1
|
Xu P, Zhou J, Xing X, Hao Y, Gao M, Li Z, Li X, Li M, Xiao Y. Melitoxin Inhibits Proliferation, Metastasis, and Invasion of Glioma U251 Cells by Down-regulating F2RL1. Appl Biochem Biotechnol 2024; 196:6234-6252. [PMID: 38252207 DOI: 10.1007/s12010-023-04841-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/23/2024]
Abstract
As the principal active component of bee venom, melittin has an anti-cancer effect in different cancers. This study was aimed to investigate the effect of melittin in glioma and explore whether F2RL1 is closely involved in glioblastoma cells proliferation. TCGA and GES databases were used to evaluate the role of F2RL1 in gliomas. The U251 cells were divided into a control lentivirus + PBS group (NC-PBS), F2RL1 intervention lentivirus + PBS group (KD-PBS), control lentivirus + melittin group (NC-melittin), and F2RL1 intervention lentivirus + melittin group (KD-melittin). Cell proliferation was detected by MTT and EDU staining assays. The apoptosis rate was assessed by flow cytometry. Expressions of genes related to apoptosis, cycle arrest, migration, and invasion were detected by qRT-PCR. Cellular LDH concentrations were detected by ELISA. The subcutaneous tumor volume of nude mice was analyzed by xenograft method. F2RL1 was significantly overexpressed in glioma tissues and were reduced in the melittin-treated group compared to the blank group. F2RL1 knockdown and melittin alone or in combination increased the proportion of cells in the G1-phase, and the combination was more pronounced. The KD-melittin group showed a decrease in the number of viable cells at 24, 48, 72, and 96 h compared to the NC-PBS group. The number of cell migration and invasion was decreased in the KD-melittin group compared to the other groups. Moreover, the genes related to cell cycle arrest and apoptosis were significantly changed in the KD-melittin group. At weeks 4, 5, and 6, the tumor volume in the KD-melittin group was smaller than that in the KD-PBS group and NC-melittin group. Interference with the target gene F2RL1 inhibited the proliferation of glioma U251 cells, and melittin treatment inhibited the proliferation of glioma U251 cells. Melittin inhibited the proliferation of glioma U251 cells by suppressing the expression of target gene F2RL1.
Collapse
Affiliation(s)
- Peng Xu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Jie Zhou
- Department of Nursing, Liaocheng Vocational and Technical College, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Xiaohui Xing
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Yuan Hao
- Department of Pathology, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Mingxu Gao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Zhongchen Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Xin Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Mengyou Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China.
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China.
| |
Collapse
|
2
|
Abstract
Carriers for RNA delivery must be dynamic, first stabilizing and protecting therapeutic RNA during delivery to the target tissue and across cellular membrane barriers and then releasing the cargo in bioactive form. The chemical space of carriers ranges from small cationic lipids applied in lipoplexes and lipid nanoparticles, over medium-sized sequence-defined xenopeptides, to macromolecular polycations applied in polyplexes and polymer micelles. This perspective highlights the discovery of distinct virus-inspired dynamic processes that capitalize on mutual nanoparticle-host interactions to achieve potent RNA delivery. From the host side, subtle alterations of pH, ion concentration, redox potential, presence of specific proteins, receptors, or enzymes are cues, which must be recognized by the RNA nanocarrier via dynamic chemical designs including cleavable bonds, alterable physicochemical properties, and supramolecular assembly-disassembly processes to respond to changing biological microenvironment during delivery.
Collapse
Affiliation(s)
- Simone Berger
- Department of Pharmacy, Pharmaceutical Biotechnology, Ludwig-Maximilians-Universität Munich, 81377Munich, Germany
- Center for NanoScience, Ludwig-Maximilians-Universität Munich, 80799Munich, Germany
| | - Ulrich Lächelt
- Center for NanoScience, Ludwig-Maximilians-Universität Munich, 80799Munich, Germany
- Department of Pharmaceutical Sciences, University of Vienna, Vienna1090, Austria
| | - Ernst Wagner
- Department of Pharmacy, Pharmaceutical Biotechnology, Ludwig-Maximilians-Universität Munich, 81377Munich, Germany
- Center for NanoScience, Ludwig-Maximilians-Universität Munich, 80799Munich, Germany
| |
Collapse
|
3
|
Lyu M, Yazdi M, Lin Y, Höhn M, Lächelt U, Wagner E. Receptor-Targeted Dual pH-Triggered Intracellular Protein Transfer. ACS Biomater Sci Eng 2024; 10:99-114. [PMID: 35802884 DOI: 10.1021/acsbiomaterials.2c00476] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein therapeutics are of widespread interest due to their successful performance in the current pharmaceutical and medical fields, even though their broad applications have been hindered by the lack of an efficient intracellular delivery approach. Herein, we fabricated an active-targeted dual pH-responsive delivery system with favorable tumor cell entry augmented by extracellular pH-triggered charge reversal and tumor receptor targeting and pH-controlled endosomal release in a traceless fashion. As a traceable model protein, the enhanced green fluorescent protein (eGFP) bearing a nuclear localization signal was covalently coupled with a pH-labile traceless azidomethyl-methylmaleic anhydride (AzMMMan) linker followed by functionalization with different molar equivalents of two dibenzocyclooctyne-octa-arginine-cysteine (DBCO-R8C)-modified moieties: polyethylene glycol (PEG)-GE11 peptide for epidermal growth factor receptor-mediated targeting and melittin for endosomal escape. The cationic melittin domain was masked with tetrahydrophthalic anhydride revertible at mild acidic pH 6.5. At the optimally balanced ratio of functional units, the on-demand charge conversion at tumoral extracellular pH 6.5 in combination with GE11-mediated targeting triggered enhanced electrostatic cellular attraction by the R8C cell-penetrating peptides and melittin, as demonstrated by strongly enhanced cellular uptake. Successful endosomal release followed by nuclear localization of the eGFP cargo was obtained by taking advantage of melittin-mediated endosomal escape and rapid traceless release from the AzMMMan linker. The effectiveness of this multifunctional bioresponsive system suggests a promising strategy for delivery of protein drugs toward intracellular targets. A possible therapeutic relevance was indicated by an example of cytosolic delivery of cytochrome c initiating the apoptosis pathway to kill cancer cells.
Collapse
Affiliation(s)
- Meng Lyu
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Yi Lin
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Ulrich Lächelt
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| |
Collapse
|
4
|
Sinani G, Durgun ME, Cevher E, Özsoy Y. Polymeric-Micelle-Based Delivery Systems for Nucleic Acids. Pharmaceutics 2023; 15:2021. [PMID: 37631235 PMCID: PMC10457940 DOI: 10.3390/pharmaceutics15082021] [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: 05/13/2023] [Revised: 07/11/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Nucleic acids can modulate gene expression specifically. They are increasingly being utilized and show huge potential for the prevention or treatment of various diseases. However, the clinical translation of nucleic acids faces many challenges due to their rapid clearance after administration, low stability in physiological fluids and limited cellular uptake, which is associated with an inability to reach the intracellular target site and poor efficacy. For many years, tremendous efforts have been made to design appropriate delivery systems that enable the safe and effective delivery of nucleic acids at the target site to achieve high therapeutic outcomes. Among the different delivery platforms investigated, polymeric micelles have emerged as suitable delivery vehicles due to the versatility of their structures and the possibility to tailor their composition for overcoming extracellular and intracellular barriers, thus enhancing therapeutic efficacy. Many strategies, such as the addition of stimuli-sensitive groups or specific ligands, can be used to facilitate the delivery of various nucleic acids and improve targeting and accumulation at the site of action while protecting nucleic acids from degradation and promoting their cellular uptake. Furthermore, polymeric micelles can be used to deliver both chemotherapeutic drugs and nucleic acid therapeutics simultaneously to achieve synergistic combination treatment. This review focuses on the design approaches and current developments in polymeric micelles for the delivery of nucleic acids. The different preparation methods and characteristic features of polymeric micelles are covered. The current state of the art of polymeric micelles as carriers for nucleic acids is discussed while highlighting the delivery challenges of nucleic acids and how to overcome them and how to improve the safety and efficacy of nucleic acids after local or systemic administration.
Collapse
Affiliation(s)
- Genada Sinani
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Altinbas University, 34147 Istanbul, Türkiye;
| | - Meltem Ezgi Durgun
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34126 Istanbul, Türkiye; (M.E.D.); (E.C.)
| | - Erdal Cevher
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34126 Istanbul, Türkiye; (M.E.D.); (E.C.)
| | - Yıldız Özsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34126 Istanbul, Türkiye; (M.E.D.); (E.C.)
| |
Collapse
|
5
|
Surface Design Options in Polymer- and Lipid-Based siRNA Nanoparticles Using Antibodies. Int J Mol Sci 2022; 23:ijms232213929. [PMID: 36430411 PMCID: PMC9692731 DOI: 10.3390/ijms232213929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
The mechanism of RNA interference (RNAi) could represent a breakthrough in the therapy of all diseases that arise from a gene defect or require the inhibition of a specific gene expression. In particular, small interfering RNA (siRNA) offers an attractive opportunity to achieve a new milestone in the therapy of human diseases. The limitations of siRNA, such as poor stability, inefficient cell uptake, and undesired immune activation, as well as the inability to specifically reach the target tissue in the body, can be overcome by further developments in the field of nanoparticulate drug delivery. Therefore, types of surface modified siRNA nanoparticles are presented and illustrate how a more efficient and safer distribution of siRNA at the target site is possible by modifying the surface properties of nanoparticles with antibodies. However, the development of such efficient and safe delivery strategies is currently still a major challenge. In consideration of that, this review article aims to demonstrate the function and targeted delivery of siRNA nanoparticles, focusing on the surface modification via antibodies, various lipid- and polymer-components, and the therapeutic effects of these delivery systems.
Collapse
|
6
|
Tuncaboylu DC, Wischke C. Opportunities and Challenges of Switchable Materials for Pharmaceutical Use. Pharmaceutics 2022; 14:2331. [PMID: 36365149 PMCID: PMC9696173 DOI: 10.3390/pharmaceutics14112331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 06/27/2024] Open
Abstract
Switchable polymeric materials, which can respond to triggering signals through changes in their properties, have become a major research focus for parenteral controlled delivery systems. They may enable externally induced drug release or delivery that is adaptive to in vivo stimuli. Despite the promise of new functionalities using switchable materials, several of these concepts may need to face challenges associated with clinical use. Accordingly, this review provides an overview of various types of switchable polymers responsive to different types of stimuli and addresses opportunities and challenges that may arise from their application in biomedicine.
Collapse
|
7
|
Abstract
Advances in microfabrication and biomaterials have enabled the development of microfluidic chips for studying tissue and organ models. While these platforms have been developed primarily for modeling human diseases, they are also used to uncover cellular and molecular mechanisms through in vitro studies, especially in the neurovascular system, where physiological mechanisms and three-dimensional (3D) architecture are difficult to reconstruct via conventional assays. An extracellular matrix (ECM) model with a stable structure possessing the ability to mimic the natural extracellular environment of the cell efficiently is useful for tissue engineering applications. Conventionally used techniques for this purpose, for example, Matrigels, have drawbacks of owning complex fabrication procedures, in some cases not efficient enough in terms of functionality and expenses. Here, we proposed a fabrication protocol for a GelMA hydrogel, which has shown structural stability and the ability to imitate the natural environment of the cell accurately, inside a microfluidic chip utilizing co-culturing of two human cell lines. The chemical composition of the synthesized GelMA was identified by Fourier transform infrared spectrophotometry (FTIR), its surface morphology was observed by field emission electron microscopy (FESEM), and the structural properties were analyzed by atomic force microscopy (AFM). The swelling behavior of the hydrogel in the microfluidic chip was imaged, and its porosity was examined for 72 h by tracking cell localization using immunofluorescence. GelMA exhibited the desired biomechanical properties, and the viability of cells in both platforms was more than 80% for seven days. Furthermore, GelMA was a viable platform for 3D cell culture studies and was structurally stable over long periods, even when prepared by photopolymerization in a microfluidic platform. This work demonstrated a viable strategy to conduct co-culturing experiments as well as modeling invasion and migration events. This microfluidic assay may have application in drug delivery and dosage optimization studies.
Collapse
|
8
|
Wang A, Zheng Y, Zhu W, Yang L, Yang Y, Peng J. Melittin-Based Nano-Delivery Systems for Cancer Therapy. Biomolecules 2022; 12:biom12010118. [PMID: 35053266 PMCID: PMC8773652 DOI: 10.3390/biom12010118] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/01/2022] [Accepted: 01/04/2022] [Indexed: 02/06/2023] Open
Abstract
Melittin (MEL) is a 26-amino acid polypeptide with a variety of pharmacological and toxicological effects, which include strong surface activity on cell lipid membranes, hemolytic activity, and potential anti-tumor properties. However, the clinical application of melittin is restricted due to its severe hemolytic activity. Different nanocarrier systems have been developed to achieve stable loading, side effects shielding, and tumor-targeted delivery, such as liposomes, cationic polymers, lipodisks, etc. In addition, MEL can be modified on nano drugs as a non-selective cytolytic peptide to enhance cellular uptake and endosomal/lysosomal escape. In this review, we discuss recent advances in MEL’s nano-delivery systems and MEL-modified nano drug carriers for cancer therapy.
Collapse
|
9
|
van Hees M, Slott S, Hansen AH, Kim HS, Ji HP, Astakhova K. New approaches to moderate CRISPR-Cas9 activity: Addressing issues of cellular uptake and endosomal escape. Mol Ther 2022; 30:32-46. [PMID: 34091053 PMCID: PMC8753288 DOI: 10.1016/j.ymthe.2021.06.003] [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] [Received: 11/26/2020] [Revised: 03/15/2021] [Accepted: 05/27/2021] [Indexed: 02/09/2023] Open
Abstract
CRISPR-Cas9 is rapidly entering molecular biology and biomedicine as a promising gene-editing tool. A unique feature of CRISPR-Cas9 is a single-guide RNA directing a Cas9 nuclease toward its genomic target. Herein, we highlight new approaches for improving cellular uptake and endosomal escape of CRISPR-Cas9. As opposed to other recently published works, this review is focused on non-viral carriers as a means to facilitate the cellular uptake of CRISPR-Cas9 through endocytosis. The majority of non-viral carriers, such as gold nanoparticles, polymer nanoparticles, lipid nanoparticles, and nanoscale zeolitic imidazole frameworks, is developed with a focus toward optimizing the endosomal escape of CRISPR-Cas9 by taking advantage of the acidic environment in the late endosomes. Among the most broadly used methods for in vitro and ex vivo ribonucleotide protein transfection are electroporation and microinjection. Thus, other delivery formats are warranted for in vivo delivery of CRISPR-Cas9. Herein, we specifically revise the use of peptide and nanoparticle-based systems as platforms for CRISPR-Cas9 delivery in vivo. Finally, we highlight future perspectives of the CRISPR-Cas9 gene-editing tool and the prospects of using non-viral vectors to improve its bioavailability and therapeutic potential.
Collapse
Affiliation(s)
- Maja van Hees
- Department of Chemistry, Technical University of Denmark, 2800 Lyngby, Denmark
| | - Sofie Slott
- Department of Chemistry, Technical University of Denmark, 2800 Lyngby, Denmark
| | | | - Heon Seok Kim
- School of Medicine, Stanford University, Stanford, CA 94350, USA
| | - Hanlee P. Ji
- School of Medicine, Stanford University, Stanford, CA 94350, USA
| | - Kira Astakhova
- Department of Chemistry, Technical University of Denmark, 2800 Lyngby, Denmark,Corresponding author: Kira Astakhova, Department of Chemistry, Technical University of Denmark, 2800 Lyngby, Denmark.
| |
Collapse
|
10
|
Manouchehri S, Zarrintaj P, Saeb MR, Ramsey JD. Advanced Delivery Systems Based on Lysine or Lysine Polymers. Mol Pharm 2021; 18:3652-3670. [PMID: 34519501 DOI: 10.1021/acs.molpharmaceut.1c00474] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Polylysine and materials that integrate lysine form promising drug delivery platforms. As a cationic macromolecule, a polylysine polymer electrostatically interacts with cells and is efficiently internalized, thereby enabling intracellular delivery. Although polylysine is intrinsically pH-responsive, the conjugation with different functional groups imparts smart, stimuli-responsive traits by adding pH-, temperature-, hypoxia-, redox-, and enzyme-responsive features for enhanced delivery of therapeutic agents. Because of such characteristics, polylysine has been used to deliver various cargos such as small-molecule drugs, genes, proteins, and imaging agents. Furthermore, modifying contrast agents with polylysine has been shown to improve performance, including increasing cellular uptake and stability. In this review, the use of lysine residues, peptides, and polymers in various drug delivery systems has been discussed comprehensively to provide insight into the design and robust manufacturing of lysine-based delivery platforms.
Collapse
Affiliation(s)
- Saeed Manouchehri
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, Oklahoma 74078, United States
| | - Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, Oklahoma 74078, United States
| | | | - Joshua D Ramsey
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, Oklahoma 74078, United States
| |
Collapse
|
11
|
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: 167] [Impact Index Per Article: 41.8] [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
|
12
|
The role of the multifunctional antimicrobial peptide melittin in gene delivery. Drug Discov Today 2021; 26:1053-1059. [DOI: 10.1016/j.drudis.2021.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 12/01/2020] [Accepted: 01/07/2021] [Indexed: 12/23/2022]
|
13
|
Lim MSH, Nishiyama Y, Ohtsuki T, Watanabe K, Kobuchi H, Kobayashi K, Matsuura E. Lactosome-Conjugated siRNA Nanoparticles for Photo-Enhanced Gene Silencing in Cancer Cells. J Pharm Sci 2021; 110:1788-1798. [PMID: 33529684 DOI: 10.1016/j.xphs.2021.01.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/14/2020] [Accepted: 01/25/2021] [Indexed: 12/13/2022]
Abstract
The A3B-type Lactosome comprised of poly(sarcosine)3-block-poly(l-lactic acid), a biocompatible and biodegradable polymeric nanomicelle, was reported to accumulate in tumors in vivo via the enhanced permeability and retention (EPR) effect. Recently, the cellular uptake of Lactosome particles was enhanced through the incorporation of a cell-penetrating peptide (CPP), L7EB1. However, the ability of Lactosome as a drug delivery carrier has not been established. Herein, we have developed a method to conjugate the A3B-type Lactosome with ATP-binding cassette transporter G2 (ABCG2) siRNA for inducing in vitro apoptosis in the cancer cell lines PANC-1 and NCI-H226. The L7EB1 peptide facilitates the cellular uptake efficiency of Lactosome but does not deliver siRNA into cytosol. To establish the photoinduced cytosolic dispersion of siRNA, a photosensitizer loaded L7EB1-Lactosome was prepared, and the photosensitizer 5,10,15,20-tetra-kis(pentafluorophenyl)porphyrin (TPFPP) showed superiority in photoinduced cytosolic dispersion. We exploited the combined effects of enhanced cellular uptake by L7EB1 and photoinduced endosomal escape by TPFPP to efficiently deliver ABCG2 siRNA into the cytosol for gene silencing. Moreover, the silencing of ABCG2, a protoporphyrin IX (PpIX) transporter, also mediated photoinduced cell death via 5-aminolevulinic acid (ALA)-mediated PpIX accumulated photodynamic therapy (PDT). The synergistic capability of the L7EB1/TPFPP/siRNA-Lactosome complex enabled both gene silencing and PDT.
Collapse
Affiliation(s)
- Melissa Siaw Han Lim
- Department of Cell Chemistry, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yuki Nishiyama
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushima-naka, Okayama 700-8530, Japan
| | - Takashi Ohtsuki
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushima-naka, Okayama 700-8530, Japan.
| | - Kazunori Watanabe
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushima-naka, Okayama 700-8530, Japan
| | - Hirotsugu Kobuchi
- Department of Cell Chemistry, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kazuko Kobayashi
- Department of Cell Chemistry, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Eiji Matsuura
- Department of Cell Chemistry, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; Neutron Therapy Research Centre, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; Collaborative Research Centre for OMIC, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
14
|
Freitag F, Wagner E. Optimizing synthetic nucleic acid and protein nanocarriers: The chemical evolution approach. Adv Drug Deliv Rev 2021; 168:30-54. [PMID: 32246984 DOI: 10.1016/j.addr.2020.03.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/10/2020] [Accepted: 03/30/2020] [Indexed: 12/20/2022]
Abstract
Optimizing synthetic nanocarriers is like searching for a needle in a haystack. How to find the most suitable carrier for intracellular delivery of a specified macromolecular nanoagent for a given disease target location? Here, we review different synthetic 'chemical evolution' strategies that have been pursued. Libraries of nanocarriers have been generated either by unbiased combinatorial chemistry or by variation and novel combination of known functional delivery elements. As in natural evolution, definition of nanocarriers as sequences, as barcode or design principle, may fuel chemical evolution. Screening in appropriate test system may not only provide delivery candidates, but also a refined understanding of cellular delivery including novel, unpredictable mechanisms. Combined with rational design and computational algorithms, candidates can be further optimized in subsequent evolution cycles into nanocarriers with improved safety and efficacy. Optimization of nanocarriers differs for various cargos, as illustrated for plasmid DNA, siRNA, mRNA, proteins, or genome-editing nucleases.
Collapse
|
15
|
Shen J, Chen J, Ma J, Fan L, Zhang X, Yue T, Yan Y, Zhang Y. Enhanced lysosome escape mediated by 1,2-dicarboxylic-cyclohexene anhydride-modified poly-l-lysine dendrimer as a gene delivery system. Asian J Pharm Sci 2020; 15:759-776. [PMID: 33363631 PMCID: PMC7750821 DOI: 10.1016/j.ajps.2019.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/07/2019] [Accepted: 12/10/2019] [Indexed: 12/23/2022] Open
Abstract
Antisense oligodeoxynucleotide (ASODN) can directly interfere a series of biological events of the target RNA derived from tumor cells through Watson-Crick base pairing, in turn, plays antitumor therapeutic roles. In the study, a novel HIF-1α ASODN-loaded nanocomposite was formulated to efficiently deliver gene to the target RNA. The physicochemical properties of nanocomposite were characterized using TEM, FTIR, DLS and zeta potentials. The mean diameter of resulting GEL-DGL-FA-ASODN-DCA nanocomposite was about 170-192 nm, and according to the agarose gel retardation assay, the loading amount of ASODN accounted for 166.7 mg/g. The results of cellular uptake showed that the nanocomposite could specifically target to HepG2 and Hela cells. The cytotoxicity assay demonstrated that the toxicity of vectors was greatly reduced by using DCA to reversibly block the cationic DGL. The subcellular distribution images clearly displayed the lysosomal escape ability of the DCA-modified nanocomposite. In vitro exploration of molecular mechanism indicated that the nanocomposite could inhibit mRNA expression and HIF-1α protein translation at different levels. In vivo optical images and quantitative assay testified that the formulation accumulated preferentially in the tumor tissue. In vivo antitumor efficacy research confirmed that this nanocomposite had significant antitumor activity and the tumor inhibitory rate was 77.99%. These results manifested that the GEL-DGL-FA-ASODN-DCA nanocomposite was promising in gene therapeutics for antitumor by interacting directly with target RNA.
Collapse
Affiliation(s)
- Jianmin Shen
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.,Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Jing Chen
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.,Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Jingbo Ma
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.,Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Linlan Fan
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Xiaoli Zhang
- Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Ting Yue
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.,Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Yaping Yan
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Yuhang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
16
|
Naik S, Shreya AB, Raychaudhuri R, Pandey A, Lewis SA, Hazarika M, Bhandary SV, Rao BSS, Mutalik S. Small interfering RNAs (siRNAs) based gene silencing strategies for the treatment of glaucoma: Recent advancements and future perspectives. Life Sci 2020; 264:118712. [PMID: 33159955 DOI: 10.1016/j.lfs.2020.118712] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/28/2020] [Accepted: 10/31/2020] [Indexed: 01/22/2023]
Abstract
RNA-interference-based mechanisms, especially the use of small interfering RNAs (siRNAs), have been under investigation for the treatment of several ailments and have shown promising results for ocular diseases including glaucoma. The eye, being a confined compartment, serves as a good target for the delivery of siRNAs. This review focuses on siRNA-based strategies for gene silencing to treat glaucoma. We have discussed the ocular structures and barriers to gene therapy (tear film, corneal, conjunctival, vitreous, and blood ocular barriers), methods of administration for ocular gene delivery (topical instillation, periocular, intracameral, intravitreal, subretinal, and suprachoroidal routes) and various viral and non-viral vectors in siRNA-based therapy for glaucoma. The components and mechanism of siRNA-based gene silencing have been mentioned briefly followed by the basic strategies and challenges faced during siRNA therapeutics development. We have emphasized different therapeutic targets for glaucoma which have been under research by scientists and the current siRNA-based drugs used in glaucoma treatment. We also mention briefly strategies for siRNA-based treatment after glaucoma surgery.
Collapse
Affiliation(s)
- Santoshi Naik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Ajjappla Basavaraj Shreya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Ruchira Raychaudhuri
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Abhijeet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Shaila A Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Manali Hazarika
- Department of Ophthalmology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Sulatha V Bhandary
- Department of Ophthalmology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Bola Sadashiva Satish Rao
- Director - Research, Directorte of Research, Manipal Academy of Higher Education, Manipal and School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India.
| |
Collapse
|
17
|
Bidram E, Esmaeili Y, Ranji-Burachaloo H, Al-Zaubai N, Zarrabi A, Stewart A, Dunstan DE. A concise review on cancer treatment methods and delivery systems. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101350] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
18
|
Rai MF, Pan H, Yan H, Sandell LJ, Pham CTN, Wickline SA. Applications of RNA interference in the treatment of arthritis. Transl Res 2019; 214:1-16. [PMID: 31351032 PMCID: PMC6848781 DOI: 10.1016/j.trsl.2019.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/02/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022]
Abstract
RNA interference (RNAi) is a cellular mechanism for post-transcriptional gene regulation mediated by small interfering RNA (siRNA) and microRNA. siRNA-based therapy holds significant promise for the treatment of a wide-range of arthritic diseases. siRNA selectively suppresses the expression of a gene product and can thus achieve the specificity that is lacking in small molecule inhibitors. The potential use of siRNA-based therapy in arthritis, however, has not progressed to clinical trials despite ample evidence for efficacy in preclinical studies. One of the main challenges to clinical translation is the lack of a suitable delivery vehicle to efficiently and safely access diverse pathologies. Moreover, the ideal targets in treatment of arthritides remain elusive given the complexity and heterogeneity of these disease pathogeneses. Herein, we review recent preclinical studies that use RNAi-based drug delivery systems to mitigate inflammation in models of rheumatoid arthritis and osteoarthritis. We discuss a self-assembling peptide-based nanostructure that demonstrates the potential of overcoming many of the critical barriers preventing the translation of this technology to the clinic.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, Missouri; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Hua Pan
- Department of Cardiovascular Sciences, University of South Florida Health Heart Institute, Morsani School of Medicine, Tampa, Florida
| | - Huimin Yan
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Linda J Sandell
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, Missouri; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Christine T N Pham
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri.
| | - Samuel A Wickline
- Department of Cardiovascular Sciences, University of South Florida Health Heart Institute, Morsani School of Medicine, Tampa, Florida
| |
Collapse
|
19
|
Anilmis NM, Kara G, Kilicay E, Hazer B, Denkbas EB. Designing siRNA-conjugated plant oil-based nanoparticles for gene silencing and cancer therapy. J Microencapsul 2019; 36:635-648. [PMID: 31509450 DOI: 10.1080/02652048.2019.1665117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In this study, the anticancer activities of two siRNA carriers were compared using a human lung adenocarcinoma epithelial cell line (A549). Firstly, poly(styrene)-graft-poly(linoleic acid) (PS-g-PLina) and poly(styrene)-graft-poly(linoleic acid)-graft-poly(ethylene glycol) (PS-g-PLina-g-PEG) graft copolymers were synthesized by free-radical polymerization. PS-PLina and PS-PLina-PEG nanoparticles (NPs) were prepared by solvent evaporation method and were then characterized. The size was found as 150 ± 10 nm for PS-PLina and 184 ± 6 nm for PS-PLina-PEG NPs. The NPs were functionalized with poly(l-lysine) (PLL) for c-myc siRNA conjugation. siRNA entrapment efficiencies were found in the range of 4-63% for PS-PLina-PLL and 6-42% for PS-PLina-PEG-PLL NPs. The short-term stability test was realised for 1 month. siRNA release profiles were also investigated. In vitro anticancer activity of siRNA-NPs was determined by MTT, flow cytometry, and fluorescence microscopy analyses. Obtained findings showed that both NPs systems were promising as siRNA delivery tool for lung cancer therapy.
Collapse
Affiliation(s)
- Nur Merve Anilmis
- Nanotechnology Engineering Division, Institute of Science and Technology, Bulent Ecevit University , Zonguldak , Turkey
| | - Goknur Kara
- Department of Chemistry, Biochemistry Division,Hacettepe University , Ankara , Turkey
| | - Ebru Kilicay
- Vocational School of Higher Education, Programme of Biomedical Device Technology, Bulent Ecevit University , Zonguldak , Turkey
| | - Baki Hazer
- Department of Aircraft Mechanic-Engine Maintenance, Cappadocia University , Urgup , Nevsehir , Turkey.,Department of Chemistry, Bulent Ecevit University, Universite Caddes , Zonguldak , Turkey.,Department of Nanotechnology Engineering, Bulent Ecevit University , Zonguldak , Turkey.,Department of Biomedical Engineering, Baskent, University , Ankara , Turkey
| | - Emir Baki Denkbas
- Department of Chemistry, Biochemistry Division,Hacettepe University , Ankara , Turkey.,Department of Biomedical Engineering, Baskent, University , Ankara , Turkey
| |
Collapse
|
20
|
Peng L, Wagner E. Polymeric Carriers for Nucleic Acid Delivery: Current Designs and Future Directions. Biomacromolecules 2019; 20:3613-3626. [DOI: 10.1021/acs.biomac.9b00999] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
21
|
Kandil R, Xie Y, Heermann R, Isert L, Jung K, Mehta A, Merkel OM. Coming in and Finding Out: Blending Receptor-Targeted Delivery and Efficient Endosomal Escape in a Novel Bio-Responsive siRNA Delivery System for Gene Knockdown in Pulmonary T Cells. ADVANCED THERAPEUTICS 2019; 2:1900047. [PMID: 31372493 PMCID: PMC6675603 DOI: 10.1002/adtp.201900047] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Indexed: 12/11/2022]
Abstract
RNA interference (RNAi) offers the potential to selectively silence disease-related genes in defined cell subsets. Translation into the clinical routine is, however, still hampered by the lack of efficient carrier systems for therapeutic siRNA, endosomal entrapment presenting a major hurdle. A promising siRNA delivery system has previously been developed on the base of polyethylenimine (PEI) and the targeting ligand transferrin (Tf) to specifically reach activated T cells in the lung. In the present work, the focus is on optimizing Tf-PEI polyplexes for gene knockdown in primary activated T cells by improving their endosomal escape properties. Blending of the conjugate with membrane lytic melittin significantly enhanced endosomal release and thereby cytoplasmic delivery, while maintaining selective T cell targeting abilities and overall cell tolerability. The gathered data furthermore demonstrate that melittin addition also distinctly improves several other essential particle characteristics, such as siRNA encapsulation efficiency and stability in lung lining fluids. In conclusion, this results in a novel upgraded siRNA delivery system that is not only able to specifically deliver its payload to the desired target cells via receptor-mediated endocytosis, but also shows enhanced release from endosomal vesicles in order to initiate RNAi in the cytoplasm.
Collapse
Affiliation(s)
- Rima Kandil
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, Butenandtstraße 5-13, 81337 Munich, Germany
| | - Yuran Xie
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI 48201, USA
| | - Ralf Heermann
- Institute for Molecular Physiology, Microbiology and Wine Research, Johannes-Gutenberg-University, Johann-Joachim-Becher-Weg 13, 55128 Mainz, Germany; Biocenter, Department Microbiology, Ludwig-Maximilians-University, Großhaderner Str. 2-4, 82152 Martinsried, Germany
| | - Lorenz Isert
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, Butenandtstraße 5-13, 81337 Munich, Germany
| | - Kirsten Jung
- Biocenter, Department Microbiology, Ludwig-Maximilians-University, Großhaderner Str. 2-4, 82152 Martinsried, Germany
| | - Aditi Mehta
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, Butenandtstraße 5-13, 81337 Munich, Germany
| | - Olivia M. Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, Butenandtstraße 5-13, 81337 Munich, Germany
| |
Collapse
|
22
|
Current Aspects of siRNA Bioconjugate for In Vitro and In Vivo Delivery. Molecules 2019; 24:molecules24122211. [PMID: 31200490 PMCID: PMC6631009 DOI: 10.3390/molecules24122211] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/03/2019] [Accepted: 06/08/2019] [Indexed: 02/07/2023] Open
Abstract
Studies on siRNA delivery have seen intense growth in the past decades since siRNA has emerged as a new class of gene therapeutics for the treatment of various diseases. siRNA bioconjugate, as one of the major delivery strategies, offers the potential to enhance and broaden pharmacological properties of siRNA, while minimizing the heterogeneity and stability-correlated toxicology. This review summarizes the recent developments of siRNA bioconjugate, including the conjugation with antibody, peptide, aptamer, small chemical, lipidoid, cell-penetrating peptide polymer, and nanoparticle. These siRNA bioconjugate, either administrated alone or formulated with other agents, could significantly improve pharmacokinetic behavior, enhance the biological half-life, and increase the targetability while maintaining sufficient gene silencing activity, with a concomitant improvement of the therapeutic outcomes and diminishment of adverse effects. This review emphasizes the delivery application of these siRNA bioconjugates, especially the conjugation strategy that control the integrity, stability and release of siRNA bioconjugates. The limitations conferred by these conjugation strategies have also been covered.
Collapse
|
23
|
Chernikov IV, Vlassov VV, Chernolovskaya EL. Current Development of siRNA Bioconjugates: From Research to the Clinic. Front Pharmacol 2019; 10:444. [PMID: 31105570 PMCID: PMC6498891 DOI: 10.3389/fphar.2019.00444] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022] Open
Abstract
Small interfering RNAs (siRNAs) acting via RNA interference mechanisms are able to recognize a homologous mRNA sequence in the cell and induce its degradation. The main problems in the development of siRNA-based drugs for therapeutic use are the low efficiency of siRNA delivery to target cells and the degradation of siRNAs by nucleases in biological fluids. Various approaches have been proposed to solve the problem of siRNA delivery in vivo (e.g., viruses, cationic lipids, polymers, nanoparticles), but all have limitations for therapeutic use. One of the most promising approaches to solve the problem of siRNA delivery to target cells is bioconjugation; i.e., the covalent connection of siRNAs with biogenic molecules (lipophilic molecules, antibodies, aptamers, ligands, peptides, or polymers). Bioconjugates are "ideal nanoparticles" since they do not need a positive charge to form complexes, are less toxic, and are less effectively recognized by components of the immune system because of their small size. This review is focused on strategies and principles for constructing siRNA bioconjugates for in vivo use.
Collapse
Affiliation(s)
- Ivan V Chernikov
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Valentin V Vlassov
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Elena L Chernolovskaya
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
24
|
Peeler DJ, Sellers DL, Pun SH. pH-Sensitive Polymers as Dynamic Mediators of Barriers to Nucleic Acid Delivery. Bioconjug Chem 2018; 30:350-365. [PMID: 30398844 DOI: 10.1021/acs.bioconjchem.8b00695] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The nonviral delivery of exogenous nucleic acids (NA) into cells for therapeutic purposes has rapidly matured into tangible clinical impact. Synthetic polymers are particularly attractive vectors for NA delivery due to their relatively inexpensive production compared to viral alternatives and their highly tailorable chemical properties; indeed, many preclinical investigations have revealed the primary biological barriers to nonviral NA delivery by systematically varying polymeric material properties. This review focuses on applications of pH-sensitive chemistries that enable polymeric vectors to serially address multiple biological barriers to NA delivery. In particular, we focus on recent innovations with in vivo evaluation that dynamically enable colloidal stability, cellular uptake, endosomal escape, and nucleic acid release. We conclude with a summary of successes to date and projected areas for impactful future research.
Collapse
Affiliation(s)
- David J Peeler
- Department of Bioengineering and Molecular Engineering and Sciences Institute , University of Washington , Seattle , Washington 98195 , United States
| | - Drew L Sellers
- Department of Bioengineering and Molecular Engineering and Sciences Institute , University of Washington , Seattle , Washington 98195 , United States
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute , University of Washington , Seattle , Washington 98195 , United States
| |
Collapse
|
25
|
Hager S, Wagner E. Bioresponsive polyplexes - chemically programmed for nucleic acid delivery. Expert Opin Drug Deliv 2018; 15:1067-1083. [PMID: 30247975 DOI: 10.1080/17425247.2018.1526922] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The whole delivery process of nucleic acids is very challenging. Appropriate carrier systems are needed, which show extracellular stability and intracellular disassembly. Viruses have developed various strategies to meet these requirements, as they are optimized by biological evolution to transfer genetic information into host cells. Taking viruses as models, smart synthetic carriers can be designed, mimicking the efficient delivery process of viral infection. These 'synthetic viruses' are pre-programmed and respond to little differences in their microenvironment, caused by either exogenous or endogenous stimuli. AREAS COVERED This review deals with polymer-based, bioresponsive nanosystems (polyplexes) for the delivery of nucleic acids. Strategies utilizing pH-responsiveness, redox-responsiveness as well as sensitivity towards enzymes will be described more in detail. Systems, which respond to other endogenous triggers (i.e. reactive oxygen species, adenosine triphosphate, hypoxia), will be briefly illustrated. Moreover, some examples for combined bioresponsiveness will be presented. EXPERT OPINION Bioresponsive polyplexes are a smart way to facilitate programmed, timely delivery of nucleic acids to desired, specific sites. Nevertheless, further optimization is necessary to improve the still moderate transfection efficiency and specificity - also in regard to medical translation. For this purpose, precise carrier structures are desirable and stability issues of bioresponsive systems must be considered.
Collapse
Affiliation(s)
- Simone Hager
- a Pharmaceutical Biotechnology, Department of Pharmacy , Ludwig-Maximilians-Universität , Munich , Germany
| | - Ernst Wagner
- a Pharmaceutical Biotechnology, Department of Pharmacy , Ludwig-Maximilians-Universität , Munich , Germany
| |
Collapse
|
26
|
Klein PM, Kern S, Lee DJ, Schmaus J, Höhn M, Gorges J, Kazmaier U, Wagner E. Folate receptor-directed orthogonal click-functionalization of siRNA lipopolyplexes for tumor cell killing in vivo. Biomaterials 2018; 178:630-642. [DOI: 10.1016/j.biomaterials.2018.03.031] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/15/2018] [Accepted: 03/17/2018] [Indexed: 12/11/2022]
|
27
|
Nakamoto K, Akao Y, Furuichi Y, Ueno Y. Enhanced Intercellular Delivery of cRGD-siRNA Conjugates by an Additional Oligospermine Modification. ACS OMEGA 2018; 3:8226-8232. [PMID: 30087937 PMCID: PMC6072241 DOI: 10.1021/acsomega.8b00850] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 06/07/2018] [Indexed: 05/07/2023]
Abstract
Small interfering RNA (siRNA), consisting a 21-mer duplex molecule, is often modified by conjugation with specific ligands to enhance its capacity for tissue-specific delivery. However, these attempts are hampered by the low permeability of negatively charged RNA molecules to enter the cell membrane. In this study, we designed and synthesized siRNA conjugates modified with cationic oligospermine and cyclic RGD (cRGD) to overcome the low-membrane permeability of siRNA. The siRNA conjugate, which contains 15 spermines and a cRGD peptide, showed sufficient gene-silencing activity at 250 nM final concentration without a transfection reagent. Under these conditions, the cationic oligospermine and cRGD-siRNA conjugate did not show any cytotoxicity.
Collapse
Affiliation(s)
- Kosuke Nakamoto
- United
Graduate School of Agricultural Science, United Graduate School of Drug
Discovery and Medical Information Sciences, and Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yukihiro Akao
- United
Graduate School of Agricultural Science, United Graduate School of Drug
Discovery and Medical Information Sciences, and Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yasuhiro Furuichi
- GeneCare
Research Institute Co., Ltd., 19-2 Kajiwara, Kamakura 247-0063, Japan
| | - Yoshihito Ueno
- United
Graduate School of Agricultural Science, United Graduate School of Drug
Discovery and Medical Information Sciences, and Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Center
of Highly Advanced Integration of Nano and Life Sciences, Gifu University (G-CHAIN), 1-1 Yanagido, Gifu 501-1193, Japan
- E-mail: . Phone: +81-58-293-2919. Fax: +81-58-293-2919 (Y.U.)
| |
Collapse
|
28
|
Klein PM, Klinker K, Zhang W, Kern S, Kessel E, Wagner E, Barz M. Efficient Shielding of Polyplexes Using Heterotelechelic Polysarcosines. Polymers (Basel) 2018; 10:E689. [PMID: 30966723 PMCID: PMC6404158 DOI: 10.3390/polym10060689] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/13/2018] [Accepted: 06/17/2018] [Indexed: 11/16/2022] Open
Abstract
Shielding agents are commonly used to shield polyelectrolyte complexes, e.g., polyplexes, from agglomeration and precipitation in complex media like blood, and thus enhance their in vivo circulation times. Since up to now primarily poly(ethylene glycol) (PEG) has been investigated to shield non-viral carriers for systemic delivery, we report on the use of polysarcosine (pSar) as a potential alternative for steric stabilization. A redox-sensitive, cationizable lipo-oligomer structure (containing two cholanic acids attached via a bioreducible disulfide linker to an oligoaminoamide backbone in T-shape configuration) was equipped with azide-functionality by solid phase supported synthesis. After mixing with small interfering RNA (siRNA), lipopolyplexes formed spontaneously and were further surface-functionalized with polysarcosines. Polysarcosine was synthesized by living controlled ring-opening polymerization using an azide-reactive dibenzo-aza-cyclooctyne-amine as an initiator. The shielding ability of the resulting formulations was investigated with biophysical assays and by near-infrared fluorescence bioimaging in mice. The modification of ~100 nm lipopolyplexes was only slightly increased upon functionalization. Cellular uptake into cells was strongly reduced by the pSar shielding. Moreover, polysarcosine-shielded polyplexes showed enhanced blood circulation times in bioimaging studies compared to unshielded polyplexes and similar to PEG-shielded polyplexes. Therefore, polysarcosine is a promising alternative for the shielding of non-viral, lipo-cationic polyplexes.
Collapse
Affiliation(s)
- Philipp Michael Klein
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | - Kristina Klinker
- Institute of Organic Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, D-55128 Mainz, Germany.
- Graduate School Materials Science in Mainz, Staudinger Weg 9, 55128 Mainz, Germany.
| | - Wei Zhang
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | - Sarah Kern
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | - Eva Kessel
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | - Ernst Wagner
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
- Nanosystems Initiative Munich, Schellingstraße 4, D-80799 Munich, Germany.
| | - Matthias Barz
- Institute of Organic Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, D-55128 Mainz, Germany.
| |
Collapse
|
29
|
Feng Y, Yin Z, Zhang D, Srivastava A, Ling C. Chinese Medicine Protein and Peptide in Gene and Cell Therapy. Curr Protein Pept Sci 2018; 20:251-264. [PMID: 29895243 DOI: 10.2174/1389203719666180612082432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 04/10/2018] [Accepted: 05/22/2018] [Indexed: 01/05/2023]
Abstract
The success of gene and cell therapy in clinic during the past two decades as well as our expanding ability to manipulate these biomaterials are leading to new therapeutic options for a wide range of inherited and acquired diseases. Combining conventional therapies with this emerging field is a promising strategy to treat those previously-thought untreatable diseases. Traditional Chinese medicine (TCM) has evolved for thousands of years in China and still plays an important role in human health. As part of the active ingredients of TCM, proteins and peptides have attracted long-term enthusiasm of researchers. More recently, they have been utilized in gene and cell therapy, resulting in promising novel strategies to treat both cancer and non-cancer diseases. This manuscript presents a critical review on this field, accompanied with perspectives on the challenges and new directions for future research in this emerging frontier.
Collapse
Affiliation(s)
- Yinlu Feng
- Department of Traditional Chinese Medicine, 401 Hospital of the Chinese People's Liberation Army, Qingdao, Shandong 266071, China.,Division of Cellular and Molecular Therapy, Department of Pediatrics, College of Medicine, University of Florida, Gainesville 32611, FL, United States
| | - Zifei Yin
- Division of Cellular and Molecular Therapy, Department of Pediatrics, College of Medicine, University of Florida, Gainesville 32611, FL, United States
| | - Daniel Zhang
- Division of Cellular and Molecular Therapy, Department of Pediatrics, College of Medicine, University of Florida, Gainesville 32611, FL, United States
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, College of Medicine, University of Florida, Gainesville 32611, FL, United States
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, College of Medicine, University of Florida, Gainesville 32611, FL, United States
| |
Collapse
|
30
|
Song YF, Liu DZ, Cheng Y, Teng ZH, Cui H, Liu M, Zhang BL, Mei QB, Zhou SY. Charge Reversible and Mitochondria/Nucleus Dual Target Lipid Hybrid Nanoparticles To Enhance Antitumor Activity of Doxorubicin. Mol Pharm 2018; 15:1296-1308. [PMID: 29432025 DOI: 10.1021/acs.molpharmaceut.7b01109] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The experiment aims to increase antitumor activity while decreasing the systemic toxicity of doxorubicin (DOX). Charge reversible and mitochondria/nucleus dual target lipid hybrid nanoparticles (LNPs) was prepared. The in vitro experimental results indicated that LNPs released more amount of DOX in acidic environment and delivered more amount of DOX to the mitochondria and nucleus of tumor cells than did free DOX, which resulted in the reduction of mitochondrial membrane potential and the enhancement of cytotoxicity of LNPs on tumor cells. Furthermore, the in vivo experimental results indicated that LNPs delivered more DOX to tumor tissue and significantly prolonged the retention time of DOX in tumor tissue as compared with free DOX, which consequently resulted in the high antitumor activity and low systemic toxicity of LNPs on tumor-bearing nude mice. The above results indicated that charge reversible mitochondria/nucleus dual targeted lipid hybrid nanoparticles greatly enhanced therapeutic efficacy of DOX for treating lung cancer.
Collapse
|
31
|
Grijalvo S, Alagia A, Jorge AF, Eritja R. Covalent Strategies for Targeting Messenger and Non-Coding RNAs: An Updated Review on siRNA, miRNA and antimiR Conjugates. Genes (Basel) 2018; 9:E74. [PMID: 29415514 PMCID: PMC5852570 DOI: 10.3390/genes9020074] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/26/2018] [Accepted: 01/26/2018] [Indexed: 12/11/2022] Open
Abstract
Oligonucleotide-based therapy has become an alternative to classical approaches in the search of novel therapeutics involving gene-related diseases. Several mechanisms have been described in which demonstrate the pivotal role of oligonucleotide for modulating gene expression. Antisense oligonucleotides (ASOs) and more recently siRNAs and miRNAs have made important contributions either in reducing aberrant protein levels by sequence-specific targeting messenger RNAs (mRNAs) or restoring the anomalous levels of non-coding RNAs (ncRNAs) that are involved in a good number of diseases including cancer. In addition to formulation approaches which have contributed to accelerate the presence of ASOs, siRNAs and miRNAs in clinical trials; the covalent linkage between non-viral vectors and nucleic acids has also added value and opened new perspectives to the development of promising nucleic acid-based therapeutics. This review article is mainly focused on the strategies carried out for covalently modifying siRNA and miRNA molecules. Examples involving cell-penetrating peptides (CPPs), carbohydrates, polymers, lipids and aptamers are discussed for the synthesis of siRNA conjugates whereas in the case of miRNA-based drugs, this review article makes special emphasis in using antagomiRs, locked nucleic acids (LNAs), peptide nucleic acids (PNAs) as well as nanoparticles. The biomedical applications of siRNA and miRNA conjugates are also discussed.
Collapse
Affiliation(s)
- Santiago Grijalvo
- Institute of Advanced Chemistry of Catalonia (IQAC, CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain.
- Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Jordi Girona 18-26, 08034 Barcelona, Spain.
| | - Adele Alagia
- Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Jordi Girona 18-26, 08034 Barcelona, Spain.
| | - Andreia F Jorge
- Coimbra Chemistry Centre, (CQC), Department of Chemistry, University of Coimbra, Rua Larga, 3004-535 Coimbra, Portugal.
| | - Ramon Eritja
- Institute of Advanced Chemistry of Catalonia (IQAC, CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain.
- Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Jordi Girona 18-26, 08034 Barcelona, Spain.
| |
Collapse
|
32
|
Matsumoto A, Miyahara Y. 'Borono-lectin' based engineering as a versatile platform for biomedical applications. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2018; 19:18-30. [PMID: 29296128 PMCID: PMC5738650 DOI: 10.1080/14686996.2017.1411143] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/26/2017] [Accepted: 11/27/2017] [Indexed: 05/23/2023]
Abstract
Boronic acids are well known for their ability to reversibly interact with the diol groups, a common motif of biomolecules including sugars and ribose. Due to their ability to interact with carbohydrates, they can be regarded as synthetic mimics of lectins, termed 'borono-lectins'. The borono-lectins can be tailored to elicit a broad profile of binding strength and specificity. This special property has been translated into many creative biomedical applications in a way interactive with biology. This review provides a brief overview of recent efforts of polymeric materials-based engineering taking advantage of such virtue of 'borono-lectins' chemistry, related to the field of biomaterials and drug delivery applications.
Collapse
Affiliation(s)
- Akira Matsumoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
- Kanagawa Institute of Industrial Science and Technology (KISTEC-KAST), Kawasaki, Japan
| | - Yuji Miyahara
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
33
|
Sun Y, Yang Z, Wang C, Yang T, Cai C, Zhao X, Yang L, Ding P. Exploring the role of peptides in polymer-based gene delivery. Acta Biomater 2017; 60:23-37. [PMID: 28778533 DOI: 10.1016/j.actbio.2017.07.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/14/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022]
Abstract
Polymers are widely studied as non-viral gene vectors because of their strong DNA binding ability, capacity to carry large payload, flexibility of chemical modifications, low immunogenicity, and facile processes for manufacturing. However, high cytotoxicity and low transfection efficiency substantially restrict their application in clinical trials. Incorporating functional peptides is a promising approach to address these issues. Peptides demonstrate various functions in polymer-based gene delivery systems, such as targeting to specific cells, breaching membrane barriers, facilitating DNA condensation and release, and lowering cytotoxicity. In this review, we systematically summarize the role of peptides in polymer-based gene delivery, and elaborate how to rationally design polymer-peptide based gene delivery vectors. STATEMENT OF SIGNIFICANCE Polymers are widely studied as non-viral gene vectors, but suffer from high cytotoxicity and low transfection efficiency. Incorporating short, bioactive peptides into polymer-based gene delivery systems can address this issue. Peptides demonstrate various functions in polymer-based gene delivery systems, such as targeting to specific cells, breaching membrane barriers, facilitating DNA condensation and release, and lowering cytotoxicity. In this review, we highlight the peptides' roles in polymer-based gene delivery, and elaborate how to utilize various functional peptides to enhance the transfection efficiency of polymers. The optimized peptide-polymer vectors should be able to alter their structures and functions according to biological microenvironments and utilize inherent intracellular pathways of cells, and consequently overcome the barriers during gene delivery to enhance transfection efficiency.
Collapse
Affiliation(s)
- Yanping Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhen Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chunxi Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, USA
| | - Cuifang Cai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaoyun Zhao
- Department of Microbiology and Cell Biology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Li Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
34
|
Ramasamy T, Ruttala HB, Gupta B, Poudel BK, Choi HG, Yong CS, Kim JO. Smart chemistry-based nanosized drug delivery systems for systemic applications: A comprehensive review. J Control Release 2017; 258:226-253. [PMID: 28472638 DOI: 10.1016/j.jconrel.2017.04.043] [Citation(s) in RCA: 246] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 04/28/2017] [Accepted: 04/30/2017] [Indexed: 12/21/2022]
|
35
|
Bhavsar D, Subramanian K, Sethuraman S, Krishnan UM. 'Nano-in-nano' hybrid liposomes increase target specificity and gene silencing efficiency in breast cancer induced SCID mice. Eur J Pharm Biopharm 2017; 119:96-106. [PMID: 28600223 DOI: 10.1016/j.ejpb.2017.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 03/06/2017] [Accepted: 06/05/2017] [Indexed: 02/06/2023]
Abstract
Gene silencing has immense potential in the treatment of cancer. However, enhancement of its efficiency requires the development of specifically targeted and safe carrier systems. Cationic carriers are generally limited by their immunogenicity. Hence, in this study, we report hybrid liposomes encapsulating Poly (L-lysine)-siRNA complex to silence epithelial cell adhesion molecule (EpCAM), highly expressed in epithelial cancers. The hybrid liposomes LL1 (Egg PC:DSPE-PEG, 10:0) and hybrid immunoliposomes LL2 (Egg PC:DSPE-PEG, 8:2) linked with EpCAM antibody as the targeting ligand showed an encapsulation efficiency of 70% and 86%, respectively. LL2 liposomes with a zeta potential of -26mV exhibited good colloidal stability in phosphate buffered saline containing bovine serum albumin and fetal bovine serum at 37°C. Cell uptake studies showed increased uptake of the LL2 when compared to LL1 liposomes. Finally, the hybrid immunoliposomes were evaluated for their efficacy in regressing the tumor volume in SCID mice. Eight doses each of 0.15mg/kg, which is among the lowest reported siRNA concentrations, were administered to the animals. About 45% reduction in tumor volume was achieved after 28days in the mice treated with LL2 when compared with the positive control and LL1 treated groups. Thus, our results demonstrate that the 'nano-in-nano' concept of encapsulating poly (l-Lysine) complexed EpCAM siRNA in immunoliposomes may be a promising strategy to treat EpCAM-positive epithelial cancers, especially as an adjuvant therapy.
Collapse
Affiliation(s)
- Dhiraj Bhavsar
- Centre for Nanotechnology & Advanced Biomaterials, School of Chemical & Biotechnology, SASTRA University, Thanjavur, India
| | - Krishnakumar Subramanian
- L&T Ophthalmic Pathology Department, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Swaminathan Sethuraman
- Centre for Nanotechnology & Advanced Biomaterials, School of Chemical & Biotechnology, SASTRA University, Thanjavur, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials, School of Chemical & Biotechnology, SASTRA University, Thanjavur, India.
| |
Collapse
|
36
|
Sustained delivery of siRNA poly- and lipopolyplexes from porous macromer-crosslinked gelatin gels. Int J Pharm 2017; 526:178-187. [DOI: 10.1016/j.ijpharm.2017.04.065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 01/15/2023]
|
37
|
Yousefpour Marzbali M, Yari Khosroushahi A. Polymeric micelles as mighty nanocarriers for cancer gene therapy: a review. Cancer Chemother Pharmacol 2017; 79:637-649. [DOI: 10.1007/s00280-017-3273-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 03/02/2017] [Indexed: 12/11/2022]
|
38
|
Zhang P, Wagner E. History of Polymeric Gene Delivery Systems. Top Curr Chem (Cham) 2017; 375:26. [PMID: 28181193 DOI: 10.1007/s41061-017-0112-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 01/24/2017] [Indexed: 12/16/2022]
Abstract
As an option for genetic disease treatment and an alternative for traditional cancer chemotherapy, gene therapy achieves significant attention. Nucleic acid delivery, however, remains a main challenge in human gene therapy. Polymer-based delivery systems offer a safer and promising route for therapeutic gene delivery. Over the past five decades, various cationic polymers have been optimized for increasingly effective nucleic acid transfer. This resulted in a chemical evolution of cationic polymers from the first-generation polycations towards bioinspired multifunctional sequence-defined polymers and nanocomposites. With the increasing of knowledge in molecular biological processes and rapid progress of macromolecular chemistry, further improvement of polymeric nucleic acid delivery systems will provide effective tool for gene-based therapy in the near future.
Collapse
Affiliation(s)
- Peng Zhang
- Pharmaceutical Biotechnology, Center for System-Based Drug Research Ludwig-Maximilians-Universität, 81377, Munich, Germany. .,Nanosystems Initiative Munich (NIM), 80799, Munich, Germany.
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug Research Ludwig-Maximilians-Universität, 81377, Munich, Germany.,Nanosystems Initiative Munich (NIM), 80799, Munich, Germany.,Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, 80799, Munich, Germany
| |
Collapse
|
39
|
Oude Blenke E, Sleszynska M, Evers MJW, Storm G, Martin NI, Mastrobattista E. Strategies for the Activation and Release of the Membranolytic Peptide Melittin from Liposomes Using Endosomal pH as a Trigger. Bioconjug Chem 2017; 28:574-582. [DOI: 10.1021/acs.bioconjchem.6b00677] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- E. Oude Blenke
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - M. Sleszynska
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - M. J. W. Evers
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - G. Storm
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - N. I. Martin
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - E. Mastrobattista
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| |
Collapse
|
40
|
Su S, Du FS, Li ZC. Synthesis and pH-dependent hydrolysis profiles of mono- and dialkyl substituted maleamic acids. Org Biomol Chem 2017; 15:8384-8392. [DOI: 10.1039/c7ob02188g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Controlled synthesis and in-depth study on pH-dependent hydrolysis profiles of substituted maleamic acid derivatives.
Collapse
Affiliation(s)
- Shan Su
- Beijing National Laboratory for Molecular Sciences (BNLMS)
- Key Laboratory of Polymer Chemistry & Physics of Ministry of Education
- Department of Polymer Science & Engineering
- College of Chemistry and Molecular Engineering
- Peking University
| | - Fu-Sheng Du
- Beijing National Laboratory for Molecular Sciences (BNLMS)
- Key Laboratory of Polymer Chemistry & Physics of Ministry of Education
- Department of Polymer Science & Engineering
- College of Chemistry and Molecular Engineering
- Peking University
| | - Zi-Chen Li
- Beijing National Laboratory for Molecular Sciences (BNLMS)
- Key Laboratory of Polymer Chemistry & Physics of Ministry of Education
- Department of Polymer Science & Engineering
- College of Chemistry and Molecular Engineering
- Peking University
| |
Collapse
|
41
|
Zhang X, Han L, Liu M, Wang K, Tao L, Wan Q, Wei Y. Recent progress and advances in redox-responsive polymers as controlled delivery nanoplatforms. MATERIALS CHEMISTRY FRONTIERS 2017; 1:807-822. [DOI: 10.1039/c6qm00135a] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Recent advances and progress in redox-responsive polymeric nanosystems for biomedical applications are discussed in this review article.
Collapse
Affiliation(s)
- Xiaoyong Zhang
- Department of Chemistry and Jiangxi Provincial Key Laboratory of New Energy Chemistry
- Nanchang University
- Nanchang 330031
- China
| | - Lu Han
- Department of Chemistry and the Tsinghua Center for Frontier Polymer Research
- Tsinghua University
- P. R. China
| | - Meiying Liu
- Department of Chemistry and Jiangxi Provincial Key Laboratory of New Energy Chemistry
- Nanchang University
- Nanchang 330031
- China
| | - Ke Wang
- Department of Chemistry and the Tsinghua Center for Frontier Polymer Research
- Tsinghua University
- P. R. China
| | - Lei Tao
- Department of Chemistry and the Tsinghua Center for Frontier Polymer Research
- Tsinghua University
- P. R. China
| | - Qing Wan
- Department of Chemistry and Jiangxi Provincial Key Laboratory of New Energy Chemistry
- Nanchang University
- Nanchang 330031
- China
| | - Yen Wei
- Department of Chemistry and the Tsinghua Center for Frontier Polymer Research
- Tsinghua University
- P. R. China
| |
Collapse
|
42
|
Quinn JF, Whittaker MR, Davis TP. Glutathione responsive polymers and their application in drug delivery systems. Polym Chem 2017. [DOI: 10.1039/c6py01365a] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Materials which respond to biological cues are the subject of intense research interest due to their possible application in smart drug delivery vehicles.
Collapse
Affiliation(s)
- John F. Quinn
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology
- Monash Institute of Pharmaceutical Sciences
- Monash University
- Melbourne
- Australia
| | - Michael R. Whittaker
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology
- Monash Institute of Pharmaceutical Sciences
- Monash University
- Melbourne
- Australia
| | - Thomas P. Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology
- Monash Institute of Pharmaceutical Sciences
- Monash University
- Melbourne
- Australia
| |
Collapse
|
43
|
Ahmed M. Peptides, polypeptides and peptide–polymer hybrids as nucleic acid carriers. Biomater Sci 2017; 5:2188-2211. [DOI: 10.1039/c7bm00584a] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Peptide, polypeptide and polymer–peptide hybrid based nucleic acid therapeutics (NAT).
Collapse
Affiliation(s)
- Marya Ahmed
- Department of Chemistry & School of Sustainable Design and Engineering
- University of Prince Edward Island
- Charlottetown
- Canada
| |
Collapse
|
44
|
Munsell EV, Ross NL, Sullivan MO. Journey to the Center of the Cell: Current Nanocarrier Design Strategies Targeting Biopharmaceuticals to the Cytoplasm and Nucleus. Curr Pharm Des 2016; 22:1227-44. [PMID: 26675220 DOI: 10.2174/1381612822666151216151420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/15/2015] [Indexed: 01/06/2023]
Abstract
New biopharmaceutical molecules, potentially able to provide more personalized and effective treatments, are being identified through the advent of advanced synthetic biology strategies, sophisticated chemical synthesis approaches, and new analytical methods to assess biological potency. However, translation of many of these structures has been significantly limited due to the need for more efficient strategies to deliver macromolecular therapeutics to desirable intracellular sites of action. Engineered nanocarriers that encapsulate peptides, proteins, or nucleic acids are generally internalized into target cells via one of several endocytic pathways. These nanostructures, entrapped within endosomes, must navigate the intracellular milieu to orchestrate delivery to the intended destination, typically the cytoplasm or nucleus. For therapeutics active in the cytoplasm, endosomal escape continues to represent a limiting step to effective treatment, since a majority of nanocarriers trapped within endosomes are ultimately marked for enzymatic degradation in lysosomes. Therapeutics active in the nucleus have the added challenges of reaching and penetrating the nuclear envelope, and nuclear delivery remains a preeminent challenge preventing clinical translation of gene therapy applications. Herein, we review cutting-edge peptide- and polymer-based design strategies with the potential to enable significant improvements in biopharmaceutical efficacy through improved intracellular targeting. These strategies often mimic the activities of pathogens, which have developed innate and highly effective mechanisms to penetrate plasma membranes and enter the nucleus of host cells. Understanding these mechanisms has enabled advances in synthetic peptide and polymer design that may ultimately improve intracellular trafficking and bioavailability, leading to increased access to new classes of biotherapeutics.
Collapse
Affiliation(s)
| | | | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, Delaware.
| |
Collapse
|
45
|
Achieving HIV-1 Control through RNA-Directed Gene Regulation. Genes (Basel) 2016; 7:genes7120119. [PMID: 27941595 PMCID: PMC5192495 DOI: 10.3390/genes7120119] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/28/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
HIV-1 infection has been transformed by combined anti-retroviral therapy (ART), changing a universally fatal infection into a controllable infection. However, major obstacles for an HIV-1 cure exist. The HIV latent reservoir, which exists in resting CD4+ T cells, is not impacted by ART, and can reactivate when ART is interrupted or ceased. Additionally, multi-drug resistance can arise. One alternate approach to conventional HIV-1 drug treatment that is being explored involves gene therapies utilizing RNA-directed gene regulation. Commonly known as RNA interference (RNAi), short interfering RNA (siRNA) induce gene silencing in conserved biological pathways, which require a high degree of sequence specificity. This review will provide an overview of the silencing pathways, the current RNAi technologies being developed for HIV-1 gene therapy, current clinical trials, and the challenges faced in progressing these treatments into clinical trials.
Collapse
|
46
|
Lee SH, Kang YY, Jang HE, Mok H. Current preclinical small interfering RNA (siRNA)-based conjugate systems for RNA therapeutics. Adv Drug Deliv Rev 2016; 104:78-92. [PMID: 26514375 DOI: 10.1016/j.addr.2015.10.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 09/01/2015] [Accepted: 10/12/2015] [Indexed: 01/01/2023]
Abstract
Recent promising clinical results of RNA therapeutics have drawn big attention of academia and industries to RNA therapeutics and their carrier systems. To improve their feasibility in clinics, systemic evaluations of currently available carrier systems under clinical trials and preclinical studies are needed. In this review, we focus on recent noticeable preclinical studies and clinical results regarding siRNA-based conjugates for clinical translations. Advantages and drawbacks of siRNA-based conjugates are discussed, compared to particle-based delivery systems. Then, representative siRNA-based conjugates with aptamers, peptides, carbohydrates, lipids, polymers, and nanostructured materials are introduced. To improve feasibility of siRNA conjugates in preclinical studies, several considerations for the rational design of siRNA conjugates in terms of cleavability, immune responses, multivalent conjugations, and mechanism of action are also presented. Lastly, we discuss lessons from previous preclinical and clinical studies related to siRNA conjugates and perspectives of their clinical applications.
Collapse
Affiliation(s)
- Soo Hyeon Lee
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Zurich, Switzerland
| | - Yoon Young Kang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Hyo-Eun Jang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea.
| |
Collapse
|
47
|
Cheng Y, Yumul RC, Pun SH. Virus-Inspired Polymer for Efficient In Vitro and In Vivo Gene Delivery. Angew Chem Int Ed Engl 2016; 55:12013-7. [PMID: 27538359 DOI: 10.1002/anie.201605958] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/03/2016] [Indexed: 01/02/2023]
Abstract
Clinical translation of nucleic acids drugs has been stunted by limited delivery options. Herein, we report a synthetic polymer designed to mimic viral mechanisms of delivery called VIPER (virus-inspired polymer for endosomal release). VIPER is composed of a polycation block for condensation of nucleic acids, and a pH-sensitive block for acid-triggered display of a lytic peptide to promote trafficking to the cell cytosol. VIPER shows superior efficiencies compared to commercial agents when delivering genes to multiple immortalized cell lines. Importantly, in murine models, VIPER facilitates effective gene transfer to solid tumors.
Collapse
Affiliation(s)
- Yilong Cheng
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, 98195, USA
| | - Roma C Yumul
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, 98195, USA
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
48
|
|
49
|
Gooding M, Malhotra M, Evans JC, Darcy R, O'Driscoll CM. Oligonucleotide conjugates - Candidates for gene silencing therapeutics. Eur J Pharm Biopharm 2016; 107:321-40. [PMID: 27521696 DOI: 10.1016/j.ejpb.2016.07.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 11/18/2022]
Abstract
The potential therapeutic and diagnostic applications of oligonucleotides (ONs) have attracted great attention in recent years. The capability of ONs to selectively inhibit target genes through antisense and RNA interference mechanisms, without causing un-intended sideeffects has led them to be investigated for various biomedical applications, especially for the treatment of viral diseases and cancer. In recent years, many researchers have focused on enhancing the stability and target specificity of ONs by encapsulating/complexing them with polymers or lipid chains to formulate nanoparticles/nanocomplexes/micelles. Also, chemical modification of nucleic acids has emerged as an alternative to impart stability to ONs against nucleases and other degrading enzymes and proteins found in blood. In addition to chemically modifying the nucleic acids directly, another strategy that has emerged, involves conjugating polymers/peptide/aptamers/antibodies/proteins, preferably to the sense strand (3'end) of siRNAs. Conjugation to the siRNA not only enhances the stability and targeting specificity of the siRNA, but also allows for the development of self-administering siRNA formulations, with a much smaller size than what is usually observed for nanoparticle (∼200nm). This review concentrates mainly on approaches and studies involving ON-conjugates for biomedical applications.
Collapse
Affiliation(s)
- Matt Gooding
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - James C Evans
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Raphael Darcy
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | | |
Collapse
|
50
|
Reinhard S, Wagner E. How to Tackle the Challenge of siRNA Delivery with Sequence-Defined Oligoamino Amides. Macromol Biosci 2016; 17. [PMID: 27328447 DOI: 10.1002/mabi.201600152] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/24/2016] [Indexed: 12/31/2022]
Abstract
RNA interference (RNAi) as a mechanism of gene regulation provides exciting opportunities for medical applications. Synthetic small interfering RNA (siRNA) triggers the knockdown of complementary mRNA sequences in a catalytic fashion and has to be delivered into the cytosol of the targeted cells. The design of adequate carrier systems to overcome multiple extracellular and intracellular roadblocks within the delivery process has utmost importance. Cationic polymers form polyplexes through electrostatic interaction with negatively charged nucleic acids and present a promising class of carriers. Issues of polycations regarding toxicity, heterogeneity, and polydispersity can be overcome by solid-phase-assisted synthesis of sequence-defined cationic oligomers. These medium-sized highly versatile nucleic acid carriers display low cytotoxicity and can be modified and tailored in multiple ways to meet specific requirements of nucleic acid binding, polyplex size, shielding, targeting, and intracellular release of the cargo. In this way, sequence-defined cationic oligomers can mimic the dynamic and bioresponsive behavior of viruses.
Collapse
Affiliation(s)
- Sören Reinhard
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig Maximilians University, 81377, Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig Maximilians University, 81377, Munich, Germany.,Nanosystems Initiative Munich (NIM), 80799, Munich, Germany
| |
Collapse
|