Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 144(9): 4091-4101, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35171598

ABSTRACT

Despite the clinical success of photodynamic therapy (PDT), the application of this medical technique is intrinsically limited by the low oxygen concentrations found in cancer tumors, hampering the production of therapeutically necessary singlet oxygen (1O2). To overcome this limitation, we report on a novel mitochondria-localized iridium(III) endoperoxide prodrug (2-O-IrAn), which, upon two-photon irradiation in NIR, synergistically releases a highly cytotoxic iridium(III) complex (2-IrAn), singlet oxygen, and an alkoxy radical. 2-O-IrAn was found to be highly (photo-)toxic in hypoxic tumor cells and multicellular tumor spheroids (MCTS) in the nanomolar range. To provide cancer selectivity and improve the pharmacological properties of 2-O-IrAn, it was encapsulated into a biotin-functionalized polymer. The generated nanoparticles were found to nearly fully eradicate the tumor inside a mouse model within a single treatment. This study presents, to the best of our knowledge, the first example of an iridium(III)-based endoperoxide prodrug for synergistic photodynamic therapy/photoactivated chemotherapy, opening up new avenues for the treatment of hypoxic tumors.


Subject(s)
Neoplasms , Photochemotherapy , Prodrugs , Animals , Cell Line, Tumor , Hypoxia/drug therapy , Iridium/pharmacology , Mice , Mitochondria , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Prodrugs/pharmacology , Prodrugs/therapeutic use , Singlet Oxygen/therapeutic use
2.
Adv Mater ; 34(1): e2100245, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34613635

ABSTRACT

The synthesis and the evaluation of the efficacy of a cycloruthenated complex, RuZ, is reported, to overcome multi-drug resistance (MDR) in cancer cells. RuZ can self-assemble into nanoaggregates in the cell culture medium, resulting in a high intracellular concentration of RuZ in MDR cancer cells. The self-assembly significantly decreases oxygen consumption and inhibits glycolysis, which decreases cellular adenosine triphosphate (ATP) levels. The decrease in ATP levels and its low affinity for the ABCB1 and ABCG2 transporters (which mediate MDR) significantly increase the retention of RuZ by MDR cancer cells. Furthermore, RuZ increases cellular oxidative stress, inducing DNA damage, and, in combination with the aforementioned effects of RuZ, increases the apoptosis of cancer cells. Proteomic profiling analysis suggests that the RuZ primarily decreases the expression of proteins that mediate glycolysis and aerobic mitochondrial respiration and increases the expression of proteins involved in apoptosis. RuZ inhibits the proliferation of 35 cancer cell lines, of which 7 cell lines are resistant to clinical drugs. It is also active in doxorubicin-resistant MDA-MB-231/Adr mouse tumor xenografts. To the best of our knowledge, the results are the first to show that self-assembled cycloruthenated complexes are efficacious in inhibiting the growth of MDR cancer cells.


Subject(s)
Antineoplastic Agents , Neoplasms , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Mice , Neoplasms/drug therapy , Proteomics
3.
Dalton Trans ; 50(40): 14332-14341, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34558567

ABSTRACT

Photodynamic therapy (PDT) provides an alternative option to root out localized triple-negative breast cancer (TNBC) and has been experiencing a surge of research interest over recent years. In this study, we put forward a paradigm of designing novel transition metal-based PSs with the following characteristics: favorable cell-permeability, significant light-harvesting ability and prominent ROS yield. A novel BODIPY-Ir(III) conjugate has been designed as a photoinduced ROS (1O2, ˙OH and ˙O2-) generator. BODIPY-Ir is highly photoactive in subduing cancer cells in the PDT regimen with PI values ranging from 172 to 519 and EC50 in the nanomolar regime. Among various cancerous cell lines, TNBC was especially sensitive to BODIPY-Ir-mediated PDT, with a stunning EC50 value of 4.32 nM (PI = 519) under a moderate flux of visible-light irradiation (500 nm, 10.5 mW cm-2). BODIPY-Ir mainly accumulates in mitochondria and induces cell apoptosis under irradiation. Furthermore, the nanomolar antiproliferative activity of BODIPY-Ir is retained under hypoxia (2.5% O2). This work sheds light on instilling the O2-independent type I mechanism and conferring a red-shift absorption to metal-based PSs which fundamentally facilitate the clinical translation of PSs.


Subject(s)
Antineoplastic Agents/pharmacology , Boron Compounds/pharmacology , Coordination Complexes/pharmacology , Ferric Compounds/pharmacology , Photosensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Boron Compounds/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Drug Screening Assays, Antitumor , Ferric Compounds/chemistry , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Photochemotherapy , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
4.
Biomaterials ; 276: 121064, 2021 09.
Article in English | MEDLINE | ID: mdl-34391019

ABSTRACT

The photodynamic therapy (PDT) of cancer is limited by tumor hypoxia as PDT efficiency depends on O2 concentration. A novel oxygen self-sufficient photosensitizer (Ru-g-C3N4) was therefore designed and synthesized via a facile one-pot method in order to overcome tumor hypoxia-induced PDT resistance. The photosensitizer is based on [Ru(bpy)2]2+ coordinated to g-C3N4 nanosheets by Ru-N bonding. Compared to pure g-C3N4, the resulting nanosheets exhibit increased water solubility, stronger visible light absorption, and enhanced biocompatibility. Once Ru-g-C3N4 is taken up by hypoxic tumor cells and exposed to visible light, the nanosheets not only catalyze the decomposition of H2O2 and H2O to generate O2, but also catalyze H2O2 and O2 concurrently to produce multiple ROS (•OH, •O2-, and 1O2). In addition, Ru-g-C3N4 affords luminescence imaging, while continuously generating O2 to alleviate hypoxia greatly improving PDT efficacy. To the best of our knowledge, this oxygen self-sufficient photosensitizer produced via grafting a metal complex onto g-C3N4 is the first of its type to be reported.


Subject(s)
Photochemotherapy , Ruthenium , Graphite , Humans , Hydrogen Peroxide , Hypoxia/drug therapy , Nitrogen Compounds , Oxygen , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species
6.
Anal Chem ; 93(22): 8062-8070, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34037386

ABSTRACT

Peroxynitrite (ONOO-) and glutathione (GSH), two unique reactive species, play an essential regulating role in the oxidation and antioxidation in the living body and are closely associated with various physiological and pathological processes, like cancer, cardiovascular disorders, diabetes, inflammation, Alzheimer's disease, and hepatotoxicity. Thus, it is crucial to study mitochondria ONOO-/GSH redox cycles by an effective molecular tool. In this work, a mitochondria-targeting and redox-reversible near-infrared (NIR) phosphorescent iridium complex, Ir-diol, has been synthesized and used for the detection and imaging of a cellular redox state by visualizing endogenous ONOO-/GSH content. Ir-diol shows excellent photophysical properties, including NIR emission (the maximum emissive wavelength for 704 nm, approximately) and high phosphorescent quantum yield (Φ = 0.136) and exhibits high sensitivity and selectivity toward ONOO-/GSH redox cycles in aqueous solution and living cells. Therefore, these features, combined with low cytotoxicity and excellent cell permeability, enable probe Ir-diol to monitor the changes of the intracellular ONOO-/GSH level induced by drug both in vitro and in vivo.


Subject(s)
Fluorescent Dyes , Iridium , Glutathione/metabolism , Mitochondria/metabolism , Oxidation-Reduction , Peroxynitrous Acid
7.
Nanoscale ; 13(16): 7590-7599, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33884385

ABSTRACT

Photodynamic therapy (PDT) is a promising noninvasive cancer treatment. PDT in the clinic faces several hurdles due to the unique tumor environment, a feature of which is high levels of glutathione (GSH). An excess amount of GSH consumes reactive oxygen species (ROS) generated by photosensitizers (PSs), reducing PDT efficiency. Herein, nano-photosensitizers (RuS1 NPs and RuS2 NPs) are reported. These consist of ruthenium complexes joined by disulfide bonds forming GSH sensitive polymer nanoparticles. The NPs achieve enhanced uptake compared to their constituent monomers. Inside cancer cells, high levels of GSH break the S-S bonds releasing PS molecules in the cell. The level of GSH is also then reduced leading to excellent PDT activity. Furthermore, RuS2 NPs functionalized with tumor targeting hyaluronic acid (HA@RuS2 NPs) assessed in vivo were highly effective with minimal side effects. To the best of our knowledge, RuS NPs are the first metal complex-based nano-assembled photosensitizers which exhibit enhanced specificity and consume endogenous GSH simultaneously, thus achieving excellent two-photon PDT efficiency in vitro and in vivo.


Subject(s)
Nanoparticles , Photochemotherapy , Photosensitizing Agents , Ruthenium , Cell Line, Tumor , Glutathione , Humans , Photosensitizing Agents/pharmacology
8.
Chem Commun (Camb) ; 57(14): 1790-1793, 2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33475648

ABSTRACT

A Ru(ii)-BODIPY conjugate has been rationally designed and exhibits an intense absorption in the NIR region to boost lysosome-targeted PDT in vitro and in vivo. The advantages of Ru(ii) and BODIPY were successfully instilled into the conjugate to yield highly effective PDT efficacy against malignant melanoma A375 cells (PI = 3448) and A375 mice xenografts.


Subject(s)
Boron Compounds/chemistry , Lysosomes/radiation effects , Photochemotherapy , Ruthenium Compounds/chemistry , Animals , Cell Line, Tumor , Humans , Infrared Rays , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/therapy , Photosensitizing Agents , Xenograft Model Antitumor Assays
9.
Angew Chem Int Ed Engl ; 60(9): 4657-4665, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33217194

ABSTRACT

Immunogenic cell death (ICD) is a vital component of therapeutically induced anti-tumor immunity. An iridium(III) complex (Ir1), containing an N,N-bis(2-chloroethyl)-azane derivate, as an endoplasmic reticulum-localized ICD inducer for non-small cell lung cancer (NSCLC) is reported. The characteristic discharge of damage-associated molecular patterns (DAMPs), that is, cell surface exposure of calreticulin (CRT), extracellular exclusion of high mobility group box 1 (HMGB1), and ATP, were generated by Ir1 in A549 lung cancer cells, accompanied by an increase in endoplasmic reticulum stress and reactive oxygen species (ROS). The vaccination of immunocompetent mice with Ir1-treated dying cells elicited an antitumor CD8+ T cell response and Foxp3+ T cell depletion, which eventually resulted in long-acting anti-tumor immunity by the activation of ICD in lung cancer cells. Ir1 is the first Ir-based complex that is capable of developing an immunomodulatory response by immunogenic cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Endoplasmic Reticulum Stress/drug effects , Immunogenic Cell Death/drug effects , Iridium/chemistry , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Calreticulin/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Endoplasmic Reticulum/metabolism , Female , HMGB1 Protein/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Survival Rate
10.
Angew Chem Int Ed Engl ; 60(8): 4150-4157, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33174359

ABSTRACT

The organoplatinum(II) complex [Pt(C^N^N)(Cl)] (C^N^N=5,6-diphenyl-2,2'-bipyridine, Pt1) can assemble into nanoaggregates via π-π stacking and complementary hydrogen bonds, rather than Pt-Pt interactions. Pt1 exhibits ratiometric dual emission, including rare blue emission (λem =445 nm) and assembly-induced yellow emission (λem =573 nm), under one- and two-photon excitation. Pt1 displays blue emission in cells with an intact membrane due to its low cellular uptake. In cells where the membrane is disrupted, uptake of the complex is increased and at higher concentrations yellow emission is observed. The ratio of yellow to blue emission shows a linear relationship to the loss of cell membrane integrity. Pt1 is, to our knowledge, the first example of an assembly-induced two-photon ratiometric dual emission organoplatinum complex. The excellent and unique characteristics of the complex enabled its use for the tracking of cell apoptosis, necrosis, and the inflammation process in zebrafish.


Subject(s)
Coordination Complexes/chemistry , Microscopy, Fluorescence, Multiphoton , Platinum/chemistry , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/metabolism , Coordination Complexes/pharmacology , Humans , Inflammation/chemically induced , Inflammation/diagnostic imaging , Larva/chemistry , Larva/metabolism , Pyridines/chemistry , Zebrafish/growth & development , Zebrafish/metabolism
11.
Angew Chem Int Ed Engl ; 59(46): 20697-20703, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32735748

ABSTRACT

The efficacy of photodynamic therapy is typically reliant on the local concentration and diffusion of oxygen. Due to the hypoxic microenvironment found in solid tumors, oxygen-independent photosensitizers are in great demand for cancer therapy. We herein report an iridium(III) anthraquinone complex as a mitochondrion-localized carbon-radical initiator. Its emission is turned on under hypoxic conditions after reduction by reductase. Furthermore, its two-photon excitation properties (λex =730 nm) are highly desirable for imaging. Upon irradiation, the reduced form of the complex generates carbon radicals, leading to a loss of mitochondrial membrane potential and cell death (IC50light =2.1 µm, IC50dark =58.2 µm, PI=27.7). The efficacy of the complex as a PDT agent was also demonstrated under hypoxic conditions in vivo. To the best of our knowledge, it is the first metal-complex-based theranostic agent which can generate carbon radicals for oxygen-independent two-photon photodynamic therapy.


Subject(s)
Carbon/chemistry , Cell Hypoxia , Mitochondria/drug effects , Neoplasms/drug therapy , Photosensitizing Agents/pharmacology , Humans , Mitochondria/metabolism , NADP/metabolism , Neoplasms/pathology , Photochemotherapy/methods , Photons , Spectrum Analysis/methods , Tumor Microenvironment
13.
ACS Appl Mater Interfaces ; 12(28): 31205-31216, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32628016

ABSTRACT

Oxidative stress induced by reactive oxygen species (ROS) is one of the major pathological mechanisms of acute kidney injury (AKI). Inorganic nanomaterial-mediated antioxidant therapy is considered a promising method for the prevention of AKI; however, currently available antioxidants for AKI exhibit limited clinical efficacy due to the glomerular filtration threshold (∼6 nm). To address this issue, we developed ultrasmall RuO2 nanoparticles (RuO2NPs) (average size ≈ 2 nm). The NPs show excellent antioxidant activity and low biological toxicity. In addition, they can pass through the glomerulus to be excreted. These properties in combination make the ultrasmall RuO2NPs promising as a nanozyme for the prevention of AKI. The NP catalytic properties mimic the activity of catalase, peroxidase, superoxide dismutase, and glutathione peroxidase. The nanozyme can be efficiently and rapidly absorbed by human embryonic kidney cells while significantly reducing ROS-induced apoptosis by eliminating excess ROS. After intravenous injection, the ultrasmall RuO2NPs significantly inhibit the development of AKI in mice. In vivo toxicity experiments demonstrate the biosafety of the NPs after long-term preventing. The multienzyme-like activity and biocompatibility of the ultrasmall RuO2NPs makes them of great interest for applications in the fields of biomedicine and biocatalysis.


Subject(s)
Acute Kidney Injury/drug therapy , Ruthenium Compounds/chemistry , Animals , Catalase/metabolism , Catalysis , Glutathione Peroxidase/metabolism , Humans , Mice , Nanoparticles/chemistry , Oxidative Stress/drug effects , Peroxidase/metabolism , Reactive Oxygen Species/metabolism , Ruthenium Compounds/therapeutic use , Superoxide Dismutase/metabolism
14.
Angew Chem Int Ed Engl ; 59(38): 16631-16637, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32533618

ABSTRACT

Inducing necroptosis in cancer cells is an effective approach to circumvent drug-resistance. Metal-based triggers have, however, rarely been reported. Ruthenium(II) complexes containing 1,1-(pyrazin-2-yl)pyreno[4,5-e][1,2,4]triazine were developed with a series of different ancillary ligands (Ru1-7). The combination of the main ligand with bipyridyl and phenylpyridyl ligands endows Ru7 with superior nucleus-targeting properties. As a rare dual catalytic inhibitor, Ru7 effectively inhibits the endogenous activities of topoisomerase (topo) I and II and kills cancer cells by necroptosis. The cell signaling pathway from topo inhibition to necroptosis was elucidated. Furthermore, Ru7 displays significant antitumor activity against drug-resistant cancer cells in vivo. To the best of our knowledge, Ru7 is the first Ru-based necroptosis-inducing chemotherapeutic agent.


Subject(s)
Coordination Complexes/pharmacology , DNA Topoisomerases, Type I/metabolism , Necroptosis/drug effects , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Ruthenium/pharmacology , Topoisomerase Inhibitors/pharmacology , Animals , Biocatalysis/drug effects , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , DNA Topoisomerases, Type II/metabolism , Humans , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Poly-ADP-Ribose Binding Proteins/metabolism , Ruthenium/chemistry , Topoisomerase Inhibitors/chemical synthesis , Topoisomerase Inhibitors/chemistry
15.
Angew Chem Int Ed Engl ; 59(37): 15987-15991, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32473088

ABSTRACT

Compared to 2PE (two-photon excitation) microscopy, 3PE microscopy has superior spatial resolution, deeper tissue penetration, and less defocused interference. The design of suitable agents with a large Stokes shift, good three-photon absorption (3PA), subcellular targeting, and fluorescence lifetime imaging (FLIM) properties, is challenging. Now, two IrIII complexes (3PAIr1 and 3PAIr2) were developed as efficient three-photon phosphorescence (3PP) agents. Calculations reveal that the introduction of a new group to the molecular scaffold confers a quadruple promotion in three-photon transition probability. Confocal and lifetime imaging of mitochondria using IrIII complexes as 3PP agents is shown. The complexes exhibit low working concentration (50 nm), fast uptake (5 min), and low threshold for three-photon excitation power (0.5 mW at 980 nm). The impressive tissue penetration depth (ca. 450 µm) allowed the 3D imaging and reconstruction of brain vasculature from a living specimen.


Subject(s)
Coordination Complexes/chemistry , Iridium/chemistry , Optical Imaging/methods , Animals , HeLa Cells , Humans , Mice , Photons , Zebrafish
16.
Chem Commun (Camb) ; 56(43): 5839-5842, 2020 May 28.
Article in English | MEDLINE | ID: mdl-32330213

ABSTRACT

Herein, we present a series of dual-targeted ruthenium-glucose conjugates that can function as two-photon absorption (TPA) PDT agents to effectively destroy tumors by preferentially targeting both tumor cells and mitochondria. The in vivo experiments revealed an excellent tumor inhibitory efficiency of the dual-targeted TPA PSs.


Subject(s)
Glucose/therapeutic use , Mitochondria/metabolism , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/therapeutic use , Ruthenium/therapeutic use , Animals , Cell Line , Cell Survival/drug effects , Glucose/pharmacology , Humans , Light , Mice, Inbred BALB C , Neoplasms/metabolism , Neoplasms/pathology , Photosensitizing Agents/pharmacology , Ruthenium/pharmacology , Tumor Burden/drug effects
17.
Anal Chem ; 92(8): 6003-6009, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32212607

ABSTRACT

Nitric oxide (NO) and superoxide anions (O2•-) are two noteworthy reactive species implicated in various physiological and pathological processes, such as ROS-induced lysosomal cell death. The interaction ("crosstalk") between them may form a new mediator peroxynitrite (ONOO-) which has implications for cancer, diabetes, Alzheimer's disease, and liver-damage. It is therefore essential to investigate lysosomal NO/O2•- crosstalk in vivo through ONOO--responsive molecular tools in order to fully comprehend the physiological and pathological mechanisms involved. In this study, a lysosome-targeting iridium(III) complex, Ir-NIR, has been investigated as a near-infrared (NIR) phosphorescent probe for visualizing NO/O2•- crosstalk by the phosphorescent detection of endogenous ONOO- levels in vivo. Ir-NIR exhibits a rapid (within 200 s), highly sensitive, and approximately 100-fold enhanced response to ONOO- in phosphorescence intensity. Thus, these characteristics, coupled with good cell permeability and low cytotoxicity, enable the probe to be used to detect intracellular ONOO- living organisms both in vitro and in vivo.


Subject(s)
Coordination Complexes/chemistry , Fluorescent Dyes/chemistry , Iridium/chemistry , Nitric Oxide/metabolism , Peroxynitrous Acid/metabolism , Superoxides/metabolism , Animals , Cells, Cultured , Coordination Complexes/chemical synthesis , Female , Fluorescent Dyes/chemical synthesis , Humans , Infrared Rays , Luminescent Measurements , Lysosomes , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Molecular Structure , Nitric Oxide/chemistry , Peroxynitrous Acid/analysis , Superoxides/chemistry
18.
Chem Commun (Camb) ; 56(25): 3698, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32186567

ABSTRACT

Correction for 'Mitochondria-targeted Ir@AuNRs as bifunctional therapeutic agents for hypoxia imaging and photothermal therapy' by Libing Ke et al., Chem. Commun., 2019, 55, 10273-10276.

19.
Chem Commun (Camb) ; 56(25): 3697, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32186575

ABSTRACT

Correction for 'Fabrication of red blood cell membrane-camouflaged Cu2-xSe nanoparticles for phototherapy in the second near-infrared window' by Zhou Liu et al., Chem. Commun., 2019, 55, 6523-6526.

20.
Chem Commun (Camb) ; 56(20): 3019-3022, 2020 Mar 10.
Article in English | MEDLINE | ID: mdl-32048647

ABSTRACT

Nucleus-targeting NPs based on RuO2 (RuO2NPs) were developed by controlling the size and the surface charge of nanoparticles (NPs). This study not only demonstrates a facile approach for the fabrication of ultrasmall CS-RuO2NPs with good biocompatibility and excellent photothermal properties but also their unique potential for the nucleus-targeted low-temperature PTT.


Subject(s)
Nanoparticles/chemistry , Optical Imaging , Oxides/chemistry , Photoacoustic Techniques , Ruthenium/chemistry , Temperature , Cell Nucleus/drug effects , Humans , Infrared Rays , MCF-7 Cells , Oxides/pharmacology , Particle Size , Phototherapy , Ruthenium/pharmacology , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...