151
|
Zhang R, Nie T, Fang Y, Huang H, Wu J. Poly(disulfide)s: From Synthesis to Drug Delivery. Biomacromolecules 2021; 23:1-19. [PMID: 34874705 DOI: 10.1021/acs.biomac.1c01210] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bioresponsive polymers have been widely used in drug delivery because of their degradability. For example, poly(disulfide)s with repeating disulfide bonds in the main chain have attracted considerable research attention. The characteristics of the disulfide bonds, including their dynamic and reversible properties and their responsiveness to stimuli such as reductants, light, heat, and mechanical force, make them ideal platforms for on-demand drug delivery. This review introduces the synthesis methods and applications of poly(disulfide)s. Furthermore, the synthesis methods of poly(disulfide)s are classified on the basis of the monomers used: oxidative step-growth polymerization with dithiols, ring-opening polymerization with cyclic disulfides, and polymerization with linear disulfides. In addition, recent advances in poly(disulfide)s for the delivery of small-molecule or biomacromolecular drugs are discussed. Quantum-dot-loaded poly(disulfide) delivery systems for imaging are also included. This review provides an overview of the various design strategies employed in the construction of poly(disulfide) platforms to inspire new applications in the field of drug delivery.
Collapse
Affiliation(s)
- Ruhe Zhang
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Tianqi Nie
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yifen Fang
- Department of Cardiology, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
152
|
Leer K, Cinar G, Solomun JI, Martin L, Nischang I, Traeger A. Core-crosslinked, temperature- and pH-responsive micelles: design, physicochemical characterization, and gene delivery application. NANOSCALE 2021; 13:19412-19429. [PMID: 34591061 DOI: 10.1039/d1nr04223h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Stimuli-responsive block copolymer micelles can provide tailored properties for the efficient delivery of genetic material. In particular, temperature- and pH-responsive materials are of interest, since their physicochemical properties can be easily tailored to meet the requirements for successful gene delivery. Within this study, a stimuli-responsive micelle system for gene delivery was designed based on a diblock copolymer consisting of poly(N,N-diethylacrylamide) (PDEAm) as a temperature-responsive segment combined with poly(aminoethyl acrylamide) (PAEAm) as a pH-responsive, cationic segment. Upon temperature increase, the PDEAm block becomes hydrophobic due to its lower critical solution temperature (LCST), leading to micelle formation. Furthermore, the monomer 2-(pyridin-2-yldisulfanyl)ethyl acrylate (PDSAc) was incorporated into the temperature-responsive PDEAm building block enabling disulfide crosslinking of the formed micelle core to stabilize its structure regardless of temperature and dilution. The cloud points of the PDEAm block and the diblock copolymer were investigated by turbidimetry and fluorescence spectroscopy. The temperature-dependent formation of micelles was analyzed by dynamic light scattering (DLS) and elucidated in detail by an analytical ultracentrifuge (AUC), which provided detailed insights into the solution dynamics between polymers and assembled micelles as a function of temperature. Finally, the micelles were investigated for their applicability as gene delivery vectors by evaluation of cytotoxicity, pDNA binding, and transfection efficiency using HEK293T cells. The investigations showed that core-crosslinking resulted in a 13-fold increase in observed transfection efficiency. Our study presents a comprehensive investigation from polymer synthesis to an in-depth physicochemical characterization and biological application of a crosslinked micelle system including stimuli-responsive behavior.
Collapse
Affiliation(s)
- Katharina Leer
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Gizem Cinar
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Jana I Solomun
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Liam Martin
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Ivo Nischang
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| |
Collapse
|
153
|
Alazzo A, Gumus N, Gurnani P, Stolnik S, Rahman R, Spriggs K, Alexander C. Investigating histidinylated highly branched poly(lysine) for siRNA delivery. J Mater Chem B 2021; 10:236-246. [PMID: 34852030 DOI: 10.1039/d1tb01793d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The temporary silencing of disease-associated genes utilising short interfering RNA (siRNA) is a potent and selective route for addressing a wide range of life limiting disorders. However, the few clinically approved siRNA therapies rely on lipid based formulations, which although potent, provide limited chemical space to tune the stability, efficacy and tissue selectivity. In this study, we investigated the role of molar mass and histidinylation for poly(lysine) based non-viral vectors, synthesised through a fully aqueous thermal condensation polymerisation. Formulation and in vitro studies revealed that higher molar mass derivatives yielded smaller polyplexes attributed to a greater affinity for siRNA at lower N/P ratios yielding greater transfection efficiency, albeit with some cytotoxicity. Histidinylation had a negligible effect on formulation size, yet imparted a moderate improvement in biocompatibility, but did not provide any meaningful improvement over silencing efficiency compared to non-histidinylated derivatives. This was attributed to a greater degree of cellular internalisation for non-histidinylated analogues, which was enhanced with the higher molar mass material.
Collapse
Affiliation(s)
- Ali Alazzo
- Department of Pharmaceutics, College of Pharmacy, University of Mosul, Mosul, Iraq.,Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Nurcan Gumus
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Pratik Gurnani
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Snjezana Stolnik
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Ruman Rahman
- BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Keith Spriggs
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
154
|
Transformable vesicles for cancer immunotherapy. Adv Drug Deliv Rev 2021; 179:113905. [PMID: 34331988 DOI: 10.1016/j.addr.2021.113905] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/22/2021] [Accepted: 07/24/2021] [Indexed: 02/06/2023]
Abstract
Immunotherapy that utilizes the human immune system to fight cancer represents a revolutionary method for cancer treatment. Immunotherapeutic agents that trigger the immune response should be carefully delivered to the desired site to maximize immunotherapy effectiveness and minimize side effects. Vesicles offer the possibility of encapsulating both hydrophilic and hydrophobic drugs and thus serve as a promising delivery tool. As multiple irreconcilable requirements exist at different transport stages, developing vesicles transformable in response to given stimuli is of great significance. In this review, we first introduced various vesicle types used for immunotherapy. Furthermore, the typical stimuli that trigger vesicle transformation and the usually generated transformation styles were described. Focusing on three aspects of antigen-presenting cell (APC)/T cell activation, tumor microenvironment (TME) amelioration, and immunogenic cell death (ICD)-induced immunotherapy, we reviewed recently reported transformable vesicles for tumor treatment. Finally, we put forward possible directions for future research and clinical translation.
Collapse
|
155
|
Anticancer potential of hexamolybdenum clusters [{Mo6I8}(L)6]2− (L = CF3COO− and C6F5COO−) incorporated into different nanoparticulate forms. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.117601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
156
|
Transmission Electron Microscopy as a Powerful Tool to Investigate the Interaction of Nanoparticles with Subcellular Structures. Int J Mol Sci 2021; 22:ijms222312789. [PMID: 34884592 PMCID: PMC8657944 DOI: 10.3390/ijms222312789] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/21/2021] [Accepted: 11/25/2021] [Indexed: 12/15/2022] Open
Abstract
Nanomedical research necessarily involves the study of the interactions between nanoparticulates and the biological environment. Transmission electron microscopy has proven to be a powerful tool in providing information about nanoparticle uptake, biodistribution and relationships with cell and tissue components, thanks to its high resolution. This article aims to overview the transmission electron microscopy techniques used to explore the impact of nanoconstructs on biological systems, highlighting the functional value of ultrastructural morphology, histochemistry and microanalysis as well as their fundamental contribution to the advancement of nanomedicine.
Collapse
|
157
|
VirPorters: Insights into the action of cationic and histidine-rich cell-penetrating peptides. Int J Pharm 2021; 611:121308. [PMID: 34800617 DOI: 10.1016/j.ijpharm.2021.121308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/30/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
The utilization of nanoparticles for the intracellular delivery of theranostic agents faces one substantial limitation. Sequestration in intracellular vesicles prevents them from reaching the desired location in the cytoplasm or nucleus to deliver their cargo. We investigated whether three different cell-penetrating peptides (CPPs), namely, octa-arginine R8, polyhistidine KH27K and histidine-rich LAH4, could promote cytosolic and/or nuclear transfer of unique model nanoparticles-pseudovirions derived from murine polyomavirus. Two types of CPP-modified pseudovirions that carry the luciferase reporter gene were created: VirPorters-IN with CPPs genetically attached to the capsid interior and VirPorters-EX with CPPs noncovalently associated with the capsid exterior. We tested their transduction ability by luciferase assay and monitored their presence in subcellular fractions. Our results confirmed the overall effect of CPPs on the intracellular destination of the particles and suggested that KH27K has the potential to improve the cytosolic release of pseudovirions. None of the VirPorters caused endomembrane damage detectable by the Galectin-3 assay. Remarkably, a noncovalent modification was required to promote high transduction of the reporter gene and cytosolic delivery of pseudovirions mediated by LAH4. Together, CPPs in different arrangements have demonstrated their potential to improve pseudovirion invasion into cells, and these findings could be useful for the development of other nanoparticle-based delivery systems.
Collapse
|
158
|
Machtakova M, Thérien-Aubin H, Landfester K. Polymer nano-systems for the encapsulation and delivery of active biomacromolecular therapeutic agents. Chem Soc Rev 2021; 51:128-152. [PMID: 34762084 DOI: 10.1039/d1cs00686j] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biomacromolecular therapeutic agents, particularly proteins, antigens, enzymes, and nucleic acids are emerging as powerful candidates for the treatment of various diseases and the development of the recent vaccine based on mRNA highlights the enormous potential of this class of drugs for future medical applications. However, biomacromolecular therapeutic agents present an enormous delivery challenge compared to traditional small molecules due to both a high molecular weight and a sensitive structure. Hence, the translation of their inherent pharmaceutical capacity into functional therapies is often hindered by the limited performance of conventional delivery vehicles. Polymer drug delivery systems are a modular solution able to address those issues. In this review, we discuss recent developments in the design of polymer delivery systems specifically tailored to the delivery challenges of biomacromolecular therapeutic agents. In the future, only in combination with a multifaceted and highly tunable delivery system, biomacromolecular therapeutic agents will realize their promising potential for the treatment of diseases and for the future of human health.
Collapse
Affiliation(s)
- Marina Machtakova
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Héloïse Thérien-Aubin
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. .,Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada.
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| |
Collapse
|
159
|
Han M, Beon J, Lee JY, Oh SS. Systematic Combination of Oligonucleotides and Synthetic Polymers for Advanced Therapeutic Applications. Macromol Res 2021; 29:665-680. [PMID: 34754286 PMCID: PMC8568687 DOI: 10.1007/s13233-021-9093-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/22/2021] [Accepted: 09/25/2021] [Indexed: 11/16/2022]
Abstract
The potential of oligonucleotides is exceptional in therapeutics because of their high safety, potency, and specificity compared to conventional therapeutic agents. However, many obstacles, such as low in vivo stability and poor cellular uptake, have hampered their clinical success. Use of polymeric carriers can be an effective approach for overcoming the biological barriers and thereby maximizing the therapeutic efficacy of the oligonucleotides due to the availability of highly tunable synthesis and functional modification of various polymers. As loaded in the polymeric carriers, the therapeutic oligonucleotides, such as antisense oligonucleotides, small interfering RNAs, microRNAs, and even messenger RNAs, become nuclease-resistant by bypassing renal filtration and can be efficiently internalized into disease cells. In this review, we introduced a variety of systematic combinations between the therapeutic oligonucleotides and the synthetic polymers, including the uses of highly functionalized polymers responding to a wide range of endogenous and exogenous stimuli for spatiotemporal control of oligonucleotide release. We also presented intriguing characteristics of oligonucleotides suitable for targeted therapy and immunotherapy, which can be fully supported by versatile polymeric carriers.
Collapse
Affiliation(s)
- Moohyun Han
- Department of Materials Science and Engineering, Pohang University of Science Technology (POSTECH), Pohang, Gyeongbuk, 37673 Korea
| | - Jiyun Beon
- Department of Materials Science and Engineering, Pohang University of Science Technology (POSTECH), Pohang, Gyeongbuk, 37673 Korea
| | - Ju Young Lee
- Research Center for Bio-based Chemistry, Korea Research Institute of Chemical Technology (KRICT), Ulsan, 44429 Korea
| | - Seung Soo Oh
- Department of Materials Science and Engineering, Pohang University of Science Technology (POSTECH), Pohang, Gyeongbuk, 37673 Korea
| |
Collapse
|
160
|
Chaudhary N, Weissman D, Whitehead KA. mRNA vaccines for infectious diseases: principles, delivery and clinical translation. Nat Rev Drug Discov 2021; 20:817-838. [PMID: 34433919 PMCID: PMC8386155 DOI: 10.1038/s41573-021-00283-5] [Citation(s) in RCA: 630] [Impact Index Per Article: 210.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
Over the past several decades, messenger RNA (mRNA) vaccines have progressed from a scepticism-inducing idea to clinical reality. In 2020, the COVID-19 pandemic catalysed the most rapid vaccine development in history, with mRNA vaccines at the forefront of those efforts. Although it is now clear that mRNA vaccines can rapidly and safely protect patients from infectious disease, additional research is required to optimize mRNA design, intracellular delivery and applications beyond SARS-CoV-2 prophylaxis. In this Review, we describe the technologies that underlie mRNA vaccines, with an emphasis on lipid nanoparticles and other non-viral delivery vehicles. We also overview the pipeline of mRNA vaccines against various infectious disease pathogens and discuss key questions for the future application of this breakthrough vaccine platform.
Collapse
Affiliation(s)
- Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
161
|
Marschall ALJ. Targeting the Inside of Cells with Biologicals: Chemicals as a Delivery Strategy. BioDrugs 2021; 35:643-671. [PMID: 34705260 PMCID: PMC8548996 DOI: 10.1007/s40259-021-00500-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Delivering macromolecules into the cytosol or nucleus is possible in vitro for DNA, RNA and proteins, but translation for clinical use has been limited. Therapeutic delivery of macromolecules into cells requires overcoming substantially higher barriers compared to the use of small molecule drugs or proteins in the extracellular space. Breakthroughs like DNA delivery for approved gene therapies and RNA delivery for silencing of genes (patisiran, ONPATTRO®, Alnylam Pharmaceuticals, Cambridge, MA, USA) or for vaccination such as the RNA-based coronavirus disease 2019 (COVID-19) vaccines demonstrated the feasibility of using macromolecules inside cells for therapy. Chemical carriers are part of the reason why these novel RNA-based therapeutics possess sufficient efficacy for their clinical application. A clear advantage of synthetic chemicals as carriers for macromolecule delivery is their favourable properties with respect to production and storage compared to more bioinspired vehicles like viral vectors or more complex drugs like cellular therapies. If biologicals can be applied to intracellular targets, the druggable space is substantially broadened by circumventing the limited utility of small molecules for blocking protein–protein interactions and the limitation of protein-based drugs to the extracellular space. An in depth understanding of the macromolecular cargo types, carrier types and the cell biology of delivery is crucial for optimal application and further development of biologicals inside cells. Basic mechanistic principles of the molecular and cell biological aspects of cytosolic/nuclear delivery of macromolecules, with particular consideration of protein delivery, are reviewed here. The efficiency of macromolecule delivery and applications in research and therapy are highlighted.
Collapse
Affiliation(s)
- Andrea L J Marschall
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Brunswick, Germany.
| |
Collapse
|
162
|
Klemm P, Behnke M, Solomun JI, Bonduelle C, Lecommandoux S, Traeger A, Schubert S. Self-assembled PEGylated amphiphilic polypeptides for gene transfection. J Mater Chem B 2021; 9:8224-8236. [PMID: 34643200 DOI: 10.1039/d1tb01495a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In the present study, three biodegradable block copolymers composed of a poly(ethylene glycol) block and a copolypeptide block with varying compositions of cationic L-lysine (L-Lys) and hydrophobic benzyl-L-glutamate (Bzl-L-Glu) were designed for gene delivery applications. The polypeptides were synthesized by ring opening polymerization (ROP) and after orthogonal deprotection of Boc-L-Lys side chains, the polymer exhibited an amphiphilic character. To bind or encapsulate plasmid DNA (pDNA), different formulations were investigated: a nanoprecipitation and an emulsion technique using various organic solvents as well as an aqueous pH-controlled formulation method. The complex and nanoparticle (NP) formations were monitored by dynamic light scattering (DLS), and pDNA interaction was shown by gel electrophoresis and subsequent controlled release with heparin. The polypeptides were further tested for their cytotoxicity as well as biodegradability. The complexes and NPs presenting the most promising size distributions and pDNA binding ability were subsequently evaluated for their transfection efficiency in HEK293T cells. The highest transfection efficiencies were obtained with an aqueous formulation of the polypeptide containing the highest L-Lys content and lowest proportion of hydrophobic, helical structures (P1*), which is therefore a promising candidate for efficient gene delivery by biodegradable gene delivery vectors.
Collapse
Affiliation(s)
- Paul Klemm
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.,Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany.
| | - Mira Behnke
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.,Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany.
| | - Jana I Solomun
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.,Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany.
| | - Colin Bonduelle
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600, Pessac, France
| | | | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany.,Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany.
| | - Stephanie Schubert
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany. .,Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, Friedrich Schiller University Jena, Lessingstrasse 8, 07743 Jena, Germany
| |
Collapse
|
163
|
Shah S, Chougule MB, Kotha AK, Kashikar R, Godugu C, Raghuvanshi RS, Singh SB, Srivastava S. Nanomedicine based approaches for combating viral infections. J Control Release 2021; 338:80-104. [PMID: 34375690 PMCID: PMC8526416 DOI: 10.1016/j.jconrel.2021.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
Millions of people die each year from viral infections across the globe. There is an urgent need to overcome the existing gap and pitfalls of the current antiviral therapy which include increased dose and dosing frequency, bioavailability challenges, non-specificity, incidences of resistance and so on. These stumbling blocks could be effectively managed by the advent of nanomedicine. Current review emphasizes over an enhanced understanding of how different lipid, polymer and elemental based nanoformulations could be potentially and precisely used to bridle the said drawbacks in antiviral therapy. The dawn of nanotechnology meeting vaccine delivery, role of RNAi therapeutics in antiviral treatment regimen, various regulatory concerns towards clinical translation of nanomedicine along with current trends and implications including unexplored research avenues for advancing the current drug delivery have been discussed in detail.
Collapse
Affiliation(s)
- Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mahavir Bhupal Chougule
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA; Department Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Arun K Kotha
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA; Department Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Rama Kashikar
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA; Department Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Rajeev Singh Raghuvanshi
- Indian Pharmacopoeia Commission, Ministry of Health & Family Welfare, Government of India, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
164
|
Richter F, Leer K, Martin L, Mapfumo P, Solomun JI, Kuchenbrod MT, Hoeppener S, Brendel JC, Traeger A. The impact of anionic polymers on gene delivery: how composition and assembly help evading the toxicity-efficiency dilemma. J Nanobiotechnology 2021; 19:292. [PMID: 34579715 PMCID: PMC8477462 DOI: 10.1186/s12951-021-00994-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/10/2021] [Indexed: 02/02/2023] Open
Abstract
Cationic polymers have been widely studied for non-viral gene delivery due to their ability to bind genetic material and to interact with cellular membranes. However, their charged nature carries the risk of increased cytotoxicity and interaction with serum proteins, limiting their potential in vivo application. Therefore, hydrophilic or anionic shielding polymers are applied to counteract these effects. Herein, a series of micelle-forming and micelle-shielding polymers were synthesized via RAFT polymerization. The copolymer poly[(n-butyl acrylate)-b-(2-(dimethyl amino)ethyl acrylamide)] (P(nBA-b-DMAEAm)) was assembled into cationic micelles and different shielding polymers were applied, i.e., poly(acrylic acid) (PAA), poly(4-acryloyl morpholine) (PNAM) or P(NAM-b-AA) block copolymer. These systems were compared to a triblock terpolymer micelle comprising PAA as the middle block. The assemblies were investigated regarding their morphology, interaction with pDNA, cytotoxicity, transfection efficiency, polyplex uptake and endosomal escape. The naked cationic micelle exhibited superior transfection efficiency, but increased cytotoxicity. The addition of shielding polymers led to reduced toxicity. In particular, the triblock terpolymer micelle convinced with high cell viability and no significant loss in efficiency. The highest shielding effect was achieved by layering micelles with P(NAM-b-AA) supporting the colloidal stability at neutral zeta potential and completely restoring cell viability while maintaining moderate transfection efficiencies. The high potential of this micelle-layer-combination for gene delivery was illustrated for the first time.
Collapse
Affiliation(s)
- Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Katharina Leer
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Liam Martin
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Prosper Mapfumo
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jana I Solomun
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Maren T Kuchenbrod
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany.
| |
Collapse
|
165
|
Lv S, Sylvestre M, Prossnitz AN, Yang LF, Pun SH. Design of Polymeric Carriers for Intracellular Peptide Delivery in Oncology Applications. Chem Rev 2021; 121:11653-11698. [PMID: 33566580 DOI: 10.1021/acs.chemrev.0c00963] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent decades, peptides, which can possess high potency, excellent selectivity, and low toxicity, have emerged as promising therapeutics for cancer applications. Combined with an improved understanding of tumor biology and immuno-oncology, peptides have demonstrated robust antitumor efficacy in preclinical tumor models. However, the translation of peptides with intracellular targets into clinical therapies has been severely hindered by limitations in their intrinsic structure, such as low systemic stability, rapid clearance, and poor membrane permeability, that impede intracellular delivery. In this Review, we summarize recent advances in polymer-mediated intracellular delivery of peptides for cancer therapy, including both therapeutic peptides and peptide antigens. We highlight strategies to engineer polymeric materials to increase peptide delivery efficiency, especially cytosolic delivery, which plays a crucial role in potentiating peptide-based therapies. Finally, we discuss future opportunities for peptides in cancer treatment, with an emphasis on the design of polymer nanocarriers for optimized peptide delivery.
Collapse
Affiliation(s)
| | | | - Alexander N Prossnitz
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | | | | |
Collapse
|
166
|
Chung SL, Yee MSL, Hii LW, Lim WM, Ho MY, Khiew PS, Leong CO. Advances in Nanomaterials Used in Co-Delivery of siRNA and Small Molecule Drugs for Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2467. [PMID: 34684908 PMCID: PMC8540385 DOI: 10.3390/nano11102467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022]
Abstract
Recent advancements in nanotechnology have improved our understanding of cancer treatment and allowed the opportunity to develop novel delivery systems for cancer therapy. The biological complexities of cancer and tumour micro-environments have been shown to be highly challenging when treated with a single therapeutic approach. Current co-delivery systems which involve delivering small molecule drugs and short-interfering RNA (siRNA) have demonstrated the potential of effective suppression of tumour growth. It is worth noting that a considerable number of studies have demonstrated the synergistic effect of co-delivery systems combining siRNA and small molecule drugs, with promising results when compared to single-drug approaches. This review focuses on the recent advances in co-delivery of siRNA and small molecule drugs. The co-delivery systems are categorized based on the material classes of drug carriers. We discuss the critical properties of materials that enable co-delivery of two distinct anti-tumour agents with different properties. Key examples of co-delivery of drug/siRNA from the recent literature are highlighted and discussed. We summarize the current and emerging issues in this rapidly changing field of research in biomaterials for cancer treatments.
Collapse
Affiliation(s)
- Shei Li Chung
- Nanotechnology Research Group, Faculty of Science and Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih 43500, Selangor, Malaysia; (S.L.C.); (P.S.K.)
- Department of Mechanical, Materials & Manufacturing Engineering, Faculty of Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih 43500, Selangor, Malaysia
| | - Maxine Swee-Li Yee
- Nanotechnology Research Group, Faculty of Science and Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih 43500, Selangor, Malaysia; (S.L.C.); (P.S.K.)
| | - Ling-Wei Hii
- Center for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur 57000, Malaysia; (L.-W.H.); (W.-M.L.)
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Wei-Meng Lim
- Center for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur 57000, Malaysia; (L.-W.H.); (W.-M.L.)
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Mui Yen Ho
- Department of Materials Engineering, Faculty of Engineering and Technology, Tunku Abdul Rahman University College, Jalan Genting Kelang, Kuala Lumpur 53300, Malaysia;
- Centre of Advanced Materials, Faculty of Engineering and Technology, Tunku Abdul Rahman University College, Jalan Genting Kelang, Kuala Lumpur 53300, Malaysia
| | - Poi Sim Khiew
- Nanotechnology Research Group, Faculty of Science and Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih 43500, Selangor, Malaysia; (S.L.C.); (P.S.K.)
| | - Chee-Onn Leong
- Center for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur 57000, Malaysia; (L.-W.H.); (W.-M.L.)
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
167
|
Faizullin BA, Strelnik ID, Dayanova IR, Gerasimova TP, Kholin KV, Nizameev IR, Voloshina AD, Gubaidullin AT, Fedosimova SV, Mikhailov MA, Sokolov MN, Sibgatullina GV, Samigullin DV, Petrov KA, Karasik AA, Mustafina AR. Structure impact on photodynamic therapy and cellular contrasting functions of colloids constructed from dimeric Au(I) complex and hexamolybdenum clusters. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112355. [PMID: 34474903 DOI: 10.1016/j.msec.2021.112355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 01/11/2023]
Abstract
Electrostatically driven self-assembly of [Au2L2]2+ (L is cyclic PNNP ligand) with [{Mo6I8}(L')6]2- (L' = I-, CH3COO-) in aqueous solutions is introduced as facile route for combination of therapeutic and cellular contrasting functions within heterometallic colloids (Mo6-Au2). The nature of L' affects the size and aggregation behavior of crystalline Mo6-Au2 aggregates, which in turn affect the luminescence of the cluster units incorporated into Mo6-Au2 colloids. The spin trap facilitated electron spin resonance spectroscopy technique indicates that the level of ROS generated by Mo6-Au2 colloids is also affected by their size. Both (L' = I-, CH3COO-) Mo6-Au2 colloids undergo cell internalization, which is enhanced by their assembly with poly-DL-lysine (PL) for L' = CH3COO-, but remains unchanged for L' = I-. The colloids PL-Mo6-Au2 (L' = CH3COO-) are visualized as huge crystalline aggregates both outside and inside the cell cytoplasm by confocal microscopy imaging of the incubated cells, while the smaller sized (30-50 nm) PL-Mo6-Au2 (L' = I-) efficiently stain the cell nuclei. Quantitative colocalization analysis of PL-Mo6-Au2 (L' = CH3COO-) in lysosomal compartments points to the fast endo-lysosomal escape of the colloids followed by their intracellular aggregation. The cytotoxicity of PL-Mo6-Au2 differs from that of Mo6 and Au2 blocks, predominantly acting through apoptotic pathway. The photodynamic therapeutic effect of the PL-Mo6-Au2 colloids on the cancer cells correlates with their intracellular trafficking and aggregation.
Collapse
Affiliation(s)
- Bulat A Faizullin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation; Kazan (Volga region) Federal University, 18 Kremlyovskaya str., 420008 Kazan, Russian Federation.
| | - Igor D Strelnik
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Irina R Dayanova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Tatyana P Gerasimova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Kirill V Kholin
- Kazan National Research Technical University named after A.N. Tupolev - KAI, 10 K. Marx str., 420111 Kazan, Russian Federation
| | - Irek R Nizameev
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Alexandra D Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Aidar T Gubaidullin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Svetlana V Fedosimova
- Kazan (Volga region) Federal University, 18 Kremlyovskaya str., 420008 Kazan, Russian Federation
| | - Maxim A Mikhailov
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., 630090 Novosibirsk, Russian Federation
| | - Maxim N Sokolov
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., 630090 Novosibirsk, Russian Federation
| | - Guzel V Sibgatullina
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevski str., 420111 Kazan, Russian Federation
| | - Dmitry V Samigullin
- Kazan National Research Technical University named after A.N. Tupolev - KAI, 10 K. Marx str., 420111 Kazan, Russian Federation; Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevski str., 420111 Kazan, Russian Federation
| | - Konstantin A Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Andrey A Karasik
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Asiya R Mustafina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| |
Collapse
|
168
|
Lee MC, Seonwoo H, Jang KJ, Pandey S, Lim J, Park S, Kim JE, Choung YH, Garg P, Chung JH. Development of novel gene carrier using modified nano hydroxyapatite derived from equine bone for osteogenic differentiation of dental pulp stem cells. Bioact Mater 2021; 6:2742-2751. [PMID: 33665505 PMCID: PMC7895645 DOI: 10.1016/j.bioactmat.2021.01.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 12/15/2022] Open
Abstract
Hydroxyapatite (HA) is a representative substance that induces bone regeneration. Our research team extracted nanohydroxyapatite (EH) from natural resources, especially equine bones, and developed it as a molecular biological tool. Polyethylenimine (PEI) was used to coat the EH to develop a gene carrier. To verify that PEI is well coated in the EH, we first observed the morphology and dispersity of PEI-coated EH (pEH) by electron microscopy. The pEH particles were well distributed, while only the EH particles were not distributed and aggregated. Then, the existence of nitrogen elements of PEI on the surface of the pEH was confirmed by EDS, calcium concentration measurement and fourier transform infrared spectroscopy (FT-IR). Additionally, the pEH was confirmed to have a more positive charge than the 25 kD PEI by comparing the zeta potentials. As a result of pGL3 transfection, pEH was better able to transport genes to cells than 25 kD PEI. After verification as a gene carrier for pEH, we induced osteogenic differentiation of DPSCs by loading the BMP-2 gene in pEH (BMP-2/pEH) and delivering it to the cells. As a result, it was confirmed that osteogenic differentiation was promoted by showing that the expression of osteopontin (OPN), osteocalcin (OCN), and runt-related transcription factor 2 (RUNX2) was significantly increased in the group treated with BMP-2/pEH. In conclusion, we have not only developed a novel nonviral gene carrier that is better performing and less toxic than 25 kD PEI by modifying natural HA (the agricultural byproduct) but also proved that bone differentiation can be effectively promoted by delivering BMP-2 with pEH to stem cells.
Collapse
Affiliation(s)
- Myung Chul Lee
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hoon Seonwoo
- Department of Industrial Machinery Engineering, Sunchon National University, 315 Maegok-dong, Sunchon, 57922, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Sunchon National University, Suncheon, 57922, Republic of Korea
| | - Kyoung Je Jang
- Division of Agro-system Engineering, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828, Republic of Korea
- Institute of Agriculture & Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Shambhavi Pandey
- Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaewoon Lim
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sangbae Park
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae Eun Kim
- Department of Biosystems Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yun-Hoon Choung
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
- Ajou University Graduate School of Medicine, Bk21 Plus Research Center for Biomedical Sciences, Suwon, 16499, Republic of Korea
| | - Pankaj Garg
- Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jong Hoon Chung
- Department of Biosystems Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- BK21 Global Smart Farm Educational Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
169
|
Voronin DV, Abalymov AA, Svenskaya YI, Lomova MV. Key Points in Remote-Controlled Drug Delivery: From the Carrier Design to Clinical Trials. Int J Mol Sci 2021; 22:9149. [PMID: 34502059 PMCID: PMC8430748 DOI: 10.3390/ijms22179149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
The increased research activity aiming at improved delivery of pharmaceutical molecules indicates the expansion of the field. An efficient therapeutic delivery approach is based on the optimal choice of drug-carrying vehicle, successful targeting, and payload release enabling the site-specific accumulation of the therapeutic molecules. However, designing the formulation endowed with the targeting properties in vitro does not guarantee its selective delivery in vivo. The various biological barriers that the carrier encounters upon intravascular administration should be adequately addressed in its overall design to reduce the off-target effects and unwanted toxicity in vivo and thereby enhance the therapeutic efficacy of the payload. Here, we discuss the main parameters of remote-controlled drug delivery systems: (i) key principles of the carrier selection; (ii) the most significant physiological barriers and limitations associated with the drug delivery; (iii) major concepts for its targeting and cargo release stimulation by external stimuli in vivo. The clinical translation for drug delivery systems is also described along with the main challenges, key parameters, and examples of successfully translated drug delivery platforms. The essential steps on the way from drug delivery system design to clinical trials are summarized, arranged, and discussed.
Collapse
Affiliation(s)
- Denis V. Voronin
- Science Medical Center, Saratov State University, Astrakhanskaya St. 83, 410012 Saratov, Russia; (A.A.A.); (Y.I.S.); (M.V.L.)
- Department of Physical and Colloid Chemistry, National University of Oil and Gas “Gubkin University”, Leninsky Prospekt 65, 119991 Moscow, Russia
| | - Anatolii A. Abalymov
- Science Medical Center, Saratov State University, Astrakhanskaya St. 83, 410012 Saratov, Russia; (A.A.A.); (Y.I.S.); (M.V.L.)
| | - Yulia I. Svenskaya
- Science Medical Center, Saratov State University, Astrakhanskaya St. 83, 410012 Saratov, Russia; (A.A.A.); (Y.I.S.); (M.V.L.)
| | - Maria V. Lomova
- Science Medical Center, Saratov State University, Astrakhanskaya St. 83, 410012 Saratov, Russia; (A.A.A.); (Y.I.S.); (M.V.L.)
| |
Collapse
|
170
|
Hausig F, Sobotta FH, Richter F, Harz DO, Traeger A, Brendel JC. Correlation between Protonation of Tailor-Made Polypiperazines and Endosomal Escape for Cytosolic Protein Delivery. ACS APPLIED MATERIALS & INTERFACES 2021; 13:35233-35247. [PMID: 34283557 DOI: 10.1021/acsami.1c00829] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Responsive polymers, which become protonated at decreasing pH, are considered a milestone in the development of synthetic cell entry vectors. Exact correlations between their properties and their ability to escape the endosome, however, often remain elusive due to hydrophobic interactions or limitations in the design of water-soluble materials with suitable basicity. Here, we present a series of well-defined, hydrophilic polypiperazines, where systematic variation of the amino moiety facilitates an unprecedented fine-tuning of the basicity or pKa value within the physiologically relevant range (pH 6-7.4). Coincubation of HEK 293T cells with various probes, including small fluorophores or functioning proteins, revealed a rapid increase of endosomal release for polymers with pKa values above 6.5 or 7 in serum-free or serum-containing media, respectively. Similarly, cytotoxic effects became severe at increased pKa values (>7). Although the window for effective transport appears narrow, the discovered correlations offer a principal guideline for the design of effective polymers for endosomal escape.
Collapse
Affiliation(s)
- Franziska Hausig
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Fabian H Sobotta
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Dominic O Harz
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| |
Collapse
|
171
|
Fattal E, Fay F. Nanomedicine-based delivery strategies for nucleic acid gene inhibitors in inflammatory diseases. Adv Drug Deliv Rev 2021; 175:113809. [PMID: 34033819 DOI: 10.1016/j.addr.2021.05.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 02/07/2023]
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.
Collapse
|
172
|
Simms BL, Ji N, Chandrasiri I, Zia MF, Udemgba CS, Kaur R, Delcamp JH, Flynt A, Tan C, Watkins DL. Physicochemical properties and bio‐interfacial interactions of surface modified
PDLLA‐PAMAM
linear dendritic block copolymers. JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1002/pol.20210448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Briana L. Simms
- Department of Chemistry and Biochemistry University of Mississippi University Oxford Mississippi USA
| | - Nan Ji
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy University of Mississippi University Oxford Mississippi USA
| | - Indika Chandrasiri
- Department of Chemistry and Biochemistry University of Mississippi University Oxford Mississippi USA
| | - Mohammad Farid Zia
- Department of Biological Sciences The University of Southern Mississippi Hattiesburg Mississippi USA
| | - Chinwe S. Udemgba
- Department of Chemistry and Biochemistry University of Mississippi University Oxford Mississippi USA
| | - Ravinder Kaur
- Department of Chemistry and Biochemistry University of Mississippi University Oxford Mississippi USA
| | - Jared H. Delcamp
- Department of Chemistry and Biochemistry University of Mississippi University Oxford Mississippi USA
| | - Alex Flynt
- Department of Biological Sciences The University of Southern Mississippi Hattiesburg Mississippi USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy University of Mississippi University Oxford Mississippi USA
| | - Davita L. Watkins
- Department of Chemistry and Biochemistry University of Mississippi University Oxford Mississippi USA
| |
Collapse
|
173
|
Azevedo A, Farinha D, Geraldes C, Faneca H. Combining gene therapy with other therapeutic strategies and imaging agents for cancer theranostics. Int J Pharm 2021; 606:120905. [PMID: 34293466 DOI: 10.1016/j.ijpharm.2021.120905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023]
Abstract
Cancer is one of the most prevalent and deadly diseases in the world, to which conventional treatment options, such as chemotherapy and radiotherapy, have been applied to overcome the disease or used in a palliative manner to enhance the quality of life of the patient. However, there is an urgent need to develop new preventive and treatment strategies to overcome the limitations of the commonly used approaches. The field of cancer nanomedicine, and more recently the field of nanotheranostics, where imaging and therapeutic agents are combined in a single platform, provide new opportunities for the treatment and the diagnosis of cancer. This combination could bring us closer to a more personalized and cared-for therapy, in opposition to the conventional and standardized approaches. Gene therapy is a promising strategy for the treatment of cancer that requires a transport system to efficiently deliver the genetic material into the target cells. Hence, the main purpose of this work was to review recent findings and developments regarding theranostic nanosystems that incorporate both gene therapy and imaging agents for cancer treatment.
Collapse
Affiliation(s)
- Alexandro Azevedo
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Calçada Martim de Freitas, 3000-393 Coimbra, Portugal
| | - Dina Farinha
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Institute of Interdisciplinary Research (III), University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Carlos Geraldes
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Calçada Martim de Freitas, 3000-393 Coimbra, Portugal; Coimbra Chemistry Center, University of Coimbra, Rua Larga Largo D. Dinis, 3004-535 Coimbra, Portugal
| | - Henrique Faneca
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Institute of Interdisciplinary Research (III), University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal.
| |
Collapse
|
174
|
Tieu T, Wei Y, Cifuentes‐Rius A, Voelcker NH. Overcoming Barriers: Clinical Translation of siRNA Nanomedicines. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100108] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Terence Tieu
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
| | - Yingkai Wei
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Anna Cifuentes‐Rius
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Nicolas H. Voelcker
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
- Melbourne Centre for Nanofabrication 151 Wellington Road Victorian Node of the Australian National Fabrication Facility Clayton VIC 3168 Australia
| |
Collapse
|
175
|
Debnath K, Pal S, Jana NR. Chemically Designed Nanoscale Materials for Controlling Cellular Processes. Acc Chem Res 2021; 54:2916-2927. [PMID: 34232016 DOI: 10.1021/acs.accounts.1c00215] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nanoparticles are widely used in various biomedical applications as drug delivery carriers, imaging probes, single-molecule tracking/detection probes, artificial chaperones for inhibiting protein aggregation, and photodynamic therapy materials. One key parameter of these applications is the ability of the nanoparticles to enter into the cell cytoplasm, target different subcellular compartments, and control intracellular processes. This is particularly the case because nanoparticles are designed to interact with subcellular components for the required biomedical performance. However, cells are protected from their surroundings by the cell membrane, which exerts strict control over entry of foreign materials. Thus, nanoparticles need to be designed appropriately so that they can readily cross the cell membrane, target subcellular compartments, and control intracellular processes.In the past few decades there have been great advancements in understanding the principles of cellular uptake of foreign materials. In particular, it has been shown that internalization of foreign materials (small molecules, macromolecules, nanoparticles) is size-dependent: endocytotic uptake of materials requires sizes greater than 10 nm, and materials with sizes of 10-100 nm usually enter into cells by energy-dependent endocytosis via biomembrane-coated vesicles. Direct access to the cytosol is limited to very specific conditions, and endosomal escape of material appears to be the most practical approach for intracellular processing.In this Account, we describe how cellular uptake and intracellular processing of nanoscale materials can be controlled by appropriate design of size and surface chemistry. We first describe the cell membrane structure and principles of cellular uptake of foreign materials followed by their subcellular trafficking. Next, we discuss the designed surface chemistry of a 5-50 nm particle that offers preferential lipid-raft/caveolae-mediated endocytosis over clathrin-mediated endocytosis with minimum endosomal/lysosomal trafficking or energy-independent direct cell membrane translocation (without endocytosis) followed by cytosolic delivery without endosomal/lysosomal trafficking. In particular, we emphasize that the zwitterionic-lipophilic surface property of a nanoparticle offers preferential interaction with the lipid raft region of the cell membrane followed by lipid raft uptake, whereas a lower number of affinity biomolecules (<25) on the nanoparticle surface offers caveolae/lipid-raft uptake, while an arginine/guanidinium-terminated surface along with a size of <10 nm offers direct cell membrane translocation. Finally, we discuss how nanoprobes can be designed by adapting these surface chemistry and size preference principles so that they can readily enter into the cell, label different subcellular compartments, and control intracellular processes such as trafficking kinetics, exocytosis, autophagy, amyloid aggregation, and clearance of toxic amyloid aggregates. The Account ends with a Conclusions and Outlook where we discuss a vision for the development of subcellular targeting nanodrugs and imaging nanoprobes by adapting to these surface chemistry principles.
Collapse
Affiliation(s)
- Koushik Debnath
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Suman Pal
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Nikhil R. Jana
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| |
Collapse
|
176
|
Hu Y, Zhu Y, Sutherland ND, Wilson DR, Pang M, Liu E, Staub JR, Berlinicke CA, Zack DJ, Green JJ, Reddy SK, Mao HQ. Size-Controlled and Shelf-Stable DNA Particles for Production of Lentiviral Vectors. NANO LETTERS 2021; 21:5697-5705. [PMID: 34228937 PMCID: PMC8283758 DOI: 10.1021/acs.nanolett.1c01421] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/03/2021] [Indexed: 06/13/2023]
Abstract
Polyelectrolyte complex particles assembled from plasmid DNA (pDNA) and poly(ethylenimine) (PEI) have been widely used to produce lentiviral vectors (LVVs) for gene therapy. The current batch-mode preparation for pDNA/PEI particles presents limited reproducibility in large-scale LVV manufacturing processes, leading to challenges in tightly controlling particle stability, transfection outcomes, and LVV production yield. Here we identified the size of pDNA/PEI particles as a key determinant for a high transfection efficiency with an optimal size of 400-500 nm, due to a cellular-uptake-related mechanism. We developed a kinetics-based approach to assemble size-controlled and shelf-stable particles using preassembled nanoparticles as building blocks and demonstrated production scalability on a scale of at least 100 mL. The preservation of colloidal stability and transfection efficiency was benchmarked against particles generated using an industry standard protocol. This particle manufacturing method effectively streamlines the viral manufacturing process and improves the production quality and consistency.
Collapse
Affiliation(s)
- Yizong Hu
- Department
of Biomedical Engineering, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
- Translational
Tissue Engineering Center, Johns Hopkins
University School of Medicine Baltimore, Maryland 21205, United States
- Institute
for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Yining Zhu
- Department
of Biomedical Engineering, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
- Translational
Tissue Engineering Center, Johns Hopkins
University School of Medicine Baltimore, Maryland 21205, United States
- Institute
for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | | | - David R. Wilson
- Department
of Biomedical Engineering, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
- Translational
Tissue Engineering Center, Johns Hopkins
University School of Medicine Baltimore, Maryland 21205, United States
- Institute
for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Marion Pang
- Department
of Biomedical Engineering, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
- Institute
for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Ester Liu
- Institute
for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21287, United States
- Department
of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Jacob R. Staub
- Institute
for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Cynthia A. Berlinicke
- Department
of Ophthalmology, Johns Hopkins University
School of Medicine Baltimore, Maryland 21205, United States
| | - Donald J. Zack
- Department
of Ophthalmology, Johns Hopkins University
School of Medicine Baltimore, Maryland 21205, United States
| | - Jordan J. Green
- Department
of Biomedical Engineering, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
- Translational
Tissue Engineering Center, Johns Hopkins
University School of Medicine Baltimore, Maryland 21205, United States
- Institute
for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21287, United States
- Department
of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21287, United States
- Department
of Ophthalmology, Johns Hopkins University
School of Medicine Baltimore, Maryland 21205, United States
- Department
of Materials Science and Engineering, Johns
Hopkins University, Baltimore, Maryland 21287, United States
| | - Sashank K. Reddy
- Department
of Plastic and Reconstructive Surgery, Johns
Hopkins University School of Medicine Baltimore, Maryland 21205, United States
| | - Hai-Quan Mao
- Department
of Biomedical Engineering, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
- Translational
Tissue Engineering Center, Johns Hopkins
University School of Medicine Baltimore, Maryland 21205, United States
- Institute
for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21287, United States
- Department
of Materials Science and Engineering, Johns
Hopkins University, Baltimore, Maryland 21287, United States
| |
Collapse
|
177
|
Xue J, Liu J, Yong J, Liang K. Biomedical Applications of Metal–Organic Frameworks at the Subcellular Level. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Jueyi Xue
- School of Chemical Engineering and Australian Centre for NanoMedicine University of New South Wales Sydney NSW 2052 Australia
| | - Jian Liu
- School of Chemical Engineering and Australian Centre for NanoMedicine University of New South Wales Sydney NSW 2052 Australia
| | - Joel Yong
- School of Chemical Engineering and Australian Centre for NanoMedicine University of New South Wales Sydney NSW 2052 Australia
| | - Kang Liang
- School of Chemical Engineering and Australian Centre for NanoMedicine University of New South Wales Sydney NSW 2052 Australia
- Graduate School of Biomedical Engineering University of New South Wales Sydney NSW 2052 Australia
| |
Collapse
|
178
|
Zheng M, Pan M, Zhang W, Lin H, Wu S, Lu C, Tang S, Liu D, Cai J. Poly(α-l-lysine)-based nanomaterials for versatile biomedical applications: Current advances and perspectives. Bioact Mater 2021; 6:1878-1909. [PMID: 33364529 PMCID: PMC7744653 DOI: 10.1016/j.bioactmat.2020.12.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 02/05/2023] Open
Abstract
Poly(α-l-lysine) (PLL) is a class of water-soluble, cationic biopolymer composed of α-l-lysine structural units. The previous decade witnessed tremendous progress in the synthesis and biomedical applications of PLL and its composites. PLL-based polymers and copolymers, till date, have been extensively explored in the contexts such as antibacterial agents, gene/drug/protein delivery systems, bio-sensing, bio-imaging, and tissue engineering. This review aims to summarize the recent advances in PLL-based nanomaterials in these biomedical fields over the last decade. The review first describes the synthesis of PLL and its derivatives, followed by the main text of their recent biomedical applications and translational studies. Finally, the challenges and perspectives of PLL-based nanomaterials in biomedical fields are addressed.
Collapse
Affiliation(s)
- Maochao Zheng
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Miao Pan
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Wancong Zhang
- The Second Affiliated Hospital of Shantou University Medical College, 69 Dongxiabei Road, Shantou, 515041, China
| | - Huanchang Lin
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Shenlang Wu
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Chao Lu
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Shijie Tang
- The Second Affiliated Hospital of Shantou University Medical College, 69 Dongxiabei Road, Shantou, 515041, China
| | - Daojun Liu
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL, 33620, USA
| |
Collapse
|
179
|
Liu Z, Wang S, Tapeinos C, Torrieri G, Känkänen V, El-Sayed N, Python A, Hirvonen JT, Santos HA. Non-viral nanoparticles for RNA interference: Principles of design and practical guidelines. Adv Drug Deliv Rev 2021; 174:576-612. [PMID: 34019958 DOI: 10.1016/j.addr.2021.05.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 02/08/2023]
Abstract
Ribonucleic acid interference (RNAi) is an innovative treatment strategy for a myriad of indications. Non-viral synthetic nanoparticles (NPs) have drawn extensive attention as vectors for RNAi due to their potential advantages, including improved safety, high delivery efficiency and economic feasibility. However, the complex natural process of RNAi and the susceptible nature of oligonucleotides render the NPs subject to particular design principles and requirements for practical fabrication. Here, we summarize the requirements and obstacles for fabricating non-viral nano-vectors for efficient RNAi. To address the delivery challenges, we discuss practical guidelines for materials selection and NP synthesis in order to maximize RNA encapsulation efficiency and protection against degradation, and to facilitate the cytosolic release of oligonucleotides. The current status of clinical translation of RNAi-based therapies and further perspectives for reducing the potential side effects are also reviewed.
Collapse
|
180
|
Cationic Dendrimers for siRNA Delivery: Computational Approaches for Characterization. Methods Mol Biol 2021. [PMID: 33928581 DOI: 10.1007/978-1-0716-1298-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Nowadays, computer simulations have been established as a fundamental tool in the design and development of new dendrimer-based nanocarriers for drug and gene delivery. Moreover, the level of detail contained in the information that can be gathered by performing atomistic-scale simulations cannot be obtained with any other available experimental technique. In this chapter we describe the main computational toolbox that can be exploited in the different stages of novel dendritic nanocarrier production-from the initial conception to the stage of biological intermolecular interactions.
Collapse
|
181
|
Hershberger KK, Gauger AJ, Bronstein LM. Utilizing Stimuli Responsive Linkages to Engineer and Enhance Polymer Nanoparticle-Based Drug Delivery Platforms. ACS APPLIED BIO MATERIALS 2021; 4:4720-4736. [PMID: 35007022 DOI: 10.1021/acsabm.1c00351] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The devastating nature of cancer continues to be one of the leading causes of death in the world. Chemotherapy is among the most common forms of cancer treatment but comes with a host of adverse effects caused by the therapeutic agents damaging healthy tissue and organs. To limit these side effects, scientists have been designing stimuli responsive drug delivery vessels for targeted release. This Review focuses on the incorporation of stimuli responsive linkages in targeted drug delivery systems to enhance therapeutic efficiency. These platforms are primarily employed to control the distribution of anticancer agents in the body to reduce the adverse side effects caused by their toxicities. We will outline how drug delivery vessels are constructed so that exposure to select environmental and external stimuli releases the enclosed drug only at the target site. Stimuli responsive components are integrated within drug delivery vessels in the form of cross-linkers, polymers, and surface modifications. The changes, these moieties undergo upon stimuli exposure, cascade into larger scale alterations to the platforms, resulting in complete disassembly, reversible morphological variations, and enhanced cellular uptake. The ability for these modes of delivery to be initiated exclusively under stimuli exposure allows for release of toxic therapeutic agents to be confined only to the affected area.
Collapse
Affiliation(s)
- Kian K Hershberger
- Indiana University, Department of Chemistry, Bloomington, 800 East Kirkwood Avenue, Indiana 47405, United States
| | - Andrew J Gauger
- Indiana University, Department of Chemistry, Bloomington, 800 East Kirkwood Avenue, Indiana 47405, United States
| | - Lyudmila M Bronstein
- Indiana University, Department of Chemistry, Bloomington, 800 East Kirkwood Avenue, Indiana 47405, United States.,A.N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, 28 Vavilov Street, Moscow, 119991 Russia.,King Abdulaziz University, Faculty of Science, Department of Physics, P.O. Box 80303, Jeddah 21589, Saudi Arabia
| |
Collapse
|
182
|
Dutta K, Das R, Medeiros J, Kanjilal P, Thayumanavan S. Charge-Conversion Strategies for Nucleic Acid Delivery. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2011103. [PMID: 35832306 PMCID: PMC9275120 DOI: 10.1002/adfm.202011103] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Indexed: 05/05/2023]
Abstract
Nucleic acids are now considered as one of the most potent therapeutic modalities, as their roles go beyond storing genetic information and chemical energy or as signal transducer. Attenuation or expression of desired genes through nucleic acids have profound implications in gene therapy, gene editing and even in vaccine development for immunomodulation. Although nucleic acid therapeutics bring in overwhelming possibilities towards the development of molecular medicines, there are significant loopholes in designing and effective translation of these drugs into the clinic. One of the major pitfalls lies in the traditional design concepts for nucleic acid drug carriers, viz. cationic charge induced cytotoxicity in delivery pathway. Targeting this bottleneck, several pioneering research efforts have been devoted to design innovative carriers through charge-conversion approaches, whereby built-in functionalities convert from cationic to neutral or anionic, or even from anionic to cationic enabling the carrier to overcome several critical barriers for therapeutics delivery, such as serum deactivation, instability in circulation, low transfection and poor endosomal escape. This review will critically analyze various molecular designs of charge-converting nanocarriers in a classified approach for the successful delivery of nucleic acids. Accompanied by the narrative on recent clinical nucleic acid candidates, the review concludes with a discussion on the pitfalls and scope of these interesting approaches.
Collapse
Affiliation(s)
- Kingshuk Dutta
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Corteva Agriscience, 9330 Zionsville Road, Indianapolis 46268, United States
| | - Ritam Das
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Pintu Kanjilal
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
183
|
Franck CO, Fanslau L, Bistrovic Popov A, Tyagi P, Fruk L. Biopolymer-based Carriers for DNA Vaccine Design. Angew Chem Int Ed Engl 2021; 60:13225-13243. [PMID: 32893932 PMCID: PMC8247987 DOI: 10.1002/anie.202010282] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Indexed: 12/16/2022]
Abstract
Over the last 30 years, genetically engineered DNA has been tested as novel vaccination strategy against various diseases, including human immunodeficiency virus (HIV), hepatitis B, several parasites, and cancers. However, the clinical breakthrough of the technique is confined by the low transfection efficacy and immunogenicity of the employed vaccines. Therefore, carrier materials were designed to prevent the rapid degradation and systemic clearance of DNA in the body. In this context, biopolymers are a particularly promising DNA vaccine carrier platform due to their beneficial biochemical and physical characteristics, including biocompatibility, stability, and low toxicity. This article reviews the applications, fabrication, and modification of biopolymers as carrier medium for genetic vaccines.
Collapse
Affiliation(s)
- Christoph O. Franck
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Luise Fanslau
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Andrea Bistrovic Popov
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Puneet Tyagi
- Dosage Form Design and DevelopmentBioPharmaceuticals DevelopmentR&DAstra ZenecaGaithersburgMD20878USA
| | - Ljiljana Fruk
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
184
|
Boonstra E, Hatano H, Miyahara Y, Uchida S, Goda T, Cabral H. A proton/macromolecule-sensing approach distinguishes changes in biological membrane permeability during polymer/lipid-based nucleic acid delivery. J Mater Chem B 2021; 9:4298-4302. [PMID: 34018540 DOI: 10.1039/d1tb00645b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
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.
Collapse
Affiliation(s)
- Eger Boonstra
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-8656, Japan.
| | - Hiroaki Hatano
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Yuji Miyahara
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Satoshi Uchida
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan.
| | - Tatsuro Goda
- Department of Biomedical Engineering, Faculty of Science and Engineering, Toyo University, 2100 Kujirai, Kawagoe, Saitama 350-8585, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-8656, Japan.
| |
Collapse
|
185
|
Zhang Z, Qiu N, Wu S, Liu X, Zhou Z, Tang J, Liu Y, Zhou R, Shen Y. Dose-Independent Transfection of Hydrophobized Polyplexes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102219. [PMID: 33991017 DOI: 10.1002/adma.202102219] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/09/2021] [Indexed: 05/14/2023]
Abstract
Cationic polymers dynamically complex DNA into complexes (polyplexes). So, upon dilution, polyplexes easily dissociate and lose transfection ability, limiting their in vivo systemic gene delivery. Herein, it is found that polyplex's stability and endocytosis pathway determine its transfection dose-dependence. The polyplexes of hydrophilic polycations have dose-dependent integrity and lysosome-trafficking endocytosis; at low doses, most of these polyplexes dissociate, and the remaining few are internalized and trapped in lysosomes, abolishing their transfection ability. In contrast, the polyplexes of the polycations with optimal hydrophobicity remain integrated even at low concentrations and enter cells via macropinocytosis directly into the cytosol evading lysosomes, so each polyplex can accomplish its infection process, leading to dose-independent DNA transfection like viral vectors. Furthermore, the tuned hydrophobicity balancing the affinity of anionic poly(γ-glutamic acid) (γ-PGA) to the polyplex surface enables γ-PGA to stick on the polyplex surface as a shielding layer but peel off on the cell membrane to release the naked polyplexes for dose-independent transfection. These findings may provide guidelines for developing polyplexes that mimick a viral vector's dose-independent transfection for effective in vivo gene delivery.
Collapse
Affiliation(s)
- Zhen Zhang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Shuling Wu
- Department of Respiratory, The First People's Hospital of Xiaoshan, Hangzhou, 311200, China
| | - Xin Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yanpeng Liu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, China
| |
Collapse
|
186
|
Abstract
To realize RNA interference (RNAi) therapeutics, it is necessary to deliver therapeutic RNAs (such as small interfering RNA or siRNA) into cell cytoplasm. A major challenge of RNAi therapeutics is the endosomal entrapment of the delivered siRNA. In this study, we developed a family of delivery vehicles called Janus base nanopieces (NPs). They are rod-shaped nanoparticles formed by bundles of Janus base nanotubes (JBNTs) with RNA cargoes incorporated inside via charge interactions. JBNTs are formed by noncovalent interactions of small molecules consisting of a base component mimicking DNA bases and an amino acid side chain. NPs presented many advantages over conventional delivery materials. NPs efficiently entered cells via macropinocytosis similar to lipid nanoparticles while presenting much better endosomal escape ability than lipid nanoparticles; NPs escaped from endosomes via a "proton sponge" effect similar to cationic polymers while presenting significant lower cytotoxicity compared to polymers and lipids due to their noncovalent structures and DNA-mimicking chemistry. In a proof-of-concept experiment, we have shown that NPs are promising candidates for antiviral delivery applications, which may be used for conditions such as COVID-19 in the future.
Collapse
|
187
|
de Braganca L, Ferguson GJ, Luis Santos J, Derrick JP. Adverse immunological responses against non-viral nanoparticle (NP) delivery systems in the lung. J Immunotoxicol 2021; 18:61-73. [PMID: 33956565 PMCID: PMC8788408 DOI: 10.1080/1547691x.2021.1902432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
There is a large, unmet medical need to treat chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis and other respiratory diseases. New modalities are being developed, including gene therapy which treats the disease at the DNA/RNA level. Despite recent innovations in non-viral gene therapy delivery for chronic respiratory diseases, unwanted or adverse interactions with immune cells, particularly macrophages, can limit drug efficacy. This review will examine the relationship between the design and fabrication of non-viral nucleic acid nanoparticle (NP) delivery systems and their ability to trigger unwanted immunogenic responses in lung tissues. NP formulated with peptides, lipids, synthetic and natural polymers provide a robust means of delivering the genetic cargos to the desired cells. However NP, or their components, may trigger local responses such as cell damage, edema, inflammation, and complement activation. These effects may be acute short-term reactions or chronic long-term effects like fibrosis, increased susceptibility to diseases, autoimmune disorders, and even cancer. This review examines the relationship between physicochemical properties, i.e. shape, charge, hydrophobicity, composition and stiffness, and interactions of NP with pulmonary immune cells. Inhalation is the ideal route of administration for direct delivery but inhaled NP encounter innate immune cells, such as alveolar macrophages (AM) and dendritic cells (DC), that perceive them as harmful foreign material, interfere with gene delivery to target cells, and can induce undesirable side effects. Recommendations for fabrication and formulation of gene therapies to avoid adverse immunological responses are given. These include fine tuning physicochemical properties, functionalization of the surface of NP to actively target diseased pulmonary cells and employing biomimetics to increase immunotolerance.
Collapse
Affiliation(s)
- Leonor de Braganca
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - G John Ferguson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jose Luis Santos
- Dosage Form Design Development, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Jeremy P Derrick
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
188
|
Chemical Manipulation of the Endosome Trafficking Machinery: Implications for Oligonucleotide Delivery. Biomedicines 2021; 9:biomedicines9050512. [PMID: 34063104 PMCID: PMC8148136 DOI: 10.3390/biomedicines9050512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 12/14/2022] Open
Abstract
Antisense oligonucleotides (ASOs), siRNA and splice switching oligonucleotides (SSOs) all have immense potential as therapeutic agents, potential that is now being validated as oligonucleotides enter the clinic. However, progress in oligonucleotide-based therapeutics has been limited by the difficulty in delivering these complex molecules to their sites of action in the cytosol or nucleus of cells within specific tissues. There are two aspects to the delivery problem. The first is that most types of oligonucleotides have poor uptake into non-hepatic tissues. The second is that much of the oligonucleotide that is taken up by cells is entrapped in endosomes where it is pharmacologically inert. It has become increasingly recognized that endosomal trapping is a key constraint on oligonucleotide therapeutics. Thus, many approaches have been devised to address this problem, primarily ones based on various nanoparticle technologies. However, recently an alternative approach has emerged that employs small molecules to manipulate intracellular trafficking processes so as to enhance oligonucleotide actions. This review presents the current status of this chemical biology approach to oligonucleotide delivery and seeks to point out possible paths for future development.
Collapse
|
189
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
190
|
Ahmadi Z, Jena H, Singh M, Dhawan G, Kumar P. Self-Assembled Biodegradable Core-Shell Nanocomposites of Amphiphilic Retinoic Acid-LMW bPEI Conjugates Exhibit Enhanced Transgene Expression in Hepatocellular Carcinoma Cells With Inherent Anticancer Properties. J Pharm Sci 2021; 110:3047-3060. [PMID: 33933435 DOI: 10.1016/j.xphs.2021.04.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/02/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022]
Abstract
Low molecular weight branched polyethylenimines (LMW bPEIs) are almost nontoxic but display poor transfection efficiency due to lack of adequate complexation ability with nucleic acids followed by transportation across the cell membrane. Here, a series of amphiphilic retinoyl-bPEI conjugates (RP-1, RP-2 and RP-3) has been synthesized by allowing the reaction between bPEI (1.8 kDa) and a bioactive and hydrophobic vitamin A metabolite, all-trans-retinoic acid (ATRA), in varying amounts. In aqueous medium, these conjugates self-assembled into core-shell RP nanocomposites with size ranging from ~113-178 nm and zeta potential from ~ +15-35 mV. Evaluation of pDNA complexes of RP nanocomposites revealed that all the complexes exhibited significantly enhanced transfection efficiency without compromising on the cytocompatibility. RP-3/pDNA complex, with the highest content of retinoic acid, exhibited the best transfection efficiency. Further, due to anticancer properties of ATRA, these nanocomposites significantly reduced the viability of cancer cells (HepG2 and MCF-7 cells) without affecting the viability of non-cancerous cells (HEK 293 cells) demonstrating the cell-selective nature of the formulated nanocomposites. The intracellular trafficking and co-localization studies involving RP-3 nanocomposites also showed their higher uptake with intracellular and nuclear accumulation properties. Altogether, the results demonstrate the promising potential of the RP conjugates that can be used in future hepatocellular carcinoma targeted gene delivery applications.
Collapse
Affiliation(s)
- Zeba Ahmadi
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi-110007, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad- 201002, India
| | - Harekrushna Jena
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi-110007, India; Department of Biomedical Sciences, Acharya Narendra Dev College (University of Delhi), Govindpuri, Kalkaji, New Delhi- 110019, India
| | - Mahak Singh
- Department of Chemistry, Ramjas College (University of Delhi), University Enclave, Delhi-110007, India
| | - Gagan Dhawan
- Department of Biomedical Sciences, Acharya Narendra Dev College (University of Delhi), Govindpuri, Kalkaji, New Delhi- 110019, India.
| | - Pradeep Kumar
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi-110007, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad- 201002, India.
| |
Collapse
|
191
|
Chen Z, Wang X, Liu S, Li Y, Zhou H, Guo T. Zn(ii)-Dipicolylamine analogues with amphiphilic side chains endow low molecular weight PEI with high transfection performance. Biomater Sci 2021; 9:3090-3099. [PMID: 33751016 DOI: 10.1039/d0bm02181d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
To investigate the effect of amphiphilic balance of Zn(ii)-dipicolylamine analogues on the transfection process, we fabricated a series of Zn(ii)-dipicolylamine functional modules (DDAC-Rs) with different hydrophilic-phobic side chains to modify low molecular weight PEI (Zn-DP-Rs) by the Michael addition reaction. Zn-DP-Rs with hydrophilic terminal hydroxy group side chains demonstrate superior overall performance compared to those of hydrophobic alkyl side chains. In terms of the influence of the chain lengths in DDAC-Rs, from Zn-DP-A/OH-3 to Zn-DP-A/OH-5, the corresponding transfection efficiency shows an upward trend as the lengths increase. However, decreasing efficacy is observed with further increase in the length of side chains. In addition, the Zn-DP-Rs with amphiphilic side chains show prominent performance in every respect, highlighting the significance of balance in the amphipathy of side chains in DDAC-Rs. This work is of great significance for the development of polycationic gene carrier materials with excellent performance.
Collapse
Affiliation(s)
- Zhaoming Chen
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Weijin Road, No. 94, Tianjin, 300071, China.
| | - Xindong Wang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Weijin Road, No. 94, Tianjin, 300071, China.
| | - Shuai Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Weijin Road, No. 94, Tianjin, 300071, China.
| | - Yumeng Li
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Weijin Road, No. 94, Tianjin, 300071, China.
| | - Hao Zhou
- Department of Biochemistry and Molecular Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Tianying Guo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Weijin Road, No. 94, Tianjin, 300071, China.
| |
Collapse
|
192
|
Singh R, Kumar P. Disaccharide-polyethylenimine organic nanoparticles as non-toxic in vitro gene transporters and their anticancer potential. Bioorg Chem 2021; 112:104918. [PMID: 33932768 DOI: 10.1016/j.bioorg.2021.104918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/26/2022]
Abstract
Polyethylenimines (PEIs) have been shown as efficient gene delivery vectors due to their unique properties, however, toxicity as well as non-specific interactions with the tissues/cells because of high charge density have hampered their use in clinical applications. To counter these concerns, here, we have prepared disachharide-PEI organic nanoparticles by mixing PEIs with non-reducing disaccharides, i.e. trehalose (TPONs) and sucrose (SPONs), under mild conditions. The fabricated nanoparticles were complexed with pDNA and size of these complexes was found in the range of ~130-162 nm with zeta potential ~ +8-25 mV. Further evaluation of these nanoparticles revealed that substitution of disaccharides on PEIs successfully augmented cell viability. Transfection efficiency exhibited by these complexes was significantly higher than the unmodified polymer and the standard, Lipofectamine, complexes. Fabrication of organic nanoparticles did not alter the buffering capacity considerably which was found to be instrumental during endosomal escape of the complexes. Among both the series of nanoparticles, trehalose-PEI organic nanoparticles (TPONs) exhibited greater pDNA transportation potential than sucrose-PEI organic nanoparticles (SPONs) which was also established by flow cytometric data, wherein percent cells expressing GFP was higher in case of TP/pDNA complexes as compared to SP/pDNA complexes. Interestingly, TPONs also showed promising anticancer activity on cancer cell lines i.e. Mg63, MCF-7 and HepG2. Overall, the results advocate promising potential of disaccharide-PEI organic nanoparticles as efficient gene delivery agents which can be used effectively in future gene therapy applications along with anti-cancer competence of TPONs.
Collapse
Affiliation(s)
- Reena Singh
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pradeep Kumar
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
193
|
Son I, Lee Y, Baek J, Park M, Han D, Min SK, Lee D, Kim BS. pH-Responsive Amphiphilic Polyether Micelles with Superior Stability for Smart Drug Delivery. Biomacromolecules 2021; 22:2043-2056. [PMID: 33835793 DOI: 10.1021/acs.biomac.1c00163] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite widespread interest in the amphiphilic polymeric micelles for drug delivery systems, it is highly desirable to achieve high loading capacity and high efficiency to reduce the side effects of therapeutic agents while maximizing their efficacy. Here, we present a novel hydrophobic epoxide monomer, cyclohexyloxy ethyl glycidyl ether (CHGE), containing an acetal group as a pH-responsive cleavable linkage. A series of its homopolymers, poly(cyclohexyloxy ethyl glycidyl ether)s (PCHGEs), and block copolymers, poly(ethylene glycol)-block-poly(cyclohexyloxy ethyl glycidyl ether)s (mPEG-b-PCHGE), were synthesized via anionic ring-opening polymerization in a controlled manner. Subsequently, the self-assembled polymeric micelles of mPEG-b-PCHGE demonstrated high loading capacity, excellent stability in biological media, tunable release efficiency, and high cell viability. Importantly, quantum mechanical calculations performed by considering prolonged hydrolysis of the acetal group in CHGE indicated that the CHGE monomer had higher hydrophobicity than three other functional epoxide monomer analogues developed. Furthermore, the preferential cellular uptake and in vivo therapeutic efficacy confirmed the enhanced stability and the pH-responsive degradation of the amphiphilic block copolymer micelles. This study provides a new platform for the development of versatile smart polymeric drug delivery systems with high loading efficiency and tailorable release profiles.
Collapse
Affiliation(s)
- Iloh Son
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Yujin Lee
- Department of PolymerNano Science and Technology, Chonbuk National University, Jeonju 54896, Republic of Korea
| | - Jinsu Baek
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Miran Park
- Department of PolymerNano Science and Technology, Chonbuk National University, Jeonju 54896, Republic of Korea
| | - Daeho Han
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seung Kyu Min
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Dongwon Lee
- Department of PolymerNano Science and Technology, Chonbuk National University, Jeonju 54896, Republic of Korea
| | - Byeong-Su Kim
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
194
|
Goswami A, Sharma PR, Agarwal R. Combatting intracellular pathogens using bacteriophage delivery. Crit Rev Microbiol 2021; 47:461-478. [PMID: 33818246 DOI: 10.1080/1040841x.2021.1902266] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Intracellular pathogens reside in specialised compartments within the host cells restricting the access of antibiotics. Insufficient intracellular delivery of antibiotics along with several other resistance mechanisms weaken the efficacy of current therapies. An alternative to antibiotic therapy could be bacteriophage (phage) therapy. Although phage therapy has been in practice for a century against various bacterial infections, the efficacy of phages against intracellular bacteria is still being explored. In this review, we will discuss the advancement and challenges in phage therapy, particularly against intracellular bacterial pathogens. Finally, we will highlight the uptake mechanisms and approaches to overcome the challenges to phage therapy against intracellular bacteria.
Collapse
Affiliation(s)
- Avijit Goswami
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Pallavi Raj Sharma
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Rachit Agarwal
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
195
|
Zhou J, Ma S, Zhang Y, He Y, Mao H, Yang J, Zhang H, Luo K, Gong Q, Gu Z. Bacterium-mimicking sequentially targeted therapeutic nanocomplexes based on O-carboxymethyl chitosan and their cooperative therapy by dual-modality light manipulation. Carbohydr Polym 2021; 264:118030. [PMID: 33910720 DOI: 10.1016/j.carbpol.2021.118030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/10/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023]
Abstract
An integrated gene nanovector capable of overcoming complicated physiological barriers in one vector is desirable to circumvent the challenges imposed by the intricate tumor microenvironment. Herein, a nuclear localization signals (NLS)-decorated element and an iRGD-functionalized element based on O-carboxymethyl chitosan were synthesized, mixed, and coated onto PEI/DNA to fabricate bacterium-mimicking sequentially targeted therapeutic nanocomplexes (STNPs) which were internalized through receptor-mediated endocytosis and other pathways and achieved nuclear translocation of DNA. The endo/lysosomal membrane disruption triggered by reactive oxygen species (ROS) after short-time illumination, together with the DNA nuclear translocation, evoked an enhanced gene expression. Alternatively, the excessive ROS from long-time irradiation induced apoptosis in tumor cells, bringing about greater anti-tumor efficacy owing to the integration of gene and photodynamic therapy. Overall, these results demonstrated bacterium-mimicking STNPs could be a potential candidate for tumor treatments.
Collapse
Affiliation(s)
- Jie Zhou
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Shengnan Ma
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Yuxin Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Yiyan He
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, 30 South Puzhu Road, Nanjing, 211816, PR China.
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, 30 South Puzhu Road, Nanjing, 211816, PR China
| | - Jun Yang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, PR China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China; Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, 30 South Puzhu Road, Nanjing, 211816, PR China.
| |
Collapse
|
196
|
Taleghani AS, Nakhjiri AT, Khakzad MJ, Rezayat SM, Ebrahimnejad P, Heydarinasab A, Akbarzadeh A, Marjani A. Mesoporous silica nanoparticles as a versatile nanocarrier for cancer treatment: A review. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.115417] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
197
|
Mdlovu NV, Lin KS, Chen Y, Wu CM. Formulation of magnetic nanocomposites for intracellular delivery of micro-RNA for MYCN inhibition in neuroblastoma. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.126264] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
198
|
Wang S. pH-Responsive Amphiphilic Carboxylate Polymers: Design and Potential for Endosomal Escape. Front Chem 2021; 9:645297. [PMID: 33834015 PMCID: PMC8021698 DOI: 10.3389/fchem.2021.645297] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/22/2021] [Indexed: 12/19/2022] Open
Abstract
The intracellular delivery of emerging biomacromolecular therapeutics, such as genes, peptides, and proteins, remains a great challenge. Unlike small hydrophobic drugs, these biotherapeutics are impermeable to the cell membrane, thus relying on the endocytic pathways for cell entry. After endocytosis, they are entrapped in the endosomes and finally degraded in lysosomes. To overcome these barriers, many carriers have been developed to facilitate the endosomal escape of these biomacromolecules. This mini-review focuses on the development of anionic pH-responsive amphiphilic carboxylate polymers for endosomal escape applications, including the design and synthesis of these polymers, the mechanistic insights of their endosomal escape capability, the challenges in the field, and future opportunities.
Collapse
Affiliation(s)
- Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
199
|
Yu T, Nie W, Hong Z, He Y, Chen J, Mi X, Yang S, Li X, Wang B, Lin Y, Gao X. Synergy of Immunostimulatory Genetherapy with Immune Checkpoint Blockade Motivates Immune Response to Eliminate Cancer. ADVANCED FUNCTIONAL MATERIALS 2021. [DOI: 10.1002/adfm.202100715] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Ting Yu
- Department of Neurosurgery and Institute of Neurosurgery State Key Laboratory of Biotherapy and cancer center West China Hospital West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Wen Nie
- Department of Neurosurgery and Institute of Neurosurgery State Key Laboratory of Biotherapy and cancer center West China Hospital West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
- Department of Medical Oncology The First Affiliated Hospital School of Medicine Zhejiang University Hangzhou 310003 P. R. China
| | - Zehua Hong
- Department of Neurosurgery and Institute of Neurosurgery State Key Laboratory of Biotherapy and cancer center West China Hospital West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Yihong He
- Department of Neurosurgery and Institute of Neurosurgery State Key Laboratory of Biotherapy and cancer center West China Hospital West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Jing Chen
- Department of Neurosurgery and Institute of Neurosurgery State Key Laboratory of Biotherapy and cancer center West China Hospital West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Xue Mi
- Department of Neurosurgery and Institute of Neurosurgery State Key Laboratory of Biotherapy and cancer center West China Hospital West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Shuping Yang
- Department of Neurosurgery and Institute of Neurosurgery State Key Laboratory of Biotherapy and cancer center West China Hospital West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Xiaoling Li
- Department of Neurosurgery and Institute of Neurosurgery State Key Laboratory of Biotherapy and cancer center West China Hospital West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Bilan Wang
- Department of Pharmacy West China Second University Hospital of Sichuan University Chengdu 610041 P. R. China
| | - Yunzhu Lin
- Department of Pharmacy West China Second University Hospital of Sichuan University Chengdu 610041 P. R. China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery State Key Laboratory of Biotherapy and cancer center West China Hospital West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| |
Collapse
|
200
|
Khazieva A, Kholin K, Nizameev I, Brylev K, Kashnik I, Voloshina A, Lyubina A, Gubaidullin A, Daminova A, Petrov K, Mustafina A. Surface modification of silica nanoparticles by hexarhenium anionic cluster complexes for pH-sensing and staining of cell nuclei. J Colloid Interface Sci 2021; 594:759-769. [PMID: 33789187 DOI: 10.1016/j.jcis.2021.03.082] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/05/2021] [Accepted: 03/13/2021] [Indexed: 11/29/2022]
Abstract
The surface deposition of luminescent anionic cluster complex [{Re6S8}(OH)6]4- advantages to the design and synthesis of composite luminescent silica nanoparticles (SNs) for intracellular imaging and sensing, while the encapsulation of the cluster units into SNs lacks for efficient luminescence. The deposition of the Re6 clusters resulted from their assembly at the silica surface functionalized by amino-groups provides the synthetic route for the composite SNs with bright cluster-centered luminescence invariable in pH range from 4.0 to 12.0. The pH-dependent supramolecular assembly of the cluster units with polyethyleneimine (PEI) at the silica surface is an alternative route for the synthesis of the composite SNs with high cluster-centered luminescence sensitive to pH-changes within 4.0-6.0. The sensitivity derives from the pH-driven conformational changes of PEI chains resulting in the release of the clusters from the PEI-based confinement under the acidification within pH 6.0-4.0. The potential of the composite SNs in cellular contrasting has been also revealed by the cell viability and flow cytometry measurements. It has been found that the PEI-supported embedding of the cluster units facilitates cell internalization of the composite SNs as well as results in specific intracellular distribution manifested by efficient staining of the cell nuclei in the confocal images.
Collapse
Affiliation(s)
- Alsu Khazieva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation.
| | - Kirill Kholin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Irek Nizameev
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Konstantin Brylev
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., 630090 Novosibirsk, Russian Federation
| | - Ilya Kashnik
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., 630090 Novosibirsk, Russian Federation
| | - Alexandra Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Anna Lyubina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Aidar Gubaidullin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Amina Daminova
- Kazan (Volga region) Federal University, 18 Kremlyovskaya str., 420008 Kazan, Russian Federation
| | - Konstantin Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| | - Asiya Mustafina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov str., 420088 Kazan, Russian Federation
| |
Collapse
|