Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
1.
Acc Chem Res ; 56(12): 1578-1590, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37220248

ABSTRACT

Theranostic nanoparticles' potential in tumor treatment has been widely acknowledged thanks to their capability of integrating multifaceted functionalities into a single nanosystem. Theranostic nanoparticles are typically equipped with an inorganic core with exploitable physical properties for imaging and therapeutic functions, bioinert coatings for improved biocompatibility and immunological stealth, controlled drug-loading-release modules, and the ability to recognize specific cell type for uptake. Integrating multiple functionalities in a single nanosized construct require sophisticated molecular design and precise execution of assembly procedures. Underlying the multifunctionality of theranostic nanoparticles, ligand chemistry plays a decisive role in translating theoretical designs into fully functionalized theranostic nanoparticles. The ligand hierarchy in theranostic nanoparticles is usually threefold. As they serve to passivate the nanoparticle's surface, capping ligands form the first layer directly interfacing with the crystalline lattice of the inorganic core. The size and shape of nanoparticles are largely determined by the molecular property of capping ligands so that they have profound influences on the nanoparticles' surface chemistry and physical properties. Capping ligands are mostly chemically inert, which necessitates the presence of additional ligands for drug loading and tumor targeting. The second layer is commonly utilized for drug loading. Therapeutic drugs can either be covalently conjugated onto the capping layer or noncovalently loaded onto nanoparticles via drug-loading ligands. Drug-loading ligands need to be equally versatile in properties to accommodate the diversity of drugs. Biodegradable moieties are often incorporated into drug-loading ligands to enable smart drug release. With the aid of targeting ligands which usually stand the tallest on the nanoparticle surface to seek and bind to their corresponding receptors on the target, theranostic nanoparticles can preferentially accumulate at the tumor site to attain a higher precision and quantity for drug delivery. In this Account, the properties and utilities of representative capping ligands, drug-loading ligands, and targeting ligands are reviewed. Since these types of ligands are often assembled in close vicinity to each other, it is essential for them to be chemically compatible and able to function in tandem with each other. Relevant conjugation strategies and critical factors with a significant impact on ligands' performance on nanoparticles are discussed. Representative theranostic nanoparticles are presented to showcase how different types of ligands function synergistically from a single nanosystem. Finally, the technological outlook of evolving ligand chemistry on theranostic nanoparticles is provided.


Subject(s)
Nanoparticles , Neoplasms , Humans , Drug Delivery Systems , Ligands , Nanoparticles/therapeutic use , Nanoparticles/chemistry , Neoplasms/drug therapy , Pharmaceutical Preparations , Precision Medicine , Theranostic Nanomedicine/methods
2.
Adv Funct Mater ; 31(5)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-34366761

ABSTRACT

Cancer is a genetic disease originating from the accumulation of gene mutations in a cellular subpopulation. Although many therapeutic approaches have been developed to treat cancer, recent studies have revealed an irrefutable challenge that tumors evolve defenses against some therapies. Gene therapy may prove to be the ultimate panacea for cancer by correcting the fundamental genetic errors in tumors. The engineering of nanoscale inorganic carriers of cancer therapeutics has shown promising results in the efficacious and safe delivery of nucleic acids to treat oncological diseases in small-animal models. When these nanocarriers are used for co-delivery of gene therapeutics along with auxiliary treatments, the synergistic combination of therapies often leads to an amplified health benefit. In this review, an overview of the inorganic nanomaterials developed for combinatorial therapies of gene and other treatment modalities is presented. First, the main principles of using nucleic acids as therapeutics, inorganic nanocarriers for medical applications and delivery of gene/drug payloads are introduced. Next, the utility of recently developed inorganic nanomaterials in different combinations of gene therapy with each of chemo, immune, hyperthermal, and radio therapy is examined. Finally, current challenges in the clinical translation of inorganic nanomaterial-mediated therapies are presented and outlooks for the field are provided.

3.
Adv Funct Mater ; 31(6)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33708035

ABSTRACT

Temozolomide (TMZ) is the standard of care chemotherapy drug for treating glioblastomas (GBMs), the most aggressive cancer that affects people of all ages. However, its therapeutic efficacy is limited by the drug resistance mediated by a DNA repair protein, O6-methylguanine-DNA methyltransferase (MGMT), which eliminates the TMZ-induced DNA lesions. Here we report the development of an iron oxide nanoparticle (NP) system for targeted delivery of siRNAs to suppress the TMZ-resistance gene (MGMT). We show that our NP is able to overcome biological barriers, bind specifically to tumor cells, and reduce MGMT expression in tumors of mice bearing orthotopic GBM serially-passaged patient-derived xenografts. The treatment with sequential administration of this NP and TMZ resulted in increased apoptosis of GBM stem-like cells, reduced tumor growth, and significantly-prolonged survival as compared to mice treated with TMZ alone. This study introduces an approach that holds great promise to improve the outcomes of GBM patients.

4.
Mater Today (Kidlington) ; 50: 149-169, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34987308

ABSTRACT

Triple negative breast cancer is difficult to treat effectively, due to its aggressiveness, drug resistance, and lack of the receptors required for hormonal therapy, particularly at the metastatic stage. Here, we report the development and evaluation of a multifunctional nanoparticle formulation containing an iron oxide core that can deliver doxorubicin, a cytotoxic agent, and polyinosinic:polycytidylic acid (Poly IC), a TLR3 agonist, in a targeted and simultaneous fashion to both breast cancer and dendritic cells. Endoglin-binding peptide (EBP) is used to target both TNBC cells and vasculature epithelia. The nanoparticle demonstrates favorable physicochemical properties and a tumor-specific targeting profile. The nanoparticle induces tumor apoptosis through multiple mechanisms including direct tumor cell killing, dendritic cell-initiated innate and T cell-mediated adaptive immune responses. The nanoparticle markedly inhibits tumor growth and metastasis and substantially extends survival in an aggressive and drug-resistant metastatic mouse model of triple negative breast cancer (TNBC). This study points to a promising platform that may substantially improve the therapeutic efficacy for treating metastatic TNBC.

5.
Acc Chem Res ; 52(6): 1496-1506, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31135134

ABSTRACT

Certain genetic mutations lead to the development of cancer through unchecked cell growth and division. Cancer is typically treated through surgical resection, radiotherapy, and small-molecule chemotherapy. A relatively recent approach to cancer therapy involves the use of a natural process wherein small RNA molecules regulate gene expression in a pathway known as RNA interference (RNAi). RNA oligomers pair with a network of proteins to form an RNA-induced silencing complex, which inhibits the translation of mRNA into proteins, thereby controlling the expression of gene products. Synthetically produced RNA oligomers may be designed to target and silence specific oncogenes to provide cancer therapy. The primary challenges facing the use of the RNAi pathway for cancer therapy are the safe and efficacious delivery of RNA payloads and their release at pertinent sites within disease-causing cells. Nucleases are abundant in the bloodstream and intracellular environment, and therapeutic RNA sequences often require a suitable carrier to provide protection from degradation prior to reaching their site of action in the body. The use of metal core nanoparticles (NPs) serving as targeted delivery vehicles able to shield and direct RNA payloads to their intended destinations have recently gained favor. Biological barriers present in the body establish a size prerequisite for drug delivery vehicles; to overcome recognition by the body's immune system and to gain access to intracellular environments, drug carriers must be small (< 100 nm). Iron oxide and gold core NPs can be synthesized with a high degree of control to create uniform ultrasmall drug delivery vehicles capable of bypassing key biological barriers. While progress is being made in size control of liposomal and polymer NPs, such advances still lag in comparison to the exquisite tunability and time stability of size engineering achievable with metal core NPs at bulk scales. Further, unlike lipid- and viral-based transfection agents, the biodistribution of metal core NPs can be traced using noninvasive imaging techniques that capitalize on the interaction of electromagnetic radiation and the inorganic atoms at the core of the NPs. Finally, metal core NPs have been shown to match the transfection efficiency of conventional RNA-delivery vehicles while also providing less immunogenicity and minimal side effects through the addition of tumor-targeting ligands on their surface. This Account reviews recent advances in the use of iron oxide and gold NPs for RNAi therapy. An overview of the different types of RNA-based therapies is provided along with a discussion of the advantages and current limitations of the technique. We highlight design considerations for the use of iron oxide and gold NP carriers in RNAi, including a discussion of the importance of size and its role in traversing biological barriers, NP surface modifications required for targeted delivery and RNA payload release, and auxiliary properties supporting imaging functionality for treatment monitoring. Applications of NPs for combination therapies including the pairing of RNAi with chemotherapy, photothermal therapy, immunotherapy, and radiotherapy are explored through examples. Finally, future perspectives are provided with a focus on the current limitations and the potential for clinical translation of iron oxide and gold NPs in RNAi therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Carriers/chemistry , Metal Nanoparticles/chemistry , MicroRNAs/therapeutic use , Neoplasms/drug therapy , RNA, Small Interfering/therapeutic use , Animals , Ferric Compounds/chemistry , Gold/chemistry , Humans , Neoplasms/diagnostic imaging , Polymers/chemistry , RNAi Therapeutics/methods , Theranostic Nanomedicine/methods
6.
Mater Today (Kidlington) ; 38: 10-23, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33716549

ABSTRACT

Systemic delivery of hydrophobic anti-cancer drugs with nanocarriers, particularly for drug-resistant and metastatic cancer, remain a challenge because of the difficulty to achieve high drug loading, while maintaining a small hydrodynamic size and colloid stability in blood to ensure delivery of an efficacious amount of drug to tumor cells. Here we introduce a new approach to address this challenge. In this approach, nanofibers of larger size with good drug loading capacity are first constructed by a self-assembly process, and upon intravascular injection and interacting with serum proteins in vivo, these nanofibers break down into ultra-fine nanoparticles of smaller size that inherit the drug loading property from their parent nanofibers. We demonstrate the efficacy of this approach with a clinically available anti-cancer drug: paclitaxel (PTX). In vitro, the PTX-loaded nanoparticles enter cancer cells and induce cellular apoptosis. In vivo, they demonstrate prolonged circulation in blood, induce no systemic toxicity, and show high potency in inhibiting tumor growth and metastasis in both mouse models of aggressive, drug-resistant breast cancer and melanoma. This study points to a new strategy toward improved anti-cancer drug delivery and therapy.

7.
Biomed Microdevices ; 21(2): 34, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30906951

ABSTRACT

Prolonged osteochondral tissue damage can result in osteoarthritis and decreased quality of life. Multiphasic scaffolds, where different layers model different microenvironments, are a promising treatment approach, yet stable joining between layers during fabrication remains challenging. Here, a bilayer scaffold for osteochondral tissue regeneration was fabricated using thermally-induced phase separation (TIPS). Two distinct polymer solutions were layered before TIPS, and the resulting porous, bilayer scaffold was characterized by seamless interfacial integration and a mechanical stiffness gradient reflecting the native osteochondral microenvironment. Chitosan is a critical component of both scaffold layers to facilitate cell attachment and the formation of polyelectrolyte complexes with other biologically relevant natural polymers. The articular cartilage region was optimized for hyaluronic acid content and stiffness, while the subchondral bone region was defined by higher stiffness and osteoconductive hydroxyapatite content. Following co-culture with chondrocyte-like (SW-1353 or mesenchymal stem cells) and osteoblast-like cells (MG63), cell proliferation and migration to the interface along with increased gene expression associated with relevant markers of osteogenesis and chondrogenesis indicates the potential of this bilayer scaffold for osteochondral tissue regeneration.


Subject(s)
Bone and Bones/physiology , Cartilage, Articular/physiology , Chitosan/chemistry , Chitosan/pharmacology , Regeneration/drug effects , Tissue Scaffolds/chemistry , Alginates/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Bone and Bones/cytology , Bone and Bones/drug effects , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cell Adhesion/drug effects , Cell Line , Cell Proliferation/drug effects , Durapatite/chemistry , Humans , Mechanical Phenomena , Tissue Engineering
8.
Bioconjug Chem ; 28(1): 194-202, 2017 01 18.
Article in English | MEDLINE | ID: mdl-27936607

ABSTRACT

Nanoparticle-mediated delivery of chemotherapeutics has demonstrated potential in improving anticancer efficacy by increasing serum half-life and providing tissue specificity and controlled drug release to improve biodistribution of hydrophobic chemotherapeutics. However, suboptimal drug loading, particularly for solid core nanoparticles (NPs), remains a challenge that limits their clinical application. In this study we formulated a NP coated with a pH-sensitive polymer of O6-methylguanine-DNA methyltransferase (MGMT) inhibitor analog, dialdehyde modified O6-benzylguanosine (DABGS) to achieve high drug loading, and polyethylene glycol (PEG) to ameliorate water solubility and maintain NP stability. The base nanovector consists of an iron oxide core (9 nm) coated with hydrazide functionalized PEG (IOPH). DABGS and PEG-dihydrazide were polymerized on the iron oxide nanoparticle surface (IOPH-pBGS) through acid-labile hydrazone bonds utilizing a rapid, freeze-thaw catalysis approach. DABGS polymerization was confirmed by FTIR and quantitated by UV-vis spectroscopy. IOPH-pBGS demonstrated excellent drug loading of 33.4 ± 5.1% by weight while maintaining small size (36.5 ± 1.8 nm). Drug release was monitored at biologically relevant pHs and demonstrated pH dependent release with maximum release at pH 5.5 (intracellular conditions), and minimal release at physiological pH (7.4). IOPH-pBGS significantly suppressed activity of MGMT and potentiated Temozolomide (TMZ) toxicity in vitro, demonstrating potential as a new treatment option for glioblastomas (GBMs).


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Guanosine/chemistry , Hydrogen-Ion Concentration , Polymers/chemistry , Cell Line, Tumor , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Humans , Microscopy, Electron, Transmission , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Temozolomide
9.
Nanomedicine ; 13(7): 2131-2139, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28614736

ABSTRACT

Glioblastoma (GBM) remains incurable, and recurrent tumors rarely respond to standard-of-care radiation and chemo-therapies. Therefore, strategies that enhance the effects of these therapies should provide significant benefits to GBM patients. We have developed a nanoparticle delivery vehicle that can stably bind and protect nucleic acids for specific delivery into brain tumor cells. These nanoparticles can deliver therapeutic siRNAs to sensitize GBM cells to radiotherapy and improve GBM treatment via systemic administration. We show that nanoparticle-mediated knockdown of the DNA repair protein apurinic endonuclease 1 (Ape1) sensitizes GBM cells to radiotherapy and extend survival in a genetic mouse model of GBM. Specific knockdown of Ape1 activity by 30% in brain tumor tissue doubled the extended survival achieved with radiotherapy alone. Ape1 is a promising target for increasing the effectiveness of radiotherapy, and nanoparticle-mediated delivery of siRNA is a promising strategy for tumor specific knockdown of Ape1.


Subject(s)
Brain Neoplasms/radiotherapy , DNA Repair , Drug Carriers/chemistry , Glioblastoma/radiotherapy , Nanoparticles/chemistry , RNA, Small Interfering/therapeutic use , Animals , Brain/metabolism , Brain/pathology , Brain/radiation effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Disease Models, Animal , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Mice , RNA, Small Interfering/administration & dosage , RNAi Therapeutics/methods
10.
Small ; 12(46): 6388-6397, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27671114

ABSTRACT

Applications of hydrophobic drug-based nanocarriers (NCs) remain largely limited because of their low loading capacity. Here, development of a multifunctional hybrid NC made of a magnetic Fe3O4 core and a mesoporous silica shell embedded with carbon dots (CDs) and paclitaxel (PTX), and covered by another layer of silica is reported. The NC is prepared via a one-pot process under mild condition. The PTX loading method introduced in this study simplifies drug loading process and demonstrates a high loading capacity due to mesoporous silica dual-shell structure, supramolecular π-stacking between conjugated rings of PTX molecules, and aromatic rings of the CDs in the hybrid NC. The CDs serve as both confocal and two-photon fluorescence imaging probes, while the Fe3O4 core serves as a magnetic resonance imaging contrast agent. Significantly, NC releases PTX in response to near infrared irradiation as a result of local heating of the embedded CDs and the heating of CDs also provides an additional therapeutic effect by thermally killing cancer cells in tumor in addition to the chemotherapeutic effect of released PTX. Both in vitro and in vivo results show that NC demonstrates high therapeutic efficacy through a synergistic effect from the combined chemo-photothermal treatments.


Subject(s)
Drug Carriers/chemistry , Drug Delivery Systems/methods , Multimodal Imaging/methods , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Doxorubicin/chemistry , Drug Liberation , Humans , Hydrophobic and Hydrophilic Interactions , Mice
11.
Small ; 12(4): 477-87, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26641029

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide. Small interfering RNA (siRNA) holds promise as a new class of therapeutics for HCC, as it can achieve sequence-specific gene knockdown with low cytotoxicity. However, the main challenge in the clinical application of siRNA lies in the lack of effective delivery approaches that need to be highly specific and thus incur low or no systemic toxicity. Here, a nonviral nanoparticle-based gene carrier is presented that can specifically deliver siRNA to HCC. The nanovector (NP-siRNA-GPC3 Ab) is made of an iron oxide core coated with chitosan-polyethylene glycol (PEG) grafted polyethyleneimine copolymer, which is further functionalized with siRNA and conjugated with a monoclonal antibody (Ab) against human glypican-3 (GPC3) receptor highly expressed in HCC. A rat RH7777 HCC cell line that coexpresses human GPC3 and firefly luciferase (Luc) is established to evaluate the nanovector. The nanoparticle-mediated delivery of siRNA against Luc effectively suppresses Luc expression in vitro without notable cytotoxicity. Significantly, NP-siLuc-GPC3 Ab administered intravenously in an orthotopic model of HCC is able to specifically bound to tumor and induce remarkable inhibition of Luc expression. The findings demonstrate the potential of using this nanovector for targeted delivery of therapeutic siRNA to HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Ferric Compounds/chemistry , Gene Transfer Techniques , Genetic Vectors/metabolism , Liver Neoplasms/metabolism , Nanoparticles/chemistry , RNA, Small Interfering/metabolism , Animals , Antibodies, Monoclonal/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Gene Knockdown Techniques , Gene Silencing , Glypicans/metabolism , Humans , Liver Neoplasms/pathology , Luciferases/metabolism , Mice , Proton Magnetic Resonance Spectroscopy , Xenograft Model Antitumor Assays
12.
Mater Today (Kidlington) ; 19(3): 157-168, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27524934

ABSTRACT

The development of nanoparticles (NPs) for use in all facets of oncological disease detection and therapy has shown great progress over the past two decades. NPs have been tailored for use as contrast enhancement agents for imaging, drug delivery vehicles, and most recently as a therapeutic component in initiating tumor cell death in magnetic and photonic ablation therapies. Of the many possible core constituents of NPs, such as gold, silver, carbon nanotubes, fullerenes, manganese oxide, lipids, micelles, etc., iron oxide (or magnetite) based NPs have been extensively investigated due to their excellent superparamagnetic, biocompatible, and biodegradable properties. This review addresses recent applications of magnetite NPs in diagnosis, treatment, and treatment monitoring of cancer. Finally, some views will be discussed concerning the toxicity and clinical translation of iron oxide NPs and the future outlook of NP development to facilitate multiple therapies in a single formulation for cancer theranostics.

13.
Pharmacol Res ; 114: 56-66, 2016 12.
Article in English | MEDLINE | ID: mdl-27771464

ABSTRACT

Compared to conventional treatments, gene therapy offers a variety of advantages for cancer treatment including high potency and specificity, low off-target toxicity, and delivery of multiple genes that concurrently target cancer tumorigenesis, recurrence, and drug resistance. In the past decades, gene therapy has undergone remarkable progress, and is now poised to become a first line therapy for cancer. Among various gene delivery systems, nanoparticles have attracted much attention because of their desirable characteristics including low toxicity profiles, well-controlled and high gene delivery efficiency, and multi-functionalities. This review provides an overview on gene therapeutics and gene delivery technologies, and highlight recent advances, challenges and insights into the design and the utility of nanoparticles in gene therapy for cancer treatment.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Nanoparticles/chemistry , Neoplasms/genetics , Neoplasms/therapy , Animals , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy/methods , Nanomedicine/methods , Nanoparticles/administration & dosage , Nanotechnology/methods
14.
Langmuir ; 31(1): 499-505, 2015.
Article in English | MEDLINE | ID: mdl-25521606

ABSTRACT

This work describes the preparation and validation of single-chain semiconducting polymer dots (sPdots), which were generated using a method based on surface immobilization, washing, and cleavage. The sPdots have an ultrasmall size of ∼3.0 nm as determined by atomic force microscopy, a size that is consistent with the anticipated diameter calculated from the molecular weight of the single-chain semiconducting polymer. sPdots should find use in biology and medicine as a new class of fluorescent probes. The FRET assay this work presents is a simple and rapid test to ensure methods developed for preparing sPdot indeed produced single-chain Pdots as designed.


Subject(s)
Polymers/chemistry , Semiconductors , Fluorescence Resonance Energy Transfer , Microscopy, Atomic Force , Particle Size , Surface Properties
15.
Biomacromolecules ; 16(10): 3362-72, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26347946

ABSTRACT

Cationic nanoparticles (NPs) for targeted gene delivery are conventionally evaluated using 2D in vitro cultures. However, this does not translate well to corresponding in vivo studies because of the marked difference in NP behavior in the presence of the tumor microenvironment. In this study, we investigated whether prostate cancer (PCa) cells cultured in three-dimensional (3D) chitosan-alginate (CA) porous scaffolds could model cationic NP-mediated gene targeted delivery to tumors in vitro. We assessed in vitro tumor cell proliferation, formation of tumor spheroids, and expression of marker genes that promote tumor malignancy in CA scaffolds. The efficacy of NP-targeted gene delivery was evaluated in PCa cells in 2D cultures, PCa tumor spheroids grown in CA scaffolds, and PCa tumors in a mouse TRAMP-C2 flank tumor model. PCa cells cultured in CA scaffolds grew into tumor spheroids and displayed characteristics of higher malignancy as compared to those in 2D cultures. Significantly, targeted gene delivery was only observed in cells cultured in CA scaffolds, whereas cells cultured on 2D plates showed no difference in gene delivery between targeted and nontarget control NPs. In vivo NP evaluation confirmed targeted gene delivery, indicating that only CA scaffolds correctly modeled NP-mediated targeted delivery in vivo. These findings suggest that CA scaffolds serve as a better in vitro platform than 2D cultures for evaluation of NP-mediated targeted gene delivery to PCa.


Subject(s)
Alginates/chemistry , Chitosan/chemistry , Genetic Therapy , Nanoparticles , Prostatic Neoplasms/therapy , Animals , Female , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , In Vitro Techniques , Male , Mice , Porosity
16.
Macromol Rapid Commun ; 36(3): 332-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25522283

ABSTRACT

Smart hydrogels play an increasingly important role in biomedical applications, since materials that are both biocompatible and multi-stimuli-responsive are highly desirable. A simple, organic solvent-free method is presented to synthesize a biocompatible hydrogel that undergoes a sol-gel transition in response to multiple stimuli. Methoxy-poly(ethylene glycol) (mPEG) is modified into carboxylic-acid-terminated-methoxy-poly(ethylene glycol) (mPEG-acid), which is then grafted onto chitosan via amide linkages yielding mPEG-g-chitosan. Grafting of mPEG onto hydrophobic chitosan imparts hydrophilic properties to the resultant polymer. The mPEG-g-chitosan gel exhibits a controllable multi-stimuli-responsive property. The balance between hydrophilicity and hydrophobicity is believed to confer mPEG-g-chitosan with stimuli-responsive behavior. The effect of salt concentration, solute concentration, temperature, and pH on the sol-gel transition of mPEG-g-chitosan is evaluated and the underlying mechanisms of mPEG-g-chitosan polymer packing and gelation property is discussed.


Subject(s)
Chitosan/chemistry , Polyethylene Glycols/chemistry , Hydrogels/chemistry
17.
Mol Pharm ; 11(7): 2134-42, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24779767

ABSTRACT

Breast cancer is a major health problem for women worldwide. Although in vitro culture of established breast cancer cell lines is the most widely used model for preclinical assessment, it poorly represents the behavior of breast cancers in vivo. Acceleration of the development of effective therapeutic strategies requires a cost-efficient in vitro model that can more accurately resemble the in vivo tumor microenvironment. Here, we report the use of a thermoreversible poly(ethylene glycol)-g-chitosan hydrogel (PCgel) as an in vitro breast cancer model. We hypothesized that PCgel could provide a tumor microenvironment that promotes cultured cancer cells to a more malignant phenotype with drug and immune resistance. Traditional tissue culture plates and Matrigel were applied as controls in our studies. In vitro cellular proliferation and morphology, the secretion of angiogenesis-related growth factors and cytokines, and drug and immune resistance were assessed. Our results show that PCgel cultures promoted tumor aggregate formation, increased secretion of various angiogenesis- and metastasis-related growth factors and cytokines, and increased tumor cell resistance to chemotherapeutic drugs and immunotherapeutic T cells. This PCgel platform may offer a valuable strategy to bridge the gap between standard in vitro and costly animal studies for a wide variety of experimental designs.


Subject(s)
Breast Neoplasms/drug therapy , Chitosan/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Tumor Microenvironment/drug effects , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Polyethylene Glycols/administration & dosage
18.
Biomacromolecules ; 15(7): 2656-62, 2014 Jul 14.
Article in English | MEDLINE | ID: mdl-24890220

ABSTRACT

The outcome for glioblastoma patients remains dismal for its invariably recrudesces within 2 cm of the resection cavity. Local immunotherapy has the potential to eradicate the residual infiltrative component of these tumors. Here, we report the development of a biodegradable hydrogel containing therapeutic T lymphocytes for localized delivery to glioblastoma cells for brain tumor immunotherapy. Thermoreversible poly(ethylene glycol)-g-chitosan hydrogels (PCgels) were optimized for steady T lymphocyte release. Nuclear magnetic resonance spectroscopy confirmed the chemical structure of poly(ethylene glycol)-g-chitosan, and rheological studies revealed that the sol-to-gel transition of the PCgel occurred around ≥32 °C. T lymphocyte invasion through the PCgel and subsequent cytotoxicity to glioblastoma were assessed in vitro. The PCgel was shown to be cellular compatible with T lymphocytes, and the T lymphocytes retain their anti-glioblastoma activity after being encapsulated in the PCgel. T lymphocytes in the PCgel were shown to be more effective in killing glioblastoma than those in the Matrigel control. This may be attributed to the optimal pore size of the PCgel allowing better invasion of T lymphocytes. Our study suggests that this unique PCgel depot may offer a viable approach for localized immunotherapy for glioblastoma.


Subject(s)
Brain Neoplasms/therapy , Chitosan/analogs & derivatives , Chitosan/chemistry , Glioblastoma/therapy , Polyethylene Glycols/chemistry , T-Lymphocytes/physiology , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Culture Media , Cytotoxicity, Immunologic , Humans , Hydrogels/chemistry , Immunotherapy, Adoptive , Tissue Scaffolds , Transition Temperature
19.
ACS Appl Mater Interfaces ; 16(22): 28263-28275, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38788694

ABSTRACT

Intervertebral disc degeneration (IDD) is a progressive condition and stands as one of the primary causes of low back pain. Cell therapy that uses nucleus pulposus (NP)-like cells derived from human induced pluripotent stem cells (hiPSCs) holds great promise as a treatment for IDD. However, the conventional two-dimensional (2D) monolayer cultures oversimplify cell-cell interactions, leading to suboptimal differentiation efficiency and potential loss of phenotype. While three-dimensional (3D) culture systems like Matrigel improve hiPSC differentiation efficiency, they are limited by animal-derived materials for translation, poorly defined composition, short-term degradation, and high cost. In this study, we introduce a new 3D scaffold fabricated using medical-grade chitosan with a high degree of deacetylation. The scaffold features a highly interconnected porous structure, near-neutral surface charge, and exceptional degradation stability, benefiting iPSC adhesion and proliferation. This scaffold remarkably enhances the differentiation efficiency and allows uninterrupted differentiation for up to 25 days without subculturing. Notably, cells differentiated on the chitosan scaffold exhibited increased cell survival rates and upregulated gene expression associated with extracellular matrix secretion under a chemically defined condition mimicking the challenging microenvironment of intervertebral discs. These characteristics qualify the chitosan scaffold-cell construct for direct implantation, serving as both a structural support and a cellular source for enhanced stem cell therapy for IDD.


Subject(s)
Cell Differentiation , Chitosan , Induced Pluripotent Stem Cells , Nucleus Pulposus , Tissue Scaffolds , Chitosan/chemistry , Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Nucleus Pulposus/cytology , Humans , Tissue Scaffolds/chemistry , Intervertebral Disc Degeneration/therapy , Intervertebral Disc Degeneration/pathology , Cells, Cultured , Cell Survival/drug effects
20.
Nanoscale Horiz ; 9(6): 976-989, 2024 05 29.
Article in English | MEDLINE | ID: mdl-38568029

ABSTRACT

Single-atom nanozymes with well-defined atomic structures and electronic coordination environments can effectively mimic the functions of natural enzymes. However, the costly and intricate preparation processes have hindered further exploration and application of these single-atom nanozymes. In this study, we presented a synthesis technique for creating Fe-N central single-atom doped graphene quantum dot (FeN/GQDs) nanozymes using a one-step solvothermal process, where individual iron atoms form strong bonds with graphene quantum dots through nitrogen coordination. Unlike previous studies, this method significantly simplifies the synthesis conditions for single-atom nanozymes, eliminating the need for high temperatures and employing environmentally friendly precursors derived from pineapple (ananas comosus) leaves. The resulting FeN/GQDs exhibited peroxidase-like catalytic activity and kinetics comparable to that of natural enzymes, efficiently converting H2O2 into hydroxyl radical species. Leveraging their excellent peroxide-like activity, FeN/GQDs nanozymes have been successfully applied to construct a colorimetric biosensor system characterized by remarkably high sensitivity for glucose detection. This achievement demonstrated a promising approach to designing single-atom nanozymes with both facile synthesis procedures and high catalytic activity, offering potential applications in wearable sensors and personalized health monitoring.


Subject(s)
Biosensing Techniques , Glucose , Graphite , Green Chemistry Technology , Hydrogen Peroxide , Iron , Quantum Dots , Quantum Dots/chemistry , Graphite/chemistry , Iron/chemistry , Glucose/analysis , Glucose/chemistry , Biosensing Techniques/methods , Green Chemistry Technology/methods , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/analysis , Colorimetry/methods , Catalysis , Humans
SELECTION OF CITATIONS
SEARCH DETAIL