Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Pharm ; 594: 120184, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33340597

ABSTRACT

D-a-tocopheryl polyethylene glycol succinate (TPGS) as a FDA-approved safe adjuvant has shown an excellent application in the targeting delivery of antitumor drugs and overcoming multidrug resistance. Beside, TPGS can result in apoptogenic activity toward many tumor types because it can induce mitochondrial dysfunction. Therefore, TPGS can serve as an antineoplastic agent. However, the current research on the selective antitumor activity of TPGS is ignored. To reveal the issue, herein we develop a mitochondria-targeting drug-free TPGS nanomicelles with the hydrodynamic diameter of about 100 nm and outstanding serum stability by weak interaction-driven self-assembly of the amphiphilic TPGS polymer. Moreover, such drug-free TPGS nanomicelles intravenously injected into tumor-bearing mice exhibit long blood circulation time, superior tumor enrichment, and inhibit the tumor growth via inducing excessive reactive oxygen species (ROS) generation within tumor cells. Further in vitro and in vivo researches jointly demonstrate that drug-free TPGS nanomicelles have more significant antitumor effect on HeLa cells compared with that of other tumor cells. On the contrary, drug-free TPGS nanomicelles display the low toxicity toward normal cells and tissues. Taken together, these new findings confirm that TPGS drug-free nanomicelles represent simple, multifunctional, safe, and efficient antineoplastic agents, which can be expected to bring new light on the development of drug-free polymers for tumor therapy.


Subject(s)
Antineoplastic Agents , Polyethylene Glycols , Animals , Antineoplastic Agents/pharmacology , Cell Death , Cell Line, Tumor , HeLa Cells , Humans , Mice , Micelles , Mitochondria , Reactive Oxygen Species , Vitamin E
2.
ACS Appl Mater Interfaces ; 12(13): 14884-14904, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32167740

ABSTRACT

Low drug payload and lack of tumor-targeting for chemodynamic therapy (CDT) result in an insufficient reactive oxygen species (ROS) generation, which seriously hinders its further clinical application. Therefore, how to improve the drug payload and tumor targeting for amplification of ROS and combine it with chemotherapy has been a huge challenge in CDT. Herein, methotrexate (MTX), gadolinium (Gd), and artesunate (ASA) were used as theranostic building blocks to be coordinately assembled into tumor-specific endogenous FeII-activated and magnetic resonance imaging (MRI)-guided self-targeting carrier-free nanoplatforms (NPs) for amplification of ROS and enhanced chemodynamic chemotherapy. The obtained ASA-MTX-GdIII NPs exhibited extremely high drug payload (∼96 wt %), excellent physiological stability, long circulating ability (half-time: ∼12 h), and outstanding tumor accumulation. Moreover, ASA-MTX-GdIII NPs could be specifically uptaken by tumor cells via folate (FA) receptors and subsequently be disassembled via lysosomal acidity-induced coordination breakage, resulting in drug burst release. Most strikingly, the produced ASA could be catalyzed by tumor-specific overexpressed endogenous FeII ions to generate sufficient ROS for enhancing the main chemodynamic efficacy, which could exert a synergistic effect with the assistant chemotherapy of MTX. Interestingly, ASA-MTX-GdIII NPs caused a lower ROS generation and toxicity on normal cell lines that seldom expressed endogenous FeII ions. Under MRI guidance with assistance of self-targeting, significantly superior synergistic tumor therapy was performed on FA receptor-overexpressed tumor-bearing mice with a higher ROS generation and an almost complete elimination of tumor. This work highlights ASA-MTX-GdIII NPs as an efficient chemodynamic-chemotherapeutic agent for MRI imaging and tumor theranostics.


Subject(s)
Antineoplastic Agents/chemistry , Ferrous Compounds/chemistry , Gadolinium/chemistry , Nanoparticles/chemistry , Theranostic Nanomedicine , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Artesunate/chemistry , Artesunate/pharmacology , Artesunate/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Imaging , Methotrexate/chemistry , Methotrexate/pharmacology , Methotrexate/therapeutic use , Mice , Mice, Nude , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/pathology , Rats , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
3.
J Mater Chem B ; 8(9): 1922-1934, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32052817

ABSTRACT

Carrier-free nanoparticles (NPs) via chemotherapeutic drug-drug conjugate assembly are a promising alternative for tumor chemotherapy. However, these NPs are still hindered via their nonspecific internalization into certain healthy cells and tissues. Herein, dual-acting methotrexate (MTX) and mannose (MAN) were conjugated via a hydrolyzable ester bond to synthesize a MTX-MAN conjugate as one molecule, which could be directly self-assembled into stimulus-responsive carrier-free NPs (MTX-MAN NPs) in aqueous solution. Such carrier-free MTX-MAN NPs with an accurate drug to sugar ratio could achieve on-demand drug release by dual stimuli of lysosomal acidity and esterase. Besides, MTX-MAN NPs could be dual-recognized by tumor cells in vitro and specifically by tumors in vivo. Moreover, the large proportion of MAN located on the NPs' surface could exert a shielding effect to avoid phagocytosis of macrophages, leading to long blood circulation. Therefore, the MTX-MAN NPs sharply reduced the drug dosage and decreased the toxicity to normal cells and tissues. Further in vitro and in vivo studies consistently confirmed that the MTX-MAN NPs exhibited superior tumor accumulation and highly synergistic chemotherapeutic effects. Furthermore, we found for the first time that MAN could enhance the antitumor activity of MTX. Considering that bi-functional MTX and MAN are approved via the FDA, and MAN is highly biosafe, the dual-self-recognizing, stimulus-responsive, and carrier-free MTX-MAN NPs might be a simple, selective, and safe chemotherapeutic strategy.


Subject(s)
Antineoplastic Agents/pharmacology , Mannose/pharmacology , Methotrexate/pharmacology , Nanoparticles/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Delivery Systems , Drug Liberation , Drug Screening Assays, Antitumor , Humans , Mannose/chemistry , Methotrexate/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Optical Imaging , Particle Size , Surface Properties
4.
ACS Appl Mater Interfaces ; 11(17): 15262-15275, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30964624

ABSTRACT

Nanoparticles camouflaged by red blood cell (RBC) membranes have attracted considerable attention owing to reservation of structure of membrane and surface proteins, endowing prominent cell-specific function including biocompatibility, prolonged circulation lifetime, and reduced reticular endothelial system (RES) uptake ability. Considering the drawbacks of carrier-free nanomedicine including the serious drug burst release, poor stability, and lack of immune escape function, herein we developed and fabricated a novel RBC membranes biomimetic combinational therapeutic system by enveloping the small molecular drug coassemblies of 10-hydroxycamptothecin (10-HCPT) and indocyanine green (ICG) in the RBC membranes for prolonged circulation, controlled drug release, and synergistic chemo-photothermal therapy (PTT). The self-reorganized RBCs@ICG-HCPT nanoparticles (NPs) exhibited a diameter of ∼150 nm with core-shell structure, high drug payload (∼92 wt %), and reduced RES uptake function. Taking advantage of the stealth functionality of RBC membranes, RBCs@ICG-HCPT NPs remarkably enhanced the accumulation at the tumor sites by passive targeting followed by cellular endocytosis. Upon the stimuli of near-infrared laser followed by acidic stimulation, RBCs@ICG-HCPT NPs showed exceptional instability by heat-mediated membrane disruption and pH change, thereby triggering the rapid disassembly and accelerated drug release. Consequently, compared with individual treatment, RBCs@ICG-HCPT NPs under dual-stimuli accomplished highly efficient apoptosis in cancer cells and remarkable ablation of tumors by chemo-PTT. This biomimetic nanoplatform based on carrier-free, small molecular drug coassemblies integrating imaging capacity as a promising theranostic system provides potential for cancer diagnosis and combinational therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Biomimetics , Camptothecin/analogs & derivatives , Cell Membrane/chemistry , Infrared Rays , Nanoparticles/chemistry , Neoplasms/therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Camptothecin/chemistry , Camptothecin/pharmacology , Camptothecin/therapeutic use , Erythrocytes/cytology , Erythrocytes/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Indocyanine Green/chemistry , Male , Mice , Mice, Nude , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Phototherapy , Rats , Rats, Sprague-Dawley
5.
Mol Pharm ; 16(5): 1982-1998, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30892898

ABSTRACT

Locating nanomedicines at the active sites plays a pivotal role in the nanoparticle-based cancer therapy field. Herein, a multifunctional nanotherapeutic is designed by using graphene oxide (GO) nanosheets with rich carboxyl groups as the supporter for hyaluronic acid (HA)-methotrexate (MTX) prodrug modification via an adipicdihydrazide cross-linker, achieving synergistic multistage tumor-targeting and combined chemo-photothermal therapy. As a tumor-targeting biomaterial, HA can increase affinity of the nanocarrier toward CD44 receptor for enhanced cellular uptake. MTX, a chemotherapeutic agent, can also serve as a tumor-targeting enhancer toward folate receptor based on its similar structure with folic acid. The prepared nanosystems possess a sheet shape with a dynamic size of approximately 200 nm and pH-responsive drug release. Unexpectedly, the physiological stability of HA-MTX prodrug-decorated GO nanosystems in PBS, serum, and even plasma is more excellent than that of HA-decorated GO nanosystems, while both of them exhibit an enhanced photothermal effect than GO nanosheets. More importantly, because of good blood compatibility as well as reduced undesired interactions with blood components, HA-MTX prodrug-decorated GO nanosystems exhibited remarkably superior accumulation at the tumor sites by passive and active targeting mechanisms, achieving highly effective synergistic chemo-photothermal therapeutic effect upon near-infrared laser irradiation, efficient ablation of tumors, and negligible systemic toxicity. Hence, the HA-MTX prodrug-decorated hybrid nanosystems have a promising potential for synergistic multistage tumor-targeting therapy.


Subject(s)
Drug Delivery Systems/methods , Graphite/chemistry , Nanoconjugates/chemistry , Photochemotherapy/methods , Uterine Cervical Neoplasms/therapy , Adipates/chemistry , Adipates/metabolism , Animals , Cell Survival/drug effects , Drug Liberation , Drug Stability , Drug Synergism , Female , Graphite/metabolism , HeLa Cells , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , MCF-7 Cells , Methotrexate/chemistry , Methotrexate/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , NIH 3T3 Cells , Prodrugs/chemistry , Prodrugs/metabolism , Tissue Distribution , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
ACS Biomater Sci Eng ; 5(9): 4442-4454, 2019 Sep 09.
Article in English | MEDLINE | ID: mdl-33438410

ABSTRACT

The medical application of nanotechnology is promising for cancer chemotherapy. However, most of the small-molecule drug assemblies still have such disadvantages as serious drug leakage and nonideal synergistic mechanisms, resulting in undesired therapeutic effect. Both nucleoside analogue-based clofarabine (CA) and methotrexate (MTX) were used as the first-line anticancer medication. However, a single-agent chemotherapy still faced many challenges including low bioavailability and toxic side effects to normal tissues due to nonspecific biodistribution of drugs. Herein, we designed and fabricated novel viral-mimicking and carry-free nanodrugs (CA-MTX NPs) via molecular recognition-driven precise self-assembly between CA and MTX. After introduction of mild acid-responsive PEG-lipid on the surface of CA-MTX NPs, the synthetic nanodrugs with a diameter of ∼150 nm exhibited tumor microenvironment-activated characteristics and self-targeting property. The results suggested that our nanodrugs could achieve superior tumor accumulation and synergistically promote the tumor suppression by collaboratively inhibiting dNTP pools. We foresaw that the well-designed smart nanodrugs delivery system would open a new avenue in synergistic cancer therapeutics.

7.
Int J Pharm ; 549(1-2): 230-238, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30071310

ABSTRACT

Poor water solubility, short half-life, and low drug efficacy posed a challenge for clinical application of curcumin (CUR). In this work, a kind of CUR prodrug was synthesized by coupling two CUR molecules with a mono-thioether linker for glutathione (GSH)-responsive drug delivery. The synthesized CUR prodrug (CUR-S-CUR dimer) could self-assemble into the homogeneous spherical nanoparticles (NPs) in aqueous solution followed by surface functionalization of trace amounts of DSPE-PEG. These CUR-S-CUR@PEG NPs exhibited a small particle size of ∼100 nm, high CUR-loading content of ∼78 wt%, and good colloid stability. Moreover, the CUR-S-CUR@PEG NPs demonstrated much more efficient cellular uptake and intracellular/nuclear drug delivery compared with free CUR, indicating the advantages of small molecular prodrug assembly. In addition, the GSH with high concentration in tumor cells could trigger the disassembly of CUR-S-CUR@PEG NPs. Furthermore, the cytotoxicity assays indicated that the CUR-S-CUR@PEG NPs exhibited the comparable inhibition effect of tumor cell proliferation with free CUR due to sustained drug release. Therefore, these stimuli-responsive CUR-S-CUR@PEG NPs might have promising potential for highly efficient intracellular drug delivery and controlled drug release in cancer therapy.


Subject(s)
Curcumin/administration & dosage , Drug Delivery Systems , Glutathione/metabolism , Nanoparticles , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Chemistry, Pharmaceutical/methods , Curcumin/pharmacokinetics , Curcumin/pharmacology , Delayed-Action Preparations , Drug Carriers/chemistry , Drug Stability , Half-Life , HeLa Cells , Humans , Particle Size , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Prodrugs , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...