Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Sci Rep ; 13(1): 2278, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36755030

RESUMO

The combined passive and active targeting of tumoral tissue remains an active and relevant cancer research field. Here, we exploit the properties of two highly magnetic nanomaterials, magnetosomes and ultramagnetic liposomes, in order to magnetically target prostate adenocarcinoma tumors, implanted orthotopically or subcutaneously, to take into account the role of tumor vascularization in the targeting efficiency. Analysis of organ biodistribution in vivo revealed that, for all conditions, both nanomaterials accumulate mostly in the liver and spleen, with an overall low tumor retention. However, both nanomaterials were more readily identified in orthotopic tumors, reflecting their higher tumor vascularization. Additionally, a 2- and 3-fold increase in nanomaterial accumulation was achieved with magnetic targeting. In summary, ultramagnetic nanomaterials show promise mostly in the targeting of highly-vascularized orthotopic murine tumor models.


Assuntos
Magnetossomos , Neoplasias da Próstata , Masculino , Humanos , Animais , Camundongos , Lipossomos , Distribuição Tecidual , Neovascularização Patológica , Fenômenos Magnéticos , Linhagem Celular Tumoral
3.
Chem Commun (Camb) ; 58(37): 5642-5645, 2022 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-35439806

RESUMO

Here, multivalent functions have been successfully integrated on a single core-shell type nanostructure, for remote-controlled and receptor-targeted intracellular delivery of doxorubicin (DOX) to breast cancer cells that overexpress biotin receptors.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Doxorrubicina/química , Sistemas de Liberação de Medicamentos , Fenômenos Magnéticos , Polímeros Molecularmente Impressos , Nanopartículas/química , Neoplasias/tratamento farmacológico
4.
ACS Appl Mater Interfaces ; 14(13): 15021-15034, 2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-35319860

RESUMO

The endosomal entrapment of functional nanoparticles is a severe limitation to their use for biomedical applications. In the case of magnetic nanoparticles (MNPs), this entrapment leads to poor heating efficiency for magnetic hyperthermia and suppresses the possibility to manipulate them in the cytosol. Current strategies to limit their entrapment include functionalization with cell-penetrating peptides to promote translocation directly across the cell membrane or facilitate endosomal escape. However, these strategies suffer from the potential release of free peptides in the cell, and to the best of our knowledge, there is currently a lack of effective methods for the cytosolic delivery of MNPs after incubation with cells. Herein, we report the conjugation of fluorescently labeled cationic peptides to γ-Fe2O3@SiO2 core-shell nanoparticles by click chemistry to improve MNP access to the cytosol. We compare the effect of Arg9 and His4 peptides. On the one hand, Arg9 is a classical cell-penetrating peptide able to enter cells by direct translocation, and on the other hand, it has been demonstrated that sequences rich in histidine residues can promote endosomal escape, possibly by the proton sponge effect. The methodology developed here allows a high colocalization of the peptides and core-shell nanoparticles in cells and confirms that grafting peptides rich in histidine residues onto nanoparticles promotes NPs' access to the cytosol. Endosomal escape was confirmed by a calcein leakage assay and by ultrastructural analysis in transmission electron microscopy. No toxicity was observed for the peptide-nanoparticles conjugates. We also show that our conjugation strategy is compatible with the addition of multiple substrates and can thus be used for the delivery of cytoplasm-targeted therapeutics.


Assuntos
Peptídeos Penetradores de Células , Nanopartículas , Peptídeos Penetradores de Células/metabolismo , Citosol/metabolismo , Endossomos/metabolismo , Fenômenos Magnéticos , Nanopartículas/química , Dióxido de Silício/metabolismo
5.
Commun Biol ; 5(1): 137, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35177769

RESUMO

How mechanical stress actively impacts the physiology and pathophysiology of cells and tissues is little investigated in vivo. The colon is constantly submitted to multi-frequency spontaneous pulsatile mechanical waves, which highest frequency functions, of 2 s period, remain poorly understood. Here we find in vivo that high frequency pulsatile mechanical stresses maintain the physiological level of mice colon stem cells (SC) through the mechanosensitive Ret kinase. When permanently stimulated by a magnetic mimicking-tumor growth analogue pressure, we find that SC levels pathologically increase and undergo mechanically induced hyperproliferation and tumorigenic transformation. To mimic the high frequency pulsatile mechanical waves, we used a generator of pulsed magnetic force stimulation in colonic tissues pre-magnetized with ultra-magnetic liposomes. We observed the pulsatile stresses using last generation ultra-wave dynamical high-resolution imaging. Finally, we find that the specific pharmacological inhibition of Ret mechanical activation induces the regression of spontaneous formation of SC, of CSC markers, and of spontaneous sporadic tumorigenesis in Apc mutated mice colons. Consistently, in human colon cancer tissues, Ret activation in epithelial cells increases with tumor grade, and partially decreases in leaking invasive carcinoma. High frequency pulsatile physiological mechanical stresses thus constitute a new niche that Ret-dependently fuels mice colon physiological SC level. This process is pathologically over-activated in the presence of permanent pressure due to the growth of tumors initiated by pre-existing genetic alteration, leading to mechanotransductive self-enhanced tumor progression in vivo, and repressed by pharmacological inhibition of Ret.


Assuntos
Neoplasias do Colo/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Animais , Biomarcadores Tumorais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Células-Tronco Neoplásicas , Proteínas Proto-Oncogênicas c-ret/genética
6.
Nanomaterials (Basel) ; 11(11)2021 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-34835858

RESUMO

Molecularly imprinted polymers (MIPs) have been widely used in nanomedicine in the last few years. However, their potential is limited by their intrinsic properties resulting, for instance, in lack of control in drug release processes or complex detection for in vivo imaging. Recent attempts in creating hybrid nanomaterials combining MIPs with inorganic nanomaterials succeeded in providing a wide range of new interesting properties suitable for nanomedicine. Through this review, we aim to illustrate how hybrid molecularly imprinted polymers may improve patient care with enhanced imaging, treatments, and a combination of both.

7.
Molecules ; 26(13)2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34210027

RESUMO

The inhibition of the protein function for therapeutic applications remains challenging despite progress these past years. While the targeting application of molecularly imprinted polymer are in their infancy, no use was ever made of their magnetic hyperthermia properties to damage proteins when they are coupled to magnetic nanoparticles. Therefore, we have developed a facile and effective method to synthesize magnetic molecularly imprinted polymer nanoparticles using the green fluorescent protein (GFP) as the template, a bulk imprinting of proteins combined with a grafting approach onto maghemite nanoparticles. The hybrid material exhibits very high adsorption capacities and very strong affinity constants towards GFP. We show that the heat generated locally upon alternative magnetic field is responsible of the decrease of fluorescence intensity.


Assuntos
Proteínas de Fluorescência Verde/química , Nanopartículas Magnéticas de Óxido de Ferro/química , Polímeros Molecularmente Impressos/química , Desnaturação Proteica
8.
Chem Commun (Camb) ; 57(48): 5945-5948, 2021 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-34019041

RESUMO

We describe a novel synthesis allowing one to enhance the load of magnetic nanoparticles and gold nanorods in nanogels. Two different structures, simple cores and core-shell, were synthesized and their heating properties upon alternating magnetic field or laser exposure are compared. Remarkably, the core-shell structure showed a greater heating capacity in the two modalities.


Assuntos
Ouro/química , Nanopartículas de Magnetita/química , Nanogéis/química , Calefação , Campos Magnéticos , Estrutura Molecular , Tamanho da Partícula , Processos Fotoquímicos , Propriedades de Superfície
9.
Nanotoxicology ; 14(10): 1342-1361, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33078975

RESUMO

We investigated the toxicity of Iron oxide and Zinc oxide engineered nanoparticles (ENPs) on Paracentrotus lividus sea urchin embryos and three species of microalgae. Morphological responses, internalization, and potential impacts of Fe2O3 and ZnO ENPs on physiology and metabolism were assessed. Both types of ENPs affected P. lividus larval development, but ZnO ENPs had a much stronger effect. While growth of the alga Micromonas commoda was severely impaired by both ENPs, Ostreococcus tauri or Nannochloris sp. were unaffected. Transmission electron microscopy showed the internalization of ENPs in sea urchin embryonic cells while only nanoparticle interaction with external membranes was evidenced in microalgae, suggesting that marine organisms react in diverse ways to ENPs. Transcriptome-wide analysis in P. lividus and M. commoda showed that many different physiological pathways were affected, some of which were common to both species, giving insights about the mechanisms underpinning toxic responses.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Nanopartículas Magnéticas de Óxido de Ferro/toxicidade , Microalgas/efeitos dos fármacos , Nanopartículas/toxicidade , Paracentrotus/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Óxido de Zinco/toxicidade , Animais , Embrião não Mamífero/metabolismo , Perfilação da Expressão Gênica , Microalgas/crescimento & desenvolvimento , Microalgas/metabolismo , Paracentrotus/genética , Paracentrotus/crescimento & desenvolvimento
10.
Int J Mol Sci ; 21(18)2020 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-32911745

RESUMO

The remote actuation of cellular processes such as migration or neuronal outgrowth is a challenge for future therapeutic applications in regenerative medicine. Among the different methods that have been proposed, the use of magnetic nanoparticles appears to be promising, since magnetic fields can act at a distance without interactions with the surrounding biological system. To control biological processes at a subcellular spatial resolution, magnetic nanoparticles can be used either to induce biochemical reactions locally or to apply forces on different elements of the cell. Here, we show that cell migration and neurite outgrowth can be directed by the forces produced by a switchable parallelized array of micro-magnetic pillars, following the passive uptake of nanoparticles. Using live cell imaging, we first demonstrate that adherent cell migration can be biased toward magnetic pillars and that cells can be reversibly trapped onto these pillars. Second, using differentiated neuronal cells we were able to induce events of neurite outgrowth in the direction of the pillars without impending cell viability. Our results show that the range of forces applied needs to be adapted precisely to the cellular process under consideration. We propose that cellular actuation is the result of the force on the plasma membrane caused by magnetically filled endo-compartments, which exert a pulling force on the cell periphery.


Assuntos
Movimento Celular/efeitos dos fármacos , Magnetismo/métodos , Nanopartículas de Magnetita/uso terapêutico , Espaço Intracelular/fisiologia , Campos Magnéticos , Nanopartículas de Magnetita/análise , Fenômenos Mecânicos , Crescimento Neuronal/efeitos dos fármacos , Fenômenos Físicos , Medicina Regenerativa/métodos
11.
Pharmaceutics ; 12(9)2020 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-32911863

RESUMO

Cationic liposomes have been considered as potential vectors for gene delivery thanks to their ability to transfect cells with high efficiency. Recently, the combination of diagnostic agent and therapeutic agents in the same particle to form a theranostic system has been reported. Magnetic liposomes are one of these examples. Due to the magnetic nanoparticles encapsulated in the liposomes, they can act as a drug delivery system and, at the same time, a magnetic resonance imaging contrast enhancement agent or hyperthermia. In this work, nucleic acid delivery systems based on magnetic cationic liposomes (MCLs) were developed. Two different techniques, reverse phase evaporation and cosolvent sonication, were employed for liposome preparation. Both strategies produced MCLs of less than 200 nm with highly positive charge. Enhancement of their transverse and longitudinal relaxivities r2and r1 was obtained with both kinds of magnetic liposomes compared to free magnetic nanoparticles. Moreover, these MCLs showed high capacity to form complexes and transfect CT-26 cells using the antibiotic-free pFAR4-luc plasmid. The transfection enhancement with magnetofection was also carried out in CT26 cells. These results suggested that our MCLs could be a promising candidate for image-guided gene therapy.

12.
Colloids Surf B Biointerfaces ; 196: 111297, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32791474

RESUMO

In this work, we describe the synthesis and characterization of the SPIONP-CUR conjugate between curcumin (CUR) and superparamagnetic iron oxide nanoparticles (SPIONPs), in addition to its application in photodynamic therapy (PDT) using a protocol free of organic solvents as a dispersant. The SPIONP-CUR conjugate was characterized by X-ray diffraction, transmission electron microscopy, zeta potential measurements, Fourier transform infrared spectroscopy, thermogravimetry, magnetometry and magnetic hyperthermia assays. The SPIONP-CUR conjugation occurred by bonding between the keto-enol moiety of CUR and the iron atoms present on the surfaces of the SPIONPs. The conjugate showed heating power under an alternating magnetic field (AMF) and photodynamic action when irradiated with blue LED light. In experiments using PDT against Staphylococcus aureus in the planktonic phase, it was demonstrated that with application of blue light at 3.12 J cm-2, the conjugate (dispersed in water) caused a total reduction of the bacterial load. In the absence of light, the reduction was insignificant, even after 24 h of contact with the bacteria.


Assuntos
Curcumina , Nanopartículas , Fotoquimioterapia , Curcumina/farmacologia , Campos Magnéticos , Nanopartículas Magnéticas de Óxido de Ferro , Fármacos Fotossensibilizantes/farmacologia
13.
Chem Commun (Camb) ; 56(70): 10255-10258, 2020 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-32756712

RESUMO

Herein, we report a facile and rapid one-step synthetic strategy for the development of magnetic doxorubicin imprinted silica nanoparticles for drug release experiments in living cells showing a remotely triggered doxorubicin release upon applying an alternating magnetic field, without temperature elevation of the medium (local heating).


Assuntos
Doxorrubicina/química , Portadores de Fármacos/química , Campos Magnéticos , Nanopartículas/química , Dióxido de Silício/química , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Liberação Controlada de Fármacos , Humanos
14.
J Control Release ; 322: 137-148, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32145266

RESUMO

Theranostic nanocarriers of antivascular drug encapsulated in thermosensitive ultramagnetic liposomes can be advantageously designed to provide a locally high concentration and an active delivery, with image-guided Magnetic Resonance Imaging (MRI) so as to reliably cure tumor. We propose a novel therapeutic strategy consisting of the magnetic accumulation of Ultra Magnetic Liposomes (UML) followed by High-Intensity Focused Ultrasound (HIFU) to trigger the release of an antivascular agent monitored by MRI. For this purpose, we co-encapsulated Combretastatin A4 phosphate (CA4P), a vascular disrupting agent, in the core of UML to obtain CA4P-loaded thermosensitive Ultra Magnetic Liposomes (CA4P-UML). To assess the HIFU parameters, the CA4P release has been triggered in vitro by local heating HIFU at the lipids transition temperature. Morphology of endothelial cells was assessed to evaluate the effect of encapsulated versus non-encapsulated CA4P. The efficiency of a treatment combining the magnetic targeting of CA4P-UML with the CA4P release triggered by HIFU was studied in CT26 murine tumors. Tumor perfusion and volume regression parameters were monitored by multiparametric quantitative anatomical and dynamic in vivo MRI at 7 T. Additionally, vascularization and cellularity were evaluated ex-vivo by histology. This thorough investigation showed that the combined treatment exhibited a full benefit. A 150-fold improvement compared with the chemotherapy alone was obtained using a magnetic targeting of CA4P-UML triggered by HIFU, and was consistent with an expected effect on vascularization 24 h after treatment.


Assuntos
Lipossomos , Estilbenos , Animais , Meios de Contraste , Células Endoteliais , Imageamento por Ressonância Magnética , Camundongos , Medicina de Precisão
15.
ACS Appl Bio Mater ; 3(10): 6802-6810, 2020 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-35019343

RESUMO

Tissue engineering aims to repair or replace deficient tissue by delivering constructs that mimic the native in vivo structure. One challenge in cardiac tissue engineering approaches is to achieve intrinsic cardiac organization, particularly the alignment of cardiomyocytes. Here, we propose a strategy for 3D manipulation and alignment of cardiomyocytes by combining magnetism and a hydrogel. The advantage of using magnetic forces is that they act remotely on the cells when these are endowed with magnetization via the internalization of magnetic nanoparticles. The magnetic actuation then allows obtaining, almost instantaneously and before gel transition, an aligned biomimetic cardiac tissue construct. Gel transition enables us to keep the cellular pattern once the magnetic field was removed. This cardiac tissue engineering approach was tested with both H9c2 cell line and primary cardiomyocytes, and with both a synthetic hydrogel and a natural one, Pluronic F-127 and fibrin, respectively. Key parameters of the anisotropic tissue formation were assessed. Hydrogel rheology is provided, and the impact of cell density and magnetic labeling on cell-cell alignment is assessed. Immunofluorescence confirms the presence of several cardiac markers upon chaining, demonstrating the functionality of the tissue-like cell alignment obtained via magnetic actuation.

16.
Sci Rep ; 10(1): 22452, 2020 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-33384447

RESUMO

The axon regeneration of neurons in the brain can be enhanced by activating intracellular signaling pathways such as those triggered by the membrane-anchored Rat sarcoma (RAS) proto-oncogene. Here we demonstrate the induction of neurite growth by expressing tagged permanently active Harvey-RAS protein or the RAS-activating catalytic domain of the guanine nucleotide exchange factor (SOS1cat), in secondary dopaminergic cells. Due to the tag, the expressed fusion protein is captured by functionalized magnetic nanoparticles in the cytoplasm of the cell. We use magnetic tips for remote translocation of the SOS1cat-loaded magnetic nanoparticles from the cytoplasm towards the inner face of the plasma membrane where the endogenous Harvey-RAS protein is located. Furthermore, we show the magnetic transport of SOS1cat-bound nanoparticles from the cytoplasm into the neurite until they accumulate at its tip on a time scale of minutes. In order to scale-up from single cells, we show the cytoplasmic delivery of the magnetic nanoparticles into large numbers of cells without changing the cellular response to nerve growth factor. These results will serve as an initial step to develop tools for refining cell replacement therapies based on grafted human induced dopaminergic neurons loaded with functionalized magnetic nanoparticles in Parkinson model systems.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Nanopartículas de Magnetita , Regeneração Nervosa , Neuritos/metabolismo , Proteína SOS1/metabolismo , Biomarcadores , Linhagem Celular , Imunofluorescência , Expressão Gênica , Vetores Genéticos/genética , Humanos , Modelos Biológicos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteína SOS1/genética
17.
ACS Appl Mater Interfaces ; 11(39): 35556-35565, 2019 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-31496222

RESUMO

Magnetic nanoparticles coated with protein-specific molecularly imprinted polymers (MIPs) are receiving increasing attention thanks to their binding abilities, robustness, and easy synthesis compared to their natural analogues also able to target proteins, such as antibodies or aptamers. Acting as tailor-made recognition systems, protein-specific MIPs can be used in many in vivo nanomedicine applications, such as targeted drug delivery, biosensing, and tissue engineering. Nonetheless, studies on their biocompatibility and long-term fate in biological environments are almost nonexistent, although these questions have to be addressed before considering clinical applications. To alleviate this lack of knowledge, we propose here to monitor the effect of a protein-specific MIP coating on the toxicity and biodegradation of magnetic iron oxide nanoparticles, both in a minimal aqueous degradation medium and in a model of cartilage tissue formed by differentiated human mesenchymal stem cells. Degradation of iron oxide nanoparticles with or without the polymer coating was monitored for a month by following their magnetic properties using vibrating sample magnetometry and their morphology by transmission electron microscopy. We showed that the MIP coating of magnetic iron oxide nanoparticles does not affect their biocompatibility or internalization inside cells. Remarkably, the imprinted polymer coating does not hinder the magnetic particle degradation but seems to slow it down, although this effect is more visible when degradation occurs in the buffer medium than in cells. Hence, the results presented in this paper are really encouraging and open up the way to future applications of MIP-coated nanoparticles into the clinic.


Assuntos
Materiais Revestidos Biocompatíveis , Sistemas de Liberação de Medicamentos , Nanopartículas de Magnetita/química , Células-Tronco Mesenquimais , Impressão Molecular , Diferenciação Celular/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacocinética , Materiais Revestidos Biocompatíveis/farmacologia , Humanos , Nanopartículas de Magnetita/ultraestrutura , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Células PC-3
18.
J Funct Biomater ; 10(3)2019 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-31315182

RESUMO

Parkinson's disease (PD) is a neurodegenerative disease associated with loss or dysfunction of dopaminergic neurons located in the substantia nigra (SN), and there is no cure available. An emerging new approach for treatment is to transplant human induced dopaminergic neurons directly into the denervated striatal brain target region. Unfortunately, neurons grafted into the substantia nigra are unable to grow axons into the striatum and thus do not allow recovery of the original connectivity. Towards overcoming this general limitation in guided neuronal regeneration, we develop here magnetic nanoparticles functionalized with proteins involved in the regulation of axonal growth. We show covalent binding of constitutive active human rat sarcoma (RAS) proteins or RAS guanine nucleotide exchange factor catalytic domain of son of sevenless (SOS) by fluorescence correlation spectroscopy and multiangle light scattering as well as the characterization of exchange factor activity. Human dopaminergic neurons were differentiated from neural precursor cells and characterized by electrophysiological and immune histochemical methods. Furthermore, we demonstrate magnetic translocation of cytoplasmic γ-Fe2O3@SiO2 core-shell nanoparticles into the neurite extensions of induced human neurons. Altogether, we developed tools towards remote control of directed neurite growth in human dopaminergic neurons. These results may have relevance for future therapeutic approaches of cell replacement therapy in Parkinson's disease.

19.
Mol Imaging Biol ; 21(2): 269-278, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29942990

RESUMO

PURPOSE: The development of theranostic nanocarriers as an innovative therapy against cancer has been improved by targeting properties in order to optimize the drug delivery to safely achieve its desired therapeutic effect. The aim of this paper is to evaluate the magnetic targeting (MT) efficiency of ultra-magnetic liposomes (UML) into CT26 murine colon tumor by magnetic resonance imaging (MRI). PROCEDURES: Dynamic susceptibility contrast MRI was applied to assess the bloodstream circulation time. A novel semi-quantitative method called %I0.25, based on the intensity distribution in T2*-weighted MRI images was developed to compare the accumulation of T2 contrast agent in tumors with or without MT. To evaluate the efficiency of magnetic targeting, the percentage of pixels under the intensity value I0.25 (I0.25 = 0.25(Imax - Imin)) was calculated on the intensity distribution histogram. RESULTS: This innovative method of processing MRI images showed the MT efficiency by a %I0.25 that was significantly higher in tumors using MT compared to passive accumulation, from 15.3 to 28.6 %. This methodology was validated by ex vivo methods with an iron concentration that is 3-fold higher in tumors using MT. CONCLUSIONS: We have developed a method that allows a semi-quantitative evaluation of targeting efficiency in tumors, which could be applied to different T2 contrast agents.


Assuntos
Neoplasias do Colo/diagnóstico por imagem , Imageamento por Ressonância Magnética , Magnetismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Lipossomos , Fígado/metabolismo , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3
20.
Adv Drug Deliv Rev ; 138: 233-246, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30414493

RESUMO

Magnetic hyperthermia which exploits the heat generated by magnetic nanoparticles (MNPs) when exposed to an alternative magnetic field (AMF) is now in clinical trials for the treatment of cancers. However, this thermal therapy requires a high amount of MNPs in the tumor to be efficient. On the contrary the hot spot local effect refers to the use of specific temperature profile at the vicinity of nanoparticles for heating with minor to no long-range effect. This magneto-thermal effect can be exploited as a relevant external stimulus to temporally and spatially trigger drug release. In this review, we focus on recent advances in magnetic hyperthermia. Indirect experimental proofs of the local temperature increase are first discussed leading to a good estimation of the temperature at the surface (from 0.5 to 6 nm) of superparamagnetic NPs. Then we highlight recent studies illustrating the hot-spot effect for drug-release. Finally, we present another recent strategy to enhance the efficacity of thermal treatment by combining photothermal therapy with magnetic hyperthermia mediated by magneto-plasmonic nanoplatforms.


Assuntos
Sistemas de Liberação de Medicamentos , Hipertermia Induzida , Campos Magnéticos , Nanopartículas/administração & dosagem , Animais , Temperatura Alta , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...