Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
J Cosmet Dermatol ; 23(5): 1816-1827, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38193246

RESUMO

BACKGROUND: The purpose of this study was to investigate the protective effect of Silibinin-loaded polymeric micelles from human hair against UV-B radiation. METHODS: Eight formulations with different concentrations of Silibinin, Pluronic F-127, and Labrasol-Labrafil were made by a solvent evaporation method, and the selected formulation was chosen by examining their properties like particle size and loading efficiency. Six groups of human hair, including a group that received the selected formulation, were exposed to UV-B radiation and by calculating its factors such as peak-to-valley roughness, RMS roughness, FTIR, and the amount of protein loss, the protective effect of the selected formulation was judged. RESULTS: According to the results, the loading efficiency and particle size of the selected formulation were 45.34% and 43.19 nm. The Silibinin release profile had two parts, fast and slow, which were suitable for creating a drug depot on hair. Its zeta potential also confirmed the minimum electrostatic interference between the formulation and hair surface. The zeta potential of selected formulation was -5.9 mv. Examination of AFM images showed that the selected formulation was able to prevent the increase in peak-to-valley roughness and RMS roughness caused by UV-B radiation. RMS roughness after 600 h of UV radiation in Groups 5 and 6 was significantly lower than the negative control group and the amount of this factor did not differ significantly between 0 and 600, so it can be concluded that the selected formulation containing Silibinin and the positive control group was able to prevent the increase of RMS roughness and hair destruction. In other hands, the two positive control groups and the selected formulation containing Silibinin were able to effectively reduce hair protein loss. CONCLUSION: Silibinin-loaded polymeric micelles were able to effectively protect hair from structural and chemical changes caused by UV-B radiation.


Assuntos
Cabelo , Micelas , Tamanho da Partícula , Silibina , Raios Ultravioleta , Humanos , Raios Ultravioleta/efeitos adversos , Silibina/farmacologia , Silibina/administração & dosagem , Silibina/química , Cabelo/efeitos dos fármacos , Cabelo/efeitos da radiação , Silimarina/farmacologia , Silimarina/administração & dosagem , Silimarina/química , Polímeros/química , Liberação Controlada de Fármacos/efeitos da radiação , Antioxidantes/farmacologia , Antioxidantes/administração & dosagem , Portadores de Fármacos/química , Portadores de Fármacos/efeitos da radiação
2.
ACS Appl Mater Interfaces ; 14(1): 57-68, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-34935343

RESUMO

Integrating chemodynamic therapy (CDT) and photodynamic therapy (PDT) into one nanoplatform can produce much more reactive oxygen species (ROS) for tumor therapy. Nevertheless, it is still a great challenge to selectively generate sufficient ROS in tumor regions. Meanwhile, CDT and PDT are restricted by insufficient H2O2 content in the tumor as well as by the limited tumor tissue penetration of the light source. In this study, a smart pH/ROS-responsive nanoplatform, Fe2+@UCM-BBD, is rationally designed for tumor combination therapy. The acidic microenvironment can induce the pH-responsive release of doxorubicin (DOX), which can induce tumor apoptosis through DNA damage. Beyond that, DOX can promote the production of H2O2, providing sufficient materials for CDT. Of note, upconversion nanoparticles at the core can convert the 980 nm light to red and green light, which are used to activate Ce6 to produce singlet oxygen (1O2) and achieve upconversion luminescence imaging, respectively. Then, the ROS-responsive linker bis-(alkylthio)alkene is cleaved by 1O2, resulting in the release of Fenton reagent (Fe2+) to realize CDT. Taken together, Fe2+@UCM-BBD exhibits on-demand therapeutic reagent release capability, excellent biocompatibility, and remarkable tumor inhibition ability via synergistic chemo/photodynamic/chemodynamic combination therapy.


Assuntos
Antineoplásicos/uso terapêutico , Doxorrubicina/uso terapêutico , Portadores de Fármacos/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Fármacos Fotossensibilizantes/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Clorofilídeos/química , Clorofilídeos/efeitos da radiação , Clorofilídeos/uso terapêutico , Terapia Combinada , Doxorrubicina/química , Portadores de Fármacos/química , Portadores de Fármacos/efeitos da radiação , Liberação Controlada de Fármacos , Tratamento Farmacológico , Érbio/química , Érbio/efeitos da radiação , Érbio/uso terapêutico , Feminino , Fluoretos/química , Fluoretos/efeitos da radiação , Fluoretos/uso terapêutico , Humanos , Ferro/química , Ferro/efeitos da radiação , Ferro/uso terapêutico , Nanopartículas Metálicas/química , Nanopartículas Metálicas/efeitos da radiação , Camundongos Endogâmicos BALB C , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Itérbio/química , Itérbio/efeitos da radiação , Itérbio/uso terapêutico , Ítrio/química , Ítrio/efeitos da radiação , Ítrio/uso terapêutico
3.
ACS Appl Mater Interfaces ; 13(37): 43855-43867, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34494809

RESUMO

Black phosphorus (BP) nanosheet is easily oxidized by oxygen and water under ambient environment, thus, reliable BP passivation techniques for biomedical applications is urgently needed. A simple and applicable passivation strategy for biomedical applications was established by encapsulating BP nanosheet into zeolitic imidazole framework-8 (ZIF-8). The resulted BP nanosheet in ZIF-8 (BP@ZIF-8) shows not only satisfied chemical stability in both water and phosphate buffered saline (PBS), but also excellent biocompatibility. Notably, BP nanosheet endows the prepared BP@ZIF-8 with prominent photothermal conversion efficiency (31.90%). Besides passivation BP, ZIF-8 provides the BP@ZIF-8 with high drug loading amount (1353.3 mg g-1). Moreover, the loaded drug can be controlled release by pH stimuli. Both in vitro and in vivo researches verified the resulted BP@ZIF-8 an ideal candidate for tumor multimodal treatments.


Assuntos
Antineoplásicos/uso terapêutico , Portadores de Fármacos/química , Estruturas Metalorgânicas/química , Nanoestruturas/química , Neoplasias/tratamento farmacológico , Fósforo/química , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Terapia Combinada , Doxorrubicina/química , Doxorrubicina/uso terapêutico , Portadores de Fármacos/efeitos da radiação , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos , Tratamento Farmacológico , Feminino , Humanos , Concentração de Íons de Hidrogênio , Raios Infravermelhos , Estruturas Metalorgânicas/efeitos da radiação , Estruturas Metalorgânicas/toxicidade , Camundongos , Nanoestruturas/efeitos da radiação , Nanoestruturas/toxicidade , Fósforo/efeitos da radiação , Fósforo/toxicidade , Terapia Fototérmica
4.
ACS Appl Mater Interfaces ; 13(37): 44124-44135, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34495627

RESUMO

Although nanoparticles based on Group 8 elements such as Fe and Ru have been developed, not much is known about Os nanoparticles. However, Os-based nanostructures might have potential in various applications including biomedical fields. Therefore, in this study, we synthesized Os-Te nanorods (OsTeNRs) by solvothermal galvanic replacement with Te nanotemplates. We explored the nanozymatic activity of the synthesized OsTeNRs and found that they exhibited superior photothermal conversion and photocatalytic activity. Along with chemotherapy (regorafenib) and immunotherapy, the nanozymatic, photothermal, and photodynamic activities of OsTeNRs were harnessed to develop a pentamodal treatment for hepatocellular carcinoma (HCC); in vitro and in vivo studies demonstrated that the pentamodal therapy could alleviate hypoxia in HCC cells by generating oxygen and reduced unintended drug accumulation in organs. Moreover, bone-marrow toxicity due to regorafenib could be reduced as the drug was released in a sustained manner. Thus, OsTeNRs can be considered as suitable nanotemplates for combinatorial cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Portadores de Fármacos/química , Neoplasias Hepáticas/tratamento farmacológico , Nanotubos/química , Animais , Catálise , Linhagem Celular Tumoral , Portadores de Fármacos/síntese química , Portadores de Fármacos/efeitos da radiação , Masculino , Camundongos Endogâmicos C57BL , Nanotubos/efeitos da radiação , Osmio/química , Osmio/efeitos da radiação , Compostos de Fenilureia/uso terapêutico , Fotoquimioterapia , Piridinas/uso terapêutico , Telúrio/química , Telúrio/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
5.
ACS Appl Mater Interfaces ; 13(31): 37563-37577, 2021 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-34338525

RESUMO

Despite its success against cancer, photothermal therapy (PTT) (>50 °C) suffers from several limitations such as triggering inflammation and facilitating immune escape and metastasis and also damage to the surrounding normal cells. Mild-temperature PTT has been proposed to override these shortcomings. We developed a nanosystem using HepG2 cancer cell membrane-cloaked zinc glutamate-modified Prussian blue nanoparticles with triphenylphosphine-conjugated lonidamine (HmPGTL NPs). This innovative approach achieved an efficient mild-temperature PTT effect by downregulating the production of intracellular ATP. This disrupts a section of heat shock proteins that cushion cancer cells against heat. The physicochemical properties, anti-tumor efficacy, and mechanisms of HmPGTL NPs both in vitro and in vivo were investigated. Moreover, the nanoparticles cloaked with the HepG2 cell membrane substantially prolonged the circulation time in vivo. Overall, the designed nanocomposites enhance the efficacy of mild-temperature PTT by disrupting the production of ATP in cancer cells. Thus, we anticipate that the mild-temperature PTT nanosystem will certainly present its enormous potential in various biomedical applications.


Assuntos
Antineoplásicos/uso terapêutico , Membrana Celular/química , Ferrocianetos/química , Mitocôndrias/efeitos dos fármacos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Portadores de Fármacos/química , Portadores de Fármacos/efeitos da radiação , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos , Feminino , Ferrocianetos/efeitos da radiação , Ferrocianetos/toxicidade , Células Hep G2 , Humanos , Indazóis/química , Indazóis/uso terapêutico , Raios Infravermelhos , Camundongos Nus , Nanocompostos/química , Nanocompostos/toxicidade , Nanopartículas/efeitos da radiação , Nanopartículas/toxicidade , Terapia Fototérmica
6.
ACS Appl Mater Interfaces ; 13(28): 32690-32702, 2021 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-34229434

RESUMO

The synergistic nanotheranostics of reactive oxygen species (ROS) augment or phototherapy has been a promising method within synergistic oncotherapy. However, it is still hindered by sophisticated design and fabrication, lack of a multimodal synergistic effect, and hypoxia-associated poor photodynamic therapy (PDT) efficacy. Herein, a kind of porous shuttle-shape platinum (IV) methylene blue (Mb) coordination polymer nanotheranostics-loaded 10-hydroxycamptothecin (CPT) is fabricated to address the abovementioned limitations. Our nanoreactors possess spatiotemporally controlled O2 self-supply, self-sufficient singlet oxygen (1O2), and outstanding photothermal effect. Once they are taken up by tumor cells, nanoreactors as a cascade catalyst can efficiently catalyze degradation of the endogenous hydrogen peroxide (H2O2) into O2 to alleviate tumor hypoxia. The production of O2 can ensure enhanced PDT. Subsequently, under both stimuli of external red light irradiation and internal lysosomal acidity, nanoreactors can achieve the on-demand release of CPT to augment in situ mitochondrial ROS and highly efficient tumor ablation via phototherapy. Moreover, under the guidance of near-infrared (NIR) fluorescent imaging, our nanoreactors exhibit strongly synergistic potency for treatment of hypoxic tumors while reducing damages against normal tissues and organs. Collectively, shuttle-shape platinum-coordinated nanoreactors with augmented ROS capacity and enhanced phototherapy efficiency can be regarded as a novel tumor theranostic agent and further promote the research of synergistic oncotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Camptotecina/análogos & derivados , Portadores de Fármacos/química , Nanoestruturas/química , Neoplasias/tratamento farmacológico , Hipóxia Tumoral/efeitos dos fármacos , Animais , Antineoplásicos/química , Camptotecina/química , Camptotecina/uso terapêutico , Catálise/efeitos da radiação , Linhagem Celular Tumoral , Portadores de Fármacos/efeitos da radiação , Liberação Controlada de Fármacos , Feminino , Humanos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Luz , Azul de Metileno/análogos & derivados , Azul de Metileno/efeitos da radiação , Camundongos Endogâmicos BALB C , Nanoestruturas/efeitos da radiação , Neoplasias/metabolismo , Oxigênio/metabolismo , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , Terapia Fototérmica , Platina/química , Platina/efeitos da radiação , Polímeros/síntese química , Polímeros/química , Polímeros/efeitos da radiação , Porosidade , Oxigênio Singlete/metabolismo , Nanomedicina Teranóstica
7.
ACS Appl Mater Interfaces ; 13(30): 35376-35388, 2021 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-34313109

RESUMO

Escorting therapeutics for malignancies by nano-encapsulation to ameliorate treatment effects and mitigate side effects has been pursued in precision medicine. However, the majority of drug delivery systems suffer from uncontrollable drug release kinetics and thus lead to unsatisfactory triggered-release efficiency along with severe side effects. Herein, we developed a unique nanovesicle delivery system that shows near-infrared (NIR) light-triggered drug release behavior and minimal premature drug release. By co-encapsulation of superparamagnetic iron oxide (SPIO) nanoparticles, the ultrasound contrast agent perfluorohexane (PFH), and cisplatin in a silicate-polyaniline vesicle, we achieved the controllable release of cisplatin in a thermal-responsive manner. Specifically, vaporization of PFH triggered by the heat generated from NIR irradiation imparts high inner vesicle pressure on the nanovesicles, leading to pressure-induced nanovesicle collapse and subsequent cisplatin release. Moreover, the multimodal imaging capability can track tumor engagement of the nanovesicles and assess their therapeutic effects. Due to its precise inherent NIR-triggered drug release, our system shows excellent tumor eradication efficacy and biocompatibility in vivo, empowering it with great prospects for future clinical translation.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Meios de Contraste/química , Portadores de Fármacos/química , Fluorocarbonos/química , Neoplasias/tratamento farmacológico , Células A549 , Compostos de Anilina/química , Compostos de Anilina/efeitos da radiação , Compostos de Anilina/toxicidade , Animais , Antineoplásicos/química , Antineoplásicos/toxicidade , Cisplatino/química , Cisplatino/toxicidade , Meios de Contraste/toxicidade , Portadores de Fármacos/efeitos da radiação , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos , Tratamento Farmacológico , Fluorocarbonos/toxicidade , Humanos , Raios Infravermelhos , Nanopartículas Magnéticas de Óxido de Ferro/química , Nanopartículas Magnéticas de Óxido de Ferro/efeitos da radiação , Nanopartículas Magnéticas de Óxido de Ferro/toxicidade , Camundongos Nus , Terapia Fototérmica , Silicatos/química , Silicatos/toxicidade , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Inorg Biochem ; 223: 111558, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34329998

RESUMO

Photo-chemotherapy (PCT) reveals great potential in hepatocellular carcinoma (HCC) treatment, therefore the construct of smart PCT nano-agents with high photothermal conversion efficiency and accurate drug delivery is of great significant. Herein, a novel hybrid nanomaterial MGO-TCA-FA has been designed and constructed by grafting the triformyl cholic acid (TCA) and folic acid (FA) on the surface of Fe3O4 modified graphene oxide (MGO). The doxorubicin hydrochloride (DOX) as a model drug could be effectively loaded on the MGO-TCA-FA via hydrogen bonding and π-π stacking (the drug loading amount was 1040 mg/g). The formed MGO-TCA-FA@DOX has been developed to be an effective PCT nanoplatform with the advantages of multiple-targeted drug delivery, near-infrared light (NIR) and pH triggered drug release, and photothermal conversion efficiency. In vitro experiments showed that compared with other cancer cells and normal liver cells, MGO-TCA-FA@DOX could specifically target liver cancer cells and presented significant killing ability to liver cancer cells. More importantly, in vivo experiments indicated that PCT synergistic therapy (MGO-TCA-FA@DOX) revealed the best tumor inhibition (the tumor inhibition rate was about 85%) compared with chemotherapy and photothermal therapy alone. Thus, this study supplied a viable multiple-targeted PCT nano-agent for chemo-photothermal combination therapy of liver cancer.


Assuntos
Antineoplásicos/uso terapêutico , Portadores de Fármacos/uso terapêutico , Grafite/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Nanocompostos/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/efeitos da radiação , Linhagem Celular Tumoral , Ácidos Cólicos/química , Terapia Combinada/métodos , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapêutico , Portadores de Fármacos/química , Portadores de Fármacos/efeitos da radiação , Liberação Controlada de Fármacos , Tratamento Farmacológico , Óxido Ferroso-Férrico/química , Ácido Fólico/química , Grafite/química , Grafite/efeitos da radiação , Humanos , Raios Infravermelhos , Fenômenos Magnéticos , Camundongos Endogâmicos BALB C , Nanocompostos/química , Nanocompostos/efeitos da radiação , Terapia Fototérmica
9.
ACS Appl Mater Interfaces ; 13(24): 28802-28817, 2021 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-34109788

RESUMO

In this study, a novel class of multifunctional responsive nanoparticles is designed and fabricated as drug nanocarriers for synergetic chemo-photothermal therapy of tumors. The proposed nanoparticles are composed of a thermo-/pH-responsive poly(N-isopropylacrylamide-co-acrylic acid) (PNA) nanogel core, a polydopamine (PDA) layer for photothermal conversion, and an outer folic acid (FA) layer as a targeting agent for the folate receptors on tumor cells. The fabricated nanoparticles show good biocompatibility and outstanding photothermal conversion efficiency. The proposed nanoparticles loaded with doxorubicin (DOX) drug molecules are stable under physiological conditions with low leakage of drugs, while rapidly release drugs in environments with low pH conditions and at high temperature. The experimental results show that the drug release process is mainly governed by Fickian diffusion. In vitro cell experimental results demonstrate that the PNA-DOX@PDA-FA nanoparticles can be phagocytized by 4T1 tumor cells and release drugs in tumor cell acidic environments, and confirm that the combined chemo and photothermal therapeutic efficacy of PNA-DOX@PDA-FA nanoparticles is higher than the photothermal therapeutic efficacy or the chemotherapeutic efficacy alone. The proposed multifunctional responsive nanoparticles in this study provide a novel class of drug nanocarriers as a promising tool for synergetic chemo-photothermal therapy of tumors.


Assuntos
Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Nanopartículas Multifuncionais/química , Acrilamidas/química , Acrilamidas/metabolismo , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Doxorrubicina/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/efeitos da radiação , Liberação Controlada de Fármacos , Endocitose/fisiologia , Ácido Fólico/análogos & derivados , Ácido Fólico/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Indóis/química , Indóis/metabolismo , Indóis/efeitos da radiação , Raios Infravermelhos , Camundongos , Nanopartículas Multifuncionais/metabolismo , Terapia Fototérmica , Polímeros/química , Polímeros/metabolismo , Polímeros/efeitos da radiação , Temperatura
10.
Carbohydr Polym ; 267: 118152, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34119127

RESUMO

Herein, we demonstrate a novel UV-induced decomposable nanocapsule of natural polysaccharide (HA-azo/PDADMAC). The nanocapsules are fabricated based on layer-by-layer co-assembly of anionic azobenzene functionalized hyaluronic acid (HA-azo) and cationic poly diallyl dimethylammonium chloride (PDADMAC). When the nanocapsules are exposed to 365 nm light, ultraviolet photons can trigger the photo-isomerization of azobenzene groups in the framework. The nanocapsules could decompose from large-sized nanocapsules to small fragments. Due to their optimized original size (~180 nm), the nanocapsules can effectively avoid biological barriers, provide a long blood circulation and achieve high tumor accumulation. It can fast eliminate nanocapsules from tumor and release the loaded drugs for chemotherapy after UV-induced dissociation. Besides, HA is an endogenous polysaccharide that shows intrinsic targetability to CD44 receptors on surface of cancer cells. The intracellular experiment shows that the HA-azo/PDADMAC nanocapsules with CD44 targeting ability and UV-controlled intracellular drug release are promising for cancer chemotherapy.


Assuntos
Compostos Azo/química , Portadores de Fármacos/química , Ácido Hialurônico/química , Nanocápsulas/química , Antineoplásicos/química , Compostos Azo/metabolismo , Compostos Azo/efeitos da radiação , Compostos Azo/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/efeitos da radiação , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos/efeitos da radiação , Endocitose/fisiologia , Células Hep G2 , Humanos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/síntese química , Ácido Hialurônico/metabolismo , Ácido Hialurônico/toxicidade , Nanocápsulas/efeitos da radiação , Nanocápsulas/toxicidade , Nanopartículas/química , Nanopartículas/metabolismo , Nanopartículas/toxicidade , Polietilenos/química , Polietilenos/toxicidade , Compostos de Amônio Quaternário/química , Compostos de Amônio Quaternário/toxicidade , Dióxido de Silício/síntese química , Dióxido de Silício/química , Dióxido de Silício/toxicidade , Estereoisomerismo , Raios Ultravioleta
11.
Eur J Pharm Biopharm ; 165: 374-382, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34038797

RESUMO

Gemcitabine and nab-paclitaxel (Abraxane®) is a standard of care chemotherapy combination used in the treatment of patients with advanced pancreatic cancer. While the combination has shown a survival benefit when compared to gemcitabine monotherapy, it is associated with significant off-target toxicity. Ultrasound targeted microbubble destruction (UTMD) has emerged as an effective strategy for the site-specific deposition of drug-payloads. However, loading a single microbubble formulation with two drug payloads can be challenging and often involves several manipulations post-microbubble preparation that can be cumbersome and generally results in low / inconsistent drug loadings. In this manuscript, we report the one-pot synthesis of a gemcitabine functionalised phospholipid and use it to successfully generate stable microbubble formulations loaded with gemcitabine (Lipid-Gem MB) or a combination of gemcitabine and paclitaxel (Lipid-Gem-PTX MB). Efficacy of the Lipid-Gem MB and Lipid-Gem-PTX MB formulations, following ultrasound (US) stimulation, was evaluated in a three-dimensional (3D) PANC-1 spheroid model of pancreatic cancer and a mouse model bearing ectopic BxPC-3 tumours. The results demonstrated a significant reduction in the cell viability in spheroids for both formulations reducing from 90 ± 10% to 62 ± 5% for Lipid-Gem MB and 84 ± 10% to 30 ± 6% Lipid-Gem-PTX MB following US irradiation. When compared with a clinically relevant dose of free gemcitabine and paclitaxel (i.e. non-particle bound) in a BxPC-3 murine pancreatic tumour model, both formulations also improved tumour growth delay with tumours 40 ± 20% and 40 ± 30% smaller than the respective free drug formulation when treated with Lipid-Gem MB and Lipid-Gem-PTX MB respectively, at the conclusion of the experiment. These results highlight the potential of UTMD mediated Gem / PTX as a treatment for pancreatic cancer and the facile preparation of Lipid-Gem-PTX MBs using a gemcitabine functionalised lipid should expedite clinical translation of this technology.


Assuntos
Albuminas/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Desoxicitidina/análogos & derivados , Portadores de Fármacos/efeitos da radiação , Paclitaxel/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Albuminas/farmacocinética , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Linhagem Celular Tumoral , Desoxicitidina/administração & dosagem , Desoxicitidina/farmacocinética , Portadores de Fármacos/química , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos/efeitos da radiação , Feminino , Humanos , Masculino , Camundongos , Microbolhas , Nanopartículas/química , Nanopartículas/efeitos da radiação , Paclitaxel/farmacocinética , Neoplasias Pancreáticas/patologia , Fosfolipídeos/química , Ondas Ultrassônicas , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
12.
J Inorg Biochem ; 220: 111458, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33857697

RESUMO

There has been growing interest in the application of gold nanorods (GNRs) to tumor therapy due to the unique properties they possess. In the past, GNRs were not used in clinical treatments as they lacked stability in vivo and were characterized by potential toxicity. Despite these issues, the significant potential for utilizing GNRs to conduct safe and effective treatments for tumors cannot be ignored. Therefore, it remains crucial to thoroughly investigate the mechanisms behind the toxicity of GNRs in order to provide the means of overcoming obstacles to its full application in the future. This review presents the toxic effects of GNRs, the factors affecting toxicity and the methods to improve biocompatibility, all of which are presently being studied. Finally, we conclude by briefly discussing the current research status of GNRs and provide additional perspective on the challenges involved along with the course of development for GNRs in the future.


Assuntos
Antineoplásicos/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Nanotubos/química , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/efeitos da radiação , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Portadores de Fármacos/química , Portadores de Fármacos/efeitos da radiação , Portadores de Fármacos/uso terapêutico , Tratamento Farmacológico , Ouro/química , Ouro/efeitos da radiação , Humanos , Nanopartículas Metálicas/química , Nanopartículas Metálicas/efeitos da radiação , Nanotubos/efeitos da radiação , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/efeitos da radiação , Fármacos Fotossensibilizantes/uso terapêutico , Terapia Fototérmica
13.
ACS Appl Mater Interfaces ; 13(9): 10674-10688, 2021 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-33621058

RESUMO

Cyclodextrins (CDs), as pharmaceutical excipients with excellent biocompatibility, non-immunogenicity, and low toxicity in vivo, are widely used to carry drugs by forming inclusion complexes for improving the solubility and stability of drugs. However, the limited space of CDs' lipophilic central cavity affects the loading of many drugs, especially with larger molecules. In this study, ß-CDs were modified by acetonization to improve the affinity for the chemotherapy drug doxorubicin (DOX), and doxorubicin-adsorbing acetalated ß-CDs (Ac-CD:DOX) self-assembled to nanoparticles, followed by coating with the amphiphilic zinc phthalocyanine photosensitizer ZnPc-(PEG)5 for antitumor therapy. The final product ZnPc-(PEG)5:Ac-CD:DOX was demonstrated to have excellent stability and pH-sensitive drug release characteristics. The cell viability and apoptosis assay showed synergistic cytotoxic effects of chemotherapy and phototherapy. The mechanism of cytotoxicity was analyzed in terms of intracellular reactive oxygen species, mitochondrial membrane potential, and subcellular localization. More importantly, in vivo experiments indicated that ZnPc-(PEG)5:Ac-CD:DOX possessed significant tumor targeting, prominent antitumor activity, and less side effects. Our strategy expands the application of CDs as drug carriers and provides new insights into the development of CD chemistry.


Assuntos
Antineoplásicos/uso terapêutico , Doxorrubicina/uso terapêutico , Portadores de Fármacos/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Doxorrubicina/química , Portadores de Fármacos/síntese química , Portadores de Fármacos/efeitos da radiação , Liberação Controlada de Fármacos , Sinergismo Farmacológico , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Indóis/síntese química , Indóis/efeitos da radiação , Indóis/uso terapêutico , Isoindóis , Luz , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Nanopartículas/química , Nanopartículas/efeitos da radiação , Compostos Organometálicos/síntese química , Compostos Organometálicos/efeitos da radiação , Compostos Organometálicos/uso terapêutico , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/efeitos da radiação , Fármacos Fotossensibilizantes/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Compostos de Zinco , beta-Ciclodextrinas/síntese química , beta-Ciclodextrinas/efeitos da radiação , beta-Ciclodextrinas/uso terapêutico
14.
ACS Appl Mater Interfaces ; 12(51): 56767-56781, 2020 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-33289550

RESUMO

The consolidation of nanovectors with biological membranes has recently been a subject of interest owing to the prolonged systemic circulation time and delayed clearance by the reticuloendothelial system of such systems. Among the different biomembranes, the macrophage membrane has a similar systemic circulation time, with an additional chemotactic aptitude, targeting integrin proteins. In this study, we aimed to establish a laser-activated, disintegrable, and deeply tumor-penetrative nanoplatform. We used a highly tumor-ablative and laser-responsive disintegrable copper sulfide nanoparticle, loaded it with paclitaxel, and camouflaged it with the macrophage membrane for the fabrication of PTX@CuS@MMNPs. The in vitro paclitaxel release profile was favorable for release in the tumor microenvironment, and the release was accelerated after laser exposure. Cellular internalization was improved by membrane encapsulation. Cellular uptake, cytotoxicity, reactive oxygen species generation, and apoptosis induction of PTX@CuS@MMNPs were further improved upon laser exposure, and boosted permeation was achieved by co-administration of the tumor-penetrating peptide iRGD. In vivo tumor accumulation, tumor inhibition rate, and apoptotic marker expression induced by PTX@CuS@MMNPs were significantly improved by laser irradiation and iRGD co-administration. PTX@CuS@MMNPs induced downregulation of cellular proliferation and angiogenic markers but no significant changes in body weight, survival, or significant toxicities in vital organs after laser exposure, suggesting their biocompatibility. The disintegrability of the nanosystem, accredited to biodegradability, favored efficient elimination from the body. In conclusion, PTX@CuS@MMNPs showed promising traits in combination therapies for excellent tumor eradication.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Membrana Celular/química , Macrófagos/química , Nanopartículas Metálicas/química , Neoplasias/tratamento farmacológico , Paclitaxel/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cobre/química , Cobre/efeitos da radiação , Cobre/toxicidade , Portadores de Fármacos/química , Portadores de Fármacos/efeitos da radiação , Portadores de Fármacos/toxicidade , Raios Infravermelhos , Nanopartículas Metálicas/efeitos da radiação , Nanopartículas Metálicas/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7
15.
Nat Commun ; 11(1): 4929, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33004789

RESUMO

Non-invasive, molecularly-specific, focal modulation of brain circuits with low off-target effects can lead to breakthroughs in treatments of brain disorders. We systemically inject engineered ultrasound-controllable drug carriers and subsequently apply a novel two-component Aggregation and Uncaging Focused Ultrasound Sequence (AU-FUS) at the desired targets inside the brain. The first sequence aggregates drug carriers with millimeter-precision by orders of magnitude. The second sequence uncages the carrier's cargo locally to achieve high target specificity without compromising the blood-brain barrier (BBB). Upon release from the carriers, drugs locally cross the intact BBB. We show circuit-specific manipulation of sensory signaling in motor cortex in rats by locally concentrating and releasing a GABAA receptor agonist from ultrasound-controlled carriers. Our approach uses orders of magnitude (1300x) less drug than is otherwise required by systemic injection and requires very low ultrasound pressures (20-fold below FDA safety limits for diagnostic imaging). We show that the BBB remains intact using passive cavitation detection (PCD), MRI-contrast agents and, importantly, also by sensitive fluorescent dye extravasation and immunohistochemistry.


Assuntos
Barreira Hematoencefálica/metabolismo , Encefalopatias/tratamento farmacológico , Portadores de Fármacos/efeitos da radiação , Agonistas de Receptores de GABA-A/administração & dosagem , Ultrassonografia de Intervenção/métodos , Animais , Barreira Hematoencefálica/diagnóstico por imagem , Barreira Hematoencefálica/efeitos da radiação , Relação Dose-Resposta à Radiação , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Feminino , Agonistas de Receptores de GABA-A/farmacocinética , Humanos , Imageamento por Ressonância Magnética , Modelos Animais , Muscimol/administração & dosagem , Muscimol/farmacocinética , Ratos , Técnicas Estereotáxicas , Ondas Ultrassônicas
16.
Theranostics ; 10(23): 10448-10465, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32929359

RESUMO

Rationale: Insufficient penetration and accumulation of theranostic payloads in solid tumors greatly challenge the clinical translation of cancer nanomedicines. To address this challenge, we synthesized natural melanin-cored and doxorubicin-loaded perfluoropentane nanodroplets with good biocompatibility and self-assembling ability. Methods: We used an opto-acoustic synergistic irradiation (OASI) method that was effective at lower energy levels than ultrasound- or laser-only irradiation to safely vaporize the nanodroplets and to cavitate the generated microbubbles for mechanically enhancing intratumoral delivery. The delivered melanin and doxorubicin inside the tumors mediated secondary chemo-photothermal therapy under laser irradiation to fully kill cancer cells. Results:In vivo animal experiments demonstrated direct mechanical disruption of tumor structures (H&E staining), enhanced intratumoral penetration of melanin (photoacoustic imaging), and efficient intratumoral accumulation of doxorubicin (fluorescent imaging). Anti-tumor experiments demonstrated that the nanodroplets combined with OASI treatment and subsequent laser irradiation could efficiently eliminate melanoma tumors. Conclusion: Melanin-cored and doxorubicin-loaded perfluoropentane nanodroplets hold great promise for translational sono-chemo-photothermal cancer therapy.


Assuntos
Doxorrubicina/administração & dosagem , Portadores de Fármacos/efeitos da radiação , Microbolhas/uso terapêutico , Neoplasias/terapia , Nanomedicina Teranóstica/métodos , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Feminino , Fluorocarbonos/química , Humanos , Hipertermia Induzida/métodos , Terapia a Laser/métodos , Melaninas/química , Melaninas/efeitos da radiação , Camundongos , Nanopartículas/química , Nanopartículas/efeitos da radiação , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Técnicas Fotoacústicas/métodos , Fotoquimioterapia/métodos , Terapia por Ultrassom/métodos , Volatilização/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Mol Pharm ; 17(10): 3885-3899, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32787269

RESUMO

Boron neutron capture therapy (BNCT) for cancer is on the rise worldwide due to recent developments of in-hospital neutron accelerators which are expected to revolutionize patient treatments. There is an urgent need for improved boron delivery agents, and herein we have focused on studying the biochemical foundations upon which a successful GLUT1-targeting strategy to BNCT could be based. By combining synthesis and molecular modeling with affinity and cytotoxicity studies, we unravel the mechanisms behind the considerable potential of appropriately designed glucoconjugates as boron delivery agents for BNCT. In addition to addressing the biochemical premises of the approach in detail, we report on a hit glucoconjugate which displays good cytocompatibility, aqueous solubility, high transporter affinity, and, crucially, an exceptional boron delivery capacity in the in vitro assessment thereby pointing toward the significant potential embedded in this approach.


Assuntos
Terapia por Captura de Nêutron de Boro/métodos , Boro/administração & dosagem , Portadores de Fármacos/efeitos da radiação , Glucose/efeitos da radiação , Isótopos/administração & dosagem , Neoplasias/radioterapia , Boro/farmacocinética , Linhagem Celular Tumoral , Portadores de Fármacos/síntese química , Portadores de Fármacos/farmacocinética , Liberação Controlada de Fármacos/efeitos da radiação , Glucose/análogos & derivados , Glucose/síntese química , Glucose/farmacocinética , Transportador de Glucose Tipo 1/metabolismo , Humanos , Isótopos/farmacocinética , Simulação de Acoplamento Molecular
18.
Mol Pharm ; 17(10): 3900-3914, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32820927

RESUMO

Erythrocyte-derived particles activated by near-infrared (NIR) light present a platform for various phototheranostic applications. We have engineered such a platform with indocyanine green as the NIR-activated agent. A particular feature of these particles is that their diameters can be tuned from micro- to nanoscale, providing a potential capability for broad clinical utility ranging from vascular to cancer-related applications. An important issue related to clinical translation of these particles is their immunogenic effects. Herein, we have evaluated the early-induced innate immune response of these particles in healthy Swiss Webster mice following tail vein injection by measurements of specific cytokines in blood serum, the liver, and the spleen following euthanasia. In particular, we have investigated the effects of particle size and relative dose, time-dependent cytokine response for up to 6 h postinjection, functionalization of the nanosized particles with folate or Herceptin, and dual injections of the particles 1 week apart. Mean concentrations of interleukin (IL)-6, IL-10, tumor necrosis factor (TNF)-α, and monocyte chemoattractant protein (MCP)-1 in response to injection of microsized particles at the investigated relative doses were significantly lower than the corresponding mean concentrations induced by lipopolysaccharide (positive control) at 2 h. All investigated doses of the nanosized particles induced significantly higher concentrations of MCP-1 in the liver and the spleen as compared to phosphate buffer saline (PBS) (negative control) at 2 h. In response to micro- and nanosized particles at the highest investigated dose, there were significantly higher levels of TNF-α in blood serum at 2 and 6 h postinjection as compared to the levels associated with PBS treatment at these times. Whereas the mean concentration of TNF-α in the liver significantly increased between 2 and 6 h postinjection in response to the injection of the microsized particles, it was significantly reduced during this time interval in response to the injection of the nanosized particles. In general, functionalization of the nanosized particles was associated with a reduction of IL-6 and MCP-1 in blood serum, the liver, and the spleen, and TNF-α in blood serum. With the exception of IL-10 in the spleen in response to nanosized particles, the second injection of micro- or nanosized particles did not lead to significantly higher concentrations of other cytokines at the investigated dose as compared to a single injection.


Assuntos
Portadores de Fármacos/efeitos adversos , Eritrócitos/química , Imunidade/efeitos dos fármacos , Fototerapia/métodos , Nanomedicina Teranóstica/métodos , Animais , Citocinas/análise , Citocinas/metabolismo , Relação Dose-Resposta Imunológica , Esquema de Medicação , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Portadores de Fármacos/efeitos da radiação , Eritrócitos/imunologia , Feminino , Raios Infravermelhos , Injeções Intravenosas , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/metabolismo , Camundongos , Modelos Animais , Nanopartículas/administração & dosagem , Nanopartículas/efeitos adversos , Nanopartículas/química , Nanopartículas/efeitos da radiação , Tamanho da Partícula , Fototerapia/efeitos adversos , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo
19.
Mol Pharm ; 17(10): 3720-3729, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32633977

RESUMO

The limited tumor tissue penetration of many nanoparticles remains a formidable challenge to their therapeutic efficacy. Although several photonanomedicines have been applied to improve tumor penetration, the first near-infrared window mediated by the low optical tissue penetration depth severely limits their anticancer effectiveness. To achieve deep optical tissue and drug delivery penetration, a near-infrared second window (NIR-II)-excited and pH-responsive ultrasmall drug delivery nanoplatform was fabricated based on BSA-stabilized CuS nanoparticles (BSA@CuS NPs). The BSA@CuS NPs effectively encapsulated doxorubicin (DOX) via strong electrostatic interactions to form multifunctional nanoparticles (BSA@CuS@DOX NPs). The BSA@CuS@DOX NPs had an ultrasmall size, which allowed them to achieve deeper tumor penetration. They also displayed stronger NIR II absorbance-mediated deep optical tissue penetration than that of the NIR I window. Moreover, the multifunctional nanoplatform preferentially accumulated in tumor sites, induced tumor hyperthermia, and generated remarkably high ROS levels in tumor sites upon NIR-II laser (1064 nm) irradiation. More importantly, our strategy achieved excellent synergistic effects of chemotherapy and phototherapy (chemophototherapy) under the guidance of photothermal imaging. The developed nanoparticles also showed good biocompatibility and bioclearance properties. Therefore, our work demonstrated a facile strategy for fabricating a multifunctional nanoplatform that is a promising candidate for deep tumor penetration as an effective antitumor therapy.


Assuntos
Doxorrubicina/administração & dosagem , Portadores de Fármacos/efeitos da radiação , Nanopartículas/efeitos da radiação , Neoplasias/tratamento farmacológico , Fototerapia/métodos , Animais , Linhagem Celular Tumoral/transplante , Sobrevivência Celular , Modelos Animais de Doenças , Doxorrubicina/farmacocinética , Portadores de Fármacos/química , Liberação Controlada de Fármacos/efeitos da radiação , Humanos , Concentração de Íons de Hidrogênio , Raios Infravermelhos , Lasers , Camundongos , Nanopartículas/química , Neoplasias/patologia , Fototerapia/instrumentação , Distribuição Tecidual
20.
J Nanobiotechnology ; 18(1): 91, 2020 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-32539777

RESUMO

BACKGROUND: Stimulus-responsive degradable mesoporous organosilica nanoparticles (MONs) have shown great promise as drug carriers via enhancing the efficiency of drug delivery and accelerating the degradation of nanocarriers. However, it remains a great challenge to develop novel light-enabled spatial and temporal degradable MONs with both superior responsiveness for efficient anti-cancer drug delivery and safe exocytosis. RESULTS: We report a novel photo-responsive degradable hollow mesoporous organosilica nanoplatform (HMONs@GOQD). The platform is based on organosilica nanoparticles (HMONs) containing singlet oxygen (1O2)-responsive bridged organoalkoxysilanes and wrapped graphene oxide quantum dots (GOQDs). The unique hollow mesoporous structure of the HMONs guarantees an excellent drug loading and release profile. During light irradiation, 1O2 produced by the GOQDs leads to the degradation of the organosilica nanoparticles, resulting in enhanced local drug release. CONCLUSIONS: We carried out in vitro and in vivo experiments using DOX as a model drug; DOX-HMONs@GOQDs exhibited high biocompatibility, accelerated degradation, and superior therapeutic efficacy during light irradiation, indicating a promising platform for clinical cancer therapy.


Assuntos
Portadores de Fármacos , Compostos de Organossilício/química , Pontos Quânticos , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/efeitos da radiação , Grafite/química , Masculino , Camundongos , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Processos Fotoquímicos , Pontos Quânticos/química , Pontos Quânticos/metabolismo , Pontos Quânticos/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA