Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Chem Soc Rev ; 50(20): 11614-11667, 2021 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-34661212

RESUMO

Magnetic hyperthermia (MHT) is a therapeutic modality for the treatment of solid tumors that has now accumulated more than 30 years of experience. In the ongoing MHT clinical trials for the treatment of brain and prostate tumors, iron oxide nanoparticles are employed as intra-tumoral MHT agents under a patient-safe 100 kHz alternating magnetic field (AMF) applicator. Although iron oxide nanoparticles are currently approved by FDA for imaging purposes and for the treatment of anemia, magnetic nanoparticles (MNPs) designed for the efficient treatment of MHT must respond to specific physical-chemical properties in terms of magneto-energy conversion, heat dose production, surface chemistry and aggregation state. Accordingly, in the past few decades, these requirements have boosted the development of a new generation of MNPs specifically aimed for MHT. In this review, we present an overview on MNPs and their assemblies produced via different synthetic routes, focusing on which MNP features have allowed unprecedented heating efficiency levels to be achieved in MHT and highlighting nanoplatforms that prevent magnetic heat loss in the intracellular environment. Moreover, we review the advances on MNP-based nanoplatforms that embrace the concept of multimodal therapy, which aims to combine MHT with chemotherapy, radiotherapy, immunotherapy, photodynamic or phototherapy. Next, for a better control of the therapeutic temperature at the tumor, we focus on the studies that have optimized MNPs to maintain gold-standard MHT performance and are also tackling MNP imaging with the aim to quantitatively assess the amount of nanoparticles accumulated at the tumor site and regulate the MHT field conditions. To conclude, future perspectives with guidance on how to advance MHT therapy will be provided.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita , Neoplasias , Humanos , Temperatura Alta , Hipertermia , Campos Magnéticos , Neoplasias/terapia
2.
Adv Funct Mater ; 30(28): 2002362, 2020 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-32684910

RESUMO

Here, cation exchange (CE) reactions are exploited to radiolabel ZnSe, ZnS, and CuFeS2 metal chalcogenide nanocrystals (NCs) with 64Cu. The CE protocol requires one simple step, to mix the water-soluble NCs with a 64Cu solution, in the presence of vitamin C used to reduce Cu(II) to Cu(I). Given the quantitative cation replacement on the NCs, a high radiochemical yield, up to 99%, is reached. Also, provided that there is no free 64Cu, no purification step is needed, making the protocol easily translatable to the clinic. A unique aspect of the approach is the achievement of an unprecedentedly high specific activity: by exploiting a volumetric CE, the strategy enables to concentrate a large dose of 64Cu (18.5 MBq) in a small NC dose (0.18 µg), reaching a specific activity of 103 TBq g-1. Finally, the characteristic dielectric resonance peak, still present for the radiolabeled 64Cu:CuFeS2 NCs after the partial-CE reaction, enables the generation of heat under clinical laser exposure (1 W cm-2). The synergic toxicity of photo-ablation and 64Cu ionization is here proven on glioblastoma and epidermoid carcinoma tumor cells, while no intrinsic cytotoxicity is seen from the NC dose employed for these dual experiments.

3.
Nano Lett ; 18(11): 6856-6866, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30336062

RESUMO

Herein, by studying a stepwise phase transformation of 23 nm FeO-Fe3O4 core-shell nanocubes into Fe3O4, we identify a composition at which the magnetic heating performance of the nanocubes is not affected by the medium viscosity and aggregation. Structural and magnetic characterizations reveal the transformation of the FeO-Fe3O4 nanocubes from having stoichiometric phase compositions into Fe2+-deficient Fe3O4 phases. The resultant nanocubes contain tiny compressed and randomly distributed FeO subdomains as well as structural defects. This phase transformation causes a 10-fold increase in the magnetic losses of the nanocubes, which remain exceptionally insensitive to the medium viscosity as well as aggregation unlike similarly sized single-phase magnetite nanocubes. We observe that the dominant relaxation mechanism switches from Néel in fresh core-shell nanocubes to Brownian in partially oxidized nanocubes and once again to Néel in completely treated nanocubes. The Fe2+ deficiencies and structural defects appear to reduce the magnetic energy barrier and anisotropy field, thereby driving the overall relaxation into Néel process. The magnetic losses of these nanoparticles remain unchanged through a progressive internalization/association to ovarian cancer cells. Moreover, the particles induce a significant cell death after being exposed to hyperthermia treatment. Here, we present the largest heating performance that has been reported to date for 23 nm iron oxide nanoparticles under intracellular conditions. Our findings clearly demonstrate the positive impacts of the Fe2+ deficiencies and structural defects in the Fe3O4 structure on the heating performance into intracellular environment.


Assuntos
Compostos Férricos/química , Hipertermia Induzida/métodos , Campos Magnéticos , Nanopartículas de Magnetita/química
4.
Int J Paediatr Dent ; 27(6): 568-573, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28387468

RESUMO

BACKGROUND: Sella turcica bridging (STB), or calcification of the interclinoid ligament of sella turcica, has been reported to be associated with some dental anomalies (palatal canine impaction and transposition). HYPOTHESIS OR AIM: The aim of the study was to find any association between canine impaction, hyperdontia or hypodontia and sellar dimensions or bridging. DESIGN: Lateral cephalometric radiographs from 78 patients with impacted canines, 68 with dental agenesis and 17 with hyperdontia were collected. Linear dimensions of sella turcica were calculated and compared to those of a control group (47 individuals). A standardize scoring scale was used to quantify the extent of STB from each radiographs. RESULTS: The frequency of partial and complete calcifications of sella in patients with dental anomalies is increased when compared to controls. STB can influence the interclinoid distance but does not affect other linear dimensions of sella. No statistically significant difference has been found in sellar dimensions and STB expression when evaluating radiographs at different ages. CONCLUSIONS: STB is frequently found in patients with dental abnormalities.


Assuntos
Calcinose/complicações , Sela Túrcica/patologia , Anormalidades Dentárias/complicações , Anodontia , Doenças Ósseas/complicações , Doenças Ósseas/diagnóstico por imagem , Calcinose/diagnóstico por imagem , Cefalometria , Humanos , Radiografia Dentária , Sela Túrcica/diagnóstico por imagem , Anormalidades Dentárias/diagnóstico por imagem , Dente Impactado , Dente Supranumerário
5.
Bioconjug Chem ; 26(4): 680-9, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25741889

RESUMO

The relationship between the positioning of ligands on the surface of nanoparticles and the structural features of nanoconjugates has been underestimated for a long time, albeit of primary importance to promote specific biological recognition at the nanoscale. In particular, it has been formerly observed that a proper molecular orientation can play a crucial role, first optimizing ligand immobilization onto the nanoparticles and, second, improving the targeting efficiency of the nanoconjugates. In this work, we present a novel strategy to afford peptide-oriented ligation using genetically modified cutinase fusion proteins, which combines the presence of a site-directed "capture" module based on an enzymatic unit and a "targeting" moiety consisting of the ligand terminal end of a genetically encoded polypeptide chain. As an example, the oriented presentation of U11 peptide, a sequence specific for the recognition of urokinase plasminogen activator receptor (uPAR), was achieved by enzyme-mediated conjugation with an irreversible inhibitor of cutinase, an alkylphosphonate p-nitrophenol ester linker, covalently bound to the surface of iron oxide nanoparticles. The targeting efficiency of the resulting protein-nanoparticle conjugates was assessed using uPAR-positive breast cancer cells exploiting confocal laser scanning microscopy and quantitative fluorescence analysis of confocal images. Ultrastructural analysis of transmission electron micrographs provided evidence of a receptor-mediated pathway of endocytosis. Our results showed that, despite the small average number of targeting peptides presented on the nanoparticles, our ligand-oriented nanoconjugates proved to be very effective in selectively binding to uPAR and in promoting the uptake in uPAR-positive cancer cells.


Assuntos
Hidrolases de Éster Carboxílico/química , Sistemas de Liberação de Medicamentos/métodos , Nanoconjugados/química , Peptídeos/química , Proteínas Recombinantes de Fusão/química , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/metabolismo , Linhagem Celular Tumoral , Endocitose , Compostos Férricos/química , Humanos , Modelos Moleculares , Nanoconjugados/ultraestrutura , Nanopartículas/química , Nanopartículas/ultraestrutura , Nitrofenóis/química , Peptídeos/genética , Peptídeos/metabolismo , Peptídeos/farmacologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Relação Estrutura-Atividade
6.
Bioconjug Chem ; 25(8): 1381-6, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25080049

RESUMO

The functionalization of colloidal nanoparticles with short peptides often fails in achieving satisfactory targeting efficiency and selectivity toward receptor-specific human cells. Here, we show that an optimized passivation of gold nanoparticle surface with a mixed self-assembled monolayer, including a targeting ligand, a fluorescent dye, and an intercalating short PEG derivative, led to a very stable, nontoxic, and efficient nanoconjugate for targeting urokinase plasminogen activator receptor-positive breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Ouro/química , Nanopartículas Metálicas/química , Terapia de Alvo Molecular , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Desenho de Fármacos , Corantes Fluorescentes/química , Humanos , Ligantes , Modelos Moleculares , Conformação Molecular , Polietilenoglicóis/química , Especificidade por Substrato
7.
Adv Sci (Weinh) ; 11(8): e2305769, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38054651

RESUMO

The application of lipid-based nanoparticles for COVID-19 vaccines and transthyretin-mediated amyloidosis treatment have highlighted their potential for translation to cancer therapy. However, their use in delivering drugs to solid tumors is limited by ineffective targeting, heterogeneous organ distribution, systemic inflammatory responses, and insufficient drug accumulation at the tumor. Instead, the use of lipid-based nanoparticles to remotely activate immune system responses is an emerging effective strategy. Despite this approach showing potential for treating hematological cancers, its application to treat solid tumors is hampered by the selection of eligible targets, tumor heterogeneity, and ineffective penetration of activated T cells within the tumor. Notwithstanding, the use of lipid-based nanoparticles for immunotherapy is projected to revolutionize cancer therapy, with the ultimate goal of rendering cancer a chronic disease. However, the translational success is likely to depend on the use of predictive tumor models in preclinical studies, simulating the complexity of the tumor microenvironment (e.g., the fibrotic extracellular matrix that impairs therapeutic outcomes) and stimulating tumor progression. This review compiles recent advances in the field of antitumor lipid-based nanoparticles and highlights emerging therapeutic approaches (e.g., mechanotherapy) to modulate tumor stiffness and improve T cell infiltration, and the use of organoids to better guide therapeutic outcomes.


Assuntos
Neuropatias Amiloides Familiares , Neoplasias , Humanos , Vacinas contra COVID-19 , Imunoterapia , Neoplasias/terapia , Lipídeos , Microambiente Tumoral
8.
Adv Sci (Weinh) ; 11(2): e2302965, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37946710

RESUMO

Interactions between living cells and nanoparticles are extensively studied to enhance the delivery of therapeutics. Nanoparticles size, shape, stiffness, and surface charge are regarded as the main features able to control the fate of cell-nanoparticle interactions. However, the clinical translation of nanotherapies has so far been limited, and there is a need to better understand the biology of cell-nanoparticle interactions. This study investigates the role of cellular mechanosensitive components in cell-nanoparticle interactions. It is demonstrated that the genetic and pharmacologic inhibition of yes-associated protein (YAP), a key component of cancer cell mechanosensing apparatus and Hippo pathway effector, improves nanoparticle internalization in triple-negative breast cancer cells regardless of nanoparticle properties or substrate characteristics. This process occurs through YAP-dependent regulation of endocytic pathways, cell mechanics, and membrane organization. Hence, the study proposes targeting YAP may sensitize triple-negative breast cancer cells to chemotherapy and increase the selectivity of nanotherapy.


Assuntos
Nanopartículas , Neoplasias de Mama Triplo Negativas , Humanos , Transdução de Sinais/fisiologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas de Sinalização YAP
9.
Matrix Biol ; 125: 12-30, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37944712

RESUMO

Extracellular matrix (ECM) tumorigenic alterations resulting in high matrix deposition and stiffening are hallmarks of adenocarcinomas and are collectively defined as desmoplasia. Here, we thoroughly analysed primary prostate cancer tissues obtained from numerous patients undergoing radical prostatectomy to highlight reproducible structural changes in the ECM leading to the loss of the glandular architecture. Starting from patient cells, we established prostate cancer tumoroids (PCTs) and demonstrated they require TGF-ß signalling pathway activity to preserve phenotypical and structural similarities with the tissue of origin. By modulating TGF-ß signalling pathway in PCTs, we unveiled its role in ECM accumulation and remodelling in prostate cancer. We also found that TGF-ß-induced ECM remodelling is responsible for the initiation of prostate cell epithelial-to-mesenchymal transition (EMT) and the acquisition of a migratory, invasive phenotype. Our findings highlight the cooperative role of TGF-ß signalling and ECM desmoplasia in prompting prostate cell EMT and promoting tumour progression and dissemination.


Assuntos
Neoplasias da Próstata , Fator de Crescimento Transformador beta , Masculino , Humanos , Fator de Crescimento Transformador beta/metabolismo , Transição Epitelial-Mesenquimal , Neoplasias da Próstata/patologia , Matriz Extracelular/metabolismo , Próstata/metabolismo , Linhagem Celular Tumoral
10.
Nanoscale ; 16(15): 7678-7689, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38533617

RESUMO

Magnetic nanoparticles (MNPs) provide new opportunities for enzyme-free biosensing of nucleic acid biomarkers and magnetic actuation by patterning on DNA origami, yet how the DNA grafting density affects their dynamics and accessibility remains poorly understood. Here, we performed surface functionalization of MNPs with single-stranded DNA (ssDNA) via click chemistry with a tunable grafting density, which enables the encapsulation of single MNPs inside a functional polymeric layer. We used several complementary methods to show that particle translational and rotational dynamics exhibit a sigmoidal dependence on the ssDNA grafting density. At low densities, ssDNA strands adopt a coiled conformation that results in minor alterations to particle dynamics, while at high densities, they organize into polymer brushes that collectively influence particle dynamics. Intermediate ssDNA densities, where the dynamics are most sensitive to changes, show the highest magnetic biosensing sensitivity for the detection of target nucleic acids. Finally, we demonstrate that MNPs with high ssDNA grafting densities are required to efficiently couple to DNA origami. Our results establish ssDNA grafting density as a critical parameter for the functionalization of MNPs for magnetic biosensing and functionalization of DNA nanostructures.


Assuntos
Nanopartículas de Magnetita , Ácidos Nucleicos , DNA/química , DNA de Cadeia Simples , Fenômenos Magnéticos , Conformação de Ácido Nucleico
11.
Adv Mater ; 34(13): e2107964, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35100658

RESUMO

Reconfiguring the structure and selectivity of existing chemotherapeutics represents an opportunity for developing novel tumor-selective drugs. Here, as a proof-of-concept, the use of high-frequency sound waves is demonstrated to transform the nonselective anthracycline doxorubicin into a tumor selective drug molecule. The transformed drug self-aggregates in water to form ≈200 nm nanodrugs without requiring organic solvents, chemical agents, or surfactants. The nanodrugs preferentially interact with lipid rafts in the mitochondria of cancer cells. The mitochondrial localization of the nanodrugs plays a key role in inducing reactive oxygen species mediated selective death of breast cancer, colorectal carcinoma, ovarian carcinoma, and drug-resistant cell lines. Only marginal cytotoxicity (80-100% cell viability) toward fibroblasts and cardiomyocytes is observed, even after administration of high doses of the nanodrug (25-40 µg mL-1 ). Penetration, cytotoxicity, and selectivity of the nanodrugs in tumor-mimicking tissues are validated by using a 3D coculture of cancer and healthy cells and 3D cell-collagen constructs in a perfusion bioreactor. The nanodrugs exhibit tropism for lung and limited accumulation in the liver and spleen, as suggested by in vivo biodistribution studies. The results highlight the potential of this approach to transform the structure and bioactivity of anticancer drugs and antibiotics bearing sono-active moieties.


Assuntos
Nanopartículas , Neoplasias Ovarianas , Antibióticos Antineoplásicos/química , Doxorrubicina/química , Doxorrubicina/farmacologia , Humanos , Nanopartículas/química , Distribuição Tecidual
12.
Nanoscale ; 14(8): 3049-3061, 2022 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-35142755

RESUMO

Real-time detection and nanoscale imaging of human immunodeficiency virus type 1 ribonucleic acid (HIV-1 RNA) in latently infected cells that persist in people living with HIV-1 on antiretroviral therapy in blood and tissue may reveal new insights needed to cure HIV-1 infection. Herein, we develop a strategy combining DNA nanotechnology and super-resolution expansion microscopy (ExM) to detect and image a 22 base sequence transcribed from the HIV-1 promoter in model live and fixed cells. We engineer a chimeric locked nucleic acid (LNA)-DNA sensor via hybridization chain reaction to probe HIV-1 RNA in the U3 region of the HIV-1 long terminal repeat (LTR) by signal amplification in live cells. We find that the viral RNA transcript of the U3 region of the HIV-1 LTR, namely PromA, is a valid and specific biomarker to detect infected live cells. The efficiency and selectivity of the LNA-DNA sensor are evaluated in combination with ExM. Unlike standard ExM methods, which rely on additional custom linkers to anchor and immobilize RNA molecules in the intracellular polymeric network, in the current strategy, we probe and image the HIV-1 RNA target at nanoscale resolution, without resorting to chemical linkers or additional preparation steps. This is achieved by physical entrapment of the HIV-1 viral transcripts in the cells post-expansion by finely tuning the mesh size of the intracellular polymeric network.


Assuntos
HIV-1 , DNA , HIV-1/genética , Humanos , Oligonucleotídeos , RNA Viral/genética
13.
Sci Rep ; 12(1): 17409, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36257968

RESUMO

Cardiovascular diseases remain the leading cause of death worldwide; hence there is an increasing focus on developing physiologically relevant in vitro cardiovascular tissue models suitable for studying personalized medicine and pre-clinical tests. Despite recent advances, models that reproduce both tissue complexity and maturation are still limited. We have established a scaffold-free protocol to generate multicellular, beating human cardiac microtissues in vitro from hiPSCs-namely human organotypic cardiac microtissues (hOCMTs)-that show some degree of self-organization and can be cultured for long term. This is achieved by the differentiation of hiPSC in 2D monolayer culture towards cardiovascular lineage, followed by further aggregation on low-attachment culture dishes in 3D. The generated hOCMTs contain multiple cell types that physiologically compose the heart and beat without external stimuli for more than 100 days. We have shown that 3D hOCMTs display improved cardiac specification, survival and metabolic maturation as compared to standard monolayer cardiac differentiation. We also confirmed the functionality of hOCMTs by their response to cardioactive drugs in long-term culture. Furthermore, we demonstrated that they could be used to study chemotherapy-induced cardiotoxicity. Due to showing a tendency for self-organization, cellular heterogeneity, and functionality in our 3D microtissues over extended culture time, we could also confirm these constructs as human cardiac organoids (hCOs). This study could help to develop more physiologically-relevant cardiac tissue models, and represent a powerful platform for future translational research in cardiovascular biology.


Assuntos
Antineoplásicos , Fármacos Cardiovasculares , Células-Tronco Pluripotentes Induzidas , Humanos , Engenharia Tecidual/métodos , Coração/fisiologia , Diferenciação Celular/fisiologia , Fármacos Cardiovasculares/metabolismo , Antineoplásicos/metabolismo , Miócitos Cardíacos/metabolismo
14.
Artigo em Inglês | MEDLINE | ID: mdl-32391340

RESUMO

The research for heart therapies is challenged by the limited intrinsic regenerative capacity of the adult heart. Moreover, it has been hampered by the poor results obtained by tissue engineering and regenerative medicine attempts at generating functional beating constructs able to integrate with the host tissue. For this reason, organ transplantation remains the elective treatment for end-stage heart failure, while novel strategies aiming to promote cardiac regeneration or repair lag behind. The recent discovery that adult cardiomyocytes can be ectopically induced to enter the cell cycle and proliferate by a combination of microRNAs and cardioprotective drugs, like anti-oxidant, anti-inflammatory, anti-coagulants and anti-platelets agents, fueled the quest for new strategies suited to foster cardiac repair. While proposing a revolutionary approach for heart regeneration, these studies raised serious issues regarding the efficient controlled delivery of the therapeutic cargo, as well as its timely removal or metabolic inactivation from the site of action. Especially, there is need for innovative treatment because of evidence of severe side effects caused by pleiotropic drugs. Biocompatible nanoparticles possess unique physico-chemical properties that have been extensively exploited for overcoming the limitations of standard medical therapies. Researchers have put great efforts into the optimization of the nanoparticles synthesis and functionalization, to control their interactions with the biological milieu and use as a viable alternative to traditional approaches. Nanoparticles can be used for diagnosis and deliver therapies in a personalized and targeted fashion. Regarding the treatment of cardiovascular diseases, nanoparticles-based strategies have provided very promising outcomes, in preclinical studies, during the last years. Efficient encapsulation of a large variety of cargos, specific release at the desired site and improvement of cardiac function are some of the main achievements reached so far by nanoparticle-based treatments in animal models. This work offers an overview on the recent nanomedical applications for cardiac regeneration and highlights how the versatility of nanomaterials can be combined with the newest molecular biology discoveries to advance cardiac regeneration therapies.

15.
Curr Med Chem ; 27(42): 7234-7255, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32586245

RESUMO

Nanodrugs represent novel solutions to reshuffle repurposed drugs for cancer therapy. They might offer different therapeutic options by combining targeted drug delivery and imaging in unique platforms. Such nanomaterials are deemed to overcome the limitations of currently available treatments, ultimately improving patients' life quality. However, despite these promises being made for over three decades, the poor clinical translation of nanoparticle- based therapies calls for deeper in vit.. and in vivo investigations. Translational issues arise very early during the development of nanodrugs, where complex and more reliable cell models are often replaced by easily accessible and convenient 2D monocultures. This is particularly true in the field of cancer therapy. In fact, 2D monocultures provide poor information about the real impact of the nanodrugs in a complex living organism, especially given the poor mimicry of the solid Tumors Microenvironment (TME). The dense and complex extracellular matrix (ECM) of solid tumors dramatically restricts nanoparticles efficacy, impairing the successful implementation of nanodrugs in medical applications. Herein, we propose a comprehensive guideline of the 3D cell culture models currently available, including their potential and limitations for the evaluation of nanodrugs activity. Advanced culture techniques, more closely resembling the physiological conditions of the TME, might give a better prediction of the reciprocal interactions between cells and nanoparticles and eventually help reconsider the use of old drugs for new applications.


Assuntos
Neoplasias , Sistemas de Liberação de Medicamentos , Humanos , Nanopartículas , Nanoestruturas , Neoplasias/tratamento farmacológico , Preparações Farmacêuticas , Microambiente Tumoral
16.
ACS Appl Mater Interfaces ; 11(45): 41957-41971, 2019 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-31584801

RESUMO

The design of magnetic nanostructures whose magnetic heating efficiency remains unaffected at the tumor site is a fundamental requirement to further advance magnetic hyperthermia in the clinic. This work demonstrates that the confinement of magnetic nanoparticles (NPs) into a sub-micrometer cavity is a key strategy to enable a certain degree of nanoparticle motion and minimize aggregation effects, consequently preserving the magnetic heat loss of iron oxide nanocubes (IONCs) under different conditions, including intracellular environments. We fabricated magnetic layer-by-layer (LbL) self-assembled polyelectrolyte sub-micrometer capsules using three different approaches, and we studied their heating efficiency as obtained in aqueous dispersions and after internalization by tumor cells. First, IONCs were added to the hollow cavities of LbL submicrocapsules, allowing the IONCs to move to a certain extent in the capsule cavities. Second, IONCs were coencapsulated into solid calcium carbonate cores coated with LbL polymer shells. Third, IONCs were incorporated within the polymer layers of the LbL capsule walls. In aqueous solution, higher specific absorption rate (SAR) values were related to those of free IONCs, while lower SAR values were recorded for capsule/core assemblies. However, after uptake by cancer cell lines (SKOV-3 cells), the SAR values of the free IONCs were significantly lower than those observed for capsule/core assemblies, especially after prolonged incubation periods (24 and 48 h). These results show that IONCs packed into submicrocavities preserve the magnetic losses, as the SAR values remained almost invariable. Conversely, free IONCs without the protective capsule shell agglomerated and their magnetic losses were strongly reduced. Indeed, IONC-loaded capsules and free IONCs reside inside endosomal and lysosomal compartments after cellular uptake and show strongly reduced magnetic losses due to the immobilization and aggregation in centrosymmetrical structures in the intracellular vesicles. The confinement of IONCs into sub-micrometer cavities is a key strategy to provide a sustained and predictable heating dose inside biological matrices.

17.
Adv Healthc Mater ; 8(3): e1801313, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30614638

RESUMO

Oil-in-water emulsions represent a promising carrier for in vivo imaging because of the possibility to convey poorly water-soluble species. To promote accumulation at the tumor site and prolong circulation time, reduction of carrier size and surface PEGylation plays a fundamental role. In this work a novel, simple method to design an oil-core/PEG-shell nanocarrier is reported. A PEG-shell is grown around a monodisperse oil-in-water nanoemulsion with a one-pot method, using the radical polymerization of poly(ethylene glycol)diacrylate. PEG polymerization is triggered by UV, obtaining a PEG-shell with tunable thickness. This core-shell nanosystem combines the eluding feature of the PEG with the ability to confine high payloads of lipophilic species. Indeed, the core is successfully loaded with a lipophilic contrast agent, namely super paramagnetic iron oxide nanocubes. Interestingly, it is demonstrated an in vitro and an in vivo MRI response of the nanocapsules. Additionally, when the nanosystem loaded with nanocubes is mixed with a fluorescent contrast agent, indo-cyanine green, a relevant in vitro photoacoustic effect is observed. Moreover, viability and cellular uptake studies show no significant cell cytotoxicity. These results, together with the choice of low cost materials and the scale up production, make this nanocarrier a potential platform for in vivo imaging.


Assuntos
Meios de Contraste , Portadores de Fármacos/química , Compostos Férricos , Imageamento por Ressonância Magnética , Nanopartículas/química , Óleos , Polietilenoglicóis , Meios de Contraste/química , Meios de Contraste/farmacologia , Compostos Férricos/química , Compostos Férricos/farmacologia , Células HT29 , Humanos , Verde de Indocianina/química , Verde de Indocianina/farmacologia , Células MCF-7 , Óleos/química , Óleos/farmacologia , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia
18.
ACS Appl Mater Interfaces ; 9(40): 35095-35104, 2017 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-28858466

RESUMO

In this work, the versatility of layer-by-layer technology was combined with the magnetic response of iron oxide nanobeads to prepare magnetic mesostructures with a degradable multilayer shell into which a dye quenched ovalbumin conjugate (DQ-OVA) was loaded. The system was specifically designed to prove the protease sensitivity of the hybrid mesoscale system and the easy detection of the ovalbumin released. The uptake of the nanostructures in the breast cancer cells was followed by the effective release of DQ-OVA upon activation via the intracellular proteases degradation of the polymer shells. Monitoring the fluorescence rising due to DQ-OVA digestion and the cellular dye distribution, together with the electron microscopy studying, enabled us to track the shell degradation and the endosomal uptake pathway that resulted in the release of the digested fragments of DQ ovalbumin in the cytosol.


Assuntos
Magnetismo , Nanopartículas , Nanoestruturas , Ovalbumina , Peptídeo Hidrolases , Polímeros
19.
ACS Nano ; 11(12): 12121-12133, 2017 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-29155560

RESUMO

Magnetic hyperthermia (MH) based on magnetic nanoparticles (MNPs) is a promising adjuvant therapy for cancer treatment. Particle clustering leading to complex magnetic interactions affects the heat generated by MNPs during MH. The heat efficiencies, theoretically predicted, are still poorly understood because of a lack of control of the fabrication of such clusters with defined geometries and thus their functionality. This study aims to correlate the heating efficiency under MH of individually coated iron oxide nanocubes (IONCs) versus soft colloidal nanoclusters made of small groupings of nanocubes arranged in different geometries. The controlled clustering of alkyl-stabilized IONCs is achieved here during the water transfer procedure by tuning the fraction of the amphiphilic copolymer, poly(styrene-co-maleic anhydride) cumene-terminated, to the nanoparticle surface. It is found that increasing the polymer-to-nanoparticle surface ratio leads to the formation of increasingly large nanoclusters with defined geometries. When compared to the individual nanocubes, we show here that controlled grouping of nanoparticles-so-called "dimers" and "trimers" composed of two and three nanocubes, respectively-increases specific absorption rate (SAR) values, while conversely, forming centrosymmetric clusters having more than four nanocubes leads to lower SAR values. Magnetization measurements and Monte Carlo-based simulations support the observed SAR trend and reveal the importance of the dipolar interaction effect and its dependence on the details of the particle arrangements within the different clusters.


Assuntos
Compostos Férricos/química , Hipertermia Induzida , Nanopartículas de Magnetita/química , Neoplasias/tratamento farmacológico , Quimioterapia Adjuvante , Coloides/química , Compostos Férricos/síntese química , Compostos Férricos/uso terapêutico , Humanos , Nanopartículas de Magnetita/uso terapêutico , Simulação de Dinâmica Molecular , Estrutura Molecular , Método de Monte Carlo , Tamanho da Partícula , Propriedades de Superfície
20.
Sci Rep ; 6: 33295, 2016 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-27665698

RESUMO

Here, we propose the use of magnetic hyperthermia as a means to trigger the oxidation of Fe1-xO/Fe3-δO4 core-shell nanocubes to Fe3-δO4 phase. As a first relevant consequence, the specific absorption rate (SAR) of the initial core-shell nanocubes doubles after exposure to 25 cycles of alternating magnetic field stimulation. The improved SAR value was attributed to a gradual transformation of the Fe1-xO core to Fe3-δO4, as evidenced by structural analysis including high resolution electron microscopy and Rietveld analysis of X-ray diffraction patterns. The magnetically oxidized nanocubes, having large and coherent Fe3-δO4 domains, reveal high saturation magnetization and behave superparamagnetically at room temperature. In comparison, the treatment of the same starting core-shell nanocubes by commonly used thermal annealing process renders a transformation to γ-Fe2O3. In contrast to other thermal annealing processes, the method here presented has the advantage of promoting the oxidation at a macroscopic temperature below 37 °C. Using this soft oxidation process, we demonstrate that biotin-functionalized core-shell nanocubes can undergo a mild self-oxidation transformation without losing their functional molecular binding activity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA