Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Nanotoxicology ; 15(9): 1151-1167, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34752713

RESUMO

This article presents an automated image-processing workflow for quantitative assessment of SPION accumulation in tissue sections stained with Prussian blue for iron detection. We utilized supervised machine learning with manually labeled features used for training the classifier. Performance of the classifier was validated by 10-fold cross-validation of obtained data and by measuring Dice and Jaccard Similarity Coefficients between manually segmented image and automated segmentation. The proposed approach provides time and cost-effective solution for quantitative imaging analysis of SPION in tissue with a precision similar to that obtained via thresholding method for stain quantification. Furthermore, we exploited the classifiers to generate segmented 3D volumes from histological slides. This enabled visualization of particles which were obscured in original 3D histology stacks. Our approach offers a powerful tool for preclinical assessment of the precise tissue-specific SPION biodistribution, which could affect both their toxicity and their efficacy as nanocarriers for medicines.


Assuntos
Processamento de Imagem Assistida por Computador , Nanopartículas Magnéticas de Óxido de Ferro , Processamento de Imagem Assistida por Computador/métodos , Distribuição Tecidual
2.
Curr Opin Pharmacol ; 56: 85-92, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33341460

RESUMO

Nanotechnology in medicine-nanomedicine-is extensively employed to diagnose, treat, and prevent pulmonary diseases. Over the last few years, this brave new world has made remarkable progress, offering opportunities to address historical clinical challenges in pulmonary diseases including multidrug resistance, adverse side effects of conventional therapeutic agents, novel imaging, and earlier disease detection. Nanomedicine is also being applied to tackle the new emerging infectious diseases, including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East Respiratory Syndrome Coronavirus (MERS-CoV), influenza A virus subtype H1N1 (A/H1N1), and more recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this review we provide both a historical overview of the application of nanomedicine to respiratory diseases and more recent cutting-edge approaches such as nanoparticle-mediated combination therapies, novel double-targeted nondrug delivery system for targeting, stimuli-responsive nanoparticles, and theranostic imaging in the diagnosis and treatment of pulmonary diseases.


Assuntos
Nanotecnologia/métodos , Pneumologia/métodos , Doenças Respiratórias/tratamento farmacológico , Animais , Infecções por Coronavirus/tratamento farmacológico , Portadores de Fármacos , Resistência a Medicamentos/fisiologia , Humanos
3.
Nanomedicine (Lond) ; 15(30): 2933-2953, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33241979

RESUMO

Aim: Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine, which has been shown to promote disease severity in cystic fibrosis. Methods: In this study, aerosolized drug-loaded nanoparticles containing SCD-19, an inhibitor of MIF's tautomerase enzymatic activity, were developed and characterized. Results: The aerosolized nanoparticles had an optimal droplet size distribution for deep lung deposition, with a high degree of biocompatibility and significant cellular uptake. Conclusion: For the first time, we have developed an aerosolized nano-formulation against MIF's enzymatic activity that achieved a significant reduction in the inflammatory response of macrophages, and inhibited Pseudomonas aeruginosa biofilm formation on airway epithelial cells. This represents a potential novel adjunctive therapy for the treatment of P. aeruginosa infection in cystic fibrosis.


Assuntos
Fatores Inibidores da Migração de Macrófagos , Nanopartículas , Preparações Farmacêuticas , Infecções por Pseudomonas , Biofilmes , Humanos , Inflamação/tratamento farmacológico , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa
4.
Nanomaterials (Basel) ; 10(3)2020 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-32213961

RESUMO

Magnetic hyperthermia involves the use of iron oxide nanoparticles to generate heat in tumours following stimulation with alternating magnetic fields. In recent times, this treatment has undergone numerous clinical trials in various solid malignancies and subsequently achieved clinical approval to treat glioblastoma and prostate cancer in 2011 and 2018, respectively. However, despite recent clinical advances, many questions remain with regard to the underlying mechanisms involved in this therapy. One such query is whether intracellular or extracellular nanoparticles are necessary for treatment efficacy. Herein, we compare the effects of intracellular and extracellular magnetic hyperthermia in BxPC-3 cells to determine the differences in efficacy between both. Extracellular magnetic hyperthermia at temperatures between 40-42.5 °C could induce significant levels of necrosis in these cells, whereas intracellular magnetic hyperthermia resulted in no change in viability. This led to a discussion on the overall relevance of intracellular nanoparticles to the efficacy of magnetic hyperthermia therapy.

5.
Nanomedicine ; 20: 101983, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30940505

RESUMO

In this paper we show that conjugation of magnetic nanoparticles (MNPs) with Gemcitabine and/or NucAnt (N6L) fostered their internalization into pancreatic tumor cells and that the coupling procedure did not alter the cytotoxic potential of the drugs. By treating tumor cells (BxPC3 and PANC-1) with the conjugated MNPs and magnetic hyperthermia (43 °C, 60 min), cell death was observed. The two pancreatic tumor cell lines showed different reactions against the combined therapy according to their intrinsic sensitivity against Gemcitabine (cell death, ROS production, ability to activate ERK 1/2 and JNK). Finally, tumors (e.g. 3 mL) could be effectively treated by using almost 4.2 × 105 times lower Gemcitabine doses compared to conventional therapies. Our data show that this combinatorial therapy might well play an important role in certain cell phenotypes with low readiness of ROS production. This would be of great significance in distinctly optimizing local pancreatic tumor treatments.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita/química , Neoplasias Pancreáticas/patologia , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Antígeno Ki-67/metabolismo , Nanopartículas de Magnetita/ultraestrutura , Camundongos Nus , Peptídeos/farmacologia , Fenótipo , Fase S/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
6.
Sci Rep ; 8(1): 12920, 2018 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-30150787

RESUMO

Evidence supports the advantages of inhalation over other drug-administration routes in the treatment of lung diseases, including cancer. Although data obtained from animal models and conventional in vitro cultures are informative, testing the efficacy of inhaled chemotherapeutic agents requires human-relevant preclinical tools. Such tools are currently unavailable. Here, we developed and characterized in vitro models for the efficacy testing of inhaled chemotherapeutic agents against non-small-cell lung cancer (NSCLC). These models recapitulated key elements of both the lung epithelium and the tumour tissue, namely the direct contact with the gas phase and the three-dimensional (3D) architecture. Our in vitro models were formed by growing, for the first time, human adenocarcinoma (A549) cells as multilayered mono-cultures at the Air-Liquid Interface (ALI). The in vitro models were tested for their response to four benchmarking chemotherapeutics, currently in use in clinics, demonstrating an increased resistance to these drugs as compared to sub-confluent monolayered 2D cell cultures. Chemoresistance was comparable to that detected in 3D hypoxic tumour spheroids. Being cultured in ALI conditions, the multilayered monocultures demonstrated to be compatible with testing drugs administered as a liquid aerosol by a clinical nebulizer, offering an advantage over 3D tumour spheroids. In conclusion, we demonstrated that our in vitro models provide new human-relevant tools allowing for the efficacy screening of inhaled anti-cancer drugs.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Células A549 , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos
7.
J Interdiscip Nanomed ; 3(1): 16-28, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29938115

RESUMO

The size-dependent optical properties of quantum dots (QDs) are frequently exploited for use in medical imaging and labelling applications. Similarly, presented here, they also elicit profound size-dependent anticoagulant properties. Cadmium telluride quantum dot (QDs) (3.2 nm) were shown to have a dramatic anticoagulant effect centred on around the intrinsic coagulation pathway, compared to their 3.6 nm counterparts. Several clinically relevant diagnostic tests were carried out over a concentration range of the QDs and demonstrated that the 3.2 nm QDs elicited their response on the intrinsic pathway as a whole, yet the activity of the individual intrinsic coagulation factors was not affected. The mechanism appears also to be strongly influenced by the concentration of calcium ions and not cadmium ions leached from the QDs. Static and shear-based primary haemostasis assays were also carried out, demonstrating a profound anticoagulant effect which was independent of platelets and phospholipids. The data presented here suggest that the physical-chemical properties of the QDs may have a role in the modulation of haemostasis and the coagulation cascade, in a yet not fully understood mechanism. This study has implications for the use of similar QDs as diagnostic or therapeutic tools in vivo, and for the occupational health and safety of those working with such materials.

8.
Tissue Cell ; 50: 15-30, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29429514

RESUMO

Alternative models such as three-dimensional (3D) cell cultures represent a distinct milestone towards capturing the realities of cancer biology in vitro and reduce animal experimentation in the preclinical stage of drug discovery. Significant work remains to be done to understand how substrates used in in vitro alternatives influence cancer cells phenotype and drug efficacy responses, so that to accurately link such models to specific in vivo disease scenarios. Our study describes how the morphological, mechanical and biochemical properties of adenocarcinoma (A549) cells change in response to a 3D environment and varying substrates. Confocal Laser Scanning (LSCM), He-Ion (HIM) and Atomic Force (AFM) microscopies, supported by ELISA and Western blotting, were used. These techniques enabled us to evaluate the shape, cytoskeletal organization, roughness, stiffness and biochemical signatures of cells grown within soft 3D matrices (PuraMatrix™ and Matrigel™), and to compare them to those of cells cultured on two-dimensional glass substrates. Cell cultures are also characterized for their biological response to docetaxel, a taxane-type drug used in Non-Small-Cell Lung Cancer (NSCLC) treatment. Our results offer an advanced biophysical insight into the properties and potential application of 3D cultures of A549 cells as in vitro alternatives in lung cancer research.


Assuntos
Adenocarcinoma/tratamento farmacológico , Fenômenos Biofísicos , Técnicas de Cultura de Células/métodos , Neoplasias Pulmonares/tratamento farmacológico , Células Tumorais Cultivadas/ultraestrutura , Células A549 , Adenocarcinoma/química , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Docetaxel , Ensaio de Imunoadsorção Enzimática , Humanos , Neoplasias Pulmonares/química , Neoplasias Pulmonares/patologia , Microscopia Confocal , Especificidade por Substrato , Taxoides/farmacologia , Células Tumorais Cultivadas/química , Células Tumorais Cultivadas/efeitos dos fármacos
9.
Artigo em Inglês | MEDLINE | ID: mdl-29430177

RESUMO

Objective: The objective of the study was to determine whether the cadmium-derived materials induce intracellular protein citrullination. Methods: Human A549 lung epithelial cells were exposed to cadmium in soluble and nanoparticulate forms represented by cadmium chloride (CdCl2) and cadmium oxide (CdO), respectively, and their combinations with ultrafine carbon black (ufCB) produced by high temperature combustion, imitating cigarette burning. Protein citrullination in cell lysates was analyzed by Western immunoblotting and verified by immunofluorescent confocal microscopy. Target citrullinated proteins were identified by proteomic analysis. Results: CdO, ufCB and its combination with CdCl2 and CdO after high temperature combustion induced protein citrullination in cultured human lung epithelial cells, as detected by immunoblotting with anti-citrullinated protein antibody. Cytokeratins of type II (1, 2, 5, 6A, 6B and 77) and type I (9, 10) were identified as major intracellular citrullination targets. Immunofluorescent staining confirmed the localization of citrullinated proteins both in the cytoplasm and cell nuclei. Conclusion: Cadmium oxide nanoparticle exposure facilitated post-translational citrullination of proteins.


Assuntos
Cloreto de Cádmio/toxicidade , Compostos de Cádmio/toxicidade , Citrulina/metabolismo , Células Epiteliais/efeitos dos fármacos , Queratinas/metabolismo , Pulmão/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Óxidos/toxicidade , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Células A549 , Citrulinação , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Pulmão/metabolismo , Pulmão/patologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Medição de Risco , Fumar/efeitos adversos
10.
Sci Rep ; 8(1): 679, 2018 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-29330439

RESUMO

Citrullination, or the post-translational deimination of polypeptide-bound arginine, is involved in several pathological processes in the body, including autoimmunity and tumorigenesis. Recent studies have shown that nanomaterials can trigger protein citrullination, which might constitute a common pathogenic link to disease development. Here we demonstrated auto-antibody production in serum of nanomaterials-treated mice. Citrullination-associated phenomena and PAD levels were found to be elevated in nanomaterials -treated cell lines as well as in the spleen, kidneys and lymph nodes of mice, suggesting a systemic response to nanomaterials injection, and validated in human pleural and pericardial malignant mesothelioma (MM) samples. The observed systemic responses in mice exposed to nanomaterials support the evidence linking exposure to environmental factors with the development of autoimmunity responses and reinforces the need for comprehensive safety screening of nanomaterials. Furthermore, these nanomaterials induce pathological processes that mimic those observed in Pleural MM, and therefore require further investigations into their carcinogenicity.


Assuntos
Autoanticorpos/sangue , Hidrolases/metabolismo , Nanofios/administração & dosagem , Níquel/química , Proteínas/metabolismo , Células A549 , Animais , Formação de Anticorpos , Linhagem Celular Tumoral , Citrulinação , Feminino , Humanos , Hidrolases/imunologia , Rim/metabolismo , Rim/patologia , Linfonodos/metabolismo , Linfonodos/patologia , Mesotelioma/metabolismo , Mesotelioma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Nanofios/química , Baço/metabolismo , Baço/patologia
11.
Int J Nanomedicine ; 11: 791-822, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27013874

RESUMO

The main objective of this study was to optimize and characterize a drug delivery carrier for doxorubicin, intended to be intravenously administered, capable of improving the therapeutic index of the chemotherapeutic agent itself, and aimed at the treatment of pancreatic cancer. In light of this goal, we report a robust one-step method for the synthesis of dicarboxylic acid-terminated polyethylene glycol (PEG)-gold nanoparticles (AuNPs) and doxorubicin-loaded PEG-AuNPs, and their further antibody targeting (anti-Kv11.1 polyclonal antibody [pAb]). In in vitro proof-of-concept studies, we evaluated the influence of the nanocarrier and of the active targeting functionality on the anti-tumor efficacy of doxorubicin, with respect to its half-maximal effective concentration (EC50) and drug-triggered changes in the cell cycle. Our results demonstrated that the therapeutic efficacy of doxorubicin was positively influenced not only by the active targeting exploited through anti-Kv11.1-pAb but also by the drug coupling with a nanometer-sized delivery system, which indeed resulted in a 30-fold decrease of doxorubicin EC50, cell cycle blockage, and drug localization in the cell nuclei. The cell internalization pathway was strongly influenced by the active targeting of the Kv11.1 subunit of the human Ether-à-go-go related gene 1 (hERG1) channel aberrantly expressed on the membrane of pancreatic cancer cells. Targeted PEG-AuNPs were translocated into the lysosomes and were associated to an increased lysosomal function in PANC-1 cells. Additionally, doxorubicin release into an aqueous environment was almost negligible after 7 days, suggesting that drug release from PEG-AuNPs was triggered by enzymatic activity. Although preliminary, data gathered from this study have considerable potential in the application of safe-by-design nano-enabled drug-delivery systems (ie, nanomedicines) for the treatment of pancreatic cancer, a disease with a poor prognosis and one of the main current burdens of today's health care bill of industrialized countries.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Ouro/química , Nanopartículas Metálicas/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Polietilenoglicóis/química , Antibióticos Antineoplásicos/química , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Doxorrubicina/química , Portadores de Fármacos/uso terapêutico , Humanos , Técnicas In Vitro , Nanopartículas Metálicas/química , Neoplasias Pancreáticas/patologia , Células Tumorais Cultivadas
12.
Clin Exp Pharmacol Physiol ; 43(3): 319-26, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26707795

RESUMO

Manufactured nanomaterials have a variety of medical applications, including diagnosis and targeted treatment of cancer. A series of experiments were conducted to determine the pharmacokinetic, biodistribution and biocompatibility of two novel magnetic nanoparticles (MNPs) in the anaesthetized pig. Dimercaptosuccinic acid (DMSA) coated superparamagnetic iron oxide nanoparticles (MF66-labelled 12 nm, core nominal diameter and OD15 15 nm); at 0.5, or 2.0 mg/kg) were injected intravenously. Particles induced a dose-dependent decrease in blood pressure following administration which recovered to control levels several minutes after injection. Blood samples were collected for a 5-h period and stored for determination of particle concentration using particle electron paramagnetic resonance (pEPR). Organs were harvested post-mortem for magnetic resonance imaging (MRI at 1.5 T field strength) and histology. OD15 (2.0 mg/kg) MNP had a plasma half-life of approximately 15 min. Both doses of the MF66 (0.5 and 2.0 mg/kg) MNP were below detection limits. MNP accumulation was observed primarily in the liver and spleen with MRI scans which was confirmed by histology. MRI also showed that both MNPs were present in the lungs. The results show that further modifications may be required to improve the biocompatibility of these particles for use as diagnostic and therapeutic agents.


Assuntos
Compostos Férricos/química , Compostos Férricos/farmacocinética , Imãs , Suínos , Anestesia , Animais , Pressão Sanguínea/efeitos dos fármacos , Compostos Férricos/efeitos adversos , Compostos Férricos/sangue , Pulmão/citologia , Pulmão/efeitos dos fármacos , Imageamento por Ressonância Magnética , Tamanho da Partícula , Distribuição Tecidual
13.
Theranostics ; 5(11): 1249-63, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26379790

RESUMO

In a report from 2008, The International Agency for Research on Cancer predicted a tripled cancer incidence from 1975, projecting a possible 13-17 million cancer deaths worldwide by 2030. While new treatments are evolving and reaching approval for different cancer types, the main prevention of cancer mortality is through early diagnosis, detection and treatment of malignant cell growth. The last decades have seen a development of new imaging techniques now in widespread clinical use. The development of nano-imaging through fluorescent imaging and magnetic resonance imaging (MRI) has the potential to detect and diagnose cancer at an earlier stage than with current imaging methods. The characteristic properties of nanoparticles result in their theranostic potential allowing for simultaneous detection of and treatment of the disease. This review provides state of the art of the nanotechnological applications for cancer therapy. Furthermore, it advances a novel concept of personalized nanomedical theranostic therapy using iron oxide magnetic nanoparticles in conjunction with MRI imaging. Regulatory and industrial perspectives are also included to outline future perspectives in nanotechnological cancer research.


Assuntos
Portadores de Fármacos/administração & dosagem , Magnetismo , Nanopartículas/administração & dosagem , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Nanomedicina Teranóstica/métodos , Compostos Férricos/administração & dosagem , Imagem Óptica/métodos , Imagem Óptica/tendências , Medicina de Precisão/métodos , Medicina de Precisão/tendências
14.
Dalton Trans ; 44(32): 14323-32, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26197944

RESUMO

A first investigation into the application of a luminescent osmium(ii) bipyridine complex to live cell imaging is presented. Osmium(ii) (bis-2,2-bipyridyl)-2(4-carboxylphenyl) imidazo[4,5f][1,10]phenanthroline was prepared and conjugated to octaarginine, a cell penetrating peptide. The photophysics, cell uptake and cytotoxicity of this osmium complex conjugate were performed and compared with its ruthenium analogue. Cell uptake and distribution of both ruthenium and osmium conjugates were very similar with rapid transmembrane transport of the osmium probe (complete within approx. 20 min) and dispersion throughout the cytoplasm and organelles. The near-infrared (NIR) emission of the osmium complex (λmax 726 nm) coincides well with the biological optical window and this facilitated luminescent and luminescence lifetime imaging of the cell which was well resolved from cell autofluorescence. The large Stokes shift of the emission also permitted resonance Raman mapping of the dye within CHO cells. Rather surprisingly, the osmium conjugate exhibited very low cytotoxicity when incubated both in the dark and under visible irradiation. This was attributed to the remarkable stability of this complex which was reflected by the complete absence of photo-bleaching of the complex even under extended continuous irradiation. In addition, when compared to its ruthenium analogue its luminescence was short-lived in water therefore rendering it insensitive to O2.


Assuntos
2,2'-Dipiridil/química , Sobrevivência Celular/efeitos dos fármacos , Compostos Organometálicos/química , Compostos Organometálicos/farmacologia , Osmio/química , Peptídeos/química , Rutênio/química , Animais , Células CHO , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Diagnóstico por Imagem , Processamento de Imagem Assistida por Computador , Espectroscopia de Ressonância Magnética , Sondas Moleculares/química , Estrutura Molecular , Oligopeptídeos/química , Análise Espectral Raman
15.
Breast Cancer Res ; 17: 66, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25968050

RESUMO

INTRODUCTION: Tumor cells can effectively be killed by heat, e.g. by using magnetic hyperthermia. The main challenge in the field, however, is the generation of therapeutic temperatures selectively in the whole tumor region. We aimed to improve magnetic hyperthermia of breast cancer by using innovative nanoparticles which display a high heating potential and are functionalized with a cell internalization and a chemotherapeutic agent to increase cell death. METHODS: The superparamagnetic iron oxide nanoparticles (MF66) were electrostatically functionalized with either Nucant multivalent pseudopeptide (N6L; MF66-N6L), doxorubicin (DOX; MF66-DOX) or both (MF66-N6LDOX). Their cytotoxic potential was assessed in a breast adenocarcinoma cell line MDA-MB-231. Therapeutic efficacy was analyzed on subcutaneous MDA-MB-231 tumor bearing female athymic nude mice. RESULTS: All nanoparticle variants showed an excellent heating potential around 500 W/g Fe in the alternating magnetic field (AMF, conditions: H=15.4 kA/m, f=435 kHz). We could show a gradual inter- and intracellular release of the ligands, and nanoparticle uptake in cells was increased by the N6L functionalization. MF66-DOX and MF66-N6LDOX in combination with hyperthermia were more cytotoxic to breast cancer cells than the respective free ligands. We observed a substantial tumor growth inhibition (to 40% of the initial tumor volume, complete tumor regression in many cases) after intratumoral injection of the nanoparticles in vivo. The proliferative activity of the remaining tumor tissue was distinctly reduced. CONCLUSION: The therapeutic effects of breast cancer magnetic hyperthermia could be strongly enhanced by the combination of MF66 functionalized with N6L and DOX and magnetic hyperthermia. Our approach combines two ways of tumor cell killing (magnetic hyperthermia and chemotherapy) and represents a straightforward strategy for translation into the clinical practice when injecting nanoparticles intratumorally.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Compostos Férricos/química , Hipertermia Induzida/métodos , Nanopartículas Metálicas/administração & dosagem , Nanopartículas Metálicas/química , Animais , Apoptose , Neoplasias da Mama/diagnóstico , Linhagem Celular Tumoral , Modelos Animais de Doenças , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Feminino , Humanos , Hipertermia Induzida/efeitos adversos , Nanopartículas Metálicas/efeitos adversos , Camundongos , Camundongos Nus , Microtomografia por Raio-X , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Nanobiotechnology ; 13: 16, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25880445

RESUMO

BACKGROUND: Different superparamagnetic iron oxide nanoparticles have been tested for their potential use in cancer treatment, as they enter into cells with high effectiveness, do not induce cytotoxicity, and are retained for relatively long periods of time inside the cells. We have analyzed the interaction, internalization and biocompatibility of dimercaptosuccinic acid-coated superparamagnetic iron oxide nanoparticles with an average diameter of 15 nm and negative surface charge in MCF-7 breast cancer cells. RESULTS: Cells were incubated with dimercaptosuccinic acid-coated superparamagnetic iron oxide nanoparticles for different time intervals, ranging from 0.5 to 72 h. These nanoparticles showed efficient internalization and relatively slow clearance. Time-dependent uptake studies demonstrated the maximum accumulation of dimercaptosuccinic acid-coated superparamagnetic iron oxide nanoparticles after 24 h of incubation, and afterwards they were slowly removed from cells. Superparamagnetic iron oxide nanoparticles were internalized by energy dependent endocytosis and localized in endosomes. Transmission electron microscopy studies showed macropinocytosis uptake and clathrin-mediated internalization depending on the nanoparticles aggregate size. MCF-7 cells accumulated these nanoparticles without any significant effect on cell morphology, cytoskeleton organization, cell cycle distribution, reactive oxygen species generation and cell viability, showing a similar behavior to untreated control cells. CONCLUSIONS: All these findings indicate that dimercaptosuccinic acid-coated superparamagnetic iron oxide nanoparticles have excellent properties in terms of efficiency and biocompatibility for application to target breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Materiais Revestidos Biocompatíveis/metabolismo , Compostos Férricos/metabolismo , Nanopartículas de Magnetita/química , Succímero/metabolismo , Mama/citologia , Mama/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/química , Citoesqueleto/efeitos dos fármacos , Endocitose , Endossomos/metabolismo , Feminino , Compostos Férricos/química , Humanos , Pinocitose , Succímero/química
17.
Nanomedicine ; 11(4): 815-24, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25652898

RESUMO

Bismuth Ferrite (BFO) nanoparticles (BFO-NP) display interesting optical (nonlinear response) and magnetic properties which make them amenable for bio-oriented diagnostic applications as intra- and extra membrane contrast agents. Due to the relatively recent availability of this material in well dispersed nanometric form, its biocompatibility was not known to date. In this study, we present a thorough assessment of the effects of in vitro exposure of human adenocarcinoma (A549), lung squamous carcinoma (NCI-H520), and acute monocytic leukemia (THP-1) cell lines to uncoated and poly(ethylene glycol)-coated BFO-NP in the form of cytotoxicity, haemolytic response and biocompatibility. Our results support the attractiveness of the functional-BFO towards biomedical applications focused on advanced diagnostic imaging. FROM THE CLINICAL EDITOR: Bismuth Ferrite nanoparticles (BFO-NP) have been recently successfully introduced as photodynamic tools and imaging probes. However, how these nanoparticles interact with various cells at the cellular level remains poorly understood. In this study, the authors performed in vitro experiments to assess the effects of uncoated and PEG-coated BFO-NP in the form of cytotoxicity, haemolytic response and biocompatibility.


Assuntos
Bismuto/química , Materiais Revestidos Biocompatíveis/química , Meios de Contraste/química , Compostos Férricos/química , Teste de Materiais , Nanopartículas/química , Linhagem Celular Tumoral , Humanos
18.
ACS Nano ; 8(6): 5682-95, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24873349

RESUMO

Despite the widespread availability of immunohistochemical and other methodologies for screening and early detection of lung and breast cancer biomarkers, diagnosis of the early stage of cancers can be difficult and prone to error. The identification and validation of early biomarkers specific to lung and breast cancers, which would permit the development of more sensitive methods for detection of early disease onset, is urgently needed. In this paper, ultra-small and bright nanoprobes based on quantum dots (QDs) conjugated to single domain anti-HER2 (human epidermal growth factor receptor 2) antibodies (sdAbs) were applied for immunolabeling of breast and lung cancer cell lines, and their performance was compared to that of anti-HER2 monoclonal antibodies conjugated to conventional organic dyes Alexa Fluor 488 and Alexa Fluor 568. The sdAbs-QD conjugates achieved superior staining in a panel of lung cancer cell lines with differential HER2 expression. This shows their outstanding potential for the development of more sensitive assays for early detection of cancer biomarkers.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias Pulmonares/metabolismo , Pontos Quânticos , Receptor ErbB-2/metabolismo , Anticorpos/química , Anticorpos Monoclonais/química , Linhagem Celular Tumoral , Técnicas de Cocultura , Citometria de Fluxo , Corantes Fluorescentes/química , Humanos , Imuno-Histoquímica , Macrófagos/metabolismo , Microscopia Confocal
19.
Biomaterials ; 35(9): 2543-57, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24429386

RESUMO

Gold nanomaterials are currently raising a significant interest for human welfare in the field of clinical diagnosis, therapeutics for chronic pathologies, as well as of many other biomedical applications. In particular, gold nanomaterials are becoming a promising technology for developing novel approaches and treatments against widespread societal diseases such as cancer. In this study, we investigated the potential of proprietary gold nanoboxes (AuNBs) as carriers for their perspective translation into multifunctional, pre-clinical nano-enabled systems for personalized medicine approaches against lung cancer. A safe-by-design, tiered approach, with systematic tests conducted in the early phases on uncoated AuNBs and more focused testing on the coated, drug-loaded nanomaterial toward the end, was adopted. Our results showed that uncoated AuNBs could effectively penetrate into human lung adenocarcinoma (A549) cells when in simple (mono-cultures) or complex (co- and three-dimensional-cultures) in vitro microenvironments mimicking the alveolar region of human lungs. Uncoated AuNBs were biologically inert in A549 cells and demonstrated signs of biodegradability. Concurrently, preliminary data revealed that coated, drug-loaded AuNBs could efficiently deliver a chemotherapeutic agent to A549 cells, corroborating the hypothesis that AuNBs could be used in the future for the development of personalized nano-enabled systems for lung cancer treatment.


Assuntos
Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Endocitose , Ouro/química , Nanoestruturas/química , Neoplasias/metabolismo , Materiais Biocompatíveis/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Técnicas de Cocultura , Gelatina/química , Ouro/toxicidade , Humanos , Nanoestruturas/toxicidade , Nanoestruturas/ultraestrutura , Neoplasias/patologia , Neoplasias/ultraestrutura , Fatores de Tempo
20.
Sci Rep ; 3: 1124, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23350031

RESUMO

Single-walled carbon nanotubes (SWCNTs) have been widely explored as potential technologies for information systems and medical applications. The impact of SWCNTs on human health is of prime concern, if SWCNTs have a future in the manufacturing industry. This study proposes a novel, inflammation-independent paradigm of toxicity for SWCNTs, identifying the protein citrullination process as early-stage indicator of inflammatory responses of macrophages (THP-1) and of subtle phenotypic damages of lung epithelial (A549) cells following exposure to chemically-treated SWCNTs. Our results showed that, while most of the cellular responses of A549 cells exposed to SWCNTs are different to those of similarly treated THP-1 cells, the protein citrullination process is triggered in a dose- and time-dependent manner in both cell lines, with thresholds comparable between inflammatory (THP-1) and non-inflammatory (A549) cell types. The cellular mechanism proposed herein could have a high impact in predicting the current risk associated with environmental exposure to SWCNTs.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Citrulina/metabolismo , Nanotubos de Carbono/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Nanotubos de Carbono/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA