51
|
Müller K, Kessel E, Klein PM, Höhn M, Wagner E. Post-PEGylation of siRNA Lipo-oligoamino Amide Polyplexes Using Tetra-glutamylated Folic Acid as Ligand for Receptor-Targeted Delivery. Mol Pharm 2016; 13:2332-45. [DOI: 10.1021/acs.molpharmaceut.6b00102] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Katharina Müller
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Eva Kessel
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
- Nanosystems Initiative Munich, Schellingstrasse 4, D-80799 Munich, Germany
| | - Philipp M. Klein
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
- Nanosystems Initiative Munich, Schellingstrasse 4, D-80799 Munich, Germany
| |
Collapse
|
52
|
Weber J, Lächelt U, Wagner E. Multifunctional Oligoaminoamides for the Receptor-Specific Delivery of Therapeutic RNA. Methods Mol Biol 2016. [PMID: 26202283 DOI: 10.1007/978-1-4939-2806-4_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Drugs with novel and versatile modes of action, such as therapeutic nucleic acids or proteins, open new possibilities for the precise therapy of different diseases. The most crucial limitation during the development of a therapeutic drug remains the safe and efficient intracellular delivery.To overcome the hurdles and to realize the successful delivery of such new biopharmaceuticals, our laboratory has recently developed a sequence-defined, cationic oligomer platform based on solid-phase synthesis. These multifunctional oligomers have displayed efficient delivery of therapeutic RNA in vitro and in vivo. In this chapter, we provide a brief background on the special features and applications of these carrier systems as well as detailed protocols for the oligomer and polyplex synthesis and their evaluation.
Collapse
Affiliation(s)
- Judith Weber
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | |
Collapse
|
53
|
Roberts TC, Ezzat K, El Andaloussi S, Weinberg MS. Synthetic SiRNA Delivery: Progress and Prospects. Methods Mol Biol 2016; 1364:291-310. [PMID: 26472459 DOI: 10.1007/978-1-4939-3112-5_23] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Small interfering RNA (siRNA) is a powerful tool for modulating gene expression by RNA interference (RNAi). Duplex RNA oligonucleotides induce cleavage of homologous target transcripts, thereby enabling posttranscriptional silencing of potentially any gene. As such, siRNAs may have utility as novel pharmaceuticals for a wide range of diseases. However, a lack of "drug-likeness," physiological barriers, and potential toxicities have meant that systemic delivery of SiRNAs in vivo remains a major challenge. Here we discuss various strategies that have been employed to solve the problem of SiRNA delivery. These include chemical modification of the SiRNA, direct conjugation to bioactive moieties, and nanoparticle formulations.
Collapse
Affiliation(s)
- Thomas C Roberts
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Kariem Ezzat
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Samir El Andaloussi
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Marc S Weinberg
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
- Antiviral Gene Therapy Research Unit, Department of Molecular Medicine and Haematology, University of the Witwatersrand Medical School, Johannesburg, WITS 2050, South Africa.
- HIV Pathogenesis Research Unit, Department of Molecular Medicine and Haematology, University of the Witwatersrand Medical School, Johannesburg, WITS 2050, South Africa.
| |
Collapse
|
54
|
Hou KK, Pan H, Schlesinger PH, Wickline SA. A role for peptides in overcoming endosomal entrapment in siRNA delivery - A focus on melittin. Biotechnol Adv 2015; 33:931-40. [PMID: 26025036 PMCID: PMC4540690 DOI: 10.1016/j.biotechadv.2015.05.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/20/2015] [Accepted: 05/23/2015] [Indexed: 12/21/2022]
Abstract
siRNA has the possibility to revolutionize medicine by enabling highly specific and efficient silencing of proteins involved in disease pathogenesis. Despite nearly 20 years of research dedicated to translating siRNA from a research tool into a clinically relevant therapeutic, minimal success has been had to date. Access to RNA interference machinery located in the cytoplasm is often overlooked, but must be considered when designing the next generation of siRNA delivery strategies. Peptide transduction domains (PTDs) have demonstrated moderate siRNA transfection, which is primarily limited by endosomal entrapment. Strategies aimed at overcoming endosomal entrapment associated with peptide vectors are reviewed here, including osmotic methods, lipid conjugation, and fusogenic peptides. As an alternative to traditional PTD, the hemolytic peptide melittin exhibits the native capacity for endosomal disruption but causes cytotoxicity. However, appropriate packaging and protection of melittin with activation and release in the endosomal compartment has allowed melittin-based strategies to demonstrate both in vitro and in vivo safety and efficacy. These data suggest that melittin's membrane disruptive properties can enable safe and effective endosomolysis, building a case for melittin as a key component in a new generation of siRNA therapeutics.
Collapse
Affiliation(s)
- Kirk K Hou
- Computational and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Hua Pan
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63108, USA
| | - Paul H Schlesinger
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Samuel A Wickline
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63108, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63108, USA; Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO 63108, USA.
| |
Collapse
|
55
|
Abstract
In this article, advances in designing polymeric nanoparticles for targeted cancer gene therapy are reviewed. Characterization and evaluation of biomaterials, targeting ligands, and transcriptional elements are each discussed. Advances in biomaterials have driven improvements to nanoparticle stability and tissue targeting, conjugation of ligands to the surface of polymeric nanoparticles enable binding to specific cancer cells, and the design of transcriptional elements has enabled selective DNA expression specific to the cancer cells. Together, these features have improved the performance of polymeric nanoparticles as targeted non-viral gene delivery vectors to treat cancer. As polymeric nanoparticles can be designed to be biodegradable, non-toxic, and to have reduced immunogenicity and tumorigenicity compared to viral platforms, they have significant potential for clinical use. Results of polymeric gene therapy in clinical trials and future directions for the engineering of nanoparticle systems for targeted cancer gene therapy are also presented.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David R. Wilson
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Camila G. Zamboni
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jordan J. Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
56
|
Niedermayer S, Weiss V, Herrmann A, Schmidt A, Datz S, Müller K, Wagner E, Bein T, Bräuchle C. Multifunctional polymer-capped mesoporous silica nanoparticles for pH-responsive targeted drug delivery. NANOSCALE 2015; 7:7953-7964. [PMID: 25865957 DOI: 10.1039/c4nr07245f] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
A highly stable modular platform, based on the sequential covalent attachment of different functionalities to the surface of core-shell mesoporous silica nanoparticles (MSNs) for targeted drug delivery is presented. A reversible pH-responsive cap system based on covalently attached poly(2-vinylpyridine) (PVP) was developed as drug release mechanism. Our platform offers (i) tuneable interactions and release kinetics with the cargo drug in the mesopores based on chemically orthogonal core-shell design, (ii) an extremely robust and reversible closure and release mechanism based on endosomal acidification of the covalently attached PVP polymer block, (iii) high colloidal stability due to a covalently coupled PEG shell, and (iv) the ability to covalently attach a wide variety of dyes, targeting ligands and other functionalities at the outer periphery of the PEG shell. The functionality of the system was demonstrated in several cell studies, showing pH-triggered release in the endosome, light-triggered endosomal escape with an on-board photosensitizer, and efficient folic acid-based cell targeting.
Collapse
Affiliation(s)
- Stefan Niedermayer
- Department of Chemistry, Nanosystems Initiative Munich (NIM) and Center for Nano Science (CeNS), University of Munich (LMU), Butenandtstr. 11 (E), 81377 Munich, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Kokil GR, Veedu RN, Ramm GA, Prins JB, Parekh HS. Type 2 diabetes mellitus: limitations of conventional therapies and intervention with nucleic acid-based therapeutics. Chem Rev 2015; 115:4719-43. [PMID: 25918949 DOI: 10.1021/cr5002832] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ganesh R Kokil
- †School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Rakesh N Veedu
- §Center for Comparative Genomics, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia.,∥Western Australian Neuroscience Research Institute, Perth, WA 6150, Australia.,‡School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane QLD 4072 Australia
| | - Grant A Ramm
- ⊥The Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia.,#Faculty of Medicine and Biomedical Sciences, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Johannes B Prins
- ∇Mater Research Institute, The University of Queensland, Brisbane, QLD 4101, Australia
| | - Harendra S Parekh
- †School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Brisbane, QLD 4102, Australia
| |
Collapse
|
58
|
Lächelt U, Wagner E. Nucleic Acid Therapeutics Using Polyplexes: A Journey of 50 Years (and Beyond). Chem Rev 2015; 115:11043-78. [DOI: 10.1021/cr5006793] [Citation(s) in RCA: 418] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ulrich Lächelt
- Pharmaceutical
Biotechnology, Department of Pharmacy, Ludwig Maximilians Universität, 81377 Munich, Germany
- Nanosystems
Initiative
Munich (NIM), 80799 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical
Biotechnology, Department of Pharmacy, Ludwig Maximilians Universität, 81377 Munich, Germany
- Nanosystems
Initiative
Munich (NIM), 80799 Munich, Germany
| |
Collapse
|
59
|
Moreno M, Giralt E. Three valuable peptides from bee and wasp venoms for therapeutic and biotechnological use: melittin, apamin and mastoparan. Toxins (Basel) 2015; 7:1126-50. [PMID: 25835385 PMCID: PMC4417959 DOI: 10.3390/toxins7041126] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/18/2015] [Accepted: 03/25/2015] [Indexed: 12/14/2022] Open
Abstract
While knowledge of the composition and mode of action of bee and wasp venoms dates back 50 years, the therapeutic value of these toxins remains relatively unexploded. The properties of these venoms are now being studied with the aim to design and develop new therapeutic drugs. Far from evaluating the extensive number of monographs, journals and books related to bee and wasp venoms and the therapeutic effect of these toxins in numerous diseases, the following review focuses on the three most characterized peptides, namely melittin, apamin, and mastoparan. Here, we update information related to these compounds from the perspective of applied science and discuss their potential therapeutic and biotechnological applications in biomedicine.
Collapse
Affiliation(s)
- Miguel Moreno
- Chemistry and Molecular Pharmacology, Institute for Research in Biomedicine (IRB Barcelona), Baldiri i Reixac, 10, Barcelona 08028, Spain.
| | - Ernest Giralt
- Chemistry and Molecular Pharmacology, Institute for Research in Biomedicine (IRB Barcelona), Baldiri i Reixac, 10, Barcelona 08028, Spain.
| |
Collapse
|
60
|
Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy. Eur J Pharm Biopharm 2015; 93:52-79. [PMID: 25813885 DOI: 10.1016/j.ejpb.2015.03.018] [Citation(s) in RCA: 1081] [Impact Index Per Article: 108.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 02/08/2023]
Abstract
Cancer is the second worldwide cause of death, exceeded only by cardiovascular diseases. It is characterized by uncontrolled cell proliferation and an absence of cell death that, except for hematological cancers, generates an abnormal cell mass or tumor. This primary tumor grows thanks to new vascularization and, in time, acquires metastatic potential and spreads to other body sites, which causes metastasis and finally death. Cancer is caused by damage or mutations in the genetic material of the cells due to environmental or inherited factors. While surgery and radiotherapy are the primary treatment used for local and non-metastatic cancers, anti-cancer drugs (chemotherapy, hormone and biological therapies) are the choice currently used in metastatic cancers. Chemotherapy is based on the inhibition of the division of rapidly growing cells, which is a characteristic of the cancerous cells, but unfortunately, it also affects normal cells with fast proliferation rates, such as the hair follicles, bone marrow and gastrointestinal tract cells, generating the characteristic side effects of chemotherapy. The indiscriminate destruction of normal cells, the toxicity of conventional chemotherapeutic drugs, as well as the development of multidrug resistance, support the need to find new effective targeted treatments based on the changes in the molecular biology of the tumor cells. These novel targeted therapies, of increasing interest as evidenced by FDA-approved targeted cancer drugs in recent years, block biologic transduction pathways and/or specific cancer proteins to induce the death of cancer cells by means of apoptosis and stimulation of the immune system, or specifically deliver chemotherapeutic agents to cancer cells, minimizing the undesirable side effects. Although targeted therapies can be achieved directly by altering specific cell signaling by means of monoclonal antibodies or small molecules inhibitors, this review focuses on indirect targeted approaches that mainly deliver chemotherapeutic agents to molecular targets overexpressed on the surface of tumor cells. In particular, we offer a detailed description of different cytotoxic drug carriers, such as liposomes, carbon nanotubes, dendrimers, polymeric micelles, polymeric conjugates and polymeric nanoparticles, in passive and active targeted cancer therapy, by enhancing the permeability and retention or by the functionalization of the surface of the carriers, respectively, emphasizing those that have received FDA approval or are part of the most important clinical studies up to date. These drug carriers not only transport the chemotherapeutic agents to tumors, avoiding normal tissues and reducing toxicity in the rest of the body, but also protect cytotoxic drugs from degradation, increase the half-life, payload and solubility of cytotoxic agents and reduce renal clearance. Despite the many advantages of all the anticancer drug carriers analyzed, only a few of them have reached the FDA approval, in particular, two polymer-protein conjugates, five liposomal formulations and one polymeric nanoparticle are available in the market, in contrast to the sixteen FDA approval of monoclonal antibodies. However, there are numerous clinical trials in progress of polymer-protein and polymer-drug conjugates, liposomal formulations, including immunoliposomes, polymeric micelles and polymeric nanoparticles. Regarding carbon nanotubes or dendrimers, there are no FDA approvals or clinical trials in process up to date due to their unresolved toxicity. Moreover, we analyze in detail the more promising and advanced preclinical studies of the particular case of polymeric nanoparticles as carriers of different cytotoxic agents to active and passive tumor targeting published in the last 5 years, since they have a huge potential in cancer therapy, being one of the most widely studied nano-platforms in this field in the last years. The interest that these formulations have recently achieved is stressed by the fact that 90% of the papers based on cancer therapeutics with polymeric nanoparticles have been published in the last 6 years (PubMed search).
Collapse
|
61
|
Kos P, Lächelt U, Herrmann A, Mickler FM, Döblinger M, He D, Krhač Levačić A, Morys S, Bräuchle C, Wagner E. Histidine-rich stabilized polyplexes for cMet-directed tumor-targeted gene transfer. NANOSCALE 2015; 7:5350-5362. [PMID: 25721131 DOI: 10.1039/c4nr06556e] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Overexpression of the hepatocyte growth factor receptor/c-Met proto oncogene on the surface of a variety of tumor cells gives an opportunity to specifically target cancerous tissues. Herein, we report the first use of c-Met as receptor for non-viral tumor-targeted gene delivery. Sequence-defined oligomers comprising the c-Met binding peptide ligand cMBP2 for targeting, a monodisperse polyethylene glycol (PEG) for polyplex surface shielding, and various cationic (oligoethanamino) amide cores containing terminal cysteines for redox-sensitive polyplex stabilization, were assembled by solid-phase supported syntheses. The resulting oligomers exhibited a greatly enhanced cellular uptake and gene transfer over non-targeted control sequences, confirming the efficacy and target-specificity of the formed polyplexes. Implementation of endosomal escape-promoting histidines in the cationic core was required for gene expression without additional endosomolytic agent. The histidine-enriched polyplexes demonstrated stability in serum as well as receptor-specific gene transfer in vivo upon intratumoral injection. The co-formulation with an analogous PEG-free cationic oligomer led to a further compaction of pDNA polyplexes with an obvious change of shape as demonstrated by transmission electron microscopy. Such compaction was critically required for efficient intravenous gene delivery which resulted in greatly enhanced, cMBP2 ligand-dependent gene expression in the distant tumor.
Collapse
Affiliation(s)
- Petra Kos
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig Maximilians University Munich, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Mandlmeier B, Niedermayer S, Schmidt A, Schuster J, Bein T. Lipid-bilayer coated nanosized bimodal mesoporous carbon spheres for controlled release applications. J Mater Chem B 2015; 3:9323-9329. [DOI: 10.1039/c5tb01635e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report the facile synthesis of mesoporous nanosized carbon spheres (MCS) featuring a very high porosity. The MCS are subsequently sealed with an active and biocompatible lipid bilayer making the SLB@MCS suitable for release-on-demand applications.
Collapse
Affiliation(s)
- Benjamin Mandlmeier
- Department of Chemistry and Center for NanoScience (CeNS)
- University of Munich (LMU)
- 81377 Munich
- Germany
| | - Stefan Niedermayer
- Department of Chemistry and Center for NanoScience (CeNS)
- University of Munich (LMU)
- 81377 Munich
- Germany
| | - Alexandra Schmidt
- Department of Chemistry and Center for NanoScience (CeNS)
- University of Munich (LMU)
- 81377 Munich
- Germany
| | - Jörg Schuster
- Department of Chemistry and Center for NanoScience (CeNS)
- University of Munich (LMU)
- 81377 Munich
- Germany
| | - Thomas Bein
- Department of Chemistry and Center for NanoScience (CeNS)
- University of Munich (LMU)
- 81377 Munich
- Germany
| |
Collapse
|
63
|
Yu Z, Yu B, Kaye JB, Tang C, Chen S, Dong C, Shen B. Perspectives and Challenges of Cell-Penetrating Peptides in Effective siRNA Delivery. ACTA ACUST UNITED AC 2014. [DOI: 10.1142/s1793984414410165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Over the last two decades, hundreds of cell penetrating peptides (CPPs) have been intensively developed as drug and nucleic acid delivery vectors. In many cases, however, the efficient delivery of exogenous bioactive molecules through the plasma membrane to their targets remains a tremendous challenging issue. CPPs have attracted tremendous research interest as efficient cellular delivery vehicles due to their intrinsic ability to enter cells and mediate uptake of a wide range of macromolecular cargos, such as proteins, peptides, nucleic acids, drugs and nanoparticle carriers. This review presents and discusses the current perspectives of CPP-mediated siRNA delivery system. We focus on the CPP-mediated siRNA delivery approaches, and particular emphasis is placed on the strategies for the advantages and disadvantages for each delivery approach. Lastly, the cellular uptake mechanisms of CPPs and the specific challenges associated with each delivery system of siRNAs are discussed.
Collapse
Affiliation(s)
- Zhiqiang Yu
- Department of Physiology, Anhui Medical University, Hefei, Anhui 230032, P. R. China
- Center for BioEnergetics, The Biodesign Institute and Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Bin Yu
- School of Pharmaceutical Sciences and New Drug Research & Development Center Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Justin Boy Kaye
- Center for BioEnergetics, The Biodesign Institute and Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Chenhong Tang
- Center for BioEnergetics, The Biodesign Institute and Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Shengxi Chen
- Center for BioEnergetics, The Biodesign Institute and Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Chenbo Dong
- Department of Chemical Engineering, West Virginia University, Morgantown, WV 26505, USA
| | - Bing Shen
- Department of Physiology, Anhui Medical University, Hefei, Anhui 230032, P. R. China
| |
Collapse
|
64
|
Novo L, Takeda KM, Petteta T, Dakwar GR, van den Dikkenberg JB, Remaut K, Braeckmans K, van Nostrum CF, Mastrobattista E, Hennink WE. Targeted Decationized Polyplexes for siRNA Delivery. Mol Pharm 2014; 12:150-61. [DOI: 10.1021/mp500499x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Luís Novo
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584, CG Utrecht, The Netherlands
| | - Kaori M. Takeda
- Department
of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tamara Petteta
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584, CG Utrecht, The Netherlands
| | - George R. Dakwar
- Laboratory for General
Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent Research Group on Nanomedicines, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Joep B. van den Dikkenberg
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584, CG Utrecht, The Netherlands
| | - Katrien Remaut
- Laboratory for General
Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent Research Group on Nanomedicines, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory for General
Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent Research Group on Nanomedicines, Harelbekestraat 72, 9000 Ghent, Belgium
- Centre
for Nano- and Biophotonics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Cornelus F. van Nostrum
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584, CG Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584, CG Utrecht, The Netherlands
| | - Wim E. Hennink
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584, CG Utrecht, The Netherlands
| |
Collapse
|
65
|
Hong CA, Nam YS. Functional nanostructures for effective delivery of small interfering RNA therapeutics. Am J Cancer Res 2014; 4:1211-32. [PMID: 25285170 PMCID: PMC4183999 DOI: 10.7150/thno.8491] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 02/23/2014] [Indexed: 02/04/2023] Open
Abstract
Small interfering RNA (siRNA) has proved to be a powerful tool for target-specific gene silencing via RNA interference (RNAi). Its ability to control targeted gene expression gives new hope to gene therapy as a treatment for cancers and genetic diseases. However, siRNA shows poor pharmacological properties, such as low serum stability, off-targeting, and innate immune responses, which present a significant challenge for clinical applications. In addition, siRNA cannot cross the cell membrane for RNAi activity because of its anionic property and stiff structure. Therefore, the development of a safe, stable, and efficient system for the delivery of siRNA therapeutics into the cytoplasm of targeted cells is crucial. Several nanoparticle platforms for siRNA delivery have been developed to overcome the major hurdles facing the therapeutic uses of siRNA. This review covers a broad spectrum of non-viral siRNA delivery systems developed for enhanced cellular uptake and targeted gene silencing in vitro and in vivo and discusses their characteristics and opportunities for clinical applications of therapeutic siRNA.
Collapse
|
66
|
Abstract
![]()
RNA
interference (RNAi) is an endogenous process in which small
noncoding RNAs, including small interfering RNAs (siRNAs) and microRNAs
(miRNAs), post-transcriptionally regulate gene expressions. In general,
siRNA and miRNA/miRNA mimics are similar in nature and activity except
their origin and specificity. Although both siRNAs and miRNAs have
been extensively studied as novel therapeutics for a wide range of
diseases, the large molecular weight, anionic surface charges, instability
in blood circulation, and intracellular trafficking to the RISC after
cellular uptake have hindered the translation of these RNAs from bench
to clinic. As a result, a great variety of delivery systems have been
investigated for safe and effective delivery of small noncoding RNAs.
Among these systems, peptides, especially cationic peptides, have
emerged as a promising type of carrier due to their inherent ability
to condense negatively charged RNAs, ease of synthesis, controllable
size, and tunable structure. In this review, we will focus on three
major types of cationic peptides, including poly(l-lysine)
(PLL), protamine, and cell penetrating peptides (CPP), as well as
peptide targeting ligands that have been extensively used in RNA delivery.
The delivery strategies, applications, and limitations of these cationic
peptides in siRNA/miRNA delivery will be discussed.
Collapse
Affiliation(s)
- Ravi S Shukla
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City , Kansas City, Missouri 64108, United States
| | | | | |
Collapse
|
67
|
Klein PM, Wagner E. Bioreducible polycations as shuttles for therapeutic nucleic acid and protein transfection. Antioxid Redox Signal 2014; 21:804-17. [PMID: 24219092 PMCID: PMC4098974 DOI: 10.1089/ars.2013.5714] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 11/12/2013] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE Nucleic acids such as gene-encoding DNAs, gene-silencing small interfering RNAs, or recombinant proteins addressing intracellular molecular targets present a major new therapeutic modality, provided efficient solutions for intracellular delivery can be found. The different physiological redox environments inside and outside the cell can be utilized for optimizing the involved transport processes. RECENT ADVANCES Intracellular delivery of nucleic acids or proteins requires dynamic carriers that discriminate between different cellular locations. Bioreducible cationic polymers can package their therapeutic cargo stably in the extracellular environment, but sense the reducing intracellular cytosolic environment. Based on disulfide cleavage, carriers are degraded into biocompatible fragments and release the cargo in functional form. Disulfide linkages between oligocations, between the carrier and the cargo, or spatial caging of complexed cargo by disulfides have been pursued, with polymers or precise sequence-defined peptides and oligomers. CRITICAL ISSUES A quantitative knowledge of the bioreductive capacities within different biological compartments and the involved cellular reduction processes would be greatly helpful for improved carriers with disulfides cleaved within the right compartment at the right time. FUTURE DIRECTIONS Novel designs of multifunctional nanocarriers will incorporate macromolecular disulfide entry mechanisms previously optimized by natural evolution of toxins and viruses. In addition to extracellular stabilization and intracellular disassembly, tuned disulfides will contribute to deshielding at the cell surface, or translocation from intracellular compartments to the cytosol.
Collapse
Affiliation(s)
- Philipp M. Klein
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-University, Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-University, Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
68
|
Targeted delivery of miRNA therapeutics for cardiovascular diseases: opportunities and challenges. Clin Sci (Lond) 2014; 127:351-65. [PMID: 24895056 DOI: 10.1042/cs20140005] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dysregulation of miRNA expression has been associated with many cardiovascular diseases in animal models, as well as in patients. In the present review, we summarize recent findings on the role of miRNAs in cardiovascular diseases and discuss the opportunities, possibilities and challenges of using miRNAs as future therapeutic targets. Furthermore, we focus on the different approaches that can be used to deliver these newly developed miRNA therapeutics to their sites of action. Since siRNAs are structurally homologous with the miRNA therapeutics, important lessons learned from siRNA delivery strategies are discussed that might be applicable to targeted delivery of miRNA therapeutics, thereby reducing costs and potential side effects, and improving efficacy.
Collapse
|
69
|
Tong R, Tang L, Ma L, Tu C, Baumgartner R, Cheng J. Smart chemistry in polymeric nanomedicine. Chem Soc Rev 2014; 43:6982-7012. [DOI: 10.1039/c4cs00133h] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
70
|
Matsumoto A, Kataoka K, Miyahara Y. New directions in the design of phenylboronate-functionalized polymers for diagnostic and therapeutic applications. Polym J 2014. [DOI: 10.1038/pj.2014.42] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
71
|
Yousefi A, Lauwers M, Nemes R, van Holten T, Babae N, Roest M, Storm G, Schiffelers R, Mastrobattista E. Hemocompatibility Assessment of two siRNA Nanocarrier Formulations. Pharm Res 2014; 31:3127-35. [DOI: 10.1007/s11095-014-1405-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/28/2014] [Indexed: 12/21/2022]
|
72
|
Parmar RG, Poslusney M, Busuek M, Williams JM, Garbaccio R, Leander K, Walsh E, Howell B, Sepp-Lorenzino L, Riley S, Patel M, Kemp E, Latham A, Leone A, Soli E, Burke RS, Carr B, Colletti SL, Wang W. Novel endosomolytic poly(amido amine) polymer conjugates for systemic delivery of siRNA to hepatocytes in rodents and nonhuman primates. Bioconjug Chem 2014; 25:896-906. [PMID: 24742200 DOI: 10.1021/bc400527e] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The application of small interfering (si)RNAs as potential therapeutic agents requires safe and effective methods for their delivery to the cytoplasm of the target cells and tissues. Recent studies have shown significant progress in the development of targeting reagents that facilitate the recognition of, and siRNA delivery to, specific cell types. Among recently reported delivery approaches, polymers with amphipathic properties have been used to enable endosome escape and cytosolic delivery. Here, we describe a linear amphipathic poly(amido amine) polymer conjugate system for the efficient siRNA delivery in vitro and in vivo. This polymer contains a novel amine bearing bis-acrylamide monomer designed for increasing amine density, which resulted in substantial improvement in liver uptake and RNAi activity compared to our previously reported poly(amido amine disulfide) polymer.1 The activity for this liver targeted delivery system was demonstrated in rodents and nonhuman primates.
Collapse
Affiliation(s)
- Rubina Giare Parmar
- Department of Medicinal Chemistry, ‡Department of Pharmaceutical Sciences, §Department of RNAi Biology, ∥Department of Pharmacokinetics, Pharmacodynamics & Drug Metabolism, ⊥Department of Process Chemistry, Merck & Co. , West Point, Pennsylvania 19438, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Cavallaro G, Licciardi M, Amato G, Sardo C, Giammona G, Farra R, Dapas B, Grassi M, Grassi G. Synthesis and characterization of polyaspartamide copolymers obtained by ATRP for nucleic acid delivery. Int J Pharm 2014; 466:246-57. [DOI: 10.1016/j.ijpharm.2014.03.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/07/2014] [Accepted: 03/09/2014] [Indexed: 01/23/2023]
|
74
|
Tschiche A, Malhotra S, Haag R. Nonviral gene delivery with dendritic self-assembling architectures. Nanomedicine (Lond) 2014; 9:667-93. [DOI: 10.2217/nnm.14.32] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In this review, we outline the concept and applicability of self-assembling dendrimers for gene-delivery applications. Low-molecular-weight, well-defined cationic dendritic arrays which have been modified with hydrophobic domains can form self-organized multivalent systems that have significant advantages over nonassembling, high-molecular-weight/polymeric gene vectors. Particular structural variations have been highlighted with respect to the individual components of the displayed dendritic amphiphiles, namely, the employed amine termini, the hydrophobic segment, the size of the dendritic array, and the integration of special features such as targeting ability and cleavability/degradability, which can all have a crucial effect on gene-transfection efficiencies. Accordingly, the scientific efforts to create new synthetic gene-delivery vectors to act as promising in vivo transfection agents in the future will be presented and discussed here.
Collapse
Affiliation(s)
- Ariane Tschiche
- Institute of Chemistry & Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
| | - Shashwat Malhotra
- Institute of Chemistry & Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
| | | |
Collapse
|
75
|
Barrett SE, Burke RS, Abrams MT, Bason C, Busuek M, Carlini E, Carr BA, Crocker LS, Fan H, Garbaccio RM, Guidry EN, Heo JH, Howell BJ, Kemp EA, Kowtoniuk RA, Latham AH, Leone AM, Lyman M, Parmar RG, Patel M, Pechenov SY, Pei T, Pudvah NT, Raab C, Riley S, Sepp-Lorenzino L, Smith S, Soli ED, Staskiewicz S, Stern M, Truong Q, Vavrek M, Waldman JH, Walsh ES, Williams JM, Young S, Colletti SL. Development of a liver-targeted siRNA delivery platform with a broad therapeutic window utilizing biodegradable polypeptide-based polymer conjugates. J Control Release 2014; 183:124-37. [PMID: 24657948 DOI: 10.1016/j.jconrel.2014.03.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 03/04/2014] [Accepted: 03/17/2014] [Indexed: 01/08/2023]
Abstract
The greatest challenge standing in the way of effective in vivo siRNA delivery is creating a delivery vehicle that mediates a high degree of efficacy with a broad therapeutic window. Key structure-activity relationships of a poly(amide) polymer conjugate siRNA delivery platform were explored to discover the optimized polymer parameters that yield the highest activity of mRNA knockdown in the liver. At the same time, the poly(amide) backbone of the polymers allowed for the metabolism and clearance of the polymer from the body very quickly, which was established using radiolabeled polymers to demonstrate the time course of biodistribution and excretion from the body. The fast degradation and clearance of the polymers provided for very low toxicity at efficacious doses, and the therapeutic window of this poly(amide)-based siRNA delivery platform was shown to be much broader than a comparable polymer platform. The results of this work illustrate that the poly(amide) platform has a promising future in the development of a siRNA-based drug approved for human use.
Collapse
Affiliation(s)
- Stephanie E Barrett
- Department of RNA Medicinal Chemistry, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA.
| | - Rob S Burke
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Marc T Abrams
- Department of RNA Biology, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Carol Bason
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Marina Busuek
- Department of RNA Medicinal Chemistry, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Edward Carlini
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Brian A Carr
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Louis S Crocker
- Department of RNAi Analytical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Haihong Fan
- Department of RNAi Analytical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Robert M Garbaccio
- Department of RNA Medicinal Chemistry, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Erin N Guidry
- Department of Process Chemistry, Merck Research Laboratories, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Jun H Heo
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Bonnie J Howell
- Department of RNA Biology, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Eric A Kemp
- Department of RNAi Analytical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Robert A Kowtoniuk
- Department of RNA Medicinal Chemistry, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Andrew H Latham
- Department of RNAi Analytical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Anthony M Leone
- Department of RNAi Analytical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Michael Lyman
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Rubina G Parmar
- Department of RNA Medicinal Chemistry, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Mihir Patel
- Department of RNAi Analytical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Sergey Y Pechenov
- Department of RNAi Analytical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Tao Pei
- Department of Process Chemistry, Merck Research Laboratories, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Nicole T Pudvah
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Conrad Raab
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Sean Riley
- Department of RNAi Analytical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Laura Sepp-Lorenzino
- Department of RNA Biology, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Sheri Smith
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Eric D Soli
- Department of Process Chemistry, Merck Research Laboratories, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Steven Staskiewicz
- Department of Process Chemistry, Merck Research Laboratories, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Melissa Stern
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Quang Truong
- Department of Process Chemistry, Merck Research Laboratories, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Marissa Vavrek
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Jacob H Waldman
- Department of Process Chemistry, Merck Research Laboratories, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Eileen S Walsh
- Department of RNA Biology, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - J Michael Williams
- Department of RNA Medicinal Chemistry, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Stephanie Young
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| | - Steven L Colletti
- Department of RNA Medicinal Chemistry, Merck Research Laboratories, Merck & Co., Inc., West Point, PA 19486, USA
| |
Collapse
|
76
|
Levenson EA, Kiick KL. DNA-polymer conjugates for immune stimulation through Toll-like receptor 9 mediated pathways. Acta Biomater 2014; 10:1134-45. [PMID: 24316364 DOI: 10.1016/j.actbio.2013.11.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 10/01/2013] [Accepted: 11/24/2013] [Indexed: 01/01/2023]
Abstract
Oligodeoxynucleotides (ODNs) containing unmethylated CpG dinucleotide motifs are agonists of Toll-like receptor 9 and are currently being investigated for use as vaccine adjuvants through the promotion of type I immunity. Several classes of ODN have been developed which differ in their propensity to aggregate, which in turn alters cytokine profiles and cellular subsets activated. Although aggregation state is correlated with the change in cytokine response, it is unknown if this results from a change in the number of ODNs available for binding and/or the possible engagement of multiple TLR9 molecules. Here, we examined the role of ligand valency on the activation of TLR9 through the synthesis of ODN-poly(acrylic acid) (PAA) conjugates. The compositions and size of the conjugates were characterized by UV-vis spectroscopy, proton nuclear magnetic resonance, gel permeation chromatography and dynamic light scattering. Enzyme-linked immunosorbent assays of cytokine secretion by murine-like macrophages indicate that these ODN-PAA polymer conjugates show enhanced immunostimulation at 100-fold lower concentrations than those required for ODN alone, for both TNF-α and IL-6 release, and are more potent than any other previously reported multivalent ODN constructs. Increasing valency was shown to significantly enhance cytokine expression, particularly for IL-6. Knockdown by siRNA demonstrates that these polymer conjugates are specific to TLR9. Our results define valency as a critical design parameter and polymer conjugation as an advantageous strategy for producing ODN immunomodulatory agents.
Collapse
Affiliation(s)
- Eric A Levenson
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA; Biomedical Engineering, University of Delaware, Newark, DE 19716, USA; The Delaware Biotechnology Institute, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
77
|
Guidry EN, Farand J, Soheili A, Parish CA, Kevin NJ, Pipik B, Calati KB, Ikemoto N, Waldman JH, Latham AH, Howell BJ, Leone A, Garbaccio RM, Barrett SE, Parmar RG, Truong QT, Mao B, Davies IW, Colletti SL, Sepp-Lorenzino L. Improving the in vivo therapeutic index of siRNA polymer conjugates through increasing pH responsiveness. Bioconjug Chem 2014; 25:296-307. [PMID: 24409989 DOI: 10.1021/bc400442p] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Polymer based carriers that aid in endosomal escape have proven to be efficacious siRNA delivery agents in vitro and in vivo; however, most suffer from cytotoxicity due in part to a lack of selectivity for endosomal versus cell membrane lysis. For polymer based carriers to move beyond the laboratory and into the clinic, it is critical to find carriers that are not only efficacious, but also have margins that are clinically relevant. In this paper we report three distinct categories of polymer conjugates that improve the selectivity of endosomal membrane lysis by relying on the change in pH associated with endosomal trafficking, including incorporation of low pKa heterocycles, acid cleavable amino side chains, or carboxylic acid pH sensitive charge switches. Additionally, we determine the therapeutic index of our polymer conjugates in vivo and demonstrate that the incorporation of pH responsive elements dramatically expands the therapeutic index to 10-15, beyond that of the therapeutic index (less than 3), for polymer conjugates previously reported.
Collapse
Affiliation(s)
- Erin N Guidry
- Department of Process Chemistry and ∥Department of Analytical Chemistry, Merck & Co. Inc. , , Rahway, New Jersey, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Abstract
For the last five decades cationic polymers have been used for nucleic acids transfection. Our understanding of polymer-nucleic acid interactions and their rational use in delivery has continuously increased. The great improvements in macromolecular chemistry and the recognition of distinct biological extra- and intracellular delivery hurdles triggered several breakthrough developments, including the discovery of natural and synthetic polycations for compaction of nucleic acids into stable nanoparticles termed polyplexes; the incorporation of targeting ligands and surface-shielding of polyplexes to enable receptor-mediated gene delivery into defined target tissues; and strongly improved intracellular transfer efficacy by better endosomal escape of vesicle-trapped polyplexes into the cytosol. These experiences triggered the development of second-generation polymers with more dynamic properties, such as endosomal pH-responsive release mechanisms, or biodegradable units for improved biocompatibility and intracellular release of the nucleic acid pay load. Despite a better biological understanding, significant challenges such as efficient nuclear delivery and persistence of gene expression persist. The therapeutic perspectives widened from pDNA-based gene therapy to application of novel therapeutic nucleic acids including mRNA, siRNA, and microRNA. The finding that different therapeutic pay loads require different tailor-made carriers complicates preclinical developments. Convincing evidence of medical efficacy still remains to be demonstrated. Bioinspired multifunctional polyplexes resembling "synthetic viruses" appear as attractive opportunity, but provide additional challenges: how to identify optimum combinations of functional delivery units, and how to prepare such polyplexes reproducibly in precise form? Design of sequence-defined polymers, screening of combinatorial polymer and polyplex libraries are tools for further chemical evolution of polyplexes.
Collapse
Affiliation(s)
- Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-University Munich, and Nanosystems Initiative Munich (NIM), Munich, Germany
| |
Collapse
|
79
|
Huang X, Sevimli SI, Bulmus V. pH-labile sheddable block copolymers by RAFT polymerization: Synthesis and potential use as siRNA conjugates. Eur Polym J 2013. [DOI: 10.1016/j.eurpolymj.2013.03.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
80
|
Is there a future for cell-penetrating peptides in oligonucleotide delivery? Eur J Pharm Biopharm 2013; 85:5-11. [DOI: 10.1016/j.ejpb.2013.03.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 11/23/2022]
|
81
|
Cass P, Knower W, Hinton T, Shi S, Grusche F, Tizard M, Gunatillake P. Synthesis and evaluation of degradable polyurea block copolymers as siRNA delivery agents. Acta Biomater 2013; 9:8299-307. [PMID: 23684725 DOI: 10.1016/j.actbio.2013.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 05/01/2013] [Accepted: 05/09/2013] [Indexed: 01/08/2023]
Abstract
Chain extension by diisocyanate condensation provides a versatile and convenient means for preparing block copolymers. We have utilized this chemistry to prepare reducible multiblock polycations for siRNA delivery. This approach, an alternative to oxidative coupling, was suitable for preparing multiblock polycations with defined molecular weight and architecture. The polymer, PEG-b-multi-(polyhexylurea-co-oligo-L-lysine)-b-PEG, was capable of electrostatically condensing siRNA to form nano-sized polyplexes across a broad compositional range. We demonstrated that the polyplexes enter the cells via endocytosis and interact with the endosome membrane leading to destabilization and hence endosome escape. Another feature of these polymers is their multiple intra-chain disulfide linkages. This enables weakening of the polyplex via chain scission within the cytosol's reductive environment. In addition to the controlled preparation of the polymer, the polyplexes were capable of delivering siRNA in vitro to silence greater than 50% green fluorescent protein expression with negligible toxicity.
Collapse
|
82
|
|
83
|
Stigliano C, Aryal S, de Tullio MD, Nicchia GP, Pascazio G, Svelto M, Decuzzi P. siRNA-chitosan complexes in poly(lactic-co-glycolic acid) nanoparticles for the silencing of aquaporin-1 in cancer cells. Mol Pharm 2013; 10:3186-94. [PMID: 23789777 DOI: 10.1021/mp400224u] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A large number of studies document the strong expression of aquaporin-1 (AQP1) in tumor microvessels and correlate this aberrant expression with higher metastatic potential and aggressiveness of the malignancy. Although small animal experiments have shown that the modulation of AQP1 expression can halt angiogenesis and induce tumor regression, effective and safe strategies for the tissue specific inhibition of AQP1 are still missing. Here, small interference RNA-chitosan complexes encapsulated in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) are proposed for the intracellular delivery of siRNA molecules against AQP1. These NPs are coated with poly(vinyl alcohol) (PVA), to improve stability under physiological conditions, and demonstrate a diameter of 160 nm. The partial neutralization of the negatively charged siRNA molecules with the cationic chitosan enhances the loading by 5-fold, as compared to that of the free siRNA molecules, and allows one to modulate the release kinetics in the pH-dependent manner. At pH = 7.4, mimicking the conditions found in the systemic circulation, only the 40% of siRNA is released at 24 h post incubation; whereas at pH = 5.0, recreating the cell endosomal environment, all siRNA molecules are released in about 3 h. These NPs show no cytotoxicity on HeLa cells up to 72 h of incubation. In the same cells, transfected to overexpress AQP1, a silencing efficiency of 70% is achieved at 24 h post treatment with siRNA-loaded NPs. Confocal microscopy analysis of NP uptake demonstrates that siRNA molecules accumulate perinuclearly and in the nucleus. Given the stability, preferential release behavior, and well-known biocompatibility properties of PLGA nanostructures, these siRNA-loaded NPs hold potential for the efficient and safe in vivo silencing of AQPs via systemic administration.
Collapse
Affiliation(s)
- Cinzia Stigliano
- Department of Translational Imaging and Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, Texas 77030, United States
| | | | | | | | | | | | | |
Collapse
|
84
|
Mackowiak SA, Schmidt A, Weiss V, Argyo C, von Schirnding C, Bein T, Bräuchle C. Targeted drug delivery in cancer cells with red-light photoactivated mesoporous silica nanoparticles. NANO LETTERS 2013; 13:2576-2583. [PMID: 23662711 DOI: 10.1021/nl400681f] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Mesoporous nanoparticles for drug delivery would benefit significantly from further improvements in targeting efficiency and endosomal release. We present a system based on colloidal mesoporous silica nanoparticles with targeting-ligands and a red-light photosensitizer. This nanoparticle system provides spatial and temporal control of the release of drugs into the cytosol of cancer cells. Furthermore, the system presents a general platform since it can be loaded with different cargos and adapted for targeting of multiple cell types.
Collapse
Affiliation(s)
- Stephan A Mackowiak
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU), Butenandtstrasse 5-13(E), 81377 Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
85
|
Trends in polymeric delivery of nucleic acids to tumors. J Control Release 2013; 170:209-18. [PMID: 23770011 DOI: 10.1016/j.jconrel.2013.05.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 11/21/2022]
Abstract
Delivery of nucleic acids to tumors has received extensive attention in the past few decades since these molecules are capable of treating disease by modulating the source of abnormalities. Although high efficiency and low toxicity of numerous delivery systems for nucleic acids have been approved frequently with in vitro assays, contradictions have been observed in many cases between these results and what has occurred in the dynamic in vivo situation. Filling this gap seems to be crucial for further preclinical development of such systems. In this paper, we discuss various barriers which polymeric DNA or siRNA nanoparticles encounter upon systemic administration with an aim to assist in designing more relevant in vitro assays. Furthermore, individual considerations concerning delivery of DNA and siRNA have been addressed.
Collapse
|
86
|
Cell-penetrating cationic siRNA and lipophilic derivatives efficient at nanomolar concentrations in the presence of serum and albumin. J Control Release 2013; 170:92-8. [PMID: 23639452 DOI: 10.1016/j.jconrel.2013.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 04/12/2013] [Accepted: 04/17/2013] [Indexed: 01/22/2023]
Abstract
Despite its considerable interest in human therapy, in vivo siRNA delivery is still suffering from hurdles of vectorization. We have shown recently efficient gene silencing by non-vectorized cationic siRNA. Here, we describe the synthesis and in vitro evaluation of new amphiphilic cationic siRNA. C₁₂-, (C₁₂)₂- and cholesteryl-spermine(x)-siRNA were capable of luciferase knockdown at nanomolar concentrations without vectorization (i.e. one to two orders of magnitude more potent than commercially available cholesteryl siRNA). Moreover, incubation in the presence of serum did not impair their efficiency. Finally, amphiphilic cationic siRNA was pre-loaded on albumin. In A549Luc cells in the presence of serum, these siRNA conjugates were highly effective and had low toxicity.
Collapse
|
87
|
Guo J, Evans JC, O’Driscoll CM. Delivering RNAi therapeutics with non-viral technology: a promising strategy for prostate cancer? Trends Mol Med 2013; 19:250-61. [DOI: 10.1016/j.molmed.2013.02.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 01/30/2013] [Accepted: 02/07/2013] [Indexed: 02/07/2023]
|
88
|
Parmar RG, Busuek M, Walsh ES, Leander KR, Howell BJ, Sepp-Lorenzino L, Kemp E, Crocker LS, Leone A, Kochansky CJ, Carr BA, Garbaccio RM, Colletti SL, Wang W. Endosomolytic Bioreducible Poly(amido amine disulfide) Polymer Conjugates for the in Vivo Systemic Delivery of siRNA Therapeutics. Bioconjug Chem 2013; 24:640-7. [PMID: 23496378 DOI: 10.1021/bc300600a] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Rubina Giare Parmar
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Marina Busuek
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Eileen S. Walsh
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Karen R. Leander
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Bonnie J. Howell
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Laura Sepp-Lorenzino
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Eric Kemp
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Louis S. Crocker
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Anthony Leone
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Christopher J. Kochansky
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Brian A. Carr
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Robert M. Garbaccio
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Steven L. Colletti
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Weimin Wang
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| |
Collapse
|
89
|
Abstract
The emergence of RNAi offers a potentially exciting new therapeutic paradigm for respiratory diseases. However, effective delivery remains a key requirement for their translation into the clinic and has been a major factor in the limited clinical success seen to date. Inhalation offers tissue-specific targeting of the RNAi to treat respiratory diseases and a diminished risk of off-target effects. In order to deliver RNAi directly to the respiratory tract via inhalation, ‘smart’ non-viral carriers are required to protect the RNAi during delivery/aerosolization and enhance cell-specific uptake to target cells. Here, we review the state-of-the-art in therapeutic aerosol bioengineering, and specifically non-viral siRNA delivery platforms, for delivery via inhalation. This includes developments in inhaler device engineering and particle engineering, including manufacturing methods and excipients used in therapeutic aerosol bioengineering that underpin the development of smart, cell type-specific delivery systems to target siRNA to respiratory epithelial cells and/or alveolar macrophages.
Collapse
|
90
|
|
91
|
|
92
|
Rational design of gold(III)-dithiocarbamato peptidomimetics for the targeted anticancer chemotherapy. J Inorg Biochem 2012; 117:248-60. [DOI: 10.1016/j.jinorgbio.2012.07.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 05/30/2012] [Accepted: 07/03/2012] [Indexed: 01/23/2023]
|
93
|
Maier K, Martin I, Wagner E. Sequence Defined Disulfide-Linked Shuttle for Strongly Enhanced Intracellular Protein Delivery. Mol Pharm 2012; 9:3560-8. [DOI: 10.1021/mp300404d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Kevin Maier
- Pharmaceutical Biotechnology, Center for System-Based Drug
Research, and Center for Nanoscience, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Irene Martin
- Pharmaceutical Biotechnology, Center for System-Based Drug
Research, and Center for Nanoscience, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug
Research, and Center for Nanoscience, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| |
Collapse
|
94
|
Fan Z, Senapati D, Singh AK, Ray PC. Theranostic magnetic core-plasmonic shell star shape nanoparticle for the isolation of targeted rare tumor cells from whole blood, fluorescence imaging, and photothermal destruction of cancer. Mol Pharm 2012; 10:857-66. [PMID: 23110457 DOI: 10.1021/mp300468q] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cancer is one of the most life-threatening diseases, which causes 7.6 million deaths and around 1 trillion dollars economic loss every year. Theranostic materials are expected to improve early detection and safe treatment through personalized medicine. Driven by the needs, we report the development of a theranostic plasmonic shell-magnetic core star shape nanomaterial based approach for the targeted isolation of rare tumor cells from the whole blood sample, followed by diagnosis and photothermal destruction. Experimental data with whole blood sample spiked with SK-BR-3 cancer cell shows that Cy3 attached S6 aptamer conjugated theranostic plasmonic/magnetic nanoparticles can be used for fluorescence imaging and magnetic separation even in 0.001% mixtures. A targeted photothermal experiment using 1064 nm near-IR light at 2-3 W/cm(2) for 10 min resulted in selective irreparable cellular damage to most of the SK-BR-3 cancer cells. We discuss the possible mechanism and operating principle for the targeted imaging, separation, and photothermal destruction using theranostic magnetic/plasmonic nanotechnology. After the optimization of different parameters, this theranostic nanotechnology-driven assay could have enormous potential for applications as contrast agent and therapeutic actuators for cancer.
Collapse
Affiliation(s)
- Zhen Fan
- Department of Chemistry, Jackson State University, Jackson, Mississippi 39217, USA
| | | | | | | |
Collapse
|
95
|
Brülisauer L, Kathriner N, Prenrecaj M, Gauthier MA, Leroux JC. Tracking the Bioreduction of Disulfide-Containing Cationic Dendrimers. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201207070] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
96
|
Brülisauer L, Kathriner N, Prenrecaj M, Gauthier MA, Leroux JC. Tracking the Bioreduction of Disulfide-Containing Cationic Dendrimers. Angew Chem Int Ed Engl 2012; 51:12454-8. [DOI: 10.1002/anie.201207070] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Indexed: 11/11/2022]
|
97
|
Erazo-Oliveras A, Muthukrishnan N, Baker R, Wang TY, Pellois JP. Improving the endosomal escape of cell-penetrating peptides and their cargos: strategies and challenges. Pharmaceuticals (Basel) 2012; 5:1177-1209. [PMID: 24223492 PMCID: PMC3816665 DOI: 10.3390/ph5111177] [Citation(s) in RCA: 308] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 10/25/2012] [Accepted: 10/26/2012] [Indexed: 12/13/2022] Open
Abstract
Cell penetrating peptides (CPPs) can deliver cell-impermeable therapeutic cargos into cells. In particular, CPP-cargo conjugates tend to accumulate inside cells by endocytosis. However, they often remain trapped inside endocytic organelles and fail to reach the cytosolic space of cells efficiently. In this review, the evidence for CPP-mediated endosomal escape is discussed. In addition, several strategies that have been utilized to enhance the endosomal escape of CPP-cargos are described. The recent development of branched systems that display multiple copies of a CPP is presented. The use of viral or synthetic peptides that can disrupt the endosomal membrane upon activation by the low pH of endosomes is also discussed. Finally, we survey how CPPs labeled with chromophores can be used in combination with light to stimulate endosomal lysis. The mechanisms and challenges associated with these intracellular delivery methodologies are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Jean-Philippe Pellois
- Author to whom correspondence should be addressed; ; Tel.: +1-979-845-0101; Fax: +1-979-862-4718
| |
Collapse
|
98
|
Naito M, Ishii T, Matsumoto A, Miyata K, Miyahara Y, Kataoka K. A phenylboronate-functionalized polyion complex micelle for ATP-triggered release of siRNA. Angew Chem Int Ed Engl 2012; 51:10751-5. [PMID: 22907668 DOI: 10.1002/anie.201203360] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 06/26/2012] [Indexed: 12/19/2022]
Affiliation(s)
- Mitsuru Naito
- Department of Materials Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8656, Japan
| | | | | | | | | | | |
Collapse
|
99
|
Ghanty U, Fostvedt E, Valenzuela R, Beal PA, Burrows CJ. Promiscuous 8-alkoxyadenosines in the guide strand of an siRNA: modulation of silencing efficacy and off-pathway protein binding. J Am Chem Soc 2012; 134:17643-52. [PMID: 23030736 DOI: 10.1021/ja307102g] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
8-Alkoxyadenosines have the potential to exist in anti or syn conformations around the glycosidic bond when paired opposite to U or G in the complementary strands, thereby placing the sterically demanding 8-alkoxy groups in the major or minor groove, respectively, of duplex RNA. These modified bases were used as "base switches" in the guide strands of an siRNA to prevent off-pathway protein binding during delivery via placement of the alkoxy group in the minor groove, while maintaining significant RNAi efficacy by orienting the alkoxy group in the major groove. 8-Alkoxyadenosine phosphoramidites were synthesized and incorporated into the guide strand of caspase 2 siRNA at four different positions: two in the seed region, one at the cleavage junction, and another nearer to the 3'-end of the guide strand. Thermal stabilities of the corresponding siRNA duplexes showed that U is preferred over G as the base-pairing partner in the complementary strand. When compared to the unmodified positive control siRNAs, singly modified siRNAs knocked down the target mRNA efficiently and with little or no loss of efficacy. Doubly modified siRNAs were found to be less effective and lose their efficacy at low nanomolar concentrations. SiRNAs singly modified at positions 6 and 10 of the guide strand were found to be effective in blocking binding to the RNA-dependent protein kinase PKR, a cytoplasmic dsRNA-binding protein implicated in sequence-independent off-target effects.
Collapse
Affiliation(s)
- Uday Ghanty
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-0850, USA
| | | | | | | | | |
Collapse
|
100
|
Lee SJ, Son S, Yhee JY, Choi K, Kwon IC, Kim SH, Kim K. Structural modification of siRNA for efficient gene silencing. Biotechnol Adv 2012; 31:491-503. [PMID: 22985697 DOI: 10.1016/j.biotechadv.2012.09.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 09/07/2012] [Accepted: 09/07/2012] [Indexed: 11/16/2022]
Abstract
Small interfering RNA (siRNA) holds a great promise for the future of genomic medicine because of its highly sequence-specific gene silencing and universality in therapeutic target. The medical use of siRNA, however, has been severely hampered by the inherent physico-chemical properties of siRNA itself, such as low charge density, high structural stiffness and rapid enzymatic degradation; therefore, the establishment of efficient and safe siRNA delivery methodology is an essential prerequisite, particularly for systemic administration. For an efficient systemic siRNA delivery, it is a critical issue to obtain small and compact siRNA polyplexes with cationic condensing reagents including cationic polymers, because the size and surface properties of the polyplexes are major determinants for achieving desirable in vivo fate. Unfortunately, synthetic siRNA is not easily condensed with cationic polymers due to its intrinsic rigid structure and low spatial charge density. Accordingly, the loose siRNA polyplexes inevitably expose siRNA to the extracellular environment during systemic circulation, resulting in low therapeutic efficiency and poor biodistribution. In this review, we highlight the innovative approaches to increase the size of siRNA via structural modification of the siRNA itself. The attempts include several methodologies such as hybridization, chemical polymerization, and micro- and nano-structurization of siRNA. Due to its increased charge density and flexibility, the structured siRNA can produce highly condensed and homogenous polyplexes compared to the classical monomeric siRNA. As a result, stable and compact siRNA polyplexes can enhance serum stability and target delivery efficiency in vivo with desirable biodistribution. The review specifically aims to provide the recent progress of structural modification of siRNA. In addition, the article also briefly and concisely explains the improved physico-chemical properties of structured siRNA with respect to stability, condensation ability and gene silencing efficiency.
Collapse
Affiliation(s)
- So Jin Lee
- 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
| | | | | | | | | | | | | |
Collapse
|