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










Database
Language
Publication year range
1.
J Pharm Sci ; 112(2): 386-403, 2023 02.
Article in English | MEDLINE | ID: mdl-36351479

ABSTRACT

The remarkable impact of mRNA vaccines on mitigating disease and improving public health has been amply demonstrated during the COVID-19 pandemic. Many new mRNA-based vaccine and therapeutic candidates are in development, yet the current reality of their stability limitations requires their frozen storage. Numerous challenges remain to improve formulated mRNA stability and enable refrigerator storage, and this review provides an update on developments to tackle this multi-faceted stability challenge. We describe the chemistry underlying mRNA degradation during storage and highlight how lipid nanoparticle (LNP) formulations are a double-edged sword: while LNPs protect mRNA against enzymatic degradation, interactions with and between LNP excipients introduce additional risks for mRNA degradation. We also discuss strategies to improve mRNA stability both as a drug substance (DS) and a drug product (DP) including the (1) design of the mRNA molecule (nucleotide selection, primary and secondary structures), (2) physical state of the mRNA-LNP complexes, (3) formulation composition and purity of the components, and (4) DS and DP manufacturing processes. Finally, we summarize analytical control strategies to monitor and assure the stability of mRNA-based candidates, and advocate for an integrated analytical and formulation development approach to further improve their storage, transport, and in-use stability profiles.


Subject(s)
COVID-19 , Nanoparticles , Humans , Pandemics , Lipids/chemistry , COVID-19/prevention & control , Nanoparticles/chemistry , Liposomes , RNA, Messenger/genetics , mRNA Vaccines
2.
J Control Release ; 317: 154-165, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31765703

ABSTRACT

Small, synthetic oligonucleotides (ON) are of great interest as potential disease modifying drugs, mainly because of their ability to modulate previously undruggable target mutations. To date, therapeutic applications of ON are, however, limited by their physicochemical properties, including poor stability, rapid excretion and low intracellular access. In order to overcome some of these shortcomings, ON are generally formulated using nanoparticle (NP) delivery systems. Alternatively, the poor stability can be circumvented by including chemical modifications to the backbone or sugars of the ON. Some of these modifications also result in better intracellular target access of these otherwise membrane-impermeable macromolecules. Therefore, complex formulation of ON into NP in order to overcome the hurdle of intracellular access might not always be needed, especially in case of local delivery. In this study, the delivery and functionality of chemically modified ON in free form was compared to polymeric NP assisted delivery, measuring their effectivity and efficiency. For this reason, phosphorothioate (PS) backbone-modified 18-mer ON with either 2'OMe or 2'MOE-modifications were selected, capable of eliciting exon-skipping of an aberrant exon in fluorescence based in vitro and in vivo model systems. The NP consisted of poly(D,L-lactic,co-glycolic acid) and poly-ß-amino-ester, previously demonstrated to successfully deliver nucleic acids via the pulmonary route. Several NP formulation parameters were tested in order to optimize the delivery of the ON, including ratio polymer:ON, NP size and concentration. The results reported here show clear differences between gymnotic and nanoparticle mediated ON delivery in terms of cellular uptake and local tissue distribution. In vitro, differences in exon-skipping efficiencies were observed with 2'OMe and 2'MOE ON either in free form or formulated in NP, with the striking observation that 2'OMe ON formulated in polymeric NP did not result in exon skipping. Gymnotic delivery of 2'MOE ON into the respiratory tract of mice resulted in functional delivery of exon-skipping ON into nasal epithelia and lungs as well as other downstream tissues and organs, pointing towards a gradual redistribution of locally delivered ONs, with limited but measurable systemic exposure. Conversely, NP-mediated delivery into the respiratory tract resulted in a more contained functional delivery at 10× lower ON doses compared to gymnotic delivery. Based on these findings we conclude that gymnotic delivery of 2'OMe or 2'MOE exon-skipping ON to the respiratory tract is effective, but that NP formulation might be advantageous in case spread of ON to non-target tissue can lead to undesired effects.


Subject(s)
Nanoparticles , Nucleic Acids , Animals , Mice , Oligonucleotides , RNA , Respiratory System
3.
Int J Pharm ; 548(2): 793-802, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-29275035

ABSTRACT

There is a very large variety in the types of nanoparticulate lipid formulations for oligonucleotides, and remarkably, also a very large heterogeneity in the methods that are used for analyzing oligonucleotide load, encapsulation efficiency and oligonucleotide release. Furthermore, a literature survey showed that the extent to which these procedures are reported in scientific literature varies greatly, with some of them not even reporting any quantification at all. This greatly hampers the reproducibility of nanoparticle preparation between different researchers and between different laboratories, which slows down the clinical translation of such nanomedicines. In this work, a standardized extraction method from liposomes is proposed, in which potential contaminants from the carrier are removed by a simple extraction of the oligonucleotides. These extracts were then analyzed with seven commonly used methods for oligonucleotide quantification, including several absorbance based methods and the most commonly applied dye binding assay. Strikingly, differences in absolute values up to fourfold were found when the same sample was analyzed using different methods which should be taken into consideration when reports using different methods are compared. Furthermore, these results indicate that the most commonly applied method, the dye binding assay, may -without adaptations- not be suitable for short oligonucleotides like siRNAs. The found differences in quantification methods as described here underscore the need for proper documentation of methods to correctly interpret published results, which -with regard to oligonucleotide analysis- is currently lacking in many reports.


Subject(s)
Nanoparticles/analysis , Nanoparticles/chemistry , Oligonucleotides/analysis , Oligonucleotides/chemistry , Drug Compounding/methods , Drug Evaluation, Preclinical/methods , Liposomes , RNA, Small Interfering/analysis , RNA, Small Interfering/chemistry
4.
J Control Release ; 244(Pt B): 139-148, 2016 12 28.
Article in English | MEDLINE | ID: mdl-27498021

ABSTRACT

The CRISPR-Cas9 gene editing system has taken the biomedical science field by storm, initiating rumors about future Nobel Prizes and heating up a fierce patent war, but also making significant scientific impact. The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR), together with CRISPR-associated proteins (Cas) are a part of the prokaryotic adaptive immune system and have successfully been repurposed for genome editing in mammalian cells. The CRISPR-Cas9 system has been used to correct genetic mutations and for replacing entire genes, opening up a world of possibilities for the treatment of genetic diseases. In addition, recently some new CRISPR-Cas systems have been discovered with interesting mechanistic variations. Despite these promising developments, many challenges have to be overcome before the system can be applied therapeutically in human patients and enabling delivery technology is one of the key challenges. Furthermore, the relatively high off-target effect of the system in its current form prevents it from being safely applied directly in the human body. In this review, the transformation of the CRISPR-Cas gene editing systems into a therapeutic modality will be discussed and the currently most realistic in vivo applications will be highlighted.


Subject(s)
CRISPR-Cas Systems , Gene Transfer Techniques , Genetic Therapy , Animals , Gene Editing , Humans
5.
Nanoscale ; 8(16): 8955-65, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-27073992

ABSTRACT

Coiled coil interactions are strong protein-protein interactions that are involved in many biological processes, including intracellular trafficking and membrane fusion. A synthetic heterodimeric coiled-coil forming peptide pair, known as E3 (EIAALEK)3 and K3 (KIAALKE)3 was used to functionalize liposomes encapsulating a splice correcting oligonucleotide or siRNA. These peptide-functionalized vesicles are highly stable in solution but start to cluster when vesicles modified with complementary peptides are mixed together, demonstrating that the peptides quickly coil and crosslink the vesicles. When one of the peptides was anchored to the cell membrane using a hydrophobic cholesterol anchor, vesicles functionalized with the complementary peptide could be docked to these cells, whereas non-functionalized cells did not show any vesicle tethering. Although the anchored peptides do not have a downstream signaling pathway, microscopy pictures revealed that after four hours, the majority of the docked vesicles were internalized by endocytosis. Finally, for the first time, it was shown that the coiled coil assembly at the interface between the vesicles and the cell membrane induces active uptake and leads to cytosolic delivery of the nucleic acid cargo. Both the siRNA and the splice correcting oligonucleotide were functionally delivered, resulting respectively in the silencing or recovery of luciferase expression in the appropriate cell lines. These results demonstrate that the docking to the cell by coiled coil interaction can induce active uptake and achieve the successful intracellular delivery of otherwise membrane impermeable nucleic acids in a highly specific manner.


Subject(s)
Liposomes/administration & dosage , Liposomes/chemistry , Nucleic Acids/administration & dosage , Amino Acid Sequence , Cell Membrane Permeability , Drug Delivery Systems , Endocytosis , HeLa Cells , Humans , Nanotechnology , Nucleic Acids/pharmacokinetics , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Protein Interaction Domains and Motifs
6.
Nanoscale ; 7(46): 19687-98, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26553270

ABSTRACT

Cationic vectors have demonstrated the potential to facilitate intracellular delivery of therapeutic oligonucleotides. However, enhanced transfection efficiency is usually associated with adverse effects, which also proves to be a challenge for vectors based on cationic peptides. In this study a series of proteolytically stable palmitoylated α-peptide/ß-peptoid peptidomimetics with a systematically varied number of repeating lysine and homoarginine residues was shown to self-assemble with small interfering RNA (siRNA). The resulting well-defined nanocomplexes were coated with anionic lipids giving rise to net anionic liposomes. These complexes and the corresponding liposomes were optimized towards efficient gene silencing and low adverse effects. The optimal anionic liposomes mediated a high silencing effect, which was comparable to that of the control (cationic Lipofectamine 2000), and did not display any noticeable cytotoxicity and immunogenicity in vitro. In contrast, the corresponding nanocomplexes mediated a reduced silencing effect with a more narrow safety window. The surface coating with anionic lipid bilayers led to partial decomplexation of the siRNA-peptidomimetic nanocomplex core of the liposomes, which facilitated siRNA release. Additionally, the optimal anionic liposomes showed efficient intracellular uptake and endosomal escape. Therefore, these findings suggest that a more efficacious and safe formulation can be achieved by surface coating of the siRNA-peptidomimetic nano-self-assemblies with anionic lipid bilayers.


Subject(s)
Gene Silencing , Lipid Bilayers , Materials Testing , Nanoparticles/chemistry , Peptidomimetics , RNA, Small Interfering , Cell Line, Tumor , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/pharmacology , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology
7.
ACS Nano ; 9(4): 3740-52, 2015.
Article in English | MEDLINE | ID: mdl-25831471

ABSTRACT

Treatment of cancer patients with taxane-based chemotherapeutics, such as paclitaxel (PTX), is complicated by their narrow therapeutic index. Polymeric micelles are attractive nanocarriers for tumor-targeted delivery of PTX, as they can be tailored to encapsulate large amounts of hydrophobic drugs and achiv prolonged circulation kinetics. As a result, PTX deposition in tumors is increased, while drug exposure to healthy tissues is reduced. However, many PTX-loaded micelle formulations suffer from low stability and fast drug release in the circulation, limiting their suitability for systemic drug targeting. To overcome these limitations, we have developed PTX-loaded micelles which are stable without chemical cross-linking and covalent drug attachment. These micelles are characterized by excellent loading capacity and strong drug retention, attributed to π-π stacking interaction between PTX and the aromatic groups of the polymer chains in the micellar core. The micelles are based on methoxy poly(ethylene glycol)-b-(N-(2-benzoyloxypropyl)methacrylamide) (mPEG-b-p(HPMAm-Bz)) block copolymers, which improved the pharmacokinetics and the biodistribution of PTX, and substantially increased PTX tumor accumulation (by more than 2000%; as compared to Taxol or control micellar formulations). Improved biodistribution and tumor accumulation were confirmed by hybrid µCT-FMT imaging using near-infrared labeled micelles and payload. The PTX-loaded micelles were well tolerated at different doses, while they induced complete tumor regression in two different xenograft models (i.e., A431 and MDA-MB-468). Our findings consequently indicate that π-π stacking-stabilized polymeric micelles are promising carriers to improve the delivery of highly hydrophobic drugs to tumors and to increase their therapeutic index.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Drug Carriers/chemistry , Mammary Neoplasms, Experimental/drug therapy , Micelles , Paclitaxel/chemistry , Paclitaxel/pharmacology , Polymers/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Drug Carriers/pharmacokinetics , Drug Stability , Female , Humans , Kinetics , Mammary Neoplasms, Experimental/diagnosis , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Methacrylates/chemistry , Mice , Multimodal Imaging , Paclitaxel/therapeutic use , Polyethylene Glycols/chemistry , Polymers/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays
8.
J Control Release ; 202: 14-20, 2015 Mar 28.
Article in English | MEDLINE | ID: mdl-25626085

ABSTRACT

The modification of liposomal surfaces is of interest for many different applications and a variety of chemistries are available that makes this possible. A major disadvantage of commonly used coupling chemistries (e.g. maleimide-thiol coupling) is the limited control over the site of conjugation in cases where multiple reactive functionalities are present, leading to heterogeneous products and in some cases dysfunctional conjugates. Bioorthogonal coupling approaches such as the well-established copper-catalyzed azide-alkyne cycloaddition (CuAAC) "click" reaction are attractive alternatives as the reaction kinetics are favorable and azide-containing reagents are widely available. In the work described here, we prepared lipids containing a reactive cyclooctyne group and, after incorporation into liposomes, demonstrated successful conjugation of both a small molecule dye (5'-TAMRA-azide) as well as a larger azide-containing model protein based upon a designed ankyrin repeat protein (azido-DARPin). By applying the strain-promoted azido-alkyne cycloaddition (SPAAC) the use of Cu(I) as a catalyst is avoided, an important advantage considering the known deleterious effects associated with copper in cell and protein studies. We demonstrate complete control over the number of ligands coupled per liposome when using a small molecule azide with conjugation occurring at a reasonable reaction rate. By comparison, the conjugation of a larger azide-modified protein occurs more slowly, however the number of protein ligands coupled was found to be sufficient for liposome targeting to cells. Importantly, these results provide a strong proof of concept for the site-specific conjugation of protein ligands to liposomal surfaces via SPAAC. Unlike conventional approaches, this strategy provides for the homogeneous coupling of proteins bearing a single site-specific azide modification and eliminates the chance of forming dysfunctional ligands on the liposome. Furthermore, the absence of copper in the reaction process should also make this approach much more compatible with cell-based and in vivo applications.


Subject(s)
Azides/chemistry , Bridged Bicyclo Compounds/chemistry , Coloring Agents/chemistry , Liposomes/chemistry , Nuclear Proteins/chemistry , Rhodamines/chemistry , Ankyrin Repeat , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Cholesterol/chemistry , Click Chemistry , Copper , Epithelial Cell Adhesion Molecule , HT29 Cells , Humans , Nuclear Proteins/administration & dosage , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry
9.
Expert Opin Drug Deliv ; 10(10): 1399-410, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23796390

ABSTRACT

INTRODUCTION: Long circulating liposomal drug carriers are widely used in experimental cancer therapy because they avoid excretion and benefit from the enhanced permeability and retention-effect to accumulate at the tumor site while simultaneously limiting systemic exposure to the cytotoxic drug due to their high stability. A drawback of the stability of the formulation is that the unloading of the drug at the target site is very poor. This opens up a new challenge to trigger drug release at the target site, while still retaining most of the drug inside the carrier while it resides in the bloodstream. AREAS COVERED: A short introduction is given about lipid polymorphism and phase behavior. To illustrate how this can be used to design triggered release systems, the development of delivery systems that are activated by tumor environment, UV or visible light and mild heat are discussed. The most recent triggered release systems have evolved even further, creating a need for more sophisticated triggers, which are as non-invasive and patient friendly as possible. EXPERT OPINION: Currently the most promising triggered release systems that have advanced furthest are thermosensitive liposomal delivery systems. As mild hyperthermia also increases tissue permeability it appears a suitable trigger for drug release while it also assists in drug accumulation. Combined with an advanced imaging system in the MR-high intensity focused ultrasound, this could be the combination of delivery system and trigger that can achieve clinical success.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Liposomes/chemistry , Neoplasms/drug therapy , Animals , Hot Temperature , Humans , Hyperthermia, Induced
SELECTION OF CITATIONS
SEARCH DETAIL
...