Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
BME Front ; 4: 0021, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37849674

RESUMO

The wide and frequent use of antibiotics in the treatment of bacterial infection can cause the occurrence of multidrug-resistant bacteria, which becomes a serious health threat. Therefore, it is necessary to develop antibiotic-independent treatment modalities. Chemodynamic therapy (CDT) is defined as the approach employing Fenton and/or Fenton-like reactions for generating hydroxyl radical (•OH) that can kill target cells. Recently, CDT has been successfully employed for antibacterial applications. Apart from the common Fe-mediated CDT strategy, antibacterial CDT strategies mediated by other metal elements such as copper, manganese, cobalt, molybdenum, platinum, tungsten, nickel, silver, ruthenium, and zinc have also been proposed. Furthermore, different types of materials like nanomaterials and hydrogels can be adopted for constructing CDT-involved antibacterial platforms. Besides, CDT can introduce some toxic metal elements and then achieve synergistic antibacterial effects together with reactive oxygen species. Finally, CDT can be combined with other therapies such as starvation therapy, phototherapy, and sonodynamic therapy for achieving improved antibacterial performance. This review first summarizes the advancements in antibacterial CDT and then discusses the present limitations and future research directions in this field, hoping to promote the development of more effective materials and strategies for achieving potentiated CDT.

2.
J Control Release ; 351: 692-702, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36150580

RESUMO

Photodynamic therapy (PDT) is a robust cancer treatment modality, and the precise spatiotemporal control of its subcellular action site is crucial for its effectiveness. However, accurate comparison of the efficacy of different organelle-targeted PDT approaches is challenging since it is difficult to find a single system that can achieve separate targeting of different organelles with separable time windows and similar binding amounts. Herein, we conjugated chlorin e6 (Ce6) with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-5000] (ammonium salt) (DSPE-PEG5000-NH2) to afford DSPE-PEG-Ce6, which could migrate from mitochondrion to lysosome and ultimately to endoplasmic reticulum (ER) after cellular internalization. Benefiting from the dynamic subcellular distribution of DSPE-PEG-Ce6 with tunable organelle-binding amounts, we accurately determined the PDT efficacy order of the molecule, i.e., mitochondrion > ER > lysosome. This work proposes an ideal model system for accurately evaluating the specific organelle-targeted PDT efficacy and may promote the future development of effective PDT strategies.


Assuntos
Fotoquimioterapia , Porfirinas , Fototerapia , Retículo Endoplasmático/metabolismo , Lisossomos/metabolismo , Mitocôndrias , Fármacos Fotossensibilizantes/química , Linhagem Celular Tumoral
3.
Small ; 18(6): e2103868, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34729913

RESUMO

Chemodynamic therapy (CDT), a novel cancer therapeutic strategy defined as the treatment using Fenton or Fenton-like reaction to produce •OH in the tumor region, was first proposed by Bu, Shi, and co-workers in 2016. Recently, with the rapid development of Fenton and Fenton-like nanomaterials, CDT has attracted tremendous attention because of its unique advantages: 1) It is tumor-selective with low side effects; 2) the CDT process does not depend on external field stimulation; 3) it can modulate the hypoxic and immunosuppressive tumor microenvironment; 4) the treatment cost of CDT is low. In addition to the Fe-involved CDT strategies, the Fenton-like reaction-mediated CDT strategies have also been proposed, which are based on many other metal elements including copper, manganese, cobalt, titanium, vanadium, palladium, silver, molybdenum, ruthenium, tungsten, cerium, and zinc. Moreover, CDT has been combined with other therapies like chemotherapy, radiotherapy, phototherapy, sonodynamic therapy, and immunotherapy for achieving enhanced anticancer effects. Besides, there have also been studies that extend the application of CDT to the antibacterial field. This review introduces the latest advancements in the nanomaterials-involved CDT from 2018 to the present and proposes the current limitations as well as future research directions in the related field.


Assuntos
Nanoestruturas , Neoplasias , Linhagem Celular Tumoral , Cobre , Humanos , Peróxido de Hidrogênio , Imunoterapia , Neoplasias/tratamento farmacológico , Microambiente Tumoral
4.
Artigo em Inglês | MEDLINE | ID: mdl-33860622

RESUMO

Chemotherapy typically involves the use of specific chemodrugs to inhibit the proliferation of cancer cells, but the frequent emergence of a variety of multidrug-resistant cancer cells poses a tremendous threat to our combat against cancer. The fundamental causes of multidrug resistance (MDR) have been studied for decades, and can be generally classified into two types: one is associated with the activation of diverse drug efflux pumps, which are responsible for translocating intracellular drug molecules out of the cells; the other is linked with some non-efflux pump-related mechanisms, such as antiapoptotic defense, enhanced DNA repair ability, and powerful antioxidant systems. To overcome MDR, intense efforts have been made to develop synergistic therapeutic strategies by introducing MDR inhibitors or combining chemotherapy with other therapeutic modalities, such as phototherapy, gene therapy, and gas therapy, in the hope that the drug-resistant cells can be sensitized toward chemotherapeutics. In particular, nanotechnology-based drug delivery platforms have shown the potential to integrate multiple therapeutic agents into one system. In this review, the focus was on the recent development of nanostrategies aiming to enhance the efficiency of chemotherapy and overcome the MDR of cancer in a synergistic manner. Different combinatorial strategies are introduced in detail and the advantages as well as underlying mechanisms of why these strategies can counteract MDR are discussed. This review is expected to shed new light on the design of advanced nanomedicines from the angle of materials and to deepen our understanding of MDR for the development of more effective anticancer strategies. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.


Assuntos
Antineoplásicos , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Nanomedicina , Neoplasias , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico
5.
Angew Chem Int Ed Engl ; 60(25): 14013-14021, 2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-33768682

RESUMO

The presence of bacteria in the tumor can cause cancer resistance to chemotherapeutics. To fight against bacterium-induced drug resistance, herein we design self-traceable nanoreservoirs that are simultaneously loaded with gemcitabine (an anticancer drug) and ciprofloxacin (an antibiotic) and are decorated with hyaluronic acid for active tumor targeting. The nanoreservoirs have a pH-sensitive gate and an enzyme-responsive gate that can be opened in the acidic and hyaluronidase-abundant tumor microenvironment to control drug release rates. Moreover, the nanoreservoirs can specifically target the tumor regions without eliciting evident toxicity to normal tissues, kill the intratumoral bacteria, and inhibit the tumor growth even in the presence of the bacteria. Unexpectedly, the nanoreservoirs can activate T cell-mediated immune responses through promoting antigen-presenting dendritic cell maturation and depleting immunosuppressive myeloid-derived suppressor cells in bacterium-infected tumors.


Assuntos
Antibacterianos/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Ciprofloxacina/farmacologia , Neoplasias do Colo/terapia , Desoxicitidina/análogos & derivados , Escherichia coli/efeitos dos fármacos , Animais , Antibacterianos/química , Antimetabólitos Antineoplásicos/química , Linhagem Celular , Ciprofloxacina/química , Neoplasias do Colo/microbiologia , Desoxicitidina/química , Desoxicitidina/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Humanos , Camundongos , Gencitabina
6.
ACS Appl Mater Interfaces ; 12(33): 36882-36894, 2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32666795

RESUMO

Clinically approved doxorubicin (Dox)-loaded liposomes (e.g., Doxil) guarantee good biosafety, but their insufficient nuclear delivery of Dox (<0.4%) after cellular uptake significantly hampers their final anticancer efficacy. Here, we report that simply doping protoporphyrin IX (PpIX, a commonly used hydrophobic photosensitizer) into the lipid bilayers of Dox-loaded liposomes (the resultant product is termed PpIX/Dox liposomes) is a feasible way to promote the nuclear delivery of Dox. This facile strategy relies on a unique property of PpIX-it presents considerably higher affinity for the real plasma membrane over its liposomal carrier, which drives the doped PpIX molecules to detach from the liposomes when encountering cancer cells. We demonstrate that this process can trigger the efficient release of the loaded Dox molecules and allow them to enter the nuclei of MCF-7 breast cancer cells without being trapped by lysosomes. Regarding the drug-resistant MCF-7/ADR cells, the aberrant activation of the efflux pumps in the plasma membranes expels the internalized Dox. However, we strikingly find that the robust drug resistance can be reversed upon mild laser irradiation because the photodynamic effect of PpIX disrupts the drug efflux system (e.g., P-glycoprotein) and facilitates the nuclear entry of Dox. As a proof-of-concept, this PpIX doping strategy is also applicable for enhancing the effectiveness of cisplatin-loaded liposomes against both A549 and A549/DDP lung cancer cells. In vivo experimental results prove that a single injection of PpIX/Dox liposomes completely impedes the growth of MCF-7 tumors in nude mice within 2 weeks and, in combination with laser irradiation, can synergistically ablate MCF-7/ADR tumors. Biosafety assessments reveal no significant systemic toxicity caused by PpIX/Dox liposomes. This work exemplifies a facile method to modulate the subcellular fate of liposomal drugs and may inspire the optimization of nanopharmaceuticals in the near future.


Assuntos
Antineoplásicos/química , Doxorrubicina/análogos & derivados , Lipossomos/química , Fármacos Fotossensibilizantes/química , Protoporfirinas/química , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Núcleo Celular/metabolismo , Colesterol/química , Terapia Combinada , Doxorrubicina/química , Doxorrubicina/farmacologia , Liberação Controlada de Fármacos , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Hipertermia Induzida , Lipossomos/metabolismo , Camundongos Nus , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Protoporfirinas/farmacologia , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Biomaterials ; 232: 119668, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31927179

RESUMO

Mitophagy is a specific self-protective autophagic process that degrades damaged or dysfunctional mitochondria, and is generally considered to reduce the effectiveness of mitochondria-targeted therapies. Here, we report an energy depletion-based anticancer strategy by selectively activating excessive mitophagy in cancer cells. We fabricate a type of mitochondria-targeting nanomicelles via the self-assembly of D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) and dc-IR825 (a near-infrared cyanine dye and a photothermal agent). The TPGS/dc-IR825 nanomicelles enable mitochondrial damage in cancer cells, which, for self-protection, activate two autophagic pathways, (1) mitophagy and (2) adenosine triphosphate (ATP) shortage-triggered autophagy. However, the excessive mitophagy/autophagy activities far surpass the degradative capacity of autolysosomes, leading to the formation of micrometer-sized vacuoles and degradation blockage. Immunofluorescence staining and Western blot analysis reveal that the nanomicelle-treated cancer cells are under severe ATP shortage, which eventually causes substantial cell death. Moreover, the nanomicelles intravenously injected into tumor-bearing mice show high tumor accumulation, long tumor retention, and inhibit the tumor growth by inducing excessive mitophagy/autophagy and energy depletion in tumor cells. Additional near-infrared laser irradiation treatment further enhances the in vitro and in vivo anticancer efficiencies of the nanomicelles, due to the excellent photothermal and photodynamic effects of dc-IR825. We believe that this work highlights the important role of mitophagy/autophagy in treating cancers.


Assuntos
Mitofagia , Nanoestruturas , Neoplasias , Fototerapia , Animais , Autofagia , Lisossomos/metabolismo , Camundongos , Micelas , Mitocôndrias , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
8.
Nanoscale ; 12(1): 210-219, 2020 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-31815993

RESUMO

Nanoparticle (NP)-based hydrogels that can introduce synergistic advantages to the novel three-dimensional scaffold have garnered much attention recently. However, the application of NP-crosslinked hydrogels still remains challenging due to the complicated synthesis and/or modification of the NPs and the changed properties of the NPs after gelation. Herein, a novel palladium nanosheet (Pd NS)-based hydrogel (Pd Gel) with Pd NSs as crosslinkers was obtained by simply mixing Pd NSs with thiol-terminated four-arm polyethylene glycol (4arm-PEG-thiol). It was found that the formed Pd Gel was injectable, possibly due to the dynamic Pd-S bonds formed between Pd NSs and 4arm-PEG-thiol. In addition, compared with free Pd NSs, the Pd NSs within the hydrogel exhibited a significantly higher stability. We have further demonstrated that the formed hydrogel could encapsulate the commonly used anticancer drug doxorubicin (DOX) to form DOX@Pd Gel for combined chemo-photothermal therapy. Particularly, Pd NSs with a high absorption in the near-infrared (NIR) region could convert the energy of NIR laser into heat with a high efficiency, which is beneficial for photothermal therapy. Moreover, DOX@Pd Gel could maintain a sustainable release of DOX and the NIR laser irradiation could accelerate this drug release process. Then, the explosively released DOX and the hyperthermia generated from Pd NSs under NIR laser irradiation acted in a synergistic way to realize the combined therapeutic effect of the chemo-photothermal treatment. Finally, the in vivo anticancer effect and safety of the combined therapy were also verified by the tumor-bearing mouse model. Taken together, this work constructs a NP-crosslinked, NIR laser-activatable and injectable photothermal hydrogel via dynamic Pd-S bonding, and demonstrates that the hydrogel allows us to release DOX more precisely, eliminate tumor more effectively and inhibit tumor metastasis more persistently, which will advance the development of novel anticancer strategies.


Assuntos
Antineoplásicos/uso terapêutico , Hidrogéis/química , Nanoestruturas/química , Neoplasias/terapia , Paládio/química , Enxofre/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Terapia Combinada , Doxorrubicina/química , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Raios Infravermelhos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fototerapia , Polietilenoglicóis/química , Espécies Reativas de Oxigênio/metabolismo
9.
Biomaterials ; 223: 119443, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31541779

RESUMO

Tumor metastasis is a key cause that leads to the failure of cancer treatment. Inhibition of metastasis, rather than the simple removal of the primary tumor, is critical to the survival improvement. Here, we report a cell-penetrating peptide-modification strategy to realize substantial perinuclear accumulation and subsequent near-infrared (NIR) laser-triggered nuclear entry of palladium nanosheets (Pd NSs) for inhibition of cancer cell metastasis and photothermal cancer therapy. Specifically, it was found that the cell-penetrating peptide TAT-modified Pd NSs (abbreviated as Pd-TAT) mainly accumulated in the perinuclear region and showed the enhanced endocytosis and reduced efflux compared with the counterpart without TAT modification. On the one hand, Pd-TAT could inhibit cell migration and invasion. It was proposed that Pd-TAT located in the perinuclear region could promote the overexpression of lamin A/C proteins (related with nuclear stiffness) and increase the mechanical stiffness of the nucleus. More importantly, the introduction of NIR laser irradiation with a laser density of 0.3 W/cm2 (below the permitted value 0.329 W/cm2 for skin exposure) significantly enhanced the inhibitory effect of Pd-TAT on cancer cell migration, which might be due to the increased nuclear stiffness caused by the enhanced nuclear entry of Pd-TAT under the effect of mild laser-induced local hyperthermia in the perinuclear region. On the other hand, the increased nuclear entry of Pd-TAT under NIR laser irradiation greatly enhanced their photothermal therapeutic efficacy due to the susceptibility of the nucleus to hyperthermia. Taken together, the Pd-TAT-based and laser-promoted perinuclear-to-intranuclear localization strategy allows us to not only destroy the primary tumor more effectively, but also inhibit cancer metastasis more persistently.


Assuntos
Movimento Celular/efeitos da radiação , Peptídeos Penetradores de Células/uso terapêutico , Hipertermia Induzida , Nanopartículas/uso terapêutico , Neoplasias/terapia , Fototerapia , Animais , Peso Corporal , Linhagem Celular Tumoral , Humanos , Raios Infravermelhos , Lasers , Células MCF-7 , Camundongos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Paládio/química
10.
Small ; 15(33): e1900501, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31282114

RESUMO

Photothermal therapy (PTT) usually requires hyperthermia >50 °C for effective tumor ablation, which inevitably induces heating damage to the surrounding normal tissues/organs. Moreover, low tumor retention and high liver accumulation are the two main obstacles that significantly limit the efficacy and safety of many nanomedicines. To solve these problems, a smart albumin-based tumor microenvironment-responsive nanoagent is designed via the self-assembly of human serum albumin (HSA), dc-IR825 (a cyanine dye and a photothermal agent), and gambogic acid (GA, a heat shock protein 90 (HSP90) inhibitor and an anticancer agent) to realize molecular targeting-mediated mild-temperature PTT. The formed HSA/dc-IR825/GA nanoparticles (NPs) can escape from mitochondria to the cytosol through mitochondrial disruption under near-infrared (NIR) laser irradiation. Moreover, the GA molecules block the hyperthermia-induced overexpression of HSP90, achieving the reduced thermoresistance of tumor cells and effective PTT at a mild temperature (<45 °C). Furthermore, HSA/dc-IR825/GA NPs show pH-responsive charge reversal, effective tumor accumulation, and negligible liver deposition, ultimately facilitating synergistic mild-temperature PTT and chemotherapy. Taken together, the NIR-activated NPs allow the release of molecular drugs more precisely, ablate tumors more effectively, and inhibit cancer metastasis more persistently, which will advance the development of novel mild-temperature PTT-based combination strategies.


Assuntos
Albuminas/administração & dosagem , Hipertermia Induzida/métodos , Terapia de Alvo Molecular , Fototerapia/métodos , Células A549 , Albuminas/farmacocinética , Animais , Terapia Combinada , Endocitose , Humanos , Camundongos , Nanopartículas/uso terapêutico , Neoplasias/terapia , Temperatura , Distribuição Tecidual
11.
Small ; 15(11): e1804575, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30761748

RESUMO

Herein, water-dispersible carbon nano-onion clusters (CNOCs) with an average hydrodynamic size of ≈90 nm are prepared by simply sonicating candle soot in a mixture of oxidizing acid. The obtained CNOCs have high photothermal conversion efficiency (57.5%), excellent aqueous dispersibility (stable in water for more than a year without precipitation), and benign biocompatibility. After polyethylenimine (PEI) and poly(ethylene glycol) (PEG) modification, the resultant CNOCs-PEI-PEG have a high photothermal conversion efficiency (56.5%), and can realize after-wash photothermal cancer cell ablation due to their ultrahigh cellular uptake (21.3 pg/cell), which is highly beneficial for the selective ablation of cancer cells via light-triggered intracellular heat generation. More interestingly, the cellular uptake of CNOCs-PEI-PEG is so high that the internalized nanoagents can be directly observed under a microscope without fluorescent labeling. Besides, in vivo experiments reveal that CNOCs-PEI-PEG can be used for photothermal/photoacoustic dual-modal imaging-guided photothermal therapy after intravenous administration. Furthermore, CNOCs-PEI-PEG can be efficiently cleared from the mouse body within a week, ensuring their excellent long-term biosafety. To the best of the authors' knowledge, the first example of using candle soot as raw material to prepare water-dispersible onion-like carbon nanomaterials for cancer theranostics is represented herein.


Assuntos
Carbono/química , Diagnóstico por Imagem , Hipertermia Induzida , Nanoestruturas/química , Neoplasias/terapia , Fototerapia , Fuligem/química , Água/química , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Nanoestruturas/ultraestrutura , Técnicas Fotoacústicas , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Polietilenoimina/síntese química , Polietilenoimina/química , Temperatura
12.
Acta Biomater ; 88: 462-476, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30735810

RESUMO

Lysosomal entrapment and liver accumulation are the two main obstacles faced by many anticancer drugs for achieving satisfactory therapeutic outcomes. Here, we develop a facile one-step hydrothermal synthetic route to prepare trace metal (M)-, N-, and O-doped carbon-dominated nanoparticles (termed as MNOCNPs, M = Ni, Pd, or Cu, metal content: <0.1 mol%) with exceptional photothermal properties (e.g., the ultrahigh extinction coefficient of 32.7 L g-1 cm-1), which can simultaneously realize preferable endoplasmic reticulum (ER) targeting and specific tumor enrichment without noticeable liver accumulation after poly(ethylene glycol) (PEG) conjugation. More interestingly, the PEG-modified MNOCNPs with nanoscale lengths exhibit considerable nucleolar delivery and increased tumor accumulation upon laser irradiation. After fluorescence labeling, these PEG-modified MNOCNPs are suitable for fluorescence/photoacoustic/thermal triple-modal imaging-guided photothermal cancer treatment. Additionally, the ultralow metal content ensures the exceptional biosafety of the nanoagents. The present work provides a novel, facile, and general synthetic method of carbon-dominated nanoparticles with superior photothermal properties for highly efficient tumor ablation, and the large-organelle (ER and nucleus)-targeted cancer therapeutic strategy may represent an alternative solution for optimizing the anticancer efficacy of nanomaterials. STATEMENT OF SIGNIFICANCE: Limited wire-like nanomaterials have been used for biomedical applications due to their lack of intrinsic photothermal properties, poor cellular uptake and tumor accumulation, and potential biotoxicity arising from their micrometer lengths and/or massive heavy metal doping. Besides, the clinical applications of many nanoagents are hindered by their tendency to accumulate in liver, which may cause severe liver toxicity. Herein, we develop for the first time a one-step hydrothermal method to prepare wire-like trace metal-, N-, and O-doped carbon-dominated nanoparticles with excellent photothermal properties, massive cellular uptake, preferable ER localization, selective tumor targeting with negligible liver deposition, laser irradiation-enhanced nucleolar delivery and tumor accumulation, and multimodal imaging-guided cancer therapy. This work opens a new window for simultaneously overcoming lysosomal entrapment and liver accumulation in cancer therapy.


Assuntos
Carbono/química , Nucléolo Celular/metabolismo , Retículo Endoplasmático/metabolismo , Lasers , Nanopartículas/química , Neoplasias/terapia , Fototerapia , Oligoelementos/química , Animais , Coloides/química , Feminino , Células HeLa , Humanos , Hipertermia Induzida , Camundongos Nus , Nanopartículas/ultraestrutura , Neoplasias/patologia , Técnicas Fotoacústicas , Polietilenoglicóis/química , Distribuição Tecidual
13.
ACS Appl Mater Interfaces ; 10(49): 42077-42087, 2018 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-30403472

RESUMO

Integration of multiple diagnostic/therapeutic modalities into a single system with ultrasmall size, excellent photothermal/photodynamic properties, high cellular uptake efficiency, nuclear delivery capacity, rapid renal clearance, and good biosafety is highly desirable for cancer theranostics, but still remains challenging. Here, a novel type of multifunctional nanodots (denoted as BCCGH) was synthesized by mixing bovine serum albumin, carbon dots, and metal ions (Cu2+ and Gd3+), followed by the conjugation with a photosensitizer (HPPH). The nanodots hold great promise for fluorescence/photoacoustic/magnetic resonance/photothermal imaging-guided synergistic photothermal/photodynamic therapy (PDT) because of their appealing properties such as high photothermal conversion efficiency (68.4%), high longitudinal relaxivity (11.84 mM-1 s-1, 7 T), and superior colloidal stability with negligible Gd3+ release. Benefiting from the massive cellular uptake, endoplasmic reticulum/mitochondrion-targeting ability, and mild near-infrared laser irradiation-promoted nuclear delivery of BCCGH, a high anticancer therapeutic efficiency is achieved in the subsequent in vitro PDT. Besides, as revealed by the in vivo/ex vivo results, the nanodots also exhibit excellent tumor accumulation, efficient renal clearance, complete tumor ablation, and exceptional biosafety. To summarize, this work develops a carbon dot-mediated and albumin-based synthetic approach for constructing ultrasmall and multifunctional nanodots, which may hold great potential for cancer theranostics and beyond.


Assuntos
Imagem Multimodal , Neoplasias Experimentais/tratamento farmacológico , Fotoquimioterapia , Pontos Quânticos , Soroalbumina Bovina/química , Animais , Carbono , Linhagem Celular Tumoral , Humanos , Camundongos , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Pontos Quânticos/química , Pontos Quânticos/uso terapêutico , Soroalbumina Bovina/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Molecules ; 23(11)2018 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-30453692

RESUMO

Mitochondria-targeted cancer phototherapy (PT), which works by delivering photoresponsive agents specifically to mitochondria, is a powerful strategy to improve the phototherapeutic efficiency of anticancer treatments. Mitochondria play an essential role in cellular apoptosis, and are relevant to the chemoresistance of cancer cells. Furthermore, mitochondria are a major player in many cellular processes and are highly sensitive to hyperthermia and reactive oxygen species. Therefore, mitochondria serve as excellent locations for organelle-targeted phototherapy. In this review, we focus on the recent advances of mitochondria-targeting materials for mitochondria-specific PT. The combination of mitochondria-targeted PT with other anticancer strategies is also summarized. In addition, we discuss both the challenges currently faced by mitochondria-based cancer PT and the promises it holds.


Assuntos
Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Terapia de Alvo Molecular , Organelas/metabolismo , Fotoquimioterapia , Fototerapia , Nanomedicina Teranóstica , Animais , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacologia , Terapia Combinada , Humanos , Mitocôndrias/imunologia , Nanopartículas/química , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Organelas/efeitos dos fármacos , Organelas/imunologia , Peptídeos/química , Peptídeos/farmacologia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Tolerância a Radiação
15.
Biomaterials ; 183: 30-42, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30149228

RESUMO

Tumor growth and metastasis are two main causes of cancer-related deaths. Here, we simultaneously investigated the effects of nanoparticles on cancer cell viability and migration using polyethylene glycol (PEG)-modified, platinum-doped (<4 mol %) carbon nanoparticles (denoted as PEG-PtCNPs). The bare PtCNPs were prepared by the facile one-step hydrothermal treatment of p-phenylenediamine and K2PtCl4 in aqueous solution. After PEGylation, the obtained PEG-PtCNPs can serve as an excellent photothermal nanoagent for cell migration inhibition, laser-triggered nuclear delivery, effective tumor accumulation, and imaging-guided tumor ablation with improved therapeutic efficacy and reduced side effects. In the absence of laser exposure, the positively charged PEG-PtCNPs with a hydrodynamic diameter of ∼19 nm easily entered the cells by endocytosis and were located in multiple organelles (including mitochondrion, endoplasmic reticulum, lysosome, and Golgi apparatus), causing a slight increase in the expression level of nuclear protein lamin A/C. Upon mild laser irradiation (0.3 W cm-2), the fragmented cytoskeletal structures and overexpression of lamin A/C were observed, thus inhibiting cancer cell migration. Furthermore, hyperthermia induced by PEG-PtCNPs plus laser irradiation at a higher power density (1.0 W cm-2) could cause irreversible damage to the nuclear membranes and then facilitate the nuclear delivery of the nanoagents without the introduction of nuclear targeting ligands. Taken together, this work develops a facile synthetic approach of platinum-based carbon nanoparticles with excellent photothermal properties, and demonstrates their potential applications for modulating tumor metastasis and realizing multi-organelle-targeted tumor ablation.


Assuntos
Carbono/química , Nanopartículas/química , Organelas/metabolismo , Platina/química , Animais , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular , Feminino , Xenoenxertos , Humanos , Hipertermia Induzida , Lamina Tipo A/metabolismo , Lasers , Camundongos Nus , Fototerapia/métodos , Polietilenoglicóis/química , Distribuição Tecidual
16.
Nanoscale ; 10(4): 2115-2127, 2018 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-29326993

RESUMO

Developing biocompatible and photodegradable photothermal agents (PTAs) holds great promise for potential clinical applications in photothermal cancer therapy. Herein, a new PTA was innovatively constructed by conjugating the hydrophobic near-infrared (NIR) heptamethine cyanine molecule IR825-NH2 with a double hydrophilic block copolymer methoxypoly(ethylene glycol)5k-block-poly(l-aspartic acid sodium salt)10 (abbreviated as PEG-PLD) via amine-carboxyl reaction. The as-designed PEG-PLD(IR825) was amphiphilic and could self-assemble into polymeric nanomicelles in aqueous solutions. Benefiting from the chemical conjugation strategy, PEG-PLD(IR825) nanomicelles realized a considerably high drug loading rate (∼21.0%) and substantially avoided the premature release of IR825 during systemic circulation. Confocal imaging revealed that the nanomicelles mainly located at mitochondria and endoplasmic reticulum after cellular internalization. In vitro photothermal therapy demonstrated the excellent cancer killing efficiency of PEG-PLD(IR825) nanomicelles due to their high light-to-heat conversion efficiency upon NIR laser irradiation. In addition, PEG-PLD(IR825) nanomicelles showed polarity-sensitive fluorescence at ∼610 nm (under 552 nm excitation) and 830 nm (under 780 nm excitation), which was especially useful for both in vitro visible fluorescence imaging and in vivo near-infrared fluorescence imaging-guided photothermal therapy (PTT). At the in vivo level, PEG-PLD(IR825) nanomicelles exhibited an excellent tumor-homing ability and a long retention time in tumor tissues as evidenced by the in vivo fluorescence imaging results. The desirable properties of PEG-PLD(IR825) nanomicelles ensured their effective tumor ablation during PTT treatment. More importantly, the PEG-PLD(IR825) nanomicelles underwent degradation after laser irradiation, which ensured their post-treatment biosafety. Therefore, the nanomicelles are promising to serve as an efficient and safe PTA for imaging-guided photothermal cancer therapy.


Assuntos
Hipertermia Induzida , Micelas , Nanopartículas , Neoplasias/terapia , Animais , Feminino , Células HeLa , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias , Polímeros , Ensaios Antitumorais Modelo de Xenoenxerto
17.
ACS Appl Mater Interfaces ; 10(2): 1544-1555, 2018 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-29260843

RESUMO

Copper-containing nanomaterials have been applied in various fields because of their appealing physical, chemical, and biomedical properties/functions. Herein, for the first time, a facile, room-temperature, and one-pot method of simply mixing copper ions and sulfur-doped carbon dots (CDs) is developed for the synthesis of copper/carbon quantum dot (or CD)-crosslinked nanosheets (CuCD NSs). The thus-obtained CuCD NSs with the size of 20-30 nm had a high photothermal conversion efficiency of 41.3% and good photothermal stability. Especially, after coating with thiol-polyethylene glycol and fluorescent molecules, the resultant CuCD NSs could selectively target tumor tissues and realize multimodal (photoacoustic, photothermal, and fluorescence) imaging-guided cancer therapy. More importantly, our CuCD NSs exhibited laser-triggered cytosolic delivery, lysosomal escape, and nuclear-targeting properties, which greatly enhanced their therapeutic efficacy. The significantly enhanced tumor accumulation of CuCD NSs after in situ tumor-site laser irradiation was also observed in in vivo experiments. These in vitro and in vivo events occurring during the continuous laser irradiation have not been observed. Overall, this work develops a CD-assisted synthetic method of photothermal nanoagents for triple-modal imaging-guided phototherapy and deepens our understanding of the action mechanism of photothermal therapy, which will promote the development of nanomedicine and beyond.


Assuntos
Neoplasias/terapia , Carbono , Cobre , Humanos , Imagem Multimodal , Nanoestruturas , Fototerapia , Pontos Quânticos
18.
ACS Appl Mater Interfaces ; 8(42): 28480-28494, 2016 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-27689441

RESUMO

Metallic nanostructures as excellent candidates for nanosensitizers have shown enormous potentials in cancer radiotherapy and photothermal therapy. Clinically, a relatively low and safe radiation dose is highly desired to avoid damage to normal tissues. Therefore, the synergistic effect of the low-dosed X-ray radiation and other therapeutic approaches (or so-called "combined therapeutic strategy") is needed. Herein, we have synthesized hollow and spike-like gold nanostructures by a facile galvanic replacement reaction. Such gold nanospikes (GNSs) with low cytotoxicity exhibited high photothermal conversion efficiency (η = 50.3%) and had excellent photostability under cyclic near-infrared (NIR) laser irradiations. We have demonstrated that these GNSs can be successfully used for in vitro and in vivo X-ray radiation therapy and NIR photothermal therapy. For the in vitro study, colony formation assay clearly demonstrated that GNS-mediated photothermal therapy and X-ray radiotherapy reduced the cell survival fraction to 89% and 51%, respectively. In contrast, the cell survival fraction of the combined radio- and photothermal treatment decreased to 33%. The synergistic cancer treatment performance was attributable to the effect of hyperthermia, which efficiently enhanced the radiosensitizing effect of hypoxic cancer cells that were resistant to ionizing radiation. The sensitization enhancement ratio (SER) of GNSs alone was calculated to be about 1.38, which increased to 1.63 when the GNS treatment was combined with the NIR irradiation, confirming that GNSs are effective radiation sensitizers to enhance X-ray radiation effect through hyperpyrexia. In vivo tumor growth study indicated that the tumor growth inhibition (TGI) in the synergistically treated group reached 92.2%, which was much higher than that of the group treated with the GNS-enhanced X-ray radiation (TGI = 29.8%) or the group treated with the GNS-mediated photothermal therapy (TGI = 70.5%). This research provides a new method to employ GNSs as multifunctional nanosensitizers for synergistic NIR photothermal and X-ray radiation therapy in vitro and in vivo.


Assuntos
Nanoestruturas , Linhagem Celular Tumoral , Ouro , Humanos , Hipertermia Induzida , Neoplasias , Fototerapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA