Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 21(1): 138, 2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-37106405

RESUMO

Since the successful clinical trial of AuroShell for photothermal therapy, there is currently intense interest in developing gold-based core-shell structures with near-infrared (NIR) absorption ranging from NIR-I (650-900 nm) to NIR-II (900-1700 nm). Here, we propose a seed-mediated successive growth approach to produce gold nanoshells on the surface of the nanoscale metal-organic framework (NMOF) of UiO-66-NH2 (UiO = the University of Oslo) in one pot. The key to this strategy is to modulate the proportion of the formaldehyde (reductant) and its regulator / oxidative product of formic acid to harness the particle nucleation and growth rate within the same system. The gold nanoshells propagate through a well-oriented and controllable diffusion growth pattern (points → facets → octahedron), which has not been identified. Most strikingly, the gold nanoshells prepared hereby exhibit an exceedingly broad and strong absorption in NIR-II with a peak beyond 1300 nm and outstanding photothermal conversion efficiency of 74.0%. Owing to such superior performance, these gold nanoshells show promising outcomes in photoacoustic (PA), computed tomography (CT), and photothermal imaging-guided photothermal therapy (PTT) for breast cancer, as demonstrated both in vitro and in vivo.


Assuntos
Nanoconchas , Nanoconchas/química , Terapia Fototérmica , Ouro/química , Imagem Multimodal , Fototerapia
2.
Nanomedicine ; 33: 102349, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33359414

RESUMO

Drug resistance and inefficient localization of chemotherapeutic agent limit the current treatment strategy in locally advanced melanoma (MEL), accounting to the 10-year survival rate from 24% to 68%. In this study we constructed anti-PD-L1 conjugated and doxorubicin loaded hollow gold nanoshell (T-HGNS-DOX) for targeted and localized chemo-photothermal therapy of MEL by the conjugation of LA-PEG-anti-PD-L1 antibody and short PEG chain on the surface of HGNS-DOX. Near infrared (NIR) as well as pH dependent drug release profile was observed. Significant uptake of DOX following NIR due to high PD-L1 receptors resulted in pronounced anticancer effect of T-HGNS-DOX. Following intratumoral administration, maximum nanoparticles retention with the significant reduction in tumor growth was observed as a result of elevated apoptosis marker (cleaved caspase-3, cleaved PARP) as well as downregulation of proliferative (Ki-67) and angiogenesis marker (CD31). Cumulatively, our system avoids the systemic toxicities of the nanosystem thereby providing maximum chemotherapeutic retention in tumor.


Assuntos
Anticorpos Monoclonais Humanizados/química , Doxorrubicina/química , Ouro/química , Melanoma/tratamento farmacológico , Melanoma/radioterapia , Nanocápsulas/química , Nanoconchas/química , Animais , Anticorpos Monoclonais Humanizados/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Proliferação de Células/efeitos dos fármacos , Doxorrubicina/farmacologia , Composição de Medicamentos , Liberação Controlada de Fármacos , Humanos , Concentração de Íons de Hidrogênio , Masculino , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fototerapia , Propriedades de Superfície
3.
ACS Appl Mater Interfaces ; 12(28): 31172-31181, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32532159

RESUMO

The development of stimuli-responsive multifunctional nanocarriers for therapeutic drug delivery is extremely desirable for highly specific treatment of disease. Herein, thiol-polyethylene glycol-folate acid-modified hollow mesoporous bismuth nanoshells (HM-Bi@PEG-FA NSs) were developed as the new dual-stimuli-responsive single-"elemental" photothermal nanocarriers for synergistic chemo-photothermal therapy of tumor. The designed hollow-mesoporous-type nanocarriers present excellent photothermal conversion capacity (∼34.72%) and good biocompatibility. Meanwhile, acidic pH and near-infrared (NIR) laser dual-stimulated doxorubicin (DOX) release is successfully achieved. More importantly, the DOX-loaded HM-Bi@PEG-FA NSs hold an efficient in vitro/in vivo antitumor effect through the synergistic chemo-photothermal therapy. Therefore, our findings provide the possibility of designing a dual-stimuli-responsive hollow mesoporous Bi-based photothermal nanocarrier for synergistically enhanced antitumor therapy.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Nanoconchas/química , Linhagem Celular Tumoral , Doxorrubicina/química , Humanos , Nanopartículas/química , Fototerapia/métodos
4.
Biomed Res Int ; 2020: 5869235, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32352001

RESUMO

The purpose of this study was to investigate the effect of photothermal treatment (PTT) with gold nanoshell (ANS) using a macrophage-mediated delivery system in a head and neck squamous cell carcinoma (HNSCC) cell line. To achieve this, ANS-loaded rat macrophages (ANS-MAs) were prepared via the coculture method with ANS. The human HNSCC (FaDu cell) and macrophage (rat macrophage; NR8383 cell) hybrid spheroid models were generated by the centrifugation method to determine the possibility of using ANS-MAs as a cancer therapy. These ANS-MAs were set into the tumor and macrophage hybrid spheroid model to measure PTT efficacy. Kinetic analysis of the spheroid growth pattern revealed that this PTT process caused a decreasing pattern in the volume of the hybrid model containing ANS-MAs (p < 0.001). Comparison with empty macrophages showed harmony between ANS and laser irradiation for the generation of PTT. An annexin V/dead cell marker assay indicated that the PTT-treated hybrid model induced increasing apoptosis and dead cells. Further studies on the toxicity of ANS-MAs are needed to reveal whether it can be considered biocompatible. In summary, the ANS was prepared with a macrophage as the delivery method and protective carrier. The ANS was successfully localized to the macrophages, and their photoabsorption property was stationary. This strategy showed significant growth inhibition of the tumor and macrophage spheroid model under NIR laser irradiation. In vivo toxicology results suggest that ANS-MA is a promising candidate for a biocompatible strategy to overcome the limitations of fabricated nanomaterials. This ANS-MA delivery and PTT strategy may potentially lead to improvements in the quality of life of patients with HNSCC by providing a biocompatible, minimally invasive modality for cancer treatment.


Assuntos
Ouro , Hipertermia Induzida , Nanoconchas , Carcinoma de Células Escamosas de Cabeça e Pescoço , Animais , Linhagem Celular Tumoral , Ouro/química , Ouro/farmacologia , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Nanoconchas/química , Nanoconchas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia
5.
ACS Nano ; 14(3): 3299-3315, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32048825

RESUMO

Due to the inability to spontaneously heal and vulnerability to bacterial infection, diabetic patients are frustrated by unexpected epithelium injuries in daily life. Notably, a drug-resistant bacterial infection may result in a long-term impact to the natural function of damaged organs. It is imperative to develop strategies that promote injury recovery and eradicate drug-resistant infection simultaneously. Here, we present a composite structured cupriferous hollow nanoshell (AuAgCu2O NS) that consists of a hollow gold-silver (AuAg) core and Cu2O shell as a photothermal therapeutic agent for a cutaneous chronic wound and nonhealing keratitis with drug-resistant bacterial infection. The controllable photothermal therapeutic effect and released silver ion from the hollow AuAg core possess a synergistic effect to eradicate multi-drug-resistant bacteria, including extended-spectrum ß-lactamase Escherichia coli (ESBL E. coli) and methicillin-resistant Staphylococcus aureus (MRSA). Meanwhile, the released copper ion from the Cu2O shell could expedite endothelial cell angiogenesis and fibroblast cell migration, thus boosting wound-healing effects. In both infection-complicated disease models, the ophthalmic clinical score, wound closure rates, and histopathology analysis demonstrate that the AuAgCu2O NSs could facilitate the re-epithelialization at the wound area and eliminate the complicated bacterial infection from diabetic mice. A primary signal path involved in the promoted healing effect was further illustrated by comprehensive assays of immunohistochemical evaluation, Western blot, and quantitative polymerase chain reaction. Taken together, our AuAgCu2O NSs are shown to be potent candidates for clinical utilization in the treatment of diabetic epithelium injuries.


Assuntos
Antibacterianos/farmacologia , Infecções Bacterianas/terapia , Ceratite/terapia , Luz , Nanoconchas/química , Cicatrização/efeitos dos fármacos , Animais , Antibacterianos/síntese química , Antibacterianos/química , Infecções Bacterianas/patologia , Cobre/química , Cobre/farmacologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Feminino , Ouro/química , Ouro/farmacologia , Ceratite/patologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Tamanho da Partícula , Fototerapia , Porosidade , Prata/química , Prata/farmacologia , Propriedades de Superfície
6.
J Nanobiotechnology ; 17(1): 99, 2019 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-31530285

RESUMO

BACKGROUND: Combined therapy has demonstrated to be an effective strategy for cancer therapy. Herein, an injectable hydrogel based on the genetically engineered polypeptide and hollow gold nanoshells (HAuNS) has been developed for chemo-photothermal therapy of HepG2 tumor. METHODS: PC10A/DOX/HAuNS nanogel was prepared with layer-by-layer through the adsorption of DOX and PC10A successively. DOX with positive charge and PC10A with negative charge were coated step by step onto the surface of negatively charged HAuNS. The multifunctional hydrogel PC10A/DOX/HAuNS were prepared via dissolving hybrid PC10A/DOX/HAuNS nanogel in polypeptide PC10A. Chemotherapy drug DOX in the PC10A/DOX/HAuNS hydrogel was absorbed on the HAuNS and directly embedded in the PC10A hydrogel, which contributes to sequentially release of the drug. Specifically, DOX adsorbed on the HAuNS could be released slowly for sustainable chemotherapy. RESULTS: The PC10A/DOX/HAuNS hydrogel could pass 26-gauge needle without clogging, indicating that it is injectable. In addition, the PC10A/DOX/HAuNS hydrogel possessed outstanding photothermal effect and photothermal stability. In both in vitro cell and in vivo tumor-bearing mice experiments, a remarkably enhance tumor inhibition was observed by the combined therapy of chemo-photothermal therapy compared with photothermal therapy or chemotherapy alone. CONCLUSIONS: The combined chemotherapy and photothermal therapy of PC10A/DOX/HAuNS hydrogels could significantly improve the therapeutic effect. Therefore, the multifunctional hydrogel PC10A/DOX/HAuNS is promising to provide a new strategy for sustained chemo-photothermal therapy.


Assuntos
Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/química , Ouro/química , Hidrogéis/administração & dosagem , Hidrogéis/química , Nanoconchas/química , Animais , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Portadores de Fármacos/química , Células Hep G2 , Humanos , Masculino , Camundongos , Nanosferas/química , Fototerapia/métodos
7.
Int J Nanomedicine ; 14: 5369-5379, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31409993

RESUMO

Purpose: Photothermal therapy (PTT) exploits the light-absorbing properties of nanomaterials such as silica-gold nanoshells (NS) to inflict tumor death through local hyperthermia. However, in in vivo studies of PTT, the heat distribution is often found to be heterogeneous throughout the tumor volume, which leaves parts of the tumor untreated and impairs the overall treatment outcome. As this challenges PTT as a one-dose therapy, this study here investigates if giving the treatment repeatedly, ie, fractionated PTT, increases the efficacy in mice bearing subcutaneous tumors. Methods: The NS heating properties were first optimized in vitro and in vivo. Two fractionated PTT protocols, consisting of two and four laser treatments, respectively, were developed and applied in a murine subcutaneous colorectal tumor model. The efficacy of the two fractionated protocols was evaluated both by longitudinal monitoring of tumor growth and, at an early time point, by positron emission tomography (PET) imaging of 18F-labeled glucose analog 18F-FDG. Results: Overall, there were no significant differences in tumor growth and survival between groups of mice receiving single-dose PTT and fractionated PTT in our study. Nonetheless, some animals did experience inhibited tumor growth or even complete tumor disappearance due to fractionated PTT, and these animals also showed a significant decrease in tumor uptake of 18F-FDG after therapy. Conclusion: This study only found an effect of giving PTT to tumors in fractions compared to a single-dose approach in a few animals. However, many factors can affect the outcome of PTT, and reliable tools for optimization of treatment protocol are needed. Despite the modest treatment effect, our results indicate that 18F-FDG PET/CT imaging can be useful to guide the number of treatment sessions necessary.


Assuntos
Hipertermia Induzida , Fototerapia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Feminino , Glicerol/química , Ouro/química , Temperatura Alta , Raios Infravermelhos , Camundongos Endogâmicos BALB C , Nanoconchas/química , Nanoconchas/ultraestrutura , Neoplasias/patologia , Neoplasias/terapia , Tomografia por Emissão de Pósitrons , Dióxido de Silício/química , Resultado do Tratamento , Carga Tumoral
8.
ACS Appl Mater Interfaces ; 11(30): 26637-26647, 2019 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-31276378

RESUMO

Stimulation of Toll-like receptors (TLRs) and/or NOD-like receptors on immune cells initiates and directs immune responses that are essential for vaccine adjuvants. The small-molecule TLR7 agonist, imiquimod, has been approved by the FDA as an immune response modifier but is limited to topical application due to its poor pharmacokinetics that causes undesired adverse effects. Nanoparticles are increasingly used with innate immune stimulators to mitigate side effects and enhance adjuvant efficacy. In this study, a potent small-molecule TLR7 agonist, 2-methoxyethoxy-8-oxo-9-(4-carboxybenzyl)adenine (1V209), was conjugated to hollow silica nanoshells (NS). Proinflammatory cytokine (IL-6, IL-12) release by mouse bone-marrow-derived dendritic cells and human peripheral blood mononuclear cells revealed that the potency of silica nanoshells-TLR7 conjugates (NS-TLR) depends on nanoshell size and ligand coating density. Silica nanoshells of 100 nm diameter coated with a minimum of ∼6000 1V209 ligands/particle displayed 3-fold higher potency with no observed cytotoxicity when compared to an unconjugated TLR7 agonist. NS-TLR activated the TLR7-signaling pathway, triggered caspase activity, and stimulated IL-1ß release, while neither unconjugated TLR7 ligands nor silica shells alone produced IL-1ß. An in vivo murine immunization study, using the model antigen ovalbumin, demonstrated that NS-TLR increased antigen-specific IgG antibody induction by 1000× with a Th1-biased immune response, compared to unconjugated TLR7 agonists. The results show that the TLR7 ligand conjugated to silica nanoshells is capable of activating an inflammasome pathway to enhance both innate immune-stimulatory and adjuvant potencies of the TLR7 agonist, thereby broadening applications of innate immune stimulators.


Assuntos
Imiquimode/imunologia , Imunidade Inata/efeitos dos fármacos , Imunoconjugados/imunologia , Receptor 7 Toll-Like/imunologia , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/farmacologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Humanos , Imiquimode/química , Imiquimode/uso terapêutico , Imunidade Inata/genética , Imunoconjugados/química , Imunoconjugados/uso terapêutico , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Camundongos , Nanoconchas/química , Transdução de Sinais/efeitos dos fármacos , Dióxido de Silício/química , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/genética
9.
J Cancer Res Clin Oncol ; 145(9): 2199-2209, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31309302

RESUMO

PURPOSE: Radiofrequency (RF) ablation therapy is of great interest in cancer therapy as it is non-ionizing radiation and can effectively penetrate into the tissue. However, the current RF ablation technique is invasive that requires RF probe insertion into the tissue and generates a non-specific heating. Recently, RF-responsive nanomaterials such as gold nanoparticles (AuNPs) and iron oxide nanoparticles (IONPs) have led to tremendous progress in this area. They have been found to be able to absorb the RF field and induce a localized heating within the target, thereby affording a non-invasive and tumor-specific RF ablation strategy. In the present study, for the first time, we used a hybrid core-shell nanostructure comprising IONPs as the core and AuNPs as the shell (IO@Au) for targeted RF ablation therapy. Due to the magnetic core, the nanohybrid can be directed toward the tumor through a magnet. Moreover, IONPs enable the nanohybrid to be used as a magnetic resonance imaging (MRI) contrast agent. RESULTS: In vitro cytotoxicity experiment showed that the combination of IO@Au and 13.56-MHz RF field significantly reduced the viability of cancer cells. Next, during an in vivo experiment, we demonstrated that magnetically targeting of IO@Au to the tumor and subsequent RF exposure dramatically suppressed the tumor growth. CONCLUSION: Therefore, the integration of targeting, imaging, and therapeutic performances into IO@Au nanohybrid could afford the promise to improve the effectiveness of RF ablation therapy.


Assuntos
Ablação por Cateter/métodos , Compostos Férricos/química , Ouro/química , Hipertermia Induzida/métodos , Nanopartículas de Magnetita/uso terapêutico , Neoplasias/cirurgia , Ablação por Radiofrequência/métodos , Animais , Compostos Férricos/uso terapêutico , Ouro/uso terapêutico , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita/química , Masculino , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Terapia de Alvo Molecular/métodos , Nanocompostos/química , Nanocompostos/uso terapêutico , Nanoconchas/química , Nanoconchas/uso terapêutico , Neoplasias/patologia , Células Tumorais Cultivadas
10.
J Exp Clin Cancer Res ; 38(1): 306, 2019 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-31299997

RESUMO

BACKGROUND: HER2-overexpressing metastatic breast cancers are challenging practice in oncology when they become resistant to anti-HER2 therapies such as trastuzumab. In these clinical situations, HER2-overexpression persists in metastatic localizations, and can thus be used for active targeting using innovative therapeutic approaches. Functionalized gold nanoparticles with anti-HER2 antibody can be stimulated by near-infrared light to induce hyperthermia. METHODS: Here, hybrid anti-HER2 gold nanoshells were engineered for photothermal therapy to overcome trastuzumab resistance in HER2-overexpressing breast cancer xenografts. RESULTS: When gold nanoshells were administered in HER2-tumor xenografts, no toxicity was observed. A detailed pharmacokinetic study showed a time-dependent accumulation of gold nanoshells within the tumors, significantly greater with functionalized gold nanoshells at 72 h. This enabled us to optimize the treatment protocol and irradiate the mice when the anti-HER2 gold nanoshells had accumulated most in the tumors. After weekly injections of anti-HER2 gold nanoshells, and repeated irradiations with a femtosecond-pulsed laser over four weeks, tumor growth was significantly inhibited. Detailed tissue microscopic analyses showed that the tumor growth inhibition was due to an anti-angiogenic effect, coherent with a preferential distribution of the nanoshells in tumor microvessels. We also showed a direct tumor cell effect with apoptosis and inhibition of proliferation, coherent with an immune-mediated targeting of tumor cells by anti-HER2 nanoshells. CONCLUSION: This preclinical study thus supports the use of anti-HER2 gold nanoshells and photothermal therapy to overcome trastuzumab resistance in HER2-overexpressing breast cancer.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Portadores de Fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Ouro , Lasers , Nanoconchas , Trastuzumab/administração & dosagem , Animais , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Dosagem de Genes , Regulação da Expressão Gênica , Ouro/química , Humanos , Imageamento por Ressonância Magnética , Camundongos , Nanoconchas/química , Neovascularização Patológica/tratamento farmacológico , Fototerapia , Receptor ErbB-2/genética , Dióxido de Silício/química , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Nanobiotechnology ; 17(1): 80, 2019 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-31277667

RESUMO

BACKGROUND: The gastric cancer is the second most malignant tumor in the world. HER-2 is one of the key targets for the gastric cancer therapy. Anti-HER-2 antibodies like trastuzumab, exhibits the satisfactory therapeutic effect in clinical. However, the drug resistance problem limits its application. METHOD: In this study, we develop a gold nanoshell (Gold Nanoshell) drug carrier for delivery and selective photo-thermal release of genes which target HER-2 and immunologic adjuvant CPG sequence in gastric tumor cells. The drug delivery system generated a multidimensional treatment strategy which includes gene-, immune- and photothermal-therapy. RESULTS: The whole gold nanoshell drug delivery system exhibits the well gene transduction ability and combined treatment effect. Both in vitro and in vivo results demonstrate the multiple therapeutic effects of the drug delivery system is better than the monotherapy. CONCLUSIONS: This study indicates the multiple combined therapy based on the gold nanoshell system would be a promising translational treatment for gastric cancer.


Assuntos
Ouro/química , Nanoconchas/química , Neoplasias Gástricas/terapia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Terapia Genética , Humanos , Hipertermia Induzida , Imunoterapia , Camundongos Nus , Terapia de Alvo Molecular , Fototerapia , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia
12.
Artif Cells Nanomed Biotechnol ; 47(1): 2316-2324, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31184218

RESUMO

Nowadays, gold nanoshells are used in targeted nano photothermal cancer therapy. This study surveyed the application of gold nanoshell (GNs) to thermal ablative therapy for melanoma cancer cells and it takes advantage of the near infrared absorption of gold nanoshells. The synthesis and characterization of glycosylated gold nanoshells (GGNs) were done. The cytotoxicity and photothermal effects of GNs on melanoma cells were evaluated using MTT assay and flow cytometry. The characterization data showed that GGNs are spherical, with a hydrodynamic size of 46.7 nm. Results suggest that the cellular uptake of GGNs was about 78%. Viability assays showed no significant toxicity at low concentrations of GNs. The higher heating rate and toxicity of cancer cells were obtained for the cells exposed to 808 nm NIR laser after incubation with GGNs rather than the GNs. The viability of these cells has dramatically decreased by 29%. Furthermore, 61% more cell lethality was achieved for A375 cells using combined photothermal therapy and treatment with GGNs in comparison to NIR radiation alone. In conclusion, our findings suggest that the synthesized gold/silica core-shell nanoparticles conjugated with glucosamine have high potentials to be considered as an efficient metal-nanoshell in the process of targeted cancer photothermal therapy.


Assuntos
Ouro/química , Ouro/farmacologia , Raios Infravermelhos , Melanoma/patologia , Nanoconchas/química , Fototerapia/métodos , Temperatura , Linhagem Celular Tumoral , Glicosilação , Humanos , Melanoma/terapia , Nanopartículas/química , Dióxido de Silício/química
13.
Mater Sci Eng C Mater Biol Appl ; 101: 505-512, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31029345

RESUMO

In this study, an antitumor drug delivery system, gold nanoshell coated wedelolactone liposomes (AuNS-Wed-Lip), were designed and synthesized. In the drug delivery system, wedelolactone liposome and gold-nanoshell were linked by l-cysteine, which had been shown an effective nanocarrier for antitumor drug delivery, on-demand drug release, and phototherapy under near-infrared (NIR) light irradiation. It was capable of absorbing 780-850 nm NIR light and converting light energy to heat rapidly. The hyperthermia promoted wedelolactone release rapidly from the systems. The release amount of AuNS-Wed-Lip under NIR irradiation reached up to 97.34% over 8 h, achieving the on-demand drug release. Moreover, a high inhibition rate up to 95.73% for 143B tumor cells by AuNS-Wed-Lip upon laser irradiation at 808 nm was observed. The excellent inhibition efficacy was also displayed in vivo antitumor study with S180 tumor-bearing mice. The results demonstrated that AuNS-Wed-Lip, as an antitumor drug delivery system, achieved chemo-photothermal synergetic effect, which has great potential in cancer therapy.


Assuntos
Antineoplásicos/química , Antineoplásicos/uso terapêutico , Cumarínicos/química , Cumarínicos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Ouro/química , Lipossomos/química , Nanoconchas/química , Animais , Camundongos , Fototerapia , Sarcoma/tratamento farmacológico
14.
Biomed Pharmacother ; 111: 1147-1155, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30841428

RESUMO

This article describes the work on synthesis, surface functionalization and characterization of hollow gold nanoshells (HGNs), and their application in a photothermal tumor therapy. The studied gold nanoshells were synthesized using a method based on a reduction of HAuCl4 onto silver nanoparticle templates. A selected aptamer - AS1411 - selective towards nucleolin, with a terminal thiol group was conjugated to HGNs. Surface functionalization of synthesized nanoparticles was necessary to gain the affinity to tumor cells, thus allowing the selective delivery of the modified nanoparticles to target cells. As expected, an increased selectivity of nanoconjugates towards tumor cells (A375) in a comparison to normal ones (HaCaT) was observed (2.67 times). In the framework of our studies the biological activity of the obtained nanoconjugates was evaluated using MTT assay. For this aim, both normal and tumor cell cultures respresenting skin tissue were exposed to gold nanoparticles solutions. It was found, that HGN-AS1411 conjugates cause less than 10% loss in a cell viability in the case of both cell lines at the highest tested concentration. The potential of the developed nanoconjugates as agents in PTT of skin cancer was investigated in a subsequent stage of our research. It was found, that A375 cell viability amounted to less than 40% for 75µM of nanoconjugates (expressed as a concentration of Au atoms) as a result of the laser irradiation of this cell culture after desired accumulation of the tested nanoconjugates inside A375 cells.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Nanoconchas/química , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Nanopartículas Metálicas/administração & dosagem , Nanoconchas/administração & dosagem , Fototerapia/métodos , Prata/química , Neoplasias Cutâneas/tratamento farmacológico , Nanomedicina Teranóstica/métodos
15.
Nano Lett ; 19(3): 2128-2137, 2019 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-30799620

RESUMO

Gallium and gallium-based alloys, typical types of liquid metals with unique physiochemical properties, are emerging as a next generation of functional materials in versatile biomedical applications. However, the exploration of their biomedical performance is currently insufficient, and their intrinsic low oxidative resistance is a key factor blocking their further clinical translation. Herein, we report on the surface engineering of liquid metal-based nanoplatforms by an inorganic silica nanoshell based on a novel but facile sonochemical synthesis for highly efficient, targeted, and near-infrared (NIR)-triggered photothermal tumor hyperthermia in the NIR-II biowindow. The inorganic silica-shell engineering of liquid metal significantly enhances the photothermal performance of the liquid metal core as reflected by enhanced NIR absorption, improved photothermal stability by oxidation protection, and abundant surface chemistry for surface-targeted engineering to achieve enhanced tumor accumulation. Systematic in vitro cell-level evaluation and in vivo tumor xenograft assessment demonstrate that (Arg-Gly-Asp) RGD-targeted and silica-coated nanoscale liquid metal substantially induces phototriggered cancer-cell death and photothermal tumor eradication, accompanied by high in vivo biocompatibility and easy excretion out of the body. This work provides the first paradigm for surface-inorganic engineering of liquid metal-based nanoplatforms for achieving multiple desirable therapeutic performances, especially for combating cancer.


Assuntos
Hipertermia Induzida/métodos , Nanoconchas/química , Neoplasias/terapia , Estresse Oxidativo/efeitos dos fármacos , Ligas/síntese química , Ligas/química , Ligas/farmacologia , Gálio/química , Gálio/farmacologia , Humanos , Compostos Inorgânicos/química , Líquidos Iônicos/química , Líquidos Iônicos/uso terapêutico , Nanoconchas/uso terapêutico , Dióxido de Silício/química
16.
ACS Appl Mater Interfaces ; 11(9): 8831-8837, 2019 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-30763070

RESUMO

As an alternative to traditional cancer treatment, photothermal therapy is a promising method with advantages such as noninvasiveness and high efficiency. Herein, we synthesized armored golden Escherichia coli (AGE) microrods as photothermal agents to evaluate the viability of cancer cell. The hollow gold nanoshell (HAuNS) was synthesized for photothermal effects under the near-infrared (NIR) region using unicellular E. coli as a framework. Coupling HAuNS onto the surface of E. coli@SiO2 enhanced temperature elevation and resulted in high conversion efficiency. The synthesized AGE microrods had excellent photothermal stability under NIR laser irradiation in the five-times recycling experiment. The temperature elevation of AGE microrod solution reached 43.7 °C, which induced hyperthermia-mediated killing of tumor cells. The results of the cytotoxicity test revealed the AGE microrod-induced T98G cell death mediated via apoptosis.


Assuntos
Ouro/química , Nanoconchas/química , Dióxido de Silício/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Escherichia coli/química , Escherichia coli/metabolismo , Humanos , Raios Infravermelhos , Nanoconchas/toxicidade , Fototerapia , Temperatura
17.
ACS Appl Mater Interfaces ; 11(7): 6777-6788, 2019 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-30668088

RESUMO

Recently, rodlike nanomaterials with specific aspect ratio for efficient cellular uptake have received enormous attention. For functional nanomaterials, such as photothermal agents, large surface areas for their rod-shaped exterior that increase the amount of light absorbed would lead to a higher absorption coefficient as well as drug-loading property. In this project, we coated rodlike mesoporous silica with gold nanoshells (MSNR@Au hybrid), modifying them with ultrasmall gadolinium (Gd)-chelated supramolecular photosensitizers, TPPS4 (MSNR@Au-TPPS4(Gd)), which could be applied to near-infrared fluorescence/multispectral optoacoustic tomography/computed tomography/magnetic resonance imaging and imaging-guided remotely controlled photothermal (PTT)/photodynamic (PDT) combined antitumor therapy. Gold nanoshells, as a perfect PTT agent, were used to assemble the rodlike mesoporous silica nanoparticles with larger superficial area and higher drug loading, thus obtaining the MSNR@Au hybrid. HS-ß-CD, which was used as the host, was adsorbed on the gold nanoshell (MSNR@Au-ß-CD) to link TPPS4(Gd) through the host-guest reaction, thus forming CD-TPPS4 supramolecular photosensitizers (supraPSs). Compared with conventional PSs, supraPSs have host screens, which could reduce the self-aggregation of TPPS4, and consequently generate 1O2 with high efficiency. The in vivo quadmodal imaging of MSNR@Au-TPPS4(Gd) nanoparticles revealed an intensive tumor uptake effect after injection. The in vivo antitumor efficacy further testified that the synergistic therapy, which was more efficient than any other monotherapy, exhibited an excellent tumor inhibition therapeutic effect. As a result, this encourages to further explore multifunctional theranostic nanoparticles based on gold shells for combined cancer therapy.


Assuntos
Meios de Contraste , Ouro , Hipertermia Induzida/métodos , Nanoconchas , Neoplasias Experimentais , Fotoquimioterapia , Fármacos Fotossensibilizantes , Dióxido de Silício , Tomografia Computadorizada por Raios X , Animais , Linhagem Celular Tumoral , Meios de Contraste/química , Meios de Contraste/farmacocinética , Meios de Contraste/farmacologia , Ouro/química , Ouro/farmacocinética , Ouro/farmacologia , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanoconchas/química , Nanoconchas/uso terapêutico , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/terapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacocinética , Fármacos Fotossensibilizantes/farmacologia , Dióxido de Silício/química , Dióxido de Silício/farmacocinética , Dióxido de Silício/farmacologia , Nanomedicina Teranóstica/métodos
18.
Nanotechnology ; 30(1): 015101, 2019 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-30370902

RESUMO

Various nanoformulations of perfluorocarbon have been developed thus far, to achieve ultrasound imaging of tumors and tumor-targeted therapy. However, their application has been greatly limited by their short sonographic duration and large size distribution. A novel theranostic agent was constructed based on gold nanoshell cerasome-encapsulated L-menthol (GNC-LM). Owing to the sustained and controllable generation of L-menthol bubbles under near-infrared laser irradiation, GNC-LM showed good performance in contrast enhancement of ultrasound imaging in vivo. GNC-LM could be imaged for 30 min, which is much longer than the imaging time of SonoVue (commercially used microbubbles). Moreover, photothermal therapy (PTT) based on the light-to-heat conversion of the nanosystem effectively ablated the tumor. Our study demonstrated the promising potential of the obtained GNC-LM to serve as a therapeutic nanoprobe for ultrasound contrast imaging and PTT of tumors.


Assuntos
Meios de Contraste/química , Ouro/química , Hipertermia Induzida , Lipídeos/química , Mentol/química , Nanoconchas/química , Neoplasias/terapia , Fototerapia , Animais , Morte Celular , Camundongos Endogâmicos BALB C , Nanoconchas/ultraestrutura , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Ultrassonografia
19.
J Mater Chem B ; 7(4): 598-610, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-32254793

RESUMO

With the rapid development of photothermal therapy (PTT) in cancer treatment, it is necessary to obtain effective plasma-responsive tunable photothermal transducing agents. Inspired by the peptide-directed hierarchical mineralized Ag nanocages (Ag NCs), scientists designed a new duel-template cascade preparation method, and novel unique multi-branched gold nanoshells (BGSs) were successfully prepared under mild conditions using green strategy. The length, density and diameter of the branches were tuned, which led to the adjustment of the surface plasma response of the nanostructure. Because of the hierarchical structure and anisotropic surface, an obvious red shift of the local surface plasmon resonance spectrum was observed for the branched Au nanoshells. The excellent photothermal conversion efficiency (70.9%) and photo-induced heating responsive curves proved the superior photothermal conversion performance and photothermal stability of BGSs. The in vitro and in vivo results indicated that the heat generated by the intense NIR absorption of BGSs can selectively destroy cancer cells under laser irradiation. The nanostructures with ultrastrong absorption have promising prospects in tumor therapy.


Assuntos
Nanoconchas , Neoplasias/terapia , Fototerapia/métodos , Animais , Sobrevivência Celular/efeitos dos fármacos , Ouro/química , Células HEK293 , Células HeLa , Humanos , Camundongos , Nanoconchas/química , Nanoconchas/uso terapêutico , Fenômenos Físicos , Prata/química
20.
Biosens Bioelectron ; 126: 214-221, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30423478

RESUMO

Herein, we present the research focused on the synthesis and application of aptamer-modified gold nanoshells for photothermal therapy (PTT). NIR-absorbing hollow gold nanoshells were synthetized and conjugated with anti-MUC1 aptamer (HGNs@anti-MUC1). MUC1 (Mucin 1) is a transmembrane glycoprotein, which is overexpressed in a variety of epithelial cancers (eg. breast, lung, pancreatic). In order to evaluate the efficiency of PTT with HGNs@anti-MUC1 we used 3D cell culture model - multicellular spheroids. The selected cell culture model is considered as the best in vitro model for cancer research (similar morphology, metabolite and oxygen gradients, cellular interactions and cell growth kinetics in the spheroids are similar to the early stage of a nonvascular tumor). We conducted our research on human normal (MRC-5, MCF-10A) and tumor (A549, MCF-7) cell lines using a microfluidic system. Aptamer-modified nanoparticles were accumulated selectively in tumor cells (A549, MCF-7) and this fact contributed to the reduction of tumor spheroids viability and size. It should be underlined, that it is the first example of photothermal therapy carried out in a microsystem on multicellular spheroids.


Assuntos
Aptâmeros de Peptídeos/química , Técnicas Biossensoriais , Mucina-1/química , Neoplasias/diagnóstico , Células A549 , Aptâmeros de Peptídeos/farmacologia , Proliferação de Células/efeitos dos fármacos , Humanos , Células MCF-7 , Microfluídica , Mucina-1/genética , Nanoconchas/química , Neoplasias/patologia , Fototerapia , Esferoides Celulares/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA