Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891819

ABSTRACT

Photothermal therapy (PTT) is a promising cancer therapy modality with significant advantages such as precise targeting, convenient drug delivery, better efficacy, and minimal adverse effects. Photothermal therapy effectively absorbs the photothermal transducers in the near-infrared region (NIR), which induces the photothermal effect to work. Although PTT has a better role in tumor therapy, it also suffers from low photothermal conversion efficiency, biosafety, and incomplete tumor elimination. Therefore, the use of nanomaterials themselves as photosensitizers, the targeted modification of nanomaterials to improve targeting efficiency, or the combined use of nanomaterials with other therapies can improve the therapeutic effects and reduce side effects. Notably, noble metal nanomaterials have attracted much attention in PTT because they have strong surface plasmon resonance and an effective absorbance light at specific near-infrared wavelengths. Therefore, they can be used as excellent photosensitizers to mediate photothermal conversion and improve its efficiency. This paper provides a comprehensive review of the key role played by noble metal nanomaterials in tumor photothermal therapy. It also describes the major challenges encountered during the implementation of photothermal therapy.


Subject(s)
Metal Nanoparticles , Neoplasms , Photothermal Therapy , Humans , Photothermal Therapy/methods , Neoplasms/therapy , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Animals , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use
2.
Mater Today Bio ; 26: 101094, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38854952

ABSTRACT

Cerebral ischemia-reperfusion injury (CIRI) is a major challenge to neuronal survival in acute ischemic stroke (AIS). However, effective neuroprotective agents remain to be developed for the treatment of CIRI. In this work, we have developed an Anti-TRAIL protein-modified and indocyanine green (ICG)-responsive nanoagent (Anti-TRAIL-ICG) to target ischemic areas and then reduce CIRI and rescue the ischemic penumbra. In vitro and in vivo experiments have demonstrated that the carrier-free nanoagent can enhance drug transport across the blood-brain barrier (BBB) in stroke mice, exhibiting high targeting ability and good biocompatibility. Anti-TRAIL-ICG nanoagent played a better neuroprotective role by reducing apoptosis and ferroptosis, and significantly improved ischemia-reperfusion injury. Moreover, the multimodal imaging platform enables the dynamic in vivo examination of multiple morphofunctional information, so that the dynamic molecular events of nanoagent can be detected continuously and in real time for early treatment in transient middle cerebral artery occlusion (tMCAO) models. Furthermore, it has been found that Anti-TRAIL-ICG has great potential in the functional reconstruction of neurovascular networks through optical coherence tomography angiography (OCTA). Taken together, our work effectively alleviates CIRI after stoke by blocking multiple cell death pathways, which offers an innovative strategy for harnessing the apoptosis and ferroptosis against CIRI.

3.
Pharmaceutics ; 16(5)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38794265

ABSTRACT

Sonodynamic therapy (SDT) has attracted significant attention in recent years as it is an innovative approach to tumor treatment. It involves the utilization of sound waves or ultrasound (US) to activate acoustic sensitizers, enabling targeted drug release for precise tumor treatment. This review aims to provide a comprehensive overview of SDT, encompassing its underlying principles and therapeutic mechanisms, the applications of nanomaterials, and potential synergies with combination therapies. The review begins by introducing the fundamental principle of SDT and delving into the intricate mechanisms through which it facilitates tumor treatment. A detailed analysis is presented, outlining how SDT effectively destroys tumor cells by modulating drug release mechanisms. Subsequently, this review explores the diverse range of nanomaterials utilized in SDT applications and highlights their specific contributions to enhancing treatment outcomes. Furthermore, the potential to combine SDT with other therapeutic modalities such as photothermal therapy (PTT) and chemotherapy is discussed. These combined approaches aim to synergistically improve therapeutic efficacy while mitigating side effects. In conclusion, SDT emerges as a promising frontier in tumor treatment that offers personalized and effective treatment options with the potential to revolutionize patient care. As research progresses, SDT is poised to play a pivotal role in shaping the future landscape of oncology by providing patients with a broader spectrum of efficacious and tailored treatment options.

4.
ChemSusChem ; : e202400339, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38440923

ABSTRACT

The pursuit of efficient host materials to address the sluggish redox kinetics of sulfur species has been a longstanding challenge in advancing the practical application of lithium-sulfur batteries. In this study, amorphous carbon layer loaded with ultrafine CoP nanoparticles prepared by a one-step in situ carbonization/phosphating method to enhance the inhibition of 2D black phosphorus (BP) on LiPSs shuttle. The carbon coating layer facilitates accelerated electron/ion transport, enabling the active involvement of BP in the conversion of soluble lithium polysulfides (LiPSs). Concurrently, the ultra-fine CoP nanoparticles enhance the chemical anchoring ability and introduce additional catalytic sites. As a result, S@BP@C-CoP electrodes demonstrate exemplary cycling stability (with a minimal capacity decay of 0.054 % over 500 cycles at 1 C) and superior rate performance (607.1 mAh g-1 at 5 C). Moreover, at a sulfur loading of 5.5 mg cm-2, the electrode maintains an impressive reversible areal capacity of 5.45 mAh cm-2 after 50 cycles at 0.1 C. This research establishes a promising approach, leveraging black phosphorus-based materials, for developing high-efficiency Li-S batteries.

5.
Pharmaceutics ; 16(1)2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38258099

ABSTRACT

Clinical drug administration aims to deliver drugs efficiently and safely to target tissues, organs, and cells, with the objective of enabling their therapeutic effects. Currently, the main approach to enhance a drug's effectiveness is ensuring its efficient delivery to the intended site. Due to the fact that there are still various drawbacks of traditional drug delivery methods, such as high toxicity and side effects, insufficient drug specificity, poor targeting, and poor pharmacokinetic performance, nanocarriers have emerged as a promising alternative. Nanocarriers possess significant advantages in drug delivery due to their size tunability and surface modifiability. Moreover, nano-drug delivery systems have demonstrated strong potential in terms of prolonging drug circulation time, improving bioavailability, increasing drug retention at the tumor site, decreasing drug resistance, as well as reducing the undesirable side effects of anticancer drugs. Numerous studies have focused on utilizing polysaccharides as nanodelivery carriers, developing delivery systems based on polysaccharides, or exploiting polysaccharides as tumor-targeting ligands to enhance the precision of nanoparticle delivery. These types of investigations have become commonplace in the academic literature. This review aims to elucidate the preparation methods and principles of polysaccharide gold nanocarriers. It also provides an overview of the factors that affect the loading of polysaccharide gold nanocarriers with different kinds of drugs. Additionally, it outlines the strategies employed by polysaccharide gold nanocarriers to improve the delivery efficiency of various drugs. The objective is to provide a reference for further development of research on polysaccharide gold nanodelivery systems.

6.
Small Methods ; : e2301405, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38168901

ABSTRACT

Currently, the copolymer of dopamine (DA) and pyrrole (PY) via chemical and electrochemical oxidation usually requires additional oxidants, and lacks flexibility in regulating the size and morphology, thereby limiting the broad applications of DA-PY copolymer in biomedicine. Herein, the semiquinone radicals produced by the self-oxidation of DA is ingeniously utilized as the oxidant to initiate the following copolymerization with PY, and a series of quinone-rich polydopamine-pyrrole copolymers (PDAm -nPY) with significantly enhanced absorption in near-infrared (NIR) region without any additional oxidant assistance is obtained. Moreover, the morphology and size of PDAm -nPY can be regulated by changing the concentration of DA and PY, thereby optimizing nanoscale PDA0.05 -0.15PY particles (≈ 150 nm) with excellent NIR absorption and surface modification activity are successfully synthesized. Such PDA0.05 -0.15PY particles show effective photoacoustic (PA) imaging and photothermal therapy (PTT) against 4T1 tumors in vivo. Furthermore, other catechol derivatives can also copolymerize with PY under the same conditions. This work by fully utilizing the semiquinone radical active intermediates produced through the self-oxidation of DA reduces the dependence on external oxidants in the synthesis of composite materials and predigests the preparation procedure, which provides a novel, simple, and green strategy for the synthesis of other newly catechol-based functional copolymers.

7.
Int J Mol Sci ; 24(23)2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38069279

ABSTRACT

Tumors are a major public health issue of concern to humans, seriously threatening the safety of people's lives and property. With the increasing demand for early and accurate diagnosis and efficient treatment of tumors, noninvasive optical imaging (including fluorescence imaging and photoacoustic imaging) and tumor synergistic therapies (phototherapy synergistic with chemotherapy, phototherapy synergistic with immunotherapy, etc.) have received increasing attention. In particular, light in the near-infrared second region (NIR-II) has triggered great research interest due to its penetration depth, minimal tissue autofluorescence, and reduced tissue absorption and scattering. Nanomaterials with many advantages, such as high brightness, great photostability, tunable photophysical properties, and excellent biosafety offer unlimited possibilities and are being investigated for NIR-II tumor imaging-guided synergistic oncotherapy. In recent years, many researchers have tried various approaches to investigate nanomaterials, including gold nanomaterials, two-dimensional materials, metal sulfide oxides, polymers, carbon nanomaterials, NIR-II dyes, and other nanomaterials for tumor diagnostic and therapeutic integrated nanoplatform construction. In this paper, the application of multifunctional nanomaterials in tumor NIR-II imaging and collaborative therapy in the past three years is briefly reviewed, and the current research status is summarized and prospected, with a view to contributing to future tumor therapy.


Subject(s)
Nanoparticles , Nanostructures , Neoplasms , Humans , Phototherapy/methods , Polymers/therapeutic use , Nanostructures/therapeutic use , Neoplasms/therapy , Neoplasms/drug therapy , Optical Imaging , Theranostic Nanomedicine/methods
8.
Pharmaceuticals (Basel) ; 16(12)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38139769

ABSTRACT

Polydopamine (PDA) as a melanin-like biomimetic material with excellent biocompatibility, full spectrum light absorption capacity and antioxidation property has been extensively applied in the biomedical field. Based on the high reactivity of dopamine (DA), exploiting new strategies to fabricate novel PDA-based nano-biomaterials with controllable size and improved performance is valuable and desirable. Herein, we reported a facile way to synthesize pyrrole-doped polydopamine-pyrrole nanoparticles (PDA-nPY NPs) with tunable size and enhanced near-infrared (NIR) absorption capacity through self-oxidative polymerization of DA with PY in an alkaline ethanol/H2O/NH4OH solution. The PDA-nPY NPs maintain excellent biocompatibility and surface reactivity as PDA. By regulating the volume of added PY, PDA-150PY NPs with a smaller size (<100 nm) and four-fold higher absorption intensity at 808 nm than that of PDA can be successfully fabricated. In vitro and in vivo experiments effectively further demonstrate that PDA-150PY NPs can effectively inhibit tumor growth and completely thermally ablate a tumor. It is believed that these PY doped PDA-nPY NPs can be a potential photothermal (PT) agent in biomedical application.

9.
Phytother Res ; 37(11): 5341-5353, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37700535

ABSTRACT

BACKGROUND AND AIM: Our previous study has revealed that OEA promotes motor function recovery in the chronic stage of ischemic stroke. However, the neuroprotective mechanism of OEA on motor function recovery after stroke still is unexplored. Therefore, the aim of this study was to explore the effects of OEA treatment on angiogenesis, neurogenesis, and white matter repair in the peri-infarct region after cerebral ischemia. EXPERIMENTAL PROCEDURE: The adult male rats were subjected to 2 h of middle cerebral artery occlusion. The rats were treated with 10 and 30 mg/kg OEA or vehicle daily starting from day 2 after ischemia induction until they were sacrificed. KEY RESULTS AND CONCLUSIONS: The results revealed that OEA increased cortical angiogenesis, neural progenitor cells (NPCs) proliferation, migration, and differentiation. OEA treatment enhanced the survival of newborn neurons and oligodendrogenesis, which eventually repaired the cortical neuronal injury and improved motor function after ischemic stroke. Meanwhile, OEA treatment promoted the differentiation of oligodendrocyte progenitor cells (OPCs) and oligodendrogenesis by activating the PPARα signaling pathway. Our results showed that OEA restores motor function by facilitating cortical angiogenesis, neurogenesis, and white matter repair in rats after ischemic stroke. Therefore, we demonstrate that OEA facilitates functional recovery after ischemic stroke and propose the hypothesis that the long-term application of OEA mitigates the disability after stroke.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , White Matter , Rats , Male , Animals , White Matter/metabolism , PPAR alpha/metabolism , Brain Ischemia/drug therapy , Stroke/drug therapy , Neurogenesis , Cell Differentiation , Oligodendroglia/metabolism
10.
Pharmaceutics ; 15(9)2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37765202

ABSTRACT

Targeted drug delivery is a precise and effective strategy in oncotherapy that can accurately deliver drugs to tumor cells or tissues to enhance their therapeutic effect and, meanwhile, weaken their undesirable side effects on normal cells or tissues. In this research field, a large number of researchers have achieved significant breakthroughs and advances in oncotherapy. Typically, nanocarriers as a promising drug delivery strategy can effectively deliver drugs to the tumor site through enhanced permeability and retention (EPR) effect-mediated passive targeting and various types of receptor-mediated active targeting, respectively. Herein, we review recent targeted drug delivery strategies and technologies for enhancing oncotherapy. In addition, we also review two mainstream drug delivery strategies, passive and active targeting, based on various nanocarriers for enhancing tumor therapy. Meanwhile, a comparison and combination of passive and active targeting are also carried out. Furthermore, we discuss the associated challenges of passive and active targeted drug delivery strategies and the prospects for further study.

11.
J Colloid Interface Sci ; 646: 649-662, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37220698

ABSTRACT

Oxidative stress-based antitumor modalities derived from reactive oxygen species (ROS) storms have attracted increasing attention. Nevertheless, low delivery efficiency, poor selectivity, hypoxia and overexpressed glutathione (GSH) have severely restricted the sustainable generation of the ROS storm in tumor cells. Herein, we design a bioengineered nanogenerator by coordination-driven co-assembly of sonosensitizer indocyanine green (ICG), Fenton-like agent copper ion (CuⅡ) and mitochondrial respiratory inhibitor metformin (MET), which is then camouflaged by a cancer cytomembrane to induce a sustainable intracellular ROS storm for on-demand self-reinforcing sono-chemodynamic oncotherapy. Such a nanogenerator with a core-shell structure, suitable diameter and outstanding stability can efficiently accumulate in tumor regions and then internalize into tumor cells through the camouflaging and homologous targeting strategy of the cancer cytomembrane. The nanogenerator shows an exceptional instability under the triple stimulations of acidic lysosomes, overexpressed GSH and ultrasound (US) radiation, thereby resulting in the rapid disassembly and burst drug release. Interestingly, the released MET significantly enhances the sonodynamic therapy (SDT) efficacy of the released ICG by inhibiting mitochondrial respiration and meanwhile the released CuⅡ obviously reduces ROS elimination by downregulating overexpressed GSH for self-amplifying and self-protecting the intracellular ROS storm. Moreover, such a nanogenerator almost completely achieves the tumor ablation in vivo in a single therapy cycle. Taken together, our bioengineered nanogenerator with a sustainable ROS storm can provide a promising strategy for ROS storm-based oncotherapy.


Subject(s)
Metformin , Neoplasms , Humans , Reactive Oxygen Species , Oxidative Stress , Biomedical Engineering , Copper/pharmacology , Drug Liberation , Glutathione , Indocyanine Green , Metformin/pharmacology , Cell Line, Tumor , Neoplasms/drug therapy , Hydrogen Peroxide
12.
J Mater Chem B ; 11(16): 3679-3692, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37042187

ABSTRACT

Although oxidative stress-based antitumor modality derived from reactive oxygen species (ROS) storm has attracted considerable attention in copper-based nanomaterials, its efficiency is still weakened by the insufficient hydrogen peroxide (H2O2) and overexpressed glutathione (GSH) in a tumor microenvironment (TME). In view of this, we designed an engineered programmable spike-like nanogenerator via the coordination-driven co-assembly of Evans Blue (EB), copper ions (CuII), and 5-hydroxy-p-naphthoquinone (HND). For programmable nanogenerators, the introduction of EB as a stabilizer-like component can not only adjust its morphology but also achieve its visual tracking. Interestingly, such programmable nanogenerators can be efficiently enriched in tumor regions and then internalized into tumor cells due to ECH with spike-like morphology. Notably, once the nanogenerator is disintegrated and burst to release the drug upon acidic lysosome and endogenous GSH triggering, the released HND can not only efficiently amplify endogenous H2O2 by intracellular oxidoreductases but also down-regulate the peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin 1) activity. In addition, the released CuII ions can efficiently catalyze the degradation of the endogenous H2O2 to amplify hydroxyl radicals (˙OH) and down-regulate the overexpressed GSH to reduce ˙OH elimination for on-demand cascade-amplifying oxidative stress. Importantly, such programmable nanogenerators show an excellent antitumor effect via down-regulating the Pin 1 activity and cascade-amplifying oxidative stress. In this study, we propose a spatiotemporally programmable cascade nanogenerator for oxidative stress-based antitumor therapy.


Subject(s)
Copper , Hydrogen Peroxide , Copper/pharmacology , Hydrogen Peroxide/metabolism , Tumor Microenvironment , Oxidative Stress , Reactive Oxygen Species/metabolism , Glutathione/metabolism
13.
ACS Appl Mater Interfaces ; 15(14): 17495-17506, 2023 Apr 12.
Article in English | MEDLINE | ID: mdl-36996342

ABSTRACT

How to efficiently synthesize toxic chemo-drugs in the hypoxia tumor microenvironment still faces a huge challenge. Herein, we have tailored engineered vehicle-free nanoreactors by coordination-driven co-assembly of photosensitizer indocyanine green (ICG), transition metal platinum (Pt), and nontoxic 1,5-dihydroxynaphthalene (DHN) to self-amplify O2 and cascade chemo-drug synthesis in tumor cells for self-reinforcing hypoxic oncotherapy. Once vehicle-free nanoreactors are internalized into tumor cells, they show a serious instability that results in rapid disassembly and on-demand drug release under the stimuli of acidic lysosome and laser radiation. Notably, the released Pt can efficiently decompose the endogenous hydrogen peroxide (H2O2) into O2 to alleviate tumor hypoxia, which is conducive to enhancing the photodynamic therapy (PDT) efficiency of the released ICG. Complementarily, a large amount of the 1O2 generated by PDT can efficiently oxidize the released nontoxic DHN into the highly toxic chemo-drug juglone. Therefore, such vehicle-free nanoreactors can achieve intracellular on-demand cascade chemo-drug synthesis and self-reinforce photo-chemotherapeutic efficacy on the hypoxic tumor. On the whole, such a simple, flexible, efficient, and nontoxic therapeutic strategy will broaden the study of on-demand chemo-drug synthesis and hypoxic oncotherapy.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Photochemotherapy , Humans , Photochemotherapy/methods , Platinum/therapeutic use , Hydrogen Peroxide , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Hypoxia/drug therapy , Nanotechnology , Cell Line, Tumor , Tumor Microenvironment
14.
J Pharm Pharmacol ; 75(1): 87-97, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36332079

ABSTRACT

OBJECTIVES: Esculetin is a coumarin derivative, which is extracted from the dried barks of fraxinus chinensis Roxb. Although it is reported esculetin possesses multiple pharmacological activities, its associated regulatory mechanism on ovarian cancer isn't well investigated. METHODS: Cytotoxicity is evaluated by MTT, clonogenic and living/dead cells staining assays. Migration and invasion effects are investigated by wound healing, and transwell assays. The effect of cell cycle and apoptosis are analyzed by flow cytometry and western blotting. Mitochondrial membrane potential and intracellular reactive oxygen species (ROS) is assessed by fluorescence microscope. Analysis of animal experiments are carried out by various pathological section assays. KEY FINDINGS: Esculetin exerts an anti- ovarian cancer effect. It is found that apoptosis induction is promoted by the accumulation of excessive ROS and inhibition of JAK2/STAT3 signalling pathway. In addition, exposure to esculetin leads to the cell viability reduction, migration and invasion capability decrease and G0/G1 phase cell cycle arrest induced by down-regulating downstream targets of STAT3. In vivo experimental results also indicate esculetin can inhibit tumour growth of mice. CONCLUSIONS: Our study provides some strong evidences to support esculetin as a potential anti-cancer agent in ovarian cancer.


Subject(s)
Apoptosis , Ovarian Neoplasms , Animals , Mice , Female , Humans , Reactive Oxygen Species/metabolism , Cell Proliferation , Cell Line, Tumor , G1 Phase Cell Cycle Checkpoints , Ovarian Neoplasms/drug therapy , Janus Kinase 2/metabolism , Janus Kinase 2/pharmacology , STAT3 Transcription Factor/metabolism
15.
J Mater Chem B ; 11(1): 119-130, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36504220

ABSTRACT

Intracellular-synthesized chemo-drugs based on the inherent characteristics of the tumor microenvironment (TME) have been extensively applied in oncotherapy. However, combining other therapeutic strategies to convert nontoxic small molecules into toxic small-molecule chemo-drugs in the TME is still a huge challenge. To address this issue, herein we have developed a biomimetic dual-responsive bioengineered nanotheranostics system via the supramolecular co-assembly of the nontoxic small-molecule 1,5-dihydroxynaphthalene (DHN) and small-molecule photosensitizer indocyanine green (ICG) followed by surface cloaking through red blood cell membranes (RBCs) for intracellular cascade-synthesizing chemo-drugs and efficient oncotherapy. Such nanotheranostics with a suitable diameter, core-shell structure, ultrahigh dual-drug payload rate, and excellent stability can efficiently accumulate in tumor regions and then internalize into tumor cells. Under the dual stimulations of near-infrared laser irradiation and acidic lysosomes, the nanotheranostics system exhibited exceptional instability under heat-primed membrane rupture and pH decrease, thereby achieving rapid disassembly and on-demand drug release. Furthermore, the released ICG can efficiently convert 3O2 into 1O2. After that, the generated 1O2 can efficiently oxidize the released nontoxic DHN into the highly toxic chemo-drug juglone, thereby realizing intracellular cascade-synthesizing chemo-drugs and synergistic photodynamic-chemotherapy while reducing detrimental side effects on normal cells or tissues. Overall, it is envisioned that RBC-cloaked nanotheranostics with intracellular cascade-synthesizing chemo-drugs can provide a promising strategy for intracellular chemo-drug synthesis-based oncotherapy.


Subject(s)
Antineoplastic Agents , Biomimetics , Theranostic Nanomedicine , Antineoplastic Agents/pharmacology , Phototherapy , Photosensitizing Agents/chemistry , Indocyanine Green/pharmacology , Indocyanine Green/chemistry
16.
Acta Biomater ; 147: 258-269, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35605954

ABSTRACT

Chemodynamic therapy (CDT) has aroused extensive attention as a potent therapeutic modality. However, its practical application is severely restricted by the strong acidity requirement for Fenton reaction and upregulated antioxidant defense within metastatic breast cancer. Herein, a copper-based single-site nanocatalyst functionalized with carbonic anhydrase inhibitor (CAI) was constructed for magnetic resonance/photoacoustic imaging (MRI/PA)-guided synergetic photothermal therapy (PTT) and CDT. Once reaching tumor sites, the nanocatalyst can be recognized by tumor cell membranes-overexpressed carbonic anhydrase IX (CA IX). Subsequently, the single-site CuII can be reduced to CuI by the tumor-overexpressed glutathione (GSH), which simultaneously impaired the tumor antioxidant defense system and triggered CAI release for inducing intracellular H+ accumulation. Further, the decreased intracellular pH can accelerate the nanocatalyst biodegradation to release more CuII and CAI to participate in next-cycle GSH-depletion and cytoplasm acidification, respectively, thereby continuously supplying CuI and H+ for self-cyclically amplified CDT. Upon laser irradiation, the nanocatalyst can generate local heat, which not only permits PTT but also enhances the nanocatalyst-mediated CDT. Moreover, the suppression of CA IX can hinder the tumor extracellular matrix degradation to prevent tumor metastasis. Overall, this work highlighted the great application prospect in enhancing CDT via tumor acidic/redox microenvironment remodeling, and provides an insightful paradigm for inhibiting breast cancer metastasis. STATEMENT OF SIGNIFICANCE: The practical application of chemodynamic therapy (CDT) is severely restricted by the strong acidity requirement for Fenton reaction and upregulated antioxidant defense within cancer. Herein, we developed a carbonic anhydrase inhibitor (CAI)-functionalized Cu-based nanocatalyst. Once reaching tumor sites, the CuII can be reduced to CuI by the tumor-overexpressed glutathione (GSH), which simultaneously impaired the tumor antioxidant system and triggered CAI release for inducing intracellular H+ accumulation. Further, the decreased intracellular pH can accelerate the nanocatalyst biodegradation to release more CuII and CAI to participate in next-cycle GSH-depletion and cytoplasm acidification, respectively, thus continuously supplying CuI and H+ for self-cyclically amplified CDT. Upon laser irradiation, the nanocatalyst not only permits PTT but also enhances the CDT.


Subject(s)
Breast Neoplasms , Nanoparticles , Neoplasms , Antioxidants , Breast Neoplasms/drug therapy , Carbonic Anhydrase Inhibitors/therapeutic use , Cell Line, Tumor , Copper/pharmacology , Female , Glutathione/metabolism , Humans , Hydrogen Peroxide , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Photothermal Therapy , Precision Medicine , Theranostic Nanomedicine , Tumor Microenvironment
17.
Biomaterials ; 283: 121452, 2022 04.
Article in English | MEDLINE | ID: mdl-35286856

ABSTRACT

Here, inspired by the concept of supramolecular inclusion complex, we successfully fabricate metformin (Met)-based supramolecular nanodrugs with the Aß-responsive on-demand drug release for synergistic Alzheimer's disease (AD) therapy via enhancing microglial Aß clearance. Interestingly, the introduction of low-dosage Met (1.1 mg/kg) can not only significantly improve the structural stability of nanodrugs but also exert a synergistic anti-dementia effect with donepezil (Don). Besides, such nanodrugs with outstanding physiological stability can selectively penetrate the blood-brain barrier (BBB), target brain, increase efficient uptake of microglia and neurons, and then achieve simultaneous spatiotemporal on-demand drug release under stimuli of the overexpressed amyloid-beta (Aß). Furthermore, Met and Don released from nanodrugs exhibit a superior synergistic anti-dementia effect by enhancing microglial phagocytosis and Aß clearance through the lysosomal pathway. Taken together, we report a synergistic strategy based on Aß-responsive supramolecular nanodrugs for AD therapy, which can be expected to provide a novel clinical therapeutic idea for ameliorating central nervous system disease.


Subject(s)
Alzheimer Disease , Metformin , Nanoparticles , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Humans , Metformin/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Microglia , Nanoparticles/therapeutic use
18.
Environ Health Perspect ; 130(2): 27002, 2022 02.
Article in English | MEDLINE | ID: mdl-35108087

ABSTRACT

BACKGROUND: Severe environmental pollution and contaminants left in the environment due to the abuse of chemicals, such as methylmercury, are associated with an increasing number of embryonic disorders. Ultrasound imaging has been widely used to investigate embryonic development malformation and dysorganoplasia in both research and clinics. However, this technique is limited by its low contrast and lacking functional parameters such as the ability to measure blood oxygen saturation (SaO2) and hemoglobin content (HbT) in tissues, measures that could be early vital indicators for embryonic development abnormality. Herein, we proposed combining two highly complementary techniques into a photoacoustic-ultrasound (PA-US) dual-modality imaging approach to noninvasively detect early mouse embryo abnormalities caused by methylmercury chloride (MMC) in real time. OBJECTIVES: This study aimed to assess the use of PA-US dual-modality imaging for noninvasive detection of embryonic toxicity at different stages of growth following prenatal MMC exposure. Additionally, we compared the PA-US imagining results to traditional histological methods to determine whether this noninvasive method could detect early developmental defects in utero. METHODS: Different dosages of MMC were administrated to pregnant mice by gavage to establish models of different levels of embryonic malformation. Ultrasound, photoacoustic signal intensity (PSI), blood oxygen saturation (SaO2), and hemoglobin content (HbT) were quantified in all experimental groups. Furthermore, the embryos were sectioned and examined for pathological changes. RESULTS: Using PA-US imaging, we detected differences in PSI, SaO2, HbT, and heart volume at embryonic day (E)14.5 and E11.5 for low and high dosages of MMC, respectively. More important, our results showed that differences between control and treated embryos identified by in utero PA-US imaging were consistent with those identified in ex vivo embryos using histological methods. CONCLUSION: Our results suggest that noninvasive dual-modality PA-US is a promising strategy for detecting developmental toxicology in the uterus. Overall, this study presents a new approach for detecting embryonic toxicities, which could be crucial in clinics when diagnosing aberrant embryonic development. https://doi.org/10.1289/EHP8907.


Subject(s)
Methylmercury Compounds , Prenatal Exposure Delayed Effects , Animals , Embryo, Mammalian/diagnostic imaging , Female , Mammals , Methylmercury Compounds/toxicity , Mice , Pregnancy , Ultrasonography
19.
ACS Appl Mater Interfaces ; 14(4): 5033-5052, 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35045703

ABSTRACT

Although nanotheranostics have displayed striking potential toward precise nanomedicine, their targeting delivery and tumor penetration capacities are still impeded by several biological barriers. Besides, the current antitumor strategies mainly focus on killing tumor cells rather than antiangiogenesis. Enlightened by the fact that the smart transformable self-targeting nanotheranostics can enhance their targeting efficiency, tumor penetration, and cellular uptake, we herein report carrier-free Trojan-horse diameter-reducible metal-organic nanotheranostics by the coordination-driven supramolecular sequential co-assembly of the chemo-drug pemetrexed (PEM), transition-metal ions (FeIII), and antiangiogenesis pseudolaric acid B. Such nanotheranostics with both a high dual-drug payload efficiency and outstanding physiological stability are responsively decomposed into numerous ultra-small-diameter nanotheranostics under stimuli of the moderate acidic tumor microenvironment and then internalized into tumor cells through tumor-receptor-mediated self-targeting, synergistically enhancing tumor penetration and cellular uptake. Besides, such nanotheranostics enable visualization of self-targeting capacity under the macroscopic monitor of computed tomography/magnetic resonance imaging, thereby realizing efficient oncotherapy. Moreover, tumor microvessels are precisely monitored by optical coherence tomography angiography/laser speckle imaging during chemo-antiangiogenic therapy in vivo, visually verifying that such nanotheranostics possess an excellent antiangiogenic effect. Our work will provide a promising strategy for further tumor diagnosis and targeted therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Biocompatible Materials/pharmacology , Metal-Organic Frameworks/pharmacology , Neovascularization, Pathologic/drug therapy , Theranostic Nanomedicine , Angiogenesis Inhibitors/chemistry , Antineoplastic Agents/chemistry , Biocompatible Materials/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Diterpenes/chemistry , Diterpenes/pharmacology , Drug Screening Assays, Antitumor , Humans , Materials Testing , Metal-Organic Frameworks/chemistry , Neovascularization, Pathologic/pathology , Particle Size , Pemetrexed/chemistry , Pemetrexed/pharmacology , Surface Properties
20.
ACS Appl Mater Interfaces ; 13(28): 32690-32702, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34229434

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Drug Carriers/chemistry , Nanostructures/chemistry , Neoplasms/drug therapy , Tumor Hypoxia/drug effects , Animals , Antineoplastic Agents/chemistry , Camptothecin/chemistry , Camptothecin/therapeutic use , Catalysis/radiation effects , Cell Line, Tumor , Drug Carriers/radiation effects , Drug Liberation , Female , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Light , Methylene Blue/analogs & derivatives , Methylene Blue/radiation effects , Mice, Inbred BALB C , Nanostructures/radiation effects , Neoplasms/metabolism , Oxygen/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photothermal Therapy , Platinum/chemistry , Platinum/radiation effects , Polymers/chemical synthesis , Polymers/chemistry , Polymers/radiation effects , Porosity , Singlet Oxygen/metabolism , Theranostic Nanomedicine
SELECTION OF CITATIONS
SEARCH DETAIL
...