Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
ACS Nano ; 14(1): 651-663, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31851488

RESUMO

The overexpression of immunomarker programmed cell death protein 1 (PD-1) and engagement of PD-1 to its ligand, PD-L1, are involved in the functional impairment of cluster of differentiation 8+ (CD8+) T cells, contributing to cancer progression. However, heterogeneities in PD-L1 expression and variabilities in biopsy-based assays render current approaches inaccurate in predicting PD-L1 status. Therefore, PD-L1 screening alone is not predictive of patient response to treatment, which motivates us to simultaneously detect multiple immunomarkers engaged in immune modulation. Here, we have developed multimodal probes, immunoactive gold nanostars (IGNs), that accurately detect PD-L1+ tumor cells and CD8+ T cells simultaneously in vivo, surpassing the limitations of current immunoimaging techniques. IGNs integrate the whole-body imaging of positron emission tomography with high sensitivity and multiplexing of Raman spectroscopy, enabling the dynamic tracking of both immunomarkers. IGNs also monitor response to immunotherapies in mice treated with combinatorial PD-L1 and CD137 agonists and distinguish responders from those nonresponsive to treatment. Our results showed a multifunctional nanoscale probe with capabilities that cannot be achieved with either modality alone, allowing multiplexed immunologic tumor profiling critical for predicting early response to immunotherapies.


Assuntos
Biomarcadores Tumorais/análise , Ouro/química , Imunoterapia , Melanoma/diagnóstico por imagem , Melanoma/terapia , Nanopartículas Metálicas/química , Imagem Óptica , Animais , Antígeno B7-H1/agonistas , Antígeno B7-H1/análise , Antígeno B7-H1/genética , Biomarcadores Tumorais/agonistas , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Camundongos , Tamanho da Partícula , Propriedades de Superfície , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/agonistas , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/análise , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética
3.
Nanoscale ; 10(27): 13092-13105, 2018 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-29961778

RESUMO

In this work, we demonstrate the targeted diagnosis of immunomarker programmed death ligand 1 (PD-L1) and simultaneous detection of epidermal growth factor receptor (EGFR) in breast cancer tumors in vivo using gold nanostars (AuNS) with multiplexed surface enhanced Raman spectroscopy (SERS). Real-time longitudinal tracking with SERS demonstrated maximum accumulation of AuNS occurred 6 h post intravenous (IV) delivery, enabling detection of both biomarkers simultaneously. Raman signal correlating to both PD-L1 and EGFR decreased by ∼30% in control tumors where receptors were pre-blocked prior to AuNS delivery, indicating both the sensitivity and specificity of SERS in distinguishing tumors with different levels of PD-L1 and EGFR expression. Our in vivo study was combined with the first demonstration of ex vivo SERS spatial maps of whole tumor lesions that provided both a qualitative and quantitative assessment of biomarker status with near cellular-level resolution. High resolution SERS maps also provided an overview of AuNS distribution in tumors which correlated well with the vascular density. Mass spectrometry showed AuNS accumulation in tumor and liver, and clearance via spleen, and electron microscopy revealed AuNS were endocytosed in tumors, Kupffer cells in the liver, and macrophages in the spleen. This study demonstrates that SERS-based diagnosis mediated by AuNS provides an accurate measure of multiple biomarkers both in vivo and ex vivo, which will ultimately enable a clinically-translatable platform for patient-tailored immunotherapies and combination treatments.


Assuntos
Neoplasias da Mama/diagnóstico , Ouro , Nanopartículas Metálicas , Análise Espectral Raman , Antígeno B7-H1/análise , Receptores ErbB/análise , Humanos , Sensibilidade e Especificidade
4.
ACS Omega ; 2(7): 3583-3594, 2017 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-28782050

RESUMO

In this study, we demonstrate the theranostic capability of actively targeted, site-specific multibranched gold nanoantennas (MGNs) in triple-negative breast cancer (TNBC) cells in vitro. By utilizing multiplexed surface-enhanced Raman scattering (SERS) imaging, enabled by the narrow peak widths of Raman signatures, we simultaneously targeted immune checkpoint receptor programmed death ligand 1 (PDL1) and the epidermal growth factor receptor (EGFR) overexpressed in TNBC cells. A 1:1 mixture of MGNs functionalized with anti-PDL1 antibodies and Raman tag 5,5-dithio-bis-(2-nitrobenzoic acid) (DTNB) and MGNs functionalized with anti-EGFR antibodies and Raman tag para-mercaptobenzoic acid (pMBA) were incubated with the cells. SERS imaging revealed a cellular traffic map of MGN localization by surface binding and receptor-mediated endocytosis, enabling targeted diagnosis of both biomarkers. Furthermore, cells incubated with anti-EGFR-pMBA-MGNs and illuminated with an 808 nm laser for 15 min at 4.7 W/cm2 exhibited photothermal cell death only within the laser spot (indicated by live/dead cell fluorescence assay). Therefore, this study not only provides an optical imaging platform that can track immunomarkers with spatiotemporal control but also demonstrates an externally controlled light-triggered therapeutic approach enabling receptor-specific treatment with biocompatible theranostic nanoprobes.

5.
ACS Omega ; 1(2): 234-243, 2016 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-27656689

RESUMO

In this work, we demonstrate controlled drug delivery from low-temperature-sensitive liposomes (LTSLs) mediated by photothermal heating from multibranched gold nanoantennas (MGNs) in triple-negative breast cancer (TNBC) cells in vitro. The unique geometry of MGNs enables the generation of mild hyperthermia (∼42 °C) by converting near-infrared light to heat and effectively delivering doxorubicin (DOX) from the LTSLs in breast cancer cells. We confirmed the cellular uptake of MGNs by using both fluorescence confocal Z-stack imaging and transmission electron microscopy (TEM) imaging. We performed a cellular viability assay and live/dead cell fluorescence imaging of the combined therapeutic effects of MGNs with DOX-loaded LTSLs (DOX-LTSLs) and compared them with free DOX and DOX-loaded non-temperature-sensitive liposomes (DOX-NTSLs). Imaging of fluorescent live/dead cell indicators and MTT assay outcomes both demonstrated significant decreases in cellular viability when cells were treated with the combination therapy. Because of the high phase-transition temperature of NTSLs, no drug delivery was observed from the DOX-NTSLs. Notably, even at a low DOX concentration of 0.5 µg/mL, the combination treatment resulted in a higher (33%) cell death relative to free DOX (17% cell death). The results of our work demonstrate that the synergistic therapeutic effect of photothermal hyperthermia of MGNs with drug delivery from the LTSLs can successfully eradicate aggressive breast cancer cells with higher efficacy than free DOX by providing a controlled light-activated approach and minimizing off-target toxicity.

6.
Nanoscale ; 6(5): 2502-30, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24445488

RESUMO

Gold nanostructures possess unique characteristics that enable their use as contrast agents, as therapeutic entities, and as scaffolds to adhere functional molecules, therapeutic cargo, and targeting ligands. Due to their ease of synthesis, straightforward surface functionalization, and non-toxicity, gold nanostructures have emerged as powerful nanoagents for cancer detection and treatment. This comprehensive review summarizes the progress made in nanomedicine with gold nanostructures (1) as probes for various bioimaging techniques including dark-field, one-photon and two-photon fluorescence, photothermal optical coherence tomography, photoacoustic tomography, positron emission tomography, and surface-enhanced Raman scattering based imaging, (2) as therapeutic components for photothermal therapy, gene and drug delivery, and radiofrequency ablation, and (3) as a theranostic platform to simultaneously achieve both cancer detection and treatment. Distinct from other published reviews, this article also discusses the recent advances of gold nanostructures as contrast agents and therapeutic actuators for inflammatory diseases including atherosclerotic plaque and arthritis. For each of the topics discussed above, the fundamental principles and progress made in the past five years are discussed. The review concludes with a detailed future outlook discussing the challenges in using gold nanostructures, cellular trafficking, and translational considerations that are imperative for rapid clinical viability of plasmonic nanostructures, as well as the significance of emerging technologies such as Fano resonant gold nanostructures in nanomedicine.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Nanomedicina , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Portadores de Fármacos/química , Humanos , Nanopartículas Metálicas/ultraestrutura , Neoplasias/diagnóstico , Neoplasias/terapia , Imagem Óptica , Tomografia
7.
RNA ; 19(8): 1078-88, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23798665

RESUMO

Despite the vast excess of cellular RNAs, precisely two copies of viral genomic RNA (gRNA) are selectively packaged into new human immunodeficiency type 1 (HIV-1) particles via specific interactions between the HIV-1 Gag and the gRNA psi (ψ) packaging signal. Gag consists of the matrix (MA), capsid, nucleocapsid (NC), and p6 domains. Binding of the Gag NC domain to ψ is necessary for gRNA packaging, but the mechanism by which Gag selectively interacts with ψ is unclear. Here, we investigate the binding of NC and Gag variants to an RNA derived from ψ (Psi RNA), as well as to a non-ψ region (TARPolyA). Binding was measured as a function of salt to obtain the effective charge (Zeff) and nonelectrostatic (i.e., specific) component of binding, Kd(1M). Gag binds to Psi RNA with a dramatically reduced Kd(1M) and lower Zeff relative to TARPolyA. NC, GagΔMA, and a dimerization mutant of Gag bind TARPolyA with reduced Zeff relative to WT Gag. Mutations involving the NC zinc finger motifs of Gag or changes to the G-rich NC-binding regions of Psi RNA significantly reduce the nonelectrostatic component of binding, leading to an increase in Zeff. These results show that Gag interacts with gRNA using different binding modes; both the NC and MA domains are bound to RNA in the case of TARPolyA, whereas binding to Psi RNA involves only the NC domain. Taken together, these results suggest a novel mechanism for selective gRNA encapsidation.


Assuntos
HIV-1/fisiologia , RNA Viral/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Genoma Viral , HIV-1/genética , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Técnicas de Amplificação de Ácido Nucleico , RNA Viral/química , RNA Viral/genética , Montagem de Vírus/genética , Montagem de Vírus/fisiologia , Dedos de Zinco/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Pequeno RNA não Traduzido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...