Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nanotoxicology ; : 1-20, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38847611

RESUMO

A novel brain-targeted and reactive oxygen species-activatable manganese dioxide containing nanoparticle system functionalized with anti-amyloid-ß antibody (named aAß-BTRA-NC) developed by our group has shown great promise as a highly selective magnetic resonance imaging (MRI) contrast agent for early detection and multitargeted disease-modifying treatment of Alzheimer's disease (AD). To further evaluate the suitability of the formulation for future clinical application, we investigated the safety, biodistribution, and pharmacokinetic profile of aAß-BTRA-NC in a transgenic TgCRND8 mouse AD model, wild type (WT) littermate, and CD-1 mice. Dose-ascending studies demonstrated that aAß-BTRA-NC was well-tolerated by the animals up to 300 µmol Mn/kg body weight [b.w.], 3 times the efficacious dose for early AD detection without apparent adverse effects; Histopathological, hematological, and biochemical analyses indicated that a single dose of aAß-BTRA-NC did not cause any toxicity in major organs. Immunotoxicity data showed that aAß-BTRA-NC was safer than commercially available gadolinium-based MRI contrast agents at an equivalent dose of 100 µmol/kg b.w. of metal ions. Intravenously administered aAß-BTRA-NC was taken up by main organs with the order of liver, kidneys, intestines, spleen, followed by other organs, and cleared after one day to one week post injection. Pharmacokinetic analysis indicated that the plasma concentration profile of aAß-BTRA-NC followed a 2-compartmental model with faster clearance in the AD mice than in the WT mice. The results suggest that aAß-BTRA-NC exhibits a strong safety profile as a nanotheranostic agent which warrants more robust preclinical development for future clinical applications.

2.
Mater Today Bio ; 25: 100954, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38304342

RESUMO

Early and precise detection of solid tumor cancers is critical for improving therapeutic outcomes. In this regard, magnetic resonance imaging (MRI) has become a useful tool for tumor diagnosis and image-guided therapy. However, its effectiveness is limited by the shortcomings of clinically available gadolinium-based contrast agents (GBCAs), i.e. poor tumor penetration and retention, and safety concerns. Thus, we have developed a novel nanoparticulate contrast agent using a biocompatible terpolymer and lipids to encapsulate manganese dioxide nanoparticles (TPL-MDNP). The TPL-MDNP accumulated in tumor tissue and produced paramagnetic Mn2+ ions, enhancing T1-weight MRI contrast via the reaction with H2O2 rich in the acidic tumor microenvironment. Compared to the clinically used GBCA, Gadovist®1.0, TPL-MDNP generated stronger T1-weighted MR signals by over 2.0-fold at 30 % less of the recommended clinical dose with well-defined tumor delineation in preclinical orthotopic tumor models of brain, breast, prostate, and pancreas. Importantly, the MRI signals were retained for 60 min by TPL-MDNP, much longer than Gadovist®1.0. Biocompatibility of TPL-MDNP was evaluated and found to be safe up to 4-fold of the dose used for MRI. A robust large-scale manufacturing process was developed with batch-to-batch consistency. A lyophilization formulation was designed to maintain the nanostructure and storage stability of the new contrast agent.

3.
Research (Wash D C) ; 6: 0247, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37795337

RESUMO

Despite substantial progress in the treatment of castration-resistant prostate cancer (CRPC), including radiation therapy and immunotherapy alone or in combination, the response to treatment remains poor due to the hypoxic and immunosuppressive nature of the tumor microenvironment. Herein, we exploited the bioreactivity of novel polymer-lipid manganese dioxide nanoparticles (PLMDs) to remodel the tumor immune microenvironment (TIME) by increasing the local oxygen levels and extracellular pH and enhancing radiation-induced immunogenic cell death. This study demonstrated that PLMD treatment sensitized hypoxic human and murine CRPC cells to radiation, significantly increasing radiation-induced DNA double-strand breaks and ultimately cell death, which enhanced the secretion of damage-associated molecular patterns, attributable to the induction of autophagy and endoplasmic reticulum stress. Reoxygenation via PLMDs also polarized hypoxic murine RAW264.7 macrophages toward the M1 phenotype, enhancing tumor necrosis factor alpha release, and thus reducing the viability of murine CRPC TRAMP-C2 cells. In a syngeneic TRAMP-C2 tumor model, intravenous injection of PLMDs suppressed, while radiation alone enhanced recruitment of regulatory T cells and myeloid-derived suppressor cells. Pretreatment with PLMDs followed by radiation down-regulated programmed death-ligand 1 and promoted the infiltration of antitumor CD8+ T cells and M1 macrophages to tumor sites. Taken together, TIME modulation by PLMDs plus radiation profoundly delayed tumor growth and prolonged median survival compared with radiation alone. These results suggest that PLMDs plus radiation is a promising treatment modality for improving therapeutic efficacy in radioresistant and immunosuppressive solid tumors.

4.
Adv Sci (Weinh) ; 10(12): e2207238, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36808713

RESUMO

Finding effective disease-modifying treatment for Alzheimer's disease remains challenging due to an array of factors contributing to the loss of neural function. The current study demonstrates a new strategy, using multitargeted bioactive nanoparticles to modify the brain microenvironment to achieve therapeutic benefits in a well-characterized mouse model of Alzheimer's disease. The application of brain-penetrating manganese dioxide nanoparticles significantly reduces hypoxia, neuroinflammation, and oxidative stress; ultimately reducing levels of amyloid ß plaques within the neocortex. Analyses of molecular biomarkers and magnetic resonance imaging-based functional studies indicate that these effects improve microvessel integrity, cerebral blood flow, and cerebral lymphatic clearance of amyloid ß. These changes collectively shift the brain microenvironment toward conditions more favorable to continued neural function as demonstrated by improved cognitive function following treatment. Such multimodal disease-modifying treatment may bridge critical gaps in the therapeutic treatment of neurodegenerative disease.


Assuntos
Doença de Alzheimer , Encéfalo , Nanopartículas Metálicas , Animais , Camundongos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Hipóxia Celular , Sistemas de Liberação de Medicamentos , Lipídeos/química , Nanopartículas Metálicas/química , Estresse Oxidativo , Polímeros/química , Encéfalo/metabolismo
5.
Radiother Oncol ; 170: 213-223, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35227733

RESUMO

INTRODUCTION: Radiation therapy (RT) is a major modality for the treatment of prostate cancer (PCa), especially castration-resistant PCa (CRPC). However, hypoxia, often seen in PCa tumors, leads to radiation-resistance. This work investigates the effect of a novel oxygen-generating polymer-lipid manganese dioxide nanoparticle (PLMDs) on improving RT outcomes in CRPC xenograft models by modulating the tumor microenvironment (TME) both before and after RT. MATERIALS AND METHODS: Human PC3 and DU145 PCa cells were used to investigate clonogenic inhibition and DNA repair pathways in vitro. Tumor hypoxia and post-RT angiogenesis were evaluated in a PC3-bearing SCID mouse model. PC3 and DU145 xenografts were used to study the efficacy of PLMD in combination with single or fractionated RT. RESULTS: PLMD plus RT significantly inhibited clonogenic potential, increased DNA double-strand breaks, and reduced DNA damage repair in hypoxic PC3 and DU145 cells as compared to RT alone. PLMD significantly reduced hypoxia-positive areas, hypoxia induced factor 1α (HIF-1α) expression, and protein carbonyl levels (a measure of oxidative stress). Application of PLMD with RT decreased RT-induced angiogenic biomarkers by up to 3-fold. Treatment of the human CRPC xenografts with PLMD plus RT (single or fractionated doses) significantly prolonged median survival of the host compared to RT alone resulting in up to a 40% curative rate. CONCLUSION: PLMD treatment modulated TME and sensitized hypoxic human CRPC cells to RT thus enhancing the efficacy of RT. These results confirmed the potential of PLMD as an adjuvant to RT for the treatment of hypoxic CRPC.


Assuntos
Nanopartículas , Neoplasias de Próstata Resistentes à Castração , Animais , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Hipóxia , Masculino , Camundongos , Camundongos SCID , Oxirredução , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/radioterapia , Microambiente Tumoral
6.
Pharm Res ; 38(11): 1897-1914, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34655006

RESUMO

PURPOSE: Chemotherapy for glioblastoma multiforme (GBM) remains ineffective due to insufficient penetration of therapeutic agents across the blood-brain barrier (BBB) and into the GBM tumor. Herein, is described, the optimization of the lipid composition and fabrication conditions for a BBB- and tumor penetrating terpolymer-lipid-hybrid nanoparticle (TPLN) for delivering doxorubicin (DOX) to GBM. METHODS: The composition of TPLNs was first screened using different lipids based on nanoparticle properties and in vitro cytotoxicity by using 23 full factorial experimental design. The leading DOX loaded TPLNs (DOX-TPLN) were prepared by further optimization of conditions and used to study cellular uptake mechanisms, in vitro cytotoxicity, three-dimensional (3D) glioma spheroid penetration, and in vivo biodistribution in a murine orthotopic GBM model. RESULTS: Among various lipids studied, ethyl arachidate (EA) was found to provide excellent nanoparticle properties e.g., size, polydispersity index (PDI), zeta potential, encapsulation efficiency, drug loading, and colloidal stability, and highest anticancer efficacy for DOX-TPLN. Further optimized EA-based TPLNs were prepared with an optimal particle size (103.8 ± 33.4 nm) and PDI (0.208 ± 0.02). The resultant DOX-TPLNs showed ~ sevenfold higher efficacy than free DOX against human GBM U87-MG-RED-FLuc cells in vitro. The interaction between the TPLNs and the low-density lipoprotein receptors also facilitated cellular uptake, deep penetration into 3D glioma spheroids, and accumulation into the in vivo brain tumor regions of DOX-TPLNs. CONCLUSION: This work demonstrated that the TPLN system can be optimized by rational selection of lipid type, lipid content, and preparation conditions to obtain DOX-TPLN with enhanced anticancer efficacy and GBM penetration and accumulation.


Assuntos
Antineoplásicos/administração & dosagem , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Sistemas de Liberação de Fármacos por Nanopartículas/química , Animais , Antineoplásicos/farmacocinética , Barreira Hematoencefálica , Neoplasias Encefálicas , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Glioblastoma/patologia , Humanos , Lipossomos/química , Camundongos , Nanopartículas/química , Tamanho da Partícula , Polímeros/química , Esferoides Celulares , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Expert Opin Drug Deliv ; 18(7): 991-1004, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33703991

RESUMO

BACKGROUND: Successful delivery of anticancer drugs to intracellular targets requires different properties of the nanocarrier to overcome multiple transport barriers. However, few nanocarrier systems, to date, possess such properties, despite knowledge about the biological fate of inorganic and polymeric nanocarriers in relation to their fixed size, shape and surface properties. Herein, a polymer-lipid hybrid nanoparticle (PLN) system is described with size and shape transformability and its mechanisms of cellular uptake and intracellular trafficking are studied. METHODS: Pharmaceutical lipids were screened for use in transformable PLN. Mechanisms of cellular uptake and the role of fatty acid-binding proteins in intracellular trafficking of PLN were investigated in breast cancer cells. Intra-tumoral penetration and retention of doxorubicin (DOX) were evaluated by confocal microscopy. RESULTS: The lead PLNs showed time-dependent size reduction and shape change from spherical to spiky shape. This transformability of PLNs and lipid trafficking pathways facilitated intracellular transport of DOX-loaded PLN (DOX-PLN) into mitochondria and nuclei. DOX-PLN significantly increased DOX penetration and retention over free DOX or non-transformable liposomal DOX particles at 4 h post-intravenous administration. CONCLUSION: Transformability of PLN and lipid-biology interplay can be exploited to design new nanocarriers for effective drug delivery to tumor cells and intracellular targets.


Assuntos
Antineoplásicos , Nanopartículas , Nanoestruturas , Antineoplásicos/farmacologia , Biologia , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Portadores de Fármacos , Humanos , Lipídeos , Polímeros
8.
J Natl Cancer Inst ; 111(4): 399-408, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30239773

RESUMO

BACKGROUND: Tumor microenvironment (TME) and associated multiple factors are found to contribute to the failures in cancer therapies, including chemo- and immunotherapy. Here we report a new multimodal strategy that uses a bioreactive multifunctional hybrid polymer-lipid encapsulated manganese dioxide nanoparticle (PLMD NP) system to remodel the TME, suppress drug resistance factors, reverse immunosuppressive conditions, and enhance chemotherapy efficacy. METHODS: The influence of PLMD NPs on enhancing cellular uptake in EMT6 mouse breast cancer cells and tumor penetration of doxorubicin (DOX) in EMT6 orthotopic breast tumor mouse model was evaluated using confocal microscopy (n = 3-4). Immunohistochemistry was employed to examine the effect of PLMD NPs on downregulating hypoxia-induced drug resistance proteins and anticancer activity of DOX (n = 3-4). The efficacy of the combination therapy with PLMD NPS and DOX was assessed in murine EMT6 (n = 15-23) and 4T1 (n = 7) orthotopic breast tumor mouse models. Rechallenge and splenocyte transfer were performed to validate the stimulation of adaptive tumor immunity in the surviving mice. RESULTS: PLMD NPs enhanced intratumoral penetration and efficacy of DOX, and reduced intratumoral expression of P-glycoprotein, p53, and carbonic anhydrase IX by 74.5%, 38.0%, and 58.8% vs saline control, respectively. Combination treatment with PLMD NPs and DOX increased the number of tumor-infiltrated CD8+ T cells and resulted in up to 60.0% complete tumor regression. Of naïve mice (n = 7) that received splenocytes from the PLMD+DOX-treated surviving mice, 57.1% completely suppressed tumor growth whereas 100% of mice that received splenocytes from DOX-treated mice (n = 3) and the control group (n = 7) showed rapid tumor growth. CONCLUSIONS: The clinically suitable PLMD NPs can effectively downregulate TME-associated drug resistance and immunosuppression. The combination therapy with PLMD NPs and DOX is a multimodal and translational treatment approach for enhancing chemotherapeutic efficacy and boosting antitumor immunity.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/imunologia , Doxorrubicina/farmacologia , Compostos de Manganês/química , Nanopartículas/administração & dosagem , Óxidos/química , Polímeros/química , Microambiente Tumoral/imunologia , Animais , Apoptose , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proliferação de Células , Quimioterapia Combinada , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Nanoscale ; 9(30): 10701-10714, 2017 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-28678269

RESUMO

A unique class of diruthenium(ii,iii) metallodrugs containing non-steroidal anti-inflammatory drug (NSAID), Ru2(NSAID), have been reported to show anticancer activity in glioma models in vitro and in vivo. This work reports the encapsulation of the lead metallodrug of ibuprofen (HIbp), [Ru2(Ibp)4Cl] or RuIbp, and also of the new analogue of naproxen (HNpx), [Ru2(Npx)4Cl] or RuNpx, in novel intravenously (i.v.) injectable solid polymer-lipid nanoparticles (SPLNs). A rationally selected composition of lipids/polymers rendered nearly spherical Ru2(NSAID)-SPLNs with a mean size of 120 nm and zeta potential of about -20 mV. The Ru2(NSAID)-SPLNs are characterized by spectroscopic techniques and the composition in terms of ruthenium-drug species is analyzed by mass spectrometry. The metallodrug-loaded nanoparticles showed high drug loading (17-18%) with ∼100% drug loading efficiency, and good colloidal stability in serum at body temperature. Fluorescence-labeled SPLNs were taken up by the cancer cells in a time- and energy-dependent manner as analyzed by confocal microscopy and fluorescence spectrometry. The Ru2(NSAID)-SPLNs showed enhanced cytotoxicity (IC50 at 60-100 µmol L-1 ) in relation to the corresponding Ru2(NSAID) metallodrugs in breast (EMT6 and MDA-MB-231) and prostate (DU145) cancer cells in vitro. The cell viability of both metallodrug nanoformulations is also compared with those of the parent NSAIDs, HIbp and HNpx, and their corresponding NSAID-SPLNs. In vivo and ex vivo fluorescence imaging revealed good biodistribution and high tumor accumulation of fluorescence-labeled SPLNs following i.v. injection in an orthotopic breast tumor model. The enhanced anticancer activity of the metallodrug-loaded SPLNs in these cell lines can be associated with the advantages of the nanoformulations, assigned mainly to the stability of the colloidal nanoparticles suitable for i.v. injection and enhanced cellular uptake. The findings of this work encourage future in vivo efficacy studies to further exploit the potential of the novel Ru2(NSAID)-SPLN nanoformulations for clinical application.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Portadores de Fármacos , Ibuprofeno/administração & dosagem , Lipídeos , Nanopartículas , Naproxeno/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Humanos , Ibuprofeno/farmacologia , Masculino , Naproxeno/farmacologia , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/farmacologia , Polímeros , Rutênio , Distribuição Tecidual
10.
Nanoscale ; 9(4): 1334-1355, 2017 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-27973629

RESUMO

Polymer-lipid hybrid nanoparticles (PLN) are an emerging nanocarrier platform made from building blocks of polymers and lipids. PLN integrate the advantages of biomimetic lipid-based nanoparticles (i.e. solid lipid nanoparticles and liposomes) and biocompatible polymeric nanoparticles. PLN are constructed from diverse polymers and lipids and their numerous combinations, which imparts PLN with great versatility for delivering drugs of various properties to their nanoscale targets. PLN can be classified into two types based on their hybrid nanoscopic structure and assembly methods: Type-I monolithic matrix and Type-II core-shell systems. This article reviews the history of PLN development, types of PLN, lipid and polymer candidates, fabrication methods, and unique properties of PLN. The applications of PLN in delivery of therapeutic or imaging agents alone or in combination for cancer treatment are summarized and illustrated with examples. Important considerations for the rational design of PLN for advanced nanoscale drug delivery are discussed, including selection of excipients, synthesis processes governing formulation parameters, optimization of nanoparticle properties, improvement of particle surface functionality to overcome macroscopic, microscopic and cellular biological barriers. Future directions and potential clinical translation of PLN are also suggested.


Assuntos
Portadores de Fármacos/química , Lipídeos/química , Nanopartículas , Neoplasias/tratamento farmacológico , Polímeros/química , Humanos
11.
J Control Release ; 246: 98-109, 2017 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-28017889

RESUMO

Brain metastasis is a fatal disease with limited treatment options and very short survival. Although systemic chemotherapy has some effect on peripheral metastases of breast cancer, it is ineffective in treating brain metastasis due largely to the blood-brain barrier (BBB). Here we developed a BBB-penetrating amphiphilic polymer-lipid nanoparticle (NP) system that efficiently delivered anti-mitotic drug docetaxel (DTX) for the treatment of brain metastasis of triple negative breast cancer (TNBC). We evaluated the biodistribution, brain accumulation, pharmacokinetics and efficacy of DTX-NP in a mouse model of brain metastasis of TNBC. Confocal fluorescence microscopy revealed extravasation of dye-loaded NPs from intact brain microvessels in healthy mice. DTX-NP also extravasated from brain microvessels and accumulated in micrometastasis lesions in the brain. Intravenously injected DTX-NPs increased the blood circulation time of DTX by 5.5-fold and the AUC0-24h in tumor-bearing brain by 5-fold compared to the clinically used DTX formulation Taxotere®. The kinetics of NPs in the brain, determined by ex vivo fluorescence imaging, showed synchronization with DTX kinetics in the brain measured by LC-MS/MS. This result confirmed successful delivery of DTX by the NPs into the brain and suggested that ex vivo fluorescence imaging of NP could be an effective and quick means for probing drug disposition in the brain. Treatment with the DTX-NP formulation delayed tumor growth by 11-fold and prolonged median survival of tumor-bearing mice by 94% compared to an equivalent dose of Taxotere®, without inducing histological changes in the major organs.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário , Portadores de Fármacos/metabolismo , Nanopartículas/metabolismo , Tensoativos/metabolismo , Taxoides/administração & dosagem , Neoplasias de Mama Triplo Negativas/patologia , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Docetaxel , Portadores de Fármacos/química , Feminino , Humanos , Camundongos SCID , Nanopartículas/química , Polímeros/metabolismo , Tensoativos/química , Taxoides/farmacocinética , Taxoides/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
12.
Cancer Res ; 76(22): 6643-6656, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27758881

RESUMO

Hypoxia in the tumor microenvironment (TME) mediates resistance to radiotherapy and contributes to poor prognosis in patients receiving radiotherapy. Here we report the design of clinically suitable formulations of hybrid manganese dioxide (MnO2) nanoparticles (MDNP) using biocompatible materials to reoxygenate the TME by reacting with endogenous H2O2 MDNP containing hydrophilic terpolymer-protein-MnO2 or hydrophobic polymer-lipid-MnO2 provided different oxygen generation rates in the TME relevant to different clinical settings. In highly hypoxic murine or human xenograft breast tumor models, we found that administering either MDNP formulation before radiotherapy modulated tumor hypoxia and increased radiotherapy efficacy, acting to reduce tumor growth, VEGF expression, and vascular density. MDNP treatment also increased apoptosis and DNA double strand breaks, increasing median host survival 3- to 5-fold. Notably, in the murine model, approximately 40% of tumor-bearing mice were tumor-free after a single treatment with MDNPs plus radiotherapy at a 2.5-fold lower dose than required to achieve the same curative treatment without MDNPs. Overall, our findings offer a preclinical proof of concept for the use of MDNP formulations as effective radiotherapy adjuvants. Cancer Res; 76(22); 6643-56. ©2016 AACR.


Assuntos
Neoplasias da Mama/radioterapia , Compostos de Manganês/metabolismo , Óxidos/metabolismo , Radioterapia/métodos , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Nanopartículas , Hipóxia Tumoral , Microambiente Tumoral
13.
J Control Release ; 209: 186-96, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25908171

RESUMO

Multifunctional nanoparticles (NPs) have found important applications in diagnosis, chemotherapy, and image-guided surgery of tumors. In this work, we have developed polymeric theranostic NPs (PTNPs) containing the anticancer drug docetaxel (DTX), a fluorescent dye, and magnetic manganese oxide (MnO) NPs for dual modal imaging and chemotherapy. PTNPs ~150 nm in diameter were synthesized by co-loading hydrophobic DTX and MnO NPs ~5 nm in diameter, into the matrix of a fluorescent dye-labeled amphiphilic polymer. The PTNPs enabled high loading efficiency and sustained in vitro release of DTX. Energy-dependent cellular uptake and extended cytoplasmic retention of the PTNPs in MDA-MB-231 human breast cancer cells were observed by fluorescence microscopy examination. DTX-loaded PTNPs exhibited higher cytotoxicity than free DTX with a 3 to 4.4-fold decrease in drug dose required for 50% cell growth inhibition. The hydrophilic backbone of the amphiphilic polymer improved the fluidity of PTNPs which enhanced the longitudinal relaxivity (r1) of loaded MnO NPs by 2.7-fold with r1=2.4mM(-1)s(-1). Whole body fluorescence imaging (FI) and magnetic resonance imaging (MRI) showed significant accumulation and prolonged retention of PTNPs in orthotopic MDA-MB-231 breast tumors. These results suggest that the new amphiphilic polymer-based PTNP system, able to simultaneously deliver a poorly soluble anticancer drug, enhance MRI contrast, and stain tumor tissue by fluorescence, is a good candidate for cancer theranostic applications.


Assuntos
Antineoplásicos , Fluoresceínas , Corantes Fluorescentes , Compostos de Manganês , Nanopartículas , Óxidos , Taxoides , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Docetaxel , Liberação Controlada de Fármacos , Feminino , Fluoresceínas/administração & dosagem , Fluoresceínas/química , Fluoresceínas/farmacocinética , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacocinética , Humanos , Fenômenos Magnéticos , Imageamento por Ressonância Magnética , Compostos de Manganês/administração & dosagem , Compostos de Manganês/química , Compostos de Manganês/farmacocinética , Camundongos SCID , Microscopia de Fluorescência , Nanopartículas/administração & dosagem , Nanopartículas/química , Nanopartículas/uso terapêutico , Óxidos/administração & dosagem , Óxidos/química , Óxidos/farmacocinética , Tamanho da Partícula , Polímeros/química , Taxoides/administração & dosagem , Taxoides/química , Taxoides/farmacocinética , Taxoides/uso terapêutico , Distribuição Tecidual , Carga Tumoral/efeitos dos fármacos
14.
Biosens Bioelectron ; 69: 40-5, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25703727

RESUMO

This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief and the Corresponding Author, Muhammad Kamran. This paper has been withdrawn as the authors did not fully consult with their project collaborators prior to publication and failed to include them as co-authors of the article. This is acknowledged by the corresponding author. The authors and the Publisher would like to apologise for any inconvenience caused.

15.
Diabetes Technol Ther ; 17(4): 255-67, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25671341

RESUMO

An implantable, glucose-responsive insulin delivery microdevice was reported previously by our group, providing rapid insulin release in response to hyperglycemic events and efficacy in vivo over a 1-week period when implanted intraperitoneally in rats with diabetes. Herein, we focused on the improvement of the microdevice prototype for long-term glycemic control by subcutaneous (SC) implantation, which allows for easy retrieval and replacement as needed. To surmount the strong immune response to the SC implant system, the microdevice was treated by surface modification with high-molecular-weight polyethylene glycol (PEG). In vitro glucose-responsive insulin release, in vivo efficacy, and biocompatibility of the microdevice were studied. Modification with 20-kDa PEG chains greatly reduced the immune response without a significant change in glucose-responsive insulin release in vitro. The fibrous capsule thickness was reduced from approximately 1,000 µm for the untreated devices to 30-300 µm for 2-kDa PEG-treated and to 30-50 µm for 20-kDa PEG-treated devices after 30 days of implantation. The integrity of the glucose-responsive bioinorganic membrane and the resistance to acute and chronic immune response were improved with the long-chain 20-kDa PEG brush layer. The 20-kDa PEG-treated microdevice provided long-term maintenance of euglycemia in a rat model of diabetes for up to 18 days. Moreover, a consistent rapid response to short-term glucose challenge was demonstrated in multiple-day tests for the first time on rats with diabetes in which the devices were implanted. The improvement of the microdevice is a promising step toward a long-acting insulin implant system for a true, closed-loop treatment of diabetes.


Assuntos
Glicemia/análise , Diabetes Mellitus Experimental/tratamento farmacológico , Hipoglicemiantes/administração & dosagem , Bombas de Infusão Implantáveis , Insulina de Ação Prolongada/administração & dosagem , Animais , Materiais Biocompatíveis , Glicemia/efeitos dos fármacos , Desenho de Equipamento , Infusões Subcutâneas , Masculino , Polietilenoglicóis/uso terapêutico , Ratos , Ratos Sprague-Dawley , Tensoativos/uso terapêutico , Resultado do Tratamento
16.
Biomaterials ; 47: 51-61, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25682160

RESUMO

Implantation of a medical implant within the body inevitably triggers a host inflammatory response that negatively impacts its function and longevity. Nevertheless, the degree and severity of this response may be reduced by selecting appropriate materials, implant geometry, surface topography and surface treatment. Here we demonstrate a strategy to improve the biocompatibility of a chemically-driven closed-loop insulin delivery implant. A microfabricated microporous, poly(ethylene glycol)-grafted polydimethylsiloxane membrane was placed on top of the glucose-responsive insulin release plug of the implant. Implant biocompatibility was assessed in healthy rats while implant function was evaluated in a type 1 diabetic rat model. The microporous membrane with a small distance to the plug provided a geometric barrier to inflammatory cell migration and prevented leukocyte-mediated degradation of the plug for at least 30 days. Membrane-protected devices elicited a significantly milder inflammatory response and formation of a well-defined fibrous capsule at the device opening compared to unprotected devices. The device's glucose-responsiveness was nearly unchanged, although the insulin release rate decreased with decreasing pore size. The microporous membrane improved biocompatibility and prolonged in vivo efficacy of the implant by ∼3-fold. This work suggests the importance of implant design in modulating inflammatory response and thereby extending the functional duration of the implant.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Sistemas de Infusão de Insulina , Insulina/administração & dosagem , Microtecnologia/métodos , Animais , Materiais Biocompatíveis/química , Glicemia/análise , Catalase/química , Modelos Animais de Doenças , Desenho de Fármacos , Fibrose , Glucose/química , Inflamação/metabolismo , Masculino , Polímeros/química , Ratos , Ratos Sprague-Dawley , Estreptozocina/química
17.
ACS Nano ; 8(4): 3202-12, 2014 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24702320

RESUMO

Insufficient oxygenation (hypoxia), acidic pH (acidosis), and elevated levels of reactive oxygen species (ROS), such as H2O2, are characteristic abnormalities of the tumor microenvironment (TME). These abnormalities promote tumor aggressiveness, metastasis, and resistance to therapies. To date, there is no treatment available for comprehensive modulation of the TME. Approaches so far have been limited to regulating hypoxia, acidosis, or ROS individually, without accounting for their interdependent effects on tumor progression and response to treatments. Hence we have engineered multifunctional and colloidally stable bioinorganic nanoparticles composed of polyelectrolyte-albumin complex and MnO2 nanoparticles (A-MnO2 NPs) and utilized the reactivity of MnO2 toward peroxides for regulation of the TME with simultaneous oxygen generation and pH increase. In vitro studies showed that these NPs can generate oxygen by reacting with H2O2 produced by cancer cells under hypoxic conditions. A-MnO2 NPs simultaneously increased tumor oxygenation by 45% while increasing tumor pH from pH 6.7 to pH 7.2 by reacting with endogenous H2O2 produced within the tumor in a murine breast tumor model. Intratumoral treatment with NPs also led to the downregulation of two major regulators in tumor progression and aggressiveness, that is, hypoxia-inducible factor-1 alpha and vascular endothelial growth factor in the tumor. Combination treatment of the tumors with NPs and ionizing radiation significantly inhibited breast tumor growth, increased DNA double strand breaks and cancer cell death as compared to radiation therapy alone. These results suggest great potential of A-MnO2 NPs for modulation of the TME and enhancement of radiation response in the treatment of cancer.


Assuntos
Acidose/tratamento farmacológico , Compostos de Manganês/farmacologia , Nanopartículas/química , Óxidos/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Soroalbumina Bovina/química , Microambiente Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Transporte Biológico , Bovinos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Estabilidade de Medicamentos , Humanos , Peróxido de Hidrogênio/metabolismo , Concentração de Íons de Hidrogênio , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Compostos de Manganês/química , Compostos de Manganês/metabolismo , Compostos de Manganês/uso terapêutico , Camundongos , Modelos Moleculares , Óxidos/química , Óxidos/metabolismo , Óxidos/uso terapêutico , Oxigênio/metabolismo , Conformação Proteica
18.
ACS Nano ; 5(12): 9668-74, 2011 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-22053744

RESUMO

Multiplexed detection of analytes is a challenge for numerous medical and biochemical applications. Many fluorescent particulate devices are being developed as ratiometric optical sensors to measure the concentration of intracellular analytes. The response of these sensors is based on changes of the emission intensity of analyte-sensitive probes, entrapped into the carrier system, which depends on the concentration of a specific analyte. However, there are a series of technical limits that prevent their use for quantitative detection of several analytes in parallel (e.g., emission crosstalk between different sensor molecules). Here we demonstrate that double-wall barcoded sensor capsules can be used for multiplexed analysis of proton, sodium, and potassium ions. The sensor detection methodology is based on porous microcapsules which carry ion-sensitive probes in their inner cavity for ion detection and a unique QD barcode in their outermost wall as tag for identification of individual sensors. The engineering of QD barcodes to capsules walls represents a promising strategy for optical multianalyte determination.


Assuntos
Eletrólitos/química , Processamento Eletrônico de Dados , Íons/análise , Técnicas de Sonda Molecular , Nanocápsulas/química , Coloração e Rotulagem/métodos
19.
Macromol Rapid Commun ; 32(2): 186-90, 2011 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-21433138

RESUMO

Here we report for the first time the design and expression of highly charged, unfolded protein polymers based on elastin-like peptides (ELPs). Positively and negatively charged variants were achieved by introducing lysine and glutamic acid residues, respectively, within the repetitive pentapeptide units. Subsequently it was demonstrated that the monodisperse protein polyelectrolytes with precisely defined amino acid compositions, sequences, and stereochemistries can be transferred into superstructures exploiting their electrostatic interactions. Hollow capsules were assembled from oppositely charged protein chains by using the layer-by-layer technique. The structures of the capsules were analyzed by various microscopy techniques revealing the fabrication of multilayer containers. Due to their low toxicity in comparison to other polyelectrolytes, supercharged ELPs are appealing candidates for the construction of electrostatically induced scaffolds in biomedicine.


Assuntos
Portadores de Fármacos/química , Oligopeptídeos/química , Animais , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/farmacologia , Elastina/química , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Ácido Glutâmico/química , Ácido Glutâmico/metabolismo , Lisina/química , Lisina/metabolismo , Camundongos , Estrutura Molecular , Células NIH 3T3 , Oligopeptídeos/biossíntese , Oligopeptídeos/farmacologia , Desdobramento de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Eletricidade Estática , Estereoisomerismo
20.
Small ; 7(3): 351-63, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-21294264

RESUMO

Micrometer-sized polyelectrolyte capsules are synthesized, which have ion-sensitive fluorophores embedded in their cavities. As the membranes of the capsules are permeable to ions, the fluorescence of the capsules changed with the ion concentration. In particular, capsules sensitive to protons, sodium, potassium, and chloride ions are fabricated and their fluorescence response analyzed. In order to allow for ratiometric measurements, additional fluorophores whose emission do not depend on the ion concentration and which emit a different wavelength are co-embedded in the capsule cavities.


Assuntos
Cápsulas/química , Cápsulas/síntese química , Nanotecnologia/métodos , Polímeros/síntese química , Modelos Teóricos , Polímeros/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...