright
2020, Wiley.
                                                                               14
L. Xiong et al.                                                                                                             Materials Today Bio 33 (2025) 102070
underscore ferrite-based MIH as a versatile platform for antitumor                 composition, hydrophobicity, molecular weight, and stiffness [139,
strategies, validated across preclinical in vitro and in vivo models.              140]. This degree of control often surpasses that attainable with
                                                                                   lipid-based nanomaterials. Specifically, in microbubble technology,
6.2. Organic nanomaterials                                                         polymer-shelled microbubbles exhibit several advantages over
                                                                                   lipid-based analogs. These liposomes can bear a greater drug payload
    Owing to their superior biodegradability, exceptional biocompati              owing to their denser shells [141,142] and can be designed to assume
bility, and facile fabrication processes, organic nanomaterials have               anisotropic forms, which boosts the efficiency of drug delivery by
garnered significant attention in biomedical research over several de             optimizing margination characteristics [143,144].
cades. In the specialized field of MHT, organic micro-/nanosystems                     Nanosystems synthesized via material engineering and synthetic
primarily comprise lipid-based and polymer-based.                                  chemistry strategies often require surface modifications to achieve
                                                                                   physiological stability, biocompatibility, and prolonged circulation for
6.2.1. Lipid-based nanomaterials                                                   disease diagnosis and treatment, particularly inorganic platforms. Sur
    Since the groundbreaking discovery of liposomes by Bangham in                  face functionalization enhances targeted accumulation at lesion sites,
1965, these lipid-based carriers have revolutionized the field of drug             enabling precise imaging or therapy with reduced dosages. This prin
delivery. Liposomes, composed primarily of lipids and fatty acids, have            ciple applies universally, including ultrasound-based nanosystems,
shown remarkable biocompatibility and biodegradability, making them                where surface engineering—such as PEGylation of MNPs—is critical for
ideal candidates for encapsulating and administering therapeutic mol              in vivo efficacy. Polymer-coated MNPs exemplify this strategy,
ecules to combat various diseases. Their unique structure consists of              combining magnetic separation with ligand-mediated targeting for ap
amphipathic molecules that self-assemble into a bilayer sphere, with               plications like circulating tumor cells (CTCs) isolation [145].
hydrophilic head groups facing the exterior aqueous environment and                    CTCs, shed from primary tumors into peripheral blood, are rare
hydrocarbon chains residing within the hydrophobic interior [133]. The             targets crucial for cancer diagnostics. Immunomagnetic separation le
versatility of liposomes as drug carriers lies in their amphiphilic nature,        verages polymer-coated MNPs for CTCs enrichment, offering high cap
which allows them to encapsulate molecules of varying polarities                   ture efficiency and specificity through ligands (e.g., antibodies [146],
effectively [134]. By encapsulating drugs within liposomes, systemic               aptamers [147]) targeting CTCs-specific biomarkers. The polymer
toxicity can be minimized, and dosing regimens can be more easily                  coating stabilizes the magnetic core, provides biocompatibility, and fa
tolerated, particularly in treatments for cancer [135]. The combination            cilitates functionalization. Upon exposure to blood, ligand-receptor in
of thermosensitive liposomes with HT could revolutionize the treatment             teractions selectively bind CTCs, while an external AMF isolates
of cancer by improving drug delivery and minimizing off-target effects.            CTCs-MNP complexes from blood components, minimizing contamina
    Ongoing research in this field continues to explore novel strategies to        tion and enhancing downstream analytical sensitivity.
enhance the efficacy and performance of temperature-sensitive lipo                    Synthetic polymers (e.g., PEG, PDA, polyvinylpyrrolidone) and nat
somes (TSLs) for targeted drug delivery applications. The use of pure              ural polymers (e.g., chitosan, alginate, and dextran) are widely used as
1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) liposomes is often              coatings, balancing stability, biocompatibility, and functional group
limited by incomplete drug release, prompting researchers to incorpo              availability. For instance, Wang et al. designed hydrogel-coated MNPs
rate additional phospholipids like DSPC and HSPC to improve drug                   functionalized with anti-EpCAM antibodies via methacrylic acid link
release rates. In a seminal study by Yatvin et al., in 1978 [136], a               ages [148], enabling CTCs capture with minimal nonspecific adhesion.
mixture of DPPC and DSPC in a 3:1 M ratio was shown to enhance drug                GSH-mediated disulfide bond cleavage facilitated MNPs release while
release significantly when exposed to serum, especially at elevated                maintaining >95 % CTC viability post-separation. Optimization of
temperatures. The encapsulation efficiency of DSPC was nearly perfect              polymer coating remains pivotal for advancing CTC-based clinical di
in experiments involving localized mild HT in human BLM melanoma                   agnostics [149].
NMRI nude mice, highlighting its potential in optimizing drug release                  In addition to CTC isolation, polymer-based hydrogel materials also
kinetics. Further advancements in liposomal formulations were made by              exhibit unique advantages in the field of cancer therapy. For example,
Needham et al. [137], who introduced lysolipids such as MPPC and                   magnetic colloidal hydrogels can achieve precise delivery of liver cancer
MSPC in conjunction with lipid-grafted polyethylene glycol (PEG) and               ablators through minimally invasive percutaneous injection [150], and
DPPC to create a novel temperature-sensitive liposomal doxorubicin                 their injectable properties can adapt to irregular tumor shapes to
(DOX) formulation known as ThermoDox®. This formulation is                         improve treatment coverage [151]. The hydrogel matrix can not only
currently undergoing phase III clinical trials and shows promise in                adsorb chemotherapeutic drugs or thermal ablators but also be guided to
improving drug release at specific temperatures. In a separate study,              the tumor site by an external magnetic field, avoiding the trauma of
Tatsuaki et al. developed a TSLs formulation composed of DPPC and                  traditional surgery. In the postoperative treatment of hepatocellular
Brij78 in a molar ratio of 96:4 and compared it to TSLs containing                 carcinoma, injectable magnetic montmorillonite colloidal hydrogels are
lysolipids [138]. The results indicated that the novel TSLs formulation            enriched in residual lesions through magnetic targeting [152] (Fig. 9),
displayed an increased drug release rate at elevated temperatures, while           utilizing the layered structure of montmorillonite for sustained drug
maintaining stability at lower temperatures. The incorporation of pal             release while enhancing local magnetic responsiveness to reduce the risk
mitoyl lysophosphatidylcholine (P-lyso-PC) in liposomes was also noted             of postoperative recurrence. The three-dimensional network structure of
to enhance stability and operational efficiency, ultimately improving the          hydrogels enables synergistic delivery of drugs and hyperthermia,
performance of liposomal drug delivery systems. Overall, the integration           providing an integrated platform for multidisciplinary cancer treatment
of additional phospholipids and innovative formulations has paved the              [153,154]. These studies demonstrate that hydrogels, as an important
way for more efficient drug release mechanisms in liposomal systems.               branch of polymer-based materials, significantly enhance the precision
The use of synthetic phospholipids like P-lyso-PC holds great promise in           and safety of cancer therapy by integrating multiple properties such as
enhancing the stability and functionality of liposomes, offering a po             magnetic targeting, thermal responsiveness, and biodegradability,
tential solution to the challenges posed by incomplete drug release in             showcasing broad prospects for translation from laboratory to clinic.
traditional liposomal formulations.
                                                                                   7. Combination of MHT with other treatments
6.2.2. Polymer-based nanomaterials
   Polymerbased nanomaterials are renowned for their versatility,                     The destruction of tumors via single modality MHT necessitates high
providing extensive control over their physicochemical properties                  temperatures, thereby elevating the risk of damage to neighboring tis
through the customization of polymer chain attributes like monomer                 sue. Furthermore, the efficacy of single MHT on the entire tumor mass is
                                                                              15
L. Xiong et al.                                                                                                                      Materials Today Bio 33 (2025) 102070
Fig. 9. Preparation and Characterization of Magnetic Composite Hydrogel (MCH). (A) Gelation process and inverted fluorescence microscope images. Green: Fe3O4
NPs, red: gelatin NPs. (B) Zeta potential values at different pH values. (C) Trend of mixed particle size. (D) Storage modules and loss modules. (E) Shear rate scanning
viscosity and injection image illustrations. (F) i) SEM images of Fe3O4 NPs and ii) SEM images of gelatin NPs. (G) Scanning electron microscope image of MCH and
corresponding elemental mapping analysis. Reproduced with permission [152]. Copyright 2024, Wiley.
often inadequate due to the uneven distribution of heat. To address these              achieve efficient PTT conversion. Moreover, gold-silica nanostructures
challenges, research has focused on magnetic hyperthermia-based                        have demonstrated biosafety in early prostate cancer trials, yet NIR’s
combination therapy (MHCT). Integrating MHT with radiotherapy,                         limited penetration depth (5–10 mm) restricts PTT to superficial tumors.
chemotherapy, immunotherapy, and other treatments is viewed as a                       The integration of MHT and PTT enables thermal ablation of
promising strategy for synergistic intervention (Fig. 10) [155–158].                   deep-seated tumors by leveraging AMF to activate MNPs, thereby cir
MHCT not only enables the complete eradication of tumors with reduced                  cumventing tissue penetration limitations. This synergistic effect origi
AMF intensity and lower drug dosages but also minimizes harm to the                    nates from the dual-modality energy-responsive characteristics of MNPs,
surrounding healthy tissue, resulting in highly effective treatment with               which facilitate efficient energy accumulation through coupled mag
diminished side effects [88,159–162]. Moreover, the combination of                     netic moment relaxation and photoabsorption. Such dual-modality
MHT with other thermal therapies can compensate for the limitations of                 design allows NPs to concurrently harness magnetic energy (deep-
individual thermal treatments and optimize their therapeutic benefits.                 tissue penetration) and photonic energy (spatially precise targeting),
We also analyze the clinical use and prospects of several combination                  establishing a spatiotemporally coordinated heating network.
therapies.                                                                                 MNPs have been confirmed to be efficient magnetic-photothermal
                                                                                       combined agents in the NIR window [166–171]. According to existing
                                                                                       research results, this concept of antitumor nanotherapy based on iron
7.1. MHT and photothermal-combinational therapy                                        oxide cores can locally increase the temperature in vitro through only
                                                                                       magnetic and light-induced hyperthermia, leading to the complete
    For standalone MHT, the currently available MNPs exhibit low                       destruction of cancer cells and achieving complete tumor ablation in
power absorption efficiency, necessitating either impractically high-                  vivo [170]. Although the iron oxide component of MNPs can already
intensity AMF or elevated NPs doses. While PTT, which generates heat                   ensure potential photothermal conversion functions, magnetic hybrid
via non-radiative electron relaxation in light-absorbing materials (e.g.,              nanoplatforms should continue to be used for cancer theranostics. By
cyanine, phthalocyanine) [163]. Early PTT agents were plasmonic ma                    decorating iron oxide cores with gold and silver nanoparticles as typical
terials such as gold (Au), silver (Ag), and copper chalcogenides (Cu2-xE,              modifiers, the performance of iron oxide cores can be significantly
E = S, Se, Te, O) [164,165], because these materials have the minimum                  enhanced, thereby generating hybrid nanosystems with synergistic
tissue scattering characteristics in the NIR (650–950 nm) and can
                                                                                  16
L. Xiong et al.                                                                                                           Materials Today Bio 33 (2025) 102070
Fig. 10. Schematic diagram of combining anti-tumor therapy with MHT and other treatments.
magnetic and light-excited properties. For combined MHT-PTT pro                deep-seated tumors remains a significant challenge. MHT offers distinct
tocols, they generally involve intratumoral administration of MNPs              advantages for solving the question due to its unrestricted tissue pene
followed by synchronized application of AMF and NIR laser exposure to           tration. The deep-seated mechanisms of this synergistic effect stem from
achieve dual-modal thermal ablation. Espinosa et al. engineered iron            the multi-faceted regulation of tumor biological characteristics by MHT:
oxide-gold hybrid nanoassemblies that enhance heating via dual
magnetic-plasmonic properties, achieving SAR up to 5000 Wg-1 in                 1. Microenvironmental reoxygenation: local HT dilates normal blood
dual-mode (AMF: H0 = 20 kAm− 1, f = 110 kHz; NIR: 808 nm laser)                    vessels to promote oxygen delivery. Meanwhile, the disordered
[172]. This approach amplifies heating efficiency 2–5 times compared               tumor blood vessels produce a thermal sealing effect, selectively
with MHT alone, inducing apoptosis-mediated tumor regression in vivo.              increasing the intertumoral temperature (ΔT>5 ◦ C) and reversing
    Despite the synergistic potential of combined photothermal and                 hypoxia-induced radiotherapy resistance [175].
magnetic heating therapies to address individual modality limitations,          2. Inhibition of DNA repair: Hyperthermia (>41 ◦ C) disrupts the ho
clinical adoption remains limited. Key barriers include: multiparametric           mologous recombination repair (HRR) pathway by ubiquitinating
optimization complexity, therapeutic consistency, metabolic variability,           and degrading BRCA2/FANCD2 proteins, reducing the repair effi
safety-efficacy trade-offs, cost constraint and specialized operator de           ciency of radiation - induced DNA double - strand breaks [175].
mands. Nonetheless, the integration of magnetic and photothermal                3. Metabolic sensitization: heat shock increases the permeability of
modalities demonstrates transformative potential for tumor ablation, as            tumor cell membranes, promoting the influx of radiotherapy sensi
evidenced by preclinical efficacy.                                                 tizers (such as oxygen free radicals). At the same time, the acidic
                                                                                   microenvironment enhances the cytotoxicity of thermal damage
                                                                                   [181].
7.2. MHT in combination with radiotherapy
                                                                                    This spatiotemporal synergy allows the radiation dose to be reduced
    Hypoxic TMEs arise from aberrant vasculature, which differs mark           without affecting the curative effect, making it particularly suitable for
edly from normal vascular networks [173]. This oxygen-deprived state            the precise targeted treatment of deep-seated tumors (such as gliomas
contributes to tumor cell radioresistance [174], a challenge addressed          and prostate cancer).
by combining radiotherapy with HT to exploit the heightened thermal                 Combination therapy necessitates precise control to optimize thera
sensitivity of hypoxic cells [175]. While normal tissues exhibit vasodi        peutic outcomes. The sequence and timing of HT and radiotherapy
lation and increased perfusion under hyperthermia, the disorganized             critically influence their synergistic capacity to damage cancer cells.
vasculature of tumors impedes heat dissipation, resulting in localized          Edward et al. demonstrated that minimizing the interval between irra
thermal entrapment. Furthermore, lactic acid accumulation in hypoxic            diation and HT maximizes tumor cell death in murine models, while
regions acidifies the TME, potentiating heat-induced cytotoxicity [176].        delays exceeding 4 h reduced radiosensitization efficacy [182]. Notably,
Preclinical and clinical studies have explored integrating radiotherapy         moderate HT (39–42 ◦ C) can induce transient resistance to subsequent
with external heat modalities [177–180]. However, effective heating of
                                                                           17
L. Xiong et al.                                                                                                               Materials Today Bio 33 (2025) 102070
heat exposure, termed thermotolerance, necessitating strategic sched               7.3. MHT in combination with chemotherapy
uling of therapies [183]. Current protocols prioritize irradiating tumors
before administering MNPs and initiating MHT to mitigate this adaptive                  In the domains of materials chemistry and pharmacology, consider
response. A pivotal phase II clinical trial by Hauff et al. (2011) evaluated        able research is focused on the development of advanced MNP-based
combined MHT and radiotherapy in 66 recurrent GBM patients [28].                    platforms. These efforts seek to leverage the dual benefits of MNPs as
Intratumoral IONPs were activated via AMF alongside fractionated                    effective thermal agents and as carriers for chemotherapeutic drugs.
stereotactic radiotherapy, demonstrating improved survival and toler               Additionally, there is an emphasis on designing intelligent magnetic
ability compared to standard care. This study established foundational              nanoplatforms capable of loading and releasing chemotherapeutic
clinical evidence for dual-modality therapy. Advances in magnetic                   agents in a more controlled and targeted manner [185–187]. On one
nanomaterials, such as gadolinium-doped iron oxide nanoparticles                    hand, the heat produced during the MHT process serves as an external
(Gd-IONPs), further enable real-time tracking of therapeutic responses              stimulus, initiating drug release through a thermally driven mechanism.
via MRI. Jiang et al. reported Gd-IONPs with enhanced SAR over con                 On the other hand, the unique tumor conditions provide a special
ventional Fe3O4 [184], facilitating simultaneous particle tracing and HT            microenvironment in which different intratumoral stimuli (e.g., acidic
in TRAMP-C1 prostate adenocarcinoma models. Gd-IONP-mediated HT                     pH or hypoxic and pro-oxidative states) can promote controlled drug
combined with radiotherapy extended tumor growth delay (10 days vs                  release. The synergistic enhancement between MHT and chemotherapy
4.5 days for radiotherapy alone) and reduced hypoxia via vascular                   is primarily based on the following aspects: 1. Heat-enhanced drug
disruption and necrosis. Mechanistically, HT enhances radiotherapy                  penetration: local HT dilates tumor blood vessels and increases vascular
efficacy through thermal ablation (>45 ◦ C) and hypothermic (39–42 ◦ C)             permeability, promoting the extravasation of nanocarriers while
reoxygenation. Collectively, MHT-Radiotherapy synergy offers three                  reducing interstitial fluid pressure (IFP) to accelerate drug diffusion
key advantages: enhanced tumor cytotoxicity, reduced radiotherapy                   [188]. 2.Thermochemosensitization: many drugs exhibit chemothera
doses and associated normal tissue toxicity, and improved targeting of              peutic sensitization to tumor cells at temperatures between 40 and
deep-seated tumors.                                                                 42 ◦ C. Experimental animal studies show that combining HT with
                                                                                    TSL-mediated drug delivery significantly increases drug concentrations
                                                                                    in tumors compared to free drugs [189]. 3. Spatiotemporal precise
Fig. 11. Characterization of the MIDENs. (A) TEM images of SPIONs and MIDENs. (B) Differential scanning calorimetry of PLGA polymers of different compositions.
(C) Dynamic light scattering of SPIONs, PLGA/SPIONs, and MIDENs. (D) FTIR spectra of PLGA, MIDENs, and SPIONs. (E)Vibrating sample magnetometer plots of
SPIONs and MIDENs. (F) T2-weighted MR images of MIDENs with various Fe concentrations. (G) T2 relaxation rate (1/T2) of MIDENs with various Fe concentrations.
Reproduced with permission [191]. Copyright 2018, Elsevier.
                                                                               18
L. Xiong et al.                                                                                                             Materials Today Bio 33 (2025) 102070
controlled release: PH/heat dual-responsive materials (such as                    magnetic curcumin nanoparticles (termed mCNPs) and per
poly-NIPAM copolymers) achieve pulsed drug release triggered by AMF,              fluoropentane (PFP) to enhance their water solubility [198]. The plat
leading to significantly higher local tumor drug concentrations than              form integrated magnetizable polymethyl methacrylate (PMMA) bone
intravenous injection while reducing systemic toxicity.                           cement, containing Fe3O4 and Nd2Fe14B magnetic particles, to reinforce
    Based on the above facts, many studies have used stimulatory                  the mechanical properties of bone tissue. This system leverages
chemical materials with thermo-responsive and/or pH-responsive                    AMF-induced magnetothermal effects to drive the release of chemo
properties to achieve on-demand selective release of drugs during                 therapeutic drugs, providing targeted magneto-thermo-chemotherapy
MHT [190]. Thirunavukkarasu et al. constructed smart NPs consisting of            and multimodal treatment for bone mechanical enhancement. Prom
poly(lactic acid)-vinyl acrylate (PLGA), SPION, and DOX to realize                ising tumor suppression effects were observed in the 143b osteosarcoma
efficient MRI-induced dual cancer therapy with AMF-induced dual                   model (Fig. 12), although further research is required to ensure
therapy NPs: magnetic fluid inducible drug-eluting nanoparticles                  long-term stability and safety.
(MIDENs) [191]. The particles can respond to external AMF-generated
heat while simultaneously releasing drugs embedded in the NPs                     7.4. MHT in combination with immunotherapy
(Fig. 11). In vitro studies have shown that MIDENs are cytocompatible
and allow for magnetic imaging, heat generation, and active release of                The fundamental goal of cancer immunotherapy is to disrupt tumor
DOX in AMF applications. Furthermore, in vivo experiments have                    immune escape mechanisms and enhance the immune system’s capacity
demonstrated that MIDENs facilitate in vivo MRI imaging and that                  to recognize and eliminate malignancies [199,200]. This concept traces
thermochemotherapy induced by AMF is superior to other materials or               back to Dr. Coley’s 1890s discovery, which first established a direct link
methods examined in suppressing the growth of highly aggressive                   between elevated body temperature (e.g., fever) and amplified immune
tumors.                                                                           responses against tumors. Both exogenous hyperthermia and endoge
    Moreover, the most cutting-edge preclinical validation of pH-                 nous fever stimulate intratumoral immune cells through the following
responsive nanosystems has been demonstrated in the field of MRI-                 mechanisms.
based signal detection. T1 (Gd-based) or T2 (IONPs) contrasts are used
to track the delivery of pH nanosystems, and this application can be              1. Local HT induces immunogenic cell death in tumor cells, triggering
further used to detect different types of solid tumors. Polymers that                the following cascade reactions: (1) Release of damage-associated
respond to changes in pH are sensitive to the acidic environment at the              molecular patterns (DAMPs), including signaling molecules such as
site of pathology, leading to alterations in T2 contrast. A similar system           high-mobility group protein B1 (HMGB1), ATP, and calreticulin
could be used to identify other regions with acidic conditions, such as              (CRT). (2) Upon recognition of DAMPs by tumor-associated dendritic
tumors, or to improve the planning of combination therapies. Specif                 cells (TADCs), DCs are driven to mature into antigen-presenting cells
ically, pH-sensitive nanosystems that modulate both pH and T2 contrast               (APCs). Among them, HSPs, as a subset of DAMPs, can directly
can provide valuable insights into drug release from carriers and enable             promote the migration of DCs to lymph nodes and activate natural
more precise predictions regarding the optimal timing for MHT to                     killer T cells (NKT cells) and T lymphocytes (T cells) [194,201]. (3)
maximize synergistic effects. Sasikala et al. engineered mussel-inspired             Additionally, calreticulin, a potent immunostimulatory protein in
poly(HEMA-co-DMA)-functionalized SPIONs for pH-responsive de                        DAMPs, is crucial for Ca2+ homeostasis and glycoprotein folding, and
livery of bortezomib (BTZ) and MHT [192]. The copolymer’s multi                     is believed to play a role in immunogenic cell death and other
dentate catechol motifs enhanced colloidal stability and ligand                      extracellular functions [202]. When calreticulin binds to lipoprotein
anchoring, enabling dual thermo-chemotherapeutic action: localized                   receptors on TADCs, it activates a key phagocytic signal to promote
BTZ release under acidic conditions and heat generation via                          the maturation of TADCs into APCs [203].
Néel/Brownian relaxation. In vitro/in vivo studies validated synergistic         2. Reprogramming of the immune checkpoint microenvironment: HT
tumor suppression, positioning these nanoplatforms as precision                      reverses the immunosuppressive state of tumors by regulating
oncology tools.                                                                      signaling axes [204]. The response rate of immune checkpoint in
    Existing MHT platforms can be broadly categorized into two types:                hibitors (ICIs) has been confirmed to be positively correlated with
MNP-based systems and liquid/solid metal implant-based platforms                     tumor mutational burden (TMB). Ultra-hyperthermia (>50 ◦ C) can
[159,193,194]. For the former, shape- and size-tunable MNPs (e.g.,                   enhance TMB by damaging DNA and promoting the release of tumor
FeNPs, Fe2O3NPs, and Fe3O4 NPs) were synthesized, and their high                     neoantigens, thereby largely inducing specific and nonspecific im
specific surface area enabled them to load drugs efficiently. Neverthe              munity [205]. Rangamuwa et al. observed that 80 % of patients
less, MNPs that rely on relaxation and hysteresis loss mechanisms to                 receiving bronchoscopic hot steam ablation showed upregulation of
produce magnetothermal effects generally fall short in achieving high                PD-L1 expression [206], suggesting and possibly validating that HT
MHT performance. This is the case even when they are synthesized                     can convert "cold tumors" into "hot tumors". Additionally, the
through elaborate, complex procedures, under rigorous reaction condi                numbers of Tregs [207] and Th17 cells [208] have been shown to
tions, and undergo subsequent modifications [195,196]. As a result,                  decrease significantly after HT, which alleviates immunosuppression
magnetothermal therapy based on MNPs requires repetitive drug de                    to a certain extent.
livery and high-power AMF, which severely hampers its biological ap              3. HT mediates bidirectional regulation of cytokines to achieve directed
plications. Conversely, large-scale (millimeter-sized) metallic implants             activation of the cytokine network: (1) Surge in pro-inflammatory
featuring continuous metallic structures demonstrate potent magneto                 cytokines: Upregulates the secretion of pro-immune factors such as
thermal capabilities, capable of producing intense heat via eddy current             IL-12 and IL-23α, activating cytotoxic T lymphocytes (CTLs) and
thermal effects even when exposed to low-power AMF [197]. None                      enhancing their tumor-infiltrating capacity; (2) Inhibition of anti-
theless, metallic implants with compact structures and minimal surface               inflammatory cytokines: Suppresses immunosuppressive cytokines
area typically necessitate intricate etching or plating procedures to                such as VEGF and PDGF-AA, blocking tumor angiogenesis and
attain a modest level of drug loading, a process that significantly com             recruitment of myeloid-derived suppressor cells (MDSCs) [209].
plicates the drug loading and diminishes its efficiency. Crafting a
magneto-thermal combination therapy platform that boasts exceptional                 HT systematically activates anti-tumor immune responses through
MHT efficiency along with convenient and effective drug loading ca               multiple synergistic mechanisms, providing a theoretical basis for the
pabilities, while satisfying clinical requirements, undoubtedly poses a           combined application of MHT and immune therapy. This multi-
significant challenge. Recently, Liang et al. developed a multifunctional         dimensional immune activation strategy not only improves local
biomimetic bone magnet (BBM) platform triggered by an AMF, utilizing              tumor control rates but also opens new pathways for the treatment of
                                                                             19
L. Xiong et al.                                                                                                              Materials Today Bio 33 (2025) 102070
Fig. 12. In vivo therapeutic MHT efficacy of BBMs alliance mCNPs. (A) Treatment and follow-up regimen. (B) 2D (left) and 3D (right) infrared thermal images of
143B tumor-bearing mice injected with PBS and BBMs exposed to AMF at different time intervals and (C) corresponding temperature–time curve for the tumors. (D)
Representative images of 143B tumor-bearing mice 21 days after various treatments. (E) Body weights. (F) Tumor growth curve. Reproduced with permission [198].
Copyright 2023, Elsevier.
metastatic tumors by inducing systemic immune memory.                              immunotherapy. Nishikawa et al. demonstrated that combining MHT
                                                                                   with glycyrrhizin (an HMGB1 inhibitor), CpG oligodeoxynucleotides,
7.4.1. DAMPs expression and MHT                                                    and checkpoint blockers (anti-PD-1/anti-CTLA-4) eradicated poorly
    DAMPs, including HMGB1, HSPs, uric acid, hyaluronic acid frag                 immunogenic B16-F10 melanoma in C57BL/6 mice by suppressing
ments, acetylheparin sulfate, and ATP, play dual roles in tumor immu              HMGB1-mediated inflammation while enhancing systemic antitumor
nology. HMGB1, released during cell death, functions both as a DAMP                immunity [212].
and a cytokine, binding to Toll-like receptors (TLR2/4) and the receptor              HSPs similarly exhibit context-dependent roles: they confer resis
for advanced glycation end products (RAGE) to stimulate pro-                       tance to hyperthermia-induced cytotoxicity and promote angiogenesis/
inflammatory cytokine release (e.g., TNF-α) [210,211]. Paradoxically,              metastasis, yet their release during necrosis activates DCs via "danger
while HMGB1 promotes vascular endothelial proliferation and neo                   signal" recognition, triggering antigen-presenting cell maturation and
intima formation—potentially driving tumor progression—its                         prolonged immune responses. Sato et al. leveraged this duality by
hyperthermia-induced       overexpression       can   synergize     with           functionalizing silane-coated Fe3O4 nanoparticles with N-propionyl-4-S-
                                                                              20
L. Xiong et al.                                                                                                             Materials Today Bio 33 (2025) 102070
cysteinylaminophenol to enhance melanoma cell uptake [213]. In                    cytokines (e.g., IL-1β, IL-2, IL-6, IL-12, IL-23α, CCL2, TNF-α) in murine
B16-OVA xenografts, MHT (118 kHz, 30.6 kAm− 1, 30 min) induced                    and human macrophages [226], though outcomes depend on SPION
tumor necrosis and HSP expression, activating DCs and achieving                   surface     chemistry     and      cellular    context.   For    example,
complete regression without recurrence—even after contralateral tumor             carboxydextran-coated SPIONs enhance IL-12 in bone marrow-derived
rechallenge. This underscores MHT’s potential to prime adaptive im               macrophages, whereas dimercaptosuccinic acid (DMSA) or 3-aminopro
munity against metastatic lesions.                                                pyltriethoxysilane (APS) coatings show no such effect [227,228].
    Contrary to the HSPs overexpression strategy, another group                   Notably, dextran-coated SPIONs induce IL-1β and TNF-α in human pe
exploited gene therapy to downregulate HSPs. Gupta et al. pioneered an            ripheral blood mononuclear cells at levels comparable to lipopolysac
immunologically synergistic glioma therapy combining HSP90 sup                   charide (LPS) stimulation [229], a finding warranting further
pression with MHCT. Differential HSP expressions in glioma cells under            investigation for intravenous applications. SPIONs also variably modu
varied immunosuppressive TME models (2D/3D monoculture, co-                       late anti-inflammatory cytokines: IL-10 expression fluctuates inconsis
culture spheroids) revealed HSP90 upregulation post-MHCT. Co-                     tently across murine (RAW264.7) and human macrophages depending
administration of the HSP90 inhibitor 17-DMAG enhanced MHCT effi                 on [228,230], while DMSA/APS-coated SPIONs elevate TGF-β in
cacy, reducing primary/secondary tumors by 65 % and 53 % within 8                 THP-1-derived macrophages. Additionally, SPIONs suppress VEGF—a
days, achieving complete suppression by day 20. Extracellular HSP90               pro-angiogenic marker—in RAW264.7 cells, suggesting potential
from necrotic cells triggered IFN-γ and calreticulin upregulation, indi          anti-angiogenic effects meriting exploration in human models.
cating systemic immunological reaction. This synergy arises from
multimodal mechanisms: AMF-driven HT, HSP90 inhibition, and                       7.4.3. Immune checkpoint Blockade(ICB) and MHT
DAMPs-mediated DC/cytotoxic T-lymphocyte (CTL) activation.                            Under normal physiological conditions, the human immune system
    To further enhance the therapeutic effect, researchers have devel            identifies and eliminates foreign pathogens and aberrant cells. To pre
oped a hybrid nanosystem [214], which combines the advantages of                  vent excessive activation, regulatory mechanisms such as immune
magnetic-thermal therapy and immunomodulation. Called SPIOs +                     checkpoints (e.g., CTLA-4 and PD-1/PD-L1) modulate immune re
Magnetic-thermal sensitive phase-transition nanodroplets (RPPs), this             sponses. CTLA-4 operates during early immune activation by binding to
nanosystem utilizes phase-transition nanodroplets with immunomodu                APC surface molecules, thereby restricting T-cell proliferation. In
lation to enhance the efficacy of mild magnetic-thermal therapy (below            contrast, PD-1 interacts with PD-L1—expressed on both normal and
44 ◦ C). The nanodroplets consist of the immunoadjuvant Resiquimod                tumor cells—during the effector phase to suppress T-cell activity. ICIs
(R848) and the phase-change agent perfluoropentane (PFP) encapsu                 are therapeutic agents designed to block these regulatory molecules.
lated in a PLGA shell to form the RPPs. In the presence of the RPPs, the          Among these, CTLA-4 and PD-1/PD-L1 inhibitors represent foundational
microbubble-induced cavitation phenomenon reduces the temperature                 ICI classes [231]. The FDA’s 2011 approval of ipilimumab, a
threshold required for MHT from 50 ◦ C to approximately 44 ◦ C.                   CTLA-4-blocking antibody, marked the clinical introduction of ICIs.
Cavitation-induced cell membrane disruption resulted in increased                 These therapies are now integral to managing malignancies such as
release and exposure of DAMPs, including passively released HMGB1,                melanoma, non-small-cell lung cancer (NSCLC), gastrointestinal can
actively secreted ATP, and surface-exposed CRT (Fig. 13) [215]. This              cers, renal cell carcinoma, breast cancer, and hematologic cancers. In
process effectively converts “cold” tumors that are less responsive to            melanoma, PD-1 inhibitors nivolumab and pembrolizumab have
immunotherapy into “hot” tumors, reversing the TME and making tu                 significantly improved survival outcomes, securing their status as
mors more amenable to immunotherapy. R848 is a potent activator of                first-line treatments. For NSCLC, combined regimens involving
TLR-7/8, which acts as immune adjuvants in this nanosystem [216,217].             PD-1/PD-L1 inhibitors and chemotherapy yield remission rates of
When bound to MHT-released DAMPs, R848 is recognized by Toll-like                 37.3–45.3 % and a median overall survival of 15.6 months. Neoadjuvant
receptors (TLRs), which activate DCs. This activation is characterized            immunotherapy demonstrates a 38 % major pathological response rate
by up regulation of the co-stimulatory factors CD80 and CD86, which               in early-stage NSCLC. In metastatic colorectal cancer, PD-1 mono
are hallmarks of mature DCs. The shift in DC phenotype from immu                 therapy shows efficacy, particularly in patients with defective mismatch
nosuppressive to immunogenic further enhances the immune response                 repair or high microsatellite instability. Dual PD-1/CTLA-4 inhibition
against tumor cells [218,219]. The mature DCs then acquire an                     outperforms monotherapy in some contexts, emerging as a preferred
enhanced ability for antigen cross-presentation and secrete various               first-line option [232,233]. However, ICIs face limitations. First, mon
pro-inflammatory cytokines [220–222], including interleukin-6 (IL-6),             otherapy efficacy is often constrained by intrinsic or acquired resistance,
interleukin-12 (IL-12), and tumor necrosis factor (TNF-), which modu             mediated by antigen loss, compensatory immunosuppressive pathways,
late the anti-tumor immune response. This cascade ultimately promotes             or infiltration of regulatory immune cells. Second, immune-related
the activation of CTLs to eliminate tumors [223]. Quantitative assay              adverse events—including pneumonitis, hepatitis, endocrine dysfunc
results can be seen in a 72.39 % increase in the exposure of CRT on the           tion (e.g., thyroid abnormalities), dermatitis, and colitis—arise from
cell membrane and a 45.84 % increase in the release of HMGB1 in vivo.             systemic immune activation against healthy tissues [232,234–237].
In addition, the maturation rate of DCs increased from 4.17 % to 61.33                The integration of MHT with ICB represents a transformative strat
% and the infiltration rate of CTLs increased from 10.44 % to 35.68 %.            egy in oncology, leveraging nanoparticle-mediated thermal ablation to
The hybrid nanosystem treatment effectively inhibited contralateral and           prime systemic antitumor immunity. Chao et al. demonstrated this
lung metastases, a result attributed to the combined impact of mild MHT           synergy using PEG/dopamine-coated IONPs (30–50 nm) co-
and immunostimulatory effects.                                                    encapsulated with imiquimod (TLR7 agonist) in PLGA nanocapsules
                                                                                  [238]. Intertumoral injection followed by AMF exposure (Hf = 1.2 × 109
7.4.2. Cytokines and MHT                                                          Am− 1s− 1, 50–52 ◦ C) induced primary tumor regression and elicited an
    Cytokines, a class of signaling proteins, critically regulate immune          abscopal effect—suppressing contralateral metastases via cytotoxic
and inflammatory responses by potentiating cytotoxic T lymphocyte                 T-cell activation. Systemic anti-CTLA-4 administration amplified this
activity. Clinically explored cytokines include interferon (IFN) variants         response, achieving complete tumor eradication. Wang et al. advanced
(α, β, γ), tumor necrosis factor (TNF), colony-stimulating factors (CSFs),        this paradigm by engineering Janus-type nanospheres (250 × 100 nm)
and interleukins (IL-2, IL-6, IL-12) [224]. Despite preclinical promise,          comprising Fe3O4 heads and disulfide-bridged mesoporous silica tails
cytokine monotherapies underperform in clinical trials due to short               loaded with chlorin e6 (Ce6) [239]. In orthotopic 4T1 breast cancer
half-lives and narrow therapeutic windows [225]. Emerging strategies              models, sequential MHT (25.8 kAm− 1, 262 kHz, 20 min) and photody
propose synergizing cytokines with MHT, leveraging SPIONs to amplify              namic therapy (PDT) (660 nm laser, 0.15 W cm− 2, 10 min) triggered
immunomodulation. SPION-based MHT upregulates pro-inflammatory                    immunogenic        cell   death,    DC     maturation,     and     CTLA-4
                                                                             21
L. Xiong et al.                                                                                                                    Materials Today Bio 33 (2025) 102070
Fig. 13. The enhanced ICD in vitro mediated by RPPs potentiated mild MHT. (A) LSCM images of calcein-AM/PI-stained 4T1 cells under different treatment
conditions. (B) Quantitative analysis of (A). (C, D) Flow cytometry analysis of cell apoptosis after different therapies. (E) Western blot analysis of HMGB1 and ATP
expression in cells after various treatments. (F, G) Quantitative levels of HMGB1 and ATP in 4T1 tumors after various treatments determined by Western blot analysis.
(H, I) ELISA kit analysis of the released HMGB1 and ATP in the cell supernatant. Reproduced with permission [214]. Copyright 2023, Springer Nature.
                                                                                 22
L. Xiong et al.                                                                                                            Materials Today Bio 33 (2025) 102070
blockade-enhanced T-cell cytotoxicity. This tri-modal therapy (MHT +              which normally block anti-tumor T-cell activity. In addition, the com
PDT + ICB) eradicated primary tumors and prevented metastasis with                bination therapy prevented distant tumor progression in an extraper
superior efficacy to monotherapies, underscoring the potential of                 itoneal tumor model. These findings underscore the synergistic potential
thermal-immunological synergy in overcoming tumor resistance. Liu                 of MHT in combination with ICB in enhancing anti-tumor immune re
et al. treated orthotopic 4T1 breast tumors with MHT every other day              sponses and preventing metastasis. And in a study by Gao et al. the re
using PEGylated vortexed watershed IONPs (150.9 nm) and intraperi                searchers developed temperature-responsive iron oxide nano-assemblies
toneal injections of anti-PD-L1 antibodies [240]. MHT was performed               (IONA) using cross-linked IONPs loaded with JQ1 (JQ1/IONA) [241],
using alternating current at f = 365 kHz, but H values were not reported          an immunomodulator known to down-regulate PD-L1. IONA + AMF
(Fig. 14). Treatments were administered in multiple cycles every two              heated to ~45 ◦ C exhibited better immune response and antitumor ef
days. The study observed that mild HT eradicated primary tumors                   fects compared to IONPs + AMF (~41 ◦ C) and unresponsive
treated with thermal injury. More importantly, it prompted an 88 %                nano-assemblies (uIONA) + AMF (~50 ◦ C).The combination of
increase in cytotoxic CD8+ T lymphocyte infiltration in distant tumors,           JQ1/IONPs + AMF further combined mild thermal therapy with
which sensitized them to PD-L1 checkpoint blockers and triggered                  controlled release of JQ1, resulting in complete elimination of primary
immunotherapy (Fig. 15). The team also found that combination ther               tumors and induced a potent immune effect of tumor suppression. This
apy attenuated the tumor’s immunosuppressive mechanisms, as evi                  triggered a potent immune effect of inhibiting distant tumor growth and
denced by a marked reduction in myeloid-derived suppressor cells,                 preventing tumor recurrence and metastasis (Fig. 16). The synergistic
Fig. 14. Characterization of PEGylated FVIOs. (A) TEM and (B) HRTEM images of PEGylated FVIOs. (C) Lorentz TEM image of PEGylated FVIOs. (D) Hydrodynamic
diameter of PEGylated FVIOs. (E) Hydrodynamic size measured as a function of time upon incubation in de-ionized (DI) water, pH 7.4 PBS, and DMEM. Reproduced
with permission [240]. Copyright 2019, American Chemical Society.
                                                                             23
L. Xiong et al.                                                                                                                Materials Today Bio 33 (2025) 102070
Fig. 15. Graph showing the proportion of T-cell-related factors. (A) Representative multispectral fluorescence images of distant tumors after staining immunoflu
orescence. (B) The percentages of related tumors infiltrating T cells in the distant tumors. Reproduced with permission [240]. Copyright 2019, American Chemi
cal Society.
effects observed in these studies can be attributed to multiple mecha              advanced therapies may pose a barrier to their widespread use. More
nisms. First, HT induced by MHT and/or PDT triggers immune cell                     and more trials continue to confirm the remarkable effectiveness of the
death, leading to the release of tumor-associated antigens. These anti             combination of MHT and ICIs in activating the immune system, not only
gens activate DCs, which then migrate to lymph nodes and prime                      to achieve better anti-tumor efficacy, but also to reduce the body’s
anti-tumor T cells. Second, ICIs can block inhibitory signals used by               resistance and adverse reactions to the use of a single ICI.
tumors to evade immune surveillance, thereby enhancing the cytotoxic                    Overall, the combined antitumor regimen of magnetothermal ther
activity of T cells. Third, the combination of thermotherapy and ICB                apy and immunization is still far from clinical popularity. Currently, two
produces a “vaccination”-like effect, where the eradication of the pri             clinical trials (NCT03757858 and NCT03393858) are in the phase I/II
mary tumor generates a powerful immune memory response. This                        validation stage, exploring the combination of immunotherapy with
response allows the immune system to recognize and eliminate distant                thermotherapy; however, neither of these studies includes MHT [242].
tumor cells, thereby preventing recurrence and metastasis.                          Over the past two decades, the integration of MHT with immunotherapy
    Despite the encouraging results of these studies, clinical translation          has gained traction as a promising strategy, known as
of these combined strategies still faces several challenges. First, the             thermos-immunotherapy, which offers a more targeted treatment. This
biodistribution and clearance of MNPs need to be carefully controlled to            approach is capable of elevating temperatures specifically at the tumor
ensure their safety and efficacy. Second, the optimal dosage and timing             site, thereby enhancing heat dissipation and stimulating the local im
of ICIs need to be determined to maximize their synergistic effects with            mune response more effectively [243]. The initial MHT study for GBM
thermotherapy. In addition, the combination of multiple therapies may               patients [244], revealed that intratumoral injection of MNPs, followed
lead to increased toxicity and side effects, necessitating careful moni            by MHT irradiation at 49.5 ◦ C, led to an increase in macrophages within
toring and management. Further, the cost and availability of these                  the tumor. This finding emphasized the impact of MHT on immune
                                                                               24
L. Xiong et al.                                                                                                                Materials Today Bio 33 (2025) 102070
Fig. 16. Metastasis and recurrence inhibition of 4T1 tumors via systemic immune effects induced by JQ1/IONAs with AMF. (A) Schematic illustration of treatments
in lung metastasis model. (B, C) Recurrent tumor growth curves and recurrence rates in various groups. (D–F) Representative lung photographs. (G) Survival curves
of mice after different treatments in 45 days. Reproduced with permission [241]. Copyright 2023, Elsevier.
response, suggesting potential benefits for patient treatment outcomes.             8. Clinical translation challenges and future research priorities
Following this, multiple studies in animal models have also substanti              of MHT in tumor treatment
ated the finding that MHT can stimulate the immune response in pa
tients [203,245,246]. Consequently, an extensive array of research into                 MHT demonstrates significant potential in tumor therapy, yet its
the fusion of MHT with tumor immunotherapy has been progressively                   clinical translation faces multiple bottlenecks, including biosafety of
conducted.                                                                          MNPs, targeted delivery efficiency, precise control, device development,
                                                                                    and regulatory/ethical challenges. Future research should focus on the
                                                                               25
L. Xiong et al.                                                                                                              Materials Today Bio 33 (2025) 102070
development of novel nanomaterials, optimization of combination                    8.2. Future research priorities of MHT
therapies, construction of intelligent systems, and interdisciplinary
collaboration to overcome technical and institutional barriers [247].              8.2.1. Development of novel MNPs
                                                                                       Multifunctional NPs: Integration of diagnostic (MRI, photoacoustic
8.1. Technical bottlenecks in MHT clinical translation                             imaging) and therapeutic functions into a single nanoplatform enables
                                                                                   precise tumor localization, real-time monitoring, and personalized
8.1.1. Biosafety of MNPs                                                           therapy. For example, fluorescent dye-loaded nanoparticles can delin
    Toxicity assessment: the evaluation of in vivo toxicity of MNPs re            eate tumor boundaries pre-treatment, while MRI-guided temperature
mains a critical barrier. Current studies indicate that certain NPs (e.g.,         monitoring enhances precision. Biodegradable NPs: Materials like PLGA
unmodified IONPs) may induce oxidative stress, elevate ROS levels and              offer enhanced biosafety by degrading into harmless metabolites post-
cause mitochondrial and DNA damage in normal tissues. Surface charge,              treatment. Tailoring degradation rates can optimize magnetic perfor
particle size, and morphology also influence toxicity. For example,                mance and therapeutic outcomes.
nanoparticles <10 nm exhibit higher membrane permeability,
increasing interactions with intracellular biomolecules and toxicity               8.2.2. Intelligent MHT systems
risks. However, existing toxicity assessments rely on in vitro or short-               Real-time monitoring and feedback: Closed-loop systems integrating
term animal models, which fail to accurately simulate human physi                 microsensors, wireless modules, and machine learning algorithms
ology, leading to discrepancies between preclinical and clinical out              enable dynamic temperature regulation. For instance, fiber-optic sen
comes. Long-term biological effects: The chronic retention and risks of            sors combined with predictive algorithms adjust magnetic parameters to
MNPs in vivo remain understudied. Preclinical data suggest prolonged               maintain optimal thermal ranges [248]. Personalized treatment: Artifi
accumulation in reticuloendothelial system (RES) organs (e.g., liver,              cial intelligence-driven analysis of tumor genomics, imaging, and clin
spleen), potentially disrupting organ function. NPs aggregation, degra            ical data facilitates tailored therapy [8]. Digital twin technology can
dation, or surface alterations, along with unknown biosafety profiles of           simulate patient-specific tumor responses, guiding clinical decisions
degradation products (e.g., iron overload from slow degradation), and              through predictive modeling.
further complicate clinical applications. The lack of long-term follow-up
data and monitoring tools poses significant safety concerns.                       9. Conclusion and prospects
8.1.2. Targeting and delivery efficiency                                               MHT has emerged as a key strategy in precision oncology due to its
    Targeting strategies: current approaches (active/passive targeting)            non-invasiveness, deep tissue penetration, and synergistic interaction
face limitations. Active targeting suffers from ligand-antigen binding             with the TME, capable of inducing multiple cell death pathways such as
specificity issues and immunogenicity, while passive targeting is hin             apoptosis, ferroptosis, and pyroptosis. Compared with traditional HT,
dered by tumor vascular heterogeneity, resulting in poor penetration               MHT enables localized thermal control while minimizing non-target
into deep tumor regions and uneven cellular coverage. Delivery opti               tissue damage. Nanomaterials are classified into inorganic (e.g., iron
mization: structural designs (core-shell, hollow architectures) and sur           oxides, cobalt ferrites), organic (liposomes, polymers), and hybrid sys
face modifications (PEGylation, thermoresponsive coatings) aim to                  tems, with heating efficiency, biodistribution, and surface functionali
improve pharmacokinetics. However, complex designs increase pro                   zation (e.g., PEGylation, targeted ligand modification) being critical to
duction costs, surface modifications may compromise magnetic perfor               enhancing efficacy and safety. Current research emphasizes multimodal
mance, and dynamic tumor microenvironments (pH, enzymatic activity)                combination strategies to enhance antitumor efficacy through syner
destabilize nanoparticle delivery, limiting efficiency.                            gistic mechanisms. However, the complexity of treatments and incon
                                                                                   sistent protocols may lead to unpredictable synergistic toxicity,
8.1.3. Precision control in MHT                                                    necessitating a dose-time-intensity optimization framework to balance
    Temperature regulation: Precise thermal control is critical to avoid           efficacy and safety.
normal tissue damage. Current monitoring techniques (magnetic reso                    Future directions for clinical translation involve interdisciplinary
nance thermometry, infrared thermography) lack spatial resolution and              material innovation and intelligent system construction, requiring
real-time feedback. Tumor heterogeneity in thermal conductivity and                strengthened integration of materials science, immunology, and engi
blood flow exacerbates temperature inhomogeneity. Suboptimal tem                  neering to develop smart responsive nanoplatforms, compact AMF
peratures either fail to kill tumor cells or damage healthy tissues.               generators compatible with MRI thermometry and adaptive parameter
Treatment depth and coverage: Although MHT holds promise for deep-                 adjustment. Designing biomimetic carriers (e.g., exosome-modified
seated tumors, clinical application requires optimization of magnetic              magnetic nanoparticles) to enhance tumor affinity, combined with
field penetration and NPs distribution. Limited penetration depth of               self-regulating thermoresponsive materials to prevent overheating.
AMF and uneven NPs dispersion lead to incomplete tumor ablation.                   Meanwhile, materials can carry multiple drugs, such as chemothera
Irregular tumor morphology further complicates precise treatment.                  peutic agents, immune adjuvants, and even tumor gene intervention
                                                                                   factors, to achieve multi-faceted synergistic therapy. Additionally, the
8.1.4. Technical limitations of MHT devices                                        platform should incorporate machine learning guidance to dynamically
    Existing MHT devices face challenges in magnetic field uniformity,             optimize AMF parameters and nanomaterial dosages through AI algo
excessive size, operational complexity, and high costs. Clinical-grade             rithms. Evaluating multicenter phase I/II clinical trials to summarize the
systems struggle to maintain uniform heating in large treatment areas,             long-term biodistribution and metabolic pathways of materials, and
while inefficient cooling systems increase thermal injury risks. High              establishing standardized efficacy indicators, so as to address the multi-
costs also hinder widespread adoption in resource-limited settings.                faceted research bottlenecks of MHT materials in safety, standardiza
                                                                                   tion, and efficiency.
8.1.5. Regulatory and ethical challenges                                               Through multidisciplinary collaboration, MHT has transformative
    MHT confronts undefined regulatory frameworks and ethical di                  potential to evolve from an experimental platform to a clinical mainstay,
lemmas. Safety and efficacy standards for MNPs remain incomplete,                  thereby becoming the fourth pillar of cancer treatment alongside sur
complicating regulatory classification. Ethical issues include insufficient        gery, radiotherapy, and chemotherapy. This paradigm shift could
patient awareness of long-term risks (information asymmetry in                     redefine the treatment landscape for malignancies refractory to con
informed consent) and the absence of standardized dosing and protocols             ventional therapies.
in clinical trials, potentially compromising patient rights.
                                                                              26
L. Xiong et al.                                                                                                                                    Materials Today Bio 33 (2025) 102070
CRediT authorship contribution statement                                                        [29] W.J. Atkinson, I.A. Brezovich, D.P. Chakraborty, IEEE (Inst. Electr. Electron.
                                                                                                     Eng.) Trans. Biomed. Eng. (1984) 70–75.
                                                                                                [30] G. Bellizzi, O.M. Bucci, G. Chirico, Int. J. Hyperther. 32 (2016) 688–703.
    Linyan Xiong: Writing – original draft, Methodology, Data curation.                         [31] K. Hayashi, W. Sakamoto, T. Yogo, Adv. Funct. Mater. 26 (2016) 1708–1718.
Bing Liang: Writing – review & editing, Supervision, Funding acquisi                           [32] K. Mahmoudi, A. Bouras, D. Bozec, R. Ivkov, C. Hadjipanayis, Int. J. Hyperther.
tion, Conceptualization. Kexiao Yu: Writing – review & editing, Su                                  34 (2018) 1316–1328.
                                                                                                [33] H. Wu, L. Song, L. Chen, Y. Huang, Y. Wu, F. Zang, Y. An, H. Lyu, M. Ma, J. Chen,
pervision, Software, Conceptualization.                                                              Nanoscale 9 (2017) 16175–16182.
                                                                                                [34] A. Hervault, N.T.K. Thanh, Nanoscale 6 (2014) 11553–11573.
                                                                                                [35] J.J. Skitzki, E.A. Repasky, S.S. Evans, Curr. Opin. Invest. Drugs 10 (2009) 550.
                                                                                                     London, England: 2000.
Declaration of competing interest
                                                                                                [36] D. Killock, Nat. Rev. Clin. Oncol. 15 (2018), 266-266.
                                                                                                [37] N. Chan, I.M. Pires, Z. Bencokova, C. Coackley, K.R. Luoto, N. Bhogal,
    The authors declare no other competing interests.                                                M. Lakshman, P. Gottipati, F.J. Oliver, T. Helleday, Cancer Res. 70 (2010)
                                                                                                     8045–8054.
                                                                                                [38] R.J. Gillies, P.A. Schomack, T.W. Secomb, N. Raghunand, Neoplasia 1 (1999)
Acknowledgement                                                                                      197–207.
                                                                                                [39] R.K. Jain, J.D. Martin, T. Stylianopoulos, Annu. Rev. Biomed. Eng. 16 (2014)
                                                                                                     321–346.
    We acknowledge financial support from the National Natural Science                          [40] H. Reinhold, B. Endrich, Int. J. Hyperther. 2 (1986) 111–137.
Foundation of China (Grant No. 82203067), the Natural Science Foun                             [41] E. Ruoslahti, Nat. Rev. Cancer 2 (2002) 83–90.
dation of Chongqing (Grant No.CSTB2022NSCQ-MSX0109), the Scien                                 [42] H. Wiig, M.A. Swartz, Physiol. Rev. 92 (2012) 1005–1060.
                                                                                                [43] L.F. Fajardo, B. Egbert, J. Marmor, G.M. Hahn, Cancer 45 (1980) 613–623.
tific and Technology Research Program of Chongqing Municipal                                    [44] S.L. Sankari, K. Masthan, N.A. Babu, T. Bhattacharjee, M. Elumalai, Asian Pac. J.
Education     Commission      (Grant   Nos.   KJQN202300458        and                               Cancer Prev. APJCP 13 (2012) 4873–4878.
KJQN202500417), the "Light of West China" Program and the Chongq                               [45] A. Pawlik, J.M. Nowak, D. Grzanka, L. Gackowska, J. Michalkiewicz, A. Grzanka,
                                                                                                     Acta Histochem. 115 (2013) 8–15.
ing High-Level Talents Program for Young and Middle-aged Medical
                                                                                                [46] Z.-G. Cui, J.-L. Piao, T. Kondo, R. Ogawa, K. Tsuneyama, Q.-L. Zhao, L.B. Feril Jr.,
Professionals by Dr. Yu. All authors commented on the manuscript. All                                H. Inadera, Chem. Biol. Interact. 215 (2014) 46–53.
scheme illustrations were created from biorender with licenses.                                 [47] C. Bonzon, L. Bouchier-Hayes, L.J. Pagliari, D.R. Green, D.D. Newmeyer, Mol.
                                                                                                     Biol. Cell 17 (2006) 2150–2157.
                                                                                                [48] L. Bouchier-Hayes, A. Oberst, G.P. McStay, S. Connell, S.W. Tait, C.P. Dillon, J.
Data availability                                                                                    M. Flanagan, H.M. Beere, D.R. Green, Mol. Cell 35 (2009) 830–840.
                                                                                                [49] S. Tu, G.P. McStay, L.-M. Boucher, T. Mak, H.M. Beere, D.R. Green, Nat. Cell Biol.
    No data was used for the research described in the article.                                      8 (2006) 72–77.
                                                                                                [50] L.J. Pagliari, T. Kuwana, C. Bonzon, D.D. Newmeyer, S. Tu, H.M. Beere, D.
                                                                                                     R. Green, Proceedings of the, vol 102, National Academy of Sciences, 2005,
References                                                                                           pp. 17975–17980.
                                                                                                [51] S. Tran, A. Meinander, T. Holmström, A. Rivero-Müller, K. Heiskanen, E. Linnau,
                                                                                                     M. Courtney, D. Mosser, L. Sistonen, J. Eriksson, Cell Death Differ. 10 (2003)
  [1] H. Sung, J. Ferlay, R.L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal,
                                                                                                     1137–1147.
      F. Bray, CA Cancer J. Clin. 71 (2021) 209–249.
                                                                                                [52] J. Yoo, Y.J. Lee, J. Cell. Biochem. 101 (2007) 619–630.
  [2] J. Ferlay, M. Colombet, I. Soerjomataram, D.M. Parkin, M. Pineros, A. Znaor,
                                                                                                [53] J. Han, S.H. Back, J. Hur, Y.-H. Lin, R. Gildersleeve, J. Shan, C.L. Yuan,
      F. Bray, Int. J. Cancer (2021).
                                                                                                     D. Krokowski, S. Wang, M. Hatzoglou, Nat. Cell Biol. 15 (2013) 481–490.
  [3] C.R. Boland, S.K. Shin, A. Goel, Yonsei Med. J. 50 (2009) 309–321.
                                                                                                [54] Y. Tabuchi, K. Ahmed, T. Kondo, Hyperthermic Oncology from Bench to Bedside,
  [4] M. DuPage, A.F. Cheung, C. Mazumdar, M.M. Winslow, R. Bronson, L.
                                                                                                     2016, pp. 7–18.
      M. Schmidt, D. Crowley, J. Chen, T. Jacks, Cancer Cell 19 (2011) 72–85.
                                                                                                [55] J.L. Skibba, E.J. Quebbeman, J.H. Kalbfleisch, Cancer Res. 46 (1986) 6000–6003.
  [5] M.R. Horsman, P. Vaupel, Front. Oncol. 6 (2016) 66.
                                                                                                [56] N.F. El-Orabi, C.B. Rogers, H.G. Edwards, D.D. Schwartz, J. Therm. Biol. 36
  [6] J.M. Brown, A.J. Giaccia, Cancer Res. 58 (1998) 1408–1416.
                                                                                                     (2011) 49–56.
  [7] A. Farzin, S.A. Etesami, J. Quint, A. Memic, A. Tamayol, Adv. Healthcare Mater. 9
                                                                                                [57] M.R. Sepand, S. Ranjbar, I.M. Kempson, M. Akbariani, W.C.A. Muganda,
      (2020) e1901058.
                                                                                                     M. Müller, M.H. Ghahremani, M. Raoufi, Nanomed. Nanotechnol. Biol. Med. 29
  [8] X. Zhu, Y. Li, N. Gu, Nano Biomedicine & Engineering 15 (2023).
                                                                                                     (2020) 102243.
  [9] Y. Yan, Y. Zhang, J. Liu, B. Chen, Y. Wang, Medical Review (2024).
                                                                                                [58] D. Laha, A. Pramanik, J. Maity, A. Mukherjee, P. Pramanik, A. Laskar,
 [10] T. Kang, G.D. Cha, O.K. Park, H.R. Cho, M. Kim, J. Lee, D. Kim, B. Lee, J. Chu,
                                                                                                     P. Karmakar, Biochim. Biophys. Acta Gen. Subj. 1840 (2014) 1–9.
      S. Koo, ACS Nano 17 (2023) 5435–5447.
                                                                                                [59] J. Lin, Y. Liu, H. Wu, Z. Huang, J. Ma, C. Guo, F. Gao, P. Jin, P. Wei, Y. Zhang,
 [11] L. Beola, N. Iturrioz-Rodríguez, C. Pucci, R. Bertorelli, G. Ciofani, ACS Nano 17
                                                                                                     Small 14 (2018) 1703711.
      (2023) 18441–18455.
                                                                                                [60] H.-Y. Lu, Y.-J. Chang, N.-C. Fan, L.-S. Wang, N.-C. Lai, C.-M. Yang, L.-C. Wu, J.-a.
 [12] B. Chen, K. Guo, X. Zhao, Z. Liu, C. Xu, N. Zhao, F.J. Xu, Exploration (Beijing) 3
                                                                                                     A. Ho, Biomaterials 42 (2015) 30–41.
      (2023) 20220140.
                                                                                                [61] Y. Song, Y. Zhu, K. Jiang, X. Liu, L. Dong, D. Li, S. Chen, H. Xing, X. Yan, Y. Lu,
 [13] A. Shakeri-Zadeh, J.W. Bulte, Nature reviews bioengineering 3 (2025) 245–260.
                                                                                                     Nanoscale 15 (2023) 365–375.
 [14] U. Jeong, X. Teng, Y. Wang, H. Yang, Y. Xia, Adv. Mater. 19 (2007) 33–60.
                                                                                                [62] D. Tang, G. Kroemer, R. Kang, Nat. Rev. Clin. Oncol. 21 (2024) 370–388.
 [15] A.P. Khandhar, R.M. Ferguson, K.M. Krishnan, J. Appl. Phys. 109 (2011).
                                                                                                [63] X.-K. Jin, J.-L. Liang, S.-M. Zhang, Q.-X. Huang, S.-K. Zhang, C.-J. Liu, X.-
 [16] M.B. Lilly, I.A. Brezovich, W. Atkinson, Radiology 154 (1985) 243–244.
                                                                                                     Z. Zhang, Mater. Today 68 (2023) 108–124.
 [17] J. Rehman, J. Landman, R.D. Tucker, D.G. Bostwick, C.P. Sundaram, R.
                                                                                                [64] C. Wang, Y. Xue, K. Lin, J. Lu, J. Chang, J. Sun, Acta Biomater. 8 (2012) 350–360.
      V. Clayman, J. Endourol. 16 (2002) 523–531.
                                                                                                [65] W.-R. Zhuang, Y. Wang, P.-F. Cui, L. Xing, J. Lee, D. Kim, H.-L. Jiang, Y.-K. Oh,
 [18] H. Shoji, Y. Ozu, F. Sato, H. Matsuki, S. Satomi, Y. Nihei, Y. Kurokawa, T. Sato,
                                                                                                     J. Contr. Release 294 (2019) 311–326.
      IEEE Trans. Magn. 41 (2005) 4167–4169.
                                                                                                [66] Y. Dai, L. Zhu, X. Li, F. Zhang, K. Chen, G. Jiao, Y. Liu, Z. Yang, Z. Guo, B. Zhang,
 [19] P. Wust, U. Gneveckow, P. Wust, U. Gneveckow, M. Johannsen, D. Böhmer,
                                                                                                     Biomaterials 305 (2024) 122455.
      T. Henkel, F. Kahmann, J. Sehouli, R. Felix, Int. J. Hyperther. 22 (2006) 673–685.
                                                                                                [67] C.H. Ries, M.A. Cannarile, S. Hoves, J. Benz, K. Wartha, V. Runza, F. Rey-Giraud,
 [20] M. Johannsen, U. Gneveckow, K. Taymoorian, B. Thiesen, N. Waldöfner,
                                                                                                     L.P. Pradel, F. Feuerhake, I. Klaman, Cancer Cell 25 (2014) 846–859.
      R. Scholz, K. Jung, A. Jordan, P. Wust, S. Loening, Int. J. Hyperther. 23 (2007)
                                                                                                [68] Z. Li, W. Li, Q. Li, M. Tang, K. Gao, L. Huang, Acta Pharm. Sin. B (2022).
      315–323.
                                                                                                [69] E.J. Ge, A.I. Bush, A. Casini, P.A. Cobine, J.R. Cross, G.M. DeNicola, Q.P. Dou, K.
 [21] M. Johannsen, U. Gneveckow, B. Thiesen, K. Taymoorian, C.H. Cho,
                                                                                                     J. Franz, V.M. Gohil, S. Gupta, Nat. Rev. Cancer 22 (2022) 102–113.
      N. Waldöfner, R. Scholz, A. Jordan, S.A. Loening, P. Wust, Eur. Urol. 52 (2007)
                                                                                                [70] V. Schulz, S. Basu, S.-A. Freibert, H. Webert, L. Boss, U. Mühlenhoff, F. Pierrel, L.-
      1653–1662.
                                                                                                     O. Essen, D.M. Warui, S.J. Booker, Nat. Chem. Biol. 19 (2023) 206–217.
 [22] O. Dudeck, K. Bogusiewicz, J. Pinkernelle, G. Gaffke, M. Pech, G. Wieners,
                                                                                                [71] L. Qiao, G. Zhu, T. Jiang, Y. Qian, Q. Sun, G. Zhao, H. Gao, C. Li, Adv. Mater. 36
      H. Bruhn, A. Jordan, J. Ricke, Investig. Radiol. 41 (2006) 527–535.
                                                                                                     (2024) 2308241.
 [23] F. Gazeau, M. Lévy, C. Wilhelm, Nanomedicine 3 (2008) 831–844.
                                                                                                [72] H. Lin, P. Zhang, J. Yu, H. Deng, S. Ge, H. Dou, J. Ruan, X. Fan, Mater. Today
 [24] S. Takamatsu, O. Matsui, T. Gabata, S. Kobayashi, M. Okuda, T. Ougi, Y. Ikehata,
                                                                                                     (2024).
      I. Nagano, H. Nagae, Radiat. Med. 26 (2008) 179–187.
                                                                                                [73] D. Liu, M. Liu, Y. Wan, X. Zhou, S. Yang, L. An, G. Huang, Q. Tian, Chem. Eng. J.
 [25] H. Shokrollahi, K. Janghorban, Mater. Sci. Eng., B 141 (2007) 91–107.
                                                                                                     422 (2021) 130098.
 [26] S. Sun, H. Zeng, D.B. Robinson, S. Raoux, P.M. Rice, S.X. Wang, G. Li, J. Am.
                                                                                                [74] M. El Hout, L. Dos Santos, A. Hamaï, M. Mehrpour, A promising new approach to
      Chem. Soc. 126 (2004) 273–279.
                                                                                                     cancer therapy: targeting iron metabolism in cancer stem cells, in: Seminars in
 [27] M. Johannsen, U. Gneveckow, L. Eckelt, A. Feussner, N. Waldöfner, R. Scholz,
                                                                                                     Cancer Biology, vol. 53, Elsevier, 2018, pp. 125–138.
      S. Deger, P. Wust, S. Loening, A. Jordan, Int. J. Hyperther. 21 (2005) 637–647.
                                                                                                [75] N.C. Andrews, P.J. Schmidt, Annu. Rev. Physiol. 69 (2007) 69–85.
 [28] K. Maier-Hauff, F. Ulrich, D. Nestler, H. Niehoff, P. Wust, B. Thiesen, H. Orawa,
      V. Budach, A. Jordan, Journal of neuro-oncology 103 (2011) 317–324.
                                                                                           27
L. Xiong et al.                                                                                                                                        Materials Today Bio 33 (2025) 102070
 [76] H. Feng, K. Schorpp, J. Jin, C.E. Yozwiak, B.G. Hoffstrom, A.M. Decker,                     [124] P. Cheah, J. Qu, Y. Li, D. Cao, X. Zhu, Y. Zhao, J. Magn. Magn Mater. 540 (2021)
      P. Rajbhandari, M.E. Stokes, H.G. Bender, J.M. Csuka, Cell Rep. 30 (2020)                         168481.
      3411–3423. e3417.                                                                           [125] M.S. Moorthy, Y. Oh, S. Bharathiraja, P. Manivasagan, T. Rajarathinam, B. Jang,
 [77] S.J. Dixon, K.M. Lemberg, M.R. Lamprecht, R. Skouta, E.M. Zaitsev, C.E. Gleason,                  T.T.V. Phan, H. Jang, J. Oh, RSC Adv. 6 (2016) 110444–110453.
      D.N. Patel, A.J. Bauer, A.M. Cantley, W.S. Yang, cell 149 (2012) 1060–1072.                 [126] E. Mazario, N. Menendez, P. Herrasti, M. Canete, V. Connord, J. Carrey, J. Phys.
 [78] N. Kayagaki, I.B. Stowe, B.L. Lee, K. O’Rourke, K. Anderson, S. Warming,                          Chem. C 117 (2013) 11405–11411.
      T. Cuellar, B. Haley, M. Roose-Girma, Q.T. Phung, Nature 526 (2015) 666–671.                [127] K. Islam, M. Haque, A. Kumar, A. Hoq, F. Hyder, S.M. Hoque, Nanomaterials 10
 [79] J. Shi, Y. Zhao, K. Wang, X. Shi, Y. Wang, H. Huang, Y. Zhuang, T. Cai, F. Wang,                  (2020) 2297.
      F. Shao, Nature 526 (2015) 660–665.                                                         [128] Z. Beji, A. Hanini, L. Smiri, J. Gavard, K. Kacem, F. Villain, J.-M. Greneche,
 [80] R. Tang, J. Xu, B. Zhang, J. Liu, C. Liang, J. Hua, Q. Meng, X. Yu, S. Shi,                       F. Chau, S. Ammar, Chem. Mater. 22 (2010) 5420–5429.
      J. Hematol. Oncol. 13 (2020) 1–18.                                                          [129] D. Mondal, C. Borgohain, N. Paul, J. Borah, Phys. B Condens. Matter 567 (2019)
 [81] S.B. Kovacs, E.A. Miao, Trends Cell Biol. 27 (2017) 673–684.                                      122–128.
 [82] Q. Wang, Y. Wang, J. Ding, C. Wang, X. Zhou, W. Gao, H. Huang, F. Shao, Z. Liu,             [130] J. Wang, F. Ren, R. Yi, A. Yan, G. Qiu, X. Liu, J. Alloys Compd. 479 (2009)
      Nature 579 (2020) 421–426.                                                                        791–796.
 [83] K.J. Waldron, J.C. Rutherford, D. Ford, N.J. Robinson, Nature 460 (2009)                    [131] E. Umut, M. Coşkun, F. Pineider, D. Berti, H. Güngüneş, J. Colloid Interface Sci.
      823–830.                                                                                          550 (2019) 199–209.
 [84] Y. Zhu, Y. Song, Z. Cao, L. Dong, Y. Lu, X. Yang, J. Wang, Adv. Funct. Mater. 31            [132] C.i.d.E. Demirci Donmez, P.K. Manna, R. Nickel, S. Akturk, J. van Lierop, ACS
      (2021) 2103655.                                                                                   Appl. Mater. Interfaces 11 (2019) 6858–6866.
 [85] R.E. Rosensweig, J. Magn. Magn Mater. 252 (2002) 370–374.                                   [133] A. Akbarzadeh, R. Rezaei-Sadabady, S. Davaran, S.W. Joo, N. Zarghami,
 [86] J.Y. Lee, Y.R. Na, C.M. Na, P.W. Im, H.W. Park, M.K. Kim, Y. Kim, J.H. You, D.                    Y. Hanifehpour, M. Samiei, M. Kouhi, K. Nejati-Koshki, Nanoscale Res. Lett. 8
      S. Kang, H.E. Moon, Theranostics 15 (2025) 2883.                                                  (2013) 1–9.
 [87] L. Sun, H. Liu, Y. Ye, Y. Lei, R. Islam, S. Tan, R. Tong, Y.B. Miao, L. Cai, Signal         [134] Y. Song, Z. Sheng, Y. Xu, L. Dong, W. Xu, F. Li, J. Wang, Z. Wu, Y. Yang, Y. Su,
      Transduct. Targeted Ther. 8 (2023) 418.                                                           Biomater. Sci. 7 (2019) 867–875.
 [88] X. Liu, Y. Zhang, Y. Wang, W. Zhu, G. Li, X. Ma, Y. Zhang, S. Chen, S. Tiwari,              [135] S. Paramshetti, M. Angolkar, S. Talath, R.A.M. Osmani, A. Spandana, A. Al
      K. Shi, Theranostics 10 (2020) 3793.                                                              Fatease, U. Hani, K. Ramesh, E. Singh, Life Sci. (2024) 122616.
 [89] X. Ma, Y. Wang, X.-L. Liu, H. Ma, G. Li, Y. Li, F. Gao, M. Peng, H.M. Fan, X.-              [136] M.B. Yatvin, J.N. Weinstein, W.H. Dennis, R. Blumenthal, Science 202 (1978)
      J. Liang, Nanoscale Horizons 4 (2019) 1450–1459.                                                  1290–1293.
 [90] R.L. Ge, P.N. Yan, Y. Liu, Z.S. Li, S.Q. Shen, Y. Yu, Adv. Funct. Mater. 33 (2023)          [137] D. Needham, G. Anyarambhatla, G. Kong, M.W. Dewhirst, Cancer Res. 60 (2000)
      2301138.                                                                                          1197–1201.
 [91] M. Wang, H. Yi, Z. Zhan, Z. Feng, G.G. Yang, Y. Zheng, D.Y. Zhang, Aggregate 5              [138] T. Tagami, M.J. Ernsting, S.-D. Li, J. Contr. Release 152 (2011) 303–309.
      (2024) e622.                                                                                [139] J. Liang, R. Cui, X. Zhang, K. Koumoto, C. Wan, Adv. Funct. Mater. 33 (2023)
 [92] Y. Zhu, Q. Li, C. Wang, Y. Hao, N. Yang, M. Chen, J. Ji, L. Feng, Z. Liu, Chem. Rev.              2208813.
      123 (2023) 7326–7378.                                                                       [140] F. Persano, L. Lamanna, M. Friuli, A.C. Stetco, A. Barca, T. Verri, C. Malitesta,
 [93] H. Keum, E. Cevik, J. Kim, Y.M. Demirlenk, D. Atar, G. Saini, R.A. Sheth, A.                      C. Demitri, E. Mazzotta, View (2025) 20250029.
      R. Deipolyi, R. Oklu, Adv. Mater. 36 (2024) 2310856.                                        [141] R.A. Barmin, A. Dasgupta, C. Bastard, L. De Laporte, S. Rutten, M. Weiler,
 [94] R.F. Casal, A.L. Tam, G.A. Eapen, Clin. Chest Med. 31 (2010) 151–163.                             F. Kiessling, T. Lammers, R.M. Pallares, Mol. Pharm. 19 (2022) 3256–3266.
 [95] H. Huang, Y. Zheng, M. Chang, J. Song, L. Xia, C. Wu, W. Jia, H. Ren, W. Feng,              [142] M.C. Cochran, J. Eisenbrey, R.O. Ouma, M. Soulen, M.A. Wheatley, Int. J. Pharm.
      Y. Chen, Chem. Rev. 124 (2024) 8307–8472.                                                         414 (2011) 161–170.
 [96] P.B. Balakrishnan, N. Silvestri, T. Fernandez-Cabada, F. Marinaro, S. Fernandes,            [143] M.A. Beach, U. Nayanathara, Y. Gao, C. Zhang, Y. Xiong, Y. Wang, G.K. Such,
      S. Fiorito, M. Miscuglio, D. Serantes, S. Ruta, K. Livesey, Adv. Mater. 32 (2020)                 Chem. Rev. 124 (2024) 5505–5616.
      2003712.                                                                                    [144] A. Dasgupta, T. Sun, R. Palomba, E. Rama, Y. Zhang, C. Power, D. Moeckel,
 [97] Y. Zhang, X. Zhang, H. Yang, L. Yu, Y. Xu, A. Sharma, P. Yin, X. Li, J.S. Kim,                    M. Liu, A. Sarode, M. Weiler, Proceedings of, vol 120, the National Academy of
      Y. Sun, Chem. Soc. Rev. 50 (2021) 11227–11248.                                                    Sciences, 2023 e2218847120.
 [98] M. Xu, Y. Qi, G. Liu, Y. Song, X. Jiang, B. Du, ACS Nano 17 (2023) 20825–20849.             [145] X. Jiang, X. Zhang, C. Guo, B. Ma, Z. Liu, Y. Du, B. Wang, N. Li, X. Huang, L. Ou,
 [99] Z. Ma, J. Mohapatra, K. Wei, J.P. Liu, S. Sun, Chem. Rev. 123 (2021) 3904–3943.                   Adv. Funct. Mater. 34 (2024) 2304426.
[100] C. Cocchi, M. Guerrini, J. Krumland, N.T. Nguyen, A.M. Valencia, J. Phys.:                  [146] D. Lin, L. Shen, M. Luo, K. Zhang, J. Li, Q. Yang, F. Zhu, D. Zhou, S. Zheng,
      Materials 6 (2022) 012001.                                                                        Y. Chen, Signal Transduct. Targeted Ther. 6 (2021) 404.
[101] K.S. Suslick, M. Fang, T. Hyeon, J. Am. Chem. Soc. 118 (1996) 11960–11961.                  [147] B. Rezaei, A. Harun, X. Wu, P.R. Iyer, S. Mostufa, S. Ciannella, I.H. Karampelas,
[102] X. Yang, J. Cheng, X. Yang, Y. Xu, H. Li, W. Sun, W. Tu, J. Zhou, Chem. Eng. J.                   J. Chalmers, I. Srivastava, J. Gómez-Pastora, Adv. Healthcare Mater. 13 (2024)
      455 (2023) 140694.                                                                                2401213.
[103] Y. Hou, S. Gao, J. Mater. Chem. 13 (2003) 1510–1512.                                        [148] Z. Wang, Z. Wu, N. Sun, Y. Cao, X. Cai, F. Yuan, H. Zou, C. Xing, R. Pei, J. Mater.
[104] J. Dupont, B.C. Leal, P. Lozano, A.L. Monteiro, P. Migowski, J.D. Scholten, Chem.                 Chem. B 9 (2021) 677–682.
      Rev. 124 (2024) 5227–5420.                                                                  [149] C.-Y. Wen, L.-L. Wu, Z.-L. Zhang, Y.-L. Liu, S.-Z. Wei, J. Hu, M. Tang, E.-Z. Sun, Y.-
[105] M. Mahmoudi, H. Hofmann, B. Rothen-Rutishauser, A. Petri-Fink, Chem. Rev. 112                     P. Gong, J. Yu, ACS Nano 8 (2014) 941–949.
      (2012) 2323–2338.                                                                           [150] W.S. Wu, X. Yan, S. Chen, Y. Du, J. Hu, Y. Song, Z. Zha, Y.J. Xu, B. Cao, S.H. Xuan,
[106] A. Kunzmann, B. Andersson, T. Thurnherr, H. Krug, A. Scheynius, B. Fadeel,                        Adv. Mater. 36 (2024) 2309770.
      Biochimica et Biophysica Acta (BBA)-general subjects 1810 (2011) 361–373.                   [151] K.-Y. Qian, Y. Song, X. Yan, L. Dong, J. Xue, Y. Xu, B. Wang, B. Cao, Q. Hou,
[107] G. Liu, J. Gao, H. Ai, X. Chen, Small 9 (2013) 1533–1545.                                         W. Peng, Biomaterials 259 (2020) 120299.
[108] O.B. Adewale, S.O. Anadozie, H. Davids, S. Roux, Potential therapeutic role of              [152] S. Chen, Y. Song, X. Yan, L. Dong, Y. Xu, S. Xuan, Q. Shu, B. Cao, J. Hu, H. Xing,
      gold nanoparticles in inflammatory diseases. Gold Nanoparticles for Drug                          J. Nanobiotechnol. 20 (2022) 381.
      Delivery, Elsevier, 2024, pp. 197–225.                                                      [153] X. Yan, T. Sun, Y. Song, W. Peng, Y. Xu, G. Luo, M. Li, S. Chen, W.-W. Fang,
[109] I. Dutta, A. Rahaman, S. Singh, N. Kumar, M.K. Tiwari, J. Drug Deliv. Therapeut.                  L. Dong, Nano Lett. 22 (2022) 2251–2260.
      15 (2025).                                                                                  [154] J. Gong, J. Hu, X. Yan, L. Xiang, S. Chen, H. Yang, Z. Chen, Q. Hou, Y. Song, Y. Xu,
[110] Y. Hou, H. Kondoh, T. Kogure, T. Ohta, Chem. Mater. 16 (2004) 5149–5152.                          Small 20 (2024) 2300733.
[111] S. Sun, C.B. Murray, D. Weller, L. Folks, A. Moser, science 287 (2000) 1989–1992.           [155] H. Jiang, H. Fu, Y. Guo, P. Hu, J. Shi, Biomaterials 289 (2022) 121799.
[112] F. Liu, J. Zhu, W. Yang, Y. Dong, Y. Hou, C. Zhang, H. Yin, S. Sun, Angew. Chem.            [156] Y. Zhang, Y. Xu, D. Sun, Z. Meng, W. Ying, W. Gao, R. Hou, Y. Zheng, X. Cai,
      Int. Ed. 53 (2014) 2176–2180.                                                                     B. Hu, Chem. Eng. J. 390 (2020) 124521.
[113] P. De La Presa, M. Multigner, M. Morales, T. Rueda, E. Fernandez-Pinel,                     [157] J. Pan, P. Hu, Y. Guo, J. Hao, D. Ni, Y. Xu, Q. Bao, H. Yao, C. Wei, Q. Wu, ACS
      A. Hernando, J. Magn. Magn Mater. 316 (2007) e753–e755.                                           Nano 14 (2020) 1033–1044.
[114] A. Baeza, D. Arcos, M. Vallet-Regí, J. Phys. Condens. Matter 25 (2013) 484003.              [158] H. Wu, L. Liu, L. Song, M. Ma, N. Gu, Y. Zhang, ACS Nano 13 (2019)
[115] M. Kaur, K. Singh, Mater. Sci. Eng. C 102 (2019) 844–862.                                         14013–14023.
[116] C.F. Geraldes, Molecules 29 (2024) 1352.                                                    [159] J. Pan, Y. Xu, Q. Wu, P. Hu, J. Shi, J. Am. Chem. Soc. 143 (2021) 8116–8128.
[117] H. Gaß, M. Sarcletti, L. Müller, S. Hübner, T. Yokosawa, H. Park, T. Przybilla,             [160] L. Zhang, Z. Liu, Y. Liu, Y. Wang, P. Tang, Y. Wu, H. Huang, Z. Gan, J. Liu, D. Wu,
      E. Spiecker, M. Halik, Adv. Sci. 10 (2023) 2302495.                                               Biomaterials 230 (2020) 119655.
[118] Y. Iqbal, H. Bae, I. Rhee, S. Hong, J. Nanosci. Nanotechnol. 16 (2016)                      [161] G. Song, M. Kenney, Y.-S. Chen, X. Zheng, Y. Deng, Z. Chen, S.X. Wang, S.
      11862–11867.                                                                                      S. Gambhir, H. Dai, J. Rao, Nat. Biomed. Eng. 4 (2020) 325–334.
[119] P. Das, M. Colombo, D. Prosperi, Colloids Surf. B Biointerfaces 174 (2019) 42–55.           [162] J. Xie, Y. Zhang, C. Yan, L. Song, S. Wen, F. Zang, G. Chen, Q. Ding, C. Yan, N. Gu,
[120] S.M. Dadfar, K. Roemhild, N.I. Drude, S. von Stillfried, R. Knüchel, F. Kiessling,                Biomaterials 35 (2014) 9126–9136.
      T. Lammers, Adv. Drug Deliv. Rev. 138 (2019) 302–325.                                       [163] X. Sun, Z. Zhang, C. Xiang, T. Qiao, X. Wang, G. Ma, D. Ding, Nano Biomedicine
[121] A. Sood, V. Arora, J. Shah, R. Kotnala, T.K. Jain, Mater. Sci. Eng. C 80 (2017)                   and Engineering (2025).
      274–281.                                                                                    [164] J. Ma, N. Li, J. Wang, Z. Liu, Y. Han, Y. Zeng, In vivo synergistic tumor therapies
[122] J. Luo Wang, Q. Fan, M. Suzuki, I.S. Suzuki, M.H. Engelhard, Y. Lin, N. Kim, J.                   based on copper sulfide photothermal therapeutic nanoplatforms, in: Exploration,
      Q. Wang, C.-J. Zhong, J. Phys. Chem. B 109 (2005) 21593–21601.                                    vol. 3, Wiley Online Library, 2023 20220161.
[123] A. Salman, S. Lupi, L. Vaccari, F. Piccirilli, M.M. Eid, Spectrochim. Acta Mol.             [165] S. Xie, H. Liu, K. Yang, T. Wang, H. Zhang, Z. Li, Nano Biomedicine & Engineering
      Biomol. Spectrosc. 287 (2023) 122087.                                                             17 (2025).
                                                                                             28
L. Xiong et al.                                                                                                                                        Materials Today Bio 33 (2025) 102070
[166] A. Espinosa, R. Di Corato, J. Kolosnjaj-Tabi, P. Flaud, T. Pellegrino, C. Wilhelm,           [214] Q. Qin, Y. Zhou, P. Li, Y. Liu, R. Deng, R. Tang, N. Wu, L. Wan, M. Ye, H. Zhou,
      ACS Nano 10 (2016) 2436–2446.                                                                      J. Nanobiotechnol. 21 (2023) 131.
[167] S. Cabana, A. Curcio, A. Michel, C. Wilhelm, A. Abou-Hassan, Nanomaterials 10                [215] W. Um, H. Ko, D.G. You, S. Lim, G. Kwak, M.K. Shim, S. Yang, J. Lee, Y. Song,
      (2020) 1548.                                                                                       K. Kim, Adv. Mater. 32 (2020) 1907953.
[168] S. Nemec, S. Kralj, C. Wilhelm, A. Abou-Hassan, M.-P. Rols, J. Kolosnjaj-Tabi,               [216] Y. Zhao, X. Zhao, X. Wang, Z. Ma, J. Yan, S. Li, N. Wang, J. Jiao, J. Cui, G. Zhang,
      Appl. Sci. 10 (2020) 7322.                                                                         Acta Biomater. 193 (2025) 417–428.
[169] E. Bertuit, E. Benassai, G. Mériguet, J.-M. Greneche, B. Baptiste, S. Neveu,                [217] K. Gowsalya, B. Rithisa, S. Shyamsivappan, R. Vivek, RSC Pharmaceutics 1 (2024)
      C. Wilhelm, A. Abou-Hassan, ACS Nano 16 (2021) 271–284.                                            441–457.
[170] C. Lozano-Pedraza, E. Plaza-Mayoral, A. Espinosa, B. Sot, A. Serrano, G. Salas,              [218] Y. Mi, C.T. Hagan IV, B.G. Vincent, A.Z. Wang, Adv. Sci. 6 (2019) 1801847.
      C. Blanco-Andujar, G. Cotin, D. Felder-Flesch, S. Begin-Colin, Nanoscale Adv. 3              [219] V. Revuri, S.K. Rajendrakumar, M.S. Park, A. Mohapatra, S. Uthaman, J. Mondal,
      (2021) 6490–6502.                                                                                  W.K. Bae, I.K. Park, Y.K. Lee, Adv. Healthcare Mater. 10 (2021) 2100907.
[171] S. Chen, M. Qiu, R. Wang, L. Zhang, C. Li, C. Ye, X. Zhou, Bioconjug. Chem. 33               [220] Z. Fan, H. Liu, Y. Xue, J. Lin, Y. Fu, Z. Xia, D. Pan, J. Zhang, K. Qiao, Z. Zhang,
      (2022) 1729–1740.                                                                                  Bioact. Mater. 6 (2021) 312–325.
[172] A. Espinosa, M. Bugnet, G. Radtke, S. Neveu, G.A. Botton, C. Wilhelm, A. Abou-               [221] H. Wang, X. Pan, X. Wang, W. Wang, Z. Huang, K. Gu, S. Liu, F. Zhang, H. Shen,
      Hassan, Nanoscale 7 (2015) 18872–18877.                                                            Q. Yuan, ACS Nano 14 (2020) 2847–2859.
[173] Y. Zhao, F. Tan, J. Zhao, S. Zhou, Y. Luo, C. Gong, MedComm 6 (2025) e70202.                 [222] B. Ding, S. Shao, C. Yu, B. Teng, M. Wang, Z. Cheng, K.L. Wong, P.a. Ma, J. Lin,
[174] R.G. Bristow, R.P. Hill, Nat. Rev. Cancer 8 (2008) 180–192.                                        Adv. Mater. 30 (2018) 1802479.
[175] M.F. Righini, A. Durham, P.G. Tsoutsou, Front. Oncol. 14 (2024) 1428065.                     [223] S. Persano, F. Vicini, A. Poggi, J.L.C. Fernandez, G.M.R. Rizzo, H. Gavilán,
[176] X. Wang, L. Wang, Q. Hao, M. Cai, X. Wang, W. An, Theranostics 14 (2024) 6831.                     N. Silvestri, T. Pellegrino, Pharmaceutics 13 (2021) 1668.
[177] T.M. Zagar, J.R. Oleson, Z. Vujaskovic, M.W. Dewhirst, O.I. Craciunescu, K.                  [224] M. Yi, T. Li, M. Niu, H. Zhang, Y. Wu, K. Wu, Z. Dai, Signal Transduct. Targeted
      L. Blackwell, L.R. Prosnitz, E.L. Jones, Int. J. Hyperther. 26 (2010) 612–617.                     Ther. 9 (2024) 176.
[178] J. Van Der Zee, M. De Bruijne, J. Mens, A. Ameziane, M. Broekmeyer-Reurink,                  [225] B. Seruga, H. Zhang, L.J. Bernstein, I.F. Tannock, Nat. Rev. Cancer 8 (2008)
      T. Drizdal, M. Linthorst, G. Van Rhoon, Int. J. Hyperther. 26 (2010) 638–648.                      887–899.
[179] S.V. Spirou, M. Basini, A. Lascialfari, C. Sangregorio, C. Innocenti, Nanomaterials          [226] A.M. Dias, A. Courteau, C. Petitot, J. Simonet, J.-M. Vrigneaud, P.-S. Bellaye,
      8 (2018) 401.                                                                                      A. Oudot, A. Kownacka, J. Paris, R. Decréau, Theranostic Properties of Iron Oxide
[180] M. Franckena, J. van der Zee, Curr. Opin. Obstet. Gynecol. 22 (2010) 9–14.                         Nanoparticles and their Reprograming Properties on Tumor-Associated
[181] Y. Cai, T. Chai, W. Nguyen, J. Liu, E. Xiao, X. Ran, Y. Ran, D. Du, W. Chen,                       Macrophages, Springer, 2024.
      X. Chen, Signal Transduct. Targeted Ther. 10 (2025) 115.                                     [227] R. Jin, L. Liu, W. Zhu, D. Li, L. Yang, J. Duan, Z. Cai, Y. Nie, Y. Zhang, Q. Gong,
[182] E.L. Gillette, B.A. Ensley, Int. J. Radiat. Oncol. Biol. Phys. 5 (1979) 209–213.                   Biomaterials 203 (2019) 23–30.
[183] P. Vaupel, H. Piazena, M. Notter, A.R. Thomsen, A.-L. Grosu, F. Scholkmann, A.               [228] J.M. Rojas, L. Sanz-Ortega, V. Mulens-Arias, L. Gutiérrez, S. Pérez-Yagüe, D.
      G. Pockley, G. Multhoff, Cancers 15 (2023) 1394.                                                   F. Barber, Nanomed. Nanotechnol. Biol. Med. 12 (2016) 1127–1138.
[184] P.-S. Jiang, H.-Y. Tsai, P. Drake, F.-N. Wang, C.-S. Chiang, Int. J. Hyperther. 33           [229] Q. Wu, T. Miao, T. Feng, C. Yang, Y. Guo, H. Li, Mol. Med. Rep. 18 (2018)
      (2017) 770–778.                                                                                    564–570.
[185] M. Arafat, M. Sakkal, R. Beiram, S. AbuRuz, Pharmaceuticals 17 (2024) 315.                   [230] D. Gonnissen, Y. Qu, K. Langer, C. Öztürk, Y. Zhao, C. Chen, G. Seebohm,
[186] Y. Zhang, F. Lei, W. Qian, C. Zhang, Q. Wang, C. Liu, H. Ji, Z. Liu, F. Wang,                      M. Düfer, H. Fuchs, H.-J. Galla, Int. J. Nanomed. (2016) 5221–5236.
      J. Contr. Release 373 (2024) 929–951.                                                        [231] K.M. Campbell, M. Amouzgar, S.M. Pfeiffer, T.R. Howes, E. Medina, M. Travers,
[187] M. Ashrafizadeh, A. Zarrabi, A. Bigham, A. Taheriazam, Y. Saghari, S. Mirzaei,                     G. Steiner, J.S. Weber, J.D. Wolchok, J. Larkin, Cancer Cell 41 (2023) 791–806.
      M. Hashemi, K. Hushmandi, H. Karimi-Maleh, E. Nazarzadeh Zare, Med. Res. Rev.                      e794.
      43 (2023) 2115–2176.                                                                         [232] L. Galluzzi, J. Humeau, A. Buqué, L. Zitvogel, G. Kroemer, Nat. Rev. Clin. Oncol.
[188] B. Lahooti, R.G. Akwii, F.T. Zahra, M.S. Sajib, M. Lamprou, A. Alobaida, M.                        17 (2020) 725–741.
      S. Lionakis, G. Mattheolabakis, C.M. Mikelis, J. Contr. Release 361 (2023)                   [233] R.M. Webster, Nat. Rev. Drug Discov. 13 (2014) 883.
      212–235.                                                                                     [234] M. de Miguel, E. Calvo, Cancer Cell 38 (2020) 326–333.
[189] G. Van Rhoon, M. Franckena, T. Ten Hagen, Adv. Drug Deliv. Rev. 163 (2020)                   [235] R.W. Jenkins, D.A. Barbie, K.T. Flaherty, Br. J. Cancer 118 (2018) 9–16.
      145–156.                                                                                     [236] J.A. Marin-Acevedo, E.O. Kimbrough, Y. Lou, J. Hematol. Oncol. 14 (2021) 1–29.
[190] Y. Song, D. Li, Y. Lu, K. Jiang, Y. Yang, Y. Xu, L. Dong, X. Yan, D. Ling, X. Yang,          [237] F. Martins, L. Sofiya, G.P. Sykiotis, F. Lamine, M. Maillard, M. Fraga,
      Natl. Sci. Rev. 7 (2020) 723–736.                                                                  K. Shabafrouz, C. Ribi, A. Cairoli, Y. Guex-Crosier, Nat. Rev. Clin. Oncol. 16
[191] G.K. Thirunavukkarasu, K. Cherukula, H. Lee, Y.Y. Jeong, I.-K. Park, J.Y. Lee,                     (2019) 563–580.
      Biomaterials 180 (2018) 240–252.                                                             [238] Y. Chao, G. Chen, C. Liang, J. Xu, Z. Dong, X. Han, C. Wang, Z. Liu, Nano Lett. 19
[192] A.R.K. Sasikala, A. GhavamiNejad, A.R. Unnithan, R.G. Thomas, M. Moon, Y.                          (2019) 4287–4296.
      Y. Jeong, C.H. Park, C.S. Kim, Nanoscale 7 (2015) 18119–18128.                               [239] Z. Wang, F. Zhang, D. Shao, Z. Chang, L. Wang, H. Hu, X. Zheng, X. Li, F. Chen,
[193] F. Liu, H. Wu, B. Peng, S. Zhang, J. Ma, G. Deng, P. Zou, J. Liu, A.T. Chen, D. Li,                Z. Tu, Adv. Sci. 6 (2019) 1901690.
      Nano Lett. 21 (2021) 8111–8118.                                                              [240] X. Liu, J. Zheng, W. Sun, X. Zhao, Y. Li, N. Gong, Y. Wang, X. Ma, T. Zhang, L.-
[194] M. Chang, Z. Hou, M. Wang, C. Li, J. Lin, Adv. Mater. 33 (2021) 2004788.                           Y. Zhao, ACS Nano 13 (2019) 8811–8825.
[195] P. Chandrasekharan, Z.W. Tay, D. Hensley, X.Y. Zhou, B.K. Fung, C. Colson, Y. Lu,            [241] M. Gao, K. Feng, X. Zhang, Y. Ruan, G. Zhao, H. Liu, X. Sun, J. Contr. Release 357
      B.D. Fellows, Q. Huynh, C. Saayujya, Theranostics 10 (2020) 2965.                                  (2023) 40–51.
[196] A.G. Roca, L. Gutiérrez, H. Gavilán, M.E.F. Brollo, S. Veintemillas-Verdaguer,             [242] M. Paulides, H.D. Trefna, S. Curto, D. Rodrigues, Adv. Drug Deliv. Rev. 163
      M. del Puerto Morales, Adv. Drug Deliv. Rev. 138 (2019) 68–104.                                    (2020) 3–18.
[197] N. Yang, F. Gong, L. Cheng, H. Lei, W. Li, Z. Sun, C. Ni, Z. Wang, Z. Liu, Natl. Sci.        [243] A.A. Petryk, A.J. Giustini, R.E. Gottesman, B.S. Trembly, P.J. Hoopes, Int. J.
      Rev. 8 (2021) nwaa122.                                                                             Hyperther. 29 (2013) 819–827.
[198] B. Liang, Y. Cao, X. Wang, H. Zhou, M. Wang, Y. Cao, W. Lu, K. Yu, Mater. Des.               [244] F.K. van Landeghem, K. Maier-Hauff, A. Jordan, K.-T. Hoffmann, U. Gneveckow,
      234 (2023) 112311.                                                                                 R. Scholz, B. Thiesen, W. Brück, A. von Deimling, Biomaterials 30 (2009) 52–57.
[199] A. Ribas, J.D. Wolchok, Science 359 (2018) 1350–1355.                                        [245] H.A. Albarqi, L.H. Wong, C. Schumann, F.Y. Sabei, T. Korzun, X. Li, M.N. Hansen,
[200] Y. Singh, V.K. Pawar, J.G. Meher, K. Raval, A. Kumar, R. Shrivastava,                              P. Dhagat, A.S. Moses, O. Taratula, ACS Nano 13 (2019) 6383–6395.
      S. Bhadauria, M.K. Chourasia, J. Contr. Release 254 (2017) 92–106.                           [246] T.J. Carter, G. Agliardi, F.Y. Lin, M. Ellis, C. Jones, M. Robson, A. Richard-Londt,
[201] H.-Y. Wang, J.C.-M. Fu, Y.-C. Lee, P.-J. Lu, Mol. Cell Biol. 33 (2013) 4889–4899.                  P. Southern, M. Lythgoe, M. Zaw Thin, Small 17 (2021) 2005241.
[202] J. Fucikova, R. Spisek, G. Kroemer, L. Galluzzi, Cell Res. 31 (2021) 5–16.                   [247] K.M. Jurczak, T.A. van der Boon, R. Devia-Rodriguez, R. Cl, J.S. Schuurmann, J.-
[203] L. Beola, V. Grazú, Y. Fernández-Afonso, R.M. Fratila, M. de Las Heras, J.s.M. de                 P.P. De Vries, L. van Huizen, P. van Rijn, On the Verge of bio-instructive Medical
      la Fuente, L. Gutiérrez, L. Asín, ACS Appl. Mater. Interfaces 13 (2021)                           Implant Surfaces, 2024, p. 93.
      12982–12996.                                                                                 [248] M. Tiwari, D. Mishra, Applications of 3D-and 4D-Printed polymer nanocomposites
[204] A. Osipov, S.J. Lim, A. Popovic, N.S. Azad, D.A. Laheru, L. Zheng, E.M. Jaffee,                    in the medical and biomedical field, polymer nanocomposites for 3D. 4D and 5D
      H. Wang, M. Yarchoan, Clin. Cancer Res. 26 (2020) 4842–4851.                                       Printing: Fundamental to Applications, Springer, 2025, pp. 293–320.
[205] J.B. Joiner, Y. Pylayeva-Gupta, P.A. Dayton, J. Immunol. 205 (2020) 2327–2341.               [249] B. Thiesen, A. Jordan, Int. J. Hyperther. 24 (2008) 467–474.
[206] K. Rangamuwa, T. Leong, S. Bozinovski, M. Christie, T. John, P. Antippa,                     [250] M. Johannsen, U. Gneveckow, L. Eckelt, A. Feussner, N. Waldofner, R. Scholz,
      L. Irving, D. Steinfort, Transl. Lung Cancer Res. 10 (2021) 2858.                                  S. Deger, P. Wust, S.A. Loening, A. Jordan, Int. J. Hyperther. 21 (2005) 637–647.
[207] Y. Wu, Q. Li, G. Shim, Y.-K. Oh, J. Contr. Release 330 (2021) 540–553.                       [251] J.L. Arias, L.H. Reddy, M. Othman, B. Gillet, D. Desmaele, F. Zouhiri, F. Dosio,
[208] D. Guo, Y. Chen, S. Wang, L. Yu, Y. Shen, H. Zhong, Y. Yang, Immunology 154                        R. Gref, P. Couvreur, ACS Nano 5 (2011) 1513–1521.
      (2018) 132–143.                                                                              [252] V. Plassat, C. Wilhelm, V. Marsaud, C. Ménager, F. Gazeau, J.M. Renoir,
[209] W.O. Maduabuchi, F.L. Tansi, R. Heller, I. Hilger, Biomedicines 11 (2023) 2256.                    S. Lesieur, Adv. Funct. Mater. 21 (2011) 83–92.
[210] K. Wang, H. Huang, Q. Zhan, H. Ding, Y. Li, MedComm 5 (2024) e549.                           [253] F. Zhang, G.B. Braun, A. Pallaoro, Y. Zhang, Y. Shi, D. Cui, M. Moskovits, D. Zhao,
[211] A. Brandli, K.A. Vessey, E.L. Fletcher, J. Neuroinflammation 21 (2024) 64.                         G.D. Stucky, Nano Lett. 12 (2012) 61–67.
[212] L. Mollica, F. De Marchis, A. Spitaleri, C. Dallacosta, D. Pennacchini, M. Zamai,            [254] S.H. Hu, R.H. Fang, Y.W. Chen, B.J. Liao, I.W. Chen, S.Y. Chen, Adv. Funct. Mater.
      A. Agresti, L. Trisciuoglio, G. Musco, M.E. Bianchi, Chem. Biol. 14 (2007)                         24 (2014) 4144–4155.
      431–441.                                                                                     [255] Y. Ma, X. Liang, S. Tong, G. Bao, Q. Ren, Z. Dai, Adv. Funct. Mater. 23 (2013)
[213] A. Sato, Y. Tamura, N. Sato, T. Yamashita, T. Takada, M. Sato, Y. Osai, M. Okura,                  815–822.
      I. Ono, A. Ito, Cancer Sci. 101 (2010) 1939–1946.                                            [256] M. Ahmed, C.L. Brace, F.T. Lee Jr., S.N. Goldberg, Radiology 258 (2011)
                                                                                                         351–369.
                                                                                              29
L. Xiong et al.                                                                                                                                  Materials Today Bio 33 (2025) 102070
[257] J.S. Rankin, G.E. Newman, T.M. Bashore, L.H. Muhlbaier, G.S. Tyson Jr, T.                 [266] Y. Du, X. Liu, Q. Liang, X.-J. Liang, J. Tian, Nano Lett. 19 (2019) 3618–3626.
      B. Ferguson Jr, J. Reves, D.C. Sabiston Jr, J. Thorac. Cardiovasc. Surg. 92 (1986)        [267] S.B. Somvanshi, P.B. Kharat, K. Jadhav, Surface functionalized
      832–846.                                                                                        superparamagnetic Zn-Mg ferrite nanoparticles for magnetic hyperthermia
[258] Z. Dong, L. Feng, Y. Hao, Q. Li, M. Chen, Z. Yang, H. Zhao, Z. Liu, Chem 6 (2020)               application towards noninvasive cancer treatment, in: Macromolecular Symposia,
      1391–1407.                                                                                      vol 400, Wiley Online Library, 2021 2100124.
[259] F. Gong, L. Cheng, N. Yang, O. Betzer, L. Feng, Q. Zhou, Y. Li, R. Chen,                  [268] Q. Tang, Y. Wang, B. Yan, J. Zhang, T. Wang, Y. Fang, Z. Ye, N. Zhang, N. Zhang,
      R. Popovtzer, Z. Liu, Adv. Mater. 31 (2019) 1900730.                                            Z. Wu, ACS Nano 18 (2024) 29804–29819.
[260] X. Han, J. Huang, X. Jing, D. Yang, H. Lin, Z. Wang, P. Li, Y. Chen, ACS Nano 12          [269] M. Wang, Q. Chen, D. Xu, Z. Yang, J. Chen, Y. Zhang, H. Chen, Nano Today 43
      (2018) 4545–4555.                                                                               (2022) 101374.
[261] S. Liang, X. Deng, G. Xu, X. Xiao, M. Wang, X. Guo, P.a. Ma, Z. Cheng, D. Zhang,          [270] A.S. Garanina, A.A. Nikitin, T.O. Abakumova, A.S. Semkina, A.O. Prelovskaya, V.
      J. Lin, Adv. Funct. Mater. 30 (2020) 1908598.                                                   A. Naumenko, A.S. Erofeev, P.V. Gorelkin, A.G. Majouga, M.A. Abakumov,
[262] X. Wang, X. Zhong, L. Bai, J. Xu, F. Gong, Z. Dong, Z. Yang, Z. Zeng, Z. Liu,                   Nanomaterials 12 (2021) 38.
      L. Cheng, J. Am. Chem. Soc. 142 (2020) 6527–6537.                                         [271] P. Zhou, H. Zhao, Q. Wang, Z. Zhou, J. Wang, G. Deng, X. Wang, Q. Liu, H. Yang,
[263] P. Zhu, Y. Chen, J. Shi, ACS Nano 12 (2018) 3780–3795.                                          S. Yang, Adv. Healthcare Mater. 7 (2018) 1701201.
[264] M.M. Sheha, A.M. Mostafa, D.G. Nasr, R.Y. Shahin, Sphinx Journal of                       [272] S. Shirvalilou, S. Khoei, S. Khoee, N.J. Raoufi, M.R. Karimi, A. Shakeri-Zadeh,
      Pharmaceutical and Medical Sciences 5 (2023) 1–32.                                              Chem. Biol. Interact. 295 (2018) 97–108.
[265] R. Hao, R. Xing, Z. Xu, Y. Hou, S. Gao, S. Sun, Adv. Mater. 22 (2010) 2729–2742.
30