Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 326
Filtrar
1.
J Mater Chem B ; 12(14): 3509-3520, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38516824

RESUMO

Both chemodynamic therapy and photodynamic therapy, based on the production of reactive oxygen (ROS), have excellent potential in cancer therapy. However, the abnormal redox homeostasis in tumor cells, especially the overexpressed glutathione (GSH) could scavenge ROS and reduce the anti-tumor efficiency. Therefore, it is essential to develop a simple and effective tumor-specific drug delivery system for modulating the tumor microenvironment (TME) and achieving synergistic therapy at the tumor site. In this study, self-assembled nanoparticles (named CDZP NPs) were developed using copper ion (Cu2+), doxorubicin (Dox), zinc phthalocyanine (ZnPc) and a trace amount of poly(2-(di-methylamino)ethylmethacrylate)-poly[(R)-3-hydroxybutyrate]-poly(2-(dimethylamino)ethylmethacrylate) (PDMAEMA-PHB-PDMAEMA) through chelation, π-π stacking and hydrophobic interaction. These triple factor-responsive (pH, laser and GSH) nanoparticles demonstrated unique advantages through the synergistic effect. Highly controllable drug release ensured its effectiveness at the tumor site, Dox-induced chemotherapy and ZnPc-mediated fluorescence (FL) imaging exhibited the distribution of nanoparticles. Meanwhile, Cu2+-mediated GSH-consumption not only reduced the intracellular ROS elimination but also produced Cu+ to catalyze hydrogen peroxide (H2O2) and generated hydroxyl radicals (˙OH), thereby enhancing the chemodynamic and photodynamic therapy. Herein, this study provides a green and relatively simple method for preparing multifunctional nanoparticles that can effectively modulate the TME and improve synergetic cancer therapy.


Assuntos
Metacrilatos , Metilmetacrilatos , Nanopartículas , Neoplasias , Nylons , Humanos , Cobre/uso terapêutico , Espécies Reativas de Oxigênio , Peróxido de Hidrogênio/uso terapêutico , Nanopartículas/química , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Neoplasias/tratamento farmacológico , Glutationa/química , Oxirredução , Microambiente Tumoral
2.
Nanoscale ; 16(7): 3755-3763, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38299362

RESUMO

The therapeutic outcome of chemodynamic therapy (CDT) is greatly hindered by the presence of oxidative damage repair proteins (MTH1) inside cancer cells. These oxidative damage repair proteins detoxify the action of radicals generated by Fenton or Fenton-like reactions. Hence, it is extremely important to develop a simple strategy for the downregulation of MTH1 protein inside cancer cells along with the delivery of metal ions into cancer cells. A one-pot host-guest supramolecular approach for the codelivery of MTH1 siRNA and metal ions into a cancer cell is reported. Our approach involves the fabrication of an inclusion complex between cationic ß-cyclodextrin and a ferrocene prodrug, which spontaneously undergoes amphiphilicity-driven self-assembly to form spherical nanoparticles (NPs) having a positively charged surface. The cationic surface of the NPs was then explored for the loading of MTH1 siRNA through electrostatic interactions. Using HeLa cells as a representative example, efficient uptake of the NPs, delivery of MTH1 siRNA and the enhanced CDT of the nanoformulation are demonstrated. This work highlights the potential of the supramolecular approach as a simple yet efficient method for the delivery of siRNA across the cell membrane for enhanced chemodynamic therapy.


Assuntos
Ciclodextrinas , Compostos Ferrosos , Nanopartículas , Neoplasias , Humanos , RNA Interferente Pequeno , Células HeLa , Metalocenos/farmacologia , Nanopartículas/uso terapêutico , Cátions , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Peróxido de Hidrogênio/uso terapêutico
3.
Nanoscale ; 16(8): 4095-4104, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38333905

RESUMO

In recent years, tumor catalytic therapy based on nanozymes has attracted widespread attention. However, its application is limited by the tumor hypoxic microenvironment (TME). In this study, we developed oxygen-supplying magnetic bead nanozymes that integrate hemoglobin and encapsulate the photosensitizer curcumin, demonstrating reactive oxygen species (ROS)-induced synergistic breast cancer therapy. Fe3O4 magnetic bead-mediated catalytic dynamic therapy (CDT) generates hydroxyl radicals (˙OH) through the Fenton reaction in the tumor microenvironment. The Hb-encapsulated Fe3O4 magnetic beads can be co-loaded with the photosensitizer/chemotherapeutic agent curcumin (cur), resulting in Fe3O4-Hb@cur. Under hypoxic conditions, oxygen molecules are released from Fe3O4-Hb@cur to overcome the TME hypoxia, resulting in comprehensive effects favoring anti-tumor responses. Upon near-infrared (NIR) irradiation, Fe3O4-Hb@cur activates the surrounding molecular oxygen to generate a certain amount of singlet oxygen (1O2), which is utilized for photodynamic therapy (PDT) in cancer treatment. Meanwhile, we validated that the O2 carried by Hb significantly enhances the intracellular ROS level, intensifying the catalytic therapy mediated by Fe3O4 magnetic beads and inflicting lethal damage to cancer cells, effectively inhibiting tumor growth. Therefore, significant in vivo synergistic therapeutic effects can be achieved through catalytic-photodynamic combination therapy.


Assuntos
Neoplasias da Mama , Curcumina , Neoplasias , Fotoquimioterapia , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Oxigênio , Espécies Reativas de Oxigênio/farmacologia , Curcumina/farmacologia , Curcumina/uso terapêutico , Linhagem Celular Tumoral , Fotoquimioterapia/métodos , Neoplasias/tratamento farmacológico , Hipóxia , Fenômenos Magnéticos , Microambiente Tumoral , Peróxido de Hidrogênio/uso terapêutico
4.
Int J Mol Sci ; 25(2)2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38279349

RESUMO

Retinal ischemia plays a vital role in vision-threatening retinal ischemic disorders, such as diabetic retinopathy, age-related macular degeneration, glaucoma, etc. The aim of this study was to investigate the effects of S-allyl L-cysteine (SAC) and its associated therapeutic mechanism. Oxidative stress was induced by administration of 500 µM H2O2 for 24 h; SAC demonstrated a dose-dependent neuroprotective effect with significant cell viability effects at 100 µM, and it concurrently downregulated angiogenesis factor PKM2 and inflammatory biomarker MCP-1. In a Wistar rat model of high intraocular pressure (HIOP)-induced retinal ischemia and reperfusion (I/R), post-administration of 100 µM SAC counteracted the ischemic-associated reduction of ERG b-wave amplitude and fluorogold-labeled RGC reduction. This study supports that SAC could protect against retinal ischemia through its anti-oxidative, anti-angiogenic, anti-inflammatory, and neuroprotective properties.


Assuntos
Glaucoma , Fármacos Neuroprotetores , Traumatismo por Reperfusão , Doenças Retinianas , Ratos , Animais , Ratos Wistar , Cisteína/farmacologia , Cisteína/uso terapêutico , Peróxido de Hidrogênio/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Doenças Retinianas/tratamento farmacológico , Isquemia/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Glaucoma/tratamento farmacológico
5.
Nanoscale ; 16(4): 1983-1998, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38189459

RESUMO

Acute lung injury (ALI) is an inflammatory disease caused by multiple factors such as infection, trauma, and chemicals. Without effective intervention during the early stages, it usually quickly progresses to acute respiratory distress syndrome (ARDS). Since ordinary pharmaceutical preparations cannot precisely target the lungs, their clinical application is limited. In response, we constructed a γ3 peptide-decorated and ROS-responsive nanoparticle system encapsulating therapeutic dexamethasone (Dex/PSB-γ3 NPs). In vitro, Dex/PSB-γ3 NPs had rapid H2O2 responsiveness, low cytotoxicity, and strong intracellular ROS removal capacity. In a mouse model of ALI, Dex/PSB-γ3 NPs accumulated at the injured lung rapidly, alleviating pulmonary edema and cytokine levels significantly. The modification of NPs by γ3 peptide achieved highly specific positioning of NPs in the inflammatory area. The ROS-responsive release mechanism ensured the rapid release of therapeutic dexamethasone at the inflammatory site. This combined approach improves treatment accuracy, and drug bioavailability, and effectively inhibits inflammation progression. Our study could effectively reduce the risk of ALI progressing to ARDS and hold potential for the early treatment of ALI.


Assuntos
Lesão Pulmonar Aguda , Nanopartículas , Síndrome do Desconforto Respiratório , Camundongos , Animais , Espécies Reativas de Oxigênio/farmacologia , Molécula 1 de Adesão Intercelular , Peróxido de Hidrogênio/uso terapêutico , Lesão Pulmonar Aguda/tratamento farmacológico , Pulmão , Síndrome do Desconforto Respiratório/tratamento farmacológico , Nanopartículas/uso terapêutico , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Dexametasona/farmacologia , Dexametasona/uso terapêutico
6.
J Clin Invest ; 134(3)2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37971886

RESUMO

While the poor prognosis of glioblastoma arises from the invasion of a subset of tumor cells, little is known of the metabolic alterations within these cells that fuel invasion. We integrated spatially addressable hydrogel biomaterial platforms, patient site-directed biopsies, and multiomics analyses to define metabolic drivers of invasive glioblastoma cells. Metabolomics and lipidomics revealed elevations in the redox buffers cystathionine, hexosylceramides, and glucosyl ceramides in the invasive front of both hydrogel-cultured tumors and patient site-directed biopsies, with immunofluorescence indicating elevated reactive oxygen species (ROS) markers in invasive cells. Transcriptomics confirmed upregulation of ROS-producing and response genes at the invasive front in both hydrogel models and patient tumors. Among oncologic ROS, H2O2 specifically promoted glioblastoma invasion in 3D hydrogel spheroid cultures. A CRISPR metabolic gene screen revealed cystathionine γ-lyase (CTH), which converts cystathionine to the nonessential amino acid cysteine in the transsulfuration pathway, to be essential for glioblastoma invasion. Correspondingly, supplementing CTH knockdown cells with exogenous cysteine rescued invasion. Pharmacologic CTH inhibition suppressed glioblastoma invasion, while CTH knockdown slowed glioblastoma invasion in vivo. Our studies highlight the importance of ROS metabolism in invasive glioblastoma cells and support further exploration of the transsulfuration pathway as a mechanistic and therapeutic target.


Assuntos
Glioblastoma , Humanos , Glioblastoma/patologia , Cistationina/uso terapêutico , Cisteína/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Peróxido de Hidrogênio/uso terapêutico , Multiômica , Hidrogéis
7.
J Mater Chem B ; 11(41): 9798-9839, 2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37842806

RESUMO

Nanomaterial-based cancer therapy faces significant limitations due to the complex nature of the tumor microenvironment (TME). Starvation therapy is an emerging therapeutic approach that targets tumor cell metabolism using glucose oxidase (GOx). Importantly, it can provide a material or environmental foundation for other diverse therapeutic methods by manipulating the properties of the TME, such as acidity, hydrogen peroxide (H2O2) levels, and hypoxia degree. In recent years, this cascade strategy has been extensively applied in nanoplatforms for ongoing synergetic therapy and still holds undeniable potential. However, only a few review articles comprehensively elucidate the rational designs of nanoplatforms for synergetic therapeutic regimens revolving around the conception of the cascade strategy. Therefore, this review focuses on innovative cascade strategies for GOx-based synergetic therapy from representative paradigms to state-of-the-art reports to provide an instructive, comprehensive, and insightful reference for readers. Thereafter, we discuss the remaining challenges and offer a critical perspective on the further advancement of GOx-facilitated cancer treatment toward clinical translation.


Assuntos
Nanopartículas , Nanoestruturas , Neoplasias , Humanos , Glucose Oxidase/uso terapêutico , Glucose Oxidase/metabolismo , Peróxido de Hidrogênio/uso terapêutico , Neoplasias/terapia , Nanoestruturas/uso terapêutico , Microambiente Tumoral
8.
Radiat Res ; 200(5): 456-461, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37758035

RESUMO

Diffuse intrinsic pontine gliomas (DIPG) are an aggressive type of pediatric brain tumor with a very high mortality rate. Surgery has a limited role given the tumor's location. Palliative radiation therapy alleviates symptoms and prolongs survival, but median survival remains less than 1 year. There is no clear role for chemotherapy in DIPGs as trials adding chemotherapy to palliative radiation therapy have failed to improve survival compared to radiation alone. Thus, there is a critical need to identify tissue-specific radiosensitizers to improve clinical outcomes for patients with DIPGs. Pharmacologic (high dose) ascorbate (P-AscH-) is a promising anticancer therapy that sensitizes human tumors, including adult high-grade gliomas, to radiation by acting selectively as a generator of hydrogen peroxide (H2O2) in cancer cells. In this study we demonstrate that in contrast to adult glioma models, P-AscH- does not radiosensitize DIPG. DIPG cells were sensitive to bolus of H2O2 but have faster H2O2 removal rates than GBM models which are radiosensitized by P-AscH-. These data support the hypothesis that P-AscH- does not enhance DIPG radiosensitivity, likely due to a robust capacity to detoxify and remove hydroperoxides.


Assuntos
Antineoplásicos , Neoplasias do Tronco Encefálico , Glioma Pontino Intrínseco Difuso , Glioma , Criança , Adulto , Humanos , Glioma Pontino Intrínseco Difuso/tratamento farmacológico , Glioma Pontino Intrínseco Difuso/patologia , Neoplasias do Tronco Encefálico/radioterapia , Neoplasias do Tronco Encefálico/patologia , Peróxidos/uso terapêutico , Peróxido de Hidrogênio/farmacologia , Peróxido de Hidrogênio/uso terapêutico , Glioma/radioterapia , Glioma/patologia , Antineoplásicos/uso terapêutico
9.
Radiat Res ; 200(5): 444-455, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37758045

RESUMO

Pharmacological ascorbate (P-AscH-, high dose, intravenous vitamin C) preferentially sensitizes human pancreas ductal adenocarcinoma (PDAC) cells to radiation-induced toxicity compared to non-tumorigenic epithelial cells. Radiation-induced G2-checkpoint activation contributes to the resistance of cancer cells to DNA damage induced toxicity. We hypothesized that P-AscH- induced radio-sensitization of PDAC cells is mediated by perturbations in the radiation induced activation of the G2-checkpoint pathway. Both non-tumorigenic pancreatic ductal epithelial and PDAC cells display decreased clonogenic survival and increased doubling times after radiation treatment. In contrast, the addition of P-AscH- to radiation increases clonogenic survival and decreases the doubling time of non-tumorigenic epithelial cells but decreasing clonogenic survival and increasing the doubling time of PDAC cells. Results from the mitotic index and propidium iodide assays showed that while the P-AscH- treatments did not affect radiation-induced G2-checkpoint activation, it enhanced G2-accumulation. The addition of catalase reverses the increases in G2-accumulation, indicating a peroxide-mediated mechanism. In addition, P-AscH- treatment of PDAC cells suppresses radiation-induced accumulation of cyclin B1 protein levels. Both translational and post-translational pathways appear to regulate cyclin B1 protein levels after the combination treatment of PDAC cells with P-AscH- and radiation. The protein changes seen are reversed by the addition of catalase suggesting that hydrogen peroxide mediates P-AscH- induced radiation sensitization of PDAC cells by enhancing G2-accumulation and reducing cyclin B1 protein levels.


Assuntos
Antineoplásicos , Neoplasias Pancreáticas , Humanos , Catalase/metabolismo , Catalase/uso terapêutico , Peróxido de Hidrogênio/farmacologia , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/uso terapêutico , Ciclina B1 , Linhagem Celular Tumoral , Neoplasias Pancreáticas/tratamento farmacológico , Antineoplásicos/farmacologia , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas
10.
Nanoscale ; 15(33): 13498-13514, 2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37578148

RESUMO

With the recent interest in the role of oxidative species/radicals in diseases, inorganic nanomaterials with redox activities have been extensively investigated for their potential use in nanomedicine. While many studies focusing on relieving oxidative stress to prevent pathogenesis and to suppress the progression of diseases have shown considerable success, another approach for increasing oxidative stress using nanomaterials to kill malignant cells has suffered from low efficiency despite its wide applicability to various targets. Chemodynamic therapy (CDT) is an emerging technique that can resolve such a problem by exploiting the characteristic tumour microenvironment to achieve high selectivity. In this review, we summarize the recent strategies and underlying mechanisms that have been used to improve the CDT performance using inorganic nanoparticles. In addition to the design of CDT agents, the effects of contributing factors, such as the acidity and the levels of hydrogen peroxide and antioxidants in the tumour microenvironment, together with their modulation and application in combination therapy, are presented. The challenges lying ahead of future clinical translation of this rapidly advancing technology are also discussed.


Assuntos
Nanopartículas , Nanoestruturas , Neoplasias , Humanos , Neoplasias/patologia , Nanopartículas/uso terapêutico , Nanomedicina , Oxirredução , Peróxido de Hidrogênio/uso terapêutico , Microambiente Tumoral , Linhagem Celular Tumoral
11.
Angew Chem Int Ed Engl ; 62(44): e202308761, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37496129

RESUMO

Enzymatic reactions can consume endogenous nutrients of tumors and produce cytotoxic species and are therefore promising tools for treating malignant tumors. Inspired by nature where enzymes are compartmentalized in membranes to achieve high reaction efficiency and separate biological processes with the environment, we develop liposomal nanoreactors that can perform enzymatic cascade reactions in the aqueous nanoconfinement of liposomes. The nanoreactors effectively inhibited tumor growth in vivo by consuming tumor nutrients (glucose and oxygen) and producing highly cytotoxic hydroxyl radicals (⋅OH). Co-compartmentalization of glucose oxidase (GOx) and horseradish peroxidase (HRP) in liposomes could increase local concentration of the intermediate product hydrogen peroxide (H2 O2 ) as well as the acidity due to the generation of gluconic acid by GOx. Both H2 O2 and acidity accelerate the second-step reaction by HRP, hence improving the overall efficiency of the cascade reaction. The biomimetic compartmentalization of enzymatic tandem reactions in biocompatible liposomes provides a promising direction for developing catalytic nanomedicines in antitumor therapy.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Lipossomos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Glucose Oxidase/farmacologia , Peroxidase do Rábano Silvestre , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Nanotecnologia , Peróxido de Hidrogênio/uso terapêutico
12.
J Mater Chem B ; 11(23): 5185-5194, 2023 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-37264903

RESUMO

The typical hypoxia of tumor microenvironments seriously affects the efficacy of starvation therapy (ST) and photodynamic therapy (PDT). Therefore, it is of great significance to prepare a multimodal combined therapy nanocomposite with the ability to relieve tumor hypoxia. It is an effective method to release oxygen into the tumor microenvironment using a nanoenzyme. In this work, two-dimensional black phosphorus nanosheets (BPNSs) were used as photothermal reagents and photosensitized agents due to the unique physical properties and also used as nano-carriers for the in situ reduction deposition of Au nanoparticles and fragmented MnO2 coatings. Finally, polyethylene glycol was introduced to obtain the nanocomposite BP@Au@MnO2-PEG (i.e., AMGP). In the tumor microenvironment, MnO2 can catalyze endogenous H2O2 to produce oxygen, improving the effect of ST and PDT. H2O2 generated via the catalytic oxidation of glucose by Au nanoparticles can continue to be used as the substrate catalyzed by MnO2. Meanwhile, black phosphorus nanosheets have excellent photothermal properties for photothermal treatment. The results of in vitro and in vivo experiments indicated that AMGP nanocomposites have good combined antitumor efficacies and biosafety.


Assuntos
Nanopartículas Metálicas , Neoplasias , Fotoquimioterapia , Humanos , Fotoquimioterapia/métodos , Fósforo/uso terapêutico , Compostos de Manganês/química , Peróxido de Hidrogênio/uso terapêutico , Ouro/farmacologia , Ouro/uso terapêutico , Nanopartículas Metálicas/química , Óxidos/química , Neoplasias/tratamento farmacológico , Oxigênio/química , Microambiente Tumoral
13.
Nanomedicine (Lond) ; 18(8): 679-694, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37264976

RESUMO

Background: Reactive oxygen species (ROS) are powerful weapons for various anticancer therapies. However, high glutathione (GSH) levels in cancer cells can significantly reduce the efficacy of such therapies. Methods: In this study, pH-responsive fluorescein-encapsulated zeolitic imidazolate framework-8 nanoparticles were synthesized for ROS-mediated combination therapy. Results: Upon blue light activation, fluorescein displayed a high singlet oxygen photogeneration ability for photodynamic therapy. Concurrently, accumulated Zn2+ from degraded zeolitic imidazolate framework-8 stimulated simultaneous ROS generation and GSH depletion, thereby successfully inducing chemodynamic therapy. This triggered a cascade of photo-physical and chemical processes culminating in the localized generation of ROS, ultimately breaking the intracellular redox equilibrium. Conclusion: This nanoformulation can potentially be used for light-activated ROS-mediated therapy for the management of superficial tumors.


Highly reactive molecules called reactive oxygen species (ROS) are known to be present in excess in cancer cells. As a result, cancer cells are more susceptible to death by any further rise in levels of these species. In the current study, fluorescein-encapsulated zeolitic imidazolate nanoparticles were prepared for blue light-activated ROS-enhancing combination therapy. The nanoparticles displayed significant toxicity against a breast cancer cell line and simultaneously induced glutathione depletion, an antioxidant known to reduce the efficacy of various cancer therapies. Thus, this study reveals the potential of fluorescein-encapsulated zeolitic imidazolate nanoparticles for light-activated ROS-mediated therapy for the treatment of superficial tumors.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Espécies Reativas de Oxigênio/metabolismo , Neoplasias/tratamento farmacológico , Glutationa/metabolismo , Fluoresceínas/uso terapêutico , Linhagem Celular Tumoral , Peróxido de Hidrogênio/uso terapêutico , Microambiente Tumoral
14.
Nanoscale ; 15(20): 8948-8971, 2023 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-37129051

RESUMO

Chemodynamic therapy (CDT), a novel therapeutic approach based on Fenton (Fenton-like) reaction, has been widely employed for tumor therapy. This approach utilizes Fe, Cu, or other metal ions (Mn, Zn, Co, or Mo) to react with the excess hydrogen peroxide (H2O2) in tumor microenvironments (TME), and form highly cytotoxic hydroxyl radical (˙OH) to kill cancer cells. Recently, nanoscale metal-organic frameworks (nMOFs) have attracted considerable attention as promising CDT agents with the rapid development of cancer CDT. This review focuses on summarizing the latest advances (2020-2022) on the design of nMOFs as nanomedicine for CDT or combination therapy of CDT and other therapies. The future development and challenges of CDT are also proposed based on recent progress. Our group hopes that this review will enlighten the research and development of nMOFs for CDT.


Assuntos
Antineoplásicos , Estruturas Metalorgânicas , Neoplasias , Humanos , Estruturas Metalorgânicas/farmacologia , Estruturas Metalorgânicas/uso terapêutico , Linhagem Celular Tumoral , Peróxido de Hidrogênio/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Metais , Neoplasias/tratamento farmacológico , Microambiente Tumoral
15.
J Mater Chem B ; 11(21): 4808-4818, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37212531

RESUMO

It remains an extreme challenge to develop multifunctional drug delivery systems with tumor specificity and a tumor microenvironment (TME) remodeling ability for achieving improved chemotherapy against malignant tumors. Herein, we report the design of diselenide-crosslinked poly(N-vinylcaprolactam) (PVCL) nanogels (NGs) co-loaded with gold (Au) nanoparticles (NPs) and methotrexate (MTX) as a multifunctional nanoplatform (for short, MTX/Au@PVCL NGs) for improved chemotherapy and computed tomography (CT) imaging of tumors. The designed MTX/Au@PVCL NGs show excellent colloidal stability under physiological conditions, while dissociating rapidly to release the incorporated Au NPs and MTX in the H2O2-abundant and slightly acidic TME. The responsive release of Au NPs and MTX effectively induces the apoptosis of cancer cells and prevents DNA replication, together contributing to the repolarization of macrophages from protumor M2-like to antitumor M1-like phenotype in vitro. The MTX/Au@PVCL NGs also enable the remodeling of tumor-associated macrophages to the M1-like phenotype in vivo in a subcutaneous mouse melanoma model, which increases the recruitment of effector T lymphocytes and reduces the content of immunosuppressive regulatory T cells to achieve synergistically enhanced antitumor efficacy when combined with MTX-mediated chemotherapy. Moreover, the MTX/Au@PVCL NGs can be used for Au-mediated CT imaging of tumors. The thus developed NG platform shows great promise as an updated nanomedicine formulation for immune modulation-enhanced tumor chemotherapy under the guidance of CT imaging.


Assuntos
Nanopartículas Metálicas , Neoplasias , Camundongos , Animais , Metotrexato , Nanogéis/uso terapêutico , Ouro/uso terapêutico , Peróxido de Hidrogênio/uso terapêutico , Neoplasias/tratamento farmacológico , Tomografia Computadorizada por Raios X/métodos , Microambiente Tumoral
16.
Biomaterials ; 297: 122109, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37058901

RESUMO

Covalent organic frameworks (COFs) have emerged as a promising platform for nanomedicine, while developing multifunctional COF nanoplatforms remains challenging due to the lack of efficient strategies for COF modification. Herein, we propose a nanozyme bridging (NZB) strategy for COF functionalization. Platinum nanoparticles (Pt NPs) as catalase mimics were in situ grown on the surface of COF NPs without reducing their drug loading capacity (CP), and thiol-terminated aptamer was further densely decorated onto CP NPs via a stable Pt-S bond (CPA). Pt nanozyme engineering and aptamer functionalization rendered the nanoplatform with excellent photothermal conversion, tumor targeting, and catalase-like catalytic performances. Using clinical-approved photosensitizer indocyanine green (ICG) as a model drug, we fabricated a nanosystem (ICPA) for tumor-targeted self-strengthening therapy. ICPA can effectively accumulate into tumor tissue and relieve the hypoxia microenvironment by decomposing the overexpressed H2O2 and generating O2. Under monowavelength NIR light irradiation, the catalase-like catalytic and singlet oxygen generation activities of ICPA can be significantly strengthened, leading to admirable photocatalytic treatment effects against malignant cells as well as tumor-bearing mice in a self-strengthening manner.


Assuntos
Nanopartículas Metálicas , Estruturas Metalorgânicas , Nanopartículas , Neoplasias , Fotoquimioterapia , Animais , Camundongos , Estruturas Metalorgânicas/química , Catalase/uso terapêutico , Nanopartículas Metálicas/química , Peróxido de Hidrogênio/uso terapêutico , Platina/química , Neoplasias/tratamento farmacológico , Nanopartículas/química , Linhagem Celular Tumoral , Microambiente Tumoral
17.
Angew Chem Int Ed Engl ; 62(23): e202302525, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-36930411

RESUMO

Carbon monoxide (CO) is an endogenous signaling molecule with broad therapeutic effects. Here, a multifunctional X-ray-triggered carbon monoxide (CO) and manganese dioxide (MnO2 ) generation nanoplatform based on metal carbonyl and scintillating nanoparticles (SCNPs) is reported. Attributed to the radioluminescent characteristic of SCNPs, UV-responsive Mn2 (CO)10 is not only indirectly activated to release CO by X-ray but can also be degraded into MnO2 . A high dose of CO can be used as a glycolytic inhibitor for tumor suppression; it will also sensitize tumor cells to radiotherapy. Meanwhile MnO2 , as the photolytic byproduct of Mn2 (CO)10 , has both glutathione (GSH) depletion and Fenton-like Mn2+ delivery properties to produce highly toxic hydroxyl radical (⋅OH) in tumors. Thus, this strategy can realize X-ray-activated CO release, GSH depletion, and ⋅OH generation for cascade cancer radiosensitization. Furthermore, X-ray-activated Mn2+ in vivo demonstrates an MRI contrast effect, making it a potential theranostic nanoplatform.


Assuntos
Nanopartículas , Neoplasias , Humanos , Compostos de Manganês/farmacologia , Compostos de Manganês/uso terapêutico , Óxidos/farmacologia , Monóxido de Carbono/farmacologia , Monóxido de Carbono/uso terapêutico , Raios X , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Linhagem Celular Tumoral , Glutationa/metabolismo , Peróxido de Hidrogênio/uso terapêutico
18.
J Mater Chem B ; 11(11): 2455-2465, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36810638

RESUMO

Nanomedicines have been widely used for cancer therapy, while controlling their activity for effective and safe treatment remains a big challenge. Herein, we report the development of a second near-infrared (NIR-II) photoactivatable enzyme-loaded nanomedicine for enhanced cancer therapy. Such a hybrid nanomedicine contains a thermoresponsive liposome shell loaded with copper sulfide nanoparticles (CuS NPs) and glucose oxidase (GOx). The CuS nanoparticles mediate the generation of local heat under 1064 nm laser irradiation, which not only can be used for NIR-II photothermal therapy (PTT), but also leads to the destruction of the thermal-responsive liposome shell to achieve the on-demand release of CuS nanoparticles and GOx. In a tumor microenvironment, GOx oxidizes glucose to produce hydrogen peroxide (H2O2) that acts as a medium to promote the efficacy of chemodynamic therapy (CDT) by CuS nanoparticles. This hybrid nanomedicine enables the synergetic action of NIR-II PTT and CDT to obviously improve efficacy without remarkable side effects via NIR-II photoactivatable release of therapeutic agents. Such a hybrid nanomedicine-mediated treatment can achieve complete ablation of tumors in mouse models. This study provides a promising nanomedicine with photoactivatable activity for effective and safe cancer therapy.


Assuntos
Neoplasias , Terapia Fototérmica , Animais , Camundongos , Nanomedicina , Lipossomos/uso terapêutico , Peróxido de Hidrogênio/uso terapêutico , Neoplasias/tratamento farmacológico , Microambiente Tumoral
19.
Theranostics ; 13(1): 267-277, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36593965

RESUMO

Rationale: Fluorescently traceable prodrugs, which can monitor their biodistribution in vivo and track the kinetics of drug delivery in living cells, are promising for constructing theranostic medicines. However, due to their charge and hydrophobicity, most of the fluorescently traceable prodrugs exhibit high protein binding and non-specific tissue retention affecting in vivo distribution and toxicity, with high background signals. Methods: Herein, the zwitterionic rhodamine (RhB) and camptothecin (CPT) were bridged with a disulfide bond to construct a tumorous heterogeneity-activatable prodrug (RhB-SS-CPT). The interaction of zwitterionic RhB-SS-CPT with proteins was detected by UV and fluorescence spectroscopy, and further demonstrated by molecular docking studies. Then, intracellular tracking and cytotoxicity of RhB-SS-CPT were determined in tumor and normal cells. Finally, the in vivo biodistribution, pharmacokinetics, and anticancer efficacy of RhB-SS-CPT were evaluated in a mouse animal model. Results: The tumorous heterogeneity-activatable RhB-SS-CPT prodrug can self-assemble into stable nanoparticles in water based on its amphiphilic structure. Particularly, the zwitterionic prodrug nanoparticles reduce the non-specific binding to generate a low background signal for better identification of cancerous lesions, achieve rapid internalization into cancer cells, selectively release bioactive CPT as a cytotoxic agent in response to high levels of GSH and H2O2, and exhibit high fluorescence that contributes to the visual chemotherapy modality. In addition, the RhB-SS-CPT prodrug nanoparticles show longer circulation time and better antitumor activity than free CPT in vivo. Interestingly, the zwitterionic nature allows RhB-SS-CPT to be excreted through the renal route, with fewer side effects. Conclusions: Zwitterionic features and responsive linkers are important considerations for constructing potent prodrugs, which provide some useful insights to design the next-generation of theranostic prodrugs for cancer.


Assuntos
Nanopartículas , Neoplasias , Pró-Fármacos , Camundongos , Animais , Pró-Fármacos/farmacologia , Pró-Fármacos/química , Peróxido de Hidrogênio/uso terapêutico , Camptotecina/farmacologia , Camptotecina/química , Rodaminas , Distribuição Tecidual , Simulação de Acoplamento Molecular , Medicina de Precisão , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Nanopartículas/química , Linhagem Celular Tumoral
20.
J Dermatolog Treat ; 34(1): 2133532, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36215682

RESUMO

Seborrheic keratoses (SKs) are benign epidermal neoplasms presenting as waxy, brown to black papules and plaques. Patients often seek removal for cosmetic reasons or irritation. The objective of this systematic review is to assess the efficacy and safety of topical treatments for SKs. Studies involving any topical medication indicated for SK removal were retrieved from Embase, Scopus, PubMed, and Cochrane. The final search was conducted on November 9, 2021, and 26 reports met inclusion criteria. A quality rating scheme was utilized to assess evidence quality. Heterogeneity of treatments and outcome measures precluded meta-analysis. Topical treatments that yielded a good-to-excellent response include hydrogen peroxide, Maxacalcitol 25 µg/g, BID Tazarotene 0.1% cream, 5% potassium dobesilate cream, 1% diclofenac sodium solution, urea-based solution, and 65% and 80% trichloroacetic acid. Local skin reactions were often mild and transient. Topical hydrogen peroxide showed the greatest evidence for clinical clearance of SKs, although there are no studies to our knowledge that directly compared hydrogen peroxide to current first-line treatments (e.g. cryotherapy or shave excision). The results of this review suggest viable and safe treatment of SK with topical therapies; however, there remains demand for topical treatments that reliably equate or exceed the efficacy of current first-line therapies.Key PointsQuestion: Are safe and efficacious topical treatments for seborrheic keratoses available?Findings: Topical treatments for seborrheic keratoses yield different responses and may be associated with local skin reactions. Topical hydrogen peroxide shows the greatest evidence for clinical clearance of seborrheic keratoses and may be a viable option for patients requesting noninvasive removal. No studies to our knowledge directly compare hydrogen peroxide to current first-line treatments.Meaning: There remains demand for topical treatments that reliably equate or exceed the efficacy of current first-line therapies.


Assuntos
Ceratose Seborreica , Humanos , Administração Tópica , Crioterapia/métodos , Peróxido de Hidrogênio/uso terapêutico , Ceratose Seborreica/tratamento farmacológico , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA