Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 263(Pt 2): 130251, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38368991

ABSTRACT

Compared with traditional papermaking, foam forming is a new papermaking technology that uses foam instead of water to disperse fibres, which can effectively solve the problem of poor evenness of ceramic paper, but the instability of foam itself affects the application of foam forming technology. Herein, a highly stable foaming agent for foam forming technology was prepared via physical reaction of lauryl dimethyl amine oxide (OB-2) with filamentous nanocellulose (cellulose nanofiber (CNF-C) and bacterial cellulose (BC)). Then, the quartz paper was prepared by foam forming technology. Firstly, hydrogen bond interactions between hydroxyl groups of the filamentous nanocellulose and hydrophilic moieties on OB-2 enabled the formation of a 3D nanonetwork layer on the surface of the bubble, which extended the half-life of the bubble and effectively prevented the bubble from bursting or coalescing. Then, the foam was extruded and cracked, and the filamentous nanocellulose was retained on the quartz fibres to prepare filamentous nanocellulose/quartz fibre paper by foam forming technology. The quartz paper exhibited excellent evenness and mechanical properties. In conclusion, the research of foam forming technology is of great significance to the application and development of special paper.


Subject(s)
Nanofibers , Quartz , Cellulose/chemistry , Nanofibers/chemistry , Surface-Active Agents , Water
2.
Acta Biomater ; 177: 414-430, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38360292

ABSTRACT

The limited therapeutic efficacy of checkpoint blockade immunotherapy against glioblastoma is closely related to the blood-brain barrier (BBB) and tumor immunosuppressive microenvironment, where the latter is driven primarily by tumor-associated myeloid cells (TAMCs). Targeting the C-X-C motif chemokine ligand-12/C-X-C motif chemokine receptor-4 (CXCL12/CXCR4) signaling orchestrates the recruitment of TAMCs and has emerged as a promising approach for alleviating immunosuppression. Herein, we developed an iRGD ligand-modified polymeric nanoplatform for the co-delivery of CXCR4 antagonist AMD3100 and the small-molecule immune checkpoint inhibitor BMS-1. The iRGD peptide facilitated superior BBB crossing and tumor-targeting abilities both in vitro and in vivo. In mice bearing orthotopic GL261-Luc tumor, co-administration of AMD3100 and BMS-1 significantly inhibited tumor proliferation without adverse effects. A reprogramming of immunosuppression upon CXCL12/CXCR4 signaling blockade was observed, characterized by the reduction of TAMCs and regulatory T cells, and an increased proportion of CD8+T lymphocytes. The elevation of interferon-γ secreted from activated immune cells upregulated PD-L1 expression in tumor cells, highlighting the synergistic effect of BMS-1 in counteracting the PD-1/PD-L1 pathway. Finally, our research unveiled the ability of MRI radiomics to reveal early changes in the tumor immune microenvironment following immunotherapy, offering a powerful tool for monitoring treatment responses. STATEMENT OF SIGNIFICANCE: The insufficient BBB penetration and immunosuppressive tumor microenvironment greatly diminish the efficacy of immunotherapy for glioblastoma (GBM). In this study, we prepared iRGD-modified polymeric nanoparticles, loaded with a CXCR4 antagonist (AMD3100) and a small-molecule checkpoint inhibitor of PD-L1 (BMS-1) to overcome physical barriers and reprogram the immunosuppressive microenvironment in orthotopic GBM models. In this nanoplatform, AMD3100 converted the "cold" immune microenvironment into a "hot" one, while BMS-1 synergistically counteracted PD-L1 inhibition, enhancing GBM immunotherapy. Our findings underscore the potential of dual-blockade of CXCL12/CXCR4 and PD-1/PD-L1 pathways as a complementary approach to maximize therapeutic efficacy for GBM. Moreover, our study revealed that MRI radiomics provided a clinically translatable means to assess immunotherapeutic efficacy.


Subject(s)
Benzylamines , Cyclams , Glioblastoma , Nanoparticles , Animals , Mice , B7-H1 Antigen , Glioblastoma/diagnostic imaging , Glioblastoma/drug therapy , Programmed Cell Death 1 Receptor/therapeutic use , Ligands , Radiomics , Immunotherapy , Nanoparticles/therapeutic use , Tumor Microenvironment , Cell Line, Tumor
3.
Acta Biomater ; 164: 511-521, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37004782

ABSTRACT

Photodynamic therapy (PDT), as a non-invasive and spatiotemporally controllable modality, exhibits great potential in cancer treatment. However, the efficiency of reactive oxygen species (ROS) production was restricted to the hydrophobic characteristics and aggregation-caused quenching (ACQ) of photosensitizers. Herein, we designed a ROS self-activatable nano system (denoted as PTKPa) based on poly(thioketal) conjugated with photosensitizers (PSs) pheophorbide A (Ppa) on the polymer side chains for suppressing ACQ and enhancing PDT. The process of self-activation is that ROS, which is derived from laser irradiated PTKPa, as an activating agent accelerates poly(thioketal) cleavage with the release of Ppa from PTKPa. This in turn generates abundant ROS, accelerates degradation of the remaining PTKPa and amplifies the efficacy of PDT with more tremendous ROS generated. Moreover, these abundant ROS can amplify PDT-induced oxidative stress, cause irreversible damage to tumor cells and achieve immunogenic cell death (ICD), thereby boosting the efficacy of photodynamic-immunotherapy. These findings provide new insights into ROS self-activatable strategy for enhancing cancer photodynamic- immunotherapy. STATEMENT OF SIGNIFICANCE: This work described an approach to utilize ROS-responsive self-activatable poly(thioketal) conjugated with pheophorbide A (Ppa) for suppressing aggregation-caused quenching (ACQ) and enhancing photodynamic-immunotherapy. The ROS, generated from the conjugated Ppa upon 660nm laser irradiation, as a triggering agent which initiates the release of Ppa with poly(thioketal) degradation. That in turn generates abundant ROS and facilitates degradation of the remaining PTKPa, resulting in oxidative stress to tumor cells and achieving immunogenic cell death (ICD). This work provides a promising solution to improve tumor photodynamic therapeutic effects.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photochemotherapy/methods , Reactive Oxygen Species/metabolism , Neoplasms/drug therapy , Immunotherapy , Cell Line, Tumor , Nanoparticles/chemistry
4.
Biomater Sci ; 11(11): 3840-3850, 2023 May 30.
Article in English | MEDLINE | ID: mdl-37074080

ABSTRACT

Low immune infiltration severely hinders the efficacy of cancer immunotherapy. Here, we developed a manganese-phenolic network platform (TMPD) to boost antitumor immunity via a stimulator of interferon gene (STING)-amplified activation cascade. TMPD is based on doxorubicin (DOX)-loaded PEG-PLGA nanoparticles and further coated with manganese (Mn2+)-tannic acid (TA) networks. Mechanistically, DOX-based chemotherapy and Mn2+-mediated chemodynamic therapy effectively promoted immunogenic cell death (ICD), characterized by abundant damage-associated molecular pattern (DAMP) exposure, which subsequently enhanced dendritic cells' (DCs) presentation of antigens. DOX-elicited DNA damage simultaneously caused cytoplasmic leakage of intracellular double-stranded DNA (dsDNA) as the STING signal initiator, while Mn2+ mediated significant upregulation in the expression of a STING pathway-related protein thereby amplifying the STING signal. Systemic intravenous administration of TMPD remarkably promoted DC maturation and CD8+ T cell infiltration, thus eliciting strong antitumor effects. Meanwhile, the released Mn2+ could serve as a contrast agent for tumor-specific T1-weighted magnetic resonance imaging (MRI). Moreover, TMPD combined with immune checkpoint blockade (ICB) immunotherapy significantly inhibited tumor growth and lung metastasis. Collectively, these findings indicate that TMPD has great potential in activating robust innate and adaptive immunity for MRI guided cancer chemo-/chemodynamic/immune therapy.


Subject(s)
Manganese , Neoplasms , Humans , Magnetic Resonance Imaging , Immunotherapy , Up-Regulation , Cell Line, Tumor , Tumor Microenvironment
5.
Small ; 19(2): e2205024, 2023 01.
Article in English | MEDLINE | ID: mdl-36398604

ABSTRACT

Mono-chemotherapy has significant side effects and unsatisfactory efficacy, limiting its clinical application. Therefore, a combination of multiple treatments is becoming more common in oncotherapy. Chemotherapy combined with the induction of ferroptosis is a potential new oncotherapy. Furthermore, polymeric nanoparticles (NPs) can improve the antitumor efficacy and decrease the toxicity of drugs. Herein, a polymeric NP, mPEG-b-PPLGFc@Dox, is synthesized to decrease the toxicity of doxorubicin (Dox) and enhance the efficacy of chemotherapy by combining it with the induction of ferroptosis. First, mPEG-b-PPLGFc@Dox is oxidized by endogenous H2 O2 and releases Dox, which leads to an increase of H2 O2 by breaking the redox balance. The Fe(II) group of ferrocene converts H2 O2 into ·OH, inducing subsequent ferroptosis. Furthermore, glutathione peroxidase 4, a biomarker of ferroptosis, is suppressed and the lipid peroxidation level is elevated in cells incubated with mPEG-b-PPLGFc@Dox compared to those treated with Dox alone, indicating ferroptosis induction by mPEG-b-PPLGFc@Dox. In vivo, the antitumor efficacy of mPEG-b-PPLGFc@Dox is higher than that of free Dox. Moreover, the loss of body weight in mice treated mPEG-b-PPLGFc@Dox is lower than in those treated with free Dox, indicating that mPEG-b-PPLGFc@Dox is less toxic than free Dox. In conclusion, mPEG-b-PPLGFc@Dox not only has higher antitumor efficacy but it reduces the damage to normal tissue.


Subject(s)
Ferroptosis , Nanoparticles , Mice , Animals , Metallocenes , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Polyethylene Glycols , Polymers
6.
Biomater Adv ; 135: 212745, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35929217

ABSTRACT

Metabolic reprogramming plays an important role in the development of prostate cancer (PCa). However, there are few reports on the effects of nanomaterials as vectors on cancer metabolic reprogramming. Herein, a type of nanoparticle with good biocompatibility was synthesized by modifying the double-stranded of DNA containing a sulfhydryl group on the surface of gold nanoparticles (AuNPs-dsDNA) through salt-aging conjugation methods. The resultant AuNPs-dsDNA complexes possessed low toxicity to PC3 and DU145 cells in vitro. There was also no obvious hepatorenal toxicity after intravenous injection of AuNPs-dsDNA complexes in vivo, which indicated that these nanoparticles had good biological compatibilities. We investigated their biological functions using prostate cancer cells. Seahorse assay showed that AuNPs-dsDNA complexes could increase glycolysis and glycolysis capacity both in PC3 and DU145 cells. We further detected the expression of glycolysis-related genes by qPCR assay, and found that PKM2, PDHA, and LDHA were significantly upregulated. Furthermore, untargeted metabolomics revealed that PC (18:2(9Z,12Z)/18:2(9Z,12Z)) and PC (18:0/18:2 (9Z,12Z)) levels were decreased and inosinic acid level was increased in PC3 cells. Whereas (3S,6E,10E)-1,6,10,14-Phytatetraen-3-ol, Plasmenyl-PE 36:5 and Cer (d18:2/18:2) were decreased, PE 21:3 and 1-pyrrolidinecarboxaldehyde were increased in DU145 cells after co-culturing with AuNPs-dsDNA. In summary, we found that AuNPs and AuNPs-dsDNA complexes possibly regulate the metabolic reprogramming of cancer cells mainly through the lipid metabolic pathways, which could compensate for the previously mentioned phenomenon of enhanced glycolysis and glycolysis capacity. This will provide an important theoretical basis for our future research on the characteristic targeted design of nanomaterials for cancer metabolism.


Subject(s)
Metal Nanoparticles , Prostatic Neoplasms , DNA/analysis , Gold/metabolism , Humans , Male , Metal Nanoparticles/toxicity , Prostate/chemistry , Prostatic Neoplasms/genetics
7.
Biomater Adv ; 134: 112711, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35581069

ABSTRACT

The pH-responsive polymeric micelles (PMs) have been widely used as smart nano drug delivery systems to treat tumors. However, synchronously manipulating these PMs' self-assembly properties, drug release dynamics and tracing their pH-dependent intracellular fate remain challenges. Herein, we have first synthesized hyaluronic acid (HA) based glycopolypeptides modified by tetraphenylethylene (TPE) and a pH-sensitive doxorubicin (DOX) prodrug through Diels-Alder reaction, respectively. Then, the pH-responsive nanocomplexes (NCs) were prepared by coassembling the two obtained glycopolypeptides with different formulations. Controllable size within the range of 60-125 nm and morphologies like spherical, vesicular and oblate micelles can be easily accomplished by using this method; High drug encapsulating and loading efficiency can be easily realized and adjusted within a range of 86-97% and 7-25%, respectively; Acid sensitive drug release dynamics of these NCs are also tunable by using this way. Additionally, the programmed drug release induced by subtle pH variations can be extracellularly self-indicated by detecting the blue AIE changes of the TPE units through fluorescence resonance energy transfer (FRET) effect between DOX and TPE. More importantly, the dynamic pH-triggered DOX release can be easily traced inside the tumor cells by visualizing blue emission changes of the TPE through the FRET effect. In addition, both the size and the shape can affect the endocytic routes of the NCs; The HA coated NCs targeting the tumor cells can effectively inhibit the proliferation of the HeLa cells. This work can provide a new route to acquire the stimuli-responsive self-indicating PMs with the ability to adjust their self-assembly properties and their pH-triggered drug release dynamics, and even to simultaneously visualize the PMs' intracellular fate in a real-time.


Subject(s)
Drug Delivery Systems , Micelles , Doxorubicin/chemistry , Drug Delivery Systems/methods , Drug Liberation , HeLa Cells , Humans , Hyaluronic Acid/chemistry , Hydrogen-Ion Concentration , Polymers/chemistry
8.
Int J Biol Macromol ; 181: 1243-1253, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34022305

ABSTRACT

To increase the drug loading and prolong the drug release time, novel hollow organic/inorganic hybrid nanoparticles based on dextran-b-poly(L-glutamate-graft-3-mercaptopropyltrimethoxysilane) (Dex-b-P(ALG-g-MTPMS)) were prepared. First, a polysaccharide block polypeptide diblock copolymer, dextran-block-poly(γ-allyl-L-glutamate) (Dex-b-PALG) bearing allyl side-groups, has been synthesized by the combination of ring-opening polymerization and alkyne-azide [2 + 3] Huisgen's cycloaddition. Next, the allyl side-groups residing in the poly(γ-allyl-L-glutamate) block were further functionalized with 3-mercaptopropyltrimethoxysilane(MPTMS) by radical "thiol-ene" addition reactions. Finally, after a sol-gel process of the obtained copolymers, the novel organic/inorganic hybrid nanoparticles were prepared. The molecular structures, physicochemical, and self-assembly of these copolymers were characterized through FTIR, 1H NMR, dynamic light scattering (DLS), transmission electron microscopy (TEM), and scanning electron microscopy (SEM). The cross-linked hybrid nanoparticles have a higher drug loading ability and slower release rate as compared to the uncross-linked counterparts. The MTT evaluation demonstrated that the organic/inorganic hybrid nanoparticles with good biocompatibility.


Subject(s)
Dextrans/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Peptides/chemistry , Humans , Microscopy, Electron, Transmission , Organosilicon Compounds/chemistry , Polymers/chemistry , Sulfhydryl Compounds/chemistry
9.
ACS Appl Mater Interfaces ; 13(12): 14004-14014, 2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33728894

ABSTRACT

Developing novel activatable photosensitizers with excellent plasma membrane targeting ability is urgently needed for smart photodynamic therapy (PDT). Herein, a tumor acidity-activatable photosensitizer combined with a two-step bioorthogonal pretargeting strategy to anchor photosensitizers on the plasma membrane for effective PDT is developed. Briefly, artificial receptors are first anchored on the cell plasma membrane using cell-labeling agents (Az-NPs) via the enhanced permeability and retention effect to achieve the tumor cell labeling. Then, pH-sensitive nanoparticles (S-NPs) modified with dibenzocyclooctyne (DBCO) and chlorin e6 (Ce6) accumulate in tumor tissue and disassemble upon protonation of their tertiary amines in response to the acidic tumor environment, exposing the contained DBCO and Ce6. The selective, highly specific click reactions between DBCO and azide groups enable Ce6 to be anchored on the tumor cell surface. Upon laser irradiation, the cell membrane is severely damaged by the cytotoxic reactive oxygen species, resulting in remarkable cellular apoptosis. Taken together, the membrane-localized PDT by our bioorthogonal pretargeting strategy to anchor activatable photosensitizers on the plasma membrane provides a simple but effective method for enhancing the therapeutic efficacy of photosensitizers in anticancer therapy.


Subject(s)
Cell Membrane/metabolism , Cyclooctanes/administration & dosage , Drug Delivery Systems , Neoplasms/drug therapy , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Animals , Cell Line, Tumor , Chlorophyllides , Cyclooctanes/pharmacokinetics , Cyclooctanes/therapeutic use , Humans , Mice , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Neoplasms/metabolism , Photochemotherapy , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use , Porphyrins/pharmacokinetics , Porphyrins/therapeutic use , Receptors, Artificial/metabolism
10.
Pharmaceutics ; 14(1)2021 Dec 27.
Article in English | MEDLINE | ID: mdl-35056948

ABSTRACT

ß-cyclodextrin(ßCD)-based star polymers have attracted much interest because of their unique structures and potential biomedical and biological applications. Herein, a well-defined folic acid (FA)-conjugated and disulfide bond-linked star polymer ((FA-Dex-SS)-ßCD-(PCL)14) was synthesized via a couple reaction between ßCD-based 14 arms poly(ε-caprolactone) (ßCD-(PCL)14) and disulfide-containing α-alkyne dextran (alkyne-SS-Dex), and acted as theranostic nanoparticles for tumor-targeted MRI and chemotherapy. Theranostic nanoparticles were obtained by loading doxorubicin (DOX), and superparamagnetic iron oxide (SPIO) particles were loaded into the star polymer nanoparticles to obtain ((FA-Dex-SS)-ßCD-(PCL)14@DOX-SPIO) theranostic nanoparticles. In vitro drug release studies showed that approximately 100% of the DOX was released from disulfide bond-linked theranostic nanoparticles within 24 h under a reducing environment in the presence of 10.0 mM GSH. DOX and SPIO could be delivered into HepG2 cells efficiently, owing to the folate receptor-mediated endocytosis process of the nanoparticles and glutathione (GSH), which triggered disulfide-bonds cleaving. Moreover, (FA-Dex-SS)-ßCD-(PCL)14@DOX-SPIO showed strong MRI contrast enhancement properties. In conclusion, folic acid-decorated reduction-sensitive star polymeric nanoparticles are a potential theranostic nanoparticle candidate for tumor-targeted MRI and chemotherapy.

11.
Int J Nanomedicine ; 15: 3023-3038, 2020.
Article in English | MEDLINE | ID: mdl-32431499

ABSTRACT

INTRODUCTION: Advanced tumor-targeted theranostic nanoparticles play a key role in tumor diagnosis and treatment research. In this study, we developed a multifunctional theranostic platform based on an amphiphilic hyaluronan/poly-(N-ε-carbobenzyloxy-L-lysine) derivative (HA-g-PZLL), superparamagnetic iron oxide (SPIO) and aggregation-induced emission (AIE) nanoparticles for tumor-targeted magnetic resonance (MR) and fluorescence (FL) dual-modal image-guided photodynamic therapy (PDT). MATERIALS AND METHODS: The amphiphilic hyaluronan acid (HA) derivative HA-g-PZLL was synthesized by grafting hydrophobic poly-(N-ε-carbobenzyloxy-L-lysine) (PZLL) blocks onto hyaluronic acid by a click conjugation reaction. The obtained HA-g-PZLLs self-assembled into nanoparticles in the presence of AIE molecules and SPIO nanoparticles to produce tumor-targeted theranostic nanoparticles (SPIO/AIE@HA-g-PZLLs) with MR/FL dual-modal imaging ability. Cellular uptake of the theranostic nanoparticles was traced by confocal laser scanning microscopy (CLSM), flow cytometry and Prussian blue staining. The intracellular reactive oxygen species (ROS) generation characteristics of the theranostic nanoparticles were evaluated with CLSM and flow cytometry. The effect of PDT was evaluated by cytotoxicity assay. The dual-mode imaging ability of the nanoparticles was evaluated by a real-time near-infrared fluorescence imaging system and magnetic resonance imaging scanning. RESULTS: The resulting theranostic nanoparticles not only emit red fluorescence for high-quality intracellular tracing but also effectively produce singlet oxygen for photodynamic tumor therapy. In vitro cytotoxicity experiments showed that these theranostic nanoparticles can be efficiently taken up and are mainly present in the cytoplasm of HepG2 cells. After internalization, these theranostic nanoparticles showed serious cytotoxicity to the growth of HepG2 cells after white light irradiation. DISCUSSION: This work provides a simple method for the preparation of theranostic nanoparticles with AIE characteristics and MR contrast enhancement, and serves as a dual-modal imaging platform for image-guided tumor PDT.


Subject(s)
Contrast Media/chemistry , Magnetic Resonance Imaging , Nanoparticles/chemistry , Optical Imaging , Photochemotherapy , Theranostic Nanomedicine , Animals , Cell Line, Tumor , Female , Fluorescence , Hep G2 Cells , Humans , Hyaluronic Acid/chemical synthesis , Hyaluronic Acid/chemistry , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , Polylysine/chemical synthesis , Polylysine/chemistry , Proton Magnetic Resonance Spectroscopy , Reactive Oxygen Species/metabolism , Spectroscopy, Fourier Transform Infrared
12.
Int J Biol Macromol ; 148: 483-492, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-31926232

ABSTRACT

Redox-responsive theranostic nanoparticles based on poly-(N-ε-carbobenzyloxy-l-lysine) (PZLL) grafted hyaluronan (HA) (HA-g-SS-PZLL) copolymers were constructed for hepatocellular carcinoma diagnosis and therapy. These hyaluronan derivatives formed nanoparticles via a self-assembly process in aqueous solution at low concentration. Theranostic nanoparticles were obtained after loading hydrophobic doxorubicin (DOX) and superparamagnetic iron oxide (SPIO) into the core of the nanoparticles via a dialysis method. Theranostic nanoparticles exhibited redox triggered DOX release behavior, and faster DOX released from theranostic nanoparticles was detected under a reducing environment compared with slow DOX release under a normal physiological environment. Confocal laser scanning microscopy (CLSM), flow cytometry and Prussian blue staining against HepG2 cells demonstrated that HA-g-SS-PZLL theranostic nanoparticles were capable of delivering DOX and SPIO into the cells. The analysis of the anticancer effect revealed that the HA-g-SS-PZLL theranostic nanoparticles shown higher cytotoxicity against HepG2 cells than DOX-loaded HA-g-PZLL nanoparticles. In vitro T2 magnetic resonance imaging (MRI) results exhibited that theranostic nanoparticles showed a good contrast enhancement effect, and the r2 relaxivity value was approximately 231 Fe mM-1 s-1. Finally, the theranostic nanoparticles acted as nanoprobes for HepG2 tumor-bearing BALB/c mice for in vivo MRI. Therefore, HA-g-SS-PZLL copolymers have great potential as theranostic nanoparticles for tumor-targeted diagnosis and treatment.


Subject(s)
Biopolymers , Disulfides , Hyaluronic Acid , Nanoparticles , Oxidation-Reduction , Polylysine , Theranostic Nanomedicine , Antineoplastic Agents/administration & dosage , Biopolymers/chemistry , Cell Survival , Disulfides/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , Hep G2 Cells , Humans , Hyaluronic Acid/chemical synthesis , Hyaluronic Acid/chemistry , Magnetic Resonance Imaging/methods , Nanoparticles/chemistry , Polylysine/chemistry , Spectroscopy, Fourier Transform Infrared
13.
Mater Sci Eng C Mater Biol Appl ; 98: 9-18, 2019 May.
Article in English | MEDLINE | ID: mdl-30813097

ABSTRACT

Tumor-targeted multifunctional nanocarriers play an important role in tumor diagnosis and treatment. Herein, disulfide bonds linked amphiphilic hyaluronan-SS-poly(ε-caprolactone) diblock copolymers (HA-SS-PCL) were synthesized and studied as theranostic nanocarriers for tumor diagnosis and treatment. The chemical structure of HA-SS-PCL was confirmed by Fourier transform infrared spectroscopy (FTIR) and proton nuclear magnetic resonance (1H NMR). The self-assembling behavior of the HA-SS-PCL into GSH-responsive micelles and their degradation were characterized by fluorescence spectroscopy, dynamic light scattering (DLS) and transmission electron microscopy (TEM). Theranostic nanocarriers encapsulating doxorubicin (DOX) and superparamagnetic iron oxide (SPIO) were formed via a dialysis. In vitro drug release results suggested that the HA-SS-PCL micelles possessed reductant-triggered doxorubicin release ability, which was confirmed by 100% of DOX release from HA-SS-PCL micelles within 12 h under 10 mM of glutathione (GSH), whereas about 40% of DOX was released under non-reductive condition within 24 h. Both flow cytometry and confocal laser scanning microscopy (CLSM) analysis revealed that the HA-SS-PCL micelles loaded with DOX were internalized in HepG2 cell via a receptor mediated mechanism between hyaluronan and the CD44 receptor. Furthermore, the MTT assay and cell apoptosis analysis revealed that the DOX-loaded HA-SS-PCL micelles exhibited pronounced antitumor ability towards HepG2 cells compared with that of the reduction-insensitive HA-PCL micelles at the same DOX dosage. The r2 relaxivity value of the DOX/SPIO loaded HA-SS-PCL micelles was up to 221.2 mM-1 s-1 (Fe). Thus, the obtained HA-SS-PCL block copolymers demonstrate promising potential as tumor targeting theranostic nanocarriers in the field of tumor diagnosis and treatment.


Subject(s)
Hyaluronic Acid/chemistry , Neoplasms/diagnosis , Polyesters/chemistry , Polymers/chemistry , Theranostic Nanomedicine/methods , Animals , Apoptosis , Flow Cytometry , Hep G2 Cells , Humans , Micelles , Microscopy, Confocal , Microscopy, Electron, Transmission , Neoplasms/therapy , Spectrometry, Fluorescence
14.
Mater Sci Eng C Mater Biol Appl ; 90: 568-575, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-29853126

ABSTRACT

The nucleus is one of the most important cellular organelles, where gene encode and transcribe at that location. However, nucleus-targeting gene delivery are rare been reported. It is important to develop a high-efficiency nucleus-targeting gene vector that can deliver targeted gene into nucleus directly for destroy of cancer cells. Here, special nucleus-targeting and size changeable deliver system based on TAT-SS-PAMAM-D3 with TAT functional on the surface and disulfide linked between D2 and D3 is designed to perform highly efficient nucleus-targeting gene delivery for effective cancer cell killing in vitro. CLSM observations reveal that more TAT-SS-PAMAM-D3 are enter into the nucleus when compare to SS-PAMAM-D3. The TAT modified vector can also act as gene deliver to reach high gene transfection efficiencies, high apoptosis and low viability in HeLa cells. This TAT functionalized and disulfide linking in the carrier may become a prospective vector for cancer gene treatment and also offered a different strategy for designing a better gene delivery system.


Subject(s)
Apoptosis/physiology , Cell Nucleus/metabolism , Cell Survival/physiology , Dendrimers/chemistry , Genetic Vectors/chemistry , Apoptosis/genetics , Cell Survival/genetics , Gene Transfer Techniques , HeLa Cells , Humans , Peptide Fragments/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Colloids Surf B Biointerfaces ; 162: 326-334, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29223647

ABSTRACT

We developed a high-efficiency nucleus-targeted co-delivery vector that delivers genes and drugs directly into the nucleus of cancer cells. The system is based on grafted poly-(N-3-carbobenzyloxy-lysine) (CPCL) with transactivator of transcription (TAT)- chitosan on the surface. It is designed to perform highly efficient nucleus- targeted gene and drug co-delivery. Confocal laser scanning microscopy (CLSM) revealed that more TAT-CPCL entered the nucleus than does CPCL alone. The TAT-modified vector serves as a gene and drug co-delivery mechanism to achieve high gene transfection efficiency, high apoptosis and low viability in HeLa cells. TAT-CPCL may become a vector for cancer gene treatment and a template for designing better co-deliver systems.


Subject(s)
Cell Nucleus/drug effects , Chitosan/chemistry , Drug Carriers , Gene Products, tat/metabolism , Gene Transfer Techniques , Genetic Vectors/chemistry , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Nucleus/metabolism , Doxorubicin/pharmacology , Gene Products, tat/genetics , Genetic Vectors/metabolism , HeLa Cells , Humans , Micelles , Particle Size , Polylysine/chemistry , Surface Properties
16.
J Biomater Appl ; 31(8): 1169-1181, 2017 03.
Article in English | MEDLINE | ID: mdl-28185478

ABSTRACT

The gene and drug co-delivery system has become one of the primary strategies to overcome cancers. Here, we designed a multifunctional magnetic co-delivery system for nasopharyngeal carcinoma-targeted therapy and MR imaging. Aldehyde sodium alginate (ASA) was used to decorate the oxide iron and load cisplain through coordinate bond to form a core complex. The polymer shell poly(l-lysine)-methoxy-polyethylene glycol-folate was used to coat the core complex through electric interaction to give this nano-medicine a target ability. And this polymer could also give the nano-medicine abilities to adhere and protect DNA, and enhance its solubleness in water. After being transfected with this nano-medicine, the plasmids which contain cancer suppressor gene TFPI2 could enter and express in HNE-1 cells. It caused a higher death and apoptosis rate, inhibited nasopharyngeal carcinoma cells' migration and cloning by the synergic effect together with cisplain. Besides, clear images of this nano-medicine could be got under T2 MR imaging. This magnetic co-delivery system demonstrates a potential as a powerful multifunctional vector for drug delivery and gene vector applications in nasopharyngeal carcinoma.


Subject(s)
Carcinoma/diagnostic imaging , Carcinoma/therapy , Folic Acid/pharmacokinetics , Genetic Therapy/methods , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Nasopharyngeal Neoplasms/diagnostic imaging , Nasopharyngeal Neoplasms/therapy , Polyethylene Glycols/chemistry , Polylysine/analogs & derivatives , Carcinoma/metabolism , Cell Line, Tumor , Coated Materials, Biocompatible/chemical synthesis , Contrast Media/administration & dosage , Folic Acid/chemistry , Humans , Molecular Targeted Therapy/methods , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/metabolism , Plasmids/administration & dosage , Plasmids/chemistry , Polylysine/chemistry , Theranostic Nanomedicine/methods , Treatment Outcome
17.
Mater Sci Eng C Mater Biol Appl ; 41: 36-41, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24907734

ABSTRACT

For the development of biomimetic carriers for stimuli-sensitive delivery of anticancer drugs, a novel amphiphilic glycopolypeptide conjugate containing the disulfide bond was prepared for the first time by the ring-opening polymerization of benzyl glutamate N-carboxy anhydride in the presence of (propargyl carbamate)ethyl dithio ethylamine and then click conjugation with α-azido dextran. Its structure was characterized by Fourier-transform infrared spectroscopy and nuclear magnetic resonance analyses. Owing to its amphiphilic nature, such a conjugate could self assemble into nanosize micelles in aqueous medium, as confirmed by fluorometry, transmission electron microscopy and dynamic light scattering. For the resultant micelles, it was found to encapsulate poorly water-soluble anticancer drug (methotrexate, MTX) with the loading efficiency of 45.2%. By the in vitro drug release tests, the release rate of encapsulated MTX was observed to be accelerated significantly in the presence of 10 mM 1,4-dithio-DL-threitol (DTT), analogous to the intracellular redox potential.


Subject(s)
Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Glycopeptides/chemistry , Micelles , Antineoplastic Agents/metabolism , Click Chemistry , Dextrans/chemistry , Dithiothreitol/chemistry , Glycopeptides/metabolism , Methotrexate/chemistry , Methotrexate/metabolism , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...