Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nanoscale Adv ; 6(2): 590-605, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38235072

ABSTRACT

Three-dimensional (3D) nanomaterials with high functional properties are emerging as the most promising artificial enzymes for overcoming the significant disadvantages of natural enzymes. Anticancer therapy using 3D-enzyme mimetic materials has emerged as an essential development for catalyzing cancer cell destruction. We report for the first time a novel 3D-based enzyme mimetic material, CaMoO4/MoS2/CuS nanoflower (CMC NF), that exhibits a large specific surface area, uniform flower-like structure, excellent biocompatibility, and high porosity, making it a suitable candidate for cancer detection and therapy. Additionally, CMC NFs were conjugated with folic acid (FA) to selectively target cancer cells, resulting in FA-CMC NFs explicitly binding to overexpressed folate receptor alpha (FRα) in MDA-MB-231 cells. Based on the peroxidase activity, the FA-CMC NFs are an effective nanoprobe for the selective detection of MDA-MB-231 cells over a wide detection range (50 to 5.5 × 104 cells per mL) with a low limit of detection (LOD) value of 10 cells per mL. In addition to their cancer detection capability, the FA-CMC NFs also effectively generated ˙OH radicals in a concentration-dependent manner to treat cancer cells. Under light conditions, the FA-CMC NFs with H2O2 solution showed efficient degradation of methylene blue (MB) dye, and the solution color appeared to fade within 15 min, indicating that they generated ˙OH radicals, which can efficiently kill cancer cells. Thus, the superior functionality of FA-CMC NFs offers cost-effective, facile, and reliable cancer cell detection, providing a new treatment option for cancer treatment and diagnosis.

2.
ACS Appl Mater Interfaces ; 15(23): 27471-27485, 2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37212791

ABSTRACT

Cancer is one of the diseases with high mortality worldwide. Various methods for cancer treatment are being developed, and among them, magnetically driven microrobots capable of minimally invasive surgery and accurate targeting are in the spotlight. However, existing medical magnetically manipulated microrobots contain magnetic nanoparticles (MNPs), which can cause toxicity to normal cells after the delivery of therapeutic drugs. In addition, there is a limitation in that cancer cells become resistant to the drug by mainly delivering only one drug, thereby reducing the treatment efficiency. In this paper, to overcome these limitations, we propose a microrobot that can separate/retrieve MNPs after precise targeting of the microrobot and can sequentially deliver dual drugs (gemcitabine (GEM) and doxorubicin (DOX)). First, after the proposed microrobot targeting, MNPs attached to the microrobot surface can be separated from the microrobot using focused ultrasound (FUS) and retrieved through an external magnetic field. Second, the active release of the first conjugated drug GEM to the surface of the microrobot is possible using near-infrared (NIR), and as the microrobot slowly decomposes over time, the release of the second encapsulated DOX is possible. Therefore, it is possible to increase the cancer cell treatment efficiency with sequential dual drugs in the microrobot. We performed basic experiments on the targeting of the proposed magnetically manipulated microrobot, separation/retrieval of MNPs, and the sequential dual-drug release and validated the performances of the microrobot through in vitro experiments using the EMA/FUS/NIR integrated system. As a result, the proposed microrobot is expected to be used as one of the methods to improve cancer cell treatment efficiency by improving the limitations of existing microrobots in cancer cell treatment.


Subject(s)
Drug Delivery Systems , Magnetite Nanoparticles , Drug Delivery Systems/methods , Drug Liberation , Doxorubicin/pharmacology , Magnetic Fields
3.
J Mater Chem B ; 11(5): 1044-1056, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36606505

ABSTRACT

Combinational therapy can improve the effectiveness of cancer treatment by overcoming individual therapy shortcomings, leading to accelerated cancer cell apoptosis. Combinational cancer therapy is attained by a single nanosystem with multiple physicochemical properties providing an efficient synergistic therapy against cancer cells. Herein, we report a folate receptor-targeting dual-therapeutic (photothermal and chemotherapy) core-shell nanoparticle (CSNP) exhibiting a molybdenum disulfide core with a barium titanate shell (MoS2@BT) to improve therapeutic efficacy against triple-negative breast cancer (TNBC) MDA-MB-231 cells. A simple hydrothermal approach was used to achieve the MoS2@BT CSNPs, and their diameter was calculated to be approximately 180 ± 25 nm. In addition to improving the photothermal efficiency and stability of the MoS2@BT CSNPs, their surface was functionalized with polydopamine (PDA) and subsequently modified with folic acid (FA) to achieve enhanced tumour-targeting CSNPs, named MoS2@BT-PDA-FA (MBPF). Then, gemcitabine (Gem) was loaded into the MBPF, and its loading and releasing efficacy were calculated to be 17.5 wt% and 64.5 ± 3%, respectively. Moreover, the photothermal conversion efficiency (PCE) of MBPF was estimated to be 35.3%, and it also showed better biocompatibility, which was determined by an MTT assay. The MBPF significantly increased the ambient temperature to 56.3 °C and triggered Gem release inside the TNBC cells when exposed to a near-infrared (NIR) laser (808 nm, 1.5 W cm-2, 5 min). Notably, the MoS2@BT-based nanosystem was used as a photothermal agent and a therapeutic drug-loading container for combating TNBC cells. Benefiting from the combined therapy, MBPF reduced TNBC cell viability to 81.3% due to its efficient synergistic effects. Thus, the proposed tumour-targeting MoS2@BT CSNP exhibits high drug loading, better biocompatibility, and improved anticancer efficacy toward TNBC cells due to its dual therapeutic approach in a single system, which opens up a new approach for dual cancer therapy.


Subject(s)
Molybdenum , Triple Negative Breast Neoplasms , Humans , Molybdenum/pharmacology , Molybdenum/chemistry , Triple Negative Breast Neoplasms/drug therapy , Barium , Nanomedicine , Phototherapy
4.
Biofabrication ; 14(1)2021 11 11.
Article in English | MEDLINE | ID: mdl-34670209

ABSTRACT

Biofabrication of organ-like engineered 3D tissue through the assembly of magnetized 3D multi-cellular spheroids has been recently investigated in tissue engineering. However, the cytotoxicity of magnetic nanoparticles (MNPs) and contraction-induced structural deformation of the constructs have been major limitations. In this study, we developed a method to fabricate composite stem cell spheroids using MNP-coated fibers, alleviating MNP-mediated toxicity and controlling structural assembly under external magnetic stimuli. The MNP-coated synthetic fibers (MSFs) were prepared by coating various amounts of MNPs on the fibers via electrostatic interactions. The MSFs showed magnetic hysteresis and no cytotoxicity on 2D-cultured adipose-derived stem cells (ADSCs). The composite spheroids containing MSFs and ADSCs were rapidly formed in which the amount of impregnated MSFs modulated the spheroid size. The fusion ofin vitrocomposite spheroids was then monitored at the contacting interface; the fused spheroids with over 10µg of MSF showed minimal contraction after 7 d, retaining around 90% of total area ratio regardless of the number of cells, indicating that the presence of fibers within the composite spheroid supported its structural maintenance. The fusion of MSF spheroids was modulated by external magnetic stimulation, and the effect of magnetic force on the movement and fusion of the spheroids was investigated using COMSOL simulation. Finally, ring and lamellar structures were successfully assembled using remote-controlled MSF spheroids, showing limited deformation and high viability up to 50 d duringin vitroculture. In addition, the MSFs demonstrated no adverse effects on ADSC osteochondral differentiation. Altogether, we envision that our magnetic assembly system would be a promising method for the tissue engineering of structurally controlled organ-like constructs.


Subject(s)
Spheroids, Cellular , Tissue Engineering , Cell Differentiation , Cells, Cultured , Stem Cells , Tissue Engineering/methods
5.
ACS Appl Mater Interfaces ; 13(17): 19633-19647, 2021 May 05.
Article in English | MEDLINE | ID: mdl-33877809

ABSTRACT

Therapeutic drug delivery microrobots capable of accurate targeting using an electromagnetic actuation (EMA) system are being developed. However, these drug delivery microrobots include a large number of magnetic nanoparticles (MNPs) for accurate EMA targeting, which causes side effects, such as problems with membrane integrity and normal cell apoptosis. Here, a biocompatible and hydrolyzable PEGDA-based drug delivery helical microrobot capable of MNP retrieval is proposed in which doxorubicin (DOX), an anticancer drug, is encapsulated and MNPs are conjugated by a disulfide bond. After being accurately delivered to the lesion of cancer cells through magnetic field manipulation, the fabricated microrobot provides rapid MNP separation and retrieval from the microrobot because of the use of dithiothreitol (DTT), a reducing agent, as an environment similar to the surrounding cancer cells and near-infrared (NIR) as an external stimulus. The characteristics of the fabricated microrobot are analyzed, and fundamental tests for active electromagnetic field manipulation, separation/retrieval of MNPs from the microrobot, and its hydrolysis are discussed. The therapeutic performance of the fabricated microrobot is verified through an in vitro test using tumor cells. Consequently, by use of an integrated system of microscope, eight-coil EMA, and NIR it is shown that the proposed microrobot can be moved to the target site by electromagnetic manipulation. The MNPs conjugated to the microrobot can be separated and retrieved, and the therapeutic effect on tumor cells by the encapsulated drug can be seen.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems , Magnetics , Magnetite Nanoparticles , Robotics/instrumentation , Antibiotics, Antineoplastic/pharmacology , Biocompatible Materials , Cell Line , Cell Survival/drug effects , Doxorubicin/pharmacology , Humans , Microscopy, Fluorescence
6.
Med Eng Phys ; 85: 87-96, 2020 11.
Article in English | MEDLINE | ID: mdl-33081968

ABSTRACT

In this paper, we propose the active delivery of multi-layer drug-loaded microneedle (MN) patches using a capsule that can be driven by an external magnetic field. Firstly, the multi-layer drug-loaded MN patches consist of three delivered MN patches which are composed of a drug-loaded MN patch and polydimethylsiloxane layer. The drug-loaded MN patch is made of a 10% gelatin solution and a drug. The multi-layer MN patches are attached to a permanent magnet in a magnetically driven capsule. Under an external magnetic field generated by an electromagnetic actuation system, the capsule with the multi-layer MN patches can reach the target lesions, and each MN patch can be delivered to the target lesions for medical treatment. The active delivery of the multi-layer MN patches using the proposed magnetically driven capsule was confirmed via phantom experiments. Accordingly, the adhesion of the three separated faces of the multi-layer MN patches and the adhesion between the porcine small intestine and the MN patch were measured using a load cell. We demonstrate the feasibility of the active delivery of the multi-layer MN patches to the target lesions on a porcine small intestine. Consequently, we expect that the active delivery of the multi-layer drug-loaded MN patches using the magnetically driven capsule presented in this study can be a useful method for drug delivery to lesions at various locations in the gastrointestinal tract.


Subject(s)
Needles , Pharmaceutical Preparations , Animals , Drug Delivery Systems , Humans , Swine
7.
Adv Healthc Mater ; 9(13): e2000118, 2020 07.
Article in English | MEDLINE | ID: mdl-32431072

ABSTRACT

By virtue of minimum invasiveness and driving ability using a magnetic field, drug delivery with the aid of a microrobot has an inherent potential for targeted treatment for the eye. The use of microrobots, however, has the limitation of leaving magnetic nanoparticles (MNPs) in the eye that can cause side effects. In this study, a bilayer hydrogel microrobot capable of retrieving MNPs after drug delivery is proposed that overcomes the limitations of existing microrobots. The bilayer hydrogel microrobot is composed of an MNPs layer and a therapeutic layer. Upon applying an alternating magnetic field (AMF) at the target point, the therapeutic layer is dissolved to deliver drug particles, and then the MNPs layer can be retrieved using a magnetic field. The targeting and MNPs retrieval tests validate the drug delivery and MNPs retrieval ability of the microrobot. The ex vivo bovine vitreous and in vitro cell tests demonstrate the potential for the vitreous migration of the microrobot and the therapeutic effect against retinoblastoma Y79 cancer cells. This bilayer hydrogel sheet-type intraocular microrobot provides a new drug delivery paradigm that overcomes the limitations of microrobot by maintaining the advantages of conventional microrobots in delivering drugs to the eye and retrieving MNPs after drug delivery.


Subject(s)
Hydrogels , Magnetite Nanoparticles , Animals , Cattle , Drug Delivery Systems , Magnetic Fields , Magnetics
8.
Nanoscale ; 12(4): 2773-2786, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31957767

ABSTRACT

Until now, magnetic hyperthermia was used to remove solid tumors by targeting magnetic nanoparticles (MNPs) to tumor sites. In this study, leukemia cells in the bloodstream were directly removed by whole-body hyperthermia, using leukemia cell-specific MNPs. An epithelial cellular adhesion molecule (EpCAM) antibody was immobilized on the surface of MNPs (EpCAM-MNPs) to introduce the specificity of MNPs to leukemia cells. The viability of THP1 cells (human monocytic leukemia cells) was decreased to 40.8% of that in control samples by hyperthermia using EpCAM-MNPs. In AKR mice, an animal model of lymphoblastic leukemia, the number of leukemia cells was measured following the intravenous injection of EpCAM-MNPs and subsequent whole-body hyperthermia treatment. The result showed that the leukemia cell number was also decreased to 43.8% of that without the treatment of hyperthermia, determined by Leishman staining of leukemia cells. To support the results, simulation analysis of heat transfer from MNPs to leukemia cells was performed using COMSOL Multiphysics simulation software. The surface temperature of leukemia cells adhered to EpCAM-MNPs was predicted to be increased to 82 °C, whereas the temperature of free cells without adhered MNPs was predicted to be 38 °C. Taken together, leukemia cells were selectively removed by magnetic hyperthermia from the bloodstream, because EpCAM-modified magnetic particles were specifically attached to leukemia cell surfaces. This approach has the potential to remove metastatic cancer cells, and pathogenic bacteria and viruses floating in the bloodstream.


Subject(s)
Hyperthermia, Induced/methods , Magnetite Nanoparticles/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Animals , Antibodies, Immobilized/administration & dosage , Antibodies, Immobilized/chemistry , Cell Line , Cell Survival , Disease Models, Animal , Epithelial Cell Adhesion Molecule/immunology , Epithelial Cell Adhesion Molecule/metabolism , Humans , Immunomagnetic Separation , Magnetite Nanoparticles/chemistry , Mice , Mice, Inbred AKR , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
9.
Biomed Microdevices ; 20(4): 103, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30535774

ABSTRACT

Currently, microrobots are receiving attention because of their small size and motility, which can be applied to minimal invasive therapy. Additionally, various microrobots using hydrogel with the characteristics of biocompatibility and biodegradability are also being developed. Among them, microrobots that swell and deswell in response to temperature changes caused by external near infrared (NIR) stimuli, focused ultrasound, and an alternating magnetic field, have been receiving a great amount of interest as drug carriers for therapeutic cell delivery. In this study, we propose a spring type medical microrobot that can be manipulated by an electromagnetic actuation (EMA) system and respond to an external stimulus (NIR). Additionally, we verified its feasibility with regard to targeting and drug delivery. There exist various methods of fabricating a spring type microrobot. In this study, we adopted a simple method that entails using a perfluoroalkoxy (PFA) microtube and a syringe pump. Moreover, we also used a hydrogel mixture composed of natural alginate, N-Isopropylacrylamide (NIPAM) for temperature responsiveness, and magnetic nanoparticles (MNPs) for electromagnetic control. Then, we fabricated a spring type alginate/NIPAM hydrogel-based soft microrobot. Additionally, we encapsulated doxorubicin (DOX) for tumor therapy in the microrobot. To verify the feasibility of the proposed spring type hydrogel-based soft microrobot's targeting and drug delivery, we developed an EMA and NIR integrated system. Finally, we observed the swelling and deswelling of the soft microrobot under NIR stimulation and verified the EMA controlled targeting. Moreover, we implemented a control function to release the encapsulated anticancer drug (DOX) through the swelling and deswelling of the soft microrobot by NIR, and evaluated the feasibility of cancer cell therapy by controlling the release of the drug from the soft microrobot.


Subject(s)
Acrylamides/chemistry , Alginates/chemistry , Drug Delivery Systems/instrumentation , Electromagnetic Phenomena , Hydrogels/chemistry , Infrared Rays , Robotics/instrumentation , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Liberation , Humans , Microtechnology/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...