1
|
Alvarez AC, Maguire D, Brannigan RP. Synthetic-polymer-assisted antisense oligonucleotide delivery: targeted approaches for precision disease treatment. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:435-463. [PMID: 40166479 PMCID: PMC11956074 DOI: 10.3762/bjnano.16.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025]
Abstract
This review explores the recent advancements in polymer-assisted delivery systems for antisense oligonucleotides (ASOs) and their potential in precision disease treatment. Synthetic polymers have shown significant promise in enhancing the delivery, stability, and therapeutic efficacy of ASOs by addressing key challenges such as cellular uptake, endosomal escape, and reducing cytotoxicity. The review highlights key studies from the past decade demonstrating how these polymers improve gene silencing efficiencies, particularly in cancer and neurodegenerative disease models. Despite the progress achieved, barriers such as immunogenicity, delivery limitations, and scalability still need to be overcome for broader clinical application. Emerging strategies, including stimuli-responsive polymers and advanced nanoparticle systems, offer potential solutions to these challenges. The review underscores the transformative potential of polymer-enhanced ASO delivery in personalised medicine, emphasising the importance of continued innovation to optimise ASO-based therapeutics for more precise and effective disease treatments.
Collapse
Affiliation(s)
- Ana Cubillo Alvarez
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Dylan Maguire
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Ruairí P Brannigan
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
2
|
Koo J, Shin Y, Jeon H, Cheong J, Cho S, Park C, Song EC, Ramsey JD, Lim C, Oh KT. Enhancing glioblastoma therapy via intranasal administration of highly potent cell-penetrating peptide decorated nanoparticles. J Control Release 2025; 378:997-1012. [PMID: 39724950 DOI: 10.1016/j.jconrel.2024.12.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/04/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Glioblastoma multiforme (GBM) is a devastating primary tumor of the central nervous system with a significantly poor prognosis. The primary challenge in treating GBM lies in the restrictive nature of the blood-brain barrier (BBB), impeding effective drug delivery to the brain. In this study, intranasal polymeric micelles encapsulating a quercetin-etoposide combination were developed to induce synergistic apoptotic effects and enhance direct drug delivery to the brain. However, the in vivo anticancer efficacy of the unmodified micelle formulation via intranasal administration remains limited. Therefore, this aims to investigate the enhancement of the formulation by conjugating the micelles with a novel and highly potent cell-penetrating peptide (CPP), RMMR1, identified using the intra-dermal delivery technology platform developed by REMEDI Co., Ltd. This modification seeks to enhance the brain-targeting capability of the micelles. The CPP-modified micelles encapsulating the quercetin-etoposide combination (CM(QE)) demonstrated superior in vivo brain-delivery efficiency and enhanced cellular uptake after intranasal administration. Furthermore, animal studies showed significant tumor reduction and increased survival rates, with no significant changes in body weight observed. These findings suggest that intranasal administration of CM(QE) holds promise as a significant advancement in chemotherapy for GBM.
Collapse
Affiliation(s)
- Jain Koo
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Yuseon Shin
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Hyewon Jeon
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Jaehyun Cheong
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Seongmin Cho
- Remedi Co., Ltd. Research center, Songdo 21990, Republic of Korea
| | - Chanho Park
- Remedi Co., Ltd. Research center, Songdo 21990, Republic of Korea
| | - Ee Chan Song
- Remedi Co., Ltd. Research center, Songdo 21990, Republic of Korea
| | - Jacob D Ramsey
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Chaemin Lim
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea; CHA Advanced Research Institute, CHA Bundang Medical Center, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 13488 Gyeonggi-do, Republic of Korea.
| | - Kyung Taek Oh
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
3
|
Wan D, Wu Y, Liu Y, Liu Y, Pan J. Advances in 2,3-Dimethylmaleic Anhydride (DMMA)-Modified Nanocarriers in Drug Delivery Systems. Pharmaceutics 2024; 16:809. [PMID: 38931929 PMCID: PMC11207803 DOI: 10.3390/pharmaceutics16060809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer represents a significant threat to human health. The cells and tissues within the microenvironment of solid tumors exhibit complex and abnormal properties in comparison to healthy tissues. The efficacy of nanomedicines is inhibited by the presence of substantial and complex physical barriers in the tumor tissue. The latest generation of intelligent drug delivery systems, particularly nanomedicines capable of charge reversal, have shown promise in addressing this issue. These systems can transform their charge from negative to positive upon reaching the tumor site, thereby enhancing tumor penetration via transcytosis and promoting cell internalization by interacting with the negatively charged cell membranes. The modification of nanocarriers with 2,3-dimethylmaleic anhydride (DMMA) and its derivatives, which are responsive to weak acid stimulation, represents a significant advance in the field of charge-reversal nanomedicines. This review provides a comprehensive examination of the recent insights into DMMA-modified nanocarriers in drug delivery systems, with a particular focus on their potential in targeted therapeutics. It also discusses the synthesis of DMMA derivatives and their role in charge reversal, shell detachment, size shift, and ligand reactivation mechanisms, offering the prospect of a tailored, next-generation therapeutic approach to overcome the diverse challenges associated with cancer therapy.
Collapse
Affiliation(s)
- Dong Wan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (D.W.); (Y.W.)
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China;
| | - Yanan Wu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (D.W.); (Y.W.)
| | - Yujun Liu
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China;
| | - Yonghui Liu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (D.W.); (Y.W.)
| | - Jie Pan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (D.W.); (Y.W.)
| |
Collapse
|
4
|
Roy PS. Complex Coacervate-Based Materials for Biomedicine: Recent Advancements and Future Prospects. Ind Eng Chem Res 2024; 63:5414-5487. [DOI: 10.1021/acs.iecr.3c03830] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Partha Sarathi Roy
- Division of Pharmaceutical Sciences, Health Sciences Building, University of Missouri─Kansas City, 2464 Charlotte St., Kansas City, Missouri 64108-2718, United States
- Department of Pharmaceutics/Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, 751 Brookside Rd., Stockton, California 95211, United States
| |
Collapse
|
5
|
Lyu M, Yazdi M, Lin Y, Höhn M, Lächelt U, Wagner E. Receptor-Targeted Dual pH-Triggered Intracellular Protein Transfer. ACS Biomater Sci Eng 2024; 10:99-114. [PMID: 35802884 DOI: 10.1021/acsbiomaterials.2c00476] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein therapeutics are of widespread interest due to their successful performance in the current pharmaceutical and medical fields, even though their broad applications have been hindered by the lack of an efficient intracellular delivery approach. Herein, we fabricated an active-targeted dual pH-responsive delivery system with favorable tumor cell entry augmented by extracellular pH-triggered charge reversal and tumor receptor targeting and pH-controlled endosomal release in a traceless fashion. As a traceable model protein, the enhanced green fluorescent protein (eGFP) bearing a nuclear localization signal was covalently coupled with a pH-labile traceless azidomethyl-methylmaleic anhydride (AzMMMan) linker followed by functionalization with different molar equivalents of two dibenzocyclooctyne-octa-arginine-cysteine (DBCO-R8C)-modified moieties: polyethylene glycol (PEG)-GE11 peptide for epidermal growth factor receptor-mediated targeting and melittin for endosomal escape. The cationic melittin domain was masked with tetrahydrophthalic anhydride revertible at mild acidic pH 6.5. At the optimally balanced ratio of functional units, the on-demand charge conversion at tumoral extracellular pH 6.5 in combination with GE11-mediated targeting triggered enhanced electrostatic cellular attraction by the R8C cell-penetrating peptides and melittin, as demonstrated by strongly enhanced cellular uptake. Successful endosomal release followed by nuclear localization of the eGFP cargo was obtained by taking advantage of melittin-mediated endosomal escape and rapid traceless release from the AzMMMan linker. The effectiveness of this multifunctional bioresponsive system suggests a promising strategy for delivery of protein drugs toward intracellular targets. A possible therapeutic relevance was indicated by an example of cytosolic delivery of cytochrome c initiating the apoptosis pathway to kill cancer cells.
Collapse
Affiliation(s)
- Meng Lyu
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Yi Lin
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Ulrich Lächelt
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| |
Collapse
|
6
|
Nakevska Z, Yokota T. Challenges and future perspective of antisense therapy for spinal muscular atrophy: A review. Eur J Cell Biol 2023; 102:151326. [PMID: 37295266 DOI: 10.1016/j.ejcb.2023.151326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Spinal muscular atrophy (SMA), the most common genetic cause of infantile death, is caused by a mutation in the survival of motor neuron 1 gene (SMN1), leading to the death of motor neurons and progressive muscle weakness. SMN1 normally produces an essential protein called SMN. Although humans possess a paralogous gene called SMN2, ∼90% of the SMN it produces is non-functional. This is due to a mutation in SMN2 that causes the skipping of a required exon during splicing of the pre-mRNA. The first treatment for SMA, nusinersen (brand name Spinraza), was approved by the FDA in 2016 and by the EMU in 2017. Nusinersen is an antisense oligonucleotide-based therapy that alters the splicing of SMN2 to make functional full-length SMN protein. Despite the recent advancements in antisense oligonucleotide therapy and SMA treatment development, nusinersen is faced with a multitude of challenges, such as intracellular and systemic delivery. In recent years, the use of peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) in antisense therapy has gained interest. These are antisense oligonucleotides conjugated to cell-penetrating peptides such as Pips and DG9, and they have the potential to address the challenges associated with delivery. This review focuses on the historic milestones, development, current challenges, and future perspectives of antisense therapy for SMA.
Collapse
Affiliation(s)
- Zorica Nakevska
- Department of Biological Sciences, Faculty of Science, University of Alberta, 116 St. and 85 Ave., Edmonton AB T6G 2E1, Canada.
| | - Toshifumi Yokota
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, 116 St. and 85 Ave., Edmonton AB T6G 2E1, Canada; Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, 116 St. and 85 Ave., Edmonton AB T6G 2E1, Canada; The Friends of Garret Cumming Research and Muscular Dystrophy Canada HM Toupin Neurological Science Research Chair, 8812 112 St., Edmonton AB T6G 2H7, Canada.
| |
Collapse
|
7
|
Foo W, Cseresnyés Z, Rössel C, Teng Y, Ramoji A, Chi M, Hauswald W, Huschke S, Hoeppener S, Popp J, Schacher FH, Sierka M, Figge MT, Press AT, Bauer M. Tuning the corona-core ratio of polyplex micelles for selective oligonucleotide delivery to hepatocytes or hepatic immune cells. Biomaterials 2023; 294:122016. [PMID: 36702000 DOI: 10.1016/j.biomaterials.2023.122016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/06/2023] [Accepted: 01/14/2023] [Indexed: 01/22/2023]
Abstract
Targeted delivery of oligonucleotides or small molecular drugs to hepatocytes, the liver's parenchymal cells, is challenging without targeting moiety due to the highly efficient mononuclear phagocyte system (MPS) of the liver. The MPS comprises Kupffer cells and specialized sinusoidal endothelial cells, efficiently clearing nanocarriers regardless of their size and surface properties. Physiologically, this non-parenchymal shield protects hepatocytes; however, these local barriers must be overcome for drug delivery. Nanocarrier structural properties strongly influence tissue penetration, in vivo pharmacokinetics, and biodistribution profile. Here we demonstrate the in vivo biodistribution of polyplex micelles formed by polyion complexation of short interfering (si)RNA with modified poly(ethylene glycol)-block-poly(allyl glycidyl ether) (PEG-b-PAGE) diblock copolymer that carries amino moieties in the side chain. The ratio between PEG corona and siRNA complexed PAGE core of polyplex micelles was chemically varied by altering the degree of polymerization of PAGE. Applying Raman-spectroscopy and dynamic in silico modeling on the polyplex micelles, we determined the corona-core ratio (CCR) and visualized the possible micellar structure with varying CCR. The results for this model system reveal that polyplex micelles with higher CCR, i.e., better PEG coverage, exclusively accumulate and thus allow passive cell-type-specific targeting towards hepatocytes, overcoming the macrophage-rich reticuloendothelial barrier of the liver.
Collapse
Affiliation(s)
- WanLing Foo
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Am Klinikum 1, 07747, Jena, Germany
| | - Zoltán Cseresnyés
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Research Group Applied Systems Biology, Beutenbergstraße 13, 07745, Jena, Germany
| | - Carsten Rössel
- Friedrich-Schiller-University, Jena Center for Soft Matter, Philosophenweg 7, 07743, Jena, Germany; Friedrich-Schiller-University, Institute of Organic Chemistry and Macromolecular Chemistry (IOMC), Humboldtstraße 10, 07743, Jena, Germany
| | - Yingfeng Teng
- Friedrich-Schiller-University, Computational Materials Science Group, Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Löbdergraben 32, 07743, Jena, Germany
| | - Anuradha Ramoji
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745, Jena, Germany; Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich-Schiller-University, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University, Am Klinikum 1, 07747, Jena, Germany
| | - Mingzhe Chi
- Friedrich-Schiller-University, Computational Materials Science Group, Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Löbdergraben 32, 07743, Jena, Germany
| | - Walter Hauswald
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745, Jena, Germany
| | - Sophie Huschke
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Am Klinikum 1, 07747, Jena, Germany
| | - Stephanie Hoeppener
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Research Group Applied Systems Biology, Beutenbergstraße 13, 07745, Jena, Germany; Friedrich-Schiller-University, Institute of Organic Chemistry and Macromolecular Chemistry (IOMC), Humboldtstraße 10, 07743, Jena, Germany
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745, Jena, Germany; Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich-Schiller-University, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University, Am Klinikum 1, 07747, Jena, Germany
| | - Felix H Schacher
- Friedrich-Schiller-University, Jena Center for Soft Matter, Philosophenweg 7, 07743, Jena, Germany; Friedrich-Schiller-University, Institute of Organic Chemistry and Macromolecular Chemistry (IOMC), Humboldtstraße 10, 07743, Jena, Germany
| | - Marek Sierka
- Friedrich-Schiller-University, Jena Center for Soft Matter, Philosophenweg 7, 07743, Jena, Germany; Friedrich-Schiller-University, Computational Materials Science Group, Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Löbdergraben 32, 07743, Jena, Germany
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Research Group Applied Systems Biology, Beutenbergstraße 13, 07745, Jena, Germany; Institute of Microbiology, Faculty of Biological Sciences, Friedrich-Schiller-University Jena, 07743, Jena, Germany; Friedrich-Schiller-University, Jena Center for Soft Matter, Philosophenweg 7, 07743, Jena, Germany
| | - Adrian T Press
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Am Klinikum 1, 07747, Jena, Germany; Friedrich-Schiller-University, Jena Center for Soft Matter, Philosophenweg 7, 07743, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University, Am Klinikum 1, 07747, Jena, Germany; Friedrich-Schiller-University, Faculty of Medicine, Kastanienstraße. 1, 07747, Jena, Germany.
| | - Michael Bauer
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Am Klinikum 1, 07747, Jena, Germany; Friedrich-Schiller-University, Jena Center for Soft Matter, Philosophenweg 7, 07743, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University, Am Klinikum 1, 07747, Jena, Germany.
| |
Collapse
|
8
|
Braatz D, Cherri M, Tully M, Dimde M, Ma G, Mohammadifar E, Reisbeck F, Ahmadi V, Schirner M, Haag R. Chemical Approaches to Synthetic Drug Delivery Systems for Systemic Applications. Angew Chem Int Ed Engl 2022; 61:e202203942. [PMID: 35575255 PMCID: PMC10091760 DOI: 10.1002/anie.202203942] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Indexed: 11/10/2022]
Abstract
Poor water solubility and low bioavailability of active pharmaceutical ingredients (APIs) are major causes of friction in the pharmaceutical industry and represent a formidable hurdle for pharmaceutical drug development. Drug delivery remains the major challenge for the application of new small-molecule drugs as well as biopharmaceuticals. The three challenges for synthetic delivery systems are: (i) controlling drug distribution and clearance in the blood; (ii) solubilizing poorly water-soluble agents, and (iii) selectively targeting specific tissues. Although several polymer-based systems have addressed the first two demands and have been translated into clinical practice, no targeted synthetic drug delivery system has reached the market. This Review is designed to provide a background on the challenges and requirements for the design and translation of new polymer-based delivery systems. This report will focus on chemical approaches to drug delivery for systemic applications.
Collapse
Affiliation(s)
- Daniel Braatz
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Mariam Cherri
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Michael Tully
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Mathias Dimde
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Guoxin Ma
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Ehsan Mohammadifar
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Felix Reisbeck
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Vahid Ahmadi
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Michael Schirner
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Rainer Haag
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| |
Collapse
|
9
|
Zhang P, Chen D, Li L, Sun K. Charge reversal nano-systems for tumor therapy. J Nanobiotechnology 2022; 20:31. [PMID: 35012546 PMCID: PMC8751315 DOI: 10.1186/s12951-021-01221-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022] Open
Abstract
Surface charge of biological and medical nanocarriers has been demonstrated to play an important role in cellular uptake. Owing to the unique physicochemical properties, charge-reversal delivery strategy has rapidly developed as a promising approach for drug delivery application, especially for cancer treatment. Charge-reversal nanocarriers are neutral/negatively charged at physiological conditions while could be triggered to positively charged by specific stimuli (i.e., pH, redox, ROS, enzyme, light or temperature) to achieve the prolonged blood circulation and enhanced tumor cellular uptake, thus to potentiate the antitumor effects of delivered therapeutic agents. In this review, we comprehensively summarized the recent advances of charge-reversal nanocarriers, including: (i) the effect of surface charge on cellular uptake; (ii) charge-conversion mechanisms responding to several specific stimuli; (iii) relation between the chemical structure and charge reversal activity; and (iv) polymeric materials that are commonly applied in the charge-reversal delivery systems.
Collapse
Affiliation(s)
- Peng Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong, People's Republic of China.
| | - Daoyuan Chen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong, People's Republic of China
| | - Lin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong, People's Republic of China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong, People's Republic of China.,State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co. Ltd, Yantai, 264003, People's Republic of China
| |
Collapse
|
10
|
Takemoto H, Nishiyama N. Construction of nanomaterials based on pH-responsive polymers for effective tumor delivery. Polym J 2021. [DOI: 10.1038/s41428-021-00542-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
11
|
Wang X, Song Z, Wei S, Ji G, Zheng X, Fu Z, Cheng J. Polypeptide-based drug delivery systems for programmed release. Biomaterials 2021; 275:120913. [PMID: 34217020 DOI: 10.1016/j.biomaterials.2021.120913] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 01/01/2023]
Abstract
Recent years have seen increasing interests in the use of ring-opening polymerization of α-amino acid N-carboxyanhydrides (NCAs) to prepare synthetic polypeptides, a class of biocompatible and versatile materials, for various biomedical applications. Because of their rich side-chain functionalities, diverse hydrophilicity/hydrophobicity profiles, and the capability of forming stable secondary structures, polypeptides can assemble into a variety of well-organized nano-structures that have unique advantages in drug delivery and controlled release. Herein, we review the design and use of polypeptide-based drug delivery system derived from NCA chemistry, and discuss the future perspectives of this exciting and important biomaterial area that may potentially change the landscape of next-generation therapeutics and diagnosis. Given the high significance of precise control over release for polypeptide-based systems, we specifically focus on the versatile designs of drug delivery systems capable of programmed release, through the changes in the chemical and physical properties controlled by the built-in molecular structures of polypeptides.
Collapse
Affiliation(s)
- Xu Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China; Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Ziyuan Song
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States; Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China.
| | - Shiqi Wei
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Guonan Ji
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Xuetao Zheng
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Zihuan Fu
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States; Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States.
| |
Collapse
|
12
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
13
|
Camal Ruggieri IN, Cícero AM, Issa JPM, Feldman S. Bone fracture healing: perspectives according to molecular basis. J Bone Miner Metab 2021; 39:311-331. [PMID: 33151416 DOI: 10.1007/s00774-020-01168-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
Fractures have a great impact on health all around the world and with fracture healing optimization; this problem could be resolved partially. To make a practical contribution to this issue, the knowledge of bone tissue, cellularity, and metabolism is essential, especially cytoskeletal architecture and its transformations according to external pressures. Special physical and chemical characteristics of the extracellular matrix (ECM) allow the transmission of mechanical stimuli from outside the cell to the plasmatic membrane. The osteocyte cytoskeleton is conformed by a complex network of actin and microtubules combined with crosslinker proteins like vinculin and fimbrin, connecting and transmitting outside stimuli through EMC to cytoplasm. Herein, critical signaling pathways like Cx43-depending ones, MAPK/ERK, Wnt, YAP/TAZ, Rho-ROCK, and others are activated due to mechanical stimuli, resulting in osteocyte cytoskeletal changes and ECM remodeling, altering the tissue and, therefore, the bone. In recent years, the osteocyte has gained more interest and value in relation to bone homeostasis as a great coordinator of other cell populations, thanks to its unique functions. By integrating the latest advances in relation to intracellular signaling pathways, mechanotransmission system of the osteocyte and bone tissue engineering, there are promising experimental strategies, while some are ready for clinical trials. This work aims to show clearly and precisely the integration between cytoskeleton and main molecular pathways in relation to mechanotransmission mechanism in osteocytes, and the use of this theoretical knowledge in therapeutic tools for bone fracture healing.
Collapse
Affiliation(s)
- Iván Nadir Camal Ruggieri
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina.
| | - Andrés Mauricio Cícero
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
| | | | - Sara Feldman
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
- Research Council of the Rosario National University (CIUNR) and CONICET, Rosario, Argentina
| |
Collapse
|
14
|
Mi P, Cabral H, Kataoka K. Ligand-Installed Nanocarriers toward Precision Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1902604. [PMID: 31353770 DOI: 10.1002/adma.201902604] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/04/2019] [Indexed: 05/20/2023]
Abstract
Development of drug-delivery systems that selectively target neoplastic cells has been a major goal of nanomedicine. One major strategy for achieving this milestone is to install ligands on the surface of nanocarriers to enhance delivery to target tissues, as well as to enhance internalization of nanocarriers by target cells, which improves accuracy, efficacy, and ultimately enhances patient outcomes. Herein, recent advances regarding the development of ligand-installed nanocarriers are introduced and the effect of their design on biological performance is discussed. Besides academic achievements, progress on ligand-installed nanocarriers in clinical trials is presented, along with the challenges faced by these formulations. Lastly, the future perspectives of ligand-installed nanocarriers are discussed, with particular emphasis on their potential for emerging precision therapies.
Collapse
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No.17 People's South Road, Chengdu, 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
- Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
15
|
Mi P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020; 10:4557-4588. [PMID: 32292515 PMCID: PMC7150471 DOI: 10.7150/thno.38069] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/24/2020] [Indexed: 02/05/2023] Open
Abstract
In recent years, much progress has been motivated in stimuli-responsive nanocarriers, which could response to the intrinsic physicochemical and pathological factors in diseased regions to increase the specificity of drug delivery. Currently, numerous nanocarriers have been engineered with physicochemical changes in responding to external stimuli, such as ultrasound, thermal, light and magnetic field, as well as internal stimuli, including pH, redox potential, hypoxia and enzyme, etc. Nanocarriers could respond to stimuli in tumor microenvironments or inside cancer cells for on-demanded drug delivery and accumulation, controlled drug release, activation of bioactive compounds, probes and targeting ligands, as well as size, charge and conformation conversion, etc., leading to sensing and signaling, overcoming multidrug resistance, accurate diagnosis and precision therapy. This review has summarized the general strategies of developing stimuli-responsive nanocarriers and recent advances, presented their applications in drug delivery, tumor imaging, therapy and theranostics, illustrated the progress of clinical translation and made prospects.
Collapse
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, 610041, China
| |
Collapse
|
16
|
Peng Y, Bariwal J, Kumar V, Tan C, Mahato RI. Organic Nanocarriers for Delivery and Targeting of Therapeutic Agents for Cancer Treatment. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Peng
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Jitender Bariwal
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Virender Kumar
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug DeliveryUniversity of Mississippi University MS 38677 USA
| | - Ram I. Mahato
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| |
Collapse
|
17
|
Rasines Mazo A, Allison-Logan S, Karimi F, Chan NJA, Qiu W, Duan W, O’Brien-Simpson NM, Qiao GG. Ring opening polymerization of α-amino acids: advances in synthesis, architecture and applications of polypeptides and their hybrids. Chem Soc Rev 2020; 49:4737-4834. [DOI: 10.1039/c9cs00738e] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This review provides a comprehensive overview of the latest advances in the synthesis, architectural design and biomedical applications of polypeptides and their hybrids.
Collapse
Affiliation(s)
- Alicia Rasines Mazo
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Stephanie Allison-Logan
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Fatemeh Karimi
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Nicholas Jun-An Chan
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Wenlian Qiu
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Wei Duan
- School of Medicine
- Deakin University
- Geelong
- Australia
| | - Neil M. O’Brien-Simpson
- Centre for Oral Health Research
- Melbourne Dental School and the Bio21 Institute of Molecular Science and Biotechnology
- University of Melbourne
- Parkville
- Australia
| | - Greg G. Qiao
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| |
Collapse
|
18
|
Weng Y, Huang Q, Li C, Yang Y, Wang X, Yu J, Huang Y, Liang XJ. Improved Nucleic Acid Therapy with Advanced Nanoscale Biotechnology. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 19:581-601. [PMID: 31927331 PMCID: PMC6957827 DOI: 10.1016/j.omtn.2019.12.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/23/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022]
Abstract
Due to a series of systemic and intracellular obstacles in nucleic acid (NA) therapy, including fast degradation in blood, renal clearance, poor cellular uptake, and inefficient endosomal escape, NAs may need delivery methods to transport to the cell nucleus or cytosol to be effective. Advanced nanoscale biotechnology-associated strategies, such as controlling the particle size, charge, drug loading, response to environmental signals, or other physical/chemical properties of delivery carriers, have provided great help for the in vivo and in vitro delivery of NA therapeutics. In this review, we introduce the characteristics of different NA modalities and illustrate how advanced nanoscale biotechnology assists NA therapy. The specific features and challenges of various nanocarriers in clinical and preclinical studies are summarized and discussed. With the help of advanced nanoscale biotechnology, some of the major barriers to the development of NA therapy will eventually be overcome in the near future.
Collapse
Affiliation(s)
- Yuhua Weng
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Institute of Engineering Medicine, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Qianqian Huang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Chunhui Li
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Institute of Engineering Medicine, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Yongfeng Yang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Xiaoxia Wang
- Institute of Molecular Medicine, Peking University, Beijing 100871, P.R. China
| | - Jie Yu
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Institute of Engineering Medicine, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P.R. China.
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China.
| |
Collapse
|
19
|
Bioreducible crosslinked cationic nanopolyplexes from clickable polyethylenimines enabling robust cancer gene therapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 24:102144. [PMID: 31838150 DOI: 10.1016/j.nano.2019.102144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/13/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
Bioreducible crosslinked polyplexes from branched polyethylenimine (BPEI, 10 kDa) were successfully constructed through DNA neutralization by disulfide-linked azidated BPEI (PAZ) and subsequent DNA condensation by azadibenzocyclooctyne-modified BPEI (PDB), following their self-crosslinking via azide-azadibenzocyclooctyne click chemistry. Click-crosslinked cationic polyplexes (c-polyplexes) revealed high extracellular colloidal stability against negative heparin and ions while intracellular bioreducible degradability for efficient gene unpacking. In vitro gene transfection in cancer cells indicated that the c-polyplexes produced markedly higher transfection efficiency than non-crosslinked counterparts in the serum. The c-polyplexes also had prolonged circulation kinetics, elevated gene accumulation level in SKOV-3 tumor xenografted in a mouse model and in turn superior transgene expression in the tumor. By small hairpin RNA for VEGF silencing, the c-polyplexes exerted significant tumor growth inhibition following with low systemic toxicity in the mouse. This study highlights the design of clickable polycations to construct crosslinked cationic nanopolyplexes for intravenous gene delivery against cancer.
Collapse
|
20
|
Kim B, Park JH, Sailor MJ. Rekindling RNAi Therapy: Materials Design Requirements for In Vivo siRNA Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1903637. [PMID: 31566258 PMCID: PMC6891135 DOI: 10.1002/adma.201903637] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/12/2019] [Indexed: 05/07/2023]
Abstract
With the recent FDA approval of the first siRNA-derived therapeutic, RNA interference (RNAi)-mediated gene therapy is undergoing a transition from research to the clinical space. The primary obstacle to realization of RNAi therapy has been the delivery of oligonucleotide payloads. Therefore, the main aims is to identify and describe key design features needed for nanoscale vehicles to achieve effective delivery of siRNA-mediated gene silencing agents in vivo. The problem is broken into three elements: 1) protection of siRNA from degradation and clearance; 2) selective homing to target cell types; and 3) cytoplasmic release of the siRNA payload by escaping or bypassing endocytic uptake. The in vitro and in vivo gene silencing efficiency values that have been reported in publications over the past decade are quantitatively summarized by material type (lipid, polymer, metal, mesoporous silica, and porous silicon), and the overall trends in research publication and in clinical translation are discussed to reflect on the direction of the RNAi therapeutics field.
Collapse
Affiliation(s)
- Byungji Kim
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Michael J Sailor
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| |
Collapse
|
21
|
Takemoto H, Wang CL, Nomoto T, Matsui M, Tomoda K, Nishiyama N. Pyruvate Responsiveness Based on α-Oxohydrazone Formation for Intracellular siRNA Release from Polyion Complex-Based Carriers. Biomacromolecules 2019; 20:2305-2314. [DOI: 10.1021/acs.biomac.9b00261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Hiroyasu Takemoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Chih-Ling Wang
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Takahiro Nomoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Makoto Matsui
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Keishiro Tomoda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| |
Collapse
|
22
|
Deirram N, Zhang C, Kermaniyan SS, Johnston APR, Such GK. pH-Responsive Polymer Nanoparticles for Drug Delivery. Macromol Rapid Commun 2019; 40:e1800917. [PMID: 30835923 DOI: 10.1002/marc.201800917] [Citation(s) in RCA: 286] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/31/2019] [Indexed: 01/06/2025]
Abstract
Stimuli-responsive nanoparticles have the potential to improve the delivery of therapeutics to a specific cell or region within the body. There are many stimuli that have shown potential for specific release of cargo, including variation of pH, redox potential, or the presence of enzymes. pH variation has generated significant interest for the synthesis of stimuli-responsive nanoparticles because nanoparticles are internalized into cells via vesicles that are acidified. Additionally, the tumor microenvironment is known to have a lower pH than the surrounding tissue. In this review, different strategies to design pH-responsive nanoparticles are discussed, focusing on the use of charge-shifting polymers, acid labile linkages, and crosslinking.
Collapse
Affiliation(s)
- Nayeleh Deirram
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Changhe Zhang
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Sarah S Kermaniyan
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Georgina K Such
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
23
|
Kim BS, Chuanoi S, Suma T, Anraku Y, Hayashi K, Naito M, Kim HJ, Kwon IC, Miyata K, Kishimura A, Kataoka K. Self-Assembly of siRNA/PEG-b-Catiomer at Integer Molar Ratio into 100 nm-Sized Vesicular Polyion Complexes (siRNAsomes) for RNAi and Codelivery of Cargo Macromolecules. J Am Chem Soc 2019; 141:3699-3709. [DOI: 10.1021/jacs.8b13641] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Beob Soo Kim
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 136-791, Republic of Korea
| | - Sayan Chuanoi
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tomoya Suma
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yasutaka Anraku
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kotaro Hayashi
- Innovation Center of NanoMedicne, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Mitsuru Naito
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hyun Jin Kim
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 136-791, Republic of Korea
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Akihiro Kishimura
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
- Center for Molecular Systems, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicne, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Policy Alternatives Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
24
|
Taguchi K, Lu H, Jiang Y, Hung TT, Stenzel MH. Safety of nanoparticles based on albumin-polymer conjugates as a carrier of nucleotides for pancreatic cancer therapy. J Mater Chem B 2018; 6:6278-6287. [PMID: 32254618 DOI: 10.1039/c8tb01613e] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Gene therapy through systemic administration is expected to offer significant therapeutic potential against intractable cancers, including pancreatic cancer. One of the requirements for in vivo gene therapy is the development of a gene carrier with a high level of safety, transfection ability and tumour accumulation. Bovine serum albumin (BSA)-poly(2-dimethylaminoethyl methacrylate) (PDMAEMA) conjugation (BSA-PDMAEMA conjugation) could result in the development of a promising gene carrier. This conjugate could preserve the BSA structure well and efficiently condense the nucleotide inside, resulting in the formation of BSA-PDMAEMA nanoparticles that have a polyion complex core and surrounding BSA corona with a size of <100 nm. The nanoparticles that were produced based on BSA-PDMAEMA conjugation possessed good characteristics for use as a gene carrier with good biocompatibility, appropriate blood retention and gene protective properties. Furthermore, the in vivo two-dimensional and three-dimensional biodistribution in a xenograft pancreatic cancer (AsPC-1) model in mice clearly showed that BSA-PDMAEMA nanoparticles accumulated at the tumour site via enhanced permeability and the retention effect. Furthermore, BSA-PDMAEMA nanoparticles, which condensed the active anti-cancer oligonucleotide, ISIS5132, inhibited the growth of cancer in AsPC-1-bearing mice compared to mice which were administered with ISIS5132 alone. The structure of the BSA-PDMAEMA nanoparticles, i.e. the polyion complex core with the BSA corona, would comprehensively contribute to these ideal characteristics for use as a gene carrier. In conclusion, BSA-PDMAEMA nanoparticles could exert a therapeutic effect on intractable pancreatic cancer in vivo, indicating their use as a promising gene carrier.
Collapse
Affiliation(s)
- Kazuaki Taguchi
- Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | | | | | |
Collapse
|
25
|
Cabral H, Miyata K, Osada K, Kataoka K. Block Copolymer Micelles in Nanomedicine Applications. Chem Rev 2018; 118:6844-6892. [PMID: 29957926 DOI: 10.1021/acs.chemrev.8b00199] [Citation(s) in RCA: 834] [Impact Index Per Article: 119.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymeric micelles are demonstrating high potential as nanomedicines capable of controlling the distribution and function of loaded bioactive agents in the body, effectively overcoming biological barriers, and various formulations are engaged in intensive preclinical and clinical testing. This Review focuses on polymeric micelles assembled through multimolecular interactions between block copolymers and the loaded drugs, proteins, or nucleic acids as translationable nanomedicines. The aspects involved in the design of successful micellar carriers are described in detail on the basis of the type of polymer/payload interaction, as well as the interplay of micelles with the biological interface, emphasizing on the chemistry and engineering of the block copolymers. By shaping these features, polymeric micelles have been propitious for delivering a wide range of therapeutics through effective sensing of targets in the body and adjustment of their properties in response to particular stimuli, modulating the activity of the loaded drugs at the targeted sites, even at the subcellular level. Finally, the future perspectives and imminent challenges for polymeric micelles as nanomedicines are discussed, anticipating to spur further innovations.
Collapse
Affiliation(s)
| | | | | | - Kazunori Kataoka
- Innovation Center of NanoMedicine , Kawasaki Institute of Industrial Promotion , 3-25-14, Tonomachi , Kawasaki-ku , Kawasaki 210-0821 , Japan.,Policy Alternatives Research Institute , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku , Tokyo 113-0033 , Japan
| |
Collapse
|
26
|
Kim HJ, Yi Y, Kim A, Miyata K. Small Delivery Vehicles of siRNA for Enhanced Cancer Targeting. Biomacromolecules 2018; 19:2377-2390. [PMID: 29864287 DOI: 10.1021/acs.biomac.8b00546] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Small interfering RNA (siRNA) drugs have been considered to treat various diseases in major organs. However, siRNA drugs developed for cancer therapy are hindered from proceeding to the clinic. To date, various delivery formulations have been developed from cationic lipids, polymers, and/or inorganic nanoparticles for systemic siRNA delivery to solid tumors. Most of these delivery vehicles do not generate small particle sizes and pharmacokinetics required for accumulation in target cancer cells compared with clinically tested anticancer drug-loaded polymeric micelles. This review describes the significance of small, long-circulating vehicles for efficient delivery of siRNA to cancer tissues via the enhanced permeability and retention (EPR) effect. We summarize recent biological evidence that supports the size effect of delivery vehicles in tumor microenvironments and introduce promising strategies for the construction of small vehicles with sizes of 10-50 nm. We then discuss the feasibility of these delivery vehicles with respect to translation to clinical trials.
Collapse
Affiliation(s)
- Hyun Jin Kim
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| | - Yu Yi
- Department of Materials Engineering, Graduate School of Engineering , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-8656 , Japan.,CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety , National Center for Nanosciecne and Technology , No. 11 Beiyitiao , Zhongguancun, Beijing 100190 , China
| | - Ahram Kim
- Department of Materials Science, Graduate School of Pure and Applied Sciences , University of Tsukuba , 1-1-1 Tennoudai , Tsukuba , Ibaraki 305-8573 , 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
| |
Collapse
|
27
|
pH-responsive Micelles from a Blend of PEG-b-PLA and PLA-b-PDPA Block Copolymers: Core Protection Against Enzymatic Degradation. CHINESE JOURNAL OF POLYMER SCIENCE 2018. [DOI: 10.1007/s10118-018-2149-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
28
|
Cai C, Lin J, Lu Y, Zhang Q, Wang L. Polypeptide self-assemblies: nanostructures and bioapplications. Chem Soc Rev 2018; 45:5985-6012. [PMID: 27722321 DOI: 10.1039/c6cs00013d] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Polypeptide copolymers can self-assemble into diverse aggregates. The morphology and structure of aggregates can be varied by changing molecular architectures, self-assembling conditions, and introducing secondary components such as polymers and nanoparticles. Polypeptide self-assemblies have gained significant attention because of their potential applications as delivery vehicles for therapeutic payloads and as additives in the biomimetic mineralization of inorganics. This review article provides an overview of recent advances in nanostructures and bioapplications related to polypeptide self-assemblies. We highlight recent contributions to developing strategies for the construction of polypeptide assemblies with increasing complexity and novel functionality that are suitable for bioapplications. The relationship between the structure and properties of the polypeptide aggregates is emphasized. Finally, we briefly outline our perspectives and discuss the challenges in the field.
Collapse
Affiliation(s)
- Chunhua Cai
- Shanghai Key Laboratory of Advanced Polymeric Materials, State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Jiaping Lin
- Shanghai Key Laboratory of Advanced Polymeric Materials, State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Yingqing Lu
- Shanghai Key Laboratory of Advanced Polymeric Materials, State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Qian Zhang
- Shanghai Key Laboratory of Advanced Polymeric Materials, State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Liquan Wang
- Shanghai Key Laboratory of Advanced Polymeric Materials, State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
29
|
Rinaldi C, Wood MJA. Antisense oligonucleotides: the next frontier for treatment of neurological disorders. Nat Rev Neurol 2017; 14:9-21. [PMID: 29192260 DOI: 10.1038/nrneurol.2017.148] [Citation(s) in RCA: 537] [Impact Index Per Article: 67.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antisense oligonucleotides (ASOs) were first discovered to influence RNA processing and modulate protein expression over two decades ago; however, progress translating these agents into the clinic has been hampered by inadequate target engagement, insufficient biological activity, and off-target toxic effects. Over the years, novel chemical modifications of ASOs have been employed to address these issues. These modifications, in combination with elucidation of the mechanism of action of ASOs and improved clinical trial design, have provided momentum for the translation of ASO-based strategies into therapies. Many neurological conditions lack an effective treatment; however, as research progressively disentangles the pathogenic mechanisms of these diseases, they provide an ideal platform to test ASO-based strategies. This steady progress reached a pinnacle in the past few years with approvals of ASOs for the treatment of spinal muscular atrophy and Duchenne muscular dystrophy, which represent landmarks in a field in which disease-modifying therapies were virtually non-existent. With the rapid development of improved next-generation ASOs toward clinical application, this technology now holds the potential to have a dramatic effect on the treatment of many neurological conditions in the near future.
Collapse
Affiliation(s)
- Carlo Rinaldi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford OX1 3QX, UK
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford OX1 3QX, UK
| |
Collapse
|
30
|
Song Z, Han Z, Lv S, Chen C, Chen L, Yin L, Cheng J. Synthetic polypeptides: from polymer design to supramolecular assembly and biomedical application. Chem Soc Rev 2017; 46:6570-6599. [PMID: 28944387 DOI: 10.1039/c7cs00460e] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Synthetic polypeptides from the ring-opening polymerization of N-carboxyanhydrides (NCAs) are one of the most important biomaterials. The unique features of these synthetic polypeptides, including their chemical diversity of side chains and their ability to form secondary structures, enable their broad applications in the field of gene delivery, drug delivery, bio-imaging, tissue engineering, and antimicrobials. In this review article, we summarize the recent advances in the design of polypeptide-based supramolecular structures, including complexes with nucleic acids, micelles, vesicles, hybrid nanoparticles, and hydrogels. We also highlight the progress in the chemical design of functional polypeptides, which plays a crucial role to manipulate their assembly behaviours and optimize their biomedical performances. Finally, we conclude the review by discussing the future opportunities in this field, including further studies on the secondary structures and cost-effective synthesis of polypeptide materials.
Collapse
Affiliation(s)
- Ziyuan Song
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Functional polymer-based siRNA delivery carrier that recognizes site-specific biosignals. J Control Release 2017; 267:90-99. [PMID: 28923764 DOI: 10.1016/j.jconrel.2017.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/24/2017] [Accepted: 09/01/2017] [Indexed: 02/07/2023]
Abstract
Responsive molecular designs to specific biosignals in microenvironments endow site-specific functionalities with associated polymers. Thus, the construction of small interfering RNA (siRNA) carriers with functional polymers enables smart programs that are triggered by sequential biosignals in a pathway to the targeted cytosol for effective gene silencing. In this review, we explain rational strategies for the design of functional polymers with responsiveness to biosignals and describe the examples of smart carriers for siRNA delivery.
Collapse
|
32
|
Amreddy N, Babu A, Muralidharan R, Munshi A, Ramesh R. Polymeric Nanoparticle-Mediated Gene Delivery for Lung Cancer Treatment. Top Curr Chem (Cham) 2017; 375:35. [PMID: 28290155 PMCID: PMC5480422 DOI: 10.1007/s41061-017-0128-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/22/2017] [Indexed: 11/28/2022]
Abstract
In recent years, researchers have focused on targeted gene therapy for lung cancer, using nanoparticle carriers to overcome the limitations of conventional treatment methods. The main goal of targeted gene therapy is to develop more efficient therapeutic strategies by improving the bioavailability, stability, and target specificity of gene therapeutics and to reduce off-target effects. Polymer-based nanoparticles, an alternative to lipid and inorganic nanoparticles, efficiently carry nucleic acid therapeutics and are stable in vivo. Receptor-targeted delivery is a promising approach that can limit non-specific gene delivery and can be achieved by modifying the polymer nanoparticle surface with specific receptor ligands or antibodies. This review highlights the recent developments in gene delivery using synthetic and natural polymer-based nucleic acid carriers for lung cancer treatment. Various nanoparticle systems based on polymers and polymer combinations are discussed. Further, examples of targeting ligands or moieties used in targeted, polymer-based gene delivery to lung cancer are reviewed.
Collapse
Affiliation(s)
- Narsireddy Amreddy
- Department of Pathology, Stanton L. Young Biomedical Research Center, University of Oklahoma Health Sciences Center, Suite 1403, 975 N.E., 10th Street, Oklahoma City, OK, 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anish Babu
- Department of Pathology, Stanton L. Young Biomedical Research Center, University of Oklahoma Health Sciences Center, Suite 1403, 975 N.E., 10th Street, Oklahoma City, OK, 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ranganayaki Muralidharan
- Department of Pathology, Stanton L. Young Biomedical Research Center, University of Oklahoma Health Sciences Center, Suite 1403, 975 N.E., 10th Street, Oklahoma City, OK, 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anupama Munshi
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rajagopal Ramesh
- Department of Pathology, Stanton L. Young Biomedical Research Center, University of Oklahoma Health Sciences Center, Suite 1403, 975 N.E., 10th Street, Oklahoma City, OK, 73104, USA.
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
33
|
Cai X, Zhu H, Zhang Y, Gu Z. Highly Efficient and Safe Delivery of VEGF siRNA by Bioreducible Fluorinated Peptide Dendrimers for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:9402-9415. [PMID: 28228013 DOI: 10.1021/acsami.6b16689] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
RNA interference (RNAi) has a great promise in treating various acquired and hereditary diseases. However, it remains highly desirable to develop new delivery system to circumvent complex extra- and intracellular barriers for successful clinical translation. Here, we report on a versatile polymeric vector, bioreducible fluorinated peptide dendrimers (BFPD), for efficient and safe small interfering RNA (siRNA) delivery. In virtue of skillfully integrating all of the unique advantages of reversible cross-linking, fluorination, and peptide dendrimers, this novel vector can surmount almost all extra- and intracellular barriers associated with local siRNA delivery through highly improved physiological stability and serum resistance, significantly increased intratumoral enrichment, cellular internalization, successful facilitation of endosomal escape, and cytosolic siRNA release. BFPD polyplexes, carrying small interfering vascular endothelial growth factor (siVEGF), demonstrated excellent VEGF silencing efficacy (∼65%) and a strong capability for inhibiting HeLa cell proliferation. More importantly, these polyplexes showed superior performance in long-term enrichment in the tumor sites and had a high level of tumor growth inhibition. Furthermore, these polyplexes not only exhibited excellent in vivo antitumor efficacy but also demonstrated superior biocompatibility, compared with LPF2000, both in vivo and in vitro. These findings indicate that BFPD is an efficient and safe siRNA delivery system and has remarkable potential for RNAi-based cancer treatment.
Collapse
Affiliation(s)
- Xiaojun Cai
- College of Materials Science and Engineering, Nanjing Tech University , 30 South Puzhu Road, Nanjing 211816, P. R. China
| | - Haofang Zhu
- College of Materials Science and Engineering, Nanjing Tech University , 30 South Puzhu Road, Nanjing 211816, P. R. China
| | - Yanmei Zhang
- College of Materials Science and Engineering, Nanjing Tech University , 30 South Puzhu Road, Nanjing 211816, P. R. China
| | - Zhongwei Gu
- College of Materials Science and Engineering, Nanjing Tech University , 30 South Puzhu Road, Nanjing 211816, P. R. China
| |
Collapse
|
34
|
Yousefpour Marzbali M, Yari Khosroushahi A. Polymeric micelles as mighty nanocarriers for cancer gene therapy: a review. Cancer Chemother Pharmacol 2017; 79:637-649. [DOI: 10.1007/s00280-017-3273-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 03/02/2017] [Indexed: 12/11/2022]
|
35
|
Li Y, Gao J, Zhang C, Cao Z, Cheng D, Liu J, Shuai X. Stimuli-Responsive Polymeric Nanocarriers for Efficient Gene Delivery. Top Curr Chem (Cham) 2017; 375:27. [DOI: 10.1007/s41061-017-0119-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/31/2017] [Indexed: 11/25/2022]
|
36
|
Liu Y, Winter HH, Perry SL. Linear viscoelasticity of complex coacervates. Adv Colloid Interface Sci 2017; 239:46-60. [PMID: 27633928 DOI: 10.1016/j.cis.2016.08.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 01/15/2023]
Abstract
Rheology is a powerful method for material characterization that can provide detailed information about the self-assembly, structure, and intermolecular interactions present in a material. Here, we review the use of linear viscoelastic measurements for the rheological characterization of complex coacervate-based materials. Complex coacervation is an electrostatically and entropically-driven associative liquid-liquid phase separation phenomenon that can result in the formation of bulk liquid phases, or the self-assembly of hierarchical, microphase separated materials. We discuss the need to link thermodynamic studies of coacervation phase behavior with characterization of material dynamics, and provide parallel examples of how parameters such as charge stoichiometry, ionic strength, and polymer chain length impact self-assembly and material dynamics. We conclude by highlighting key areas of need in the field, and specifically call for the development of a mechanistic understanding of how molecular-level interactions in complex coacervate-based materials affect both self-assembly and material dynamics.
Collapse
Affiliation(s)
- Yalin Liu
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - H Henning Winter
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Sarah L Perry
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
37
|
Yu X, Hou J, Shi Y, Su C, Zhao L. Preparation and characterization of novel chitosan-protamine nanoparticles for nucleus-targeted anticancer drug delivery. Int J Nanomedicine 2016; 11:6035-6046. [PMID: 27881917 PMCID: PMC5115688 DOI: 10.2147/ijn.s117066] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
It is well known that most anticancer drugs commonly show high toxicity to the DNA of tumor cells and exert effects by combining with the DNA or associated enzymes in the nucleus. Most developed drugs are first delivered into the cytoplasm and then transferred to the nucleus through the membrane pores. Sometimes, the transportation of drugs from cytoplasm to nucleus is not efficient and often results in poor therapeutic effects. In this study, we developed special and novel nanoparticles (NPs) made of chitosan and protamine for targeted nuclear capture of drugs to enhance anticancer effects. The anticancer effects of nuclear targeted-delivery of drugs in NPs were also evaluated by investigating cytotoxicity, cellular uptake mechanism, and cell apoptosis on cells. Chitosan–protamine NPs were characterized by good drug entrapment, sustained release, small average particle size, low polydispersity index, and high encapsulation efficiency; and accomplished the efficient nuclear delivery of fluorouracil (5-Fu). Compared with free 5-Fu and 5-Fu-loaded chitosan NPs, treatment of A549 cells and HeLa cells with 5-Fu-loaded chitosan–protamine NPs showed the highest cytotoxicity and further induced the significant apoptosis of cells. In addition, 5-Fu-loaded chitosan–protamine NPs exhibited the best efficiency in inhibiting tumor growth than the other three formulations. 5-Fu-loaded chitosan–protamine NPs enhanced antitumor efficacy through the targeted nuclear capture of drugs and showed promising potential as a nanodelivery system for quickly locating drugs in the nucleus of cells.
Collapse
Affiliation(s)
| | | | | | - Chang Su
- School of Veterinary Medicine, Jinzhou Medical University, Jinzhou, People's Republic of China
| | | |
Collapse
|
38
|
Blocher WC, Perry SL. Complex coacervate-based materials for biomedicine. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [DOI: 10.1002/wnan.1442] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/10/2016] [Accepted: 10/02/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Whitney C. Blocher
- Department of Chemical Engineering; University of Massachusetts Amherst; Amherst MA USA
| | - Sarah L. Perry
- Department of Chemical Engineering; University of Massachusetts Amherst; Amherst MA USA
| |
Collapse
|
39
|
Dimde M, Steinhilber D, Neumann F, Li Y, Paulus F, Ma N, Haag R. Synthesis of pH-Cleavable dPG-Amines for Gene Delivery Application. Macromol Biosci 2016; 17. [PMID: 27430195 DOI: 10.1002/mabi.201600190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 06/16/2016] [Indexed: 01/05/2023]
Abstract
The development of effective nonviral vectors for gene therapy is still a challenge in research, due to the high toxicity of many existing polycationic nanocarriers. In this paper, the development of two pH-cleavable polyglycerol-amine-based nanocarriers is described. The benz-acetal bond represents the pH-sensitive cleavage site between dendritic polyglycerol (dPG) and glycerol-based 1,2-diamines that can complex genetic material. Due to the acid lability of the acetal moiety, the cleavable dPG-amines are less toxic in vitro. Cell-mediated degradation results in non-toxic dPG with low amine functionalization and low molecular weight cleavage products (cp). The genetic material is released because of the loss of multivalent amine groups. Interestingly, the release kinetics at the endosomal pH could be controlled by simple chemical modification of the acetals. In vitro experiments demonstrate the ability of the cleavable dPG-amine to transfect HeLa cells with GFP-DNA, which resulted in cell-compatible cleavage products.
Collapse
Affiliation(s)
- Mathias Dimde
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, Berlin, 14195, Germany
| | - Dirk Steinhilber
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, Berlin, 14195, Germany
| | - Falko Neumann
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, Berlin, 14195, Germany
| | - Yan Li
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, Berlin, 14195, Germany
| | - Florian Paulus
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, Berlin, 14195, Germany
| | - Nan Ma
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, Berlin, 14195, Germany.,Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies Helmholtz-Zentrum Geesthacht, Kantstrasse 55, Teltow, 14513, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, Berlin, 14195, Germany
| |
Collapse
|
40
|
Wu J, Zhang J, Deng C, Meng F, Zhong Z. Vitamin E-Oligo(methyl diglycol l-glutamate) as a Biocompatible and Functional Surfactant for Facile Preparation of Active Tumor-Targeting PLGA Nanoparticles. Biomacromolecules 2016; 17:2367-74. [PMID: 27305935 DOI: 10.1021/acs.biomac.6b00380] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Poly(d,l-lactide-co-glycolide) (PLGA) nanoparticles have attracted an enormous interest for controlled drug delivery. Their clinical applications are, however, partly hindered by lack of biocompatible, biodegradable and functional surfactants. Here, we designed and developed a novel biocompatible surfactant based on amphiphilic vitamin E-oligo(methyl diglycol l-glutamate) (VEOEG) for facile fabrication of robust and tumor-targeting PLGA-based nanomedicines. VEOEG was prepared with controlled Mn of 1.7-2.6 kg/mol and low molecular weight distribution (Đ = 1.04-1.16) via polymerization of methyl diglycol l-glutamate N-carboxyanhydride using vitamin E-ethylenediamine derivative (VE-NH2) as an initiator. VEOEG had a hydrophile-lipophile balance data of 13.8-16.1 and critical micellar concentration of 189.3-203.8 mg/L depending on lengths of oligopeptide. Using VEOEG as a surfactant, PLGA nanoparticles could be obtained via nanoprecipitation method with a small and uniform hydrodynamic size of 135 nm and positive surface charge of +26.6 mV, in accordance with presence of amino groups at the surface. The resulting PLGA nanoparticles could be readily coated with hyaluronic acid (HA) to form highly stable, small-sized (143 nm), monodisperse, and negatively charged nanoparticles (HA-PLGA NPs). Notably, paclitaxel-loaded HA-PLGA NPs (PTX-HA-PLGA NPs) exhibited better antitumor effects in CD44-positive MCF-7 breast tumor cells than Taxol (a clinical paclitaxel formulation). The in vivo pharmacokinetics assay in nude mice displayed that PTX-HA-PLGA NPs possessed a long plasma half-life of 3.14 h. The in vivo biodistribution studies revealed that PTX-HA-PLGA NPs had a high tumor PTX level of 8.4% ID/g, about 6 times better than that of Taxol. Interestingly, therapeutic studies showed that PTX-HA-PLGA NPs caused significantly more effective tumor growth inhibition, better survival rate and lower adverse effect than Taxol. VEOEG has emerged as a versatile and functional surfactant for the fabrication of advanced anticancer nanomedicines.
Collapse
Affiliation(s)
- Jintian Wu
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Jian Zhang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Chao Deng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| |
Collapse
|
41
|
Florinas S, Liu M, Fleming R, Van Vlerken-Ysla L, Ayriss J, Gilbreth R, Dimasi N, Gao C, Wu H, Xu ZQ, Chen S, Dirisala A, Kataoka K, Cabral H, Christie RJ. A Nanoparticle Platform To Evaluate Bioconjugation and Receptor-Mediated Cell Uptake Using Cross-Linked Polyion Complex Micelles Bearing Antibody Fragments. Biomacromolecules 2016; 17:1818-33. [PMID: 27007881 DOI: 10.1021/acs.biomac.6b00239] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Targeted nanomedicines are a promising technology for treatment of disease; however, preparation and characterization of well-defined protein-nanoparticle systems remain challenging. Here, we describe a platform technology to prepare antibody binding fragment (Fab)-bearing nanoparticles and an accompanying real-time cell-based assay to determine their cellular uptake compared to monoclonal antibodies (mAbs) and Fabs. The nanoparticle platform was composed of core-cross-linked polyion complex (PIC) micelles prepared from azide-functionalized PEG-b-poly(amino acids), that is, azido-PEG-b-poly(l-lysine) [N3-PEG-b-PLL] and azido-PEG-b-poly(aspartic acid) [N3-PEG-b-PAsp]. These PIC micelles were 30 nm in size and contained approximately 10 polymers per construct. Fabs were derived from an antibody binding the EphA2 receptor expressed on cancer cells and further engineered to contain a reactive cysteine for site-specific attachment and a cleavable His tag for purification from cell culture expression systems. Azide-functionalized micelles and thiol-containing Fab were linked using a heterobifunctional cross-linker (FPM-PEG4-DBCO) that contained a fluorophenyl-maleimide for stable conjugation to Fabs thiols and a strained alkyne (DBCO) group for coupling to micelle azide groups. Analysis of Fab-PIC micelle conjugates by fluorescence correlation spectroscopy, size exclusion chromatography, and UV-vis absorbance determined that each nanoparticle contained 2-3 Fabs. Evaluation of cellular uptake in receptor positive cancer cells by real-time fluorescence microscopy revealed that targeted Fab-PIC micelles achieved higher cell uptake than mAbs and Fabs, demonstrating the utility of this approach to identify targeted nanoparticle constructs with unique cellular internalization properties.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ze-Qi Xu
- SynChem, Inc., Elk Grove Village, Illinois 60007, United States
| | | | | | - Kazunori Kataoka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo , 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.,The Innovation Center of Nanomedicine, 66-20 Horikawa-cho, Saiwai-ku, Kawasaki 212-0013, Japan
| | | | | |
Collapse
|
42
|
Affiliation(s)
- Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo
| |
Collapse
|