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











Base de dados
Intervalo de ano de publicação
1.
Angew Chem Int Ed Engl ; : e202410919, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38995663

RESUMO

Despite numerous screening tools for colorectal cancer (CRC), 25% of patients are diagnosed with advanced disease.  Novel diagnostic technologies that are early, accurate, and rapid are imperative to assess the therapeutic efficacy of clinical drugs and identify new biomarkers of treatment response. Here Raman spectroscopy (RS) was used to track metabolic reprogramming in KRAS-mutant HCT116 and SW837 cells, and KRAS wild-type CC cells. RS combined with multivariate analysis methods distinguished nonresponsive, partially responsive, and responsive cells treated with cetuximab, a monoclonal antibody for EGFR inhibition, sotorasib, a clinically approved KRAS inhibitor, and various doses of trametinib, an inhibitor of the MAPK pathway. Cells treated with a combination of subtoxic doses of trametinib and BKM120, an inhibitor of the PI3K pathway, showed a synergistic response between the two pathways. Using a supervised machine learning regression model, we established a scoring methodology trained to a priori predict therapeutic response to new treatment combinations. RS metabolites were verified with mass spectrometry, and enrichment pathways were identified, including amino acid, purine, and nicotinate and nicotinamide metabolism that differentiated monotherapy from combination therapy. Our approach may ultimately be applicable to patient-derived primary cells and cultures of patient tumors to predict effective drugs for individualized care.

2.
Biomaterials ; 308: 122531, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38531198

RESUMO

Radiation therapy (RT) is essential for triple negative breast cancer (TNBC) treatment. However, patients with TNBC continue to experience recurrence after RT. The role of the extracellular matrix (ECM) of irradiated breast tissue in tumor recurrence is still unknown. In this study, we evaluated the structure, molecular composition, and mechanical properties of irradiated murine mammary fat pads (MFPs) and developed ECM hydrogels from decellularized tissues (dECM) to assess the effects of RT-induced ECM changes on breast cancer cell behavior. Irradiated MFPs were characterized by increased ECM deposition and fiber density compared to unirradiated controls, which may provide a platform for cell invasion and proliferation. ECM component changes in collagens I, IV, and VI, and fibronectin were observed following irradiation in both MFPs and dECM hydrogels. Encapsulated TNBC cell proliferation and invasive capacity was enhanced in irradiated dECM hydrogels. In addition, TNBC cells co-cultured with macrophages in irradiated dECM hydrogels induced M2 macrophage polarization and exhibited further increases in proliferation. Our study establishes that the ECM in radiation-damaged sites promotes TNBC invasion and proliferation as well as an immunosuppressive microenvironment. This work represents an important step toward elucidating how changes in the ECM after RT contribute to breast cancer recurrence.


Assuntos
Proliferação de Células , Matriz Extracelular , Hidrogéis , Neoplasias de Mama Triplo Negativas , Microambiente Tumoral , Animais , Matriz Extracelular/metabolismo , Microambiente Tumoral/efeitos da radiação , Hidrogéis/química , Feminino , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Linhagem Celular Tumoral , Camundongos , Humanos , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/radioterapia , Macrófagos/metabolismo , Glândulas Mamárias Animais/efeitos da radiação
3.
Biomater Sci ; 11(21): 7188-7202, 2023 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-37750339

RESUMO

Magnetic hyperthermia has attracted considerable attention for efficient cancer therapy because of its noninvasive nature, deep tissue penetration, and minimal damage to healthy tissues. Herein, we have fused cancer cell membrane fragments with lipids and cloaked them on magnetic nanorings to form targeted Fe nanorings (TF) for tumor-targeted magnetic hyperthermia-induced tumor ablation. In our approach, cell membrane fragments from cancer cells were fused with lipids to form vesicles, which could efficiently encapsulate magnetic nanorings, thereby forming TF. We observed that TF have high tumor uptake via homotypic targeting, where cancer cells take up TF through membrane fusion. Under an external alternating magnetic field (AMF), TF accumulated in the tumors are heated, driving magnetic-hyperthermia-induced tumor cell death. Our in vitro studies show that self-targeting TF efficiently localized in cancer cells and induced cell death with an AMF, which was shown by a live/dead assay. Our findings demonstrate the potential of TF in tumor ablation, thereby making them promising and efficient nanosystems for tumor-targeted theranostics.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita , Linhagem Celular Tumoral , Membrana Celular , Fenômenos Magnéticos , Lipídeos , Campos Magnéticos
4.
Anal Chem ; 95(35): 13172-13184, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37605298

RESUMO

Resistance to clinical therapies remains a major barrier in cancer management. There is a critical need for rapid and highly sensitive diagnostic tools that enable early prediction of treatment response to allow accurate clinical decisions. Here, Raman spectroscopy was employed to monitor changes in key metabolites as early predictors of response in KRAS-mutant colorectal cancer (CRC) cells, HCT116, treated with chemotherapies. We show at the single cell level that HCT116 is resistant to cetuximab (CTX), the first-line treatment in CRC, but this resistance can be overcome with pre-sensitization of cells with oxaliplatin (OX). In combination treatment of CTX + OX, sequential delivery of OX followed by CTX rather than simultaneous administration of drugs was observed to be critical for effective therapy. Our results demonstrated that metabolic changes are well aligned to cellular mechanical changes where Young's modulus decreased after effective treatment, indicating that both changes in mechanical properties and metabolism in cells are likely responsible for cancer proliferation. Raman findings were verified with mass spectrometry (MS) metabolomics, and both platforms showed changes in lipids, nucleic acids, and amino acids as predictors of resistance/response. Finally, key metabolic pathways enriched were identified when cells are resistant to CTX but downregulated with effective treatment. This study highlights that drug-induced metabolic changes both at the single cell level (Raman) and ensemble level (MS) have the potential to identify mechanisms of response to clinical cancer therapies.


Assuntos
Antifibrinolíticos , Neoplasias , Humanos , Análise Espectral Raman , Metabolômica , Aminoácidos , Cetuximab/farmacologia , Oxaliplatina/farmacologia
5.
Chem Rev ; 123(13): 8297-8346, 2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37318957

RESUMO

Omics technologies have rapidly evolved with the unprecedented potential to shape precision medicine. Novel omics approaches are imperative toallow rapid and accurate data collection and integration with clinical information and enable a new era of healthcare. In this comprehensive review, we highlight the utility of Raman spectroscopy (RS) as an emerging omics technology for clinically relevant applications using clinically significant samples and models. We discuss the use of RS both as a label-free approach for probing the intrinsic metabolites of biological materials, and as a labeled approach where signal from Raman reporters conjugated to nanoparticles (NPs) serve as an indirect measure for tracking protein biomarkers in vivo and for high throughout proteomics. We summarize the use of machine learning algorithms for processing RS data to allow accurate detection and evaluation of treatment response specifically focusing on cancer, cardiac, gastrointestinal, and neurodegenerative diseases. We also highlight the integration of RS with established omics approaches for holistic diagnostic information. Further, we elaborate on metal-free NPs that leverage the biological Raman-silent region overcoming the challenges of traditional metal NPs. We conclude the review with an outlook on future directions that will ultimately allow the adaptation of RS as a clinical approach and revolutionize precision medicine.


Assuntos
Medicina de Precisão , Análise Espectral Raman , Medicina de Precisão/métodos , Proteômica/métodos , Metabolômica/métodos , Biomarcadores/metabolismo
6.
Biomater Sci ; 11(1): 298-306, 2022 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-36448579

RESUMO

In an immunosuppressive tumor microenvironment, tumor-associated macrophages (TAMs) are the most abundant cells displaying pro-tumorigenic M2-like phenotypes, encouraging tumor growth and influencing the development of resistance against conventional therapies. TAMs are highly malleable. They can be repolarized into tumoricidal M1-like cells. In this study, we report the synthesis of novel co-operative immuno-photodynamic nanoparticles involving TAM self-targeting acrylic acid grafted mannan (a polysaccharide) conjugated with the chlorin e6 (Ce6) photosensitizer and then loaded with resiquimod (R848), a toll-like receptor (TLR7/8) agonist. The mannan conjugated Ce6 loaded with R848 (MCR) as bioconjugate nanoparticles demonstrated selective targeting of anti-inflammatory M2-like cells. Using photodynamic therapy they were repolarized to pro-inflammatory M1-like cells with combined effects of reactive oxygen species (ROS)-triggered intracellular signaling and a small-molecule immunostimulant. The MCR also demonstrated a TAM-directed adaptive immune response, inhibited tumor growth, and prevented metastasis. Our results indicate that these MCR nanoparticles can effectively target TAMs and modulate them for cancer immunotherapy.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Mananas , Macrófagos Associados a Tumor , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/uso terapêutico , Fotoquimioterapia/métodos , Neoplasias/tratamento farmacológico , Linhagem Celular Tumoral , Microambiente Tumoral
7.
Artigo em Inglês | MEDLINE | ID: mdl-36006784

RESUMO

Natural killer (NK) cells are an important component of the tumor immunosurveillance; activated NK cells can recognize and directly lyse tumor cells eliciting a potent antitumor immune response. Due to their intrinsic ability to unleash cytotoxicity against tumor cells, NK cell-based adoptive cell therapies have gained rapid clinical significance, and many clinical trials are ongoing. However, priming and activating NK cells, infiltration of activated NK cells in the immunosuppressive tumor microenvironment, and tracking the infiltrated NK cells in the tumors remain a critical challenge. To address these challenges, NK cells have been successfully interfaced with nanomaterials where the morphology, composition, and surface characteristics of nanoparticles (NPs) were leveraged to enable longitudinal tracking of NK cells in tumors or deliver therapeutics to prime NK cells. Distinct from other published reviews, in this tutorial review, we summarize the recent findings in the past decade where NPs were used to label NK cells for immunoimaging or deliver treatment to activate NK cells and induce long-term immunity against tumors. We discuss the NP properties that are key to surmounting the current challenges in NK cells and the different strategies employed to advance NK cells-based diagnostics and therapeutics. We conclude the review with an outlook on future directions in NP-NK cell hybrid interfaces, and overall clinical impact and patient response to such interfaces that need to be addressed to enable their clinical translation.

8.
Chem Sci ; 11(36): 9863-9874, 2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34094246

RESUMO

Rapid and accurate response to targeted therapies is critical to differentiate tumors that are resistant to treatment early in the regimen. In this work, we demonstrate a rapid, noninvasive, and label-free approach to evaluate treatment response to molecular inhibitors in breast cancer (BC) cells with Raman spectroscopy (RS). Metabolic reprogramming in BC was probed with RS and multivariate analysis was applied to classify the cells into responsive or nonresponsive groups as a function of drug dosage, drug type, and cell type. Metabolites identified with RS were then validated with mass spectrometry (MS). We treated triple-negative BC cells with Trametinib, an inhibitor of the extracellular-signal-regulated kinase (ERK) pathway. Changes measured with both RS and MS corresponding to membrane phospholipids, amino acids, lipids and fatty acids indicated that these BC cells were responsive to treatment. Comparatively, minimal metabolic changes were observed post-treatment with Alpelisib, an inhibitor of the mammalian target of rapamycin (mTOR) pathway, indicating treatment resistance. These findings were corroborated with cell viability assay and immunoblotting. We also showed estrogen receptor-positive MCF-7 cells were nonresponsive to Trametinib with minimal metabolic and viability changes. Our findings support that oncometabolites identified with RS will ultimately enable rapid drug screening in patients ensuring patients receive the most effective treatment at the earliest time point.

9.
Trends Biotechnol ; 38(4): 388-403, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31812371

RESUMO

Dynamic immunoimaging in vivo is crucial in patient-tailored immunotherapies to identify patients who will benefit from immunotherapies, monitor therapeutic efficacy post treatment, and determine alternative strategies for nonresponders. Nanoparticles have played a major role in the immunotherapy landscape. In this review, we summarize recent findings in immunoimaging where smart nanoparticles target, detect, stimulate, and deliver therapeutic dose in vivo. Nanoparticles interfaced with an immunoimaging toolbox enable the use of multiple modalities and achieve depth-resolved whole-body tracking of immunomarkers with high accuracy both before and after treatment. We highlight how functional nanoparticles track T cells, dendritic cells (DCs), tumor-associated macrophages (TAMs), and immune checkpoint receptors (ICRs), and facilitate image-guided interventions.


Assuntos
Rastreamento de Células/métodos , Ouro/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Imagem Molecular/métodos , Neoplasias/diagnóstico por imagem , Animais , Células Dendríticas/imunologia , Ouro/química , Humanos , Proteínas de Checkpoint Imunológico/imunologia , Imunoterapia , Nanopartículas Metálicas/química , Neoplasias/imunologia , Neoplasias/terapia , Polímeros Responsivos a Estímulos , Linfócitos T/imunologia , Macrófagos Associados a Tumor/imunologia
10.
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
11.
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
12.
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.

13.
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.

14.
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
15.
Nanomedicine (Lond) ; 9(8): 1209-22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24063415

RESUMO

AIM: We report a magneto-fluorescent theranostic nanocomplex targeted to neutrophil gelatinase-associated lipocalin (NGAL) for imaging and therapy of pancreatic cancer. MATERIALS & METHODS: Gold nanoshells resonant at 810 nm were encapsulated in silica epilayers doped with iron oxide and the near-infrared (NIR) dye indocyanine green, resulting in theranostic gold nanoshells (TGNS), which were subsequently conjugated with antibodies targeting NGAL in AsPC-1-derived xenografts in nude mice. RESULTS: Anti-NGAL-conjugated TGNS specifically targeted pancreatic cancer cells in vitro and in vivo providing contrast for both NIR fluorescence and T2-weighted MRI with higher tumor contrast than can be obtained using long-circulating, but nontargeted, PEGylated nanoparticles. The nanocomplexes also enabled highly specific cancer cell death via NIR photothermal therapy in vitro. CONCLUSION: TGNS with embedded NIR and magnetic resonance contrasts can be specifically targeted to pancreatic cancer cells with expression of early disease marker NGAL, and enable molecularly targeted imaging and photothermal therapy.


Assuntos
Ouro/uso terapêutico , Nanoconchas/uso terapêutico , Pâncreas/patologia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Proteínas de Fase Aguda/metabolismo , Animais , Linhagem Celular Tumoral , Meios de Contraste/química , Meios de Contraste/uso terapêutico , Sistemas de Liberação de Medicamentos , Feminino , Corantes Fluorescentes/química , Corantes Fluorescentes/uso terapêutico , Ouro/química , Humanos , Hipertermia Induzida , Lipocalina-2 , Lipocalinas/metabolismo , Imageamento por Ressonância Magnética , Imãs/química , Camundongos Nus , Nanoconchas/química , Proteínas Oncogênicas/metabolismo , Imagem Óptica , Neoplasias Pancreáticas/patologia , Fototerapia
16.
Cancer Nanotechnol ; 3(1-6): 47-54, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23205151

RESUMO

As systemic cancer therapies improve and are able to control metastatic disease outside the central nervous system, the brain is increasingly the first site of relapse. The blood-brain barrier (BBB) represents a major challenge to the delivery of therapeutics to the brain. Macrophages originating from circulating monocytes are able to infiltrate brain metastases while the BBB is intact. Here, we show that this ability can be exploited to deliver both diagnostic and therapeutic nanoparticles specifically to experimental brain metastases of breast cancer.

17.
Acc Chem Res ; 44(10): 936-46, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-21612199

RESUMO

Recent advances in nanoscience and biomedicine have expanded our ability to design and construct multifunctional nanoparticles that combine targeting, therapeutic, and diagnostic functions within a single nanoscale complex. The theranostic capabilities of gold nanoshells, spherical nanoparticles with silica cores and gold shells, have attracted tremendous attention over the past decade as nanoshells have emerged as a promising tool for cancer therapy and bioimaging enhancement. This Account examines the design and synthesis of nanoshell-based theranostic agents, their plasmon-derived optical properties, and their corresponding applications. We discuss the design and preparation of nanoshell complexes and their ability to enhance the photoluminescence of fluorophores while maintaining their properties as MR contrast agents. In this Account, we discuss the underlying physical principles that contribute to the photothermal response of nanoshells. We then elucidate the photophysical processes that induce nanoshells to enhance the fluorescence of weak near-infrared fluorophores. Nanoshells illuminated with resonant light are either strong optical absorbers or scatterers, properties that give rise to their unique capabilities. These physical processes have been harnessed to visualize and eliminate cancer cells. We describe the application of nanoshells as a contrast agent for optical coherence tomography of breast carcinoma cells in vivo. Our recent studies examine nanoshells as a multimodal theranostic probe, using these nanoparticles for near-infrared fluorescence and magnetic resonance imaging (MRI) and for the photothermal ablation of cancer cells. Multimodal nanoshells show theranostic potential for imaging subcutaneous breast cancer tumors in animal models and the distribution of tumors in various tissues. Nanoshells also show promise as light-triggered gene therapy vectors, adding temporal control to the spatial control characteristic of nanoparticle-based gene therapy approaches. We describe the fabrication of DNA-conjugated nanoshell complexes and compare the efficiency of light-induced and thermally-induced release of DNA. Double-stranded DNA nanoshells also provide a way to deliver small molecules into cells: we describe the delivery and light-triggered release of DAPI (4',6-diamidino-2-phenylindole), a dye molecule used to stain DNA in the nuclei of cells.


Assuntos
Imagem Molecular/métodos , Sondas Moleculares/uso terapêutico , Nanoconchas/uso terapêutico , Neoplasias/diagnóstico , Neoplasias/terapia , Animais , Linhagem Celular Tumoral , Desenho de Fármacos , Humanos , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Nanoconchas/química , Neoplasias/metabolismo , Fenômenos Ópticos
18.
Nano Lett ; 10(12): 4920-8, 2010 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-21090693

RESUMO

Nanoparticle-based therapeutics with local delivery and external electromagnetic field modulation holds extraordinary promise for soft-tissue cancers such as breast cancer; however, knowledge of the distribution and fate of nanoparticles in vivo is crucial for clinical translation. Here we demonstrate that multiple diagnostic capabilities can be introduced in photothermal therapeutic nanocomplexes by simultaneously enhancing both near-infrared fluorescence and magnetic resonance imaging (MRI). We track nanocomplexes in vivo, examining the influence of HER2 antibody targeting on nanocomplex distribution over 72 h. This approach provides valuable, detailed information regarding the distribution and fate of complex nanoparticles designed for specific diagnostic and therapeutic functions.


Assuntos
Neoplasias da Mama/terapia , Nanopartículas , Animais , Linhagem Celular Tumoral , Campos Eletromagnéticos , Feminino , Fluorescência , Humanos , Imageamento por Ressonância Magnética , Camundongos , Espectroscopia de Luz Próxima ao Infravermelho
19.
Mol Cancer Ther ; 9(4): 1028-38, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20371708

RESUMO

Overexpression of the human epidermal growth factor receptor (HER) family has been implicated in ovarian cancer because of its participation in signaling pathway regulating cellular proliferation, differentiation, motility, and survival. Currently, effective diagnostic and therapeutic schemes are lacking for treating ovarian cancer, and consequently ovarian cancer has a high mortality rate. Although HER2 receptor expression does not usually affect the survival rates of ovarian cancer to the same extent as in breast cancer, it can be used as a docking site for directed nanotherapies in cases with de novo or acquired chemotherapy resistance. In this study, we have exploited a novel gold nanoshell-based complex (nanocomplex) for targeting, dual modal imaging, and photothermal therapy of HER2-overexpressing and drug-resistant ovarian cancer OVCAR3 cells in vitro. The nanocomplexes are engineered to simultaneously provide contrast as fluorescence optical imaging probe and a magnetic resonance imaging agent. Immunofluorescence staining and magnetic resonance imaging successfully show that nanocomplex-anti-HER2 conjugates specifically bind to OVCAR3 cells as opposed to the control, MDA-MB-231 cells, which have low HER2 expression. In addition, nanocomplexes targeted to OVCAR3 cells, when irradiated with near-IR laser, result in selective destruction of cancer cells through photothermal ablation. We also show that near-IR light therapy and the nanocomplexes by themselves are noncytotoxic in vitro. To the best of our knowledge, this is the first successful integration of dual modal bioimaging with photothermal cancer therapy for treatment of ovarian cancer. Based on their efficacy in vitro, these nanocomplexes are highly promising for image-guided photothermal therapy of ovarian cancer, as well as other HER2-overexpressing cancers. Mol Cancer Ther; 9(4); 1028-38. (c)2010 AACR.


Assuntos
Sondas Moleculares , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/terapia , Temperatura , Técnicas de Ablação , Morte Celular , Linhagem Celular Tumoral , Meios de Contraste , Feminino , Humanos , Imageamento por Ressonância Magnética , Microscopia de Fluorescência , Nanoconchas , Neoplasias Ovarianas/patologia , Receptor ErbB-2/metabolismo
20.
Nano Lett ; 7(12): 3759-65, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17979310

RESUMO

Destruction of hypoxic regions within tumors, virtually inaccessible to cancer therapies, may well prevent malignant progression. The tumor's recruitment of monocytes into these regions may be exploited for nanoparticle-based delivery. Monocytes containing therapeutic nanoparticles could serve as "Trojan Horses" for nanoparticle transport into these tumor regions. Here we report the demonstration of several key steps toward this therapeutic strategy: phagocytosis of Au nanoshells, and photoinduced cell death of monocytes/macrophages as isolates and within tumor spheroids.


Assuntos
Antineoplásicos/administração & dosagem , Nanopartículas/administração & dosagem , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico , Transporte Biológico , Hipóxia Celular , Sistemas de Liberação de Medicamentos/métodos , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/ultraestrutura , Monócitos/efeitos dos fármacos , Monócitos/ultraestrutura , Nanopartículas/uso terapêutico , Espectrofotometria
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA