Controlled Release Diary
Banda 335,
July 10, 2021
, pages 465-480
Author links open overlay panel, ,
Abstract
Non-viral vehicles hold therapeutic promise for enhancing delivery of a variety of payloadsin vitroElive, including small molecule drugs, biologics and especiallynucleic acids. However, its effectiveness at the cellular level is limited by several delivery barriers, the most important being endolysosomal degradation. Trapping of vehicles and their cargo in the acidified endosome prevents access to the cytosol, nucleus, and other subcellular compartments. Understanding the factors that contribute to intracellular uptake and transport, particularly endosomal entrapment and release, is critical to overcoming transport obstacles within cells. In this review, we summarize and compare experimental techniques to assess the extent of endosomal leakage from a variety of non-viral vehicles and describe proposed leakage mechanisms for different classes of lipid-, polymer-, and peptide-based drugs. Based on this assessment, we present forward-looking strategies that use information from mechanistic studies to inform the rational design of efficient delivery vehicles.
introduction
Nanomedicine in the form of non-viral carriers has enormous potential to treat a wide range of diseases. Viral vectors have been widely used for gene delivery due to their high transfection efficiency, but they also carry the risk of eliciting immune responses and causing faulty insertional mutagenesis [1]. Therefore, the search for alternative non-viral methods has expanded. Advances in these delivery systems include improvements in biocompatibility, targeting ability, and the ability to deliver larger payloads [[2], [3], [4]]. For example, due to their modifiable properties, polymeric nanoparticles (NPs) have been used to enhance central nervous system delivery [5] and intravenous delivery of anticancer drugs [6,7]. Furthermore, there is great interest in using polymeric NPs and lipid NPs (LNPs) to deliver therapeutic nucleic acids [8,9], gene editing agents such as CRISPR/Cas9 [10,11] and, more recently, mRNA-based vaccines [12]. The first two FDA-approved SARS-COV-2 vaccines, Moderna's mRNA-1273 [13] and Pfizer's BNT162b2 [14], rely on LNP carriers for safe and effective delivery [15,16].
Despite continued advances, clinical translation of non-viral vectors continues to be hampered by several delivery challenges. Both extra- and intracellular delivery barriers after systemic administration have been extensively reviewed [17,18], and the ability to control delivery to target tissues, cell types, and even specific sites within cells remains a desirable trait for many formulations [19] . . At the intracellular level, cellular uptake and endosomal leakage are major obstacles for nanodrugs [20]. For example, polymeric NPs are typically taken up by cells through the endocytic pathway and therefore pass through the endolysosomal pathway [21] (Fig. 1). The relative acidity of endosomes - compared to interstitial fluid or cytoplasm - can lead to significant degradation of NPs and their cargo, and therefore they must be able to exit this compartment, i. H. "escape" from the endosome to reach their intracellular target viable state. This process remains the impeding barrier to efficient gene delivery at the cellular level [20]. There are several proposed underlying mechanisms to describe how non-viral carriers are released into the cytosol. However, understanding the intracellular delivery mechanism is complicated by the likelihood that it is not only vehicle-dependent but also cell-type dependent. Different cell types have different gene expression profiles and endosomal transport patterns (Fig. 1), so each interacts with vehicles in unique ways. Other factors, such as cell density, polarity and differentiation, can also modulate traffic patterns [22,23]. These complications increase the complexity of the endocytosis process itself, as questions remain about the number of possible pathways for cells. Although endocytosis has been the subject of studies for many decades, new pathways and interactions, particularly for clathrin-independent endocytosis, are still being elucidated [22,24]. After vehicle uptake, additional crosstalk between the endolysosomal pathway and other processes, such as autophagy and exosome biogenesis [25], further complicates the picture. Therefore, a one-size-fits-all generic mechanism for nanomedicine trade may not exist. In order to pursue rational design strategies that improve endosomal escape, it is important to understand each nanocarrier in its intended delivery context, particularly in relation to the target cell type of interest.
In this review, we focus on quantitative and qualitative methods to assess the intracellular fate of non-viral vehicles. We describe the most common techniques for quantifying endosomal leakage and use this methodological understanding to assess what is currently known about the intracellular transport mechanisms of lipid-, polymer-, and peptide-based vehicles. With this in mind, we describe ways in which current knowledge of endosomal escape mechanisms can be used to design smarter and more efficient delivery systems.
section clippings
Experimental methods to evaluate mechanisms of endosomal escape and cell uptake
There is no standard technique to determine the mechanisms of intracellular transport or endosomal escape. Instead, commonly used experimental tools were used to shed light on various aspects of the process. Current methods for assessing endosomal leakage can be divided into the following categories: leakage assays, complementation assays, cytosolic activation assays, genetic and inhibitory screens, and co-localization assessments. These experimental methods are described below;
Endosomal escape mechanisms of non-viral delivery vehicles
Several proposed endosomal escape mechanisms for non-viral vehicles have appeared in the literature. However, despite decades of intense research, the topic remains controversial, likely due to the influence that different vehicle compositions and surface chemistries can have on the escape route (Fig. 3). We summarize current understanding of endosomal escape and absorption mechanisms below and organize the information by type of delivery vehicle (lipid, polymer, and peptide based).
Improved vehicle design for improved endosomal exhaust
Endosomal entrapment represents a key delivery bottleneck, and therefore the development of vehicles with enhanced endosomal escape properties is of growing interest, particularly in the context of promising strategies for the rational design of nanomedicines. Some were inspired by strategies used by viruses, while others used expanding synthetic approaches. In the following section, we briefly describe some recently developed strategies to improve delivery vehicles.
Conclusion and future prospects
The endolysosomal pathway represents a formidable barrier to efficient non-viral therapeutic delivery into cells. To realize the full potential of nanomedicines, a more complete understanding of endosomal escape mechanisms is needed, along with more research on structure-function relationships between vehicle properties and transport routes. The first step towards achieving this goal is the development of uncomplicated, high-throughput techniques to measure endosomal leakage.
Declaration of Competing Interests
The authors declare that there are no conflicting interests.
thanks
This work was supported by a grant from theNational Institute of Health(NIH;UG3 HL147352). A.S.P was supported by twoNIH National Research Service Awards(NRSAs: umT32 GM86287training scholarship andF32 HL142144individual postdoctoral fellowship) and a postdoctoral research fellowship (PIOTRO20F0) fromCystic Fibrosis Foundation(CPF).
references(210)
- R.M.Straubingerand others
Liposome endocytosis and intracellular fate of encapsulated molecules: Finding a low pH compartment after internalization into coated vesicles
cell
(1983)
- MAFriedrichand others
Cardiotoxicity with rituximab, cyclophosphamide, non-pegylated liposomal doxorubicin, vincristine, and prednisolone compared with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone in the first-line treatment of patients with diffuse large B-cell lymphoma: a phase III randomized trial by Group Austrian Work on Cancer AGMT (NHL-14)
EUR. J. Cancer
(2016)
- JKHemorrhoidsand others
Quantification of the strength of nanoparticle-cell membrane interactions and their influence on intracellular transport of nanoparticles
(Video) Lipid Nanoparticle Portfolio for Enabling Non-Viral Delivery - Dr. Anitha Thomasbiomaterials
(2008)
- S.I am sandand others
A new series of amino acids for mRNA delivery: improved endosomal escape and sustained pharmacology and safety in non-human primates
Suggest. Day.
(2018)
- N.L.Horseand others
Histone-driven polyplexes prevent endosomal escape and enter the nucleus during postmitotic redistribution of ER membranes
Mol. ther. nucleic acids
(2015)
- S.Ben Djemaaand others
Cell penetration peptide gH625 promotes endosomal escape of nanovectored siRNA in a triple negative breast cancer cell line
Biomacromolecules
(2019)
- A.Meckeand others
Direct observation of lipid bilayer disruption by poly(amidoamine) dendrimers
chem. lipid physics
(2004)
- ClockTiwariand others
Improved intracellular translocation and biodistribution of gold nanoparticles functionalized with a cell penetrating peptide (VG-21) from the vesicular stomatitis virus
biomaterials
(2014)
- R.N.Majzouband others
Microscopic colocalization of fluorescence of nucleic acid lipid nanoparticles with wild and mutant Rab5-GFP: a platform to study early endosomal events
Biochim. Biophys. Akt
(2015)
- S.N.OKand others
The effect of nanoparticle size and NLS density on nuclear targeting in cancer and normal cells; Impaired nuclear import and aberrant intracellular transport of nanoparticles in gliomas
Control J. release
(2017)
Intracellular delivery of a dynein-based nanoparticle designed for gene delivery
EUR. J. Pharm.Science.
(2018)
Cellular uptake mechanism and intracellular fate of hydrophobically modified glycolic chitosan nanoparticles
Control J. release
(2009)
The intracellular uptake and fate of PLGA nanoparticles in epithelial cells
biomaterials
(2009)
Array CRISPR screening identifies novel targets that enhance productive mRNA delivery by MC3-based lipid nanoparticles
SLAS Discovery
(2020)
The use of inhibitors to study endocytic pathways of gene carriers: optimization and pitfalls
Suggest. Day.
(2010)
endocytose of nanodrugs
Control J. release
(2010)
Elucidation of the mechanisms of action of cell penetrating peptides for membrane interaction, cellular uptake and translocation using the hydrophobic counteranion pyrene butyrate
Biochim. Biophys. Akt
(2009)
A plug-and-play ratiometric pH sensor nanoprobe for high-throughput investigation of endosomal leakage
biomaterials
(2015)
To determine the effects of PEI adsorption on the permeability of 1,2-dipalmitoylphosphatidylcholine/bis(monoacylglycero)phosphate membranes under osmotic stress
Acta Biomater.
(2018)
Diblock copolymers with adjustable pH transitions for gene delivery
biomaterials
(2012)
Concepts of cellular uptake of nanoparticles, intracellular transport and kinetics in nanomedicine
Medication administration for adults Rev.
(2019)
Polymeric nucleic acid delivery vehicle
Medication administration for adults Rev.
(2020)
Debugging the Genetic Code: In Vivo Non-Viral Delivery of Therapeutic Genome Editing Technologies
Act. Opinion. Biomed. Closely.
(2018)
Polymeric nanoparticles for siRNA delivery: production and applications
Int. J. Pharm.
(2017)
Advances and problems in the use of viral vectors for gene therapy
Nat. Rev. Genet.
(2003)
Non-viral vectors for gene-based therapy
Nat. Rev. Genet.
(2014)
Delivery technologies for cancer immunotherapy
Nat. Rev. Drug Discovery.
(2019)
Nanoparticles as carriers for drug delivery from macromolecules across the blood-brain barrier
Document. drug delivery
(2020)
Polymer nanoparticles as smart carriers for enhanced therapeutic delivery to the CNS
agir. Farm. Des.
(2017)
Polymer nanoparticles for intravenous administration of anticancer drugs: the checkpoints on the way from synthesis to clinical implementation
Nanomaßstab
(2018)
Nanomedicine as an emerging platform for metastatic lung cancer therapy
Krebsmetastasen Rev.
(2015)
Lipid nanoparticles enabling gene therapies: from concept to clinical utility
Nucleic Acid Therapist.
(2018)
CRISPR/Cas9 delivery for therapeutic genome editing
J. GeneMed.
(2019)
Vaccines based on nanoparticles against respiratory viruses
Front. immune
(2019)
An mRNA vaccine against SARS-CoV-2 - preliminary report
N. Engl. J.Med.
(2020)
Phase I/II Study of the BNT162b1 COVID-19 RNA Vaccine in Adults
Nature
(2020)
Efficacy and safety of mRNA-1273 SARS-CoV-2 vaccine
N. Engl. J.Med.
(2020)
Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine
N. Engl. J.Med.
(2020)
Overcoming physiological barriers to nanoparticle delivery - are we there yet?
Front. Bioeng. Biotechnology.
(2019)
Challenges in carrier-mediated intracellular delivery: crossing endosomal barriers
KABEL Nanomed. Nanobiotecnologia.
(2016)
Nanoescapology: advances in the understanding of endosomal escape of polymeric nanoparticles
KABEL Nanomed. Nanobiotecnologia.
(2017)
Clathrin-independent endocytosis: increasing complexity
Histochemistry. Zellbiol.
(2018)
Endocytosis in proliferating, quiescent, and terminally differentiated cells
J. CellSci.
(2018)
Mechanisms of clathrin-mediated endocytose
night. Rev. Moth.
(2018)
The interaction of exosomes and autophagy - partners in crime
J. CellSci.
(2018)
In vitro and in vivo mRNA delivery using lipid-coated pH-responsive polymer nanoparticles
Mol. farm.
(2011)
High-resolution imaging and tracking of protein-protein interactions in the cellular space below diffraction
nat. Together.
(2014)
Light-activated localization microscopy with bimolecular fluorescence complementation (BiFC-PALM) for nanoscale imaging of protein-protein interactions in cells
Another
(2014)
Versatile labeling of proteins in cells with cleaved fluorescent protein
nat. Together.
(2016)
Green fluorescent protein nanopolygons as monodisperse supramolecular assemblies of functional proteins with defined valence
nat. Together.
(2015)
Quoted by (30)
Coactivatable dual-factor fluorescence nanosensor with enhanced accessibility of cytoplasmic biomarkers for selective tumor imaging
2023, Biosensors and Bioelectronics
Fluorescent nanosensor-based tumor imaging holds great promise in cancer diagnosis and treatment support, however, signal contrast is severely impaired by unspecific/unwanted activation in microscopic regions with a severely limited local abundance of biomarkers. Here we developed an activation enhancement strategy by integrating and manipulating the coactivation of two lysosomal detection and escape factors, facilitating increased accessibility of the cytosolic biomarker. Using hybrid DNA probes in gold nanoquenchers, ATP detection triggered a conformational switch of the corresponding aptamer units, triggering the exposure of a hidden loop in a loop structure. Subsequently, miRNA-21 recognition was triggered by toehold-mediated strand displacement and shedding of binding complexes. Application of the lysosomotropic agent chloroquine at optimized time intervals facilitated the release of nanosensors into the cytosol and an approximately 10.5-fold increase in intracellular fluorescencein vitro, while coactivation improved the signaling rate from cancer cells to normal cells by 5.9 times. The synergistic effects resulted in a high tumor-to-normal tissue ratio of approximately 7.9 imliveimage results. This strategy establishes a new paradigm of fluorescent nanosensors for selective and specific tumor imaging.
Generation of Poly and Micellar Complexes for Nucleic Acid Transport - A Reflection on Their Endosomal Escape
2023, Controlled Release Journal
In the field of nucleic acid delivery, there has long been skepticism as to whether or not polycationic drug delivery systems would make it into clinical practice. However, with the disclosure of patents on polyethyleneimine-based RNA transporters by leaders in the field of nucleic acid therapeutics such as BioNTech SE, and with clinical trials moving beyond Phase I trials, this reluctance appears to be diminishing. As one of the most distinguishing properties of polymer-based arrays, exceptional adjustability can be both a blessing and a curse. Knowing the setscrews and how they affect drug carrier performance gives the formulation scientist committed to their development a head start. Here we provide the reader with a toolbox - a toolbox intended to advise and assist the developer in designing a state-of-the-art poly or micellar system for the delivery of therapeutic nucleic acids; specifically, to manipulate the vector for maximum endosomal escape performance with minimal toxicity. So, after briefly outlining the constraints of polymer vector design, let's dive into the topic of endosomal delivery. Not only will we discuss the most recent findings about the endolysosomal compartment, but we will also present several hypotheses and mechanisms that facilitate endosomal escape from polyplex systems. Finally, we will combine the different facets presented in the previous chapters with the basic building blocks of polymer vector design and evaluate the advantages and disadvantages. Throughout the article, special emphasis is placed on cellular specificities, not only as an additional barrier, but also as inspiration for how these cell-specific properties can be exploited.
Non-modulating therapies: developing a therapy for every CF patient
2022, Thoracic Medicine Clinics
Nanoparticle-mediated gene therapy as a new strategy for the treatment of retinoblastoma
2022, Colloids and B Surfaces: Biointerfaces
Over the past two decades, nanoparticle delivery systems have revolutionized cancer treatment, achieving targeted delivery, enhanced bioavailability, and an improved toxicity profile. The growing interest in nanotechnology for the treatment of cancer stems from the unique physicochemical properties of nanoparticles (e.g. small size, surface properties,etc.). Indeed, several anti-cancer drugs can now be effectively delivered through nano-delivery systems. However, the application of such delivery systems in the field of gene therapy is still in its infancy. Furthermore, the treatment of retinoblastoma (RB), an aggressive childhood eye cancer, is a major concern in developing countries due to the late diagnosis of this type of cancer. While delivery strategies based on adeno-associated viruses remain the mainstay of the gene delivery method due to their high efficiency, other delivery systems such as e.g. g., non-viral nanoparticles (NPs), have been developed as alternative therapeutic modalities. Indeed, various nanoparticle formulations such as lipid-based nanoparticles, polymer nanoparticles, and gold nanoparticles have shown improved gene delivery efficiency in retinal diseases. This review article focuses on nanoparticle-mediated gene therapy approaches in the treatment of RB and highlights the attempts made to develop improved formulations for the treatment of RB. We describe the current state of nanoparticles as a vehicle for gene delivery and cover the future prospects of this exciting area of research. We also discuss the achievements, challenges and opportunities of nanomedicine to treat RB and mention new technical approaches that leverage our growing understanding of tumor biology and NP uptake mechanisms to design more effective nanotherapeutics for patients with RB.
Enhanced antitumor efficacy against hepatocellular carcinoma by harmine-loaded liposomes targeting mitochondrial and legume response
2022, Journal of Drug Delivery Science and Technology
Harmine (HM) is an alkaloid found in the seeds ofPagan HarmalaL., which has antimalarial and anticancer effects. However, its low bioavailability and toxic side effects have limited its clinical applications. This study used a novel mitochondrial targeting peptide (KA) to modify the HM-loaded liposome (KA-HM-LPS) by the pre-insertion method. The KA peptide consists of a mitochondrial targeting sequence (KLA) and a legume cleavable sequence (AAN). KA-HM-LPS had a small particle size of 246.2 ± 5.482 nm, a polydispersion index of 0.275 ± 0.038, a zeta potential of -15.5 ± 0.687 mV, and an encapsulation efficiency of 93.8% ± 0.96%. The drug release experiment showed that KA-HM-LPS was released more slowly than free HM in PBS buffer (pH 7.4). The kit-8 cell count assay and intracellular uptake experiments showed that KA-HM-LPS had greater cytotoxicity and mitochondrial targeting efficiency than HM-LPS in SK-Hep-1 overexpressing LGMN cells. The fluorescence observations of tetramethylrhodamine ethyl ester suggested that KA-HM-LPS had a reduced mitochondrial membrane potential. The tumor growth inhibition rate of KA-HM-LPS in vivo was as high as 67.74% ± 15.20%, far exceeding that of free HM-LPS (33.12% ± 8.96%). In addition, hematoxylin and eosin staining and blood compatibility assays showed that KA-HM-LPS had excellent biocompatibility and hemocompatibility. Therefore, KA-HM-LPS showed leguminous responsiveness and mitochondrial targeting and may represent a safe and efficient therapeutic strategy for hepatocellular carcinoma.
Recent advances in versatile inverse lyotropic liquid crystals
2022, Controlled Release Journal
Due to rapid and significant advances in advanced materials and life sciences, nanotechnology is gaining increasing popularity. Among the numerous biomimetic transporters, inverse lyotropic liquid crystals are known for their unique properties. Due to their complicated topologies, these carriers allow the inclusion of molecules with different properties. Furthermore, the versatile symmetries of inverse LCNPs (lyotropic crystalline nanoparticles) and their aggregated phases allow their application in a variety of fields including drug delivery, food, cosmetics, materials science, etc. In this review, a detailed summary, discussion, and perspective on inverse lyotropic liquid crystals is provided.
Featured Articles (6)
Research Article
Dual-response polyplexes with improved disassembly and endosomal escape for efficient siRNA delivery
Biomaterialien, Band 162, 2018, S. 47-59
Although extracellular barriers to siRNA delivery have been overcome through the use of advanced nanoparticle delivery systems, key intracellular barriers after internalization, including efficient siRNA degradation and endosomal escape, still remain a challenge. To address these issues, we designed a unique pH- and redox-responsive polyplex delivery system based on mPEG-b-PLA-PHis-ssPEI1.8k copolymer composed of a pH-responsive PEG-b -PLA-PHis copolymer ( Mw 5k) and a branched PEI (Mw 1.8k) linked to a redox cleavable disulfide bond. The copolymer showed excellent siRNA complexation and protection against endogenous substances at the relatively low N/P ratio of 6. The release of siRNA from the polyplexes (N/P 6) increased significantly from 13.62% to 58.67% under conditions simulating the endosomal microenvironment. The fluorescence resonance energy transfer (FRET) assay also showed a higher level of siRNA disassembly from the copolymer. Accelerated release of siRNA from polyplexes was significantly restricted when the N/P ratio was increased above 10 due to increased electrostatic interactions. Efficient endosomal escape of siRNA after internalization was confirmed by confocal microscopy, which was attributed to membrane destabilization of cleaved PEI strands, the “proton sponge effect” of PHis and PEI, and the relatively small size after disassembly. Increased disassembly and endosomal escape were elucidated as the main cause of polyplexes (N/P 6) showing more efficient Bcl-2 silencing (85.45%) than polyplexes with higher N/P ratios (N/P 10 and 15) . In vivo results further demonstrated that delivery of siBcl-2 by polyplexes (N/P 6) significantly inhibited MCF-7 breast tumor growth compared to its counterparts. Incorporation of non-electrical convertible interactions in equilibrium with electrostatic interactions in siRNA complexes proved to be an effective strategy to achieve efficient disassembly of stable polyplexes. Furthermore, polyplexes endowed with improved disassembly and endosomal escape offer a potential new way to address the intracellular delivery bottleneck for siRNA delivery.
Research Article
pH-responsive cationic liposome for endosomal escape-mediated drug delivery
Colloids and B Surfaces: Biointerfaces, Volume 188, 2020, Article 110804
Endosomal nanoparticle degradation is one of the main biological barriers associated with the drug delivery system. Nanoparticles are internalized into the cell via various endocytic pathways, where they are first delivered to early endosomes, which mature into the late endosome and lysosome. During this journey, the NP encounters a hostile chemical environment that leads to degradation of the NP and its contents. This process is collectively referred to as the intracellular defense against foreign substances. Therefore, to avoid this degrading fate, the endosomal escape technique has been studied for membrane fusion or membrane destabilization mechanisms. However, these methods are limited in application due to non-specific membrane fusion. To overcome this limitation, we designed a pH-responsive liposome composed of 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol hydrochloride (DC-liposome) in which only the cationic nitrogen and ammonium moiety occupies ~ 2.5% of the molecule. Such a small percentage of the cationic fraction is sufficient to exhibit pH responsive properties while maintaining DC liposome biocompatibility. DC-liposomes showed pH-dependent cationic properties due to protonation of the DC moiety at acidic pH. The fluorescence-based experiment confirmed the pH-dependent Fusogenic properties of DC liposomes. Furthermore, the endosomal colocalization study revealed a greater localization of DC-liposomes in the early endosome compared to the late endosome, suggesting a possible endosomal escape. Enhanced cationic and fusogenic properties of DC liposomes at acidic pH may mediate membrane fusion with the anionic endosomal membrane through electrostatic interaction, causing endosomal leakage. Furthermore, DC-liposome loaded with doxorubicin showed greater cytotoxicity than free doxorubicin, further supporting our endosomal escape. These results indicate the potential of DC liposomes to disrupt endosomal barriers to increase therapeutic efficacy, thereby guiding us in design considerations in the field of stimulus-responsive delivery devices.
Research Article
Endosomal escape cell-penetrating peptides significantly enhance the pharmacological potency and CNS activity of systemically administered antisense oligonucleotides
International Journal of Pharmaceutics, Band 599, 2021, Artikel 120398
Antisense oligonucleotides (ASOs) are a new class of gene-specific therapies for diseases associated with the central nervous system (CNS). However, the transport of ASO across the blood-brain barrier (BBB) to CNS target cells remains a major challenge. Since ASOs are taken up at the cerebral capillary endothelial cell interface primarily through endosomal pathways, entrapment in the endosomal compartment is a major obstacle to the efficient delivery of ASOs to the CNS. Therefore, we evaluated the efficacy of a panel of cell-penetrating peptides (CPPs) that target multiple endosomal escape domains for the intracellular delivery, endosomal delivery, and antisense activity of the FDA-approved spinraza (Nusinersen), an ASO for the treatment of muscle wasting. spinal cord (AME). We identified a CPP, HA2-ApoE(131–150), which, when conjugated with nusinersen, exhibited an efficient endosomal escape capability and significantly increased the level of complete functional survival of the motoneuron 2 mRNA (SMN2) in fibroblasts derived from SMA patients. Treatment ofSMN2Transgenic adult mice with this CPP-PMO conjugate resulted in a significant increase in complete contentSMN2in the brain and spinal cord. This work provides proof of principle that integration of endosomal escape domains with CPPs allows for enhanced cytosolic delivery of ASOs and, more importantly, improves the efficiency of BBB permeability and CNS activity of systemically administered ASOs.
Research Article
Brief update on nanodrug endocytosis
Advanced Drug Delivery Reviews, Band 144, 2019, S. 90-111
The complexity of nanoscale interactions between biomaterials and cells has limited the realization of the ultimate vision of nanotechnology in diagnostics and therapy. Therefore, considerable effort has been expended to advance our understanding of the biophysical interactions of myriad nanoparticles. Endocytosis with nanomedicine has attracted great interest in the last decade. Here, we highlight the pervasive barriers to efficient intracellular delivery of nanoparticles and current advances and strategies employed to break through these barriers. We are also introducing new barriers that have been largely overlooked, such as the glycocalyx and macromolecular clumping. In addition, we draw attention to possible complications arising from disruption of the newly discovered functions of lysosomes. New strategies are being explored to exploit the intracellular defects inherent in disease states to improve the delivery and use of exosomes for bioanalytical and drug delivery. Additionally, we discuss advances in imaging techniques such as electron microscopy, high-resolution fluorescence microscopy, and single-particle tracking, which have significantly contributed to our growing understanding of intracellular pathways and nanoparticle transport. Finally, we endorse the push for a more intravital analysis of nanoparticle transport phenomena using the variety of techniques at our disposal. Unraveling the underlying mechanisms that govern cellular barriers to nanoparticle delivery and biological interactions will guide innovations that can break through these barriers.
Research Article
The Proton Sponge Hypothesis: Fable or Fact?
European Journal of Pharmaceutics and Biopharmaceutics, Band 129, 2018, S. 184-190
In non-viral gene therapy, cationic polymers and lipids are commonly used to encapsulate macromolecular therapeutics in nanoparticles. Many biological barriers must be overcome on its journey to get the cargo to its intended intracellular destination. One of the main bottlenecks for efficient transfection is the endosomal barrier, as nanoparticles generally remain trapped in endosomes and are transported to lysosomes, where the cargo is broken down. The proton sponge hypothesis was introduced for cationic polymers in the late 1990s to explain their endosomal escape properties. However, due to many conflicting reports, no consensus on the validity of this hypothesis has been reached in the scientific community to date. Here we review the sometimes conflicting reports that have been published about the proton sponge hypothesis. We also discuss membrane destabilization and polymer swelling as additional factors that may affect endosomal escape from polyplexes. Based on the most important publications on the subject, we want to build a consensus on the role of the proton sponge hypothesis in endosomal escape.
Research Article
Polymeric nucleic acid delivery vehicle
Advanced Drug Delivery Reviews, Band 156, 2020, S. 119-132
Polymeric carriers are versatile tools for delivering therapeutic genes. Many polymers - when mounted on vehicles with nucleic acids - can protect cargo from degradation and releaseliveand facilitate its transport to intracellular compartments. Design options in polymer synthesis result in a comprehensive range of molecules and resulting vehicle formulations. These properties can be manipulated to achieve a stronger association with nucleic acid and cell loading, enhanced endosomal escape, or sustained delivery, depending on the application. Here we describe current approaches to using polymers and related strategies for gene delivery in preclinical and clinical applications. Polymeric vehicles that deliver genetic material have already reached important therapeutic parametersin vitroand in animal models. In our view, with pre-clinical trials that mimic those bestliveEnvironment, improved strategies for target specificity, and scalable techniques for polymer synthesis will further enhance the impact of this therapeutic approach.
(Video) Lunch & Learn: Intro to Viral Vectors
© 2021 Published by Elsevier B.V.