Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 576
Filter
1.
Int J Mol Sci ; 25(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39125950

ABSTRACT

In vitro alternative therapy of human epidermoid squamous carcinoma (A431) by superparamagnetic hyperthermia (SPMHT) using Fe3O4 (magnetite) superparamagnetic nanoparticles (SPIONs) with an average diameter of 15.8 nm, bioconjugated with hydroxypropyl gamma-cyclodextrins (HP-γ-CDs) by means of polyacrylic acid (PAA) biopolymer, is presented in this paper. The therapy was carried out at a temperature of 43 °C for 30 min using the concentrations of Fe3O4 ferrimagnetic nanoparticles from nanobioconjugates of 1, 5, and 10 mg/mL nanoparticles in cell suspension, which were previously found by us to be non-toxic for healthy cells (cell viabilities close to 100%), according to ISO standards (cell viability must be greater than 70%). The temperature for the in vitro therapy was obtained by the safe application (without exceeding the biological limit and cellular damage) of an alternating magnetic field with a frequency of 312.4 kHz and amplitudes of 168, 208, and 370 G, depending on the concentration of the magnetic nanoparticles. The optimal concentration of magnetic nanoparticles in suspension was found experimentally. The results obtained after the treatment show its high effectiveness in destroying the A431 tumor cells, up to 83%, with the possibility of increasing even more, which demonstrates the viability of the SPMHT method with Fe3O4-PAA-(HP-γ-CDs) nanobioconjugates for human squamous cancer therapy.


Subject(s)
Carcinoma, Squamous Cell , Hyperthermia, Induced , Magnetite Nanoparticles , Skin Neoplasms , gamma-Cyclodextrins , Humans , Hyperthermia, Induced/methods , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Carcinoma, Squamous Cell/therapy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , gamma-Cyclodextrins/chemistry , Skin Neoplasms/therapy , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Cell Survival/drug effects , Nanoconjugates/chemistry
2.
Biomater Adv ; 163: 213948, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38959651

ABSTRACT

The use of nanoparticles has increased significantly over the past few years in a number of fields, including diagnostics, biomedicine, environmental remediation, and water treatment, generating public interest. Among various types of nanoparticles, magnetic nanoparticles (MNPs) have emerged as an essential tool for biomedical applications due to their distinct physicochemical properties compared to other nanoparticles. This review article focuses on the recent growth of MNPs and comprehensively reviews the advantages, multifunctional approaches, biomedical applications, and latest research on MNPs employed in various biomedical techniques. Biomedical applications of MNPs hold on to their ability to rapidly switch magnetic states under an external field at room temperature. Ideally, these MNPs should be highly susceptible to magnetization when the field is applied and then lose that magnetization just as quickly once the field is removed. This unique property allows MNPs to generate heat when exposed to high-frequency magnetic fields, making them valuable tools in developing treatments for hyperthermia and other heat-related illnesses. This review underscores the role of MNPs as tools that hold immense promise in transforming various aspects of healthcare, from diagnostics and imaging to therapeutic treatments, with discussion on a wide range of peer-reviewed articles published on the subject. At the conclusion of this work, challenges and potential future advances of MNPs in the biomedical field are highlighted.


Subject(s)
Magnetite Nanoparticles , Humans , Magnetite Nanoparticles/therapeutic use , Magnetite Nanoparticles/chemistry , Animals , Magnetic Fields , Hyperthermia, Induced/methods
3.
Biomed Phys Eng Express ; 10(5)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38981447

ABSTRACT

Magnetic nanoparticle (MNP)-mediated precision magnet therapy plays a crucial role in treating various diseases. This therapeutic strategy compensates for the limitations of low spatial resolution and low focusing of magnetic stimulation, and realizes the goal of wireless teletherapy with precise targeting of focal areas. This paper summarizes the preparation methods of magnetic nanomaterials, the properties of magnetic nanoparticles, the biological effects, and the measurement methods for detecting magnetism; discusses the research progress of precision magnetotherapy in the treatment of psychiatric disorders, neurological injuries, metabolic disorders, and bone-related disorders, and looks forward to the future development trend of precision magnet therapy.


Subject(s)
Magnetite Nanoparticles , Humans , Magnetite Nanoparticles/therapeutic use , Magnetite Nanoparticles/chemistry , Animals , Nanostructures , Magnetic Field Therapy/methods , Precision Medicine/methods , Mental Disorders/therapy
4.
Nanotheranostics ; 8(4): 442-457, 2024.
Article in English | MEDLINE | ID: mdl-38961886

ABSTRACT

The global incidence of cancer continues to rise, posing a significant public health concern. Although numerous cancer therapies exist, each has limitations and complications. The present study explores alternative cancer treatment approaches, combining hyperthermia and photodynamic therapy (PDT). Magnetic nanoparticles (MNPs) and amine-functionalized carbon quantum dots (A-CQDs) were synthesized separately and then covalently conjugated to form a single nanosystem for combinational therapy (M-CQDs). The successful conjugation was confirmed using zeta potential, Fourier transform infrared spectroscopy (FT-IR), and UV-visible spectroscopy. Morphological examination in transmission electron microscopy (TEM) further verified the conjugation of CQDs with MNPs. Energy dispersive X-ray spectroscopy (EDX) revealed that M-CQDs contain approximately 12 weight percentages of carbon. Hyperthermia studies showed that both MNP and M-CQDs maintain a constant therapeutic temperature at lower frequencies (260.84 kHz) with high specific absorption rates (SAR) of 118.11 and 95.04 W/g, respectively. In vitro studies demonstrated that MNPs, A-CQDs, and M-CQDs are non-toxic, and combinational therapy (PDT + hyperthermia) resulted in significantly lower cell viability (~4%) compared to individual therapies. Similar results were obtained with Hoechst and propidium iodide (PI) staining assays. Hence, the combination therapy of PDT and hyperthermia shows promise as a potential alternative to conventional therapies, and it could be further explored in combination with existing conventional treatments.


Subject(s)
Carbon , Hyperthermia, Induced , Magnetite Nanoparticles , Neoplasms , Photochemotherapy , Quantum Dots , Quantum Dots/chemistry , Photochemotherapy/methods , Humans , Carbon/chemistry , Hyperthermia, Induced/methods , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Neoplasms/therapy , Neoplasms/drug therapy , Cell Survival/drug effects , Cell Line, Tumor , Combined Modality Therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology
5.
Biomed Phys Eng Express ; 10(5)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39025085

ABSTRACT

Intratumoral multi-injection strategy enhances the efficacy of magnetic nanoparticle hyperthermia therapy (MNPH). In this study, criteria for the selection of injections and their location depending on the tumor shape/geometry are developed. The developed strategy is based on the thermal dosimetry results of different invasive 3D tumor models during MNPH simulation. MNPH simulations are conducted on physical tumor tissue models encased within healthy tissue. The tumor shapes are geometrically divided into a central tumor region containing maximum tumor volume and a peripheral tumor portion protruding in any random direction. The concepts of core and invasive radius are used to geometrically divide the tumor volume. Primary & secondary injections are used to inject MNP fluid into these respective tumor regions based on the invasiveness of the tumor. The optimization strategy is devised based on the zone of influence of primary & secondary injection. Results indicate that the zone of influence of secondary injection lies between 0.7 and 0.8 times the radial distance between the center of the tumor core and branch node point (extreme far endpoint on the invasive tumor surface). Additionally, the multi-injection strategy is more effective when the protrusion volume exceeds10%of the total volume. The proposed algorithm is used to devise multi-injection strategies for arbitrarily shaped tumors and will assist in pre-planning magnetic nanoparticle hyperthermia therapy.


Subject(s)
Hyperthermia, Induced , Neoplasms , Hyperthermia, Induced/methods , Humans , Neoplasms/therapy , Neoplasms/pathology , Magnetite Nanoparticles/therapeutic use , Computer Simulation , Algorithms , Models, Biological , Tumor Burden
6.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892406

ABSTRACT

According to data from the World Health Organization (WHO), cancer is considered to be one of the leading causes of death worldwide, and new therapeutic approaches, especially improved novel cancer treatment regimens, are in high demand. Considering that many chemotherapeutic drugs tend to have poor pharmacokinetic profiles, including rapid clearance and limited on-site accumulation, a combined approach with tumor-homing peptide (THP)-functionalized magnetic nanoparticles could lead to remarkable improvements. This is confirmed by an increasing number of papers in this field, showing that the on-target peptide functionalization of magnetic nanoparticles improves their penetration properties and ensures tumor-specific binding, which results in an increased clinical response. This review aims to highlight the potential applications of THPs in combination with magnetic carriers across various fields, including a pharmacoeconomic perspective.


Subject(s)
Antineoplastic Agents , Neoplasms , Peptides , Humans , Neoplasms/drug therapy , Peptides/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Animals , Drug Delivery Systems/methods , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Economics, Pharmaceutical , Drug Carriers/chemistry
7.
Biomed Phys Eng Express ; 10(4)2024 May 10.
Article in English | MEDLINE | ID: mdl-38692266

ABSTRACT

Magnetic nanoparticle hyperthermia (MNPH) has emerged as a promising cancer treatment that complements conventional ionizing radiation and chemotherapy. MNPH involves injecting iron-oxide nanoparticles into the tumor and exposing it to an alternating magnetic field (AMF). Iron oxide nanoparticles produce heat when exposed to radiofrequency AMF due to hysteresis loss. Minimizing the non-specific heating in human tissues caused by exposure to AMF is crucial. A pulse-width-modulated AMF has been shown to minimize eddy-current heating in superficial tissues. This project developed a control strategy based on a simplified mathematical model in MATLAB SIMULINK®to minimize eddy current heating while maintaining a therapeutic temperature in the tumor. A minimum tumor temperature of 43 [°C] is required for at least 30 [min] for effective hyperthermia, while maintaining the surrounding healthy tissues below 39 [°C]. A model predictive control (MPC) algorithm was used to reach the target temperature within approximately 100 [s]. As a constrained MPC approach, a maximum AMF amplitude of 36 [kA/m] and increment of 5 [kA/m/s] were applied. MPC utilized the AMF amplitude as an input and incorporated the open-loop response of the eddy current heating in its dynamic matrix. A conventional proportional integral (PI) controller was implemented and compared with the MPC performance. The results showed that MPC had a faster response (30 [s]) with minimal overshoot (1.4 [%]) than PI controller (115 [s] and 5.7 [%]) response. In addition, the MPC method performed better than the structured PI controller in its ability to handle constraints and changes in process parameters.


Subject(s)
Algorithms , Hyperthermia, Induced , Neoplasms , Hyperthermia, Induced/methods , Humans , Neoplasms/therapy , Magnetite Nanoparticles/therapeutic use , Magnetite Nanoparticles/chemistry , Computer Simulation , Magnetic Fields , Models, Theoretical , Temperature , Magnetic Iron Oxide Nanoparticles/chemistry , Models, Biological
8.
ACS Nano ; 18(23): 15284-15302, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38814737

ABSTRACT

Magnetic hyperthermia holds significant therapeutic potential, yet its clinical adoption faces challenges. One obstacle is the large-scale synthesis of high-quality superparamagnetic iron oxide nanoparticles (SPIONs) required for inducing hyperthermia. Robust and scalable manufacturing would ensure control over the key quality attributes of SPIONs, and facilitate clinical translation and regulatory approval. Therefore, we implemented a risk-based pharmaceutical quality by design (QbD) approach for SPION production using flame spray pyrolysis (FSP), a scalable technique with excellent batch-to-batch consistency. A design of experiments method enabled precise size control during manufacturing. Subsequent modeling linked the SPION size (6-30 nm) and composition to intrinsic loss power (ILP), a measure of hyperthermia performance. FSP successfully fine-tuned the SPION composition with dopants (Zn, Mn, Mg), at various concentrations. Hyperthermia performance showed a strong nonlinear relationship with SPION size and composition. Moreover, the ILP demonstrated a stronger correlation to coercivity and remanence than to the saturation magnetization of SPIONs. The optimal operating space identified the midsized (15-18 nm) Mn0.25Fe2.75O4 as the most promising nanoparticle for hyperthermia. The production of these nanoparticles on a pilot scale showed the feasibility of large-scale manufacturing, and cytotoxicity investigations in multiple cell lines confirmed their biocompatibility. In vitro hyperthermia studies with Caco-2 cells revealed that Mn0.25Fe2.75O4 nanoparticles induced 80% greater cell death than undoped SPIONs. The systematic QbD approach developed here incorporates process robustness, scalability, and predictability, thus, supporting the clinical translation of high-performance SPIONs for magnetic hyperthermia.


Subject(s)
Hyperthermia, Induced , Humans , Magnetic Iron Oxide Nanoparticles/chemistry , Particle Size , Cell Survival/drug effects , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use
9.
Ultrason Sonochem ; 107: 106928, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38820932

ABSTRACT

Rheumatoid arthritis (RA) is a chronic systemic inflammatory disease characterized by infiltration of inflammatory cells, hyperplasia of synovium, and destruction of the joint cartilage. Owing to the low drug delivery efficiency and limited immunosuppression effect, complete cure for RA remains a formidable challenge. Here, we show that live macrophages (Mφs) carrying protoporphyrin-loaded Fe3O4 nanoparticles can migrate to the RA tissues and inhibit the inflammation by sonodynamic therapy. The inflammation of RA leads to the release of cytokines, which guides the migration of the Mφs into the RA tissues, realizing precise delivery of therapeutics. The following sonodynamic therapy induced by ultrasound and protoporphyrin destructs the proliferating synovial cells and also infiltrated inflammatory cells, demonstrating significant therapeutic effect for RA. Meanwhile, the cytokines and relapse of RA can be remarkably suppressed because of the efficient damage to the resident inflammatory cells.


Subject(s)
Arthritis, Rheumatoid , Macrophages , Protoporphyrins , Ultrasonic Therapy , Arthritis, Rheumatoid/therapy , Macrophages/drug effects , Macrophages/metabolism , Protoporphyrins/chemistry , Protoporphyrins/pharmacology , Animals , Ultrasonic Therapy/methods , Mice , RAW 264.7 Cells , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Cytokines/metabolism , Humans
10.
Sci Rep ; 14(1): 10646, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38724530

ABSTRACT

Individual theranostic agents with dual-mode MRI responses and therapeutic efficacy have attracted extensive interest due to the real-time monitor and high effective treatment, which endow the providential treatment and avoid the repeated medication with side effects. However, it is difficult to achieve the integrated strategy of MRI and therapeutic drug due to complicated synthesis route, low efficiency and potential biosafety issues. In this study, novel self-assembled ultrasmall Fe3O4 nanoclusters were developed for tumor-targeted dual-mode T1/T2-weighted magnetic resonance imaging (MRI) guided synergetic chemodynamic therapy (CDT) and chemotherapy. The self-assembled ultrasmall Fe3O4 nanoclusters synthesized by facilely modifying ultrasmall Fe3O4 nanoparticles with 2,3-dimercaptosuccinic acid (DMSA) molecule possess long-term stability and mass production ability. The proposed ultrasmall Fe3O4 nanoclusters shows excellent dual-mode T1 and T2 MRI capacities as well as favorable CDT ability due to the appropriate size effect and the abundant Fe ion on the surface of ultrasmall Fe3O4 nanoclusters. After conjugation with the tumor targeting ligand Arg-Gly-Asp (RGD) and chemotherapy drug doxorubicin (Dox), the functionalized Fe3O4 nanoclusters achieve enhanced tumor accumulation and retention effects and synergetic CDT and chemotherapy function, which serve as a powerful integrated theranostic platform for cancer treatment.


Subject(s)
Magnetic Resonance Imaging , Theranostic Nanomedicine , Magnetic Resonance Imaging/methods , Theranostic Nanomedicine/methods , Animals , Mice , Humans , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Cell Line, Tumor , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/therapy , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Succimer/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology
11.
Biomolecules ; 14(5)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38785928

ABSTRACT

The combination of magnetic fields and magnetic nanoparticles (MNPs) to kill cancer cells by magneto-mechanical force represents a novel therapy, offering advantages such as non-invasiveness, among others. Pulsed magnetic fields (PMFs) hold promise for application in this therapy due to advantages such as easily adjustable parameters; however, they suffer from the drawback of narrow pulse width. In order to fully exploit the potential of PMFs and MNPs in this therapy, while maximizing therapeutic efficacy within the constraints of the narrow pulse width, a feature-matching theory is proposed, encompassing the matching of three aspects: (1) MNP volume and critical volume of Brownian relaxation, (2) relaxation time and pulse width, and (3) MNP shape and the intermittence of PMF. In the theory, a microsecond-PMF generator was developed, and four kinds of MNPs were selected for in vitro cell experiments. The results demonstrate that the killing rate of the experimental group meeting the requirements of the theory is at least 18% higher than the control group. This validates the accuracy of our theory and provides valuable guidance for the further application of PMFs in this therapy.


Subject(s)
Magnetic Fields , Melanoma , Humans , Cell Line, Tumor , Melanoma/pathology , Melanoma/therapy , Cell Survival/drug effects , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use
12.
Nanoscale ; 16(21): 10428-10440, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38742446

ABSTRACT

Due to the relatively low efficiency of magnetic hyperthermia and photothermal conversion, it is rather challenging for magneto-photothermal nanoagents to be used as an effective treatment during tumor hyperthermal therapy. The advancement of magnetic nanoparticles exhibiting a vortex-domain structure holds great promise as a viable strategy to enhance the application performance of conventional magnetic nanoparticles while retaining their inherent biocompatibility. Here, we report the development of Mn0.5Zn0.5Fe2O4 nanoflowers with ellipsoidal magnetic cores, and show them as effective nanoagents for magneto-photothermal synergistic therapy. Comparative studies were conducted on the heating performance of anisometric Mn0.5Zn0.5Fe2O4 (MZF) nanoparticles, including nanocubes (MZF-C), hollow spheres (MZF-HS), nanoflowers consisting of ellipsoidal magnetic cores (MZF-NFE), and nanoflowers consisting of needle-like magnetic cores (MZF-NFN). MZF-NFE exhibits an intrinsic loss parameter (ILP) of up to 15.3 N h m2 kg-1, which is better than that of commercial equivalents. Micromagnetic simulations reveal the magnetization configurations and reversal characteristics of the various MZF shapes. Additionally, all nanostructures displayed a considerable photothermal conversion efficiency rate of more than 18%. Our results demonstrated that by combining the dual exposure of MHT and PTT for hyperthermia treatments induced by MZF-NFE, BT549, MCF-7, and 4T1 cell viability can be significantly decreased by ∼95.7% in vitro.


Subject(s)
Photothermal Therapy , Mice , Animals , Humans , Cell Line, Tumor , Hyperthermia, Induced , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Cell Survival/drug effects , Infrared Rays , Neoplasms/therapy , Neoplasms/pathology , Female , MCF-7 Cells
13.
Mater Horiz ; 11(13): 3157-3165, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38629215

ABSTRACT

Magnetic propulsion of nano-/micro-robots is an effective way to treat implant-associated infections by physically destroying biofilm structures to enhance antibiotic killing. However, it is hard to precisely control the propulsion in vivo. Magnetic-nanoparticle coating that can be magnetically pulled off does not need precise control, but the requirement of adhesion stability on an implant surface restricts its magnetic responsiveness. Moreover, whether the coating has been fully pulled-off or not is hard to ensure in real-time in vivo. Herein, composited silk fibroins (SFMA) are optimized to stabilize Fe3O4 nanoparticles on a titanium surface in a dry environment; while in an aqueous environment, the binding force of SFMA on titanium is significantly reduced due to hydrophilic interaction, making the coating magnetically controllable by an externally-used magnet but still stable in the absence of a magnet. The maximum working distance of the magnet can be calculated using magnetomechanical simulation in which the yielding magnetic traction force is strong enough to pull Fe3O4 nanoparticles off the surface. The pulling-off removes the biofilms that formed on the coating and enhances antibiotic killing both in vitro and in a rat sub-cutaneous implant model by up to 100 fold. This work contributes to the practical knowledge of magnetic propulsion for biofilm treatment.


Subject(s)
Biofilms , Fibroins , Titanium , Biofilms/drug effects , Animals , Rats , Titanium/pharmacology , Titanium/chemistry , Fibroins/chemistry , Fibroins/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Magnetite Nanoparticles/therapeutic use , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/chemistry , Prostheses and Implants , Rats, Sprague-Dawley , Surface Properties , Staphylococcus aureus/drug effects
14.
Molecules ; 29(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38675647

ABSTRACT

This study aimed to develop multifunctional nanoplatforms for both cancer imaging and therapy using superparamagnetic iron oxide nanoparticles (SPIONs). Two distinct synthetic methods, reduction-precipitation (MR/P) and co-precipitation at controlled pH (MpH), were explored, including the assessment of the coating's influence, namely dextran and gold, on their magnetic properties. These SPIONs were further functionalized with gadolinium to act as dual T1/T2 contrast agents for magnetic resonance imaging (MRI). Parameters such as size, stability, morphology, and magnetic behavior were evaluated by a detailed characterization analysis. To assess their efficacy in imaging and therapy, relaxivity and hyperthermia experiments were performed, respectively. The results revealed that both synthetic methods lead to SPIONs with similar average size, 9 nm. Mössbauer spectroscopy indicated that samples obtained from MR/P consist of approximately 11-13% of Fe present in magnetite, while samples obtained from MpH have higher contents of 33-45%. Despite coating and functionalization, all samples exhibited superparamagnetic behavior at room temperature. Hyperthermia experiments showed increased SAR values with higher magnetic field intensity and frequency. Moreover, the relaxivity studies suggested potential dual T1/T2 contrast agent capabilities for the coated SPpH-Dx-Au-Gd sample, thus demonstrating its potential in cancer diagnosis.


Subject(s)
Contrast Media , Magnetic Iron Oxide Nanoparticles , Magnetic Resonance Imaging , Magnetite Nanoparticles , Theranostic Nanomedicine , Magnetic Iron Oxide Nanoparticles/chemistry , Magnetic Resonance Imaging/methods , Contrast Media/chemistry , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Humans , Gold/chemistry , Dextrans/chemistry , Gadolinium/chemistry , Surface Properties , Hyperthermia, Induced/methods , Particle Size
15.
J Biomater Appl ; 39(1): 3-23, 2024 07.
Article in English | MEDLINE | ID: mdl-38606627

ABSTRACT

Hyperthermia therapy refers to the elevating of a region in the body for therapeutic purposes. Different techniques have been applied for hyperthermia therapy including laser, microwave, radiofrequency, ultrasonic, and magnetic nanoparticles and the latter have received great attention in recent years. Magnetic hyperthermia in cancer therapy aims to increase the temperature of the body tissue by locally delivering heat from the magnetic nanoparticles to cancer cells with the aid of an external alternating magnetic field to kill the cancerous cells or prevent their further growth. This review introduces magnetic hyperthermia with magnetic nanoparticles. It includes the mechanism of the operation and magnetism behind the magnetic hyperthermia phenomenon. Different synthesis methods and surface modification to enhance the biocompatibility, water solubility, and stability of the nanoparticles in physiological environments have been discussed. Recent research on versatile types of magnetic nanoparticles with their ability to increase the local temperature has been addressed.


Subject(s)
Hyperthermia, Induced , Magnetite Nanoparticles , Neoplasms , Humans , Hyperthermia, Induced/methods , Neoplasms/therapy , Animals , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Magnetic Fields
16.
Nanoscale ; 16(16): 7892-7907, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38568096

ABSTRACT

Magnetic hyperthermia-based cancer therapy (MHCT) holds great promise as a non-invasive approach utilizing heat generated by an alternating magnetic field for effective cancer treatment. For an efficacious therapeutic response, it is crucial to deliver therapeutic agents selectively at the depth of tumors. In this study, we present a new strategy using the naturally occurring tumor-colonizing bacteria Escherichia coli (E. coli) as a carrier to deliver magnetic nanoparticles to hypoxic tumor cores for effective MHCT. Self-propelling delivery agents, "nano-bacteriomagnets" (BacMags), were developed by incorporating anisotropic magnetic nanocubes into E. coli which demonstrated significantly improved hyperthermic performance, leading to an impressive 85% cell death in pancreatic cancer. The in vivo anti-cancer response was validated in a syngeneic xenograft model with a 50% tumor inhibition rate within 20 days and a complete tumor regression within 30 days. This proof-of-concept study demonstrates the potential of utilizing anaerobic bacteria for the delivery of magnetic nanocarriers as a smart therapeutic approach for enhanced MHCT.


Subject(s)
Escherichia coli , Hyperthermia, Induced , Magnetite Nanoparticles , Pancreatic Neoplasms , Animals , Mice , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Humans , Cell Line, Tumor , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays
17.
IEEE Trans Biomed Eng ; 71(8): 2528-2536, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38498750

ABSTRACT

Magnetic Particle Imaging (MPI)-guided Magnetic Fluid Hyperthermia (MFH) has the potential for widespread utilization, as it allows for the prediction of magnetothermal dosage, real-time visualization of the thermal therapy process, and precise localization of the lesion area. However, the existing MPI-guided MFH (MPI-MFH) method is insensitive to concentration gradients of magnetic nanoparticles (MNPs) and is susceptible to causing damage to normal tissues with high MNP concentrations during MFH treatment, while inadequately heating tumor tissues with lower MNP concentrations. In this work, we established a relationship between MNP concentration and heating efficiency through simulations and phantom measurements, enabling the optimal selection of MFH parameters guided by MPI. Based on these findings, we developed a high-gradient field MPI-MFH method using a field-free point (FFP) approach to achieve precise local heating. Phantom experiments and in vivo glioma model experiments were conducted to validate this proposed method. The results demonstrated that the proposed method of MPI-MFH can improve the MNP concentration gradient sensitivity to ±1 mg/ml, thereby enabling more effective lesion-site heating without damaging normal tissues. This method not only reduced glioma size effectively but also holds promise for application in various other types of cancers.


Subject(s)
Glioma , Hyperthermia, Induced , Magnetite Nanoparticles , Phantoms, Imaging , Hyperthermia, Induced/methods , Animals , Glioma/diagnostic imaging , Glioma/therapy , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Mice , Cell Line, Tumor , Rats , Brain Neoplasms/diagnostic imaging
18.
Adv Mater ; 36(26): e2309770, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38447017

ABSTRACT

Percutaneous thermotherapy, a minimally invasive operational procedure, is employed in the ablation of deep tumor lesions by means of target-delivering heat. Conventional thermal ablation methods, such as radiofrequency or microwave ablation, to a certain extent, are subjected to extended ablation time as well as biosafety risks of unwanted overheating. Given its effectiveness and safety, percutaneous thermotherapy gains a fresh perspective, thanks to magnetic hyperthermia. In this respect, an injectable- and magnetic-hydrogel-construct-based thermal ablation agent is likely to be a candidate for the aforementioned clinical translation. Adopting a simple and environment-friendly strategy, a magnetic colloidal hydrogel injection is introduced by a binary system comprising super-paramagnetic Fe3O4 nanoparticles and gelatin nanoparticles. The colloidal hydrogel constructs, unlike conventional bulk hydrogel, can be easily extruded through a percutaneous needle and then self-heal in a reversible manner owing to the unique electrostatic cross-linking. The introduction of magnetic building blocks is exhibited with a rapid magnetothermal response to an alternating magnetic field. Such hydrogel injection is capable of generating heat without limitation of deep penetration. The materials achieve outstanding therapeutic results in mouse and rabbit models. These findings constitute a new class of locoregional interventional thermal therapies with minimal collateral damages.


Subject(s)
Carcinoma, Hepatocellular , Colloids , Hydrogels , Liver Neoplasms , Animals , Rabbits , Mice , Hydrogels/chemistry , Liver Neoplasms/therapy , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/pathology , Colloids/chemistry , Gelatin/chemistry , Humans , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Hyperthermia, Induced/methods , Cell Line, Tumor , Injections , Magnetic Iron Oxide Nanoparticles/chemistry
19.
Adv Sci (Weinh) ; 11(21): e2308993, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38516757

ABSTRACT

Neural stem cells (NSCs) transplantation is an attractive and promising treatment strategy for spinal cord injury (SCI). Various pathological processes including the severe inflammatory cascade and difficulty in stable proliferation and differentiation of NSCs limit its application and translation. Here, a novel physico-chemical bifunctional neural stem cells delivery system containing magnetic nanoparticles (MNPs and methylprednisolone (MP) is designed to repair SCI, the former regulates NSCs differentiation through magnetic mechanical stimulation in the chronic phase, while the latter alleviates inflammatory response in the acute phase. The delivery system releases MP to promote microglial M2 polarization, inhibit M1 polarization, and reduce neuronal apoptosis. Meanwhile, NSCs tend to differentiate into functional neurons with magnetic mechanical stimulation generated by MNPs in the static magnetic field, which is related to the activation of the PI3K/AKT/mTOR pathway. SCI mice achieve better functional recovery after receiving NSCs transplantation via physico-chemical bifunctional delivery system, which has milder inflammation, higher number of M2 microglia, more functional neurons, and axonal regeneration. Together, this bifunctional NSCs delivery system combined physical mechanical stimulation and chemical drug therapy is demonstrated to be effective, which provides new treatment insights into clinical transformation of SCI repair.


Subject(s)
Disease Models, Animal , Magnetite Nanoparticles , Methylprednisolone , Neural Stem Cells , Spinal Cord Injuries , Animals , Spinal Cord Injuries/therapy , Methylprednisolone/pharmacology , Mice , Neural Stem Cells/transplantation , Neural Stem Cells/drug effects , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Cell Differentiation/drug effects , Stem Cell Transplantation/methods
20.
Expert Opin Drug Deliv ; 21(4): 521-535, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38555483

ABSTRACT

INTRODUCTION: Central nervous system (CNS)-related disorders are increasingly being recognized as a global health challenge worldwide. There are significant challenges for effective diagnosis and treatment due to the presence of the CNS barriers which impede the management of neurological diseases. Combination of nanovesicles (NVs) and magnetic nanoparticles (MNPs), referred to as magnetic nanovesicles (MNVs), is now well suggested as a potential theranostic option for improving the management of neurological disorders with increased targeting efficiency and minimized side effects. AREAS COVERED: This review provides a summary of major CNS disorders and the physical barriers limiting the access of imaging/therapeutic agents to the CNS environment. A special focus on the unique features of MNPs and NV is discussed which make them attractive candidates for neuro-nanomedicine. Furthermore, a deeper understanding of MNVs as a promising combined strategy for diagnostic and/or therapeutic purposes in neurological disorders is provided. EXPERT OPINION: The multifunctionality of MNVs offers the ability to overcome the CNS barriers and can be used to monitor the effectiveness of treatment. The insights provided will guide future research toward better outcomes and facilitate the development of next-generation, innovative treatments for CNS disorders.


Subject(s)
Central Nervous System Diseases , Drug Delivery Systems , Magnetite Nanoparticles , Theranostic Nanomedicine , Humans , Central Nervous System Diseases/diagnosis , Central Nervous System Diseases/drug therapy , Animals , Magnetite Nanoparticles/therapeutic use , Magnetite Nanoparticles/chemistry , Theranostic Nanomedicine/methods , Blood-Brain Barrier/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL