Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Lab Chip ; 23(24): 5139-5150, 2023 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-37942508

RESUMO

3D in vitro biological systems are progressively replacing 2D systems to increase the physiological relevance of cellular studies. Microfluidics-based approaches can be powerful tools towards such biomimetic systems, but often require high-end complicated and expensive processes and equipment for microfabrication. Herein, a drug screening platform is proposed, minimizing technicality and manufacturing steps. It provides an alternate way of spheroid generation in droplets in tubes. Droplet microfluidics then elicit multiple droplets merging events at programmable times, to submit sequentially the spheroids to chemotherapy and to reagents for cytotoxicity screening. After a comprehensive study of tumorogenesis within the droplets, the system is validated for drug screening (IC50) with chemotherapies in cancer cell lines as well as cells from a patient-derived-xenografts (PDX). As compared to microtiter plates methods, our system reduces the initial number of cells up to 10 times and opens new avenues towards primary tumors drug screening approaches.


Assuntos
Microfluídica , Neoplasias , Humanos , Microfluídica/métodos , Avaliação Pré-Clínica de Medicamentos , Detecção Precoce de Câncer , Esferoides Celulares , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico
2.
Mater Horiz ; 10(11): 4757-4775, 2023 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-37740347

RESUMO

With their distinctive physicochemical features, nanoparticles have gained recognition as effective multifunctional tools for biomedical applications, with designs and compositions tailored for specific uses. Notably, magnetic nanoparticles stand out as first-in-class examples of multiple modalities provided by the iron-based composition. They have long been exploited as contrast agents for magnetic resonance imaging (MRI) or as anti-cancer agents generating therapeutic hyperthermia through high-frequency magnetic field application, known as magnetic hyperthermia (MHT). This review focuses on two more recent applications in oncology using iron-based nanomaterials: photothermal therapy (PTT) and ferroptosis. In PTT, the iron oxide core responds to a near-infrared (NIR) excitation and generates heat in its surrounding area, rivaling the efficiency of plasmonic gold-standard nanoparticles. This opens up the possibility of a dual MHT + PTT approach using a single nanomaterial. Moreover, the iron composition of magnetic nanoparticles can be harnessed as a chemotherapeutic asset. Degradation in the intracellular environment triggers the release of iron ions, which can stimulate the production of reactive oxygen species (ROS) and induce cancer cell death through ferroptosis. Consequently, this review emphasizes these emerging physical and chemical approaches for anti-cancer therapy facilitated by magnetic nanoparticles, combining all-in-one functionalities.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita , Fotoquimioterapia , Fototerapia/métodos , Hipertermia Induzida/métodos , Nanopartículas de Magnetita/uso terapêutico , Nanopartículas de Magnetita/química , Fotoquimioterapia/métodos , Ferro
3.
Nat Commun ; 14(1): 4637, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37532698

RESUMO

The Fe(II)-induced ferroptotic cell death pathway is an asset in cancer therapy, yet it calls into question the biocompatibility of magnetic nanoparticles. In the latter, Fe(II) is sequestered within the crystal structure and is released only upon nanoparticle degradation, a transition that is not well understood. Here, we dissect the chemical environment necessary for nanoparticle degradation and subsequent Fe(II) release. Importantly, temperature acts as an accelerator of the process and can be triggered remotely by laser-mediated photothermal conversion, as evidenced by the loss of the nanoparticles' magnetic fingerprint. Remarkably, the local hot-spot temperature generated at the nanoscale can be measured in operando, in the vicinity of each nanoparticle, by comparing the photothermal-induced nanoparticle degradation patterns with those of global heating. Further, remote photothermal irradiation accelerates degradation inside cancer cells in a tumor spheroid model, with efficiency correlating with the endocytosis progression state of the nanoparticles. High-throughput imaging quantification of Fe2+ release, ROS generation, lipid peroxidation and cell death at the spheroid level confirm the synergistic thermo-ferroptotic therapy due to the photothermal degradation at the nanoparticle level.


Assuntos
Ferroptose , Nanopartículas , Neoplasias , Humanos , Fototerapia/métodos , Nanopartículas/química , Temperatura Alta , Compostos Ferrosos , Neoplasias/patologia , Linhagem Celular Tumoral
4.
J Mater Chem B ; 11(24): 5574-5585, 2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37040257

RESUMO

Hyperthermia, as the process of heating a malignant site above 42 °C to trigger cell death, has emerged as an effective and selective cancer therapy strategy. Various modalities of hyperthermia have been proposed, among which magnetic and photothermal hyperthermia are known to benefit from the use of nanomaterials. In this context, we introduce herein a hybrid colloidal nanostructure comprising plasmonic gold nanorods (AuNRs) covered by a silica shell, onto which iron oxide nanoparticles (IONPs) are subsequently grown. The resulting hybrid nanostructures are responsive to both external magnetic fields and near-infrared irradiation. As a result, they can be applied for the targeted magnetic separation of selected cell populations - upon targeting by antibody functionalization - as well as for photothermal heating. Through this combined functionality, the therapeutic effect of photothermal heating can be enhanced. We demonstrate both the fabrication of the hybrid system and its application for targeted photothermal hyperthermia of human glioblastoma cells.


Assuntos
Hipertermia Induzida , Nanopartículas , Humanos , Calefação , Hipertermia Induzida/métodos , Fototerapia/métodos , Nanopartículas/química , Campos Magnéticos
5.
J Mater Chem B ; 11(4): 787-801, 2023 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-36472454

RESUMO

Magnetic nanoparticles (NPs) are powerful agents to induce hyperthermia in tumours upon the application of an alternating magnetic field or an infrared laser. Dopants have been investigated to alter different properties of materials. Herein, the effect of zinc doping into iron oxide NPs on their magnetic properties and structural characteristics has been investigated in-depth. A high temperature reaction with autogenous pressure was used to prepare iron oxide and zinc ferrite NPs of same size and morphology for direct comparison. Pressure was key in obtaining high quality nanocrystals with reduced lattice strain (27% less) and enhanced magnetic properties. Zn0.4Fe2.6O4 NPs with small size of 10.2 ± 2.5 nm and very high saturation magnetisation of 142 ± 9 emu gFe+Zn-1 were obtained. Aqueous dispersion of the NPs showed long term magnetic (up to 24 months) and colloidal stability (at least 6 d) at physiologically mimicking conditions. The samples had been kept in the fridge and had been stable for four years. The biocompatibility of Zn0.4Fe2.6O4 NPs was next evaluated by metabolic activity, membrane integrity and clonogenic assays, which show an equivalence to that of iron oxide NPs. Zinc doping decreased the bandgap of the material by 22% making it a more efficient photothermal agent than iron oxide-based ones. Semiconductor photo-hyperthermia was shown to outperform magneto-hyperthermia in cancer cells, reaching the same temperature 17 times faster whilst using 20 times less material (20 mgFe+Zn ml-1vs. 1 mgFe+Zn ml-1). Magnetothermal conversion was minimally hindered in the cellular confinement whilst photothermal efficiency remained unchanged. Photothermia treatment alone achieved 100% cell death after 10 min of treatment compared to only 30% cell death achieved with magnetothermia at clinically relevant settings for each at their best performing concentration. Altogether, these results suggest that the biocompatible and superparamagnetic zinc ferrite NPs could be a next biomaterial of choice for photo-hyperthermia, which could outperform current iron oxide NPs for magnetic hyperthermia.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita , Neoplasias , Linhagem Celular Tumoral , Hipertermia Induzida/métodos , Nanopartículas de Magnetita/química , Zinco/farmacologia
6.
ACS Nano ; 16(1): 271-284, 2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-34963049

RESUMO

Magnetite and maghemite multicore nanoflowers (NFs) synthesized using the modified polyol-mediated routes are to date among the most effective nanoheaters in magnetic hyperthermia (MHT). Recently, magnetite NFs have also shown high photothermal (PT) performances in the most desired second near-infrared (NIR-II) biological window, making them attractive in the field of nanoparticle-activated thermal therapies. However, what makes magnetic NFs efficient heating agents in both modalities still remains an open question. In this work, we investigate the role of many parameters of the polyol synthesis on the final NFs' size, shape, chemical composition, number of cores, and crystallinity. These nanofeatures are later correlated to the magnetic, optical, and electronic properties of the NFs as well as their collective macroscopic thermal properties in MHT and PT to find relationships between their structure, properties, and function. We evidence the critical role of iron(III) and heating ramps on the elaboration of well-defined NFs with a high number of multicores. While MHT efficiency is found to be proportional to the average number of magnetic cores within the assemblies, the optical responses of the NFs and their collective photothermal properties depend directly on the mean volume of the NFs (as supported by optical cross sections numerical simulations) and strongly on the structural disorder in the NFs, rather than the stoichiometry. The concentration of defects in the nanostructures, evaluated by photoluminescence and Urbach energy (EU), evidence a switch in the optical behavior for a limit value of EU = 0.4 eV where a discontinuous transition from high to poor PT efficiency is also observed.


Assuntos
Compostos Férricos , Hipertermia Induzida , Compostos Férricos/química , Óxido Ferroso-Férrico , Fenômenos Magnéticos
7.
ACS Nano ; 15(6): 9782-9795, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34032115

RESUMO

Despite efforts in producing nanoparticles with tightly controlled designs and specific physicochemical properties, these can undergo massive nano-bio interactions and bioprocessing upon internalization into cells. These transformations can generate adverse biological outcomes and premature loss of functional efficacy. Hence, understanding the intracellular fate of nanoparticles is a necessary prerequisite for their introduction in medicine. Among nanomaterials devoted to theranostics is copper sulfide (CuS), which provides outstanding optical properties along with easy synthesis and low cost. Herein, we performed a long-term multiscale study on the bioprocessing of hollow CuS nanoparticles (CuS NPs) and rattle-like iron oxide nanoflowers@CuS core-shell hybrids (IONF@CuS NPs) when inside stem cells and cancer cells, cultured as spheroids. In the spheroids, both CuS NPs and IONF@CuS NPs are rapidly dismantled into smaller units (day 0 to 3), and hair-like nanostructures are generated (day 9 to 21). This bioprocessing triggers an adaptation of the cellular metabolism to the internalized metals without impacting cell viability, differentiation, or oxidative stress response. Throughout the remodeling, a loss of IONF-derived magnetism is observed, but, surprisingly, the CuS photothermal potential is preserved, as demonstrated by a full characterization of the photothermal conversion across the bioprocessing process. The maintained photothermal efficiency correlated well with synchrotron X-ray absorption spectroscopy measurements, evidencing a similar chemical phase for Cu but not for Fe over time. These findings evidence that the intracellular bioprocessing of CuS nanoparticles can reshape them into bioengineered nanostructures without reducing the photothermal function and therapeutic potential.


Assuntos
Nanopartículas , Nanoestruturas , Cobre , Fototerapia , Sulfetos
8.
Nano Lett ; 21(1): 769-777, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33382624

RESUMO

Nanoparticle-mediated thermal treatments have demonstrated high efficacy and versatility as a local anticancer strategy beyond traditional global hyperthermia. Nanoparticles act as heating generators that can trigger therapeutic responses at both the cell and tissue level. In some cases, treatment happens in the absence of a global temperature rise, damaging the tumor cells even more selectively than other nanotherapeutic strategies. The precise determination of the local temperature in the vicinity of such nanoheaters then stands at the heart of thermal approaches to better adjust the therapeutic thermal onset and reduce potential toxicity-related aspects. Herein, we describe an experimental procedure by X-ray absorption spectroscopy, which directly and accurately infers the local temperature of gold-based nanoparticles, single and hybrid nanocrystals, upon laser photoexcitation, revealing significant nanothermal gradients. Such nanothermometric methodology based on the temperature-dependency of atomic parameters of nanoparticles can be extended to any nanosystem upon remote hyperthermal conditions.


Assuntos
Hipertermia Induzida , Nanopartículas , Ouro , Lasers , Temperatura , Espectroscopia por Absorção de Raios X
9.
Acc Chem Res ; 53(10): 2212-2224, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-32935974

RESUMO

Considerable knowledge has been acquired in inorganic nanoparticles' synthesis and nanoparticles' potential use in biomedical applications. Among different materials, iron oxide nanoparticles remain unrivaled for several reasons. Not only do they respond to multiple physical stimuli (e.g., magnetism, light) and exert multifunctional therapeutic and diagnostic actions but also they are biocompatible and integrate endogenous iron-related metabolic pathways. With the aim to optimize the use of (magnetic) iron oxide nanoparticles in biomedicine, different biophysical phenomena have been recently identified and studied. Among them, the concept of a "nanoparticle's identity" is of particular importance. Nanoparticles' identities evolve in distinct biological environments and over different periods of time. In this Account, we focus on the remodeling of magnetic nanoparticles' identities following their journey inside cells. For instance, nanoparticles' functions, such as heat generation or magnetic resonance imaging, can be highly impacted by endosomal confinement. Structural degradation of nanoparticles was also evidenced and quantified in cellulo and correlates with the loss of magnetic nanoparticle properties. Remarkably, in human stem cells, the nonmagnetic products of nanoparticles' degradation could be subsequently reassembled into neosynthesized, endogenous magnetic nanoparticles. This stunning occurrence might account for the natural presence of magnetic particles in human organs, especially the brain. However, mechanistic details and the implication of such phenomena in homeostasis and disease have yet to be completely unraveled.This Account aims to assess the short- and long-term transformations of magnetic iron oxide nanoparticles in living cells, particularly focusing on human stem cells. Precisely, we herein overview the multiple and ever-evolving chemical, physical, and biological magnetic nanoparticles' identities and emphasize the remarkable intracellular fate of these nanoparticles.


Assuntos
Endossomos/metabolismo , Nanopartículas Magnéticas de Óxido de Ferro/química , Encéfalo/diagnóstico por imagem , Cristalização , Eletroencefalografia , Humanos , Hipertermia Induzida , Ferro/metabolismo , Imageamento por Ressonância Magnética , Nanomedicina , Células-Tronco/química , Células-Tronco/citologia , Células-Tronco/metabolismo , Engenharia Tecidual
10.
Small ; 16(11): e1904960, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32077633

RESUMO

Progress of thermal tumor therapies and their translation into clinical practice are limited by insufficient nanoparticle concentration to release therapeutic heating at the tumor site after systemic administration. Herein, the use of Janus magneto-plasmonic nanoparticles, made of gold nanostars and iron oxide nanospheres, as efficient therapeutic nanoheaters whose on-site delivery can be improved by magnetic targeting, is proposed. Single and combined magneto- and photo-thermal heating properties of Janus nanoparticles render them as compelling heating elements, depending on the nanoparticle dose, magnetic lobe size, and milieu conditions. In cancer cells, a much more effective effect is observed for photothermia compared to magnetic hyperthermia, while combination of the two modalities into a magneto-photothermal treatment results in a synergistic cytotoxic effect in vitro. The high potential of the Janus nanoparticles for magnetic guiding confirms them to be excellent nanostructures for in vivo magnetically enhanced photothermal therapy, leading to efficient tumor growth inhibition.


Assuntos
Hipertermia Induzida , Nanopartículas Multifuncionais , Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Ouro , Campos Magnéticos , Magnetismo , Neoplasias/terapia , Fototerapia
11.
Theranostics ; 9(20): 5924-5936, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31534529

RESUMO

Targeting TRAIL (Tumor necrosis factor (TNF)-Related Apoptosis-Inducing Ligand) receptors for cancer therapy remains challenging due to tumor cell resistance and poor preparations of TRAIL or its derivatives. Herein, to optimize its therapeutic use, TRAIL was grafted onto iron oxide nanoclusters (NCs) with the aim of increasing its pro-apoptotic potential through nanoparticle-mediated magnetic hyperthermia (MHT) or photothermia (PT). Methods: The nanovector, NC@TRAIL, was characterized in terms of size, grafting efficiency, and potential for MHT and PT. The therapeutic function was assessed on a TRAIL-resistant breast cancer cell line, MDA-MB-231, wild type (WT) or TRAIL-receptor-deficient (DKO), by combining complementary methylene blue assay and flow cytometry detection of apoptosis and necrosis. Results: Combined with MHT or PT under conditions of "moderate hyperthermia" at low concentrations, NC@TRAIL acts synergistically with the TRAIL receptor to increase the cell death rate beyond what can be explained by the mere global elevation of temperature. In contrast, all results are consistent with the idea that there are hotspots, close to the nanovector and, therefore, to the membrane receptor, which cause disruption of the cell membrane. Furthermore, nanovectors targeting other membrane receptors, unrelated to the TNF superfamily, were also found to cause tumor cell damage upon PT. Indeed, functionalization of NCs by transferrin (NC@Tf) or human serum albumin (NC@HSA) induces tumor cell killing when combined with PT, albeit less efficiently than NC@TRAIL. Conclusions: Given that magnetic nanoparticles can easily be functionalized with molecules or proteins recognizing membrane receptors, these results should pave the way to original remote-controlled antitumoral targeted thermal therapies.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Compostos Férricos/química , Compostos Férricos/farmacologia , Hipertermia Induzida/métodos , Ligante Indutor de Apoptose Relacionado a TNF/química , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Apoptose/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Microscopia Eletrônica de Transmissão , Fator de Necrose Tumoral alfa/metabolismo
12.
Theranostics ; 9(5): 1288-1302, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30867831

RESUMO

Innovative synthesis routes revolutionized nanomaterial combination and design possibilities resulting in a new generation of fine-tuned nanoparticles featuring exquisite shape and constitution control. However, there is still room for improvement when it comes to the development of multi-functional nanoparticle agents merging a plurality of therapeutic functions to tackle tumors simultaneously by synergic mechanisms. Herein, we report the design of an optimized nanohybrid for cancer tri-therapy featuring a maghemite (γ-Fe2O3) nanoflower-like multicore nanoparticle conceived for efficient magnetic hyperthermia (MHT) and a spiky copper sulfide shell (IONF@CuS) with a high near-infrared (NIR) absorption coefficient suitable for photothermal (PTT) and photodynamic therapy (PDT). Methods: Spiky-like IONF@CuS nanohybrids were obtained through a straightforward and scalable water-based template sacrificial synthesis, which allows the shell shape control by tuning polyvinylpyrrolidone (PVP) concentration. A comprehensive characterization of nanohybrid size, shape and structural properties was carried out by combining complementary TEM, SEM, HR-TEM, EELS, XRD and NTA. The all-in-one therapeutic multi-functionality was assessed on cancer cells and on tumor-bearing nude mice. Results: Tests carried out on IONF@CuS nanohybrid aqueous dispersion demonstrated their impressive efficiency to convert light (conversion coefficient = 42 ± 6 %) and magnetic stimulation (SAR ~ 350 W g-1) into heat as well as to induce concurrent reactive oxygen species (ROS) formation upon laser irradiation. Such capabilities were further confirmed in cellular environment by in vitro tests and at the organism level by in vivo tests in a murine tumor model. Notably, complete tumor regression was obtained for the PTT mode at low Cu concentration. Overall, these results allowed determining windows of applicability for each therapy individually or in combination. Conclusions: Altogether, the obtained data evidence the successful synthesis of a unique tri-therapeutic nanoparticle featuring highly relevant assets for clinical translation such as reduced nanoparticle administered dose, reduced laser power exposure, reduced magnetic field frequency, and the possibility of serial heating cycles and therapy monitoring by photoacoustic (PA) and magnetic resonance imaging (MRI). Furthermore, the integration of the dual heating capability (MHT + PTT) with the PDT insult offers a unique asset to tackle tumors by multiple cytotoxic strategies in order to improve the therapeutic outcome in a broader spectrum of clinical conditions.


Assuntos
Antineoplásicos/administração & dosagem , Hipertermia Induzida/métodos , Magnetoterapia/métodos , Nanocompostos/administração & dosagem , Neoplasias/terapia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/administração & dosagem , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cobre/administração & dosagem , Compostos Férricos/administração & dosagem , Camundongos Nus , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
13.
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
14.
ACS Nano ; 12(7): 6523-6535, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29906096

RESUMO

Despite their highly efficient plasmonic properties, gold nanoparticles are currently preferred to silver nanoparticles for biomedical applications such as photothermal therapy due to their high chemical stability in the biological environment. To confer protection while preserving their plasmonic properties, we allied the advantages of both materials and produced hybrid nanoparticles made of an anisotropic silver nanoplate core coated with a frame of gold. The efficiency of these hybrid nanoparticles (Ag@AuNPs) in photothermia was compared to monometallic silver nanoplates (AgNPs) or gold nanostars (AuNPs). The structural and functional properties of AuNPs, AgNPs, and Ag@AuNPs were investigated in environments of increasing complexity, in water suspensions, in cells, and in tumors in vivo. While AgNPs showed the greatest heating efficiency in suspension (followed by Ag@AuNPs and AuNPs), this trend was reversed intracellularly within a tissue-mimetic model. In this setup, AgNPs failed to provide consistent photothermal conversion over time, due to structural damage induced by the intracellular environment. Remarkably, the degraded Ag was found to be stored within the iron-storage ferritin protein. By contrast, the Au shell provided the Ag@AuNPs with total Ag biopersistence. As a result, photothermal therapy was successful with Ag@AuNPs in vivo in a mouse tumor model, providing the ultimate proof on Au shell's capability to shield the Ag core from the harsh biological environment and preserve its excellent heating properties.


Assuntos
Ferritinas/metabolismo , Ouro/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Neoplasias da Próstata/terapia , Prata/uso terapêutico , Animais , Linhagem Celular , Ouro/metabolismo , Humanos , Hipertermia Induzida/métodos , Masculino , Nanopartículas Metálicas/ultraestrutura , Camundongos Nus , Células PC-3 , Fototerapia/métodos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Prata/metabolismo
15.
ACS Appl Mater Interfaces ; 9(31): 25775-25788, 2017 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-28723064

RESUMO

Hybrid nanogels, composed of thermoresponsive polymers and superparamagnetic nanoparticles, are attractive nanocarriers for biomedical applications, being able-as a polymer matrix-to uptake and release high quantities of chemotherapeutic agents and-as magnetic nanoparticles-to be heated when exposed to an alternative magnetic field (AMF), better known as magnetic hyperthermia. Herein, biocompatible, pH-responsive, magnetoresponsive, and thermoresponsive nanogels, based on oligo(ethylene glycol) methyl ether methacrylate monomers and a methacrylic acid comonomer were prepared by conventional precipitation radical copolymerization in water, post-assembled by complexation with iron oxide magnetic nanoparticles (MNPs) of maghemite (γ-Fe2O3), and loaded with an anticancer drug (doxorubicin, DOX), for remotely controlled drug release by a "hot spot", as an athermal magnetic hyperthermia strategy against cancer. These nanogels, denoted MagNanoGels, with a hydrodynamic diameter from 328 to 460 nm, as a function of the MNP content, have a swelling-deswelling behavior at their volume phase temperature transition around 47 °C in a physiological medium (pH 7.5), which is above the human body temperature (37 °C). Applying an alternative magnetic field increases the release of DOX by 2-fold, while no macroscopic heating was recorded. This enhanced drug release is due to a shrinking of the polymer network by local heating, as illustrated by the MagNanoGel size decrease under an AMF. In cancer cells, not only do the DOX-MagNanoGels internalize DOX more efficiently than free DOX, but also DOX intracellular release can be remotely triggered under an AMF, in athermal conditions, thus enhancing DOX cytotoxicity.


Assuntos
Nanopartículas , Doxorrubicina , Éter , Etilenoglicol , Compostos Férricos , Humanos , Concentração de Íons de Hidrogênio , Hipertermia Induzida , Metacrilatos , Éteres Metílicos
16.
Nanoscale ; 9(27): 9467-9480, 2017 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-28660946

RESUMO

The design of compact nanoprobes for multimodal bioimaging is a current challenge and may have a major impact on diagnostics and therapeutics. Multicomponent gold-iron oxide nanoparticles have shown high potential as contrast agents in numerous imaging techniques due to the complementary features of iron oxide and gold nanomaterials. In this paper we describe novel gold-iron oxide Janus magnetic-plasmonic nanoparticles as versatile nanoprobes for multimodal imaging. The nanoparticles are characterized as contrast agents for different imaging techniques, including X-ray computed tomography (CT), T2-weighted nuclear magnetic resonance imaging (MRI), photoacoustic imaging (PA), dark-field and bright-field optical microscopy, transmission electron microscopy (TEM), and surface enhanced Raman spectroscopy (SERS). We discuss the effect of particle size and morphology on their performance as contrast agents and show the advantage of a Janus configuration. Additionally, the uptake of nanoparticles by cells can be simultaneously visualized in dark- and bright-field optical microscopy, SERS mapping, and electron microscopy. These complementary techniques allow a complete view of cell uptake in an artifact-free manner, with multiplexing capabilities, and with extra information regarding the nanoparticles' fate inside the cells. Altogether, the results obtained with these non-invasive techniques show the high versatility of these nanoparticles, the advantages of a Janus configuration, and their high potential in multipurpose biomedical applications.


Assuntos
Meios de Contraste/química , Compostos Férricos/química , Ouro/química , Imagem Multimodal , Nanopartículas/química , Imageamento por Ressonância Magnética , Microscopia Eletrônica de Transmissão , Técnicas Fotoacústicas , Análise Espectral Raman , Tomografia Computadorizada por Raios X
18.
Adv Healthc Mater ; 5(9): 1040-8, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26990061

RESUMO

Gold nanoparticles are prime candidates for cancer thermotherapy. However, while the ultimate target for nanoparticle-mediated photothermal therapy is the cancer cell, heating performance has not previously been evaluated in the tumoral environment. A systematic investigation of gold nanostar heat-generating efficiency in situ is presented: not only in cancer cells in vitro but also after intratumoral injection in vivo. It is demonstrated that (i) in aqueous dispersion, heat generation is governed by particle size and exciting laser wavelength; (ii) in cancer cells in vitro, heat generation is still very efficient, but irrespective of both particle size and laser wavelength; and (iii) heat generation by nanostars injected into tumors in vivo evolves with time, as the nanostars are trafficked from the extracellular matrix into endosomes. The plasmonic heating response thus serves as a signature of nanoparticle internalization in cells, bringing the ultimate goal of nanoparticle-mediated photothermal therapy a step closer.


Assuntos
Ouro , Hipotermia Induzida , Nanopartículas Metálicas , Fototerapia , Neoplasias da Próstata/terapia , Microambiente Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral , Ouro/química , Ouro/farmacocinética , Ouro/farmacologia , Humanos , Masculino , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia
19.
ACS Nano ; 10(2): 2436-46, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26766814

RESUMO

The pursuit of innovative, multifunctional, more efficient, and safer treatments is a major challenge in preclinical nanoparticle-mediated thermotherapeutic research. Here, we report that iron oxide nanoparticles have the dual capacity to act as both magnetic and photothermal agents. We further explore every key aspect of this magnetophotothermal approach, choosing iron oxide nanocubes for their high efficiency for the magnetic hyperthermia modality itself. In aqueous suspension, the nanocubes' exposure to both: an alternating magnetic field and near-infrared laser irradiation (808 nm), defined as the DUAL-mode, amplifies the heating effect 2- to 5-fold by comparison with magnetic stimulation alone, yielding unprecedented heating powers (specific loss powers) up to 5000 W/g. In cancer cells, the laser excitation restores the optimal efficiency of magnetic hyperthermia, otherwise inhibited by intracellular confinement, resulting in a remarkable heating efficiency in the DUAL-mode (up to 15-fold amplification), with respect to the magnetophotothermal mode. As a consequence, the dual action yielded complete apoptosis-mediated cell death. In solid tumors in vivo, single-mode treatments (magnetic or laser hyperthermia) reduced tumor growth, while DUAL-mode treatment resulted in complete tumor regression, mediated by heat-induced tumoral cell apoptosis and massive denaturation of the collagen fibers, and a long-lasting thermal efficiency over repeated treatments.


Assuntos
Temperatura Alta , Hipertermia Induzida/métodos , Terapia com Luz de Baixa Intensidade/métodos , Campos Magnéticos , Nanopartículas Metálicas/química , Fotoquimioterapia/métodos , Apoptose , Linhagem Celular Tumoral , Compostos Férricos/química , Humanos , Nanopartículas Metálicas/efeitos da radiação
20.
Nanoscale ; 7(45): 18872-7, 2015 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-26468627

RESUMO

Multifunctional hybrid-design nanomaterials appear to be a promising route to meet the current therapeutics needs required for efficient cancer treatment. Herein, two efficient heat nano-generators were combined into a multifunctional single nanohybrid (a multi-core iron oxide nanoparticle optimized for magnetic hyperthermia, and a gold branched shell with tunable plasmonic properties in the NIR region, for photothermal therapy) which impressively enhanced heat generation, in suspension or in vivo in tumours, opening up exciting new therapeutic perspectives.


Assuntos
Hipertermia Induzida/métodos , Campos Magnéticos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapêutico , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA