Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Acta Pharm Sin B ; 14(1): 133-154, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38239235
2.
Nano Lett ; 23(21): 9963-9971, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37729438

RESUMO

Given the key roles of cancer associated fibroblasts (CAFs) in shaping tumor stroma, this study shows a CAF-associated ITGB1-inactivating peptide-enriched membrane nanodelivery system (designated as PMNPs-D) to simultaneously target CAFs and tumor cells for boosted chemotherapy through promoted drug perfusion. In the structure of PMNPs-D, the PLGA-based inner core is loaded with the chemotherapeutic drug doxorubicin, and the outer surface is cloaked by hybrid biomembranes with the insertion of integrin ß1 (ITGB1) inhibiting peptide (i.e., FNIII14). After prolonged blood circulation and actively targeting in tumor sites, PMNPs-D can respond to CAF-overexpressed fibroblast activation protein-α (FAP-α) to trigger the release of FNIII14, which will bind to ITGB1 and inhibit CAFs' biological function in producing the stromal matrix, thereby loosening the condensed stromal structure and enhancing the permeability of nanotherapeutics in tumors. As a result, this tailor-designed nanosystem shows substantial tumor inhibition and metastasis retardation in aggressive adenoid cystic carcinoma (ACC) tumor-harboring mice.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias , Animais , Camundongos , Fibroblastos Associados a Câncer/patologia , Neoplasias/patologia , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Membranas , Peptídeos/metabolismo , Microambiente Tumoral , Linhagem Celular Tumoral , Fibroblastos/metabolismo
3.
ACS Nano ; 17(17): 17217-17232, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37584451

RESUMO

Macrophage-mediated cellular phagocytosis (MMCP) plays a critical role in conducting antitumor immunotherapy but is usually impaired by the intrinsic phagocytosis evading ability of tumor cells and the immunosuppressive tumor microenvironment (TME). Herein, a MMCP-boosting hydrogel (TCCaGM) was elaborately engineered by encapsulating granulocyte-macrophage colony-stimulating factor (GM-CSF) and a therapeutic nanoplatform (TCCaN) that preloaded with the tunicamycin (Tuni) and catalase (CAT) with the assistance of CaCO3 nanoparticles (NPs). Strikingly, the hypoxic/acidic TME was efficiently alleviated by the engineered hydrogel, "eat me" signal calreticulin (CRT) was upregulated, while the "don't eat me" signal CD47 was downregulated on tumor cells, and the infiltrated DCs were recruited and activated, all of which contributed to boosting the macrophage-mediated phagocytosis and initiating tumor-specific CD8+ T cells responses. Meanwhile, the remodeled TME was beneficial to accelerate the polarization of tumor-associated macrophages (TAMs) to the antitumoral M1-like phenotype, further heightening tumoricidal immunity. With the combination of PD-1 antibody (αPD-1), the designed hydrogel significantly heightened systemic antitumor immune responses and long-term immunological effects to control the development of primary and distant tumors as well as suppress tumor metastasis and recurrence, which established an optimal strategy for high-performance antitumor immunotherapy.


Assuntos
Adjuvantes Imunológicos , Neoplasias , Humanos , Adjuvantes Imunológicos/farmacologia , Microambiente Tumoral , Linfócitos T CD8-Positivos , Hidrogéis/farmacologia , Macrófagos , Neoplasias/terapia , Neoplasias/patologia , Fagocitose , Antígeno CD47 , Imunoterapia
4.
Nano Lett ; 22(21): 8735-8743, 2022 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-36286590

RESUMO

The chemotherapy efficacy of nanodrugs is restricted by poor tumor targeting and uptake. Here, an engineered biohybrid living material (designated as EcN@HPB) is constructed by integrating paclitaxel and BAY-876 bound human serum albumin nanodrugs (HPB) with Escherichia coli Nissle 1917 (EcN). Due to the inherent tumor tropism of EcN, EcN@HPB could actively target the tumor site and competitively deprive glucose through bacterial respiration. Thus, albumin would be used as an alternative nutrient source for tumor metabolism, which significantly promotes the internalization of HPB by tumor cells. Subsequently, BAY-876 internalized along with HPB nanodrugs would further depress glucose uptake of tumor cells via inhibiting glucose transporter 1 (GLUT1). Together, the decline of glucose bioavailability of tumor cells would activate and promote the macropinocytosis in an AMP-activated protein kinase (AMPK)-dependent manner, resulting in more uptake of HPB by tumor cells and boosting the therapeutic outcome of paclitaxel.


Assuntos
Infecções por Escherichia coli , Nanopartículas , Neoplasias , Humanos , Disponibilidade Biológica , Escherichia coli/genética , Escherichia coli/metabolismo , Glucose/metabolismo , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico
5.
ACS Nano ; 15(11): 17870-17884, 2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34747172

RESUMO

The excessive lactate in the tumor microenvironment always leads to poor therapeutic outcomes of chemotherapy. In this study, a self-driven bioreactor (defined as SO@MDH, where SO is Shewanella oneidensis MR-1 and MDH is MIL-101 metal-organic framework nanoparticles/doxorubicin/hyaluronic acid) is rationally constructed via the integration of doxorubicin (DOX)-loaded metal-organic framework (MOF) MIL-101 nanoparticles with SO to sensitize chemotherapy. Owing to the intrinsic tumor tropism and electron-driven respiration of SO, the biohybrid SO@MDH could actively target and colonize hypoxic and eutrophic tumor regions and anaerobically metabolize lactate accompanied by the transfer of electrons to Fe3+, which is the key component of the MIL-101 nanoparticles. As a result, the intratumoral lactate would undergo continuous catabolism coupled with the reduction of Fe3+ to Fe2+ and the subsequent degradation of MIL-101 frameworks, leading to an expeditious drug release for effective chemotherapy. Meanwhile, the generated Fe2+ will be promptly oxidized by the abundant hydrogen peroxide in the tumor microenvironment to reproduce Fe3+, which is, in turn, beneficial to circularly catabolize lactate and boost chemotherapy. More importantly, the consumption of intratumoral lactic acid could significantly inhibit the expression of multidrug resistance-related ABCB1 protein (also named P-glycoprotein (P-gp)) for conquering drug-resistant tumors. SO@MDH demonstrated here holds high tumor specificity and promising chemotherapeutic efficacy for suppressing tumor growth and overcoming multidrug resistance, confirming its potential prospects in cancer therapy.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Neoplasias , Humanos , Doxorrubicina/farmacologia , Neoplasias/terapia , Reatores Biológicos , Lactatos , Microambiente Tumoral
6.
Chem Commun (Camb) ; 57(74): 9398-9401, 2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34528964

RESUMO

A tumor cell membrane-camouflaged therapeutic system was fabricated to eliminate tumors by embedding apyrase and glucose oxidase (GOx) into zeolitic imidazolate framework-8 (ZIF-8) nanoparticles for tumor-targeted metabolic therapy. Experimental results demonstrated that these functional nanoparticles could disturb the energy supply of tumor cells by depleting ATP and glucose and efficiently induce tumor cell death.


Assuntos
Apirase/metabolismo , Materiais Biomiméticos/metabolismo , Glucose Oxidase/metabolismo , Estruturas Metalorgânicas/metabolismo , Nanopartículas/metabolismo , Neoplasias/metabolismo , Trifosfato de Adenosina/metabolismo , Materiais Biomiméticos/química , Morte Celular/efeitos dos fármacos , Glucose/metabolismo , Humanos , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Nanopartículas/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia
7.
Adv Mater ; 33(31): e2007630, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34050564

RESUMO

Immunotherapy that can activate immunity or enhance the immunogenicity of tumors has emerged as one of the most effective methods for cancer therapy. Nevertheless, single-mode immunotherapy is still confronted with several critical challenges, such as the low immune response, the low tumor infiltration, and the complex immunosuppression tumor microenvironment. Recently, the combination of immunotherapy with other therapeutic modalities has emerged as a powerful strategy to augment the therapeutic outcome in fighting against cancer. In this review, recent research advances of the combination of immunotherapy with chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, metabolic therapy, and microwave thermotherapy are summarized. Critical challenges and future research direction of immunotherapy-based cancer therapeutic strategy are also discussed.


Assuntos
Imunoterapia , Microambiente Tumoral , Terapia Combinada , Humanos , Terapia de Imunossupressão , Fototerapia
8.
Chem Soc Rev ; 50(2): 945-985, 2021 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-33226037

RESUMO

Cell primitive-based functional materials that combine the advantages of natural substances and nanotechnology have emerged as attractive therapeutic agents for cancer therapy. Cell primitives are characterized by distinctive biological functions, such as long-term circulation, tumor specific targeting, immune modulation etc. Moreover, synthetic nanomaterials featuring unique physical/chemical properties have been widely used as effective drug delivery vehicles or anticancer agents to treat cancer. The combination of these two kinds of materials will catalyze the generation of innovative biomaterials with multiple functions, high biocompatibility and negligible immunogenicity for precise cancer therapy. In this review, we summarize the most recent advances in the development of cell primitive-based functional materials for cancer therapy. Different cell primitives, including bacteria, phages, cells, cell membranes, and other bioactive substances are introduced with their unique bioactive functions, and strategies in combining with synthetic materials, especially nanoparticulate systems, for the construction of function-enhanced biomaterials are also summarized. Furthermore, foreseeable challenges and future perspectives are also included for the future research direction in this field.


Assuntos
Antineoplásicos/farmacologia , Materiais Biomiméticos/farmacologia , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Materiais Biomiméticos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Neoplasias/patologia
9.
J Am Chem Soc ; 141(39): 15567-15576, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31478647

RESUMO

In this study, we report on a redox-controllable and reversible complete "ON"/"OFF"-switchable aptamer binding to ATP. A series of methylene blue-modified ATP-aptamers was synthesized, revealing improved binding affinities toward ATP as compared to the nonmodified aptamer. These binding affinities were dependent on the conjugation site of the redox label on the aptamer scaffold. Importantly, we find that the oxidized methylene blue-modified aptamers bind to ATP with micromolar affinity, while the reduced form lacks binding affinity toward ATP, resulting in an unprecedented complete "ON"/"OFF" redox-controllable aptamer switch. We demonstrate the cyclic "ON"/"OFF" binding of ATP to the methylene blue-functionalized aptamer through cyclic oxidation and reduction of the redox label using both chemical and electrochemical means. Molecular dynamics and docking simulations were performed to account for the redox-switchable properties of the conjugated aptamers and to rationalize the enhanced binding affinities of the different aptamer designs.


Assuntos
Trifosfato de Adenosina/química , Aptâmeros de Nucleotídeos/química , Técnicas Biossensoriais/métodos , Fenômenos Químicos , Técnicas Eletroquímicas , Modelos Moleculares , Conformação de Ácido Nucleico , Oxirredução
10.
Small ; 15(17): e1900935, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30920730

RESUMO

UiO-68 metal-organic framework nanoparticles (NMOFs) are loaded with a doxorubicin drug (fluorescent dye analogs) and locked by means of structurally engineered duplex nucleic acid structures, where one strand is covalently linked to the NMOFs and the second strand is hybridized with the anchor strand. Besides the complementarity of the second strand to the anchor sequence, it includes the complementary sequence to the microRNAs (miRNA)-21 or miRNA-221 that is specific miRNA biomarker for MCF-7 breast cancer cells or OVCAR-3 ovarian cancer cells. In the presence of the respective miRNA biomarkers, the miRNA-induced displacement of the strand associated with the anchor strand proceeds, resulting in the release of DNA/miRNA duplexes. The released duplexes are, however, engineered to be digested in the presence of exonuclease III, Exo III, a process that recycles the miRNAs and provides the autonomous amplified unlocking of the NMOFs and the release of the doxorubicin load (or the fluorescent dye analogs) even at low concentrations of miRNA. Preliminary cell experiments reveal that the respective NMOFs are unlocked by the miRNA-21 or miRNA-221, resulting in selective cytotoxicity toward MCF-7 breast cancer cells or OVCAR-3 ovarian cancer cells.


Assuntos
Sistemas de Liberação de Medicamentos , Estruturas Metalorgânicas , MicroRNAs/química , Nanomedicina/métodos , Neoplasias/tratamento farmacológico , Apoptose , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular , Doxorrubicina/química , Feminino , Humanos , Ligantes , Células MCF-7 , MicroRNAs/metabolismo , Nanopartículas/química , Compostos Orgânicos , Neoplasias Ovarianas/tratamento farmacológico
11.
Adv Mater ; 31(3): e1802725, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30260521

RESUMO

Recently, diverse functional materials that take subcellular structures as therapeutic targets are playing increasingly important roles in cancer therapy. Here, particular emphasis is placed on four kinds of therapies, including chemotherapy, gene therapy, photodynamic therapy (PDT), and hyperthermal therapy, which are the most widely used approaches for killing cancer cells by the specific destruction of subcellular organelles. Moreover, some non-drug-loaded nanoformulations (i.e., metal nanoparticles and molecular self-assemblies) with a fatal effect on cells by influencing the subcellular functions without the use of any drug molecules are also included. According to the basic principles and unique performances of each treatment, appropriate strategies are developed to meet task-specific applications by integrating specific materials, ligands, as well as methods. In addition, the combination of two or more therapies based on multifunctional nanostructures, which either directly target specific subcellular organelles or release organelle-targeted therapeutics, is also introduced with the intent of superadditive therapeutic effects. Finally, the related challenges of critical re-evaluation of this emerging field are presented.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Animais , Humanos , Neoplasias/metabolismo , Organelas
12.
Small ; 13(18)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28266809

RESUMO

The nanoplatform GNR-ACPP-PpIX (designated as GNR-ACPI) is designed for dual image guided combined activatable photodynamic therapy (PDT) and photothermal therapy (PTT). In GNR-ACPI, gold nanorods (GNRs) are modified with a protoporphyrin (PpIX, a PDT agent) conjugated activatable cell penetrating peptide (ACPP), which consists of the matrix metalloproteinases-2 (MMP-2) sensitive peptide sequence GPLGLAG. First, the photoactivity of PpIX is effectively quenched by GNRs due to the strong near infrared region light absorption of GNR and the special "U type" structure of ACPP induced close contact between PpIX and GNR. However, once arriving at the tumor site, the GPLGLAG sequence is hydrolyzed by the MMP-2 overexpressed by tumor cells, resulting in the release of the residual cell membrane penetrating peptide (CPP) attached PpIX (CPP-PpIX) with the recovery of photoactivity of PpIX. In addition, with the help of CPP, more efficient cellular uptake of PpIX by tumor cells can be achieved, which will greatly improve the PDT efficacy. Moreover, the GNR can also be utilized for photothermic imaging as well as PTT for tumors. It is found that the combination of PTT and PDT under the guidance of dual-mode imaging greatly enhances the antitumor effects, while possessing negligible systematic toxicity.


Assuntos
Ouro/química , Nanotubos/química , Fotoquimioterapia/métodos , Peptídeos Penetradores de Células/química
13.
ACS Nano ; 11(2): 1419-1431, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28107631

RESUMO

In this study, we developed a general method to decorate plasmonic gold nanorods (GNRs) with a CD44-targeting functional polymer, containing a hyaluronic acid (HA)-targeting moiety and a small molecule Glut1 inhibitor of diclofenac (DC), to obtain GNR/HA-DC. This nanosystem exhibited the superiority of selectively sensitizing tumor cells for photothermal therapy (PTT) by inhibiting anaerobic glycolysis. Upon specifically targeting CD44, sequentially time-dependent DC release could be achieved by the trigger of hyaluronidase (HAase), which abundantly existed in tumor tissues. The released DC depleted the Glut1 level in tumor cells and induced a cascade effect on cellular metabolism by inhibiting glucose uptake, blocking glycolysis, decreasing ATP levels, hampering heat shock protein (HSP) expression, and ultimately leaving malignant cells out from the protection of HSPs to stress (e.g., heat), and then tumor cells were more easy to kill. Owing to the sensitization effect of GNR/HA-DC, CD44 overexpressed tumor cells could be significantly damaged by PTT with an enhanced therapeutic efficiency in vitro and in vivo.


Assuntos
Anaerobiose/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Temperatura Alta , Fototerapia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Células COS , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Chlorocebus aethiops , Diclofenaco/química , Diclofenaco/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Ouro/química , Ouro/farmacologia , Células HeLa , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Células MCF-7 , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7
14.
Biomaterials ; 117: 54-65, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27936417

RESUMO

Supramolecular photosensitizers (supraPSs) have emerged as effective photodynamic therapy (PDT) agents. Here, we propose the assembling capacity of supraPSs as a new strategy to construct theranostic nanoplatform with versatile functions aming at high-performance tumor therapy. By coating tirapazamine (TPZ)-loaded mesoporous silica nanoparticles (MSNs) with layer-by-layer (LbL) assembled multilayer, the versatile nanoplatform (TPZ@MCMSN-Gd3+) was obtained with the formation of supraPSs via host-guest interaction and the chelation with paramagnetic Gd3+. The TPZ@MCMSN-Gd3+ could be specifically uptaken by CD44 receptor overexpressed tumor cells and respond to hyaluronidase (HAase) to trigger the release of therapeutics. As confirmed by in vivo studies, TPZ@MCMSN-Gd3+ showed preferential accumulation in tumor site and significantly inhibited the tumor progression by the collaboration of PDT and bioreductive chemotherapy under NIR fluorescence/MR imaging guidance. Taken together, this supraPSs based strategy paves a new paradigm of the way for the construction of theranostic nanoplatform.


Assuntos
Antineoplásicos/administração & dosagem , Nanocápsulas/química , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/tratamento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/administração & dosagem , Dióxido de Silício/química , Animais , Células COS , Chlorocebus aethiops , Cristalização/métodos , Feminino , Humanos , Células MCF-7 , Imageamento por Ressonância Magnética/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia de Fluorescência/métodos , Nanocápsulas/administração & dosagem , Nanocápsulas/ultraestrutura , Nanoporos/ultraestrutura , Impressão Tridimensional , Nanomedicina Teranóstica/métodos , Tirapazamina , Resultado do Tratamento , Triazinas/administração & dosagem
15.
Biomaterials ; 76: 87-101, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26519651

RESUMO

In this paper, a multifunctional theranostic magnetic mesoporous silica nanoparticle (MMSN) with magnetic core was developed for magnetic-enhanced tumor-targeted MR imaging and precise therapy. The gatekeeper ß-cyclodextrin (ß-CD) was immobilized on the surface of mesoporous silica shell via platinum(IV) prodrug linking for reduction-triggered intracellular drug release. Then Arg-Gly-Asp (RGD) peptide ligand was further introduced onto the gatekeeper ß-CD via host-guest interaction for cancer targeting purpose. After active-targeting endocytosis by cancer cells, platinum(IV) prodrug in MMSNs would be restored to active platinum(II) drug in response to the innative reducing microenvironment in cancer cells, resulting in the detachment of ß-CD gatekeeper and thus simultaneously triggering the in situ release of anticancer drug doxorubicin (DOX) entrapped in the MMSNs to kill cancer cells. It was found that with the aid of an external magnetic field, drug loaded MMSNs showed high contrast in MR imaging in vivo and exhibited magnetically enhanced accumulation in the cancer site, leading to significant inhibition of cancer growth with minimal side effects. This multifunctional MMSN will find great potential as a theranostic nanoplatform for cancer treatment.


Assuntos
Imageamento por Ressonância Magnética/métodos , Nanopartículas , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Dióxido de Silício , Animais , Antibióticos Antineoplásicos/uso terapêutico , Células COS , Chlorocebus aethiops , Doxorrubicina/uso terapêutico , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Nanomedicina Teranóstica
16.
Small ; 12(6): 733-44, 2016 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26708101

RESUMO

In this work, a ZnO based nanococktail with programmed functions is designed and synthesized for self-synergistic tumor targeting therapy. The nanococktail can actively target tumors via specific interaction of hyaluronic acid (HA) with CD44 receptors and respond to HAase-rich tumor microenvironment to induce intracellular cascade reaction for controlled therapy. The exposed cell-penetrating peptide (R8) potentiates the cellular uptake of therapeutic nanoparticles into targeted tumor cells. Then ZnO cocktail will readily degrade in acidic endo/lysosomes and induce the production of desired reactive oxygen species (ROS) in situ. The destructive ROS not only leads to serious cell damage but also triggers the on-demand drug release for precise chemotherapy, thus achieving enhanced antitumor efficiency synergistically. After tail vein injection of ZnO cocktail, a favorable tumor apoptosis rate (71.2 ± 8.2%) is detected, which is significantly superior to that of free drug, doxorubicin (12.9 ± 5.2%). Both in vitro and in vivo studies demonstrate that the tailor-made ZnO cocktail with favorable biocompatibility, promising tumor specificity, and self-synergistically therapeutic capacity opens new avenues for cancer therapy.


Assuntos
Espaço Intracelular/metabolismo , Nanopartículas/química , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Citometria de Fluxo , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Espécies Reativas de Oxigênio/metabolismo
17.
Adv Healthc Mater ; 4(15): 2247-59, 2015 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-26333115

RESUMO

Nanomaterials that integrate diagnostic and therapeutic functions within a single nanoplatform promise great advances in revolutionizing cancer therapy. A smart multifunctional theranostic drug-delivery system (DDS) based on gold nanorods (abbreviated as GNR/TSDOX) is designed for cancer-targeted imaging and imaging-guided therapy. In this intelligent theranostic DDS, the active targeting ligand biotin is introduced to track cancer sites in vivo. With the aid of photothermal/photoacoustic imaging, GNR/TSDOX can ablate cancer specifically and effectively. When stimulated with a single near-infrared (NIR) light source, this NIR light energy is effectively absorbed and converted into heat by GNR/TSDOX for localized photothermal therapy and the increase in temperature also further triggers the cascaded release of the anticancer drug for combined thermo-chemotherapy. More importantly, the in vivo cure effect can be well guided by regulating the irradiation time and intensity of the NIR light.


Assuntos
Antineoplásicos/farmacologia , Nanotubos/química , Neoplasias/tratamento farmacológico , Nanomedicina Teranóstica/métodos , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Terapia Combinada , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Feminino , Ouro/química , Células HeLa , Humanos , Raios Infravermelhos , Terapia a Laser , Camundongos
18.
Small ; 11(39): 5230-42, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26285687

RESUMO

Nanotechnology-based drug delivery has a great potential to revolutionize cancer treatment by enhancing anticancer drug efficacy and reducing drug toxicity. Here, a bioinspired nano-prodrug (BiNp) assembled by an antineoplastic peptidic derivative (FA-KLA-Hy-DOX), a folate acid (FA)-incorporated proapoptotic peptide (KLAKLAK)(2) (KLA) to doxorubicin (DOX) via an acid-labile hydrozone bond (Hy) is constructed. The hydrophobic antineoplastic agent DOX is efficiently shielded in the core of nano-prodrug. With FA targeting moieties on the surface, the obtained BiNp shows significant tumor-targeting ability and enhances the specific uptake of cancer cells. Upon the trigger by the intracellular acidic microenvironment of endosomes, the antineoplastic agent DOX is released on-demand and promotes the apoptosis of cancer cells. Simultaneously, the liberated FA-KLA can induce the dysfunction of mitochondria and evoke mitochondria-dependent apoptosis. In vitro and in vivo results show that the nano-prodrug BiNp with integrated programmed functions exhibits remarkable inhibition of tumor and achieves a maximized therapeutic efficiency with a minimized side effect.


Assuntos
Doxorrubicina/administração & dosagem , Ácido Fólico/farmacocinética , Nanocápsulas/administração & dosagem , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Pró-Fármacos/administração & dosagem , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/química , Apoptose/efeitos dos fármacos , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/síntese química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Sinergismo Farmacológico , Feminino , Ácido Fólico/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanocápsulas/química , Nanocápsulas/ultraestrutura , Neoplasias Experimentais/química , Neoplasias Experimentais/patologia , Pró-Fármacos/síntese química
19.
ACS Appl Mater Interfaces ; 7(31): 17171-80, 2015 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-26192215

RESUMO

A versatile gold nanoparticle-based multifunctional nanocomposite AuNP@CD-AD-DOX/RGD was constructed flexibly via host-guest interaction for targeted cancer chemotherapy. The pH-sensitive anticancer prodrug AD-Hyd-DOX and the cancer-targeted peptide AD-PEG8-GRGDS were modified on the surface of AuNP@CD simultaneously, which endowed the resultant nanocomposite with the capability to selectively eliminate cancer cells. In vitro studies indicated that the AuNP@CD-AD-DOX/RGD nanocomposite was preferentially uptaken by cancer cells via receptor-mediated endocytosis. Subsequently, anticancer drug DOX was released rapidly upon the intracellular trigger of the acid microenvirenment of endo/lysosomes, inducing apoptosis in cancer cells. As the ideal drug nanocarrier, the multifunctional gold nanoparticles with the active targeting and controllable intracellular release ability hold the great potential in cancer therapy.


Assuntos
Portadores de Fármacos/química , Ouro/química , Nanopartículas Metálicas/química , Nanocompostos/química , Adamantano/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Doxorrubicina/química , Doxorrubicina/farmacologia , Portadores de Fármacos/metabolismo , Liberação Controlada de Fármacos , Endocitose , Grafite/química , Humanos , Microscopia Confocal , beta-Ciclodextrinas/química
20.
ACS Appl Mater Interfaces ; 7(17): 9078-87, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25893819

RESUMO

Functional mesoporous silica particles have attracted growing research interest for controlled drug delivery in targeted cancer therapy. For the purpose of efficient targeting tumor cells and reducing the adverse effect of antitumor drug doxorubicin (DOX), biocompatible and enzyme-responsive mesoporous silica nanoparticles (MSNs) with tumor specificity were desired. To construct these functional MSNs, the classic rotaxane structure formed between alkoxysilane tether and α-cyclodextrin (α-CD) was employed to anchor onto the orifices of MSNs as gatekeeper in this work. After subsequent modification by multifunctional peptide (azido-GFLGR7RGDS with tumor-targeting, membrane-penetrating, and cathepsin B-responsive functions) to stabilize the gatekeeper, the resulting functional MSNs showed a strong ability to load and seal DOX in their nanopores. When incubating these DOX-loaded MSNs with tumor and normal cells, the nanoparticles could efficiently employ their surface-encoded RGDS and continuous seven arginine (R7) sequences to target tumor cells, penetrate the cell membrane, and enter tumor cells. Because cathepsin B overexpressed in late endosomes and lysosomes of tumor cells could specifically hydrolyze GFLG sequences of the nanovalves, the DOX-loaded MSNs showed an "off-on" drug release behavior that ∼80% loaded DOX could be released within 24 h and thus showed a high rate of apoptosis. Furthermore, in vitro cellular experiments indicated that DOX-loaded MSNs (DOX@MSN-GFLGR7RGDS/α-CD) had high growth inhibition toward αvß3-positive HeLa cancerous cells. The research might offer a practical way for designing the tumor-targeted and enzyme-induced drug delivery system for cancer therapy.


Assuntos
Catepsina B/metabolismo , Preparações de Ação Retardada/administração & dosagem , Doxorrubicina/administração & dosagem , Nanocápsulas/química , Nanoporos/ultraestrutura , Dióxido de Silício/química , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/química , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Sobrevivência Celular/efeitos dos fármacos , Preparações de Ação Retardada/síntese química , Difusão , Doxorrubicina/química , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/química , Células HeLa , Humanos , Nanocápsulas/administração & dosagem , Nanocápsulas/ultraestrutura , Nanocompostos/administração & dosagem , Nanocompostos/química , Nanocompostos/ultraestrutura , Tamanho da Partícula , Porosidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA