Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nano Lett ; 24(19): 5690-5698, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38700237

RESUMEN

Long-term tumor starvation may be a potential strategy to elevate the antitumor immune response by depriving nutrients. However, combining long-term starvation therapy with immunotherapy often yields limited efficacy due to the blockage of immune cell migration pathways. Herein, an intelligent blood flow regulator (BFR) is first established through photoactivated in situ formation of the extravascular dynamic hydrogel to compress blood vessels, which can induce long-term tumor starvation to elicit metabolic stress in tumor cells without affecting immune cell migration pathways. By leveraging methacrylate-modified nanophotosensitizers (HMMAN) and biodegradable gelatin methacrylate (GelMA), the developed extravascular hydrogel dynamically regulates blood flow via enzymatic degradation. Additionally, aPD-L1 loaded into HMMAN continuously blocks immune checkpoints. Systematic in vivo experiments demonstrate that the combination of immune checkpoint blockade (ICB) and BFR-induced metabolic stress (BIMS) significantly delays the progression of Lewis lung and breast cancers by reshaping the tumor immunogenic landscape and enhancing antitumor immune responses.


Asunto(s)
Hidrogeles , Hidrogeles/química , Animales , Ratones , Humanos , Línea Celular Tumoral , Femenino , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Inmunoterapia , Gelatina/química , Metacrilatos/química , Metacrilatos/farmacología , Neoplasias de la Mama/inmunología
2.
Biochem Biophys Res Commun ; 679: 82-89, 2023 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-37677981

RESUMEN

BACKGROUND AND PURPOSE: Targeting and uptake are the most important strategies for enhancing the efficacy of cancer photothermal therapy (PTT) and reducing damage to surrounding normal tissues. In this study, a kind of nanophotosensitizer based on nanobubbles and TiN was prepared for synergetic therapy for hepatocellular carcinoma. METHODS: The photothermal agent titanium nitride (TiN) was wrapped in nanobubbles by membrane hydration method and verified in cells and animals. CCK-8, cell death staining, and JC-1 were used to verify the pernicious effect of photothermal combined with Ultrasound Targeted Nanobubble Destruction (UTND) and then injected into animals through the tail vein to observe its photothermal effect and in vivo inhibitory effect. A hemolysis test and body weight change verified its safety. RESULTS: The average diameter of the novel nanophotosensitizer was 300.3 ± 12.7 nm, with a consistent nanospheres morphology. The UTND technology was utilized to improve the penetration of TiN into tumor cells through the physical energy of ultrasound irradiation. The therapeutic effects of the synergistic therapy of UTND and PTT were verified in vitro and in vivo. CONCLUSION: The research has established NBs@C3F8-TiN as a suitable ultrasound photothermal agent due to its appropriate size and efficient photothermal efficacy for visual photothermal therapy for HCC.

3.
Small ; 16(14): e2000011, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32174021

RESUMEN

Singlet oxygen (1 O2 ) generation has been observed from ultrasmall luminescent gold nanoparticles (AuNPs), but regulation of 1 O2 generation ability from the nanosized noble metals has remained challenging. Herein, the 1 O2 generation ability of ultrasmall AuNPs (d ≈ 1.8 nm) is reported to be highly correlated to the surface factors including the amount of Au(I) species and surface charge. By taking the advantages of facile in situ PEGylation, it is discovered that a high amount of Au(I) species and surface charge results in strong ability in generation of 1 O2 , whereas a relative low amount of Au(I) species and surface charge leads to weak ability in 1 O2 production. A feasible general strategy is then developed to controllably regulate the 1 O2 generation efficiency of the AuNPs through facile ligand exchange with positively-charged or negatively-charged thiolated ligands. The AuNPs as nanophotosensitizer for 1 O2 generation in the cellular level is also demonstrated to be highly controllable through surface ligand exchange with synergistical effects of 1 O2 generation ability and subcellular distribution to lysosome or mitochondria. The strategy in the bidirectional regulation of 1 O2 generation from ultrasmall AuNPs provides guidance for future design of nanosized metal nanomedicine toward specific disease diagnosis and treatment.

4.
Angew Chem Int Ed Engl ; 56(36): 10717-10720, 2017 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-28667692

RESUMEN

Photodynamic therapy (PDT) is widely used to treat diverse diseases, but its dependence on oxygen to produce cytotoxic reactive oxygen species (ROS) diminishes the therapeutic effect in a hypoxic environment, such as solid tumors. Herein, we developed a ROS-producing hybrid nanoparticle-based photosensitizer capable of maintaining high levels of ROS under both normoxic and hypoxic conditions. Conjugation of a ruthenium complex (N3) to a TiO2 nanoparticle afforded TiO2 -N3. Upon exposure of TiO2 -N3 to light, the N3 injected electrons into TiO2 to produce three- and four-fold more hydroxyl radicals and hydrogen peroxide, respectively, than TiO2 at 160 mmHg. TiO2 -N3 maintained three-fold higher hydroxyl radicals than TiO2 under hypoxic conditions via N3-facilitated electron-hole reduction of adsorbed water molecules. The incorporation of N3 transformed TiO2 from a dual type I and II PDT agent to a predominantly type I photosensitizer, irrespective of the oxygen content.


Asunto(s)
Hipoxia/tratamiento farmacológico , Nanopartículas/química , Fármacos Fotosensibilizantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Rutenio/farmacología , Titanio/farmacología , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Rutenio/química , Titanio/química
5.
Int J Biol Macromol ; 266(Pt 2): 131359, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38580018

RESUMEN

The combination of photothermal therapy (PTT) and photodynamic therapy (PDT) has emerged as a promising strategy for cancer treatment. However, the poor photostability and photothermal conversion efficiency (PCE) of organic small-molecule photosensitizers, and the intracellular glutathione (GSH)-mediated singlet oxygen scavenging largely decline the antitumor efficacy of PTT and PDT. Herein, a versatile nanophotosensitizer (NPS) system is developed by ingenious incorporation of indocyanine green (ICG) into the PEGylated chitosan (PEG-CS)-coated polydopamine (PDA) nanoparticles via multiple π-π stacking, hydrophobic and electrostatic interactions. The PEG-CS-covered NPS showed prominent colloidal and photothermal stability as well as high PCE (ca 62.8 %). Meanwhile, the Michael addition between NPS and GSH can consume GSH, thus reducing the GSH-induced singlet oxygen scavenging. After being internalized by CT26 cells, the NPS under near-infrared laser irradiation produced massive singlet oxygen with the aid of thermo-enhanced intracellular GSH depletion to elicit mitochondrial damage and lipid peroxide formation, thus leading to ferroptosis and apoptosis. Importantly, the combined PTT and PDT delivered by NPS effectively inhibited CT26 tumor growth in vivo by light-activated intense hyperthermia and redox homeostasis disturbance. Overall, this work presents a new tactic of boosting antitumor potency of ICG-mediated phototherapy by PEG-CS-covered NPS.


Asunto(s)
Quitosano , Glutatión , Nanopartículas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Terapia Fototérmica , Polietilenglicoles , Quitosano/química , Fotoquimioterapia/métodos , Animales , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Glutatión/metabolismo , Polietilenglicoles/química , Ratones , Nanopartículas/química , Terapia Fototérmica/métodos , Línea Celular Tumoral , Verde de Indocianina/química , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Oxígeno Singlete/metabolismo , Humanos , Apoptosis/efectos de los fármacos , Indoles/química , Indoles/farmacología , Polímeros/química
6.
ACS Appl Mater Interfaces ; 15(18): 21746-21753, 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37126007

RESUMEN

Effective clinical practice of precise photodynamic therapy (PDT) is severely impeded by the inherent drawbacks and aggregation propensity of conventional photosensitizers. An all-in-one approach is highly desired to optimize structural features, photophysical properties, and pharmacokinetic behaviors of photosensitizers. Herein, we have fabricated mesoporous boron dipyrromethene-bridged coordination polymer nanophotosensitizers (BCP-NPs) for high-performance PDT via a unique solvent-assisted assembly strategy. Distinctive photophysical and structural characteristics of BCP-NPs confer enhanced photodynamic activities, together with high cellular uptake and ultrahigh stability. Moreover, BCP-NPs showed excellent tumor accumulation and prolonged tumor retention, achieving eradication of the triple-negative breast cancer (TNBC) model under low-power-density LED irradiation. This work has provided a valuable paradigm for the construction of mesoporous photoactive nanomaterials for biophotonic applications.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Fármacos Fotosensibilizantes/química , Polímeros/química , Nanopartículas/química , Neoplasias/tratamiento farmacológico
7.
Acta Biomater ; 157: 408-416, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36549634

RESUMEN

Photothermal therapy has become a promising approach as precision medicine to allow spatial control of therapeutic effect only in the site of interest. However, the full potential of PTT has not been realized due to the lack of simple photosensitizers (PSs) that can overcome multistage biological barriers and improve theranostic efficiency. Here, we develop a small molecule-based PS to enhance tumor-specific PTT by programming multistage transport and activation properties in molecular architecture. This PS can self-assemble into stable nanoparticles that accumulate passively in tumor, and then actively internalize through ligand-mediated endocytosis. Subsequently, the programmable degradable linkers are selectively cleaved, enabling size shrinkage for better tumor penetration, binding albumin to enhance the near-infrared fluorescence for low-background imaging, and activating photothermal conversion for tumor suppression. The self-delivery process can be programmed, representing the first multistage small-molecule nano-photosensitizer that overcomes multiple biological barriers and improves the PTT index of tumor. STATEMENT OF SIGNIFICANCE: Photothermal therapy has become a promising approach as precision medicine, but has not been realized due to the lack of simple photosensitizers that can overcome multistage biological barriers and improve theranostic efficiency. In this contribution, we solve this dilemma by developing a small molecule-based photosensitizer by programming multistage transport and activation properties in molecular architecture, which could self-assemble into stable nanoparticles that accumulate passively in tumor, and actively internalized through ligand-mediated endocytosis. Subsequently, the programmable activation by ROS triggered size reduction for tumor penetration and minimized the phototoxicity to normal tissue. The activatable fluorescence and photothermal properties made the photosensitizer intrinsically suitable for multimodal imaging-guided PTT, providing a promising supramolecular nanomedicine towards tumor precise diagnosis and therapy.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Fármacos Fotosensibilizantes/química , Terapia Fototérmica , Línea Celular Tumoral , Ligandos , Nanopartículas/uso terapéutico , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Imagen Multimodal , Nanomedicina Teranóstica/métodos , Fototerapia/métodos
8.
Biomaterials ; 293: 121953, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36521428

RESUMEN

Carbon dots (CDs) have emerged as promising nanomaterials for bioimaging-guided photodynamic therapy (PDT). However, designing red-emissive CDs (RCDs) with tunable type I and type II reactive oxygen species (ROS) generation to simultaneously meet PDT applications in aerobic and hypoxic scenarios still remain major challenges. Herein, three types of RCDs with maximum emission at approximately 680 nm are successfully prepared. It is noteworthy that they exhibit the adjustable ROS production with equal superoxide anion (via type I PDT) and incremental singlet oxygen (via type II PDT). Detailed structural and optical characterizations along with theoretical calculation reveal that the unique type I/II ROS formation mainly depends on the core sizes and surface states of RCDs, which determine their identical redox potentials and tapering energy gaps between singlet- and triplet states, respectively. Additionally, due to the inherent mitochondria targeting capability, RCDs enable themselves to induce cell programmed death via activating mitochondrion-mediated apoptotic pathways. This work exploits the unprecedented RCDs with tunable type I and type II ROS generation that could ensure highly efficient tumor eradication both in vitro and in vivo, even under the harsh tumor microenvironment, providing a new prospect for CDs as nanophotosensitizers to conquer the limitations of single type PDT.


Asunto(s)
Neoplasias , Fotoquimioterapia , Humanos , Fotoquimioterapia/métodos , Especies Reactivas de Oxígeno/metabolismo , Carbono/química , Neoplasias/tratamiento farmacológico , Mitocondrias/metabolismo , Fármacos Fotosensibilizantes/química , Línea Celular Tumoral , Microambiente Tumoral
9.
Biomaterials ; 292: 121944, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36495801

RESUMEN

The microenvironment in solid tumors drives the fate of cancer cells to ferroptosis, yet the underlying mechanism remains incompletely understood. Herein, we report a metal-free polymer photosensitizer (BDPB) as a new type ferroptosis inducer of starved cancer cells. The polymer consists of boron difluoride dipyrromethene dye as the photosensitizing unit and diisopropyl-ethyl amine as the electron-donating unit. Ultrafast spectroscopy and electron spin resonance mechanistically revealed the prolonged charge-separation process in BDPB, enabling complex-I like one-electron transfer effect to produce O2●-. Unexpectedly, the O2●--generating BDPB nanoparticles (NPs) served to deactivate the AMPK-mTOR signaling pathway in normal-state cancer cells to initiate cell repair activity and survive low-dose phototherapy. However, for cancer cells in a starved state, BDPB NPs triggered glutathione peroxidase 4 downregulation, lipid peroxides accumulation, and death to cancer cells, which was identified as ferroptosis but not apoptosis, necroptosis, or autosis. The application of BDPB NPs sheds new light on the design of individualized ferroptosis inducers for combating cancer progression.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Fármacos Fotosensibilizantes/farmacología , Polímeros/farmacología , Neoplasias/tratamiento farmacológico , Apoptosis , Microambiente Tumoral
10.
ACS Appl Mater Interfaces ; 14(10): 12050-12058, 2022 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-35234031

RESUMEN

Mitochondria-targeting photodynamic therapy (PDT) can block mitochondrial function and trigger the inherent proapoptotic cascade signal of mitochondria, which has been considered to have the potential to amplify the efficiency of PDT. However, the dynamic change of mitochondrial membrane potential (MMP) makes most cationic photosensitizers easily fall off from the mitochondria, which greatly limits the efficiency of PDT. Here, we have developed a smart liposome encapsulation method based on a mitochondria-stapling photosensitizer for efficient theranostic photodynamic therapy. The stapling photosensitizer can be covalently bound inside mitochondria via two reaction sites without a falloff effect, regardless of the change of MMP. As a result, the liposome-based nanophotosensitizer showed a high efficiency of PDT (IC50 = 0.98 µM) under 630 nm light. At the same time, the nanophotosensitizer had fluorescence imaging-guided ability to monitor abnormal mitochondrial morphology during PDT. Importantly, the results of mice experiments also showed that the liposome-based nanophotosensitizer possessed excellent antitumor PDT activity because the released photosensitizer can stay inside mitochondria during the whole process of PDT.


Asunto(s)
Fotoquimioterapia , Fármacos Fotosensibilizantes , Animales , Línea Celular Tumoral , Liposomas/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Mitocondrias/metabolismo , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/uso terapéutico
11.
Biomaterials ; 291: 121916, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36410110

RESUMEN

Cancer treatment currently still faces crucial challenges in therapeutic effectiveness, precision, and complexity. Photodynamic therapy (PDT) as a non-invasive tactic has earned widespread popularity for its excellent therapeutic output, flexibility, and restrained toxicity. Nonetheless, drawbacks, including low efficiency, poor cancer specificity, and limited therapeutic depth, remain considerable during the cancer treatment. Although great effort has been made to improve the performance, the overall efficiency and biosafety are still ambiguous and unable to meet urgent clinical needs. Herein, this study integrates merits from previous PDT strategies and develops a cancer-targeting, activatable, biosafe photosensitizer. Owing to excellent self-assembly ability, this photosensitizer can be conveniently prepared as multifunctional nano-photosensitizers, namely MBNPs, and applied to in vivo cancer phototheranostics in "all-in-one" mode. This study successfully verifies the mechanism of MBNPs, then deploys them to cell-based and in vivo cancer PDT. Based on the unique cancer microenvironment, MBNPs achieve precise distribution, accumulation, and activation towards the tumor, releasing methylene blue as a potent photosensitizer for phototherapy. The PDT outcome demonstrates MBNPs' superior cancer specificity, remarkable PDT efficacy, and negligible toxicity. Meanwhile, in vivo NIR fluorescence and photoacoustic imaging have been utilized to guide the PDT treatment synergistically. Additionally, the biosafety of the MBNPs-based PDT treatment is ensured, thus providing potential for future clinical studies.


Asunto(s)
Neoplasias , Fotoquimioterapia , Humanos , Fármacos Fotosensibilizantes/uso terapéutico , Contención de Riesgos Biológicos , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
12.
Nanomicro Lett ; 14(1): 123, 2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35513555

RESUMEN

The great promise of photodynamic therapy (PDT) has thrusted the rapid progress of developing highly effective photosensitizers (PS) in killing cancerous cells and bacteria. To mitigate the intrinsic limitations of the classical molecular photosensitizers, researchers have been looking into designing new generation of nanomaterial-based photosensitizers (nano-photosensitizers) with better photostability and higher singlet oxygen generation (SOG) efficiency, and ways of enhancing the performance of existing photosensitizers. In this paper, we review the recent development of nano-photosensitizers and nanoplasmonic strategies to enhance the SOG efficiency for better PDT performance. Firstly, we explain the mechanism of reactive oxygen species generation by classical photosensitizers, followed by a brief discussion on the commercially available photosensitizers and their limitations in PDT. We then introduce three types of new generation nano-photosensitizers that can effectively produce singlet oxygen molecules under visible light illumination, i.e., aggregation-induced emission nanodots, metal nanoclusters (< 2 nm), and carbon dots. Different design approaches to synthesize these nano-photosensitizers were also discussed. To further enhance the SOG rate of nano-photosensitizers, plasmonic strategies on using different types of metal nanoparticles in both colloidal and planar metal-PS systems are reviewed. The key parameters that determine the metal-enhanced SOG (ME-SOG) efficiency and their underlined enhancement mechanism are discussed. Lastly, we highlight the future prospects of these nanoengineering strategies, and discuss how the future development in nanobiotechnology and theoretical simulation could accelerate the design of new photosensitizers and ME-SOG systems for highly effective image-guided photodynamic therapy.

13.
Photodiagnosis Photodyn Ther ; 36: 102597, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34699982

RESUMEN

Photodynamic therapy (PDT) utilizes photosensitizers (PSs) together with irradiation light of specific wavelength interacting with oxygen to generate cytotoxic reactive oxygen species (ROS), which could trigger apoptosis and/or necrosis-induced cell death in target tissues. During the past two decades, multifunctional nano-PSs employing nanotechnology and nanomedicine developed, which present not only photosensitizing properties but additionally accurate drug release abilities, efficient response to optical stimuli and hypoxia resistance. Further, nano-PSs have been developed to enhance PDT efficacy by improving the ROS yield. In addition, nano-PSs with additive or synergistic therapies are significant for both currently preclinical study and future clinical practice, given their capability of considerable higher therapeutic efficacy under safer systemic drug dosage. In this review, nano-PSs that allow precise drug delivery for efficient absorption by target cells are introduced. Nano-PSs boosting sensitivity and conversion efficiency to PDT-activating stimuli are highlighted. Nano-PSs developed to address the challenging hypoxia conditions during PDT of deep-sited tumors are summarized. Specifically, PSs capable of synergistic therapy and the emerging novel types with higher ROS yield that further enhance PDT efficacy are presented. Finally, future demands for ideal nano-PSs, emphasizing clinical translation and application are discussed.


Asunto(s)
Antineoplásicos , Neoplasias , Fotoquimioterapia , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/uso terapéutico , Especies Reactivas de Oxígeno
14.
Materials (Basel) ; 15(1)2021 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-35009284

RESUMEN

The aim of this study is to fabricate nanophotosensitizers composed of methoxy poly(ethylene glycol) (mPEG), chlorin e6 (Ce6), and phenylboronic acid pinacol ester (PBAP) with diselenide linkages for reactive oxygen species (ROS)-sensitive photodynamic therapy (PDT) of cervical cancer cells. To fabricate nanophotosensitizers, Ce6 was conjugated with mPEG via selenocystamine linkage and then remaining carboxylic acid groups of Ce6 was attached to PBAP (mPEGseseCe6PBAP conjugates). Nanophotosensitizers of mPEGseseCe6PBAP conjugates were prepared by dialysis method. In transmission electron microscope (TEM) observation, nanophotosensitizers of mPEGseseCe6PBAP conjugates have spherical shapes and their diameters were less than 150 nm. The average diameter of mPEGseseCe6PBAP nanophotosensitizers was 92.7 ± 9.6 nm in particle size analysis. When H2O2 was added to the nanophotosensitizer solution, nanophotosensitizers were sensitively disintegrated according to the H2O2 concentration and then changed from monomodal distribution to multimodal distribution in particle size distribution. Furthermore, Ce6 release from nanophotosensitizers also increased according to the H2O2 concentration. When H2O2 was added to cell culture of HeLa human cervical cancer cells, intracellular Ce6 uptake of nanophotosensitizers were gradually increased according to the H2O2 concentration, indicating that nanophotosensitizers showed ROS-sensitive delivery of Ce6 against cancer cells.As well as free Ce6, nanophotosensitizers in the absence of light irradiation have low intrinsic cytotoxicity against RAW264.7 cells and HeLa cells. However, nanophotosensitizers induced cell death dose-dependently under light irradiation. Especially, nanophotosensitizers showed significantly higher ROS generation and phototoxicity against HeLa cells in vitro. When nanophotosensitizers were intravenously administered to animal tumor xenograft model of HeLa cells, tumor tissues revealed stronger fluorescence intensity than other tissues by light irradiation while absence of light irradiation induced relatively lower fluorescence intensity in tumor tissues, indicating that nanophotosensitizers have sensitivity against oxidative stress in tumor tissues. We suggest that nanophotosensitizers of mPEGseseCe6PBAP conjugates are promising vehicle for PDT of cervical cancer cells.

15.
Cells ; 10(9)2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34571839

RESUMEN

The aim of this study was to fabricate a reactive oxygen species (ROS)-sensitive and folate-receptor-targeted nanophotosensitizer for the efficient photodynamic therapy (PDT) of cervical carcinoma cells. Chlorin e6 (Ce6) as a model photosensitizer was conjugated with succinyl ß-cyclodextrin via selenocystamine linkages. Folic acid (FA)-poly(ethylene glycol) (PEG) (FA-PEG) conjugates were attached to these conjugates and then FA-PEG-succinyl ß-cyclodextrin-selenocystamine-Ce6 (FAPEGbCDseseCe6) conjugates were synthesized. Nanophotosensitizers of FaPEGbCDseseCe6 conjugates were fabricated using dialysis membrane. Nanophotosensitizers showed spherical shapes with small particle sizes. They were disintegrated in the presence of hydrogen peroxide (H2O2) and particle size distribution changed from monomodal distribution pattern to multimodal pattern. The fluorescence intensity and Ce6 release rate also increased due to the increase in H2O2 concentration, indicating that the nanophotosensitizers displayed ROS sensitivity. The Ce6 uptake ratio, ROS generation and cell cytotoxicity of the nanophotosensitizers were significantly higher than those of the Ce6 itself against HeLa cells in vitro. Furthermore, the nanophotosensitizers showed folate-receptor-specific delivery capacity and phototoxicity. The intracellular delivery of nanophotosensitizers was inhibited by folate receptor blocking, indicating that they have folate-receptor specificity in vitro and in vivo. Nanophotosensitizers showed higher efficiency in inhibition of tumor growth of HeLa cells in vivo compared to Ce6 alone. These results show that nanophotosensitizers of FaPEGbCDseseCe6 conjugates are promising candidates as PDT of cervical cancer.


Asunto(s)
Receptores de Folato Anclados a GPI/metabolismo , Nanopartículas/administración & dosificación , Oxidación-Reducción/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Porfirinas/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , beta-Ciclodextrinas/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Clorofilidas , Femenino , Ácido Fólico/metabolismo , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Tamaño de la Partícula , Fotoquimioterapia/métodos , Neoplasias del Cuello Uterino/metabolismo
16.
J Inorg Biochem ; 217: 111371, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33588279

RESUMEN

To develop highly efficient photosensitizers for photodynamic therapy, herein a zinc(II) phthalocyanine-folate conjugate (PcN-FA) used to construct an activatable nanophotosensitizer (NanoPcN-FA) through a facile self-assembly. The self-assembled nanophotosensitizer (NanoPcN) without folate-modification was used as a negative control. After self-assembly, the photoactivities of NanoPcN-FA was quenched. The in vitro studies showed that NanoPcN-FA could be taken in by folate-receptor (FR)-positive SKOV3 cells and activated in the cells. It also exhibited slightly higher photocytotoxicity against SKOV3 cells than NanoPcN. Moreover, the competitive assay confirmed that the cellular uptake of NanoPcN-FA was through a FR-mediated process. Finally, the in vivo results indicated that NanoPcN-FA could target tumor tissue of S180 rat ascitic tumor-bearing mice due to the folic acid (FA) ligand, leading to a highly efficient antitumor photodynamic efficacy with the tumor inhibition rate of 95%.


Asunto(s)
Antineoplásicos/uso terapéutico , Indoles/uso terapéutico , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/efectos de la radiación , Línea Celular Tumoral , Humanos , Indoles/síntesis química , Indoles/efectos de la radiación , Luz , Ratones , Fotoquimioterapia , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/efectos de la radiación , Ratas
17.
Adv Sci (Weinh) ; 8(3): 2002253, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33552856

RESUMEN

Active-targeted nanoparticles are attractive carriers due to their potentials to facilitate specific delivery of drugs into tumor cells while sparing normal cells. However, the therapeutic outcomes of active-targeted nanomedicines are hampered by the multiple physiological barriers in the tumor microenvironment (TME). Herein, an epidermal growth factor receptor-targeted ultra-pH-sensitive nanophotosensitizer is fabricated, and the regulation of the TME to augment the active targeting ability and therapeutic efficacy is pinpointed. The results reveal that tumor vasculature normalization with thalidomide indiscriminately enhance the tumor accumulation of passive and active targeted nanoparticles, both of which are sequestered in the stromal bed of tumor mass. Whereas, photoablation of stromal cells located in perivascular regions significantly improves the accessibility of antibody-modified nanophotosensitizer to receptor-overexpressed cancer cells. After sequential regulation of TME, the antitumor efficacy of antibody-modified nanophotosensitizer is drastically enhanced through synergistic enhancements of tumor accumulation and cancer cell accessibility of active-targeted nanoparticles. The study offers deep insights about the intratumoral barriers that hinder the active-targeted nanoparticles delivery, and provides a basis for developing more effective strategies to accelerate the clinical translation of active-targeted nanomedicines.

18.
Cell Rep ; 33(11): 108499, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33326787

RESUMEN

By incorporating an artificial reactive oxygen species (ROS) generation mechanism, a biotic/abiotic integration is designed to improve the anti-tumor effect of neutrophils by artificially potentiating their ROS effector mechanism in a remotely controlled route. Specifically, the photosensitizer Ce6 is nano-packaged by the albumin BSA to achieve biocompatible and efficient integration with neutrophils (NEs). Reinfusion of the engineered NEs into 4T1 tumor-bearing mice led to more Ce6 accumulation in tumors relative to Ce6 nanoformulation. At the peak of accumulation, tumor illumination activates the embedded Ce6 for ROS generation and NETosis formation. Because of the ROS-intensified cytolytic effect, the growth of 4T1 tumors is inhibited significantly. The photo-controlled process largely avoids the off-target effects observed frequently in current cell therapies. The strategy directly generates ROS effector molecules with spatiotemporal precision. This engineering approach is able to potentiate the native capacity of immune cells independent of the tumor microenvironment.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Neutrófilos/metabolismo , Animales , Humanos , Ratones
19.
ACS Nano ; 13(6): 6702-6710, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31184131

RESUMEN

A major challenge in cancer treatment is the development of effective tumor-specific therapeutic methods that have minimal side effects. Recently, a photodynamic therapy (PDT) technique using activatable photosensitizers (aPSs) has shown great potential for cancer-specific treatment. Here, we develop a sequential protein-responsive aPS (PcC4-MSN-O1) that is based on zinc(II) phthalocyanine derivative (PcC4)-entrapped mesoporous silica nanoparticles (MSNs) and a wrapping DNA (O1) as a biogate. Inside the nanostructure of PcC4-MSN-O1, PcC4 shows self-quenching photoactivity. However, when PcC4-MSN-O1 sequentially reacts with telomerase and albumin, its photoactivity is dramatically turned on. Therefore, PcC4-MSN-O1 displays selective phototoxicity against cancer cells ( e.g., HeLa) over normal cells ( e.g., HEK-293). Following systemic PcC4-MSN-O1 administration, there is an obvious accumulation in HeLa tumors of xenograft-bearing mice, and laser irradiation clearly induces the inhibition of tumor growth. Moreover, the time-modulated activation process in tumors and the relatively fast excretion of PcC4-MSN-O1 indicate its advantages in reducing potential side effects.


Asunto(s)
Indoles/química , Nanopartículas/química , Compuestos Organometálicos/química , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Albúminas/metabolismo , Animales , ADN/química , Femenino , Células HEK293 , Células HeLa , Humanos , Isoindoles , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Nanopartículas/uso terapéutico , Neoplasias Experimentales/tratamiento farmacológico , Dióxido de Silicio/química , Telomerasa/metabolismo , Compuestos de Zinc
20.
ACS Appl Mater Interfaces ; 11(46): 42873-42884, 2019 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-31553151

RESUMEN

Ferroptosis is an iron-dependent cell death caused by accumulation of lipid peroxidation (LPO), which is a new strategy for cancer treatment. Th current ferroptosis therapy nanodevices have low efficiency and side effects generally. Hence, we developed a Black Hole Quencher (BHQ)-based fluorescence "off-on" nanophotosensitizer complex assembly (CSO-BHQ-IR780-Hex/MIONPs/Sor). CSO-connected BHQ-IR780-Hex and -loaded magnetic iron oxide nanoparticles (MIONPs) and sorafenib (Sor) formed a very concise functionalized delivery system. CSO-BHQ-IR780-Hex disassembled by GSH attack and released IR780-Hex, MIONPs, and sorafenib. IR780-Hex anchored to the mitochondrial membrane, which would contribute to amplifying the efficiency of the photosensitizer. When NIR irradiation was given to CSO-BHQ-IR780-Hex/MIONPs/Sor-treated cells, iron supply increased, the xCT/GSH/GPX-4 system was triggered, and a lot of LPO burst. A malondialdehyde test showed that LPO in complex assembly-treated cells was explosive and increased about 18-fold compared to the control. The accumulation process of particles was monitored by an IR780-Hex photosensitizer, which showed an excellent tumor target ability by magnetic of nanodevice in vivo. Interestingly, the half-life of sorafenib in a nanodevice was increased about 26-fold compared to the control group. Importantly, the complex assembly effectively inhibits tumor growth in the breast tumor mouse model. This work would provide ideas in designing nanomedicines for the ferroptosis treatment of cancer.


Asunto(s)
Alcanosulfonatos , Compuestos Azo , Neoplasias de la Mama , Ferroptosis/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Nanopartículas de Magnetita , Sorafenib , Alcanosulfonatos/química , Alcanosulfonatos/farmacología , Animales , Compuestos Azo/química , Compuestos Azo/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Humanos , Células MCF-7 , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratas , Ratas Sprague-Dawley , Sorafenib/química , Sorafenib/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA