Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 403
Filter
1.
Biomed Pharmacother ; 174: 116546, 2024 May.
Article in English | MEDLINE | ID: mdl-38603885

ABSTRACT

Nanomaterials possess unusual physicochemical properties including unique optical, magnetic, electronic properties, and large surface-to-volume ratio. However, nanomaterials face some challenges when they were applied in the field of biomedicine. For example, some nanomaterials suffer from the limitations such as poor selectivity and biocompatibility, low stability, and solubility. To address the above-mentioned obstacles, functional nucleic acid has been widely served as a powerful and versatile ligand for modifying nanomaterials because of their unique characteristics, such as ease of modification, excellent biocompatibility, high stability, predictable intermolecular interaction and recognition ability. The functionally integrating functional nucleic acid with nanomaterials has produced various kinds of nanocomposites and recent advances in applications of functional nucleic acid decorated nanomaterials for cancer imaging and therapy were summarized in this review. Further, we offer an insight into the future challenges and perspectives of functional nucleic acid decorated nanomaterials.


Subject(s)
Nanostructures , Neoplasms , Nucleic Acids , Humans , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Nanostructures/chemistry , Nucleic Acids/administration & dosage , Nucleic Acids/chemistry , Animals
2.
Expert Opin Ther Pat ; 34(3): 171-186, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38578253

ABSTRACT

INTRODUCTION: Nucleic acid-based therapeutics offer groundbreaking potential for treating genetic diseases and advancing next-generation vaccines. Despite their promise, challenges in efficient delivery persist due to the properties of nucleic acids. Nanoparticles (NPs) serve as vital carriers, facilitating effective delivery to target cells, and addressing these challenges. Understanding the global landscape of patents in this field is essential for fostering innovation and guiding decision-making for researchers, the pharmaceutical industry, and regulatory agencies. AREAS COVERED: This review provides a comprehensive overview of patent compositions, applications, and manufacturing aspects concerning NPs as nucleic acid delivery systems. It delves into temporal trends, protection locations, market dynamics, and the most influential technological domains. In this work, we provide valuable insights into the advancements and potential of NP-based nucleic acid delivery systems, with a special focus on their pivotal role in advancing cutting-edge therapeutic solutions. EXPERT OPINION: Investment in NPs for nucleic acid delivery has significantly surged in recent years. However, translating these therapies into clinical practice faces obstacles, including the need for robust clinical evidence, regulatory compliance, and streamlined manufacturing processes. To address these challenges, our review article summarizes recent advances. We aim to engage researchers worldwide in the development of these promising technologies.


Subject(s)
Drug Delivery Systems , Nanoparticles , Nucleic Acids , Patents as Topic , Humans , Nucleic Acids/administration & dosage , Animals , Drug Carriers/chemistry , Genetic Therapy/methods
3.
Adv Sci (Weinh) ; 11(18): e2309748, 2024 May.
Article in English | MEDLINE | ID: mdl-38460157

ABSTRACT

Pulmonary delivery of therapeutic agents has been considered the desirable administration route for local lung disease treatment. As the latest generation of therapeutic agents, nucleic acid has been gradually developed as gene therapy for local diseases such as asthma, chronic obstructive pulmonary diseases, and lung fibrosis. The features of nucleic acid, specific physiological structure, and pathophysiological barriers of the respiratory tract have strongly affected the delivery efficiency and pulmonary bioavailability of nucleic acid, directly related to the treatment outcomes. The development of pharmaceutics and material science provides the potential for highly effective pulmonary medicine delivery. In this review, the key factors and barriers are first introduced that affect the pulmonary delivery and bioavailability of nucleic acids. The advanced inhaled materials for nucleic acid delivery are further summarized. The recent progress of platform designs for improving the pulmonary delivery efficiency of nucleic acids and their therapeutic outcomes have been systematically analyzed, with the application and the perspectives of advanced vectors for pulmonary gene delivery.


Subject(s)
Genetic Therapy , Nucleic Acids , Humans , Nucleic Acids/administration & dosage , Genetic Therapy/methods , Transfection/methods , Administration, Inhalation , Lung Diseases/therapy , Lung Diseases/genetics , Gene Transfer Techniques , Lung/metabolism , Animals
4.
Int J Biol Macromol ; 241: 124582, 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37116843

ABSTRACT

In the past few decades, substantial advancement has been made in nucleic acid (NA)-based therapies. Promising treatments include mRNA, siRNA, miRNA, and anti-sense DNA for treating various clinical disorders by modifying the expression of DNA or RNA. However, their effectiveness is limited due to their concentrated negative charge, instability, large size, and host barriers, which make widespread application difficult. The effective delivery of these medicines requires safe vectors that are efficient & selective while having non-pathogenic qualities; thus, nanomaterials have become an attractive option with promising possibilities despite some potential setbacks. Nanomaterials possess ideal characteristics, allowing them to be tuned into functional bio-entity capable of targeted delivery. In this review, current breakthroughs in the non-viral strategy of delivering NAs are discussed with the goal of overcoming challenges that would otherwise be experienced by therapeutics. It offers insight into a wide variety of existing NA-based therapeutic modalities and techniques. In addition to this, it provides a rationale for the use of non-viral vectors and a variety of nanomaterials to accomplish efficient gene therapy. Further, it discusses the potential for biomedical application of nanomaterials-based gene therapy in various conditions, such as cancer therapy, tissue engineering, neurological disorders, and infections.


Subject(s)
Genetic Therapy , Nanoparticle Drug Delivery System , Nanostructures , Nucleic Acids , Animals , Humans , Dendrimers/chemistry , Drug Stability , Genetic Therapy/methods , Hydrogels/chemistry , Liposomes/chemistry , Nanostructures/administration & dosage , Nanostructures/chemistry , Nanostructures/therapeutic use , Nucleic Acids/administration & dosage , Nucleic Acids/genetics , Nucleic Acids/metabolism , Nucleic Acids/therapeutic use , Transfection
5.
Drug Deliv ; 29(1): 386-398, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35075948

ABSTRACT

The potential of nucleic acid therapeutics to treat diseases by targeting specific cells has resulted in its increasing number of uses in clinical settings. However, the major challenge is to deliver bio-macromolecules into target cells and/or subcellular locations of interest ahead in the development of delivery systems. Although, supercharged residues replaced protein 36 + GFP can facilitate itself and cargoes delivery, its efficiency is still limited. Therefore, we combined our recent progress to further improve 36 + GFP based delivery efficiency. We found that the penetration efficacy of 36 + GFP protein was significantly improved by fusion with CPP-Dot1l or treatment with penetration enhancer dimethyl sulfoxide (DMSO) in vitro. After safely packaged with plasmid DNA, we found that the efficacy of in vitro and in vivo transfection mediated by 36 + GFP-Dot1l fusion protein is also significantly improved than 36 + GFP itself. Our findings illustrated that fusion with CPP-Dot1l or incubation with DMSO is an alternative way to synergically promote 36 + GFP mediated plasmid DNA delivery in vitro and in vivo.


Subject(s)
Cell-Penetrating Peptides/pharmacokinetics , Drug Delivery Systems/methods , Green Fluorescent Proteins/pharmacokinetics , Histone-Lysine N-Methyltransferase/pharmacokinetics , Nucleic Acids/administration & dosage , Animals , Cell Line, Tumor , Cell Survival/drug effects , Dimethyl Sulfoxide/chemistry , Green Fluorescent Proteins/chemistry , Hemolysis/drug effects , Humans , Mice , Particle Size , Surface Properties , Transfection/methods
6.
Int J Biol Macromol ; 194: 384-394, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34822829

ABSTRACT

Many challenges, such as virus infection, extreme weather and long cultivation periods, during the development of fish larvae have been observed, especially in aquaculture. Gene delivery is a useful method to express functional genes to defend against these challengers. However, the methods for fish larvae are insufficient. In our earlier report, low-molecular-weight chitosan (LMWCS) showed a strong positive charge and may be useful for polyplex formulation. Herein, we present a simple self-assembly of LMWCS polyplexes (LMWCSrNPs) for gene delivery into zebrafish larvae. Different weight ratios of LMWCS/gamma-polyglutamic acid (γ-PGA)/plasmid DNA were analyzed by gel mobility assay. Delivery efficiency determined by green fluorescent protein (GFP) expression in zebrafish liver (ZFL) cells showed that delivery efficiency at a weight ratio of 20:8:1 was higher than others. Zeta potential and transmission electron microscopy (TEM) analysis showed that the round shape of the particle size varied. In our earlier reports, IRF9S2C could induce interferon-stimulated gene (ISG) expression to induce innate immunity in zebrafish and pufferfish. Further delivery of pcDNA3-IRF9S2C-HA plasmid DNA into ZFL cells and zebrafish larvae by LMWCSrNP successfully induced ISG expression. Collectively, LMWCSrNP could be a novel gene delivery system for zebrafish larvae and might be used to improve applications in aquaculture.


Subject(s)
Chitosan/chemistry , Drug Carriers/chemistry , Gene Transfer Techniques , Nucleic Acids/administration & dosage , Polyglutamic Acid/analogs & derivatives , Animals , Cell Survival , Cells, Cultured , Chemical Phenomena , Drug Carriers/chemical synthesis , Gene Expression , Genes, Reporter , Larva , Molecular Weight , Polyglutamic Acid/chemical synthesis , Polyglutamic Acid/chemistry , Spectrum Analysis , Zebrafish
7.
Mol Pharm ; 19(1): 67-79, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34931518

ABSTRACT

The development of endosomal disruptive agents is a major challenge in the field of drug delivery and pharmaceutical chemistry. Current endosomal disruptive agents are composed of polymers, peptides, and nanoparticles and have had limited clinical impact. Alternatives to traditional endosomal disruptive agents are therefore greatly needed. In this report, we introduce a new class of low molecular weight endosomal disruptive agents, termed caged surfactants, that selectively disrupt endosomes via reversible PEGylation under acidic endosomal conditions. The caged surfactants have the potential to address several of the limitations hindering the development of current endosomal disruptive agents, such as high toxicity and low excretion, and are amenable to traditional medicinal chemistry approaches for optimization. In this report, we synthesized three generations of caged surfactants and demonstrated that they can enhance the ability of cationic lipids to deliver mRNA into primary cells. We also show that caged surfactants can deliver siRNA into cells when modified with the RNA-binding dye thiazole orange. We anticipate that the caged surfactants will have numerous applications in pharmaceutical chemistry and drug delivery given their versatility.


Subject(s)
Drug Delivery Systems , Nucleic Acids/administration & dosage , Surface-Active Agents/therapeutic use , Drug Delivery Systems/methods , Endosomes/drug effects , Hemolysis/drug effects , Humans , Hydrogen-Ion Concentration , RNA, Messenger/administration & dosage , RNA, Small Interfering/administration & dosage , Structure-Activity Relationship , Surface-Active Agents/administration & dosage , Surface-Active Agents/chemistry
8.
ACS Appl Mater Interfaces ; 13(43): 50802-50811, 2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34665600

ABSTRACT

Type 1 diabetes (T1D) is caused by breakdowns of central and peripheral immune tolerance and destructions of insulin-producing ß-cells. Conventional insulin injection cannot cure the disease. Regulatory immune cells, including regulatory T-cells (Tregs) and regulatory B-cells (Bregs), play critical roles in immune tolerance. Inducing regulatory immune cells to halt the progress of T1D and restore immune tolerance is the promising approach in T1D immunotherapy. Here, tetrahedral framework nucleic acids (tFNAs) were utilized to treat T1D in non-obese diabetic (NOD) mice. 250 nM tFNA treatment was adopted in the experiment to reverse hyperglycemia and protect insulin-secreting ß-cells in diabetic NOD mice. In addition, 250 nM tFNA treatment could induce Tregs and Bregs and suppress helper T (Th)-cells in the pancreas. In the pancreas, cytokines, as a significant signal during CD4+ T-cell differentiation, directly direct the differentiation programs. Apart from cytokines directing the differentiation of T-cells, the signal transducer and activator of transcription (STAT) signal is strongly associated with T-cell differentiation and T1D progression. We demonstrated tFNA treatment inducing regulatory immune cells probably by increasing TGF-ß levels and the STAT signal. To sum up, 250 nM tFNA treatment could protect the diabetic NOD mice from hyperglycemia and preserve the functions of ß-cells by restoring peripheral immune tolerance. The possible mechanism of inducing immune tolerance was related to the STAT signal and cytokine changes in the pancreas. Moreover, immunoregulation capabilities of tFNAs were demonstrated in the experiment, which set the foundation of tFNAs participating in further antigen-specific immunotherapies.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Nucleic Acids/therapeutic use , Animals , Diabetes Mellitus, Type 1/immunology , Female , Immune Tolerance/drug effects , Immune Tolerance/immunology , Immunotherapy , Injections, Intravenous , Mice , Mice, Inbred ICR , Mice, Inbred NOD , Nucleic Acids/administration & dosage , Nucleic Acids/chemistry
9.
EBioMedicine ; 73: 103624, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34688033

ABSTRACT

Immune checkpoints are regulatory molecules responsible for determining the magnitude and nature of the immune response. The aim of immune checkpoint targeting immunotherapy is to manipulate these interactions, engaging the immune system in treatment of cancer. Clinically, the use of monoclonal antibodies to block immunosuppressive interactions has proven itself to be a highly effective immunotherapeutic intervention. Within the literature there are numerous candidates for next generation of immune checkpoint targeting strategies. One such example is the use of nucleic acid to alter expression levels of immune checkpoint molecules, either as antisense oligo nucleotides/siRNA, to downregulate inhibitory molecules, or mRNA/DNA, to express co-stimulatory molecules. A significant component of nucleic acid delivery is its formulation within a nanoparticulate system. In this review we discuss the progress of the preclinical application of nucleic acid-based immunotherapies to target a selection of co-inhibitory/co-stimulatory molecules. Furthermore, we identify the potential and current gaps within the literature which may form the basis of future work.


Subject(s)
Drug Delivery Systems , Gene Expression Regulation , Immune Checkpoint Proteins/genetics , Nanoparticles , Nucleic Acids/administration & dosage , Theranostic Nanomedicine , Animals , Clinical Studies as Topic , Drug Evaluation, Preclinical , Humans , Immune Checkpoint Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/pathology , Nucleic Acids/genetics , Plasmids/administration & dosage , Plasmids/chemistry , RNA Interference , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Treatment Outcome
10.
Adv Drug Deliv Rev ; 178: 113834, 2021 11.
Article in English | MEDLINE | ID: mdl-34492233

ABSTRACT

Recent medical advances have exploited the ability to address a given disease at the underlying level of transcription and translation. These treatment paradigms utilize nucleic acids - including short interfering RNA (siRNA), microRNA (miRNA), antisense oligonucleotides (ASO), and messenger RNA (mRNA) - to achieve a desired outcome ranging from gene knockdown to induced expression of a selected target protein. Towards this end, numerous strategies for encapsulation or stabilization of various nucleic acid structures have been developed in order to achieve intracellular delivery. In this review, we discuss several therapeutic applications of nucleic acids directed towards specific diseases and tissues of interest, in particular highlighting recent technologies which have reached late-stage clinical trials and received FDA approval.


Subject(s)
Drug Delivery Systems/trends , Gene Transfer Techniques/trends , Nucleic Acids/administration & dosage , Nucleic Acids/genetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Animals , COVID-19/genetics , COVID-19/metabolism , COVID-19/therapy , Clinical Trials as Topic/methods , Drug Approval , Drug Delivery Systems/methods , Hepatitis/genetics , Hepatitis/metabolism , Hepatitis/therapy , Humans , MicroRNAs/administration & dosage , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Nucleic Acids/metabolism , Oligonucleotides, Antisense/metabolism , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
11.
Front Immunol ; 12: 722411, 2021.
Article in English | MEDLINE | ID: mdl-34497612

ABSTRACT

Nonviral systems, such as lipid nanoparticles, have emerged as reliable methods to enable nucleic acid intracellular delivery. The use of cationic lipids in various formulations of lipid nanoparticles enables the formation of complexes with nucleic acid cargo and facilitates their uptake by target cells. However, due to their small size and highly charged nature, these nanocarrier systems can interact in vivo with antigen-presenting cells (APCs), such as dendritic cells (DCs) and macrophages. As this might prove to be a safety concern for developing therapies based on lipid nanocarriers, we sought to understand how they could affect the physiology of APCs. In the present study, we investigate the cellular and metabolic response of primary macrophages or DCs exposed to the neutral or cationic variant of the same lipid nanoparticle formulation. We demonstrate that macrophages are the cells affected most significantly and that the cationic nanocarrier has a substantial impact on their physiology, depending on the positive surface charge. Our study provides a first model explaining the impact of charged lipid materials on immune cells and demonstrates that the primary adverse effects observed can be prevented by fine-tuning the load of nucleic acid cargo. Finally, we bring rationale to calibrate the nucleic acid load of cationic lipid nanocarriers depending on whether immunostimulation is desirable with the intended therapeutic application, for instance, gene delivery or messenger RNA vaccines.


Subject(s)
Cations/chemistry , Gene Transfer Techniques , Lipids/chemistry , Liposomes/chemistry , Nanoparticles/chemistry , Nucleic Acids/administration & dosage , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Line , Cell Survival , Chemical Phenomena , Cytokines/chemistry , Drug Carriers , Lipopolysaccharides/chemistry , Mice , Mitochondria/metabolism , Reactive Oxygen Species
12.
Adv Drug Deliv Rev ; 176: 113885, 2021 09.
Article in English | MEDLINE | ID: mdl-34324886

ABSTRACT

Gene therapy is a promising novel method of tissue regeneration by stimulating or inhibiting key signaling pathways. However, their therapeutic applications in vivo are largely limited by several physiological obstacles, such as degradation of nucleases, impermeability of cell membranes, and transport to the desired intracellular compartments. Biomaterial-based gene delivery systems can overcome the problems of stability and local drug delivery, and can temporarily control the overexpression of therapeutic genes, leading to the local production of physiologically relevant levels of regulatory factors. But the gene delivery of biomaterials for tissue regeneration relies on multi-factor design. This review aims to outline the impact of gene delivery methods, therapeutic genes and biomaterials selection on this strategy, emphatically introduce the latest developments in the design of gene delivery vehicles based on biomaterials, summarize the mechanism of nucleic acid for tissue regeneration, and explore the strategies of nucleic acid delivery vehicles for various tissue regeneration.


Subject(s)
Biocompatible Materials/administration & dosage , Gene Transfer Techniques , Hydrogels/administration & dosage , Nanoparticles/administration & dosage , Nucleic Acids/administration & dosage , Tissue Engineering/methods , Animals , Humans
13.
Adv Drug Deliv Rev ; 176: 113835, 2021 09.
Article in English | MEDLINE | ID: mdl-34144087

ABSTRACT

With recent advances in nanotechnology and therapeutic nucleic acids (TNAs), various nucleic acid nanoparticles (NANPs) have demonstrated great promise in diagnostics and therapeutics. However, the full realization of NANPs' potential necessitates the development of a safe, efficient, biocompatible, stable, tissue-specific, and non-immunogenic delivery system. Exosomes, the smallest extracellular vesicles and an endogenous source of nanocarriers, offer these advantages while avoiding complications associated with manufactured agents. The lipid membranes of exosomes surround a hydrophilic core, allowing for the simultaneous incorporation of hydrophobic and hydrophilic drugs, nucleic acids, and proteins. Additional capabilities for post-isolation exosome surface modifications with imaging agents, targeting ligands, and covalent linkages also pave the way for their diverse biomedical applications. This review focuses on exosomes: their biogenesis, intracellular trafficking, transportation capacities, and applications with emphasis on the delivery of TNAs and programmable NANPs. We also highlight some of the current challenges and discuss opportunities related to the development of therapeutic exosome-based formulations and their clinical translation.


Subject(s)
Exosomes/metabolism , Nanoparticles , Nucleic Acids/administration & dosage , Animals , Drug Delivery Systems , Humans , Lipid Metabolism/physiology , Nanotechnology
14.
Nat Nanotechnol ; 16(6): 630-643, 2021 06.
Article in English | MEDLINE | ID: mdl-34059811

ABSTRACT

The increasing number of approved nucleic acid therapeutics demonstrates the potential to treat diseases by targeting their genetic blueprints in vivo. Conventional treatments generally induce therapeutic effects that are transient because they target proteins rather than underlying causes. In contrast, nucleic acid therapeutics can achieve long-lasting or even curative effects via gene inhibition, addition, replacement or editing. Their clinical translation, however, depends on delivery technologies that improve stability, facilitate internalization and increase target affinity. Here, we review four platform technologies that have enabled the clinical translation of nucleic acid therapeutics: antisense oligonucleotides, ligand-modified small interfering RNA conjugates, lipid nanoparticles and adeno-associated virus vectors. For each platform, we discuss the current state-of-the-art clinical approaches, explain the rationale behind its development, highlight technological aspects that facilitated clinical translation and provide an example of a clinically relevant genetic drug. In addition, we discuss how these technologies enable the development of cutting-edge genetic drugs, such as tissue-specific nucleic acid bioconjugates, messenger RNA and gene-editing therapeutics.


Subject(s)
Genetic Vectors/therapeutic use , Nanoparticles/therapeutic use , Nucleic Acids/therapeutic use , Acetylgalactosamine/analogs & derivatives , Acetylgalactosamine/therapeutic use , Gene Editing/methods , Gene Expression Regulation/drug effects , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Humans , Lipids/chemistry , Nanoparticles/chemistry , Nucleic Acids/administration & dosage , Nucleic Acids/pharmacology , Oligonucleotides/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Pyrrolidines/therapeutic use , RNA, Small Interfering/chemistry , RNA, Small Interfering/therapeutic use
15.
Biomed Mater ; 16(4)2021 06 29.
Article in English | MEDLINE | ID: mdl-34116517

ABSTRACT

Nucleic acid-based gene therapy has recently made important progress toward clinical implementation, and holds tremendous promise for the treatment of some life-threatening diseases, such as cancer and inflammation. However, the on-demand delivery of nucleic acid therapeutics in target cells remains highly challenging. The development of delivery systems responsive to specific pathological cues of diseases is expected to offer promising alternatives for overcoming this problem. Among them, the reactive oxygen species (ROS)-responsive delivery systems, which in response to elevated ROS in cancer cells or activated inflammatory cells, can deliver nucleic acid therapeutics on-demand via ROS-induced structural and assembly behavior changes, constitute a promising approach for cancer and anti-inflammation therapies. In this short review, we briefly introduce the ROS-responsive chemical structures, ROS-induced release mechanisms and some representative examples to highlight the current progress in constructing ROS-responsive delivery systems. We aim to provide new insights into the rational design of on-demand gene delivery vectors.


Subject(s)
Genetic Therapy , Inflammation/therapy , Nanomedicine/methods , Neoplasms/therapy , Reactive Oxygen Species , Animals , Humans , Mice , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/metabolism , Nanoparticles/therapeutic use , Nucleic Acids/administration & dosage , Nucleic Acids/chemistry , Nucleic Acids/metabolism , Nucleic Acids/therapeutic use , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism
16.
Adv Drug Deliv Rev ; 174: 348-368, 2021 07.
Article in English | MEDLINE | ID: mdl-33964356

ABSTRACT

Extracellular vesicles (EVs) are membranous nanovesicles secreted from living cells, shuttling macromolecules in intercellular communication and potentially possessing intrinsic therapeutic activity. Due to their stability, low immunogenicity, and inherent interaction with recipient cells, EVs also hold great promise as drug delivery vehicles. Indeed, they have been used to deliver nucleic acids, proteins, and small molecules in preclinical investigations. Furthermore, EV-based drugs have entered early clinical trials for cancer or neurodegenerative diseases. Despite their appeal as delivery vectors, however, EV-based drug delivery progress has been hampered by heterogeneity of sample types and methods as well as a persistent lack of standardization, validation, and comprehensive reporting. This review highlights specific requirements for EVs in drug delivery and describes the most pertinent approaches for separation and characterization. Despite residual uncertainties related to pharmacodynamics, pharmacokinetics, and potential off-target effects, clinical-grade, high-potency EV drugs might be achievable through GMP-compliant workflows in a highly standardized environment.


Subject(s)
Drug Delivery Systems , Drug Development/methods , Extracellular Vesicles/metabolism , Animals , Cell Communication/physiology , Humans , Neoplasms/drug therapy , Neurodegenerative Diseases/drug therapy , Nucleic Acids/administration & dosage , Proteins/administration & dosage
17.
J Mater Chem B ; 9(21): 4298-4302, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34018540

ABSTRACT

Endosomal escape is crucial for the delivery of nucleic acids. However, the understanding of the underlying mechanisms is still deficient. In this work, we explored the effects of lipid- and polymer-based transfection reagents on the permeability of cellular membranes through an innovative method combining a proton-sensing transistor and a cytosolic LDH leakage assay, which allows us to distinguish between modes of molecule permeation that may occur during endosomal escape. By testing the commercial reagents lipofectin and in vivo JetPEI under physiological and endosomal pH conditions, we found that both lipid- and polymer-based transfection reagents have pH-dependent pore-forming activity, with the former creating smaller pores than the latter. This versatile approach of assessing carrier-membrane interactions is expected to contribute to the development of next-generation nucleic acid delivery systems.


Subject(s)
Cell Membrane Permeability , Lipids/chemistry , Nucleic Acids/administration & dosage , Polymers/chemistry , Hep G2 Cells , Humans , L-Lactate Dehydrogenase/metabolism , Protons
18.
Adv Drug Deliv Rev ; 175: 113809, 2021 08.
Article in English | MEDLINE | ID: mdl-34033819

ABSTRACT

Thanks to their abilities to modulate the expression of virtually any genes, RNA therapeutics have attracted considerable research efforts. Among the strategies focusing on nucleic acid gene inhibitors, antisense oligonucleotides and small interfering RNAs have reached advanced clinical trial phases with several of them having recently been marketed. These successes were obtained by overcoming stability and cellular delivery issues using either chemically modified nucleic acids or nanoparticles. As nucleic acid gene inhibitors are promising strategies to treat inflammatory diseases, this review focuses on the barriers, from manufacturing issues to cellular/subcellular delivery, that still need to be overcome to deliver the nucleic acids to sites of inflammation other than the liver. Furthermore, key examples of applications in rheumatoid arthritis, inflammatory bowel, and lung diseases are presented as case studies of systemic, oral, and lung nucleic acid delivery.


Subject(s)
Inflammation/drug therapy , Nanomedicine/methods , Nanoparticle Drug Delivery System , Nucleic Acids/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Drug Delivery Systems/methods , Genes/drug effects , Humans , Inflammation/genetics , Nucleic Acids/therapeutic use , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/therapeutic use , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use
19.
Eur J Clin Microbiol Infect Dis ; 40(8): 1623-1631, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33666790

ABSTRACT

In this study, immunoregulation and desensitization therapies were jointly applied in the treatment of asthma, in which chitosan (CS) nanoparticles were used. BALB/c mice were selected and mouse models of asthma were constructed. Mice were divided into 7 groups. A double-chamber plethysmograph, MTT, hematoxylin-eosin staining, and ELISA were used. The expression levels of IL-4 and IL-5 in lung tissue cells were detected. CS-BCG-PSN-OVA sustained-release vaccines significantly alleviated airway hyperresponsiveness (AHR) in asthmatic mice. The numbers of total lymphocytes and eosinophils in BALF were remarkably reduced. The expression levels of IL-4 and IL-5 in lung tissue cells of the treatment groups were dramatically decreased. CS-BCG-PSN-OVA was found in vitro to be able to inhibit OVA-induced T-cell proliferation and upregulate the proportion of CD4+CD25+Foxp3+ T cells. CS-BCG-PSN-OVA sustained-release vaccine could significantly attenuate AHR and airway inflammation in asthmatic mice. Thus, it has a promising application prospect for the treatment of bronchial asthma.


Subject(s)
Asthma/drug therapy , BCG Vaccine/administration & dosage , Nanoparticles , Nucleic Acids/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , Chitosan , Drug Liberation , Female , Inflammation , Interleukin-4/metabolism , Interleukin-5/metabolism , Lung/pathology , Mice, Inbred BALB C , Ovalbumin , Polysaccharides
20.
Biochem Pharmacol ; 189: 114492, 2021 07.
Article in English | MEDLINE | ID: mdl-33647260

ABSTRACT

CRISPR-based therapeutics have entered clinical trials but no methods to inhibit Cas enzymes have been demonstrated in a clinical setting. The ability to inhibit CRISPR-based gene editing or gene targeting drugs should be considered a critical step in establishing safety standards for many CRISPR-Cas therapeutics. Inhibitors can act as a failsafe or as an adjuvant to reduce off-target effects in patients. In this review we discuss the need for clinical inhibition of CRISPR-Cas systems and three existing inhibitor technologies: anti-CRISPR (Acr) proteins, small molecule Cas inhibitors, and small nucleic acid-based CRISPR inhibitors, CRISPR SNuBs. Due to their unique properties and the recent successes of other nucleic acid-based therapeutics, CRISPR SNuBs appear poised for clinical application in the near-term.


Subject(s)
CRISPR-Cas Systems/drug effects , Gene Editing/methods , Gene Targeting/methods , Nucleic Acids/administration & dosage , Animals , CRISPR-Cas Systems/physiology , Humans , Nucleic Acids/genetics , Nucleic Acids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...