Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Cell Immunol ; 386: 104691, 2023 04.
Article in English | MEDLINE | ID: mdl-36822152

ABSTRACT

COVID-19 has caused significant morbidity and mortality worldwide but also accelerated the clinical use of emerging vaccine formulations. To address the current shortcomings in the prevention and treatment of SARS-CoV-2 infection, this study developed a novel vaccine platform that closely mimics dendritic cells (DCs) in antigen presentation and T-cell stimulation in a cell-free and tunable manner. Genetically engineered DCs that express the SARS-CoV-2 spike protein (S) were chemically converted into extracellular blebs (EBs). The resulting EBs elicited potentially protective humoral immunity in vivo, indicated by the production of antibodies that potently neutralized S-pseudotyped virus, presenting EBs as a promising and safe vaccine.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , Dendritic Cells , Spike Glycoprotein, Coronavirus/genetics , Vaccination
2.
Methods ; 177: 135-145, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31734187

ABSTRACT

Extracellular vesicles (EVs) have emerged as promising biologic and comprehensive therapies for precision medicine. Despite their potential demonstrated at the benchtop, few EV formulations have made it to the clinic due to challenges in regulatory compliant scalable production; including purity, homogeneity, and reproducibility. For translation of this technology, there is a strong need for novel production methods that can meet clinical production criteria. Initial research aimed to address these challenges by taking advantage of natural pathways to increase EV yields. Such "conventional" approaches moderately increased yields but produced inhomogeneous EVs. Additionally, as there are currently no standard methods for isolation, characterization, or quantification, isolated EVs were often impure, contaminated with proteins and other biomacromolecules, and highly diverse in function. The use of shear stress and extrusion methods for EV-like vesicle production has also been investigated. While these processes can produce large EV-like vesicle yields nearly immediately, the harsh processes still result in inhomogeneous loading, and still suffer from poor purity. Chemically-induced membrane blebbing is a promising alternative production method that has the potential to overcome the previously insurmountable barriers of these current methods. This technique produces pure, and well defined EV-like vesicles, termed extracellular blebs (EBs), in clinically relevant scales over the course of minutes to hours. Furthermore, blebbing agents act on the cell in a way which locks the current surface properties and contents, preventing change, allowing for homogeneous EB production, and further preventing post-production changes. EBs may provide a promising pathway for clinical translation of EV technology.


Subject(s)
Cell Membrane/drug effects , Dithiothreitol/pharmacology , Drug Delivery Systems/methods , Ethylmaleimide/pharmacology , Extracellular Vesicles/metabolism , Formaldehyde/pharmacology , Polymers/pharmacology , Bioengineering/methods , Cell Membrane/chemistry , Cell Membrane/metabolism , Centrifugation, Density Gradient , Drug Compounding/methods , Extracellular Vesicles/chemistry , Extracellular Vesicles/transplantation , Humans , Hydrogen-Ion Concentration , Precision Medicine/methods , Translational Research, Biomedical/trends
3.
Methods ; 177: 80-94, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31626895

ABSTRACT

Cells secrete extracellular vesicles (EVs) to external environments to achieve cellular homeostasis and cell-to-cell communication. Their therapeutic potential has been constantly spotlighted since they mirror both cytoplasmic and membranous components of parental cells. Meanwhile, growing evidence suggests that EV engineering could further promote EVs with a maximized capacity. In this review, a range of engineering techniques as well as upscaling approaches to exploit EVs and their mimetics are introduced. By laying out the pros and cons of each technique from different perspectives, we sought to provide an overview potentially helpful for understanding the current state of the art EV engineering and a guideline for choosing a suitable technique for engineering EVs. Furthermore, we envision that the advances in each technique will give rise to the combinatorial engineering of EVs, taking us a step closer to a clinical translation of EV-based therapeutics.


Subject(s)
Drug Delivery Systems/methods , Extracellular Vesicles/metabolism , Neoplasms/therapy , Neurodegenerative Diseases/therapy , Translational Research, Biomedical/methods , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Biological Transport , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Cell Communication , Chemical Engineering/methods , Drug Compounding/methods , Electroporation/methods , Endocytosis , Extracellular Vesicles/chemistry , Extracellular Vesicles/transplantation , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Sonication/methods , Transfection/methods
4.
Bioconjug Chem ; 31(3): 673-684, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31986014

ABSTRACT

Targeting the inability of cancerous cells to adapt to metabolic stress is a promising alternative to conventional cancer chemotherapy. FTY720 (Gilenya), an FDA-approved drug for the treatment of multiple sclerosis, has recently been shown to inhibit cancer progression through the down-regulation of essential nutrient transport proteins, selectively starving cancer cells to death. However, the clinical use of FTY720 for cancer therapy is prohibited because of its capability of inducing immunosuppression (lymphopenia) and bradycardia when phosphorylated upon administration. A prodrug to specifically prevent phosphorylation during circulation, hence avoiding bradycardia and lymphopenia, was synthesized by capping its hydroxyl groups with polyethylene glycol (PEG) via an acid-cleavable ketal linkage. Improved aqueous solubility was also accomplished by PEGylation. The prodrug reduces to fully potent FTY720 upon cellular uptake and induces metabolic stress in cancer cells. Enhanced release of FTY720 at a mildly acidic endosomal pH and the ability to substantially down-regulate cell-surface nutrient transporter proteins in leukemia cells only by an acid-cleaved drug were confirmed. Importantly, the prodrug demonstrated nearly identical efficacy to FTY720 in an animal model of BCR-Abl-driven leukemia without inducing bradycardia or lymphopenia in vivo, highlighting its potential clinical value. The prodrug formulation of FTY720 demonstrates the utility of precisely engineering a drug to avoid undesirable effects by tackling specific molecular mechanisms as well as a financially favorable alternative to new drug development. A multitude of existing cancer therapeutics may be explored for prodrug formulation to avoid specific side effects and preserve or enhance therapeutic efficacy.


Subject(s)
Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/pharmacology , Leukemia/drug therapy , Polyethylene Glycols/chemistry , Acetals/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Leukemia/pathology , Phosphorylation
5.
Biomacromolecules ; 19(5): 1508-1516, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29562124

ABSTRACT

Despite its promises for biomedical applications, the lack of solubility in a physiological solution, the limited molecular interactions with nucleic acids due to the rigid backbone, and the inefficient intracellular release limit the use of chitosan, a natural cationic polysaccharide, for gene delivery. In this study, a flexible, aqueous-soluble aminoethoxy branch was conjugated to the primary hydroxyl group of chitosan via an acid-cleavable ketal linkage, resulting in acid-transforming chitosan (ATC) with greatly increased aqueous solubility, improved siRNA complexation, and degradability in response to an acidic pH. Acid-hydrolysis of ketal linkages, which triggers the loss of the flexible, cationic aminoethoxy branch, transforms ATC to the native form of chitosan with low water solubility, reduces molecular interaction with siRNA, and cooperatively facilitates the cytosolic release of siRNA. The siRNA complexation by ATC resulted in stable polyplexes under a neutral physiological condition, rapid cytosolic siRNA release from the mildly acidic endosome/lysosome, and substantial silencing of GFP expression in cells, notably with minimal cytotoxicity. This study demonstrates a molecularly engineered natural polymer for a biomedical application.


Subject(s)
Chitosan/analogs & derivatives , Gene Silencing , RNA, Small Interfering/genetics , Transfection/methods , Acids/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , RNA, Small Interfering/chemistry , Solubility
6.
Methods ; 106: 21-8, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27109056

ABSTRACT

It has been more than two decades since the first aptamer molecule was discovered. Since then, aptamer molecules have gain much attention in the scientific field. This increasing traction can be attributed to their many desirable traits, such as 1) their potentials to bind a wide range of molecules, 2) their malleability, and 3) their low cost of production. These traits have made aptamer molecules an ideal platform to pursue in the realm of pharmaceuticals and bio-sensors. Despite the broad applications of aptamers, tedious procedure, high resource consumption, and limited nucleobase repertoire have hindered aptamer in application usage. To address these issues, new innovative methodologies, such as automation and single round SELEX, are being developed to improve the outcomes and rates in which aptamers are discovered.


Subject(s)
Aptamers, Nucleotide/genetics , Biosensing Techniques/methods , SELEX Aptamer Technique/methods
8.
J Neurooncol ; 118(1): 29-37, 2014 May.
Article in English | MEDLINE | ID: mdl-24610460

ABSTRACT

Despite advances in surgery, chemotherapy and radiotherapy, the outcomes of patients with GBM have not significantly improved. Tumor recurrence in the resection margins occurs in more than 80% of cases indicating aggressive treatment modalities, such as gene therapy are warranted. We have examined photochemical internalization (PCI) as a method for the non-viral transfection of the cytosine deaminase (CD) suicide gene into glioma cells. The CD gene encodes an enzyme that can convert the nontoxic antifungal agent, 5-fluorocytosine, into the chemotherapeutic drug, 5-fluorouracil. Multicell tumor spheroids derived from established rat and human glioma cell lines were used as in vitro tumor models. Plasmids containing either the CD gene alone or together with the uracil phosphoribosyl transferase (UPRT) gene combined with the gene carrier protamine sulfate were employed in all experiments.PCI was performed with the photosensitizer AlPcS2a and 670 nm laser irradiance. Protamine sulfate/CD DNA polyplexes proved nontoxic but inefficient transfection agents due to endosomal entrapment. In contrast, PCI mediated CD gene transfection resulted in a significant inhibition of spheroid growth in the presence of, but not in the absence of, 5-FC. Repetitive PCI induced transfection was more efficient at low CD plasmid concentration than single treatment. The results clearly indicate that AlPcS2a-mediated PCI can be used to enhance transfection of a tumor suicide gene such as CD, in malignant glioma cells and cells transfected with both the CD and UPRT genes had a pronounced bystander effect.


Subject(s)
Antifungal Agents/pharmacology , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Flucytosine/pharmacology , Cell Line, Tumor/drug effects , Dose-Response Relationship, Drug , Glioma/pathology , Humans , Indoles/pharmacology , Organometallic Compounds/pharmacology , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Photochemical Processes/drug effects , Photosensitizing Agents/pharmacology , Transfection
9.
J Mater Chem B ; 12(27): 6577-6586, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38872501

ABSTRACT

Vaccines aim to efficiently and specifically activate the immune system via a cascade of antigen uptake, processing, and presentation by antigen-presenting cells (APCs) to CD4 and CD8 T cells, which in turn drive humoral and cellular immune responses. The specific formulation of vaccine carriers can not only shield the antigens from premature sequestering before reaching APCs but also favorably promote intracellular antigen presentation and processing. This study compares two different acid-degradable polymeric nanoparticles that are capable of encapsulating a moderately immunogenic antigen, GFP, at nearly full efficacy via electrostatic interactions or molecular affinity between His tag and Ni-NTA-conjugated monomners. This resulted in GFP-encapsulating NPs composed of ketal monomers and crosslinkers (KMX/GFP NPs) and NTA-conjugated ketal monomers and crosslinkers (NKMX/GFP NPs), respectively. Encapsulated GFP was found to be released more rapidly from NKMX/GFP NPs (electrostatic encapsulation) than from KMX/GFP NPs (affinity-driven encapsulation). In vivo vaccination studies demonstrated that while repeated injections of either NP formulation resulted in poorer generation of anti-GFP antibodies than injections of the GFP antigen itself, sequential injections of NPs and GFP as prime and booster vaccines, respectively, restored the humoral response. We proposed that NPs primarily assist APCs in antigen presentation by T cells, and B cells need to be further stimulated by free protein antigens to produce antibodies. The findings of this study suggest that the immune response can be modulated by varying the chemistry of vaccine carriers and the sequences of vaccination with free antigens and antigen-encapsulating NPs.


Subject(s)
Antigens , Nanoparticles , Polymers , Nanoparticles/chemistry , Animals , Polymers/chemistry , Mice , Antigens/immunology , Antigens/chemistry , Vaccination , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/immunology , Female , Mice, Inbred C57BL , Particle Size , Vaccines/immunology , Vaccines/chemistry , Vaccines/administration & dosage
10.
Biomater Sci ; 12(12): 3045-3067, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38712883

ABSTRACT

Various strategies at the microscale/nanoscale have been developed to improve oral absorption of therapeutics. Among them, gastrointestinal (GI)-transporter/receptor-mediated nanosized drug delivery systems (NDDSs) have drawn attention due to their many benefits, such as improved water solubility, improved chemical/physical stability, improved oral absorption, and improved targetability of their payloads. Their therapeutic potential in disease animal models (e.g., solid tumors, virus-infected lungs, metastasis, diabetes, and so on) has been investigated, and could be expanded to disease targeting after systemic/lymphatic circulation, although the detailed paths and mechanisms of endocytosis, endosomal escape, intracellular trafficking, and exocytosis through the epithelial cell lining in the GI tract are still unclear. Thus, this review summarizes and discusses potential GI transporters/receptors, their absorption and distribution, in vivo studies, and potential sequential targeting (e.g., oral absorption and disease targeting in organs/tissues).


Subject(s)
Nanoparticles , Humans , Animals , Administration, Oral , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Drug Delivery Systems , Nanoparticle Drug Delivery System/chemistry
11.
Acc Chem Res ; 45(7): 1077-88, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22103667

ABSTRACT

Silencing the expression of a target gene by RNA interference (RNAi) shows promise as a potentially revolutionizing strategy for manipulating biological (pathological) pathways at the translational level. However, the lack of reliable, efficient, versatile, and safe means for the delivery of small interfering RNA (siRNA) molecules, which are large in molecular weight, negatively charged, and subject to degradation, has impeded their use in basic research and therapy. Polyplexes of siRNA and polymers are the predominant mode of siRNA delivery, but innovative synthetic strategies are needed to further evolve them to generate the desired biological and therapeutic effects. This Account focuses on the design of polymeric vehicles for siRNA delivery based on an understanding of the molecular interactions between siRNA and cationic polymers. Ideal siRNA/polymer polyplexes should address an inherent design dilemma for successful gene silencing: (1) Cationic polymers must form tight complexes with siRNA via attractive electrostatic interactions during circulation and cellular internalization and (2) siRNA must dissociate from its cationic carrier in the cytoplasm before they are loaded into RNA-induced silencing complex (RISC) and initiate gene silencing. The physicochemical properties of polymers, which dictate their molecular affinity to siRNA, can be programmed to be altered by intracellular stimuli, such as acidic pH in the endosome and cytosolic reducers, subsequently inducing the siRNA/polymer polyplex to disassemble. Specific design goals include the reduction of the cationic density and the molecular weight, the loss of branched structure, and changes in the hydrophilicity/hydrophobicity of the polymeric siRNA carriers, via acid-responsive degradation and protonation processes within the endosome and glutathione (GSH)-mediated reduction in the cytoplasm, possibly in combination with gradual stimuli-independent hydrolysis. Acetals/ketals are acid-cleavable linkages that have been incorporated into polymeric materials for stimuli-responsive gene and drug delivery. Tailoring the ketalization ratio and the molecular weight of ketalized branched PEI (K-BPEI) offers molecular control of the intracellular trafficking of siRNA/polymer polyplexes and, therefore, the gene silencing efficiency. The ketalization of linear PEI (K-LPEI) enhances gene silencing in vitro and in vivo by improving siRNA complexation with the polymer during circulation and cellular internalization, supplementing proton buffering efficiency of the polymer in the endosome, and facilitating siRNA dissociation from the polymer in the cytoplasm, in a serum-resistant manner. Spermine polymerization via ketalization and esterification for multistep intracellular degradations provides an additional polymeric platform for improved siRNA delivery and highly biocompatible gene silencing. The chemistry presented in this Account will help lay the foundation for the development of innovative and strategic approaches that advance RNAi technology.


Subject(s)
Endosomes/metabolism , Polymers/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Cytoplasm/metabolism , Gene Transfer Techniques , Polyethyleneimine/chemistry , RNA, Small Interfering/chemistry
12.
Adv Ther (Weinh) ; 6(1)2023 Jan.
Article in English | MEDLINE | ID: mdl-36733607

ABSTRACT

Dendritic cells (DCs) are prime targets for vaccination and immunotherapy. However, limited control over antigen presentation at a desired maturation status in these plastic materials remains a fundamental challenge in efficiently orchestrating a controlled immune response. DC-derived extracellular vesicles (EVs) can overcome some of these issues, but have significant production challenges. Herein, we employ a unique chemically-induced method for production of DC-derived extracellular blebs (DC-EBs) that overcome the barriers of DC and DC-derived EV vaccines. DC-EBs are molecular snapshots of DCs in time, cell-like particles with fixed stimulatory profiles for controlled immune signalling. DC-EBs were produced an order of magnitude more quickly and efficiently than conventional EVs and displayed stable structural integrity and antigen presentation compared to live DCs. Multi-omic analysis confirmed DC-EBs are majorly pure plasma membrane vesicles that are homogeneous at the single-vesicle level, critical for safe and effective vaccination. Immature vs. mature molecular profiles on DC-EBs exhibited molecularly modulated immune responses compared to live DCs, improving remission and survival of tumor-challenged mice via generation of antigen-specific T cells. For the first time, DC-EBs make their case for use in vaccines and for their potential in modulating other immune responses, potentially in combination with other immunotherapeutics.

13.
J Control Release ; 354: 91-108, 2023 02.
Article in English | MEDLINE | ID: mdl-36572154

ABSTRACT

Cancer-targeted therapy by a chemotherapeutic agent formulated in a nanoscale platform has been challenged by complex and inefficient manufacturing, low drug loading, difficult characterization, and marginally improved therapeutic efficacy. This study investigated facile-to-produce nanocomplexes of doxorubicin (DOX), a widely used cancer drug, and clinically approved DNA fragments that are extracted from a natural source. DOX was found to self-assemble DNA fragments into relatively monodispersed nanocomplexes with a diameter of ∼70 nm at 14.3% (w/w) drug loading by simple and scalable mixing. The resulting DOX/DNA nanocomplexes showed sustained DOX release, unlike overly stable Doxil®, cellular uptake via multiple endocytosis pathways, and high hematological and immunological compatibility. DOX/DNA nanocomplexes eradicated EL4 T lymphoma cells in a time-dependent manner, eventually surpassing free DOX. Extended circulation of DOX/DNA nanocomplexes, while avoiding off-target accumulation in the lung and being cleared from the liver, resulted in rapid accumulation in tumor and lowered cardio toxicity. Finally, tumor growth of EL4-challenged C57BL/6 mice (syngeneic model) and OPM2-challenged NSG mice (human xenograft model) were efficiently inhibited by DOX/DNA nanocomplexes with enhanced overall survival, in comparison with free DOX and Doxil®, especially upon repeated administrations. DOX/DNA nanocomplexes are a promising chemotherapeutics delivery platform for their ease of manufacturing, high biocompatibility, desired drug release and accumulation, efficient tumor eradication with improved safety, and further engineering versatility for extended therapeutic applications.


Subject(s)
Doxorubicin , Neoplasms , Humans , Mice , Animals , Cell Line, Tumor , Mice, Inbred C57BL , Doxorubicin/pharmacology , Drug Delivery Systems/methods , DNA Adducts , Neoplasms/drug therapy
14.
Lasers Surg Med ; 44(9): 746-54, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23018764

ABSTRACT

BACKGROUND AND OBJECTIVE: One of many limitations for cancer gene therapy is the inability of the therapeutic gene to transfect a sufficient number of tumor cells. Photochemical internalization (PCI) is a photodynamic therapy-based approach for improving the delivery of macromolecules and genes into the cell cytosol. The utility of PCI for the delivery of the GFP reporter gene on the same plasmid as a tumor suppressor gene (PTEN) was investigated in monolayers of U251 human glioma cells and muticell U87 glioma spheroids. MATERIALS AND METHODS: U251 monolayers or U87 spheroids were incubated in AlPcS(2a) and non-viral vector polyplexes for 18 hours. In all cases, light treatment was performed with a diode laser at a wavelength of 670 nm. The non-viral transfection agents, branched polyethylenimine (bPEI), or protamine sulfate (PS), were used with the plasmid constructs GFP/PTEN or GFP. RESULTS: PS/GFP polyplexes were much less toxic to the glioma cells compared to bPEI/GFP polyplexes but were highly inefficient at gene transfection if used alone. PCI resulted in a 5- to 10-fold increase in GFP protein expression compared to controls. PCI-bPEI/PTEN or PCI-PS/PTEN transfection of either U251 monolayers or U87 spheroids significantly inhibited their growth. but had no effect on MCF-7 cells containing a wild-type PTEN gene. In addition PCI-GFP transfection of gliomas cells had no effect on their growth pattern. CONCLUSIONS: Collectively, the results suggest that AlPcS(2a) -mediated PCI can be used to enhance cell growth inhibition via transfection of tumor suppressor genes in glioma cells containing mutant PTEN genes.


Subject(s)
Genetic Therapy/methods , Glioblastoma/therapy , Lasers, Semiconductor/therapeutic use , PTEN Phosphohydrolase/genetics , Photochemotherapy , Transfection/methods , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Glioblastoma/genetics , Green Fluorescent Proteins/genetics , Humans , Indoles/therapeutic use , Organometallic Compounds/therapeutic use , Photosensitizing Agents/therapeutic use , Polyethyleneimine , Protamines
15.
ChemMedChem ; 17(7): e202100718, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35060681

ABSTRACT

Nanoparticles consisting of a condensed nucleic acid core surrounded by protective layers which aid to overcome extracellular and intracellular hurdles to gene delivery (i. e., core-shell nanoparticles, CSNPs) synthetically mimic viruses. The outer shells shield the core and are particularly designed to enable facilitated release of the gene payload into the cytoplasm, the major limiting step in intracellular gene delivery. The hypothetical proton sponge effect and degradability in response to a stimulus (i. e., mildly acidic pH in the endosome) are two prevailing, although contested, principles in designing effective carriers for intracellular gene delivery via endosomal escape. Utilizing the highly flexible chemical-tuning of the polymeric shell via surface-initiated photo-polymerization of the various monomers at different molecular ratios, the effects of proton buffering capacity, acid-degradability, and endosomal membrane-lysis property on intracellular delivery of plasmid DNA by CSNPs were investigated. This study demonstrated the equivalently critical roles of proton buffering and acid-degradability in achieving efficient intracellular gene delivery, independent of cellular uptake. Extended proton buffering resulted in further improved transfection as long as the core structure was not compromised. The results of the study present a promising synthetic strategy to the development of an efficient, chemically-tunable gene delivery carrier.


Subject(s)
Nanoparticles , Protons , Endosomes , Nanoparticles/chemistry , Polymers/chemistry , Transfection
16.
Biomater Sci ; 10(11): 2817-2830, 2022 May 31.
Article in English | MEDLINE | ID: mdl-35384946

ABSTRACT

Extracellular vesicles (EVs) have emerged as biocompatible nanocarriers for efficient delivery of various therapeutic agents, with intrinsic long-term blood circulatory capability and low immunogenicity. Here, indocyanine green (ICG)- and paclitaxel (PTX)-loaded EVs [EV(ICG/PTX)] were developed as a biocompatible nanoplatform for safe and efficient cancer treatment through near-infrared (NIR) light-triggered combination chemo/photothermal/photodynamic therapy. High dual drug encapsulation in EVs was achieved for both the hydrophilic ICG and hydrophobic PTX by simple incubation. The EVs substantially improved the photostability and cellular internalization of ICG, thereby augmenting the photothermal effects and reactive oxygen species production in breast cancer cells upon NIR light irradiation. Hence, ICG-loaded EVs activated by NIR light irradiation showed greater cytotoxic effects than free ICG. EV(ICG/PTX) showed the highest anticancer activity owing to the simultaneous chemo/photothermal/photodynamic therapy when compared with EV(ICG) and free ICG. In vivo study revealed that EV(ICG/PTX) had higher accumulation in tumors and improved pharmacokinetics compared to free ICG and PTX. In addition, a single intravenous administration of EV(ICG/PTX) exhibited a considerable inhibition of tumor proliferation with negligible systemic toxicity. Thus, this study demonstrates the potential of EV(ICG/PTX) for clinical translation of combination chemo-phototherapy.


Subject(s)
Extracellular Vesicles , Hyperthermia, Induced , Nanoparticles , Cell Line, Tumor , Indocyanine Green/chemistry , Nanoparticles/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Pharmaceutical Preparations , Phototherapy
17.
Bioconjug Chem ; 22(2): 151-5, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21280590

ABSTRACT

Conjugation of desired molecules onto retroviral surfaces through the ease of the bioorthogonal functionalization method was demonstrated. Oxidation of surface sialic acids using periodate and further p-anisidine-catalyzed conjugation with aminooxy-bearing molecules were used to directly label retroviral envelope with a fluorescent dye. The retroviral particles that were produced from a bioorthogonally functionalized virus producing cell surface and further tethered with magnetic nanoparticles were efficiently purified by simple magnetic column separation and capable of magnet-directed transduction.


Subject(s)
Retroviridae/metabolism , Tissue Engineering , Viral Proteins/biosynthesis , Amines/chemistry , Aniline Compounds/chemistry , Animals , Catalysis , Fluorescent Dyes/chemistry , Magnetics , Mice , NIH 3T3 Cells , Nanoparticles/chemistry , Oxidation-Reduction , Retroviridae/chemistry , Sialic Acids/chemistry , Surface Properties , Viral Proteins/chemistry
18.
Nano Converg ; 8(1): 34, 2021 Nov 02.
Article in English | MEDLINE | ID: mdl-34727233

ABSTRACT

Modern medicine has been waging a war on cancer for nearly a century with no tangible end in sight. Cancer treatments have significantly progressed, but the need to increase specificity and decrease systemic toxicities remains. Early diagnosis holds a key to improving prognostic outlook and patient quality of life, and diagnostic tools are on the cusp of a technological revolution. Nanotechnology has steadily expanded into the reaches of cancer chemotherapy, radiotherapy, diagnostics, and imaging, demonstrating the capacity to augment each and advance patient care. Nanomaterials provide an abundance of versatility, functionality, and applications to engineer specifically targeted cancer medicine, accurate early-detection devices, robust imaging modalities, and enhanced radiotherapy adjuvants. This review provides insights into the current clinical and pre-clinical nanotechnological applications for cancer drug therapy, diagnostics, imaging, and radiation therapy.

19.
Adv Drug Deliv Rev ; 170: 1-25, 2021 03.
Article in English | MEDLINE | ID: mdl-33359141

ABSTRACT

Due to the high prevalence and long incubation periods often without symptoms, the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has infected millions of individuals globally, causing the coronavirus disease 2019 (COVID-19) pandemic. Even with the recent approval of the anti-viral drug, remdesivir, and Emergency Use Authorization of monoclonal antibodies against S protein, bamlanivimab and casirimab/imdevimab, efficient and safe COVID-19 vaccines are still desperately demanded not only to prevent its spread but also to restore social and economic activities via generating mass immunization. Recent Emergency Use Authorization of Pfizer and BioNTech's mRNA vaccine may provide a pathway forward, but monitoring of long-term immunity is still required, and diverse candidates are still under development. As the knowledge of SARS-CoV-2 pathogenesis and interactions with the immune system continues to evolve, a variety of drug candidates are under investigation and in clinical trials. Potential vaccines and therapeutics against COVID-19 include repurposed drugs, monoclonal antibodies, antiviral and antigenic proteins, peptides, and genetically engineered viruses. This paper reviews the virology and immunology of SARS-CoV-2, alternative therapies for COVID-19 to vaccination, principles and design considerations in COVID-19 vaccine development, and the promises and roles of vaccine carriers in addressing the unique immunopathological challenges presented by the disease.


Subject(s)
Antiviral Agents/administration & dosage , COVID-19 Vaccines/administration & dosage , COVID-19/epidemiology , COVID-19/prevention & control , Drug Development/methods , SARS-CoV-2/drug effects , Animals , Antiviral Agents/immunology , COVID-19/immunology , COVID-19 Vaccines/chemical synthesis , COVID-19 Vaccines/immunology , Drug Development/trends , Humans , Immunization Programs/methods , Immunization Programs/trends , SARS-CoV-2/immunology
20.
Biomed Pharmacother ; 143: 112144, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34509823

ABSTRACT

Glioblastoma multiforme (GBM) is a grade IV malignant brain tumor with a median survival time of approximately 12-16 months. Because of its highly aggressive and heterogeneous nature it is very difficult to remove by surgical resection. Herein we have reported dual stimuli-responsive and biodegradable in situ hydrogels of oligosulfamethazine-grafted gelatin and loaded with anticancer drug paclitaxel (PTX) for preventing the progress of Glioblastoma. The oligosulfamethazine (OSM) introduced to the gelatin backbone for the formation of definite and stable in situ hydrogel. The hydrogels transformed from a sol to a gel state upon changes in stimuli. pH and temperature and retained a distinct shape after subcutaneous administration in BALB/c mice. The viscosity of the sol state hydrogels was tuned by varying the feed molar ratio between gelatin and OSM. The porosity of the hydrogels was confirmed to be lower in higher degree OSM by SEM. Sustained release of PTX from hydrogels in physiological environments (pH 7.4) was further retarded up to 63% in 9th days in tumor environments (pH 6.5). While the empty hydrogels were non-toxic in cultured cells, the hydrogels loaded with PTX showed antitumor efficacy in orthotopic-GBM xenograft mice. Collectively, the gelatin-OSM formed porous hydrogels and released the cargo in a sustained manner in tumor environments efficiently suppressing the progress of GBM. Thus, gelatin-OSM hydrogels are a potential candidate for the direct delivery of therapeutics to the local areas in brain diseases.


Subject(s)
Brain Neoplasms/drug therapy , Drug Carriers , Gelatin/chemistry , Glioblastoma/drug therapy , Paclitaxel/pharmacology , Stimuli Responsive Polymers/chemistry , Sulfamethazine/chemistry , Temperature , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Delayed-Action Preparations , Drug Compounding , Drug Liberation , Glioblastoma/pathology , Humans , Hydrogels , Hydrogen-Ion Concentration , Mice, Inbred BALB C , Mice, Nude , Neoplasm Recurrence, Local , Paclitaxel/chemistry , Porosity , Time Factors , Viscosity , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL