Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Angew Chem Int Ed Engl ; : e202411725, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39045805

ABSTRACT

The strategy of in vivo self-assembly has been developed for improved enrichment and long-term retention of anticancer drug in tumor tissues. However, most self-assemblies with non-covalent bonding interactions are susceptible to complex physiological environments, leading to weak stability and loss of biological function. Here, we develop a coupling-induced assembly (CIA) strategy to generate covalently crosslinked nanofibers, which is applied for in situ constructing artificial shell on mitochondria. The oxidation-responsive peptide-porphyrin conjugate P1 is synthesized, which self-assemble into nanoparticles. Under the oxidative microenvironment of mitochondria, the coupling of thiols in P1 causes the formation of dimers, which is further ordered and stacked into crosslinked nanofibers. As a result, the artificial shell is constructed on the mitochondria efficiently through multivalent cooperative interactions due to the increased binding sites. Under ultrasound (US) irradiation, the porphyrin molecules in the shell produce a large amount of reactive oxygen species (ROS) that act on the adjacent mitochondrial membrane, exhibiting ~2-fold higher antitumor activity than nanoparticles in vitro and in vivo. Therefore, the mitochondria-targeted CIA strategy provides a novel perspective on improved sonodynamic therapy (SDT) and shows potential applications in antitumor therapies.

2.
Bioorg Chem ; 139: 106746, 2023 10.
Article in English | MEDLINE | ID: mdl-37506624

ABSTRACT

Tumour cells show a higher level of reactive oxygen species (ROS) than normal cells. On the basis of this difference, we designed an oxidation-responsive G-quadruplex proligand PDS-B by installing borolanylbenzyls on a well-known pyridostatin (PDS) ligand PDS-S to response high level ROS in tumour cells. The rapid oxidative degradation of the proligand to its active form PDS-S in the presence of H2O2 confirms the oxidation-responsive design. According to Förster resonance energy transfer (FRET) assays, circular dichroism (CD) spectra and confocal fluorescence imaging, PDS-B stabilizes telomeric G4 structures after oxidation with H2O2 or intracellular ROS. Apoptosis assays and cell cycle assays showed significant selectivity of PDS-B in inhibiting the proliferation of tumour cells over normal cells through responses to a high level of ROS in the formers. Further assays confirmed higher level of relative Caspase-3 activity in tumour cells than normal cells, consequently the enhanced apoptosis of the tumour cells induced by PDS-B. In summary, the results demonstrate a modification approach to solve the poor selectivity of the G4 ligand in tumour cells and cytotoxicity in normal cells.


Subject(s)
G-Quadruplexes , Neoplasms , Humans , Ligands , Hydrogen Peroxide , Reactive Oxygen Species , Cell Proliferation , Circular Dichroism
3.
Biopolymers ; 111(1): e23336, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31724750

ABSTRACT

The pharmacological therapy for gastrointestinal (GI) diseases, such as inflammatory bowel diseases, continues to present challenges in targeting efficacy. The need for maximal local drug exposure at the inflamed regions of the GI tract has led research to focus on a disease-targeted drug delivery approach. Smart nanomaterials responsive to the reactive oxygen species (ROS) concentrated in the inflamed areas, can be formulated into nanoplatforms to selectively release the active compounds, avoiding unspecific drug delivery to healthy tissues and limiting systemic absorption. Recent developments of ROS-responsive nanoplatforms include combination with other materials to obtain multi-responsive systems and modifications/derivatization to increase the interactions with biological tissues, cell uptake and targeting. This review describes the applications of ROS-responsive nanosystems for on-demand drug delivery to the GI tract.


Subject(s)
Drug Carriers/chemistry , Gastrointestinal Tract/drug effects , Inflammatory Bowel Diseases/drug therapy , Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , Drug Carriers/metabolism , Drug Carriers/pharmacology , Drug Carriers/therapeutic use , Drug Delivery Systems/methods , Enzymes , Gastrointestinal Tract/pathology , Humans , Hydrogen-Ion Concentration , Inflammation/drug therapy , Inflammation/metabolism , Nanoparticles/metabolism , Nanoparticles/therapeutic use , Nanoparticles/ultrastructure , Reactive Oxygen Species/chemistry
4.
Small ; 15(45): e1904017, 2019 11.
Article in English | MEDLINE | ID: mdl-31538412

ABSTRACT

Microenvironment-responsive supramolecular assemblies have attracted great interest in the biomedical field due to their potential applications in controlled drug release. In this study, oxidation-responsive supramolecular polycationic assemblies named CPAs are prepared for nucleic acid delivery via the host-guest interaction of ß-cyclodextrin based polycations and a ferrocene-functionalized zinc tetraaminophthalocyanine core. The reactive oxygen species (ROS) can accelerate the disassembly of CPA/pDNA complexes, which would facilitate the release of pDNA in the complexes and further benefit the subsequent transfection. Such improvement in transfection efficiency is proved in A549 cells with high H2 O2 concentration. Interestingly, the transfection efficiencies mediated by CPAs are also different in the presence or absence of light in various cell lines such as HEK 293 and 4T1. The single oxygen (1 O2 ), produced by photosensitizers in the core of CPAs under light, increases the ROS amount and accelerates the disassembly of CPAs/pDNA complexes. In vitro and in vivo studies further illustrate that suppressor tumor gene p53 delivered by CPAs exhibits great antitumor effects under illumination. This work provides a promising strategy for the design and fabrication of oxidation-responsive nanoassemblies with light-enhanced gene transfection performance.


Subject(s)
Genetic Therapy/methods , Cell Line, Tumor , Delayed-Action Preparations , Drug Carriers/chemistry , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Polyelectrolytes/chemistry , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , beta-Cyclodextrins/chemistry
5.
Mol Pharm ; 15(11): 4985-4994, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30274517

ABSTRACT

Nanosized drug delivery systems based on polymeric structures have been proven to be promising approaches for cancer treatments. However, few have been effective at selectively targeting cancer cells and releasing drug at desired tumor sites. Here, we report a "smart" polymeric nanoplatform, which could actively accumulate at tumor sites and dissociate to release encapsulated cargos upon the irradiation of a near-infrared (NIR) laser. This nanoplatform composed of a novel amphiphilic block copolymer poly(propylene sulfide)-poly( N-isopropylacrylamide- co- N, N-dimethylacrylamide) (PPS-P(NIPAM- co-DMAA)) formed spherical structures in aqueous solution and responded to both oxidants and elevated temperature. Upon laser irradiation at 808 nm, the NIR light was efficiently converted to local heat by the doxorubicin (DOX) and indocyanine green (ICG) co-loaded micelles for enhanced cell uptake and therapeutic efficacy. It showed that the micelles effectively accumulated at the tumor sites guided by the application of an NIR laser in in vivo studies, exhibiting a 6-time greater and much faster targeting effect compared to the nonirradiation group. The effective tumor growth inhibition by the drug-loaded micelles upon laser irradiation demonstrated significant tumor inhibition without regrowth in 16 days. This micellar nanoplatform for precise NIR-guided cancer targeting and combination therapy provides a novel and robust strategy for cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Delivery Systems/methods , Drug Liberation/radiation effects , Infrared Rays , Neoplasms/drug therapy , A549 Cells , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Humans , Indocyanine Green/administration & dosage , Indocyanine Green/pharmacokinetics , Mice, Nude , Micelles , Nanoparticles/chemistry , Polymers/chemistry , Polymers/radiation effects , Time Factors , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Angew Chem Int Ed Engl ; 57(28): 8463-8467, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29757483

ABSTRACT

The approach of concurrent-to-synchronous chemoradiation has now been advanced by well-designed nanovesicles that permit X-ray irradiation-triggered instant drug release. The nanovesicles consist of Au nanoparticles tethered with irradiation labile linoleic acid hydroperoxide (LAHP) molecules and oxidation-responsive poly(propylene sulfide)-poly(ethylene glycol) (PPS-PEG) polymers, where DOX were loaded in the inner core of the vesicles (Au-LAHP-vDOX). Upon irradiation, the in situ formation of hydroxyl radicals from LAHP molecules triggers the internal oxidation of PPS from being hydrophobic to hydrophilic, leading to degradation of the vesicles and burst release of cargo drugs. In this manner, synchronous chemoradiation showed impressive anticancer efficacy both in vitro and in a subcutaneous mouse tumor model by one-dose injection and one-time irradiation.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Drug Carriers/chemistry , Drug Carriers/radiation effects , Drug Liberation/radiation effects , Gold/chemistry , Nanoparticles/chemistry , X-Rays , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Humans , Linoleic Acids/chemistry , Lipid Peroxides/chemistry , Mice , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Particle Size , Polymers/chemistry , Positron-Emission Tomography , Surface Properties
7.
Macromol Rapid Commun ; 36(22): 2012-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26297612

ABSTRACT

Polymer-drug conjugates have attracted great interest as one category of various promising nanomedicines due to the advantages of high drug-loading capacity, negligible burst release, and improved pharmacokinetics as compared with the small molecular weight drugs or the polymeric delivery systems with physically encapsulated drugs. Herein, a new type of oxidation-responsive polymer-drug conjugates composed of a poly(ethylene glycol) (PEG) block and a hydrophobic polyacrylate block to which Naproxen is attached through a phenylboronic ester linker is reported. The amphiphilic block copolymers are synthesized through the reversible addition-fragmentation chain transfer polymerization of the Naproxen-containing acrylic monomer using a PEG chain transfer agent. In neutral aqueous buffer, the conjugates formed nanoparticles with diameters of ≈150-300 nm depending on the length of the hydrophobic segment. The dynamic covalent bond of the phenylboronic ester is stabilized due to the hydrophobic microenvironment inside the nanoparticles. Upon exposure to H2 O2 , the phenylboronic ester is oxidized rapidly into the phenol derivative which underwent a 1,6-elimination reaction, releasing the intact Naproxen. The rate of drug release is influenced by the concentration of H2 O2 and the hydrophobic block length. This type of oxidation-responsive polymer-drug conjugate is feasible for other drugs containing hydroxyl group or amino group.


Subject(s)
Acrylic Resins/chemistry , Boronic Acids/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Naproxen/chemistry , Polyethylene Glycols/chemistry , Drug Compounding , Drug Liberation , Esters , Hydrogen Peroxide/chemistry , Hydrophobic and Hydrophilic Interactions , Kinetics , Micelles , Nanomedicine/methods , Nanoparticles/ultrastructure , Oxidation-Reduction , Particle Size , Polymerization
8.
ACS Nano ; 17(10): 9374-9387, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37141569

ABSTRACT

Stimulator of interferon genes (STING) activation by STING agonists has been recognized as one of the potent and promising immunotherapy strategies. However, the immunosuppressive tumor microenvironment always hinders the therapeutic efficacy of cancer immunotherapy. In this report, we present polymeric metal-organic framework (PMOF) nanoparticles (NPs) for the combination of photodynamic therapy (PDT) and enhanced STING activation to improve the immunotherapeutic efficacy. The PMOF NPs with poly(ethylene glycol) (PEG) shells were obtained via coordination between the block copolymer ligand PEG-b-PABDA consisting of 1,4-bezenedicarboxylic acid-bearing polyacrylamide (PABDA), meso-tetra(carboxyphenyl)porphyrin (TCPP), thioketal diacetic acid, and zirconyl chloride. Subsequently, the STING agonist SR-717 was loaded into the porous structure of PMOF to obtain SR@PMOF NPs which show excellent stability under the physiological conditions. After intravenous injection and tumor accumulation, light irradiation on the tumor sites results in efficient singlet oxygen (1O2) production from TCPP and cellular apoptosis to release fragmented DNA and tumor-associated antigens. Simultaneously, thioketal bonds can be broken by 1O2 to destroy the PMOF structure and rapidly release SR717. SR-717 and PDT synergistically enhance the antitumor immunity via combination photodynamic-immunotherapy due to reversal of the immunosuppressive tumor microenvironment and enhanced endogenous STING activation, which can suppress the growth of the primary and distant tumors efficiently. The oxidation-responsive SR@PMOF NPs represent a promising delivery system of STING agonists and efficient PDT NPs for simultaneous suppression of the primary and metastatic tumors via the rational combination of PDT and enhanced STING activation.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Humans , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemistry , Cell Line, Tumor , Neoplasms/therapy , Nanoparticles/chemistry , Polymers , Immunotherapy , Tumor Microenvironment
9.
Polymers (Basel) ; 15(9)2023 Apr 23.
Article in English | MEDLINE | ID: mdl-37177138

ABSTRACT

Micellar-nanocarrier-based drug delivery systems possessing characteristics such as an excellent circulation stability, inhibited premature release and on-demand site-specific release are urgently needed for enhanced therapeutic efficacy. Therefore, a novel kind of shell-sheddable core-crosslinked polymeric micelles with pH and oxidation dual-triggered on-demand drug release behavior was facilely constructed. The multifunctional micelles were self-assembled from a carefully designed amphiphilic triblock PEGylated polyurethane (PEG-acetal-PUBr-acetal-PEG) employing an acid-labile acetal linker at the hydrophilic-hydrophobic interface and pendant reactive bromo-containing polyurethane (PU) as the hydrophobic block, followed by a post-crosslinking via oxidation-cleavable diselenide linkages. These well-defined micelles exhibited an enhanced structural stability against dilution, achieved through the incorporation of diselenide crosslinkers. As expected, they were found to possess dual pH- and oxidation-responsive dissociation behaviors when exposure to acid pH (~5.0) and 50 mM H2O2 conditions, as evidenced using dynamic light-scattering (DLS) and atomic force microscopy (AFM) analyses. An in vitro drug release investigation showed that the drug indomethacin (IND) could be efficiently encapsulated in the micelles, which demonstrated an inhibited premature release compared to the non-crosslinked ones. It is noteworthy that the resulting micelles could efficiently release entrapped drugs at a fast rate in response to either pH or oxidation stimuli. Moreover, the release could be significantly accelerated in the presence of both acid pH and oxidation conditions, relative to a single stimulus, owing to the synergetic degradation of micelles through pH-induced dePEGylation and oxidation-triggered decrosslinking processes. The proposed shell-sheddable core-crosslinked micelles with a pH and oxidation dual-response could be potential candidates as drug carriers for on-demand drug delivery.

10.
Asian J Pharm Sci ; 17(2): 241-252, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35582643

ABSTRACT

PEGylation has been widely used to improve the pharmacokinetic properties of prodrug self-assembled nanoparticles (prodrug-SANPs). However, the impacts of the amount of PEG on the self-assemble stability, cellular uptake, pharmacokinetics, and antitumor efficacy of prodrug-SANPs are still unknown. Herein, selenoether bond bridged docetaxel dimeric prodrug was synthesized as the model prodrug. Five prodrug-SANPs were designed by using different mass ratios of prodrugs to PEG (Wprodrug/WDSPE-mPEG2000 = 10:0, 9:1, 8:2, 7:3 and 6:4), and defined as Pure drug NPs, 9:1NPs, 8:2NPs, 7:3 NPs and 6:4 NPs, respectively. Interestingly, 8:2 NPs formed the most compact nanostructure, thus improving the self-assemble stability and pharmacokinetics behavior. In addition, the difference of these prodrug-SANPs in cellular uptake was investigated, and the influence of PEG on cytotoxicity and antitumor efficacy was also clarified in details. The 8:2 NPs exhibited much better antitumor efficacy than other prodrug-SANPs and even commercial product. Our findings demonstrated the pivotal role of the amount of PEG on prodrug-SANPs.

11.
Nanomicro Lett ; 13(1): 147, 2021 Jun 19.
Article in English | MEDLINE | ID: mdl-34146147

ABSTRACT

Creating a single surfactant that is open to manipulation, while maintaining its surface activity, robustness, and compatibility, to expand the landscape of surfactant-dependent assays is extremely challenging. We report an oxidation-responsive precursor with thioethers and multiple 1,2-diols for creating a variety of functional surfactants from one parent surfactant. Using these multifunctional surfactants, we stabilize microfluidics-generated aqueous droplets. The droplets encapsulate different components and immerse in a bioinert oil with distinct interfaces where an azide-bearing surfactant allow fishing of biomolecules from the droplets, aldehyde-bearing surfactant allow fabrication of microcapsules, and hydroxyl-bearing surfactants, with/without oxidized thioethers, allow monitoring of single-cell gene expression. Creating multifunctional surfactants poses opportunities for broad applications, including adsorption, bioanalytics, catalysis, formulations, coatings, and programmable subset of emulsions.

12.
ACS Appl Mater Interfaces ; 13(30): 35431-35443, 2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34304556

ABSTRACT

Stent implantation is the primary method used to treat coronary heart disease. However, it is associated with complications such as restenosis and late thrombosis. Despite surface modification being an effective way to improve the biocompatibility of stents, the current research studies are not focused on changes in the vascular microenvironment at the implantation site. In the present study, an adaptive drug-loaded coating was constructed on the surface of vascular stent materials that can respond to oxidative stress at the site of vascular lesions. Two functional molecules, epigallocatechin gallate (EGCG) and cysteine hydrochloride, were employed to fabricate a coating on the surface of 316L stainless steel. In addition, the coating was used as a drug carrier to load pitavastatin calcium. EGCG has antioxidant activity, and pitavastatin calcium can inhibit smooth muscle cell proliferation. Therefore, EGCG and pitavastatin calcium provided a synergistic anti-inflammatory effect. Moreover, the coating was cross-linked using disulfide bonds, which accelerated the release of the drug in response to reactive oxygen species. A positive correlation was observed between the rate of drug release and the degree of oxidative stress. Collectively, this drug-loaded oxidative stress-responsive coating has been demonstrated to significantly inhibit inflammation, accelerate endothelialization, and reduce the risk of restenosis of vascular stents in vivo.


Subject(s)
Drug-Eluting Stents , Reactive Oxygen Species/antagonists & inhibitors , Animals , Catechin/administration & dosage , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Coronary Restenosis/prevention & control , Cystamine/administration & dosage , Cystamine/chemistry , Drug Liberation , Endothelial Cells/cytology , Endothelial Cells/drug effects , Macrophages/cytology , Macrophages/drug effects , Male , Myocytes, Smooth Muscle/drug effects , Neovascularization, Physiologic/drug effects , Oxidation-Reduction/drug effects , Quinolines/administration & dosage , Quinolines/chemistry , Quinolines/pharmacology , Rabbits , Rats, Sprague-Dawley , Stainless Steel/chemistry
13.
Adv Healthc Mater ; 9(16): e2000533, 2020 08.
Article in English | MEDLINE | ID: mdl-32603033

ABSTRACT

Singlet oxygen is regarded as the primary cytotoxic agent in cancer photodynamic therapy (PDT). Despite the advances in optical methods to image singlet oxygen, it remains a challenge for in vivo application due to the limited tissue penetration depth of light. Up to date, no singlet oxygen-specific magnetic resonance imaging (MRI) probe has been reported. Herein, a T2 -weighted MRI probe is reported to visually detect singlet oxygen generated in PDT in vitro and in vivo. The MRI probe Ce6/Fe3 O4 -M is constructed by co-encapsulation of photosensitizer Ce6 and Fe3 O4 nanoparticles in mPEG2000 -TK-C16 micelles. Thioketal (TK) linker in the probe is highly sensitive to singlet oxygen, but lowly sensitive to other reactive oxygen species (ROS) existing in physiological and pathological environments. Singlet oxygen, generated with light irradiation, triggers the cleavage of TK, which leads to loss of surface polyethylene glycol, increment of the hydrophobicity, and aggregation of Fe3 O4 nanoparticles. Subsequently, negatively enhanced T2 -weighted MRI signal is obtained for visual detection of singlet oxygen in the solution, cancer cells, and in vivo. This oxidation responsive MRI probe is expected to hold great promise in evaluating the ability of photosensitizers to generate singlet oxygen and in predicting the therapeutic efficacies of PDT in vivo.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Cell Line, Tumor , Magnetic Resonance Imaging , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Singlet Oxygen/therapeutic use
14.
Int J Nanomedicine ; 15: 137-149, 2020.
Article in English | MEDLINE | ID: mdl-32021170

ABSTRACT

PURPOSE: Traditional chemotherapy is accompanied by significant side effects, which, in many aspects, limits its treatment efficacy and clinical applications. Herein, we report an oxidative responsive polymersome nanosystem mediated by near infrared (NIR) light which exhibited the combination effect of photodynamic therapy (PDT) and chemotherapy. METHODS: In our study, poly (propylene sulfide)20-bl-poly (ethylene glycol)12 (PPS20-b-PEG12) block copolymer was synthesized and employed to prepare the polymersome. The hydrophobic photosensitizer zinc phthalocyanine (ZnPc) was loaded in the shell and the hydrophilic doxorubicin hydrochloride (DOX·HCl) in the inner aqueous space of the polymersome. RESULTS: Under the irradiation of 660 nm NIR light, singlet oxygen 1O2 molecules were generated from ZnPc to oxidize the neighbouring sulfur atoms on the PPS block which eventually ruptured the intact structure of polymersomes, leading to the release of encapsulated DOX·HCl. The released DOX and the 1O2 could achieve a combination effect for cancer therapy if the laser activation and drug release occur at the tumoral sites. In vitro studies confirmed the generation of singlet oxygen and DOX release by NIR irradiation. In vivo studies showed that such a combined PDT-chemotherapy nanosystem could accumulate in A375 tumors efficiently, thus leading to significant inhibition on tumor growth as compared to PDT (PZ group) or chemotherapy alone (DOX group). CONCLUSION: In summary, this oxidation-sensitive nanosystem showed excellent anti-tumor effects by synergistic chemophotodynamic therapy, indicating that this novel drug delivery strategy could potentially provide a new means for cancer treatments in clinic.


Subject(s)
Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Melanoma, Experimental/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Drug Liberation , Humans , Indoles/administration & dosage , Indoles/chemistry , Isoindoles , Lasers , Male , Mice, Inbred BALB C , Nanostructures/administration & dosage , Nanostructures/chemistry , Organometallic Compounds/administration & dosage , Organometallic Compounds/chemistry , Photosensitizing Agents/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Singlet Oxygen/pharmacokinetics , Sulfides/chemistry , Zinc Compounds
15.
ACS Appl Mater Interfaces ; 12(50): 55584-55595, 2020 Dec 16.
Article in English | MEDLINE | ID: mdl-33259182

ABSTRACT

Magnetic nanostructures (MNS) have a wide range of biological applications due to their biocompatibility, superparamagnetic properties, and customizable composition that includes iron oxide (Fe3O4), Zn2+, and Mn2+. However, several challenges to the biomedical usage of MNS must still be addressed, such as formulation stability, inability to encapsulate therapeutic payloads, and variable clearance rates in vivo. Here, we enhance the utility of MNS during controlled delivery applications via encapsulation within polymeric bicontinuous nanospheres (BCNs) composed of poly(ethylene glycol)-block-poly(propylene sulfide) (PEG-b-PPS) copolymers. PEG-b-PPS BCNs have demonstrated versatile encapsulation and delivery capabilities for both hydrophilic and hydrophobic payloads due to their unique and highly organized cubic phase nanoarchitecture. MNS-embedded BCNs (MBCNs) were thus coloaded with physicochemically diverse molecular payloads using the technique of flash nanoprecipitation and characterized in terms of their structure and in vivo biodistribution following intravenous administration. Retention of the internal aqueous channels and cubic architecture of MBCNs were verified using cryogenic transmission electron microscopy and small-angle X-ray scattering, respectively. MBCNs demonstrated improvement in magnetic resonance imaging (MRI) contrast enhancement (r2 relaxivity) as compared to free MNS, which in combination with scanning transmission electron microscopy and energy-dispersive X-ray spectroscopy evidenced the clustering and continued access to water of MNS following encapsulation. Furthermore, MBCNs were found to be noncytotoxic and able to deliver their hydrophilic and hydrophobic small-molecule payloads both in vitro and in vivo. Finally, the oxidation sensitivity of the hydrophobic PPS block allowed MBCNs to undergo a unique, triggerable transition in morphology into MNS-bearing micellar nanocarriers. In summary, MBCNs are an attractive platform for the delivery of molecular and nanoscale payloads for diverse on-demand and sustained drug delivery applications.


Subject(s)
Magnetite Nanoparticles/chemistry , Nanospheres/chemistry , Animals , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Carriers/toxicity , Female , Ferrosoferric Oxide/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Liver/chemistry , Liver/metabolism , MCF-7 Cells , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Nanospheres/metabolism , Nanospheres/toxicity , Oxidation-Reduction , Polyethylene Glycols/chemistry , Sulfides/chemistry , Tissue Distribution
16.
ACS Biomater Sci Eng ; 5(5): 2577-2586, 2019 May 13.
Article in English | MEDLINE | ID: mdl-33405763

ABSTRACT

Polymeric micelles with stimuli-triggered drug release and AIE active bioimaging have emerged as potential candidates for theranostics. Herein, a curcumin (Cur) loaded oxidation-responsive mPEG-b-PLG (Se)-TP polymeric micelle system with great aggregation-induced emission (AIE) active and two-photon imaging property has been developed for simultaneous antitumor treatment and bioimaging. Cur-loaded polymeric micelles with a core-shell structure and a homogeneous size of 136 nm show great physiological stability while rapidly disassemble under oxidation environment with accelerated drug release. The excellent biocompatibility and great AIE property and two-photon excitation endow these functional mPEG-b-PLG (Se)-TP micelles as bioprobes for the two-photon imaging of cells and deeper tissues. Furthermore, the biodistribution of nanocarriers and intracellular drug delivery can also be traced. Moreover, the Cur-loaded micelles also show great tumor inhibition ability and minimal side effects in vivo compared with free drug. These novel polymeric micelles are expected to be potential candidates for cancer theranostics.

17.
ACS Appl Mater Interfaces ; 11(21): 18914-18922, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31055911

ABSTRACT

Although environment-sensitive prodrug-based nanoparticles (NPs) have developed rapidly, lots of prodrug NPs still show poor selectivity and efficiency of parent drug bioactivation because of tumor heterogeneity. Herein, self-strengthened bioactivating prodrug-based NPs are fabricated via co-encapsulation of oxidation-responsive thioether-linked linoleic acid-paclitaxel conjugates (PTX-S-LA) and ß-lapachone (LPC) into polymeric micelles (PMs). Following cellular uptake, PMs first release LPC to significantly elevate the reactive oxidative species (ROS) level through NAD(P)H: quinone oxidoreductase-1 (NQO1) catalysis. Then, NQO1-generated ROS in combination with endogenous high ROS levels in tumor cells could synergistically facilitate PTX-S-LA to release the active cytotoxic agent PTX. Such a novel prodrug nanosystem exhibits self-strengthened prodrug bioactivation, ultraselective release, and cytotoxicity between cancer and normal cells, prolonged circulation time, and enhanced tumor accumulation, leading to high antitumor efficiency and superior biosafety. Our findings pave the new way for the rational design of oxidation-responsive prodrug NPs for high-efficacy cancer chemotherapy.


Subject(s)
Breast Neoplasms/drug therapy , Drug Liberation , Prodrugs/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Endocytosis , Female , Humans , Linoleic Acid/chemistry , Mice , Mice, Inbred BALB C , NAD(P)H Dehydrogenase (Quinone)/metabolism , NIH 3T3 Cells , Nanoparticles/chemistry , Optical Imaging , Oxidation-Reduction , Paclitaxel/blood , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Prodrugs/pharmacology , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Tissue Distribution/drug effects
18.
J Control Release ; 282: 90-100, 2018 07 28.
Article in English | MEDLINE | ID: mdl-29601932

ABSTRACT

Polymer vesicles, i.e. polymersomes (PS), present unique nanostructures with an interior aqueous core that can encapsulate multiple independent cargos concurrently. However, the sequential release of such co-loaded actives remains a challenge. Here, we report the rational design and synthesis of oxidation-responsive shell-crosslinked PS with capability for the controlled, sequential release of encapsulated hydrophilic molecules and hydrogels. Amphiphilic brush block copolymers poly(oligo(ethylene glycol) methyl ether methacrylate)-b-poly(oligo(propylene sulfide) methacrylate) (POEGMA-POPSMA) were prepared to fabricate PS via self-assembly in aqueous solution. As a type of unique drug delivery vehicle, the interior of the PS was co-loaded with hydrophilic molecules and water-soluble poly(N-isopropylacrylamide) (PNIPAM) conjugates. Due to the thermosensitivity of PNIPAM, PNIPAM conjugates within the PS aqueous interior underwent a phase transition to form hydrogels in situ when the temperature was raised above the lower critical solution temperature (LCST) of PNIPAM. Via control of the overall shell permeability by oxidation, we realized the sequential release of two water-soluble payloads based on the assumption that hydrogels have much smaller membrane permeability than that of molecular cargos. The ability to control the timing of release of molecular dyes and PNIPAM-based hydrogels was also observed within live cells. Furthermore, leakage of hydrogels from the PS was effectively alleviated in comparison to molecular cargos, which would facilitate intracellular accumulation and prolonged retention of hydrogels within the cell cytoplasm. Thus, we demonstrate that the integration of responsive hydrogels into PS with crosslinkable membranes provides a facile and versatile technique to control the stability and release of water-soluble cargos for drug delivery purposes.


Subject(s)
Acrylic Resins/chemistry , Delayed-Action Preparations/chemistry , Methacrylates/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Sulfides/chemistry , Cell Line , Coloring Agents/administration & dosage , Cross-Linking Reagents/chemistry , Drug Liberation , Humans , Hydrogels/chemistry , Hydrophobic and Hydrophilic Interactions , Micelles , Oxidation-Reduction , Solubility , Water/chemistry
19.
ACS Appl Mater Interfaces ; 10(9): 7798-7810, 2018 Mar 07.
Article in English | MEDLINE | ID: mdl-29424527

ABSTRACT

Reactive oxygen species (ROS)-responsive theranostic nanomedicines have attracted wide interest in recent years because ROS stress is implicated in some pathological disorders such as inflammatory diseases and cancers. In this article, we report a kind of innovative ROS-responsive theranostic polymeric nanoparticles that are able to load hydrophobic drugs and to fluorescently self-report the in vitro or intracellular drug release under ROS triggering. The fluorescent nanoparticles were formed by amphiphilic block copolymers consisting of a poly(ethylene glycol) (PEG) segment and an oxidation-responsive hydrophobic block. The copolymers with different hydrophobic block lengths were synthesized by the atom transfer radical polymerization of a phenylboronic ester-containing acrylic monomer with a small fraction of a ROS-activatable 1,8-naphthalimide-based fluorescent monomer, using PEG-Br as the macroinitiator. The copolymer nanoparticles were stable in neutral phosphate buffer but degraded upon H2O2 triggering, with the degradation rate depending on the hydrophobic block length and the concentration of H2O2. The degradation of nanoparticles was accompanied by a colorimetric change of the fluorophore from blue to green, which affords the nanoparticles the ability to detecting H2O2 by a ratiometric fluorescent approach. Moreover, the nanoparticles could encapsulate doxorubicin (DOX) and the H2O2-triggered DOX release was well associated with the change in ratiometric fluorescence. Confocal laser scanning microscope results reveal that the fluorescent nanoparticles were internalized into A549 cells through the endocytosis pathway. The ROS-stimulated degradation of the nanoparticles and intracellular DOX release and the fate of the degraded polymers could be monitored by ratiometric fluorescent imaging. Finally, the naked nanoparticles and the degradation products are cytocompatible, whereas the DOX-loaded ones exhibit concentration-dependent cytotoxicity. Of importance, the stimulation with exogenous H2O2 or lipopolysaccharide enhanced obviously the cell-killing capability of the DOX-loaded nanoparticles because of the ROS-enhanced intracellular DOX release.


Subject(s)
Nanoparticles , Doxorubicin , Drug Carriers , Drug Delivery Systems , Hydrogen Peroxide , Micelles , Polyethylene Glycols , Reactive Oxygen Species
20.
Carbohydr Polym ; 181: 964-973, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29254061

ABSTRACT

In the present study, citrus low-methoxyl pectin was modified by conjugating cysteine via amide bonds, and the resultant polymer (CYS-PEC) was characterized. CYS-PEC conjugates with thiol contents varying from 77.8µmol/g to 296µmol/g were synthesized, and the successful conjugation was evidenced by elemental, and FT-IR analyses. The sulfur in CYS-PEC is predominately in the thiol form, with a minor fraction forming disulfide bonds (∼15%), which occur when thiol/disulfide interchange interrupts the intended thiolation. Both native and modified pectin dispersions exhibited strong pseudoplastic properties, and the frequency sweeps revealed them to be dispersions containing microgel particles. Dynamic viscoelastic analysis was used to determine the oxidation-response gelling capacities of polymer dispersions containing H2O2, especially those that are highly thiolated and have cross-linked gel properties. For oxidation-induced CYS-PEC gels, their gelation time, hardness, viscosity and elastic moduli and swelling-disintegration ratio are dependent on the thiol group content, H2O2 concentration and polymer concentration.

SELECTION OF CITATIONS
SEARCH DETAIL