1
|
Agrohia DK, Goswami R, Jantarat T, Çiçek YA, Thongsukh K, Jeon T, Bell JM, Rotello VM, Vachet RW. Suborgan Level Quantitation of Proteins in Tissues Delivered by Polymeric Nanocarriers. ACS NANO 2024; 18:16808-16818. [PMID: 38870478 PMCID: PMC11497159 DOI: 10.1021/acsnano.4c02344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Amidst the rapid growth of protein therapeutics as a drug class, there is an increased focus on designing systems to effectively deliver proteins to target organs. Quantitative monitoring of protein distributions in tissues is essential for optimal development of delivery systems; however, existing strategies can have limited accuracy, making it difficult to assess suborgan dosing. Here, we describe a quantitative imaging approach that utilizes metal-coded mass tags and laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) to quantify the suborgan distributions of proteins in tissues that have been delivered by polymeric nanocarriers. Using this approach, we measure nanomole per gram levels of proteins as delivered by guanidinium-functionalized poly(oxanorborneneimide) (PONI) polymers to various tissues, including the alveolar region of the lung. Due to the multiplexing capability of the LA-ICP-MS imaging, we are also able to simultaneously quantify protein and polymer distributions, obtaining valuable information about the relative excretion pathways of the protein cargo and carrier. This imaging approach will facilitate quantitative correlations between nanocarrier properties and protein cargo biodistributions.
Collapse
Affiliation(s)
- Dheeraj K. Agrohia
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Teerapong Jantarat
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Yağız Anil Çiçek
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Korndanai Thongsukh
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Taewon Jeon
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Jonathan M. Bell
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Richard W. Vachet
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
2
|
Hartl N, Gabold B, Uhl P, Kromer A, Xiao X, Fricker G, Mier W, Liu R, Merkel OM. ApoE-functionalization of nanoparticles for targeted brain delivery-a feasible method for polyplexes? Drug Deliv Transl Res 2024; 14:1660-1677. [PMID: 38087181 PMCID: PMC11052808 DOI: 10.1007/s13346-023-01482-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 04/28/2024]
Abstract
The blood-brain barrier (BBB) poses a major obstacle in the treatment of all types of central nervous system (CNS) diseases. Small interfering RNA (siRNA) offers in principle a promising therapeutic approach by downregulating disease-related genes via RNA interference. However, the BBB is a formidable barrier for macromolecules such as nucleic acids. In an effort to develop a brain-targeted strategy for siRNA delivery systems formed by electrostatic interactions with cationic polymers (polyplexes (PXs)), we investigated the suitability of the well-known surfactant-based approach for Apolipoprotein E (ApoE)-functionalization of nanoparticles (NPs). The aim of this present work was to investigate if ApoE coating of siRNA PXs formed with cationic branched 25-kDa poly(ethyleneimine) (b-PEI) and nylon-3 polymers without or after precoating with polysorbate 80 (PS 80) would promote successful delivery across the BBB. We utilized highly hydrophobic NM0.2/CP0.8 nylon-3 polymers to evaluate the effects of hydrophobic cyclopentyl (CP) subunits on ApoE binding efficacy and observed successful ApoE binding with and without PS 80 precoating to the nylon-3 but not the PEI polyplexes. Accordingly, ApoE-coated nylon-3 polyplexes showed significantly increased uptake and gene silencing in U87 glioma cells but no benefit in vivo. In conclusion, further optimization of ApoE-functionalized polyplexes and more sophisticated in vitro models are required to achieve more successful in vitro-in vivo translation in future approaches.
Collapse
Affiliation(s)
- Natascha Hartl
- Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Bettina Gabold
- Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Philipp Uhl
- Pharmaceutical Technology and Biopharmaceutics, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120, Heidelberg, Germany
| | - Adrian Kromer
- Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Ximian Xiao
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Gert Fricker
- Pharmaceutical Technology and Biopharmaceutics, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120, Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Olivia M Merkel
- Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
3
|
Binder J, Winkeljann J, Steinegger K, Trnovec L, Orekhova D, Zähringer J, Hörner A, Fell V, Tinnefeld P, Winkeljann B, Frieß W, Merkel OM. Closing the Gap between Experiment and Simulation─A Holistic Study on the Complexation of Small Interfering RNAs with Polyethylenimine. Mol Pharm 2024; 21:2163-2175. [PMID: 38373164 PMCID: PMC7616749 DOI: 10.1021/acs.molpharmaceut.3c00747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Rational design is pivotal in the modern development of nucleic acid nanocarrier systems. With the rising prominence of polymeric materials as alternatives to lipid-based carriers, understanding their structure-function relationships becomes paramount. Here, we introduce a newly developed coarse-grained model of polyethylenimine (PEI) based on the Martini 3 force field. This model facilitates molecular dynamics simulations of true-sized PEI molecules, exemplified by molecules with molecular weights of 1.3, 5, 10, and 25 kDa, with degrees of branching between 50.0 and 61.5%. We employed this model to investigate the thermodynamics of small interfering RNA (siRNA) complexation with PEI. Our simulations underscore the pivotal role of electrostatic interactions in the complexation process. Thermodynamic analyses revealed a stronger binding affinity with increased protonation, notably in acidic (endosomal) pH, compared to neutral conditions. Furthermore, the molecular weight of PEI was found to be a critical determinant of binding dynamics: smaller PEI molecules closely enveloped the siRNA, whereas larger ones extended outward, facilitating the formation of complexes with multiple RNA molecules. Experimental validations, encompassing isothermal titration calorimetry and single-molecule fluorescence spectroscopy, aligned well with our computational predictions. Our findings not only validate the fidelity of our PEI model but also accentuate the importance of in silico data in the rational design of polymeric drug carriers. The synergy between computational predictions and experimental validations, as showcased here, signals a refined and precise approach to drug carrier design.
Collapse
Affiliation(s)
- Jonas Binder
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
| | - Joshua Winkeljann
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 80799 München, Germany
- Chair of Experimental Physics I, University of Augsburg, Universitätsstraße 1, 86519 Augsburg, Germany
| | - Katharina Steinegger
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
| | - Lara Trnovec
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
| | - Daria Orekhova
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 80799 München, Germany
| | - Jonas Zähringer
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 80799 München, Germany
| | - Andreas Hörner
- Chair of Experimental Physics I, University of Augsburg, Universitätsstraße 1, 86519 Augsburg, Germany
| | - Valentin Fell
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
| | - Philip Tinnefeld
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 80799 München, Germany
| | - Benjamin Winkeljann
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 80799 München, Germany
| | - Wolfgang Frieß
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
| | - Olivia M Merkel
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, Haus B, 81377 München, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 80799 München, Germany
| |
Collapse
|
4
|
Hartl N, Gabold B, Adams F, Uhl P, Oerter S, Gätzner S, Metzger M, König AC, Hauck SM, Appelt-Menzel A, Mier W, Fricker G, Merkel OM. Overcoming the blood-brain barrier? - prediction of blood-brain permeability of hydrophobically modified polyethylenimine polyplexes for siRNA delivery into the brain with in vitro and in vivo models. J Control Release 2023; 360:613-629. [PMID: 37437848 DOI: 10.1016/j.jconrel.2023.07.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/23/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
The blood-brain barrier (BBB) is a highly selective biological barrier that represents a major bottleneck in the treatment of all types of central nervous system (CNS) disorders. Small interfering RNA (siRNA) offers in principle a promising therapeutic approach, e.g., for brain tumors, by downregulating brain tumor-related genes and inhibiting tumor growth via RNA interference. In an effort to develop efficient siRNA nanocarriers for crossing the BBB, we utilized polyethyleneimine (PEI) polymers hydrophobically modified with either stearic-acid (SA) or dodecylacrylamide (DAA) subunits and evaluated their suitability for delivering siRNA across the BBB in in vitro and in vivo BBB models depending on their structure. Physicochemical characteristics of siRNA-polymer complexes (polyplexes (PXs)), e.g., particle size and surface charge, were measured by dynamic light scattering and laser Doppler anemometry, whereas siRNA condensation ability of polymers and polyplex stability was evaluated by spectrophotometric methods. The composition of the biomolecule corona that absorbs on polyplexes upon encountering physiological fluids was investigated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and by a liquid chromatography-tandem mass spectrometry (LC-MS-MS) method. Cellular internalization abilities of PXs into brain endothelial cells (hCMEC/D3) was confirmed, and a BBB permeation assay using a human induced pluripotent stem cell (hiPSC)-derived BBB model revealed similar abilities to cross the BBB for all formulations under physiological conditions. However, biodistribution studies of radiolabeled PXs in mice were inconsistent with in vitro results as the detected amount of radiolabeled siRNA in the brain delivered with PEI PXs was higher compared to PEI-SA PXs. Taken together, PEI PXs were shown to be a suitable nanocarrier to deliver small amounts of siRNA across the BBB into the brain but more sophisticated human BBB models that better represent physiological conditions and biodistribution are required to provide highly predictive in vitro data for human CNS drug development in the future.
Collapse
Affiliation(s)
- Natascha Hartl
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Bettina Gabold
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Friederike Adams
- University of Stuttgart, Institute of Polymer Chemistry, Macromolecular Materials and Fiber Chemistry, Pfaffenwaldring 55, 70569 Stuttgart, Germany
| | - Philipp Uhl
- University Hospital Heidelberg, Department of Nuclear Medicine, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Sabrina Oerter
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany; University Hospital Würzburg, Chair of Tissue Engineering and Regenerative Medicine (TERM), 97070 Würzburg, Germany
| | - Sabine Gätzner
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany; University Hospital Würzburg, Chair of Tissue Engineering and Regenerative Medicine (TERM), 97070 Würzburg, Germany
| | - Ann-Christine König
- Helmholtz Centrum Munich - German Research Center for Environmental Health, Research Unit Protein Science, Heidemannsstr. 1, 80939, Munich, Germany
| | - Stefanie M Hauck
- Helmholtz Centrum Munich - German Research Center for Environmental Health, Research Unit Protein Science, Heidemannsstr. 1, 80939, Munich, Germany
| | - Antje Appelt-Menzel
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany; University Hospital Würzburg, Chair of Tissue Engineering and Regenerative Medicine (TERM), 97070 Würzburg, Germany
| | - Walter Mier
- University Hospital Heidelberg, Department of Nuclear Medicine, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Gert Fricker
- University of Heidelberg, Institute for Pharmacy & Molekular Biotechnology, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Olivia M Merkel
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
5
|
Uchida S, Lau CYJ, Oba M, Miyata K. Polyplex designs for improving the stability and safety of RNA therapeutics. Adv Drug Deliv Rev 2023; 199:114972. [PMID: 37364611 DOI: 10.1016/j.addr.2023.114972] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
Nanoparticle-based delivery systems have contributed to the recent clinical success of RNA therapeutics, including siRNA and mRNA. RNA delivery using polymers has several distinct properties, such as enabling RNA delivery into extra-hepatic organs, modulation of immune responses to RNA, and regulation of intracellular RNA release. However, delivery systems should overcome safety and stability issues to achieve widespread therapeutic applications. Safety concerns include direct damage to cellular components, innate and adaptive immune responses, complement activation, and interaction with surrounding molecules and cells in the blood circulation. The stability of the delivery systems should balance extracellular RNA protection and controlled intracellular RNA release, which requires optimization for each RNA species. Further, polymer designs for improving safety and stability often conflict with each other. This review covers advances in polymer-based approaches to address these issues over several years, focusing on biological understanding and design concepts for delivery systems rather than material chemistry.
Collapse
Affiliation(s)
- Satoshi Uchida
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto, 606-0823, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| | - Chun Yin Jerry Lau
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Makoto Oba
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto, 606-0823, Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| |
Collapse
|
6
|
Jin Y, Adams F, Möller J, Isert L, Zimmermann CM, Keul D, Merkel OM. Synthesis and Application of Low Molecular Weight PEI-Based Copolymers for siRNA Delivery with Smart Polymer Blends. Macromol Biosci 2023; 23:e2200409. [PMID: 36446588 DOI: 10.1002/mabi.202200409] [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: 09/28/2022] [Revised: 11/17/2022] [Indexed: 12/05/2022]
Abstract
Polyethylenimine (PEI) is a commonly used cationic polymer for small-interfering RNA (siRNA) delivery due to its high transfection efficiency at low commercial cost. However, high molecular weight PEI is cytotoxic and thus, its practical application is limited. In this study, different formulations of low molecular weight PEI (LMW-PEI) based copolymers polyethylenimine-g-polycaprolactone (PEI-PCL) (800 Da-40 kDa) and PEI-PCL-PEI (5-5-5 kDa) blended with or without polyethylene glycol-b-polycaprolactone (PEG-PCL) (5 kDa-4 kDa) are investigated to prepare nanoparticles via nanoprecipitation using a solvent displacement method with sizes ≈100 nm. PEG-PCL can stabilize the nanoparticles, improve their biocompatibility, and extend their circulation time in vivo. The nanoparticles composed of PEI-PCL-PEI and PEG-PCL show higher siRNA encapsulation efficiency than PEI-PCL/PEG-PCL based nanoparticles at low N/P ratios, higher cellular uptake, and a gene silencing efficiency of ≈40% as a result of the higher molecular weight PEI blocks. These results suggest that the PEI-PCL-PEI/PEG-PCL nanoparticle system could be a promising vehicle for siRNA delivery at minimal synthetic effort.
Collapse
Affiliation(s)
- Yao Jin
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Friederike Adams
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany.,Department of Ophthalmology, University Eye Hospital Tübingen, Center for Ophthalmology, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Judith Möller
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Lorenz Isert
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Christoph M Zimmermann
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany.,Department for Chemistry, University of Bern, Biochemistry and Pharmacy, Freiestrasse 3, Bern, 3012, Switzerland
| | - David Keul
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Olivia M Merkel
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| |
Collapse
|
7
|
Huang H, Ali A, Liu Y, Xie H, Ullah S, Roy S, Song Z, Guo B, Xu J. Advances in image-guided drug delivery for antibacterial therapy. Adv Drug Deliv Rev 2023; 192:114634. [PMID: 36503884 DOI: 10.1016/j.addr.2022.114634] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/20/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022]
Abstract
The emergence of antibiotic-resistant bacterial strains is seriously endangering the global healthcare system. There is an urgent need for combining imaging with therapies to realize the real-time monitoring of pathological condition and treatment progress. It also provides guidance on exploring new medicines and enhance treatment strategies to overcome the antibiotic resistance of existing conventional antibiotics. In this review, we provide a thorough overview of the most advanced image-guided approaches for bacterial diagnosis (e.g., computed tomography imaging, magnetic resonance imaging, photoacoustic imaging, ultrasound imaging, fluorescence imaging, positron emission tomography, single photon emission computed tomography imaging, and multiple imaging), and therapies (e.g., photothermal therapy, photodynamic therapy, chemodynamic therapy, sonodynamic therapy, immunotherapy, and multiple therapies). This review focuses on how to design and fabricate photo-responsive materials for improved image-guided bacterial theranostics applications. We present a potential application of different image-guided modalities for both bacterial diagnosis and therapies with representative examples. Finally, we highlighted the current challenges and future perspectives image-guided approaches for future clinical translation of nano-theranostics in bacterial infections therapies. We envision that this review will provide for future development in image-guided systems for bacterial theranostics applications.
Collapse
Affiliation(s)
- Haiyan Huang
- Institute of Low-Dimensional Materials Genome Initiative, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China; School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Arbab Ali
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano Safety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yi Liu
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui Xie
- Institute of Low-Dimensional Materials Genome Initiative, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China; Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences, Chengdu 610041, China
| | - Sana Ullah
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan; Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box: 33, PC: 616, Oman
| | - Shubham Roy
- School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Zhiyong Song
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan 430070, China.
| | - Bing Guo
- School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Jian Xu
- Institute of Low-Dimensional Materials Genome Initiative, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
8
|
Polyethyleneimine-Based Drug Delivery Systems for Cancer Theranostics. J Funct Biomater 2022; 14:jfb14010012. [PMID: 36662059 PMCID: PMC9862060 DOI: 10.3390/jfb14010012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
With the development of nanotechnology, various types of polymer-based drug delivery systems have been designed for biomedical applications. Polymer-based drug delivery systems with desirable biocompatibility can be efficiently delivered to tumor sites with passive or targeted effects and combined with other therapeutic and imaging agents for cancer theranostics. As an effective vehicle for drug and gene delivery, polyethyleneimine (PEI) has been extensively studied due to its rich surface amines and excellent water solubility. In this work, we summarize the surface modifications of PEI to enhance biocompatibility and functionalization. Additionally, the synthesis of PEI-based nanoparticles is discussed. We further review the applications of PEI-based drug delivery systems in cancer treatment, cancer imaging, and cancer theranostics. Finally, we thoroughly consider the outlook and challenges relating to PEI-based drug delivery systems.
Collapse
|
9
|
Brito J, Andrianov AK, Sukhishvili SA. Factors Controlling Degradation of Biologically Relevant Synthetic Polymers in Solution and Solid State. ACS APPLIED BIO MATERIALS 2022; 5:5057-5076. [PMID: 36206552 DOI: 10.1021/acsabm.2c00694] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The field of biodegradable synthetic polymers, which is central for regenerative engineering and drug delivery applications, encompasses a multitude of hydrolytically sensitive macromolecular structures and diverse processing approaches. The ideal degradation behavior for a specific life science application must comply with a set of requirements, which include a clinically relevant kinetic profile, adequate biocompatibility, benign degradation products, and controlled structural evolution. Although significant advances have been made in tailoring materials characteristics to satisfy these requirements, the impacts of autocatalytic reactions and microenvironments are often overlooked resulting in uncontrollable and unpredictable outcomes. Therefore, roles of surface versus bulk erosion, in situ microenvironment, and autocatalytic mechanisms should be understood to enable rational design of degradable systems. In an attempt to individually evaluate the physical state and form factors influencing autocatalytic hydrolysis of degradable polymers, this Review follows a hierarchical analysis that starts with hydrolytic degradation of water-soluble polymers before building up to 2D-like materials, such as ultrathin coatings and capsules, and then to solid-state degradation. We argue that chemical reactivity largely governs solution degradation while diffusivity and geometry control the degradation of bulk materials, with thin "2D" materials remaining largely unexplored. Following this classification, this Review explores techniques to analyze degradation in vitro and in vivo and summarizes recent advances toward understanding degradation behavior for traditional and innovative polymer systems. Finally, we highlight challenges encountered in analytical methodology and standardization of results and provide perspective on the future trends in the development of biodegradable polymers.
Collapse
Affiliation(s)
- Jordan Brito
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas77843, United States
| | - Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland20850, United States
| | - Svetlana A Sukhishvili
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas77843, United States
| |
Collapse
|
10
|
Tan S, Chen Z, Mironchik Y, Mori N, Penet MF, Si G, Krishnamachary B, Bhujwalla ZM. VEGF Overexpression Significantly Increases Nanoparticle-Mediated siRNA Delivery and Target-Gene Downregulation. Pharmaceutics 2022; 14:pharmaceutics14061260. [PMID: 35745832 PMCID: PMC9229257 DOI: 10.3390/pharmaceutics14061260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 02/01/2023] Open
Abstract
The availability of nanoparticles (NPs) to deliver small interfering RNA (siRNA) has significantly expanded the specificity and range of ‘druggable’ targets for precision medicine in cancer. This is especially important for cancers such as triple negative breast cancer (TNBC) for which there are no targeted treatments. Our purpose here was to understand the role of tumor vasculature and vascular endothelial growth factor (VEGF) overexpression in a TNBC xenograft in improving the delivery and function of siRNA NPs using in vivo as well as ex vivo imaging. We used triple negative MDA-MB-231 human breast cancer xenografts derived from cells engineered to overexpress VEGF to understand the role of VEGF and vascularization in NP delivery and function. We used polyethylene glycol (PEG) conjugated polyethylenimine (PEI) NPs to deliver siRNA that downregulates choline kinase alpha (Chkα), an enzyme that is associated with malignant transformation and tumor progression. Because Chkα converts choline to phosphocholine, effective delivery of Chkα siRNA NPs resulted in functional changes of a significant decrease in phosphocholine and total choline that was detected with 1H magnetic resonance spectroscopy (MRS). We observed a significant increase in NP delivery and a significant decrease in Chkα and phosphocholine in VEGF overexpressing xenografts. Our results demonstrated the importance of tumor vascularization in achieving effective siRNA delivery and downregulation of the target gene Chkα and its function.
Collapse
Affiliation(s)
- Shanshan Tan
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Zhihang Chen
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Noriko Mori
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21205, USA
| | - Ge Si
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Zaver M. Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21205, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Correspondence:
| |
Collapse
|
11
|
Merkel OM. Can pulmonary RNA delivery improve our pandemic preparedness? J Control Release 2022; 345:549-556. [PMID: 35358609 PMCID: PMC8958776 DOI: 10.1016/j.jconrel.2022.03.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 03/20/2022] [Indexed: 12/17/2022]
Abstract
The coronavirus pandemic has changed our perception of RNA medicines, and RNA vaccines have revolutionized our pandemic preparedness. But are we indeed prepared for the next variant or the next emerging virus? How can we prepare? And what does the role of inhaled antiviral RNA play in this regard? When the pandemic started, I rerouted much of the ongoing inhaled RNA delivery research in my group towards the inhibition and treatment of respiratory viral infections. Two years later, I have taken the literature, past and ongoing clinical trials into consideration and have gained new insights based on our collaborative research which I will discuss in this oration.
Collapse
Affiliation(s)
- Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-University of Munich, Butenandtstraße 5, 81377 Munich, Germany.
| |
Collapse
|
12
|
miRNA Delivery by Nanosystems: State of the Art and Perspectives. Pharmaceutics 2021; 13:pharmaceutics13111901. [PMID: 34834316 PMCID: PMC8619868 DOI: 10.3390/pharmaceutics13111901] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/25/2021] [Accepted: 11/04/2021] [Indexed: 01/09/2023] Open
Abstract
MicroRNAs (miRNAs) are short (~21-23 nucleotides), non-coding endogenous RNA molecules that modulate gene expression at the post-transcriptional level via the endogenous RNA interference machinery of the cell. They have emerged as potential biopharmaceuticals candidates for the treatment of various diseases, including cancer, cardiovascular and metabolic diseases. However, in order to advance miRNAs therapeutics into clinical settings, their delivery remains a major challenge. Different types of vectors have been investigated to allow the delivery of miRNA in the diseased tissue. In particular, non-viral delivery systems have shown important advantages such as versatility, low cost, easy fabrication and low immunogenicity. Here, we present a general overview of the main types of non-viral vectors developed for miRNA delivery, with their advantages, limitations and future perspectives.
Collapse
|
13
|
Zhang D, Zhuang R, Li J, Lv Y, Yang X, Pan W, Zhang X. MicroSPECT Imaging-Guided Treatment of Idiopathic Pulmonary Fibrosis in Mice with a Vimentin-Targeting 99mTc-Labeled N-Acetylglucosamine-Polyethyleneimine. Mol Pharm 2021; 18:4140-4147. [PMID: 34657437 DOI: 10.1021/acs.molpharmaceut.1c00545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic disease with poor prognosis. Evidence has shown that vimentin is a key regulator of lung fibrogenesis. 99mTc-labeled N-acetylglucosamine-polyethyleneimine (NAG-PEI), a vimentin-targeting radiotracer, was used for the early diagnosis of IPF, and NAG-PEI was also used as a therapeutic small interfering RNA (siRNA) delivery vector for the treatment of IPF in this study. Single-photon emission-computed tomography (SPECT) imaging of bleomycin (BM)- and silica-induced IPF mice with 99mTc-labeled NAG-PEI was performed to visualize pulmonary fibrosis and monitor the treatment efficiency of siRNA-loaded NAG-PEI, lipopolysaccharide (LPS, a tolerogenic adjuvant), or zymosan (ZYM, an immunostimulant). The lung uptakes of 99mTc-NAG-PEI in the BM- and silica-induced IPF mice were clearly and directly correlated with IPF progression. The lung uptake of 99mTc-NAG-PEI in the NAG-PEI/TGF-β1-siRNA treatment group or LPS treatment group was evidently lower than that in the control group, while the lung uptake of 99mTc-NAG-PEI was significantly higher in the ZYM treatment group compared to that in the control group. These results demonstrate that NAG-PEI is a potent MicroSPECT imaging-guided theranostic platform for IPF diagnosis and therapy.
Collapse
Affiliation(s)
- Deliang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.,Department of Nuclear Medicine, Xiang'an Hospital Affiliated to Xiamen University, Xiamen 361102, China
| | - Rongqiang Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jindian Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yuting Lv
- Department of Nuclear Medicine, Xiang'an Hospital Affiliated to Xiamen University, Xiamen 361102, China.,School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xia Yang
- Department of Nuclear Medicine, Xiang'an Hospital Affiliated to Xiamen University, Xiamen 361102, China.,School of Medicine, Xiamen University, Xiamen 361102, China
| | - Weimin Pan
- Department of Nuclear Medicine, Xiang'an Hospital Affiliated to Xiamen University, Xiamen 361102, China
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
14
|
Sharifiaghdam M, Shaabani E, Sharifiaghdam Z, De Keersmaecker H, Lucas B, Lammens J, Ghanbari H, Teimoori-Toolabi L, Vervaet C, De Beer T, Faridi-Majidi R, De Smedt SC, Braeckmans K, Fraire JC. Macrophage reprogramming into a pro-healing phenotype by siRNA delivered with LBL assembled nanocomplexes for wound healing applications. NANOSCALE 2021; 13:15445-15463. [PMID: 34505619 DOI: 10.1039/d1nr03830c] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Excessive inflammatory responses in wounds are characterized by the presence of high levels of pro-inflammatory M1 macrophages rather than pro-healing M2 macrophages, which leads to delayed wound healing. Macrophage reprogramming from the M1 to M2 phenotype through knockdown of interferon regulatory factor 5 (irf5) has emerged as a possible therapeutic strategy. While downregulation of irf5 could be achieved by siRNA, it very much depends on successful intracellular delivery by suitable siRNA carriers. Here, we report on highly stable selenium-based layer-by-layer (LBL) nanocomplexes (NCs) for siRNA delivery with polyethyleneimine (PEI-LBL-NCs) as the final polymer layer. PEI-LBL-NCs showed good protection of siRNA with only 40% siRNA release in a buffer of pH = 8.5 after 72 h or in simulated wound fluid after 4 h. PEI-LBL-NCs also proved to be able to transfect RAW 264.7 cells with irf5-siRNA, resulting in successful reprogramming to the M2 phenotype as evidenced by a 3.4 and 2.6 times decrease in NOS-2 and TNF-α mRNA expression levels, respectively. Moreover, irf5-siRNA transfected cells exhibited a 2.5 times increase of the healing mediator Arg-1 and a 64% increase in expression of the M2 cell surface marker CD206+. Incubation of fibroblast cells with conditioned medium isolated from irf5-siRNA transfected RAW 264.7 cells resulted in accelerated wound healing in an in vitro scratch assay. These results show that irf5-siRNA loaded PEI-LBL-NCs are a promising therapeutic approach to tune macrophage polarization for improved wound healing.
Collapse
Affiliation(s)
- Maryam Sharifiaghdam
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Elnaz Shaabani
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zeynab Sharifiaghdam
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Herlinde De Keersmaecker
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
- Center for Advanced Light Microscopy, Ghent University, 9000 Ghent, Belgium
| | - Bart Lucas
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
| | - Joris Lammens
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Hossein Ghanbari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | | | - Chris Vervaet
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Thomas De Beer
- Laboratory of Pharmaceutical Process Analytical Technology (LPPAT), Department of Pharmaceutical Analysis, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Reza Faridi-Majidi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
- Center for Advanced Light Microscopy, Ghent University, 9000 Ghent, Belgium
| | - Juan C Fraire
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
| |
Collapse
|
15
|
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: 46] [Impact Index Per Article: 15.3] [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
|
16
|
Porfiryeva NN, Moustafine RI, Khutoryanskiy VV. PEGylated Systems in Pharmaceutics. POLYMER SCIENCE SERIES C 2020. [DOI: 10.1134/s181123822001004x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
Norouzi P, Amini M, Dinarvand R, Arefian E, Seyedjafari E, Atyabi F. Co-delivery of gemcitabine prodrug along with anti NF-κB siRNA by tri-layer micelles can increase cytotoxicity, uptake and accumulation of the system in the cancers. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111161. [PMID: 32806226 DOI: 10.1016/j.msec.2020.111161] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/21/2020] [Accepted: 06/04/2020] [Indexed: 12/17/2022]
Abstract
Combination treatment based on gene and chemotherapy is a promising strategy for effective cancer treatment due to the limited therapeutic efficacy of anticancer drugs. Dual functional polymeric micelles (PMs) have been emerged as potent nanocarriers for combinational cancer therapy. In the present study, the potential of tri-layer PMs loaded with anti-nuclear factor-κB (NF-κB) siRNA and 4-(N)-stearoyl gemcitabine (GemC18) has been investigated for cancer treatment. PMs with different core hydrophobicity were prepared by using poly(ε-caprolactone), polyethyleneimine and polyethylene glycol (PCL-PEI-PEG) copolymers and evaluated. The results revealed that GemC18-loaded PMs were significantly more cytotoxic than free drug on breast and pancreatic cancer cells. However, the cytotoxicity of drug loaded micelles was decreased by increasing the micellar core hydrophobicity because of decreasing drug release rate. Moreover, siRNA loaded PMs could considerably inhibit NF-κB expression. PMs loaded with both GemC18 and siRNA exhibited higher capability to induce apoptosis and inhibit migration of both cells. PMs with the most hydrophobic core indicated higher tumor accumulation efficiency via in-vivo imaging study. In conclusion, the prepared PMs hold a promise as an attractive dual functional delivery system for an effective cancer therapy.
Collapse
Affiliation(s)
- Parisa Norouzi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran
| | - Ehsan Arefian
- Department of Microbiology, Faculty of Biology, College of Science, University of Tehran, P.O. Box 14155-6455, Tehran, Iran
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, P.O. Box 14155-6455, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran.
| |
Collapse
|
18
|
Keil TWM, Baldassi D, Merkel OM. T-cell targeted pulmonary siRNA delivery for the treatment of asthma. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1634. [PMID: 32267622 DOI: 10.1002/wnan.1634] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 12/31/2022]
Abstract
Despite the large number of drugs available for the treatment of asthma, in 5-10% of the patients this disease is not well controlled. While most treatments palliate symptoms, those suffering from severe and uncontrolled asthma could benefit more from a therapeutic approach addressing the root problem. An siRNA-based therapy targeting the transcription factor GATA3 in activated T helper cells subtype 2 (TH 2 cells), one of the key upstream factors involved in asthma, could therefore represent a promising strategy. However, the difficult-to-transfect cell type has not extensively been explored for nucleic acid therapeutics. In this regard, our group first identified a suitable pathway, that is, transferrin receptor mediated uptake, to target efficiently and specifically activated TH 2 cells with a transferrin-polyethyleneimine (PEI) conjugate which forms polyplexes with siRNA. This system, despite efficient uptake in activated T cells (ATCs) in vivo, suffered from poor endosomal release and was later improved by a combination with a melittin-PEI conjugate. The new formulation showed improved endosomal escape and gene silencing efficacy. Additionally, in order to develop a clinically relevant dosage form for pulmonary delivery of siRNA we have lately focused on a dry powder formulation by spray drying (SD) for the production of inhalable nano-in-microparticles. In proof-of-concept experiments, DNA/PEI polyplexes were used in order to implement analytics and engineer process parameters to pave the way for SD also siRNA containing polyplexes and more sophisticated systems in general. Ultimately, our efforts are devoted to the development of a novel treatment of asthma that can be translated from bench to bedside and are reviewed and discussed here in the context of the current literature. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Respiratory Disease Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Tobias W M Keil
- Pharmaceutical Technology and Biopharmaceutics, LMU Munich, Munich, Germany
| | - Domizia Baldassi
- Pharmaceutical Technology and Biopharmaceutics, LMU Munich, Munich, Germany
| | - Olivia M Merkel
- Pharmaceutical Technology and Biopharmaceutics, LMU Munich, Munich, Germany
| |
Collapse
|
19
|
Wang Y, Luo J, Truebenbach I, Reinhard S, Klein PM, Höhn M, Kern S, Morys S, Loy DM, Wagner E, Zhang W. Double Click-Functionalized siRNA Polyplexes for Gene Silencing in Epidermal Growth Factor Receptor-Positive Tumor Cells. ACS Biomater Sci Eng 2020; 6:1074-1089. [DOI: 10.1021/acsbiomaterials.9b01904] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yanfang Wang
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Jie Luo
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ines Truebenbach
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Sören Reinhard
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Philipp Michael Klein
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Miriam Höhn
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Sarah Kern
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Stephan Morys
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Dominik M. Loy
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ernst Wagner
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
- Nanosystems Initiative Munich (NIM), Schellingstrasse 4, 80799 Munich, Germany
| | - Wei Zhang
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, Butenandtstrasse 5-13, 81377 Munich, Germany
| |
Collapse
|
20
|
The Impact of Nylon-3 Copolymer Composition on the Efficiency of siRNA Delivery to Glioblastoma Cells. NANOMATERIALS 2019; 9:nano9070986. [PMID: 31288448 PMCID: PMC6669510 DOI: 10.3390/nano9070986] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023]
Abstract
Glioblastoma multiforme is a devastating disease that has attracted enormous attention due to poor prognosis and high recurrence. Small interfering RNA (siRNA) in principle offers a promising therapeutic approach by the downregulation of disease-related genes via RNA interference. For efficient siRNA delivery to target sites, cationic polymers are often used in preclinical studies for the protection of siRNA and complex formation based on electrostatic interactions. In an effort to develop biocompatible and efficient nanocarriers with a translational outlook for optimal gene silencing at reduced toxicity, we synthesized two sets of nylon-3 copolymers with variable cationic content (DM or NM monomer) and hydrophobic subunits (CP monomer) and evaluated their suitability for in vitro siRNA delivery into glioblastoma cells. DM0.4/CP0.6 and NM0.4/CP0.6 polymers with similar subunit ratios were synthesized to compare the effect of different cationic subunits. Additionally, we utilized NM0.2/CP0.8 polymers to evaluate the impact of the different hydrophobic content in the polymer chain. The siRNA condensation ability and polymer–siRNA complex stability was evaluated by unmodified and modified SYBR gold assays, respectively. Further physicochemical characteristics, e.g., particle size and surface charge, were evaluated by dynamic light scattering and laser Doppler anemometry, whereas a relatively new method for polyplex size distribution analysis—tunable resistive pulse sensing—was additionally developed and compared to DLS measurements. Transfection efficiencies, the route of cell internalization, and protein knockdown abilities in glioblastoma cells were investigated by flow cytometry. Furthermore, cellular tolerability was evaluated by MTT and LDH assays. All the polymers efficiently condensed siRNA at N/P ratios of three, whereas polymers with NM cationic subunits demonstrated smaller particle size and lower polyplex stability. Furthermore, NM0.2/CP0.8 polyplexes with the highest hydrophobic content displayed significantly higher cellular internalization in comparison to more cationic formulations and successful knockdown capabilities. Detailed investigations of the cellular uptake route demonstrated that these polyplexes mainly follow clathrin-mediated endocytotic uptake mechanisms, implying high interaction capacity with cellular membranes. Taken together with conducive toxicity profiles, highly hydrophobic nylon-3 polymers provide an appropriate siRNA delivery agent for the potential treatment of glioblastoma.
Collapse
|
21
|
Phillips HR, Tolstyka ZP, Hall BC, Hexum JK, Hackett PB, Reineke TM. Glycopolycation–DNA Polyplex Formulation N/P Ratio Affects Stability, Hemocompatibility, and in Vivo Biodistribution. Biomacromolecules 2019; 20:1530-1544. [DOI: 10.1021/acs.biomac.8b01704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Haley R. Phillips
- Center for Genome Engineering and Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Zachary P. Tolstyka
- Center for Genome Engineering and Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Bryan C. Hall
- Center for Genome Engineering and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K. Hexum
- Center for Genome Engineering and Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Perry B. Hackett
- Center for Genome Engineering and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M. Reineke
- Center for Genome Engineering and Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
22
|
Clarke D, Idris A, McMillan NAJ. Development of novel lipidic particles for siRNA delivery that are highly effective after 12 months storage. PLoS One 2019; 14:e0211954. [PMID: 30735545 PMCID: PMC6368384 DOI: 10.1371/journal.pone.0211954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/24/2019] [Indexed: 01/30/2023] Open
Abstract
Liposomes are versatile and well-proven as a means to deliver nucleic acids into cells. Most of the formulation procedures used are labour intensive and result in unstable end products. We have previously reported on the development of a simple, yet efficient, hydration-of-freeze-dried-matrix (HFDM) method to entrap siRNA within lipid particles. Here we show that the particles are stable up to 12 months after storage at room temperature (RT), 4°C or -20°C. While RT storage results in changes in particle size and polydispersity, gene silencing of all particles was similar to freshly prepared particles following storage for 3, 6, 9 or 12 months at all temperatures. This is the first report of such long-term stability in siRNA-loaded liposomes.
Collapse
Affiliation(s)
- Daniel Clarke
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Australia
| | - Adi Idris
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Australia
- * E-mail:
| | - Nigel A. J. McMillan
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Australia
| |
Collapse
|
23
|
Jackson MA, Bedingfield SK, Yu F, Stokan ME, Miles RE, Curvino EJ, Hoogenboezem EN, Bonami RH, Patel SS, Kendall PL, Giorgio TD, Duvall CL. Dual carrier-cargo hydrophobization and charge ratio optimization improve the systemic circulation and safety of zwitterionic nano-polyplexes. Biomaterials 2019; 192:245-259. [PMID: 30458360 PMCID: PMC6534819 DOI: 10.1016/j.biomaterials.2018.11.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/27/2022]
Abstract
While polymeric nano-formulations for RNAi therapeutics hold great promise for molecularly-targeted, personalized medicine, they possess significant systemic delivery challenges including rapid clearance from circulation and the potential for carrier-associated toxicity due to cationic polymer or lipid components. Herein, we evaluated the in vivo pharmacokinetic and safety impact of often-overlooked formulation parameters, including the ratio of carrier polymer to cargo siRNA and hydrophobic siRNA modification in combination with hydrophobic polymer components (dual hydrophobization). For these studies, we used nano-polyplexes (NPs) with well-shielded, zwitterionic coronas, resulting in various NP formulations of equivalent hydrodynamic size and neutral surface charge regardless of charge ratio. Doubling nano-polyplex charge ratio from 10 to 20 increased circulation half-life five-fold and pharmacokinetic area under the curve four-fold, but was also associated with increased liver enzymes, a marker of hepatic damage. Dual hydrophobization achieved by formulating NPs with palmitic acid-modified siRNA (siPA-NPs) both reduced the amount of carrier polymer required to achieve optimal pharmacokinetic profiles and abrogated liver toxicities. We also show that optimized zwitterionic siPA-NPs are well-tolerated upon long-term, repeated administration in mice and exhibit greater than two-fold increased uptake in orthotopic MDA-MB-231 xenografts compared to commercial transfection reagent, in vivo-jetPEI®. These data suggest that charge ratio optimization has important in vivo implications and that dual hydrophobization strategies can be used to maximize both NP circulation time and safety.
Collapse
Affiliation(s)
- Meredith A Jackson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sean K Bedingfield
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Mitchell E Stokan
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Rachel E Miles
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Elizabeth J Curvino
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ella N Hoogenboezem
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Rachel H Bonami
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shrusti S Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Peggy L Kendall
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Todd D Giorgio
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
24
|
Yin Y, Lee MS, Lee JE, Lim SY, Kim ES, Jeong J, Kim D, Kim J, Lee DS, Jeong JH. Effective systemic siRNA delivery using dual-layer protected long-circulating nanohydrogel containing an inorganic core. Biomater Sci 2019; 7:3297-3306. [DOI: 10.1039/c9bm00369j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PEG-dex-dopa nanohydrogel containing a CaP/siRNA core could achieve extended circulation with reduced RES accumulation, resulting in increased tumor accumulation.
Collapse
|
25
|
CY C, GY L, L Z, XH H, D C, SC W, CZ X, JH Z, L X. MicroRNA delivery mediated by PEGylated polyethylenimine for prostate cancer therapy. OPEN CHEM 2018. [DOI: 10.1515/chem-2018-0138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractA microRNA (miRNA) nanomedicine PEG-PEI/miR-221/222 was synthesized based on PEGylated polyethylenimine PEG-PEI and used to transfect prostate cancer cells (PC-3) in vitro. Gel retardation assay confirmed the formation of nanomedicine, of which the zeta potential and particle size were determined by dynamic light scattering. Its cytotoxicity was analyzed by CCK-8 assay-while its transfection efficiency was analyzed by flow cytometry. Cell uptake and intracellular distribution of nanoparticles were evaluated using laser confocal microscopy. RT-PCR and western-blot assays were conducted to verify the regulation of SIRT1 target gene. We found that the properties of the nanocomplexes of miRNA and PEG-PEI depended on N/P ratios. At higher N/P ratio, accompanied by higher zeta potential and higher cytotoxicity, PEG-PEI is needed to completely condense the miRNA into small particles with uniform size distribution. Under an N/P ratio of 20, high transfection efficiency and low carrier cytotoxicity were obtained simultaneously in PC-3 cells in vitro. Consequently, the SIRT1 expression was up-regulated due to the nanoparticle-delivered miR-221/222, which resulted in effective inhibition of PC-3 cells. Our study revealed the PEG-PEI/miR-221/222 nanomedicine as a prospective alternative for treatment of advanced prostate cancer and also lays a foundation for future in vivo investigation.
Collapse
Affiliation(s)
- Chen CY
- Longgang District People’s Hospital of Shenzhen, Guangdong518000, China
| | - Li GY
- Longgang District People’s Hospital of Shenzhen, Guangdong518000, China
| | - Zhang L
- School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou510275, China
| | - Huang XH
- Longgang District People’s Hospital of Shenzhen, Guangdong518000, China
| | - Cheng D
- School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou510275, China
| | - Wu SC
- School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou510275, China
| | - Xu CZ
- School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou510275, China
| | - Zhou JH
- Longgang District People’s Hospital of Shenzhen, Guangdong518000, China
| | - Xun L
- Longgang District People’s Hospital of Shenzhen, Guangdong518000, China
| |
Collapse
|
26
|
Jones SK, Douglas K, Shields AF, Merkel OM. Correlating quantitative tumor accumulation and gene knockdown using SPECT/CT and bioluminescence imaging within an orthotopic ovarian cancer model. Biomaterials 2018; 178:183-192. [PMID: 29935386 PMCID: PMC6056733 DOI: 10.1016/j.biomaterials.2018.06.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/27/2018] [Accepted: 06/11/2018] [Indexed: 10/28/2022]
Abstract
Using an orthotopic model of ovarian cancer, we studied the delivery of siRNA in nanoparticles of tri-block copolymers consisting of hyperbranched polyethylenimine-graft-polycaprolactone-block-poly(ethylene glycol) (hyPEI-g-PCL-b-PEG) with and without a folic acid targeting ligand. A SKOV-3/LUC FRα overexpressing cell line was employed to mimic the clinical manifestations of ovarian cancer. Both targeted and non-targeted micelleplexes were able to effectively deliver siRNA to the primary tumor and its metastases, as measured by gamma scintillation counting and confocal microscopy. Stability of the micelleplexes was demonstrated with a serum albumin binding study. Regarding biodistribution, intravenous (I.V.) administration showed a slight advantage of FRα targeted over non-targeted micelleplex accumulation within the tumor. However, both formulations displayed significant liver uptake. On the other hand, intraperitoneally (I.P.) injected mice showed a modest 6% of the injected dose per gram (ID/g) uptake within the primary and most interestingly also in the metastatic lesions which subsequently resulted in a 62% knockdown of firefly luciferase expression in the tumor after a single injection. While this is, to the best of our knowledge, the first paper that correlates quantitative tumor accumulation in an orthotopic tumor model with in vivo gene silencing, these data demonstrate that PEI-g-PCL-b-PEG-Fol conjugates are a promising option for gene knockdown in ovarian cancer.
Collapse
Affiliation(s)
- Steven K Jones
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kirk Douglas
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Olivia M Merkel
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA; Department of Pharmaceutical Sciences, Wayne State University School of Pharmacy and Health Sciences, Detroit, MI, USA; Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
27
|
Mohammadinia S, Abedi SM, Noaparast Z. St. John’s Wort accelerates the liver clearance of technetium-99-sestamibi in rats. Nucl Med Commun 2018; 39:839-844. [DOI: 10.1097/mnm.0000000000000880] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
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: 3.5] [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
|
Tai W, Li J, Corey E, Gao X. A ribonucleoprotein octamer for targeted siRNA delivery. Nat Biomed Eng 2018; 2:326-337. [PMID: 30936447 PMCID: PMC6136846 DOI: 10.1038/s41551-018-0214-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 03/02/2018] [Indexed: 01/09/2023]
Abstract
Hurdles in cell-specific delivery of small interfering RNA (siRNA) in vivo hinder the clinical translation of RNA interference (RNAi). A fundamental problem concerns conflicting requirements for the design of the delivery vehicles: cationic materials facilitate cargo condensation and endosomolysis, yet hinder in vivo targeting and colloidal stability. Here, we describe a self-assembled, compact (~30 nm) and biocompatible ribonucleoprotein-octamer nanoparticle that achieves endosomal destabilization and targeted delivery. The protein octamer consists of a poly(ethylene glycol) scaffold, a sterically masked endosomolytic peptide and a double-stranded RNA-binding domain, providing a discrete number of siRNA loading sites and a high siRNA payload (>30 wt%), and offering flexibility in both siRNA and targeting-ligand selection. We show that a ribonucleoprotein octamer against the polo-like kinase 1 gene and bearing a ligand that binds to prostate-specific membrane antigen leads to efficient gene silencing in prostate tumour cells in vitro and when intravenously injected in mouse models of prostate cancer. The octamer's versatile nanocarrier design should offer opportunities for the clinical translation of therapies based on intracellularly acting biologics.
Collapse
Affiliation(s)
- Wanyi Tai
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Junwei Li
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Xiaohu Gao
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
30
|
Chen Y, Li J, Oupický D. Conjugate Polyplexes with Anti-Invasive Properties and Improved siRNA Delivery In Vivo. Bioconjug Chem 2018; 29:296-305. [PMID: 29338191 DOI: 10.1021/acs.bioconjchem.7b00622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This study reports on a simple method to prepare siRNA-polycation conjugate polyplexes by in situ thiol-disulfide exchange reaction. The conjugate polyplexes are prepared using thiol-terminated siRNA and a bioreducible branched polycationic inhibitor of the CXCR4 chemokine receptor (rPAMD). The rPAMD-SS-siRNA conjugate polyplexes exhibit improved colloidal stability and resistance against disassembly with heparin, serum, and physiological salt concentrations when compared with control conventional rPAMD/siRNA polyplexes. Coating the polyplexes with human serum albumin masks the positive surface charge and contributes to the enhanced in vitro gene silencing and improved safety in vivo. The conjugate polyplexes display improved in vivo reporter gene silencing following intravenous injection in tumor-bearing mice. Because the conjugate polyplexes retained the ability of rPAMD to inhibit CXCR4 and restrict cancer cell invasion, the developed systems show promise for future combination anti-metastatic siRNA therapies of cancer.
Collapse
Affiliation(s)
- Yi Chen
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| |
Collapse
|
31
|
Safari F, Rahmani Barouji S, Tamaddon AM. Strategies for Improving siRNA-Induced Gene Silencing Efficiency. Adv Pharm Bull 2017; 7:603-609. [PMID: 29399550 PMCID: PMC5788215 DOI: 10.15171/apb.2017.072] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 11/29/2022] Open
Abstract
Purpose: Human telomerase reverse transcriptase (hTERT)
plays a crucial role in tumorigenesis and progression of cancers. Gene silencing of hTERT
by short interfering RNA (siRNA) is considered as a promising strategy for cancer gene
therapy. Various algorithms have been devised for designing a high efficient siRNA which
is a significant issue in the clinical usage. Thereby, in the present study, the relation
of siRNA designing criteria and the gene silencing efficiency was evaluated. Methods: The siRNA sequences were designed and
characterized by using on line soft wares. Cationic co-polymer (polyethylene
glycol-g-polyethylene imine (PEG-g-PEI)) was used for the construction of polyelectrolyte
complexes (PECs) containing siRNAs. The cellular uptake of the PECs was evaluated. The
gene silencing efficiency of different siRNA sequences was investigated and the effect of
observing the rational designing on the functionality of siRNAs was assessed. Results: The size of PEG-g-PEI siRNA with N/P
(Nitrogen/Phosphate) ratio of 2.5 was 114 ± 0.645 nm. The transfection efficiency of PECs
was desirable (95.5% ± 2.4%.). The results of Real-Time PCR showed that main sequence (MS)
reduced the hTERT expression up to 90% and control positive sequence (CPS) up to 63%.
These findings demonstrated that the accessibility to the target site has priority than
the other criteria such as sequence preferences and thermodynamic features. Conclusion: siRNA opens a hopeful window in cancer therapy
which provides a convenient and tolerable therapeutic approach. Thereby, using the set of
criteria and rational algorithms in the designing of siRNA remarkably affect the gene
silencing efficiency.
Collapse
Affiliation(s)
- Fatemeh Safari
- Medical Biotechnology Department, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Rahmani Barouji
- Department of Traditional Medicine, Faculty of Traditional Medicine, University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammad Tamaddon
- Center for Pharmaceutical Nanotechnology and Biomaterials, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
32
|
Singhsa P, Diaz-Dussan D, Manuspiya H, Narain R. Well-Defined Cationic N-[3-(Dimethylamino)propyl]methacrylamide Hydrochloride-Based (Co)polymers for siRNA Delivery. Biomacromolecules 2017; 19:209-221. [PMID: 29195038 DOI: 10.1021/acs.biomac.7b01475] [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
Cationic glycopolymers have shown to be excellent candidates for the fabrication of gene delivery devices due to their ability to electrostatically interact with negatively charged nucleic acids and the carbohydrate residues ensure enhanced stability and low toxicity of the polyplexes. The ability to engineer the polymers for optimized compositions, molecular weights, and architectures is critical in the design of effective gene delivery vehicles. Therefore, in this study, the aqueous reversible addition-fragmentation chain transfer polymerization (RAFT) was used to synthesize well-defined cationic glycopolymers with various cationic segments. For the preparation of cationic parts, N-[3-(dimethylamino)propyl]methacrylamide hydrochloride (DMAPMA·HCl), water-soluble methacrylamide monomer containing tertiary amine, was polymerized to produce DMAPMA·HCl homopolymer, which was then used as macroCTA in the block copolymerization with two other methacrylamide monomers containing different pendant groups, namely, 2-aminoethyl methacrylamide hydrochloride (AEMA) (with primary amine) and N-(3-aminopropyl) morpholine methacrylamide (MPMA) (with morpholine ring). In addition, statistical copolymers of DMAPMA.HCl with either AEMA or MPMA were also synthesized. All resulting cationic polymers were utilized as macroCTA for the RAFT copolymerization with 2-lactobionamidoethyl methacrylamide (LAEMA), which consists of the pendent galactose residues to achieve DMAPMA·HCl-based glycopolymers. From the in vitro cytotoxicity study, the cationic glycopolymers showed better cell viabilities than the corresponding cationic homopolymers. Furthermore, complexation of the cationic polymers with siRNA, cellular uptake of the resulting polyplexes, and gene knockdown efficiencies were evaluated. All cationic polymers/glycopolymers demonstrated good complexation ability with siRNA at low weight ratios. Among these cationic polymer-siRNA polyplexes, the polyplexes prepared from the two glycopolymers, P(DMAPMA65-b-LAEMA15) and P[(DMAPMA65-b-MPMA63)-b-LAEMA16], showed outstanding results in the cellular uptake, high EGFR knockdown, and low post-transfection toxicity, suggesting the great potential in siRNA delivery of these novel glycopolymers.
Collapse
Affiliation(s)
- Pratyawadee Singhsa
- The Petroleum and Petrochemical College, Center of Excellence on Petrochemical and Materials Technology, Chulalongkorn University , Soi Chulalongkorn 12, Pathumwan, Bangkok 10330, Thailand.,Department of Chemical and Materials Engineering, Donadeo Innovation Centre for Engineering , 116 Street and 85 Avenue, Edmonton, AB T6G 2G6, Canada
| | - Diana Diaz-Dussan
- Department of Chemical and Materials Engineering, Donadeo Innovation Centre for Engineering , 116 Street and 85 Avenue, Edmonton, AB T6G 2G6, Canada
| | - Hathaikarn Manuspiya
- The Petroleum and Petrochemical College, Center of Excellence on Petrochemical and Materials Technology, Chulalongkorn University , Soi Chulalongkorn 12, Pathumwan, Bangkok 10330, Thailand
| | - Ravin Narain
- Department of Chemical and Materials Engineering, Donadeo Innovation Centre for Engineering , 116 Street and 85 Avenue, Edmonton, AB T6G 2G6, Canada
| |
Collapse
|
33
|
Shakouri A, Adljouy N, Balkani S, Mohamadi M, Hamishehkar H, Abdolalizadeh J, Kazem Shakouri S. Effectiveness of topical gel of medical leech (Hirudo medicinalis) saliva extract on patients with knee osteoarthritis: A randomized clinical trial. Complement Ther Clin Pract 2017; 31:352-359. [PMID: 29246745 DOI: 10.1016/j.ctcp.2017.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/29/2017] [Accepted: 12/05/2017] [Indexed: 01/07/2023]
Abstract
Leech saliva extract (LSE) in the liposome-based gel was used as a supplementary treatment to relief the signs and symptoms of osteoarthritis (OA). The saliva of medical leech was extracted and nano liposomes were used to formulate the supplement to enhance skin absorption. A clinical trial was designed to evaluate the therapeutic effects of LSE liposomal gel. Lenquesne and VAS questionnaires were used as indexes of this supplement therapy efficacy for 30 days. Questionnaires analysis showed that after one-month administration of LSE liposomal gel, patients' pain was relieved approximately up to 50%; also, due to reduction in joint inflammation and stiffness, the range of motion was increased and the patients' quality of life was enhanced (p < 0.001). LSE nano scaled liposomal gel as an innovative supplement therapy in OA patients, makes desirable therapeutic approach, which seems to make a significant impact on patient's quality of life and self-care capability.
Collapse
Affiliation(s)
- Amir Shakouri
- Traditional Medicine Faculty, Tabriz university of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasim Adljouy
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanaz Balkani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Mohamadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Paramedical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Seyed Kazem Shakouri
- Physical Medicine & Rehabilitation Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Stealth magnetic nanocarriers of siRNA as platform for breast cancer theranostics. Int J Pharm 2017; 532:660-668. [DOI: 10.1016/j.ijpharm.2017.05.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 01/08/2023]
|
35
|
Krhac Levacic A, Morys S, Wagner E. Solid-phase supported design of carriers for therapeutic nucleic acid delivery. Biosci Rep 2017; 37:BSR20160617. [PMID: 28963371 PMCID: PMC5662914 DOI: 10.1042/bsr20160617] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 12/21/2022] Open
Abstract
Nucleic acid molecules are important therapeutic agents in the field of antisense oligonucleotide, RNA interference, and gene therapies. Since nucleic acids are not able to cross cell membranes and enter efficiently into cells on their own, the development of efficient, safe, and precise delivery systems is the crucial challenge for development of nucleic acid therapeutics. For the delivery of nucleic acids to their intracellular site of action, either the cytosol or the nucleus, several extracellular and intracellular barriers have to be overcome. Multifunctional carriers may handle the different special requirements of each barrier. The complexity of such macromolecules however poses a new hurdle in medical translation, which is the chemical production in reproducible and well-defined form. Solid-phase assisted synthesis (SPS) presents a solution for this challenge. The current review provides an overview on the design and SPS of precise sequence-defined synthetic carriers for nucleic acid cargos.
Collapse
Affiliation(s)
- Ana Krhac Levacic
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, D-81377 Munich, Germany
| | - Stephan Morys
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, D-81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, D-81377 Munich, Germany
- Nanosystems Initiative Munich, Schellingstrasse 4, D-80799 Munich, Germany
| |
Collapse
|
36
|
Xin Y, Huang M, Guo WW, Huang Q, Zhang LZ, Jiang G. Nano-based delivery of RNAi in cancer therapy. Mol Cancer 2017; 16:134. [PMID: 28754120 PMCID: PMC5534073 DOI: 10.1186/s12943-017-0683-y] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/20/2017] [Indexed: 12/31/2022] Open
Abstract
Background RNA interference (RNAi), a newly developed method in which RNA molecules inhibit gene expression, has recently received considerable research attention. In the development of RNAi-based therapies, nanoparticles, which have distinctive size effects along with facile modification strategies and are capable of mediating effective RNAi with targeting potential, are attracting extensive interest. Objective This review presents an overview of the mechanisms of RNAi molecules in gene therapy and the different nanoparticles used to deliver RNAi molecules; briefly describes the current uses of RNAi in cancer therapy along with the nano-based delivery of RNA molecules in previous studies; and highlights some other carriers that have been applied in clinical settings. Finally, we discuss the nano-based delivery of RNAi therapeutics in preclinical development, including the current status and limitations of anti-cancer treatment. Conclusion With the growing number of RNAi therapeutics entering the clinical phase, various nanocarriers are expected to play important roles in the delivery of RNAi molecules for cancer therapeutics.
Collapse
Affiliation(s)
- Yong Xin
- Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, People's Republic of China
| | - Min Huang
- Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, People's Republic of China
| | - Wen Wen Guo
- Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, People's Republic of China
| | - Qian Huang
- Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, People's Republic of China
| | - Long Zhen Zhang
- Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, People's Republic of China
| | - Guan Jiang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
37
|
Feldmann DP, Xie Y, Jones SK, Yu D, Moszczynska A, Merkel OM. The impact of microfluidic mixing of triblock micelleplexes on in vitro / in vivo gene silencing and intracellular trafficking. NANOTECHNOLOGY 2017; 28:224001. [PMID: 28488596 PMCID: PMC5539944 DOI: 10.1088/1361-6528/aa6d15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The triblock copolymer polyethylenimine-polycaprolactone-polyethylene glycol (PEI-PCL-PEG) has been shown to spontaneously assemble into nano-sized particulate carriers capable of complexing with nucleic acids for gene delivery. The objective of this study was to investigate micelleplex characteristics, their in vitro and in vivo fate following microfluidic preparation of siRNA nanoparticles compared to the routinely used batch reactor mixing technique. Herein, PEI-PCL-PEG nanoparticles were prepared with batch reactor or microfluidic mixing techniques and characterized by various biochemical assays and in cell culture. Microfluidic nanoparticles showed a reduction of overall particle size as well as a more uniform size distribution when compared to batch reactor pipette mixing. Confocal microscopy, flow cytometry and qRT-PCR displayed the subcellular delivery of the microfluidic formulation and confirmed the ability to achieve mRNA knockdown. Intratracheal instillation of microfluidic formulation resulted in a significantly more efficient (p < 0.05) knockdown of GAPDH compared to treatment with the batch reactor formulation. The use of microfluidic mixing techniques yields an overall smaller and more uniform PEG-PCL-PEI nanoparticle that is able to more efficiently deliver siRNA in vivo. This preparation method may prove to be useful when a scaled up production of well-defined polyplexes is required.
Collapse
Affiliation(s)
- Daniel P Feldmann
- Department of Oncology, Wayne State University School of Medicine, 4100 John R St, Detroit, MI 48201, United States of America
| | | | | | | | | | | |
Collapse
|
38
|
Cavallaro G, Sardo C, Craparo EF, Porsio B, Giammona G. Polymeric nanoparticles for siRNA delivery: Production and applications. Int J Pharm 2017; 525:313-333. [DOI: 10.1016/j.ijpharm.2017.04.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 02/06/2023]
|
39
|
Synthesis and in vitro characterization of a pH-responsive chitosan- polyethylenimine nanosystem for the delivery of therapeutic proteins. J Drug Deliv Sci Technol 2017. [DOI: 10.1016/j.jddst.2017.03.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
40
|
Venault A, Huang YC, Lo JW, Chou CJ, Chinnathambi A, Higuchi A, Chen WS, Chen WY, Chang Y. Tunable PEGylation of branch-type PEI/DNA polyplexes with a compromise of low cytotoxicity and high transgene expression: in vitro and in vivo gene delivery. J Mater Chem B 2017; 5:4732-4744. [PMID: 32264316 DOI: 10.1039/c7tb01046j] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although PEGylated polyplexes for gene delivery are widespread, there is a need for an in-depth investigation of the role of the PEGylation degree on the delivery efficiency of the systems. For this, a low-toxicity series of polymers for gene delivery were designed via Michael addition of poly(ethylene glycol)methyl ether methacrylate (PEGMA) onto branched polyethylenimine PEI. The goal was to finely tune the PEGylation degree in order to determine the system offering the best compromise between low cytotoxicity and high transfection efficiency under both in vitro and in vivo conditions. From dynamic light scattering tests, zeta potential measurements and gel retardation assay, it was found that nanoparticle assembly of PEI-g-PEGMA and DNA exhibited stable complex formation when the PEGylation degree was below 2.9%. In addition, complexes formed from polymers with a PEGylation degree of at least 1.67% (from PEI-g-PEGMA-6 to PEI-g-PEGMA-18) all showed very low hemolysis activity. Transfection efficiencies of the prepared complexes were determined using the pEGFP-C3 vector and β-galactosidase. Complexes made of PEI-g-PEGMA-6 and PEI-g-PEGMA-10 at a polymer nitrogen/DNA phosphorus weight ratio (Wn/Wp) of 5 led to the best transfection efficiencies. Moreover, PEGylation ensured low cytotoxicity of the complexes in particular at high Wn/Wp ratios. In vivo tests in a mouse model confirmed the in vitro results obtained for PEI-g-PEGMA-6-based complexes, at all Wn/Wp ratios tested, but also showed that a high PEGylation degree (5.2% for PEI-g-PEGMA-18), though inefficient in vitro could still lead to successful delivery in vivo, due to a prolonged contact time between the complex and the cells, and to the change in the biological environment. Overall, provided a fine tuning of the grafting density of PEGMA onto PEI and the polymer nitrogen/DNA phosphorus weight ratio, our results prove that PEI-g-PEGMA polymers constitute an efficient platform for successful in vitro and in vivo gene delivery, and ensure low cytotoxicity and prolonged cell viability.
Collapse
Affiliation(s)
- A Venault
- R&D Center for Membrane Technology and Department of Chemical Engineering, Chung Yuan Christian University, Jhong-Li, Taoyuan 320, Taiwan, Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Influence of Defined Hydrophilic Blocks within Oligoaminoamide Copolymers: Compaction versus Shielding of pDNA Nanoparticles. Polymers (Basel) 2017; 9:polym9040142. [PMID: 30970822 PMCID: PMC6432433 DOI: 10.3390/polym9040142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/07/2017] [Accepted: 04/11/2017] [Indexed: 01/07/2023] Open
Abstract
Cationic polymers are promising components of the versatile platform of non-viral nucleic acid (NA) delivery agents. For a successful gene delivery system, these NA vehicles need to comprise several functionalities. This work focuses on the modification of oligoaminoamide carriers with hydrophilic oligomer blocks mediating nanoparticle shielding potential, which is necessary to prevent aggregation or dissociation of NA polyplexes in vitro, and hinder opsonization with blood components in vivo. Herein, the shielding agent polyethylene glycol (PEG) in three defined lengths (12, 24, or 48 oxyethylene repeats) is compared with two peptidic shielding blocks composed of four or eight repeats of sequential proline-alanine-serine (PAS). With both types of shielding agents, we found opposing effects of the length of hydrophilic segments on shielding and compaction of formed plasmid DNA (pDNA) nanoparticles. Two-arm oligoaminoamides with 37 cationizable nitrogens linked to 12 oxyethylene units or four PAS repeats resulted in very compact 40⁻50 nm pDNA nanoparticles, whereas longer shielding molecules destabilize the investigated polyplexes. Thus, the balance between sufficiently shielded but still compact and stable particles can be considered a critical optimization parameter for non-viral nucleic acid vehicles based on hydrophilic-cationic block oligomers.
Collapse
|
42
|
Werfel TA, Jackson MA, Kavanaugh TE, Kirkbride KC, Miteva M, Giorgio TD, Duvall C. Combinatorial optimization of PEG architecture and hydrophobic content improves ternary siRNA polyplex stability, pharmacokinetics, and potency in vivo. J Control Release 2017; 255:12-26. [PMID: 28366646 DOI: 10.1016/j.jconrel.2017.03.389] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 03/06/2017] [Accepted: 03/23/2017] [Indexed: 12/21/2022]
Abstract
A rationally-designed library of ternary siRNA polyplexes was developed and screened for gene silencing efficacy in vitro and in vivo with the goal of overcoming both cell-level and systemic delivery barriers. [2-(dimethylamino)ethyl methacrylate] (DMAEMA) was homopolymerized or copolymerized (50mol% each) with butyl methacrylate (BMA) from a reversible addition - fragmentation chain transfer (RAFT) chain transfer agent, with and without pre-conjugation to polyethylene glycol (PEG). Both single block polymers were tested as core-forming units, and both PEGylated, diblock polymers were screened as corona-forming units. Ternary siRNA polyplexes were assembled with varied amounts and ratios of core-forming polymers to PEGylated corona-forming polymers. The impact of polymer composition/ratio, hydrophobe (BMA) placement, and surface PEGylation density was correlated to important outcomes such as polyplex size, stability, pH-dependent membrane disruptive activity, biocompatibility, and gene silencing efficiency. The lead formulation, DB4-PDB12, was optimally PEGylated not only to ensure colloidal stability (no change in size by DLS between 0 and 24h) and neutral surface charge (0.139mV) but also to maintain higher cell uptake (>90% positive cells) than the most densely PEGylated particles. The DB4-PDB12 polyplexes also incorporated BMA in both the polyplex core- and corona-forming polymers, resulting in robust endosomolysis and in vitro siRNA silencing (~85% protein level knockdown) of the model gene luciferase across multiple cell types. Further, the DB4-PDB12 polyplexes exhibited greater stability, increased blood circulation time, reduced renal clearance, increased tumor biodistribution, and greater silencing of luciferase compared to our previously-optimized, binary parent formulation following intravenous (i.v.) delivery. This polyplex library approach enabled concomitant optimization of the composition and ratio of core- and corona-forming polymers (indirectly tuning PEGylation density) and identification of a ternary nanomedicine optimized to overcome important siRNA delivery barriers in vitro and in vivo.
Collapse
Affiliation(s)
- Thomas A Werfel
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN, USA
| | - Meredith A Jackson
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN, USA
| | - Taylor E Kavanaugh
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN, USA
| | - Kellye C Kirkbride
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN, USA
| | - Martina Miteva
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN, USA
| | - Todd D Giorgio
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN, USA
| | - Craig Duvall
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN, USA.
| |
Collapse
|
43
|
Li Y, Lee RJ, Yu K, Bi Y, Qi Y, Sun Y, Li Y, Xie J, Teng L. Delivery of siRNA Using Lipid Nanoparticles Modified with Cell Penetrating Peptide. ACS APPLIED MATERIALS & INTERFACES 2016; 8:26613-26621. [PMID: 27617513 DOI: 10.1021/acsami.6b09991] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Clinical development of siRNA has been hindered by the lack of an effective delivery system. Here, we report the construction of a novel siRNA delivery system, sTOLP, which is based on cell penetrating peptide oleoyl-octaarginine (OA-R8) modified multifunctional lipid nanoparticles. sTOLP nanoparticles are composed of a protamine complexed siRNA core, OA-R8, cationic and PEGylated lipids, and transferrin as a targeting ligand. sTOLP formulation was optimized and characterized in vitro and showed excellent gene silencing activity. In vivo, siRNA encapsulated in sTOLP exhibited potent tumor inhibition (61.7%) and was preferentially taken up by hepatocytes and tumor cells in HepG2-bearing nude mice without inducing immunogenicity or hepatic or renal toxicity. Furthermore, sTOLP-loaded siRNA had stability in circulation greater than that of free siRNA. These data demonstrated potential utility of sTOLP-mediated siRNA delivery in cancer therapy.
Collapse
Affiliation(s)
- Yuhuan Li
- School of Life Sciences, Jilin University , Changchun 130012, China
| | - Robert J Lee
- School of Life Sciences, Jilin University , Changchun 130012, China
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University , Columbus, Ohio 43210, United States
| | - Kongtong Yu
- School of Life Sciences, Jilin University , Changchun 130012, China
| | - Ye Bi
- School of Life Sciences, Jilin University , Changchun 130012, China
| | - Yuhang Qi
- School of Life Sciences, Jilin University , Changchun 130012, China
| | - Yating Sun
- School of Life Sciences, Jilin University , Changchun 130012, China
| | - Yujing Li
- School of Life Sciences, Jilin University , Changchun 130012, China
| | - Jing Xie
- School of Life Sciences, Jilin University , Changchun 130012, China
| | - Lesheng Teng
- School of Life Sciences, Jilin University , Changchun 130012, China
| |
Collapse
|
44
|
Wang Y, Kumar S, Rachagani S, Sajja BR, Xie Y, Hang Y, Jain M, Li J, Boska MD, Batra SK, Oupický D. Polyplex-mediated inhibition of chemokine receptor CXCR4 and chromatin-remodeling enzyme NCOA3 impedes pancreatic cancer progression and metastasis. Biomaterials 2016; 101:108-120. [PMID: 27267632 PMCID: PMC4921319 DOI: 10.1016/j.biomaterials.2016.05.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/22/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer (PC) is one of the most aggressive malignancies due to intense desmoplasia, extreme hypoxia and inherent chemoresistance. Studies have implicated the expression of chemokine receptor CXCR4 and nuclear receptor co-activator-3 (NCOA3) in the development of desmoplasia and metastatic spread of PC. Using a series of polymeric CXCR4 antagonists (PCX), we optimized formulation of PCX/siNCOA3 polyplexes to simultaneously target CXCR4 and NCOA3 in PC. Cholesterol-modified PCX showed maximum CXCR4 antagonism, NCOA3 silencing and inhibition of PC cell migration in vitro. The optimized PCX/siNCOA3 polyplexes were used in evaluating antitumor and antimetastatic activity in orthotopic mouse model of metastatic PC. The polyplexes displayed significant inhibition of primary tumor growth, which was accompanied by a decrease in tumor necrosis and increased tumor perfusion. The polyplexes also showed significant antimetastatic effect and effective suppression of metastasis to distant organs. Overall, dual-function PCX/siNCOA3 polyplexes can effectively regulate tumor microenvironment to decrease progression and dissemination of PC.
Collapse
Affiliation(s)
- Yan Wang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Nebraska, United States
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Nebraska, United States
| | | | - Ying Xie
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Yu Hang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Nebraska, United States
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Michael D. Boska
- Department of Radiology, University of Nebraska Medical Center, Nebraska, United States
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Nebraska, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
45
|
Aguirre G, Ramos J, Forcada J. Advanced design of t and pH dual-responsive PDEAEMA-PVCL core-shell nanogels for siRNA delivery. ACTA ACUST UNITED AC 2016. [DOI: 10.1002/pola.28207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Garbiñe Aguirre
- POLYMAT, Bionanoparticles Group, Department of Applied Chemistry, UFI 11/56, Faculty of Chemistry; University of the Basque Country UPV/EHU; Apdo. 1072 Donostia-San Sebastián 20080 Spain
| | - Jose Ramos
- POLYMAT, Bionanoparticles Group, Department of Applied Chemistry, UFI 11/56, Faculty of Chemistry; University of the Basque Country UPV/EHU; Apdo. 1072 Donostia-San Sebastián 20080 Spain
| | - Jacqueline Forcada
- POLYMAT, Bionanoparticles Group, Department of Applied Chemistry, UFI 11/56, Faculty of Chemistry; University of the Basque Country UPV/EHU; Apdo. 1072 Donostia-San Sebastián 20080 Spain
| |
Collapse
|
46
|
Huang Y, Cheng Q, Ji JL, Zheng S, Du L, Meng L, Wu Y, Zhao D, Wang X, Lai L, Cao H, Xiao K, Gao S, Liang Z. Pharmacokinetic Behaviors of Intravenously Administered siRNA in Glandular Tissues. Am J Cancer Res 2016; 6:1528-41. [PMID: 27446488 PMCID: PMC4955053 DOI: 10.7150/thno.15246] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/18/2016] [Indexed: 02/05/2023] Open
Abstract
The pharmacokinetics of small interfering RNAs (siRNAs) is a pivotal issue for siRNA-based drug development. In this study, we comprehensively investigated the behavior of siRNAs in vivo in various tissues and demonstrated that intravenously-injected naked siRNA accumulated remarkably in the submandibular gland, bulbourethral gland, and pancreas, with a respective half-life of ~22.7, ~45.6, and ~30.3 h. This was further confirmed by gel separation of tissue homogenates and/or supernatants. In vivo imaging and cryosectioning suggested that delivery carriers significantly influence the distribution and elimination profiles of siRNA. Gene-silencing assays revealed that neither naked nor liposome-formulated siRNA resulted in gene knockdown in the submandibular and bulbourethral glands after systemic administration, suggesting that these glands function as drug reservoirs that enable slow siRNA release into the circulation. But robust gene-silencing was achieved by local injection of liposome-encapsulated siRNA into the submandibular gland. Our results enhance understanding of the pharmacokinetic properties of siRNAs and we believe that they will facilitate the development of siRNA therapy, especially for the submandibular gland.
Collapse
|
47
|
Zhang W, Müller K, Kessel E, Reinhard S, He D, Klein PM, Höhn M, Rödl W, Kempter S, Wagner E. Targeted siRNA Delivery Using a Lipo-Oligoaminoamide Nanocore with an Influenza Peptide and Transferrin Shell. Adv Healthc Mater 2016; 5:1493-504. [PMID: 27109317 DOI: 10.1002/adhm.201600057] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 02/23/2016] [Indexed: 01/06/2023]
Abstract
Developing RNA-interference-based therapeutic approaches with efficient and targeted cytosolic delivery of small interfering RNA (siRNA) is remaining a critical challenge since two decades. Herein, a multifunctional transferrin receptor (TfR)-targeted siRNA delivery system (Tf&INF7) is designed based on siRNA complexes formed with the cationic lipo-oligoamino amide 454, sequentially surface-modified with polyethylene glycol-linked transferrin (Tf) for receptor targeting and the endosomolytic peptide INF7 for efficient cytosolic release of the siRNA. Effective Tf&INF7 polyplex internalization and target gene silencing are demonstrated for the TfR overexpressing tumor cell lines (K562, D145, and N2a). Treatment with antitumoral EG5 siRNA results in a block of tumor cell growth and triggered apoptosis. Tf-modified polyplexes are far more effective than the corresponding albumin- (Alb) or nonmodified 454 polyplexes. Competition experiments with excess of Tf demonstrate TfR target specificity. As alternative to the ligand Tf, an anti-murine TfR antibody is incorporated into the polyplexes for specific targeting and gene silencing in the murine N2a cell line. In vivo distribution studies not only demonstrate an enhanced tumor residence of siRNA in N2a tumor-bearing mice with the Tf&INF7 as compared to the 454 polyplex group but also a reduced siRNA nanoparticle stability limiting the in vivo performance.
Collapse
Affiliation(s)
- Wei Zhang
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Katharina Müller
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Eva Kessel
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
- Nanosystems Initiative Munich Schellingstrasse 4 D‐80799 Munich Germany
| | - Sören Reinhard
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Dongsheng He
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
- Nanosystems Initiative Munich Schellingstrasse 4 D‐80799 Munich Germany
| | - Philipp M. Klein
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Wolfgang Rödl
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Susanne Kempter
- Department of Physics Ludwig‐Maximilians‐Universität München Geschwister‐Scholl‐Platz 1 80539 Munich Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
- Nanosystems Initiative Munich Schellingstrasse 4 D‐80799 Munich Germany
| |
Collapse
|
48
|
A novel tyrosine-modified low molecular weight polyethylenimine (P10Y) for efficient siRNA delivery in vitro and in vivo. J Control Release 2016; 230:13-25. [DOI: 10.1016/j.jconrel.2016.03.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/23/2016] [Accepted: 03/25/2016] [Indexed: 11/17/2022]
|
49
|
Sarett SM, Werfel TA, Chandra I, Jackson MA, Kavanaugh TE, Hattaway ME, Giorgio TD, Duvall CL. Hydrophobic interactions between polymeric carrier and palmitic acid-conjugated siRNA improve PEGylated polyplex stability and enhance in vivo pharmacokinetics and tumor gene silencing. Biomaterials 2016; 97:122-32. [PMID: 27163624 DOI: 10.1016/j.biomaterials.2016.04.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 12/11/2022]
Abstract
Formation of stable, long-circulating siRNA polyplexes is a significant challenge in translation of intravenously-delivered, polymeric RNAi cancer therapies. Here, we report that siRNA hydrophobization through conjugation to palmitic acid (siPA) improves stability, in vivo pharmacokinetics, and tumor gene silencing of PEGylated nanopolyplexes (siPA-NPs) with balanced cationic and hydrophobic content in the core relative to the analogous polyplexes formed with unmodified siRNA, si-NPs. Hydrophobized siPA loaded into the NPs at a lower charge ratio (N(+):P(-)) relative to unmodified siRNA, and siPA-NPs had superior resistance to siRNA cargo unpackaging in comparison to si-NPs upon exposure to the competing polyanion heparin and serum. In vitro, siPA-NPs increased uptake in MDA-MB-231 breast cancer cells (100% positive cells vs. 60% positive cells) but exhibited equivalent silencing of the model gene luciferase relative to si-NPs. In vivo in a murine model, the circulation half-life of intravenously-injected siPA-NPs was double that of si-NPs, resulting in a >2-fold increase in siRNA biodistribution to orthotopic MDA-MB-231 mammary tumors. The increased circulation half-life of siPA-NPs was dependent upon the hydrophobic interactions of the siRNA and the NP core component and not just siRNA hydrophobization, as siPA did not contribute to improved circulation time relative to unmodified siRNA when delivered using polyplexes with a fully cationic core. Intravenous delivery of siPA-NPs also achieved significant silencing of the model gene luciferase in vivo (∼40% at 24 h after one treatment and ∼60% at 48 h after two treatments) in the murine MDA-MB-231 tumor model, while si-NPs only produced a significant silencing effect after two treatments. These data suggest that stabilization of PEGylated siRNA polyplexes through a combination of hydrophobic and electrostatic interactions between siRNA cargo and the polymeric carrier improves in vivo pharmacokinetics and tumor gene silencing relative to conventional formulations that are stabilized solely by electrostatic interactions.
Collapse
Affiliation(s)
- Samantha M Sarett
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA
| | - Thomas A Werfel
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA
| | - Irene Chandra
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA
| | - Meredith A Jackson
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA
| | - Taylor E Kavanaugh
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA
| | - Madison E Hattaway
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA
| | - Todd D Giorgio
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA.
| |
Collapse
|
50
|
Abstract
Small interfering RNA (siRNA) is emerging as a class of therapeutic with extremely high potential, particularly in the field of oncology. Despite this growing interest, further understanding of how siRNA behaves in vivo is still required before significant uptake into clinical application. To this end, many molecular imaging modalities have been utilised to gain a better understanding of the biodistribution and pharmacokinetics of administered siRNA and delivery vehicles. This highlight aims to provide an overview of the current state of the field for preclinical imaging of siRNA delivery.
Collapse
|