Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biomech Eng ; 146(1)2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37773642

RESUMO

High-grade gliomas (HGG) are the most common primary brain malignancy and continue to be associated with a dismal prognosis (median survival rate of 15-18 months) with standard of care therapy. Magnetic hyperthermia therapy (MHT) is an emerging intervention that leverages the ferromagnetic properties of magnetic iron-oxide nanoparticles (MIONPs) to target cancer cells that are otherwise left behind after resection. We report a novel port device to facilitate localization, delivery, and temperature measurement of MIONPs within a target lesion for MHT therapy. We conducted an in-depth literature and intellectual property review to define specifications of the conceived port device. After setting the design parameters, a thorough collaboration with neurological surgeons guided the iterative modeling process. A prototype was developed using Fusion 360 (Autodesk, San Rafael, CA) and printed on a Form 3 printer (Formlabs, Medford, MA) in Durable resin. The prototype was then tested in a phantom skull printed on a Pro-Jet 660Pro 3D printer (3D Systems, Rock Hill, SC) and a brain model based on mechanical and electrochemical properties of native brain tissue. This phantom underwent MHT heating tests using an alternating magnetic field (AMF) sequence based on current MHT workflow. Successful localization, delivery, and temperature measurement were demonstrated. The purpose of this study was twofold: first, to create and validate the procedural framework for a novel device, providing the groundwork for an upcoming comprehensive animal trial and second, to elucidate a cooperative approach between engineers and clinicians that propels advancements in medical innovation.


Assuntos
Neoplasias Encefálicas , Glioma , Hipertermia Induzida , Animais , Glioma/terapia , Neoplasias Encefálicas/terapia , Encéfalo , Fenômenos Magnéticos
2.
Int J Hyperthermia ; 40(1): 2272067, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37875265

RESUMO

PURPOSE: Magnetic particle hyperthermia is an approved cancer treatment that harnesses thermal energy generated by magnetic nanoparticles when they are exposed to an alternating magnetic field (AMF). Thermal stress is either directly cytotoxic or increases the susceptibility of cancer cells to standard therapies, such as radiation. As with other thermal therapies, the challenge with nanoparticle hyperthermia is controlling energy delivery. Here, we describe the design and implementation of a prototype pre-clinical device, called HYPER, that achieves spatially confined nanoparticle heating within a user-selected volume and location. DESIGN: Spatial control of nanoparticle heating was achieved by placing an AMF generating coil (340 kHz, 0-15 mT), between two opposing permanent magnets. The relative positions between the magnets determined the magnetic field gradient (0.7 T/m-2.3 T/m), which in turn governed the volume of the field free region (FFR) between them (0.8-35 cm3). Both the gradient value and position of the FFR within the AMF ([-14, 14]x, [-18, 18]y, [-30, 30]z) mm are values selected by the user via the graphical user interface (GUI). The software then controls linear actuators that move the static magnets to adjust the position of the FFR in 3D space based on user input. Within the FFR, the nanoparticles generate hysteresis heating; however, outside the FFR where the static field is non-negligible, the nanoparticles are unable to generate hysteresis loss power. VERIFICATION: We verified the performance of the HYPER to design specifications by independently heating two nanoparticle-rich areas of a phantom placed within the volume occupied by the AMF heating coil.


Assuntos
Antineoplásicos , Hipertermia Induzida , Nanopartículas , Temperatura Alta , Campos Magnéticos
3.
Int J Mol Sci ; 24(8)2023 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-37108659

RESUMO

Liver transplantation is the only treatment for hepatic insufficiency as a result of acute and chronic liver injuries/pathologies that fail to recover. Unfortunately, there remains an enormous and growing gap between organ supply and demand. Although recipients on the liver transplantation waitlist have significantly higher mortality, livers are often not allocated because they are (i) classified as extended criteria or marginal livers and (ii) subjected to longer cold preservation time (>6 h) with a direct correlation of poor outcomes with longer cold ischemia. Downregulating the recipient's innate immune response to successfully tolerate a graft having longer cold ischemia times or ischemia-reperfusion injury through induction of immune tolerance in the graft and the host would significantly improve organ utilization and post-transplant outcomes. Broadly, technologies proposed for development aim to extend the life of the transplanted liver through post-transplant or recipient conditioning. In this review, we focus on the potential benefits of nanotechnology to provide unique pre-transplant grafting and recipient conditioning of extended criteria donor livers using immune tolerance induction and hyperthermic pre-conditioning.


Assuntos
Falência Hepática , Transplante de Fígado , Traumatismo por Reperfusão , Humanos , Fígado , Doadores de Tecidos , Traumatismo por Reperfusão/terapia , Preservação de Órgãos
4.
Antimicrob Agents Chemother ; 66(4): e0239921, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35293784

RESUMO

Cryptococcosis is a devastating fungal disease associated with high morbidity and mortality even when treated with antifungal drugs. Bionized nanoferrite (BNF) nanoparticles are powerful immunomodulators, but their efficacy for infectious diseases has not been investigated. Administration of BNF nanoparticles to mice with experimental cryptococcal pneumonia altered the outcome of infection in a dose response manner as measured by CFU and survival. The protective effects were higher at lower doses, with reductions in IL-2, IL-4, and TNF-α, consistent with immune modulation whereby reductions in inflammation translate into reduced host damage, clearance of infection, and longer survival.


Assuntos
Criptococose , Cryptococcus neoformans , Animais , Criptococose/tratamento farmacológico , Criptococose/microbiologia , Inflamação , Camundongos , Fator de Necrose Tumoral alfa
5.
Int J Mol Sci ; 23(24)2022 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-36555306

RESUMO

A promise of cancer nanomedicine is the "targeted" delivery of therapeutic agents to tumors by the rational design of nanostructured materials. During the past several decades, a realization that in vitro and in vivo preclinical data are unreliable predictors of successful clinical translation has motivated a reexamination of this approach. Mathematical models of drug pharmacokinetics (PK) and biodistribution (BD) are essential tools for small-molecule drugs development. A key assumption underlying these models is that drug-target binding kinetics dominate blood clearance, hence recognition by host innate immune cells is not explicitly included. Nanoparticles circulating in the blood are conspicuous to phagocytes, and inevitable interactions typically trigger active biological responses to sequester and remove them from circulation. Our recent findings suggest that, instead of referring to nanoparticles as designed for active or passive "tumor targeting", we ought rather to refer to immune cells residing in the tumor microenvironment (TME) as active or passive actors in an essentially "cell-mediated tumor retention" process that competes with active removal by other phagocytes. Indeed, following intravenous injection, nanoparticles induce changes in the immune compartment of the TME because of nanoparticle uptake, irrespective of the nature of tumor targeting moieties. In this study, we propose a 6-compartment PK model as an initial mathematical framework for modeling this tumor-associated immune cell-mediated retention. Published in vivo PK and BD results obtained with bionized nanoferrite® (BNF®) nanoparticles were combined with results from in vitro internalization experiments with murine macrophages to guide simulations. As a preliminary approximation, we assumed that tumor-associated macrophages (TAMs) are solely responsible for active retention in the TME. We model the TAM approximation by relating in vitro macrophage uptake to an effective macrophage avidity term for the BNF® nanoparticles under consideration.


Assuntos
Nanopartículas , Nanoestruturas , Neoplasias , Camundongos , Animais , Distribuição Tecidual , Macrófagos/metabolismo , Neoplasias/terapia , Nanopartículas/química , Microambiente Tumoral
6.
Int J Hyperthermia ; 38(1): 611-622, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33853493

RESUMO

PURPOSE: Alternating magnetic field (AMF) tissue interaction models are generally not validated. Our aim was to develop and validate a coupled electromagnetic and thermal model for estimating temperatures in large organs during magnetic nanoparticle hyperthermia (MNH). MATERIALS AND METHODS: Coupled finite element electromagnetic and thermal model validation was performed by comparing the results to experimental data obtained from temperatures measured in homogeneous agar gel phantoms exposed to an AMF at fixed frequency (155 ± 10 kHz). The validated model was applied to a three-dimensional (3D) rabbit liver built from computed tomography (CT) images to investigate the contribution of nanoparticle heating and nonspecific eddy current heating as a function of AMF amplitude. RESULTS: Computed temperatures from the model were in excellent agreement with temperatures calculated using the analytical method (error < 1%) and temperatures measured in phantoms (maximum absolute error <2% at each probe location). The 3D rabbit liver model for a fixed concentration of 5 mg Fe/cm3 of tumor revealed a maximum temperature ∼44 °C in tumor and ∼40 °C in liver at AMF amplitude of ∼12 kA/m (peak). CONCLUSION: A validated coupled electromagnetic and thermal model was developed to estimate temperatures due to eddy current heating in homogeneous tissue phantoms. The validated model was successfully used to analyze temperature distribution in complex rabbit liver tumor geometry during MNH. In future, model validation should be extended to heterogeneous tissue phantoms, and include heat sink effects from major blood vessels.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita , Animais , Fenômenos Eletromagnéticos , Hipertermia , Coelhos , Temperatura
7.
Int J Hyperthermia ; 37(3): 100-107, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33426988

RESUMO

BACKGROUND: Magnetic fluid heating has great potential in the fields of thermal medicine and cryopreservation. However, variations among experimental parameters, analysis methods and experimental uncertainty make quantitative comparisons of results among laboratories difficult. Herein, we focus on the impact of calculating the specific absorption rate (SAR) using Time-Rise and Box-Lucas fitting. Time-Rise assumes adiabatic conditions, which is experimentally unachievable, but can be reasonably assumed (quasi-adiabatic) only for specific and limited evaluation times when heat loss is negligible compared to measured heating rate. Box-Lucas, on the other hand, accounts for heat losses but requires longer heating. METHODS: Through retrospective analysis of data obtained from two laboratories, we demonstrate measurement time is a critical parameter to consider when calculating SAR. Volumetric SAR were calculated using the two methods and compared across multiple iron-oxide nanoparticles. RESULTS: We observed the lowest volumetric SAR variation from both fitting methods between 1-10 W/mL, indicating an ideal SAR range for heating measurements. Furthermore, our analysis demonstrates that poorly chosen fitting method can generate reproducible but inaccurate SAR. CONCLUSION: We provide recommendations to select measurement time for data analysis with either Modified Time-Rise or Box-Lucas method, and suggestions to enhance experimental precision and accuracy when conducting heating experiments.


Assuntos
Nanopartículas de Magnetita , Calefação , Temperatura Alta , Magnetismo , Estudos Retrospectivos
8.
Int J Hyperthermia ; 37(3): 108-119, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33426990

RESUMO

PURPOSE: Tumor volume largely determines the success of local control of borderline resectable and locally advanced pancreatic cancer with current therapy. We hypothesized that a tumor-mass normalized dose of magnetic nanoparticle hyperthermia (MNPH) with alternating magnetic fields (AMFs) reduces the effect of tumor volume for treatment. METHODS: 18 female athymic nude mice bearing subcutaneous MiaPaCa02 human xenograft tumors were treated with MNPH following intratumor injections of 5.5 mg Fe/g tumor of an aqueous suspension of magnetic iron-oxide nanoparticles. Mice were randomly divided into control (n = 5) and treated groups having small (0.15 ± 0.03 cm3, n = 4) or large (0.30 ± 0.06 cm3, n = 5) tumors. We assessed the clinical feasibility of this approach and of pulsed AMF to minimize eddy current heating using a finite-element method to solve a bioheat equation for a human-scale multilayer model. RESULTS: Compared to the control group, both small and large MiaPaCa02 subcutaneous tumors showed statistically significant growth inhibition. Conversely, there was no significant difference in tumor growth between large and small tumors. Both computational and xenograft models demonstrated higher maximum tumor temperatures for large tumors compared to small tumors. Computational modeling demonstrates that pulsed AMF can minimize nonspecific eddy current heating. CONCLUSIONS: MNPH provides an advantage to treat large tumors because the MION dose can be adjusted to increase power. Pulsed AMF, with adjusted treatment time, can enhance MNPH in challenging cases such as low MION dose in the target tissue and/or large patients by minimizing nonspecific eddy current heating without sacrificing thermal dose to the target. Nanoparticle heterogeneity in tumors remains a challenge for continued research.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita , Neoplasias Pancreáticas , Animais , Feminino , Calefação , Humanos , Hipertermia , Nanopartículas de Magnetita/uso terapêutico , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/terapia
9.
Int J Hyperthermia ; 37(3): 59-75, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33426997

RESUMO

OBJECTIVE: Toxicity from off-target heating with magnetic hyperthermia (MHT) is generally assumed to be understood. MHT research focuses on development of more potent heating magnetic iron oxide nanoparticles (MIONs), yet our understanding of factors that define biodistribution following systemic delivery remains limited. Preclinical development relies on mouse models, thus understanding off-target heating with MHT in mice provides critical knowledge for clinical development. METHODS: Eight-week old female nude mice received a single tail vein injection of bionized nanoferrite (BNF) MIONs or a counterpart labeled with a polyclonal human antibody (BNF-IgG) at 1 mg, 3 mg or 5 mg Fe/mouse on day 1. On day 3, mice were exposed to an alternating magnetic field (AMF) having amplitude of 32, 48 or 64 kA/m at ∼145 kHz for 20 min. Twenty-four hours later, blood, livers and spleens were harvested and analyzed. RESULTS: Damage to livers was apparent by histology and serum liver enzymes following MHT with BNF or BNF-IgG at doses ≥3 mg Fe and AMF amplitudes ≥48 kA/m. Differences between effects with BNF vs. BNF-IgG at a dose of 3 mg Fe were noted in all measures, with less damage and increased survival occurring in mice injected with BNF-IgG. Necropsies revealed severe damage to duodenum and upper small intestines, likely the immediate cause of death at the highest MHT doses. CONCLUSION: Results demonstrate that the MION coating affects biodistribution, which in turn determines off-target effects. Developments to improve heating capabilities of MIONs may be clinically irrelevant without better control of biodistribution.


Assuntos
Hipertermia Induzida , Nanopartículas , Animais , Feminino , Camundongos , Hipertermia Induzida/efeitos adversos , Campos Magnéticos , Nanopartículas Magnéticas de Óxido de Ferro , Camundongos Nus , Distribuição Tecidual
10.
Int J Hyperthermia ; 37(1): 1-14, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31918595

RESUMO

Purpose: We describe a modified Helmholtz induction coil, or Maxwell coil, that generates alternating magnetic fields (AMF) having field uniformity (≤10%) within a = 3000 cm3 volume of interest for magnetic hyperthermia research.Materials and methods: Two-dimensional finite element analysis (2D-FEA) was used for electromagnetic design of the induction coil set and to develop specifications for the required matching network. The matching network and induction coil set were fabricated using best available practices and connected to a 120 kW industrial induction heating power supply. System performance was evaluated by magnetic field mapping with a magnetic field probe, and tests were performed using gel phantoms.Results: Tests verified that the system generated a target peak AMF amplitude along the coil axis of ∼35 kA/m (peak) at a frequency of 150 ± 10 kHz while maintaining field uniformity to >90% of peak for a volume of ∼3000 cm3.Conclusions: The induction coil apparatus comprising three independent loops, i.e., Maxwell-type improves upon the performance of simple solenoid and Helmholtz coils by providing homogeneous flux density fields within a large volume while minimizing demands on power and stray fields. Experiments with gel phantoms and analytical calculations show that future translational research efforts should be devoted to developing strategies to reduce the impact of nonspecific tissue heating from eddy currents; and, that an inductor producing a homogeneous field has significant clinical potential for deep-tissue magnetic fluid hyperthermia.


Assuntos
Fenômenos Eletromagnéticos , Nanopartículas de Magnetita/normas , Humanos , Hipertermia Induzida/métodos
11.
Int J Hyperthermia ; 37(1): 711-741, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32579419

RESUMO

The therapeutic application of heat is very effective in cancer treatment. Both hyperthermia, i.e., heating to 39-45 °C to induce sensitization to radiotherapy and chemotherapy, and thermal ablation, where temperatures beyond 50 °C destroy tumor cells directly are frequently applied in the clinic. Achievement of an effective treatment requires high quality heating equipment, precise thermal dosimetry, and adequate quality assurance. Several types of devices, antennas and heating or power delivery systems have been proposed and developed in recent decades. These vary considerably in technique, heating depth, ability to focus, and in the size of the heating focus. Clinically used heating techniques involve electromagnetic and ultrasonic heating, hyperthermic perfusion and conductive heating. Depending on clinical objectives and available technology, thermal therapies can be subdivided into three broad categories: local, locoregional, or whole body heating. Clinically used local heating techniques include interstitial hyperthermia and ablation, high intensity focused ultrasound (HIFU), scanned focused ultrasound (SFUS), electroporation, nanoparticle heating, intraluminal heating and superficial heating. Locoregional heating techniques include phased array systems, capacitive systems and isolated perfusion. Whole body techniques focus on prevention of heat loss supplemented with energy deposition in the body, e.g., by infrared radiation. This review presents an overview of clinical hyperthermia and ablation devices used for local, locoregional, and whole body therapy. Proven and experimental clinical applications of thermal ablation and hyperthermia are listed. Methods for temperature measurement and the role of treatment planning to control treatments are discussed briefly, as well as future perspectives for heating technology for the treatment of tumors.


Assuntos
Hipertermia Induzida , Neoplasias , Calefação , Temperatura Alta , Humanos , Neoplasias/terapia , Tecnologia
12.
Int J Hyperthermia ; 36(1): 115-129, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30541354

RESUMO

PURPOSE: To study, with computational models, the utility of power modulation to reduce tissue temperature heterogeneity for variable nanoparticle distributions in magnetic nanoparticle hyperthermia. METHODS: Tumour and surrounding tissue were modeled by elliptical two- and three-dimensional computational phantoms having six different nanoparticle distributions. Nanoparticles were modeled as point heat sources having amplitude-dependent loss power. The total number of nanoparticles was fixed, and their spatial distribution and heat output were varied. Heat transfer was computed by solving the Pennes' bioheat equation using finite element methods (FEM) with temperature-dependent blood perfusion. Local temperature was regulated using a proportional-integral-derivative (PID) controller. Tissue temperature, thermal dose and tissue damage were calculated. The required minimum thermal dose delivered to the tumor was kept constant, and heating power was adjusted for comparison of both the heating methods. RESULTS: Modulated power heating produced lower and more homogeneous temperature distributions than did constant power heating for all studied nanoparticle distributions. For a concentrated nanoparticle distribution, located off-center within the tumor, the maximum temperatures inside the tumor were 16% lower for modulated power heating when compared to constant power heating. This resulted in less damage to surrounding normal tissue. Modulated power heating reached target thermal doses up to nine-fold more rapidly when compared to constant power heating. CONCLUSIONS: Controlling the temperature at the tumor-healthy tissue boundary by modulating the heating power of magnetic nanoparticles demonstrably compensates for a variable nanoparticle distribution to deliver effective treatment.


Assuntos
Nanopartículas/química , Simulação por Computador , Humanos , Hipertermia Induzida/métodos , Magnetismo
13.
Int J Hyperthermia ; 36(1): 712-720, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31345068

RESUMO

Purpose: A proposed mechanism for the enhanced effectiveness of hyperthermia and doxorubicin (Dox) combinations is increased intracellular Dox concentrations resulting from heat-induced cell stress. The purpose of this study was to determine whether specific varied Dox and heat combinations produce measurable effects greater than the additive combination, and whether these effects can be attributed to heat-induced increases in intracellular Dox concentrations. Methods: HCT116, HT29 and CT26 cells were exposed to Dox and water bath heating independently. A clonogenic survival assay was used to determine cell killing and intracellular Dox concentrations were measured in HCT116 cells with mass spectrometry. Cells were exposed to heating at 42 °C (60 min) and 0.5 µg/ml of Dox at varying intervals. Synergy was determined by curve-fitting and isobologram analysis. Results: All cell lines displayed synergistic effects of combined heating and Dox. A maximum synergistic effect was achieved with simultaneous cell exposure to Dox and heat. For exposures at 42 °C, the synergistic effect was most pronounced at Dox concentrations <0.5 µg/ml. Increased intracellular concentrations of Dox in HCT116 cells caused by heat-stress did not generate a concomitant thermal enhancement. Conclusions: Simultaneous exposure of HCT116 cells to heating and Dox is more effective than sequential exposure. Heat-induced cell responses are accompanied by increased intracellular Dox concentrations; however, clonogenic survival data do not support this as the cause for synergistic cytotoxicity.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Temperatura Alta , Transporte Biológico , Morte Celular , Linhagem Celular Tumoral , Humanos
14.
Int J Hyperthermia ; 36(sup1): 47-63, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31795835

RESUMO

Purpose: Enhancing immune responses in triple negative breast cancers (TNBCs) remains a challenge. Our study aimed to determine whether magnetic iron oxide nanoparticle (MION) hyperthermia (HT) can enhance abscopal effects with radiotherapy (RT) and immune checkpoint inhibitors (IT) in a metastatic TNBC model.Methods: One week after implanting 4T1-luc cells into the mammary glands of BALB/c mice, tumors were treated with RT (3 × 8 Gy)±local HT, mild (HTM, 43 °C/20 min) or partially ablative (HTAbl, 45 °C/5 min plus 43 °C/15 min),±IT with anti-PD-1 and anti-CTLA-4 antibodies (both 4 × 10 mg/kg, i.p.). Tumor growth was measured daily. Two weeks after treatment, lungs and livers were harvested for histopathology evaluation of metastases.Results: Compared to untreated controls, all treatment groups demonstrated a decreased tumor volume; however, when compared against surgical resection, only RT + HTM+IT, RT + HTAbl+IT and RT + HTAbl had similar or smaller tumors. These cohorts showed more infiltration of CD3+ T-lymphocytes into the primary tumor. Tumor growth effects were partially reversed with T-cell depletion. Combinations that proved most effective for primary tumors generated modest reductions in numbers of lung metastases. Conversely, numbers of lung metastases showed potential to increase following HT + IT treatment, particularly when compared to RT. Compared to untreated controls, there was no improvement in survival with any treatment.Conclusions: Single-fraction MION HT added to RT + IT improved local tumor control and recruitment of CD3+ T-lymphocytes, with only a modest effect to reduce lung metastases and no improvement in overall survival. HT + IT showed potential to increase metastatic dissemination to lungs.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Animais , Anticorpos Monoclonais/farmacologia , Terapia Combinada , Modelos Animais de Doenças , Feminino , Humanos , Nanopartículas de Magnetita , Camundongos , Metástase Neoplásica , Transfecção
15.
Nanotechnology ; 29(37): 375101, 2018 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-29920184

RESUMO

HepG2 cell death with magnetic hyperthermia (MHT) using hydroxyapatite nanoparticles (mHAPs) and alternating magnetic fields (AMF) was investigated in vitro. The mHAPs were synthesized as thermo-seeds by co-precipitation with the addition of Fe2+. The grain size of the HAPs and iron oxide magnetic were 39.1 and 19.5 nm and were calculated by the Scherrer formula. The HepG2 cells were cultured with mHAPs and exposed to an AMF for 30 min yielding maximum temperatures of 43 ± 0.5 °C. After heating, the cell viability was reduced by 50% relative to controls, lactate dehydrogenase (LDH) concentrations measured in media were three-fold greater than those measured in all control groups. Readouts of toxicity by live/dead staining were consistent with cell viability and LDH assay results. Measured reactive oxygen species (ROS) in cells exposed to MHT were two-fold greater than in control groups. Results of cDNA microarray and Western blotting revealed tantalizing evidence of ATM and GADD45 downregulation with possible MKK3/MKK6 and ATF-2 of p38 MAPK inhibition upon exposure to mHAPs and AMF combinations. These results suggest that the combination of mHAPs and AMF can increase intracellular concentrations of ROS to cause DNA damage, which leads to cell death that complement heat stress related biological effects.


Assuntos
Durapatita/química , Hipertermia Induzida , Nanopartículas de Magnetita/química , Espécies Reativas de Oxigênio/metabolismo , Morte Celular , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Campos Magnéticos , Nanopartículas de Magnetita/ultraestrutura , Regulação para Cima/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Int J Hyperthermia ; 34(8): 1316-1328, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29353516

RESUMO

Hyperthermia therapy (HT) is the exposure of a region of the body to elevated temperatures to achieve a therapeutic effect. HT anticancer properties and its potential as a cancer treatment have been studied for decades. Techniques used to achieve a localised hyperthermic effect include radiofrequency, ultrasound, microwave, laser and magnetic nanoparticles (MNPs). The use of MNPs for therapeutic hyperthermia generation is known as magnetic hyperthermia therapy (MHT) and was first attempted as a cancer therapy in 1957. However, despite more recent advancements, MHT has still not become part of the standard of care for cancer treatment. Certain challenges, such as accurate thermometry within the tumour mass and precise tumour heating, preclude its widespread application as a treatment modality for cancer. MHT is especially attractive for the treatment of glioblastoma (GBM), the most common and aggressive primary brain cancer in adults, which has no cure. In this review, the application of MHT as a therapeutic modality for GBM will be discussed. Its therapeutic efficacy, technical details, and major experimental and clinical findings will be reviewed and analysed. Finally, current limitations, areas of improvement, and future directions will be discussed in depth.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Hipertermia Induzida , Fenômenos Magnéticos , Animais , Humanos , Resultado do Tratamento
17.
Int J Hyperthermia ; 34(4): 373-381, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28758530

RESUMO

We report the development and optimisation of an assay for quantitating iron from iron oxide nanoparticles in biological matrices by using ferene-s, a chromogenic compound. The method is accurate, reliable and can be performed with basic equipment common to many laboratories making it convenient and inexpensive. The assay we have developed is suited for quantitation of iron in cell culture studies with iron oxide nanoparticles, which tend to manifest low levels of iron. The assay was validated with standard reference materials and with inductively coupled plasma-mass spectrometry (ICP-MS) to accurately measure iron concentrations ∼1 × 10-6 g in about 1 × 106 cells (∼1 × 10-12 g Fe per cell). The assay requires preparation and use of a working solution to which samples can be directly added without further processing. After overnight incubation, the absorbance can be measured with a standard UV/Vis spectrophotometer to provide iron concentration. Alternatively, for expedited processing, samples can be digested with concentrated nitric acid before addition to the working solution. Optimization studies demonstrated significant deviations accompany variable digestion times, highlighting the importance to ensure complete iron ion liberation from the nanoparticle or sample matrix to avoid underestimating iron concentration. When performed correctly, this method yields reliable iron ion concentration measurements to ∼2 × 10-6 M (1 × 10-7 g/ml sample).


Assuntos
Compostos Férricos , Ferro/análise , Nanopartículas Metálicas , Triazinas , Bioensaio , Linhagem Celular Tumoral , Colorimetria , Humanos , Espectrometria de Massas , Ácido Nítrico/química , Espectrofotometria
18.
Int J Hyperthermia ; 32(5): 543-57, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27151045

RESUMO

PURPOSE/OBJECTIVE: The aim of this study was to develop and investigate the properties of a magnetic iron oxide nanoparticle-ethiodised oil formulation for image-guided thermal therapy of liver cancer. MATERIALS AND METHODS: The formulation comprises bionised nano-ferrite (BNF) nanoparticles suspended in ethiodised oil, emulsified with polysorbate 20 (BNF-lip). Nanoparticle size was measured via photon correlation spectroscopy and transmission electron microscopy. In vivo thermal therapy capability was tested in two groups of male Foxn1(nu) mice bearing subcutaneous HepG2 xenograft tumours. Group I (n = 12) was used to screen conditions for group II (n = 48). In group II, mice received one of BNF-lip (n = 18), BNF alone (n = 16), or PBS (n = 14), followed by alternating magnetic field (AMF) hyperthermia, with either varied duration (15 or 20 min) or amplitude (0, 16, 20, or 24 kA/m). Image-guided fluoroscopic intra-arterial injection of BNF-lip was tested in New Zealand white rabbits (n = 10), bearing liver VX2 tumours. The animals were subsequently imaged with CT and 3 T MRI, up to 7 days post-injection. The tumours were histopathologically evaluated for distribution of BNF-lip. RESULTS: The BNF showed larger aggregate diameters when suspended in BNF-lip, compared to clear solution. The BNF-lip formulation produced maximum tumour temperatures with AMF >20 kA/m and showed positive X-ray visibility and substantial shortening of T1 and T2 relaxation time, with sustained intratumoural retention up to 7 days post-injection. On pathology, intratumoural BNF-lip distribution correlated well with CT imaging of intratumoural BNF-lip distribution. CONCLUSION: The BNF-lip formulation has favourable thermal and dual imaging capabilities for image-guided thermal therapy of liver cancer, suggesting further exploration for clinical applications.


Assuntos
Compostos Férricos/administração & dosagem , Hipertermia Induzida , Neoplasias Hepáticas/terapia , Nanopartículas Metálicas/administração & dosagem , Animais , Linhagem Celular Tumoral , Óleo Etiodado/administração & dosagem , Óleo Etiodado/uso terapêutico , Estudos de Viabilidade , Compostos Férricos/uso terapêutico , Células Hep G2 , Humanos , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/patologia , Fenômenos Magnéticos , Imageamento por Ressonância Magnética , Masculino , Nanopartículas Metálicas/uso terapêutico , Camundongos Nus , Polissorbatos/administração & dosagem , Polissorbatos/uso terapêutico , Coelhos , Tomografia Computadorizada por Raios X , Carga Tumoral , Ultrassonografia
20.
Int J Hyperthermia ; 31(4): 359-74, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25811736

RESUMO

PURPOSE: We aimed to characterise magnetic nanoparticle hyperthermia (mNPH) with radiation therapy (RT) for prostate cancer. METHODS: Human prostate cancer subcutaneous tumours, PC3 and LAPC-4, were grown in nude male mice. When tumours measured 150 mm3 magnetic iron oxide nanoparticles (MIONPs) were injected into tumours to a target dose of 5.5 mg Fe/cm3 tumour, and treated 24 h later by exposure to alternating magnetic field (AMF). Mice were randomly assigned to one of four cohorts to characterise (1) intratumour MIONP distribution, (2) effects of variable thermal dose mNPH (fixed AMF peak amplitude 24 kA/m at 160 ± 5 kHz) with/without RT (5 Gy), (3) effects of RT (RT5: 5 Gy; RT8: 8 Gy), and (4) fixed thermal dose mNPH (43 °C for 20 min) with/without RT (5 Gy). MIONP concentration and distribution were assessed following sacrifice and tissue harvest using inductively coupled plasma mass spectrometry (ICP-MS) and Prussian blue staining, respectively. Tumour growth was monitored and compared among treated groups. RESULTS: LAPC-4 tumours retained higher MIONP concentration and more uniform distribution than did PC3 tumours. AMF power modulation provided similar thermal dose for mNPH and combination therapy groups (CEM43: LAPC-4: 33.6 ± 3.4 versus 25.9 ± 0.8, and PC3: 27.19 ± 0.7 versus 27.50 ± 0.6), thereby overcoming limitations of MIONP distribution and yielding statistically significant tumour growth delay. CONCLUSION: PC3 and LAPC-4 tumours represent two biological models that demonstrate different patterns of nanoparticle retention and distribution, offering a model to make comparisons of these effects for mNPH. Modulating power for mNPH offers potential to overcome limitations of MIONP distribution to enhance mNPH.


Assuntos
Hipertermia Induzida/métodos , Nanopartículas de Magnetita/administração & dosagem , Neoplasias da Próstata/terapia , Radiossensibilizantes/farmacologia , Animais , Linhagem Celular Tumoral , Terapia Combinada , Humanos , Magnetoterapia , Nanopartículas de Magnetita/uso terapêutico , Masculino , Espectrometria de Massas , Camundongos , Neoplasias da Próstata/radioterapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA