1
|
Sakai A, Singh G, Khoshbakht M, Bittner S, Löhr CV, Diaz-Tapia R, Warang P, White K, Luo LL, Tolbert B, Blanco M, Chow A, Guttman M, Li C, Bao Y, Ho J, Maurer-Stroh S, Chatterjee A, Chanda S, García-Sastre A, Schotsaert M, Teijaro JR, Moulton HM, Stein DA. Inhibition of SARS-CoV-2 growth in the lungs of mice by a peptide-conjugated morpholino oligomer targeting viral RNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102331. [PMID: 39376996 PMCID: PMC11456799 DOI: 10.1016/j.omtn.2024.102331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
Further development of direct-acting antiviral agents against human SARS-CoV-2 infections remains a public health priority. Here, we report that an antisense peptide-conjugated morpholino oligomer (PPMO) named 5'END-2, targeting a highly conserved sequence in the 5' UTR of SARS-CoV-2 genomic RNA, potently suppressed SARS-CoV-2 growth in vitro and in vivo. In HeLa-ACE 2 cells, 5'END-2 produced IC50 values of between 40 nM and 1.15 μM in challenges using six genetically disparate strains of SARS-CoV-2, including JN.1. In vivo, using K18-hACE2 mice and the WA-1/2020 virus isolate, two doses of 5'END-2 at 10 mg/kg, administered intranasally on the day before and the day after infection, produced approximately 1.4 log10 virus titer reduction in lung tissue at 3 days post-infection. Under a similar dosing schedule, intratracheal administration of 1.0-2.0 mg/kg 5'END-2 produced over 3.5 log10 virus growth suppression in mouse lungs. Electrophoretic mobility shift assays characterized specific binding of 5'END-2 to its complementary target RNA. Furthermore, using reporter constructs containing SARS-CoV-2 5' UTR leader sequence, in an in-cell system, we observed that 5'END-2 could interfere with translation in a sequence-specific manner. The results demonstrate that direct pulmonary delivery of 5'END-2 PPMO is a promising antiviral strategy against SARS-CoV-2 infections and warrants further development.
Collapse
Affiliation(s)
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mahsa Khoshbakht
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Scott Bittner
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Christiane V. Löhr
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Randy Diaz-Tapia
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kris White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Luke Le Luo
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Blanton Tolbert
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mario Blanco
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Amy Chow
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mitchell Guttman
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Cuiping Li
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
| | - Yiming Bao
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Joses Ho
- GISAID @ A∗STAR Bioinformatics Institute, Singapore 138632, Singapore
| | | | | | - Sumit Chanda
- Scripps Research Institute, La Jolla, CA 92037, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Hong M. Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - David A. Stein
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
2
|
Rihon J, Mattelaer CA, Montalvão RW, Froeyen M, Pinheiro VB, Lescrinier E. Structural insights into the morpholino nucleic acid/RNA duplex using the new XNA builder Ducque in a molecular modeling pipeline. Nucleic Acids Res 2024; 52:2836-2847. [PMID: 38412249 PMCID: PMC11014352 DOI: 10.1093/nar/gkae135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/19/2024] [Indexed: 02/29/2024] Open
Abstract
The field of synthetic nucleic acids with novel backbone structures [xenobiotic nucleic acids (XNAs)] has flourished due to the increased importance of XNA antisense oligonucleotides and aptamers in medicine, as well as the development of XNA processing enzymes and new XNA genetic materials. Molecular modeling on XNA structures can accelerate rational design in the field of XNAs as it contributes in understanding and predicting how changes in the sugar-phosphate backbone impact on the complementation properties of the nucleic acids. To support the development of novel XNA polymers, we present a first-in-class open-source program (Ducque) to build duplexes of nucleic acid analogs with customizable chemistry. A detailed procedure is described to extend the Ducque library with new user-defined XNA fragments using quantum mechanics (QM) and to generate QM-based force field parameters for molecular dynamics simulations within standard packages such as AMBER. The tool was used within a molecular modeling workflow to accurately reproduce a selection of experimental structures for nucleic acid duplexes with ribose-based as well as non-ribose-based nucleosides. Additionally, it was challenged to build duplexes of morpholino nucleic acids bound to complementary RNA sequences.
Collapse
Affiliation(s)
- Jérôme Rihon
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49, Box 1030, B-3000 Leuven, Belgium
| | - Charles-Alexandre Mattelaer
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49, Box 1030, B-3000 Leuven, Belgium
- Quantum Chemistry and Physical Chemistry, Celestijnenlaan 200f, Box 2404, B-3001, Leuven, Belgium
| | - Rinaldo Wander Montalvão
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49, Box 1030, B-3000 Leuven, Belgium
- Gain Therapeutics sucursal en España, Barcelona Science Park, Baldiri Reixac 4-10, 08028 Barcelona, Spain
| | - Mathy Froeyen
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49, Box 1030, B-3000 Leuven, Belgium
| | - Vitor Bernardes Pinheiro
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49, Box 1030, B-3000 Leuven, Belgium
| | - Eveline Lescrinier
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49, Box 1030, B-3000 Leuven, Belgium
| |
Collapse
|
3
|
Lessl AL, Pöhmerer J, Lin Y, Wilk U, Höhn M, Hörterer E, Wagner E, Lächelt U. mCherry on Top: A Positive Read-Out Cellular Platform for Screening DMD Exon Skipping Xenopeptide-PMO Conjugates. Bioconjug Chem 2023; 34:2263-2274. [PMID: 37991502 PMCID: PMC10739591 DOI: 10.1021/acs.bioconjchem.3c00408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023]
Abstract
Phosphorodiamidate morpholino oligomers (PMOs) are a special type of antisense oligonucleotides (ASOs) that can be used as therapeutic modulators of pre-mRNA splicing. Application of nucleic-acid-based therapeutics generally requires suitable delivery systems to enable efficient transport to intended tissues and intracellular targets. To identify potent formulations of PMOs, we established a new in vitro-in vivo screening platform based on mdx exon 23 skipping. Here, a new in vitro positive read-out system (mCherry-DMDEx23) is presented that is sensitive toward the PMO(Ex23) sequence mediating DMD exon 23 skipping and, in this model, functional mCherry expression. After establishment of the reporter system in HeLa cells, a set of amphiphilic, ionizable xenopeptides (XPs) was screened in order to identify potent carriers for PMO delivery. The identified best-performing PMO formulation with high splice-switching activity at nanomolar concentrations in vitro was then translated to in vivo trials, where exon 23 skipping in different organs of healthy BALB/c mice was confirmed. The predesigned in vitro-in vivo workflow enables evaluation of PMO(Ex23) carriers without change of the PMO sequence and formulation composition. Furthermore, the identified PMO-XP conjugate formulation was found to induce highly potent exon skipping in vitro and redistributed PMO activity in different organs in vivo.
Collapse
Affiliation(s)
- Anna-Lina Lessl
- Pharmaceutical
Biotechnology, Department of Pharmacy, LMU
Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Jana Pöhmerer
- Pharmaceutical
Biotechnology, Department of Pharmacy, LMU
Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Yi Lin
- Pharmaceutical
Biotechnology, Department of Pharmacy, LMU
Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ulrich Wilk
- Pharmaceutical
Biotechnology, Department of Pharmacy, LMU
Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Miriam Höhn
- Pharmaceutical
Biotechnology, Department of Pharmacy, LMU
Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Elisa Hörterer
- Pharmaceutical
Biotechnology, Department of Pharmacy, LMU
Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical
Biotechnology, Department of Pharmacy, LMU
Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
- Center
for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Ulrich Lächelt
- Pharmaceutical
Biotechnology, Department of Pharmacy, LMU
Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
- Center
for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
- Department
of Pharmaceutical Sciences, University of
Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| |
Collapse
|
4
|
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: 3.0] [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
|
5
|
Aslesh T, Erkut E, Ren J, Lim KRQ, Woo S, Hatlevig S, Moulton HM, Gosgnach S, Greer J, Maruyama R, Yokota T. DG9-conjugated morpholino rescues phenotype in SMA mice by reaching the CNS via a subcutaneous administration. JCI Insight 2023; 8:160516. [PMID: 36719755 PMCID: PMC10077475 DOI: 10.1172/jci.insight.160516] [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: 04/05/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Antisense oligonucleotide-mediated (AO-mediated) therapy is a promising strategy to treat several neurological diseases, including spinal muscular atrophy (SMA). However, limited delivery to the CNS with AOs administered intravenously or subcutaneously is a major challenge. Here, we demonstrate a single subcutaneous administration of cell-penetrating peptide DG9 conjugated to an AO called phosphorodiamidate morpholino oligomer (PMO) reached the CNS and significantly prolonged the median survival compared with unconjugated PMO and R6G-PMO in a severe SMA mouse model. Treated mice exhibited substantially higher expression of full-length survival of motor neuron 2 in both the CNS and systemic tissues compared with nontreated and unmodified AO-treated mice. The treatment ameliorated the atrophic musculature and improved breathing function accompanied by improved muscle strength and innervation at the neuromuscular junction with no signs of apparent toxicity. We also demonstrated DG9-conjugated PMO localized in nuclei in the spinal cord and brain after subcutaneous injections. Our data identify DG9 peptide conjugation as a powerful way to improve the efficacy of AO-mediated splice modulation. Finally, DG9-PMO is a promising therapeutic option to treat SMA and other neurological diseases, overcoming the necessity for intrathecal injections and treating body-wide tissues without apparent toxicity.
Collapse
Affiliation(s)
| | | | - Jun Ren
- Neuroscience and Mental Health Institute.,Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Susan Hatlevig
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Hong M Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Simon Gosgnach
- Neuroscience and Mental Health Institute.,Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John Greer
- Neuroscience and Mental Health Institute.,Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Toshifumi Yokota
- Neuroscience and Mental Health Institute.,Department of Medical Genetics, and
| |
Collapse
|
6
|
Shurpik RV, Shurpik DN, Gerasimov AV, Stoikov II. Modification of Silicon Dioxide with Variously Substituted minothiacalix[4]arenes: Organic−Inorganic Nanoparticles or Nucleic Acid Binding. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1070428022080103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
7
|
Kupryushkin MS, Filatov AV, Mironova NL, Patutina OA, Chernikov IV, Chernolovskaya EL, Zenkova MA, Pyshnyi DV, Stetsenko DA, Altman S, Vlassov VV. Antisense oligonucleotide gapmers containing phosphoryl guanidine groups reverse MDR1-mediated multiple drug resistance of tumor cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:211-226. [PMID: 34976439 PMCID: PMC8693280 DOI: 10.1016/j.omtn.2021.11.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/28/2021] [Indexed: 10/26/2022]
Abstract
Antisense gapmer oligonucleotides containing phosphoryl guanidine (PG) groups, e.g., 1,3-dimethylimidazolidin-2-imine, at three to five internucleotidic positions adjacent to the 3' and 5' ends were prepared via the Staudinger chemistry, which is compatible with conditions of standard automated solid-phase phosphoramidite synthesis for phosphodiester and, notably, phosphorothioate linkages, and allows one to design a variety of gapmeric structures with alternating linkages, and deoxyribose or 2'-O-methylribose backbone. PG modifications increased nuclease resistance in serum-containing medium for more than 21 days. Replacing two internucleotidic phosphates by PG groups in phosphorothioate-modified oligonucleotides did not decrease their cellular uptake in the absence of lipid carriers. Increasing the number of PG groups from two to seven per oligonucleotide reduced their ability to enter the cells in the carrier-free mode. Cationic liposomes provided similar delivery efficiency of both partially PG-modified and unmodified oligonucleotides. PG-gapmers were designed containing three to four PG groups at both wings and a central "window" of seven deoxynucleotides with either phosphodiester or phosphorothioate linkages targeted to MDR1 mRNA providing multiple drug resistance of tumor cells. Gapmers efficiently silenced MDR1 mRNA and restored the sensitivity of tumor cells to chemotherapeutics. Thus, PG-gapmers can be considered as novel, promising types of antisense oligonucleotides for targeting biologically relevant RNAs.
Collapse
Affiliation(s)
- Maxim S Kupryushkin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| | - Anton V Filatov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| | - Nadezhda L Mironova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| | - Olga A Patutina
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| | - Ivan V Chernikov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| | - Elena L Chernolovskaya
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| | - Marina A Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| | - Dmitrii V Pyshnyi
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| | - Dmitry A Stetsenko
- Department of Physics, Novosibirsk State University, Pirogov Str. 2, Novosibirsk 630090, Russia.,Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Lavrentiev Ave. 10, Novosibirsk 630090, Russia
| | - Sidney Altman
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA.,Life Sciences, Arizona State University, Tempe, AZ 85281, USA.,Montreal Clinical Research Institute, Montreal QC H2W 1R7, Canada
| | - Valentin V Vlassov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| |
Collapse
|
8
|
Bersani M, Rizzuti M, Pagliari E, Garbellini M, Saccomanno D, Moulton HM, Bresolin N, Comi GP, Corti S, Nizzardo M. Cell-penetrating peptide-conjugated Morpholino rescues SMA in a symptomatic preclinical model. Mol Ther 2022; 30:1288-1299. [PMID: 34808387 PMCID: PMC8899506 DOI: 10.1016/j.ymthe.2021.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 07/07/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a motor neuron disease and the leading genetic cause of infant mortality. Recently approved SMA therapies have transformed a deadly disease into a survivable one, but these compounds show a wide spectrum of clinical response and effective rescue only in the early stages of the disease. Therefore, safe, symptomatic-suitable, non-invasive treatments with high clinical impact across different phenotypes are urgently needed. We conjugated antisense oligonucleotides with Morpholino (MO) chemistry, which increase SMN protein levels, to cell-penetrating peptides (CPPs) for better cellular distribution. Systemically administered MOs linked to r6 and (RXRRBR)2XB peptides crossed the blood-brain barrier and increased SMN protein levels remarkably, causing striking improvement of survival, neuromuscular function, and neuropathology, even in symptomatic SMA animals. Our study demonstrates that MO-CPP conjugates can significantly expand the therapeutic window through minimally invasive systemic administration, opening the path for clinical applications of this strategy.
Collapse
Affiliation(s)
- Margherita Bersani
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Mafalda Rizzuti
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| | - Elisa Pagliari
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Manuela Garbellini
- Healthcare Professionals Department - Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Domenica Saccomanno
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| | - Hong M. Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Nereo Bresolin
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy,Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| | - Giacomo P. Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy,Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy,Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy,Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| | - Monica Nizzardo
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy.
| |
Collapse
|
9
|
Potential of cell-penetrating peptides (CPPs) in delivery of antiviral therapeutics and vaccines. Eur J Pharm Sci 2021; 169:106094. [PMID: 34896590 DOI: 10.1016/j.ejps.2021.106094] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
Viral infections are a great threat to human health. Currently, there are no effective vaccines and antiviral drugs against the majority of viral diseases, suggesting the need to develop novel and effective antiviral agents. Since the intracellular delivery of antiviral agents, particularly the impermeable molecules, such as peptides, proteins, and nucleic acids, are essential to exert their therapeutic effects, using a delivery system is highly required. Among various delivery systems, cell-penetrating peptides (CPPs), a group of short peptides with the unique ability of crossing cell membrane, offer great potential for the intracellular delivery of various biologically active cargoes. The results of numerous in vitro and in vivo studies with CPP conjugates demonstrate their promise as therapeutic agents in various medical fields including antiviral therapy. The CPP-mediated delivery of various antiviral agents including peptides, proteins, nucleic acids, and nanocarriers have been associated with therapeutic efficacy both in vitro and in vivo. This review describes various aspects of viruses including their biology, pathogenesis, and therapy and briefly discusses the concept of CPP and its potential in drug delivery. Particularly, it will highlight a variety of CPP applications in the management of viral infections.
Collapse
|
10
|
Liu J, Guo ZN, Yan XL, Yang Y, Huang S. Brain Pathogenesis and Potential Therapeutic Strategies in Myotonic Dystrophy Type 1. Front Aging Neurosci 2021; 13:755392. [PMID: 34867280 PMCID: PMC8634727 DOI: 10.3389/fnagi.2021.755392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/20/2021] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy that affects multiple systems including the muscle and heart. The mutant CTG expansion at the 3'-UTR of the DMPK gene causes the expression of toxic RNA that aggregate as nuclear foci. The foci then interfere with RNA-binding proteins, affecting hundreds of mis-spliced effector genes, leading to aberrant alternative splicing and loss of effector gene product functions, ultimately resulting in systemic disorders. In recent years, increasing clinical, imaging, and pathological evidence have indicated that DM1, though to a lesser extent, could also be recognized as true brain diseases, with more and more researchers dedicating to develop novel therapeutic tools dealing with it. In this review, we summarize the current advances in the pathogenesis and pathology of central nervous system (CNS) deficits in DM1, intervention measures currently being investigated are also highlighted, aiming to promote novel and cutting-edge therapeutic investigations.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Xiu-Li Yan
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Shuo Huang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
11
|
Markowska A, Markowski AR, Jarocka-Karpowicz I. The Importance of 6-Aminohexanoic Acid as a Hydrophobic, Flexible Structural Element. Int J Mol Sci 2021; 22:12122. [PMID: 34830000 PMCID: PMC8618066 DOI: 10.3390/ijms222212122] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 11/17/2022] Open
Abstract
6-aminohexanoic acid is an ω-amino acid with a hydrophobic, flexible structure. Although the ω-amino acid in question is mainly used clinically as an antifibrinolytic drug, other applications are also interesting and important. This synthetic lysine derivative, without an α-amino group, plays a significant role in chemical synthesis of modified peptides and in the polyamide synthetic fibers (nylon) industry. It is also often used as a linker in various biologically active structures. This review concentrates on the role of 6-aminohexanoic acid in the structure of various molecules.
Collapse
Affiliation(s)
- Agnieszka Markowska
- Department of Analytical Chemistry, Medical University of Bialystok, 15-089 Bialystok, Poland;
| | - Adam Roman Markowski
- Department of Internal Medicine and Gastroenterology, Polish Red Cross Memorial Municipal Hospital, 79 Henryk Sienkiewicz Street, 15-003 Bialystok, Poland;
| | - Iwona Jarocka-Karpowicz
- Department of Analytical Chemistry, Medical University of Bialystok, 15-089 Bialystok, Poland;
| |
Collapse
|
12
|
Klabenkova K, Fokina A, Stetsenko D. Chemistry of Peptide-Oligonucleotide Conjugates: A Review. Molecules 2021; 26:5420. [PMID: 34500849 PMCID: PMC8434111 DOI: 10.3390/molecules26175420] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/25/2022] Open
Abstract
Peptide-oligonucleotide conjugates (POCs) represent one of the increasingly successful albeit costly approaches to increasing the cellular uptake, tissue delivery, bioavailability, and, thus, overall efficiency of therapeutic nucleic acids, such as, antisense oligonucleotides and small interfering RNAs. This review puts the subject of chemical synthesis of POCs into the wider context of therapeutic oligonucleotides and the problem of nucleic acid drug delivery, cell-penetrating peptide structural types, the mechanisms of their intracellular transport, and the ways of application, which include the formation of non-covalent complexes with oligonucleotides (peptide additives) or covalent conjugation. The main strategies for the synthesis of POCs are viewed in detail, which are conceptually divided into (a) the stepwise solid-phase synthesis approach and (b) post-synthetic conjugation either in solution or on the solid phase, especially by means of various click chemistries. The relative advantages and disadvantages of both strategies are discussed and compared.
Collapse
Affiliation(s)
- Kristina Klabenkova
- Faculty of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia; (K.K.); (D.S.)
- Institute of Cytology and Genetics, Russian Academy of Sciences, Siberian Branch, 630090 Novosibirsk, Russia
| | - Alesya Fokina
- Faculty of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia; (K.K.); (D.S.)
- Institute of Cytology and Genetics, Russian Academy of Sciences, Siberian Branch, 630090 Novosibirsk, Russia
| | - Dmitry Stetsenko
- Faculty of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia; (K.K.); (D.S.)
- Institute of Cytology and Genetics, Russian Academy of Sciences, Siberian Branch, 630090 Novosibirsk, Russia
| |
Collapse
|
13
|
A targeted antisense therapeutic approach for Hutchinson-Gilford progeria syndrome. Nat Med 2021; 27:536-545. [PMID: 33707773 PMCID: PMC10158310 DOI: 10.1038/s41591-021-01274-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare accelerated aging disorder characterized by premature death from myocardial infarction or stroke. It is caused by de novo single-nucleotide mutations in the LMNA gene that activate a cryptic splice donor site, resulting in the production of a toxic form of lamin A, which is termed progerin. Here we present a potential genetic therapeutic strategy that utilizes antisense peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) to block pathogenic splicing of mutant transcripts. Of several candidates, PPMO SRP-2001 provided the most significant decrease in progerin transcripts in patient fibroblasts. Intravenous delivery of SRP-2001 to a transgenic mouse model of HGPS produced significant reduction of progerin transcripts in the aorta, a particularly critical target tissue in HGPS. Long-term continuous treatment with SRP-2001 yielded a 61.6% increase in lifespan and rescue of vascular smooth muscle cell loss in large arteries. These results provide a rationale for proceeding to human trials.
Collapse
|
14
|
Facile Preparation of PNA-Peptide Conjugates with a Polar Maleimide-Thioether Linkage. Methods Mol Biol 2021; 2105:97-118. [PMID: 32088866 DOI: 10.1007/978-1-0716-0243-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Conjugation of a delivery peptide containing a thiol functionality (e.g., a cysteine residue) with a PNA oligomer displaying a single unprotected aliphatic primary amine (e.g., the N-terminus or a C-terminal lysine residue) can be achieved via a one-pot modification with a bisfunctional maleimide linker also displaying a reactive N-hydroxysuccinimidyl ester group (e.g., Mal-PEG2-OSu). Here, an optimized protocol with respect to ratios between the reactants as well as recommended reaction times is presented. Formation and conversion of the maleimide-PNA intermediate was followed by analytical HPLC as exemplified by its conjugation to (KFF)3K-Cys-NH2. In addition, the reaction time required for direct conversion of a preformed Mal-(CH2)2-(C=O)-PNA oligomer in the presence of a slight excess of thiol-modified peptide (with a varying degree of sterical hindrance: HS-(CH2)2-CONH-(KFF)3K-NH2, (KFF)3K-hCys-NH2 and (KFF)3K-Cys-NH2) is provided.
Collapse
|
15
|
Varnamkhasti BS, Jafari S, Taghavi F, Alaei L, Izadi Z, Lotfabadi A, Dehghanian M, Jaymand M, Derakhshankhah H, Saboury AA. Cell-Penetrating Peptides: As a Promising Theranostics Strategy to Circumvent the Blood-Brain Barrier for CNS Diseases. Curr Drug Deliv 2020; 17:375-386. [DOI: 10.2174/1567201817666200415111755] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/09/2019] [Accepted: 03/18/2020] [Indexed: 12/14/2022]
Abstract
The passage of therapeutic molecules across the Blood-Brain Barrier (BBB) is a profound challenge for the management of the Central Nervous System (CNS)-related diseases. The ineffectual nature of traditional treatments for CNS disorders led to the abundant endeavor of researchers for the design the effective approaches in order to bypass BBB during recent decades. Cell-Penetrating Peptides (CPPs) were found to be one of the promising strategies to manage CNS disorders. CPPs are short peptide sequences with translocation capacity across the biomembrane. With special regard to their two key advantages like superior permeability as well as low cytotoxicity, these peptide sequences represent an appropriate solution to promote therapeutic/theranostic delivery into the CNS. This scenario highlights CPPs with specific emphasis on their applicability as a novel theranostic delivery system into the brain.
Collapse
Affiliation(s)
- Behrang Shiri Varnamkhasti
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical, Sciences, Kermanshah, Iran
| | - Samira Jafari
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical, Sciences, Kermanshah, Iran
| | - Fereshteh Taghavi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Loghman Alaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical, Sciences, Kermanshah, Iran
| | - Zhila Izadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical, Sciences, Kermanshah, Iran
| | - Alireza Lotfabadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical, Sciences, Kermanshah, Iran
| | - Mojtaba Dehghanian
- Department of Biotechnology, Shahr-e Kord Branch, Islamic Azad University, Shahr-e Kord, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical, Sciences, Kermanshah, Iran
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
16
|
Peptide Nucleic Acids: Applications in Biomedical Sciences. Molecules 2020; 25:molecules25153317. [PMID: 32707859 PMCID: PMC7435668 DOI: 10.3390/molecules25153317] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
The DNA mimic, PNA (peptide nucleic acid), has been with us now for almost 3 decades [...].
Collapse
|
17
|
Honcharenko M, Honcharenko D, Strömberg R. Attachment of Peptides to Oligonucleotides on Solid Support Using Copper(I)-Catalyzed Huisgen 1,3-Dipolar Cycloaddition. Methods Mol Biol 2020; 2036:165-171. [PMID: 31410796 DOI: 10.1007/978-1-4939-9670-4_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In vivo bioavailability and delivery of nucleic acids to the site of action is a severe limitation in oligonucleotide (ON) therapeutics. Equipping the ONs with cell penetrating, homing or endosomal escape peptides can enhance specificity and/or uptake efficiencies. We describe here a general procedure for the preparation of peptide-oligonucleotide conjugates (POCs) on solid support utilizing a novel activated alkyne containing linker which enhances the Cu(I) catalyzed Huisgen 1,3-dipolar cycloaddition. Conjugation reaction is efficient in millimolar concentration and submicromolar amounts at ambient temperature. The route for POC preparation involves two subsequent conjugation steps: to solid-supported ONs containing a 5'-amino modifier (1) the triple bond donor (p-(N-propynoylamino)toluic acid (PATA), p-([2-(propynyloxy)acetamido]methyl)benzoic acid (PAMBA) or 2-(propynyloxy)acetic acid (PAA)) is first coupled and then (2) an azido-functionalized peptide is attached via a triazole linkage by copper(I) catalyzed Huisgen 1,3-dipolar cycloaddition. The fragment-conjugated POC is released from the solid support by concentrated ammonia. The method gives high conversion of ON to the POC and only involves a single purification step after complete assembly and release from the solid support. The synthesis is flexible and designed to utilize commercially available oligonucleotide and peptide derivatives without the need for specific automated synthesizers.
Collapse
Affiliation(s)
| | - Dmytro Honcharenko
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Roger Strömberg
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
18
|
Hammond SM, Abendroth F, Gait MJ, Wood MJA. Evaluation of Cell-Penetrating Peptide Delivery of Antisense Oligonucleotides for Therapeutic Efficacy in Spinal Muscular Atrophy. Methods Mol Biol 2020; 2036:221-236. [PMID: 31410800 DOI: 10.1007/978-1-4939-9670-4_13] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Antisense oligonucleotides (ASOs) are a widely used form of gene therapy, which is translatable to multiple disorders. A major obstacle for ASO efficacy is its bioavailability for in vivo and in vitro studies. To overcome this challenge we use cell-penetrating peptides (CPPs) for systemic delivery of ASOs. One of the most advanced clinical uses of ASOs is for the treatment of spinal muscular atrophy (SMA). In this chapter, we describe the techniques used for in vitro screening and analysing in vivo biodistribution of CPP-conjugated ASOs targeting the survival motor neuron 2, SMN2, the dose-dependent modifying gene for SMA.
Collapse
Affiliation(s)
- Suzan M Hammond
- Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Frank Abendroth
- Laboratory of Molecular Biology, Medical Research Council, Cambridge, UK
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg-University of Mainz, Staudingerweg 5, D-55128, Mainz, Germany
| | - Michael J Gait
- Laboratory of Molecular Biology, Medical Research Council, Cambridge, UK
| | | |
Collapse
|
19
|
Stasińska AR, Putaj P, Chmielewski MK. Disulfide bridge as a linker in nucleic acids' bioconjugation. Part II: A summary of practical applications. Bioorg Chem 2019; 95:103518. [PMID: 31911308 DOI: 10.1016/j.bioorg.2019.103518] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/11/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022]
Abstract
Disulfide conjugation invariably remains a key tool in research on nucleic acids. This versatile and cost-effective method plays a crucial role in structural studies of DNA and RNA as well as their interactions with other macromolecules in a variety of biological systems. In this article we review applications of disulfide-bridged conjugates of oligonucleotides with other (bio)molecules such as peptides, proteins etc. and present key findings obtained with their help.
Collapse
Affiliation(s)
- Anna R Stasińska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, ul. Noskowskiego 12/14, 61-704 Poznań, Poland; FutureSynthesis sp. z o.o. ul. Rubież 46H, 61-612 Poznań, Poland
| | - Piotr Putaj
- FutureSynthesis sp. z o.o. ul. Rubież 46H, 61-612 Poznań, Poland
| | - Marcin K Chmielewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, ul. Noskowskiego 12/14, 61-704 Poznań, Poland; FutureSynthesis sp. z o.o. ul. Rubież 46H, 61-612 Poznań, Poland.
| |
Collapse
|
20
|
Li Z, Buck M. Computational Design of Myristoylated Cell-Penetrating Peptides Targeting Oncogenic K-Ras.G12D at the Effector-Binding Membrane Interface. J Chem Inf Model 2019; 60:306-315. [DOI: 10.1021/acs.jcim.9b00690] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
21
|
Kardani K, Milani A, H Shabani S, Bolhassani A. Cell penetrating peptides: the potent multi-cargo intracellular carriers. Expert Opin Drug Deliv 2019; 16:1227-1258. [PMID: 31583914 DOI: 10.1080/17425247.2019.1676720] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Cell penetrating peptides (CPPs) known as protein translocation domains (PTD), membrane translocating sequences (MTS), or Trojan peptides (TP) are able to cross biological membranes without clear toxicity using different mechanisms, and facilitate the intracellular delivery of a variety of bioactive cargos. CPPs could overcome some limitations of drug delivery and combat resistant strains against a broad range of diseases. Despite delivery of different therapeutic molecules by CPPs, they lack cell specificity and have a short duration of action. These limitations led to design of combined cargo delivery systems and subsequently improvement of their clinical applications. Areas covered: This review covers all our studies and other researchers in different aspects of CPPs such as classification, uptake mechanisms, and biomedical applications. Expert opinion: Due to low cytotoxicity of CPPs as compared to other carriers and final degradation to amino acids, they are suitable for preclinical and clinical studies. Generally, the efficiency of CPPs was suitable to penetrate the cell membrane and deliver different cargos to specific intracellular sites. However, no CPP-based therapeutic approach has approved by FDA, yet; because there are some disadvantages for CPPs including short half-life in blood, and nonspecific CPP-mediated delivery to normal tissue. Thus, some methods were used to develop the functions of CPPs in vitro and in vivo including the augmentation of cell specificity by activatable CPPs, specific transport into cell organelles by insertion of corresponding localization sequences, incorporation of CPPs into multifunctional dendrimeric or liposomal nanocarriers to improve selectivity and efficiency especially in tumor cells.
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Samaneh H Shabani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| |
Collapse
|
22
|
Soudah T, Khawaled S, Aqeilan RI, Yavin E. AntimiR-155 Cyclic Peptide-PNA Conjugate: Synthesis, Cellular Uptake, and Biological Activity. ACS OMEGA 2019; 4:13954-13961. [PMID: 31497713 PMCID: PMC6714607 DOI: 10.1021/acsomega.9b01697] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/25/2019] [Indexed: 06/10/2023]
Abstract
Efficient delivery of nucleic acids into cells still remains a great challenge. Peptide nucleic acids (PNAs) are DNA analogues with a neutral backbone and are synthesized by solid phase peptide chemistry. This allows a straightforward synthetic route to introduce a linear short peptide (a.k.a. cell-penetrating peptide) to the PNA molecule as a means of facilitating cellular uptake of PNAs. Herein, we have devised a synthetic route in which a cyclic peptide is prepared on a solid support and is extended with the PNA molecule, where all syntheses are accomplished on the solid phase. This allows the conjugation of the cyclic peptide to the PNA molecule with the need of only one purification step after the cyclic peptide-PNA conjugate (C9-PNA) is cleaved from the solid support. The PNA sequence chosen is an antimiR-155 molecule that is complementary to mature miR-155, a well-established oncogenic miRNA. By labeling C9-PNA with fluorescein isothiocyanate, we observe efficient cellular uptake into glioblastoma cells (U87MG) at a low concentration (0.5 μM), as corroborated by fluorescence-activated cell sorting (FACS) analysis and confocal microscopy. FACS analysis also suggests an uptake mechanism that is energy-dependent. Finally, the antimiR activity of C9-PNA was shown by analyzing miR155 levels by quantitative reverse transcription polymerase chain reaction and by observing a reduction in cell viability and proliferation in U87MG cells, as corroborated by XTT and colony formation assays. Given the added biological stability of cyclic versus linear peptides, this synthetic approach may be a useful and straightforward approach to synthesize cyclic peptide-PNA conjugates.
Collapse
Affiliation(s)
- Terese Soudah
- The
Institute for Drug Research, The School of Pharmacy,
and Lautenberg Center
for Immunology and Cancer Research, Institute for Medical Research
Israel-Canada, The Hebrew University of
Jerusalem, Hadassah Ein-Kerem, Jerusalem 9112102, Israel
| | - Saleh Khawaled
- The
Institute for Drug Research, The School of Pharmacy,
and Lautenberg Center
for Immunology and Cancer Research, Institute for Medical Research
Israel-Canada, The Hebrew University of
Jerusalem, Hadassah Ein-Kerem, Jerusalem 9112102, Israel
| | - Rami I. Aqeilan
- The
Institute for Drug Research, The School of Pharmacy,
and Lautenberg Center
for Immunology and Cancer Research, Institute for Medical Research
Israel-Canada, The Hebrew University of
Jerusalem, Hadassah Ein-Kerem, Jerusalem 9112102, Israel
| | - Eylon Yavin
- The
Institute for Drug Research, The School of Pharmacy,
and Lautenberg Center
for Immunology and Cancer Research, Institute for Medical Research
Israel-Canada, The Hebrew University of
Jerusalem, Hadassah Ein-Kerem, Jerusalem 9112102, Israel
| |
Collapse
|
23
|
Nguyen KV. Potential epigenomic co-management in rare diseases and epigenetic therapy. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2019; 38:752-780. [PMID: 31079569 DOI: 10.1080/15257770.2019.1594893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The purpose of this review is to highlight the impact of the alternative splicing process on human disease. Epigenetic regulation determines not only what parts of the genome are expressed but also how they are spliced. The recent progress in the field of epigenetics has important implications for the study of rare diseases. The role of epigenetics in rare diseases is a key issue in molecular physiology and medicine because not only rare diseases can benefit from epigenetic research, but can also provide useful principles for other common and complex disorders such as cancer, cardiovascular, type 2 diabetes, obesity, and neurological diseases. Predominantly, epigenetic modifications include DNA methylation, histone modification, and RNA-associated silencing. These modifications in the genome regulate numerous cellular activities. Disruption of epigenetic regulation process can contribute to the etiology of numerous diseases during both prenatal and postnatal life. Here, I discuss current knowledge about this matter including some current epigenetic therapies and future directions in the field by emphasizing on the RNA-based therapy via antisense oligonucleotides to correct splicing defects.
Collapse
Affiliation(s)
- Khue Vu Nguyen
- a Department of Medicine, Biochemical Genetics and Metabolism, The Mitochondrial and Metabolic Disease Center, School of Medicine, University of California, San Diego , San Diego , CA , USA.,b Department of Pediatrics, UC San Diego School of Medicine , La Jolla , CA , USA
| |
Collapse
|
24
|
MacCulloch T, Buchberger A, Stephanopoulos N. Emerging applications of peptide-oligonucleotide conjugates: bioactive scaffolds, self-assembling systems, and hybrid nanomaterials. Org Biomol Chem 2019; 17:1668-1682. [PMID: 30483688 DOI: 10.1039/c8ob02436g] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Peptide-oligonucleotide conjugates (POCs) are covalent constructs that link a molecule like DNA to a synthetic peptide sequences. These materials merge the programmable self-assembly of oligonucleotides with the bioactivity and chemical diversity of polypeptides. Recent years have seen the widespread use of POCs in a range of fields, driven the by relative advantages of each molecular type. In this review, we will present an overview of the synthesis and application of POCs, with an emphasis on emerging areas where these molecules will have a unique impact. We first discuss two main strategies for synthesizing POCs from synthetic monomers such as phosphoramidites and functionalized amino acids. We then describe four key fields of research in POCs: (1) biomaterials for interfacing with, and controlling the behavior of cells; (2) hybrid self-assembling systems that balance peptide and oligonucleotide intermolecular forces; (3) template-enhanced coupling of POCs into larger molecules; and (4) display of peptides on self-assembled oligonucleotide scaffolds. We also highlight several promising areas for future applications in each of these four directions, and anticipate ever increasing uses of POCs in interdisciplinary research.
Collapse
Affiliation(s)
- Tara MacCulloch
- School of Molecular Sciences, Arizona State University, Tempe AZ, USA.
| | | | | |
Collapse
|
25
|
Hansen AM, Bonke G, Hogendorf WFJ, Björkling F, Nielsen J, Kongstad KT, Zabicka D, Tomczak M, Urbas M, Nielsen PE, Franzyk H. Microwave-assisted solid-phase synthesis of antisense acpP peptide nucleic acid-peptide conjugates active against colistin- and tigecycline-resistant E. coli and K. pneumoniae. Eur J Med Chem 2019; 168:134-145. [PMID: 30807888 DOI: 10.1016/j.ejmech.2019.02.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 02/05/2019] [Accepted: 02/07/2019] [Indexed: 11/26/2022]
Abstract
Recent discovery of potent antibacterial antisense PNA-peptide conjugates encouraged development of a fast and efficient synthesis protocol that facilitates structure-activity studies. The use of an Fmoc/Boc protection scheme for both PNA monomers and amino acid building blocks in combination with microwave-assisted solid-phase synthesis proved to be a convenient procedure for continuous assembly of antisense PNA-peptide conjugates. A validated antisense PNA oligomer (CTCATACTCT; targeting mRNA of the acpP gene) was linked to N-terminally modified drosocin (i.e., RXR-PRPYSPRPTSHPRPIRV; X = aminohexanoic acid) or to a truncated Pip1 peptide (i.e., RXRRXR-IKILFQNRRMKWKK; X = aminohexanoic acid), and determination of the antibacterial effects of the resulting conjugates allowed assessment of the influence of different linkers as well as differences between the L- and D-forms of the peptides. The drosocin-derived compound without a linker moiety exhibited highest antibacterial activity against both wild-type Escherichia coli and Klebsiella pneumoniae (MICs in the range 2-4 μg/mL ∼ 0.3-0.7 μM), while analogues displaying an ethylene glycol (eg1) moiety or a polar maleimide linker also possessed activity toward wild-type K. pneumoniae (MICs of 4-8 μg/mL ∼ 0.6-1.3 μM). Against two colistin-resistant E. coli strains the linker-deficient compound proved most potent (with MICs in the range 2-4 μg/mL ∼ 0.3-0.7 μM). The truncated all-L Pip1 peptide had moderate inherent activity against E. coli, and this was unaltered or reduced upon conjugation to the antisense PNA oligomer. By contrast, this peptide was 8-fold less potent against K. pneumoniae, but in this case some PNA-peptide conjugates exhibited potent antisense activity (MICs of 2-8 μg/mL ∼ 0.3-1.2 μM). Most interestingly, the antibacterial activity of the D-form peptide itself was 2- to 16-fold higher than that of the L-form, even for the colistin- and tigecycline-resistant E. coli strains (MIC of 1-2 μg/mL ∼ 0.25-0.5 μM). Low activity was found for conjugates with a two-mismatch PNA sequence corroborating an antisense mode of action. Conjugates containing a D-form peptide were also significantly less active. In conclusion, we have designed and synthesized antisense PNA-drosocin conjugates with potent antibacterial activity against colistin- and tigecycline-resistant E. coli and K. pneumonia without concomitant hemolytic properties. In addition, a truncated D-form of Pip1 was identified as a peptide exhibiting potent activity against both wild-type and multidrug-resistant E. coli, P. aeruginosa, and A. baumannii (MICs within the range 1-4 μg/mL ∼ 0.25-1 μM) as well as toward wild-type Staphylococcus aureus (MIC of 2-4 μg/mL ∼ 0.5-1.0 μM).
Collapse
Affiliation(s)
- Anna Mette Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Denmark
| | - Gitte Bonke
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Denmark
| | - Wouter Frederik Johan Hogendorf
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Denmark
| | - Fredrik Björkling
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Denmark
| | - John Nielsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Denmark
| | - Kenneth T Kongstad
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Denmark
| | - Dorota Zabicka
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, ul. Chełmska 30/34, 00-725, Warsaw, Poland
| | - Magdalena Tomczak
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, ul. Chełmska 30/34, 00-725, Warsaw, Poland
| | - Malgorzata Urbas
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, ul. Chełmska 30/34, 00-725, Warsaw, Poland
| | - Peter E Nielsen
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2100, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Denmark.
| |
Collapse
|
26
|
Nan Y, Zhang YJ. Antisense Phosphorodiamidate Morpholino Oligomers as Novel Antiviral Compounds. Front Microbiol 2018; 9:750. [PMID: 29731743 PMCID: PMC5920040 DOI: 10.3389/fmicb.2018.00750] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/03/2018] [Indexed: 01/23/2023] Open
Abstract
Phosphorodiamidate morpholino oligomers (PMO) are short single-stranded DNA analogs that are built upon a backbone of morpholine rings connected by phosphorodiamidate linkages. As uncharged nucleic acid analogs, PMO bind to complementary sequences of target mRNA by Watson–Crick base pairing to block protein translation through steric blockade. PMO interference of viral protein translation operates independently of RNase H. Meanwhile, PMO are resistant to a variety of enzymes present in biologic fluids, a characteristic that makes them highly suitable for in vivo applications. Notably, PMO-based therapy for Duchenne muscular dystrophy (DMD) has been approved by the United States Food and Drug Administration which is now a hallmark for PMO-based antisense therapy. In this review, the development history of PMO, delivery methods for improving cellular uptake of neutrally charged PMO molecules, past studies of PMO antagonism against RNA and DNA viruses, PMO target selection, and remaining questions of PMO antiviral strategies are discussed in detail and new insights are provided.
Collapse
Affiliation(s)
- Yuchen Nan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, MD, United States
| | - Yan-Jin Zhang
- Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, MD, United States
| |
Collapse
|
27
|
Wolfe JM, Fadzen CM, Holden RL, Yao M, Hanson GJ, Pentelute* BL. Perfluoroaryl Bicyclic Cell-Penetrating Peptides for Delivery of Antisense Oligonucleotides. Angew Chem Int Ed Engl 2018; 57:4756-4759. [PMID: 29479836 PMCID: PMC6248909 DOI: 10.1002/anie.201801167] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Indexed: 12/12/2022]
Abstract
Exon-skipping antisense oligonucleotides are effective treatments for genetic diseases, yet exon-skipping activity requires that these macromolecules reach the nucleus. While cell-penetrating peptides can improve delivery, proteolytic instability often limits efficacy. It is hypothesized that the bicyclization of arginine-rich peptides would improve their stability and their ability to deliver oligonucleotides into the nucleus. Two methods were introduced for the synthesis of arginine-rich bicyclic peptides using cysteine perfluoroarylation chemistry. Then, the bicyclic peptides were covalently linked to a phosphorodiamidate morpholino oligonucleotide (PMO) and assayed for exon skipping activity. The perfluoroaryl cyclic and bicyclic peptides improved PMO activity roughly 14-fold over the unconjugated PMO. The bicyclic peptides exhibited increased proteolytic stability relative to the monocycle, demonstrating that perfluoroaryl bicyclic peptides are potent and stable delivery agents.
Collapse
Affiliation(s)
- Justin M. Wolfe
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA),
| | - Colin M. Fadzen
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA),
| | - Rebecca L. Holden
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA),
| | - Monica Yao
- Research Chemistry, Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142 (USA)
| | - Gunnar J. Hanson
- Research Chemistry, Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142 (USA)
| | - Bradley L. Pentelute*
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA),
| |
Collapse
|
28
|
Wolfe JM, Fadzen CM, Holden RL, Yao M, Hanson GJ, Pentelute BL. Perfluoroaryl Bicyclic Cell‐Penetrating Peptides for Delivery of Antisense Oligonucleotides. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201801167] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Justin M. Wolfe
- Department of Chemistry Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Colin M. Fadzen
- Department of Chemistry Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Rebecca L. Holden
- Department of Chemistry Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Monica Yao
- Research Chemistry Sarepta Therapeutics, Inc. 215 First Street Cambridge MA 02142 USA
| | - Gunnar J. Hanson
- Research Chemistry Sarepta Therapeutics, Inc. 215 First Street Cambridge MA 02142 USA
| | - Bradley L. Pentelute
- Department of Chemistry Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| |
Collapse
|
29
|
van der Bent ML, Paulino da Silva Filho O, van Luijk J, Brock R, Wansink DG. Assisted delivery of antisense therapeutics in animal models of heritable neurodegenerative and neuromuscular disorders: a systematic review and meta-analysis. Sci Rep 2018; 8:4181. [PMID: 29520012 PMCID: PMC5843643 DOI: 10.1038/s41598-018-22316-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
Antisense oligonucleotide (AON)-based therapies hold promise for a range of neurodegenerative and neuromuscular diseases and have shown benefit in animal models and patients. Success in the clinic is nevertheless still limited, due to unfavourable biodistribution and poor cellular uptake of AONs. Extensive research is currently being conducted into the formulation of AONs to improve delivery, but thus far there is no consensus on which of those strategies will be the most effective. This systematic review was designed to answer in an unbiased manner which delivery strategies most strongly enhance the efficacy of AONs in animal models of heritable neurodegenerative and neuromuscular diseases. In total, 95 primary studies met the predefined inclusion criteria. Study characteristics and data on biodistribution and toxicity were extracted and reporting quality and risk of bias were assessed. Twenty studies were eligible for meta-analysis. We found that even though the use of delivery systems provides an advantage over naked AONs, it is not yet possible to select the most promising strategies. Importantly, standardisation of experimental procedures is warranted in order to reach conclusions about the most efficient delivery strategies. Our best practice guidelines for future experiments serve as a step in that direction.
Collapse
Affiliation(s)
- M Leontien van der Bent
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
| | - Omar Paulino da Silva Filho
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
- CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Judith van Luijk
- Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE), Department of Health Evidence, Radboud university medical center, Nijmegen, The Netherlands
| | - Roland Brock
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
| | - Derick G Wansink
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands.
| |
Collapse
|
30
|
Xue XY, Mao XG, Zhou Y, Chen Z, Hu Y, Hou Z, Li MK, Meng JR, Luo XX. Advances in the delivery of antisense oligonucleotides for combating bacterial infectious diseases. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:745-758. [PMID: 29341934 DOI: 10.1016/j.nano.2017.12.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/12/2017] [Accepted: 12/31/2017] [Indexed: 12/22/2022]
Abstract
Discovery and development of new antibacterial drugs against multidrug resistant bacterial strains have become more and more urgent. Antisense oligonucleotides (ASOs) show immense potential to control the spread of resistant microbes due to its high specificity of action, little risk to human gene expression, and easy design and synthesis to target any possible gene. However, efficient delivery of ASOs to their action sites with enough concentration remains a major obstacle, which greatly hampers their clinical application. In this study, we reviewed current progress on delivery strategies of ASOs into bacteria, focused on various non-virus gene vectors, including cell penetrating peptides, lipid nanoparticles, bolaamphiphile-based nanoparticles, DNA nanostructures and Vitamin B12. The current review provided comprehensive understanding and novel perspective for the future application of ASOs in combating bacterial infections.
Collapse
Affiliation(s)
- Xiao-Yan Xue
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, China.
| | - Xing-Gang Mao
- Department of Neurosurgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ying Zhou
- Department of Pharmacology, Xi'an Medical University, Xi'an, China
| | - Zhou Chen
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yue Hu
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Zheng Hou
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Ming-Kai Li
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Jing-Ru Meng
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Xiao-Xing Luo
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
31
|
Phumesin P, Junking M, Panya A, Yongpitakwattana P, Noisakran S, Limjindaporn T, Yenchitsomanus PT. Vivo-morpholino oligomers strongly inhibit dengue virus replication and production. Arch Virol 2017; 163:867-876. [PMID: 29260328 DOI: 10.1007/s00705-017-3666-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 11/16/2017] [Indexed: 01/07/2023]
Abstract
Dengue virus (DENV) infection is a worldwide public health problem, which can cause severe dengue hemorrhagic fever (DHF) and life-threatening dengue shock syndrome (DSS). There are currently no anti-DENV drugs available, and there has been an intensive search for effective anti-DENV agents that can inhibit all four DENV serotypes. In this study, we tested whether vivo-morpholino oligomers (vivo-MOs), whose effect on DENV infection has not previously been studied, can inhibit DENV infection. Vivo-MOs were designed to target the top of 3' stem-loop (3' SL) in the 3' UTR of the DENV genome and tested for inhibition of DENV infection in monkey kidney epithelial (Vero) cells and human lung epithelial carcinoma (A549) cells. The results showed that vivo-MOs could bind to a DENV RNA sequence and markedly reduce DENV-RNA, protein, and virus production in infected Vero and A549 cells. Vivo-MOs at a concentration of 4 µM could inhibit DENV production by more than 104-fold when compared to that of an untreated control. In addition, vivo-MOs also inhibited DENV production in U937 cells and primary human monocytes. Therefore, vivo-MOs targeting to the 3' SL in the 3' UTR of DENV genomes are effective and have the potential to be developed as anti-DENV agents.
Collapse
Affiliation(s)
- Patta Phumesin
- Siriraj Center of Research Excellence for Molecular Medicine (SiCORE-MM), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Mutita Junking
- Siriraj Center of Research Excellence for Molecular Medicine (SiCORE-MM), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Aussara Panya
- Siriraj Center of Research Excellence for Molecular Medicine (SiCORE-MM), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Petlada Yongpitakwattana
- Siriraj Center of Research Excellence for Molecular Medicine (SiCORE-MM), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sansanee Noisakran
- Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok, 10700, Thailand
| | - Thawornchai Limjindaporn
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Molecular Medicine (SiCORE-MM), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
32
|
Soudah T, Mogilevsky M, Karni R, Yavin E. CLIP6-PNA-Peptide Conjugates: Non-Endosomal Delivery of Splice Switching Oligonucleotides. Bioconjug Chem 2017; 28:3036-3042. [PMID: 29211451 DOI: 10.1021/acs.bioconjchem.7b00638] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Efficient delivery of oligonucleotides still remains a challenge in the field of oligonucleotide based therapy. Peptide nucleic acid (PNA), a DNA analogue that is typically synthesized by solid phase peptide chemistry, has been conjugated to a variety of cell penetrating peptides (CPP) as a means of improving its cellular uptake. These CPPs typically deliver their cargoes into cells by an endosomal-dependent mechanism resulting in lower bioavailability of the cargo. Herein, we designed and synthesized PNA-peptide conjugates as splice switching oligonucleotides (SSO) targeting the Mnk2 gene, a therapeutic target in cancer. In humans, the MKNK2 gene, is alternatively spliced, generating isoforms with opposite biological activities: Mnk2a and Mnk2b. It was found that the Mnk2a isoform is down-regulated in breast, lung, brain, and colon tumors and is a tumor suppressor, whereas MnK2b is oncogenic. We have designed and synthesized PNAs that were conjugated to either of the following peptides: a nuclear localization sequence (NLS) or a cytosol localizing internalization peptide (CLIP6). CLIP6-PNA demonstrates effective cellular uptake and exclusively employs a nonendosomal mechanism to cross the cellular membranes of glioblastoma cells (U87). Simple incubation of PNA-peptide conjugates in human glioblastoma cells up-regulates the Mnk2a isoform leading to cancer cell death.
Collapse
Affiliation(s)
- Terese Soudah
- The Institute for Drug Research, The School of Pharmacy and ‡Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, The Faculty of Medicine, The Hebrew University of Jerusalem , Hadassah Ein-Kerem, Jerusalem 91120, Israel
| | - Maxim Mogilevsky
- The Institute for Drug Research, The School of Pharmacy and ‡Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, The Faculty of Medicine, The Hebrew University of Jerusalem , Hadassah Ein-Kerem, Jerusalem 91120, Israel
| | - Rotem Karni
- The Institute for Drug Research, The School of Pharmacy and ‡Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, The Faculty of Medicine, The Hebrew University of Jerusalem , Hadassah Ein-Kerem, Jerusalem 91120, Israel
| | - Eylon Yavin
- The Institute for Drug Research, The School of Pharmacy and ‡Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, The Faculty of Medicine, The Hebrew University of Jerusalem , Hadassah Ein-Kerem, Jerusalem 91120, Israel
| |
Collapse
|
33
|
Jiang K, Gao X, Shen Q, Zhan C, Zhang Y, Xie C, Wei G, Lu W. Discerning the composition of penetratin for safe penetration from cornea to retina. Acta Biomater 2017; 63:123-134. [PMID: 28927928 DOI: 10.1016/j.actbio.2017.09.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/12/2017] [Accepted: 09/15/2017] [Indexed: 12/13/2022]
Abstract
Delivery of biomacromolecules into the eye is greatly hindered by several protective barriers. The cell-penetrating peptide, penetratin, has been found to be an effective absorption enhancer for noninvasive intraocular gene delivery. To discern the composition of penetratin for safe penetration from cornea to retina, we designed a series of penetratin derivatives by varying the hydrophobicity and evaluated their potency for retina-targeted delivery. The hydrophilic amino acids of penetratin, excluding the conserved basic amino acid residues, were respectively replaced with tryptophan. Secondary structure of the resultant derivatives was analyzed by computer simulation and circular dichroism, exhibiting that the hydrophobic derivatives had a propensity to form high content of helix and entered corneal and conjunctival cells more easily than did penetratin. As expected, the hydrophobic derivatives showed improved permeability in excised rabbit cornea and sclera, and kept intact after penetration. When instilled topically in the conjunctival sac of mice eyes, the hydrophobic derivatives distributed safely and rapidly into both cornea and retina, with increased amount and prolonged retention time in comparison to penetratin. In conclusion, we demonstrated that the ocular permeability of penetratin derivatives closely correlated with their hydrophobicity, and introducing hydrophobic amino acids in penetratin was a feasible approach to develop more powerful ocular absorption enhancers. STATEMENT OF SIGNIFICANCE Due to the defensive barriers of the eye, efficient and safe absorption enhancers are indispensable for noninvasive delivery of exogenous biomacromolecules to the posterior segment. In this manuscript, we designed a series of penetratin derivatives and validated they had significantly improved penetration ability from cornea to retina than wild-type penetratin, without increasing toxicity. More importantly, we provided a sequence of solid evidences that the ocular permeability of penetratin derivatives closely correlated with their hydrophobicity, and introducing hydrophobic amino acids in penetratin was a feasible approach to develop more powerful ocular absorption enhancers. We also demonstrated that the penetratin derivatives permeated through cornea and sclera with intact structure, and might enter the eye by non-corneal pathway.
Collapse
|
34
|
Kolevzon N, Hashoul D, Naik S, Rubinstein A, Yavin E. Single point mutation detection in living cancer cells by far-red emitting PNA-FIT probes. Chem Commun (Camb) 2016; 52:2405-7. [PMID: 26735489 DOI: 10.1039/c5cc07502e] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peptide nucleic acid bis-quinoline conjugates are reported as attractive far-red emitting probes that detect mutated mRNA in living cells at SNP resolution.
Collapse
Affiliation(s)
- N Kolevzon
- The Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Hadassah Ein-Kerem, Jerusalem 91120, Israel.
| | - D Hashoul
- The Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Hadassah Ein-Kerem, Jerusalem 91120, Israel.
| | - S Naik
- The Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Hadassah Ein-Kerem, Jerusalem 91120, Israel.
| | - A Rubinstein
- The Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Hadassah Ein-Kerem, Jerusalem 91120, Israel.
| | - E Yavin
- The Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Hadassah Ein-Kerem, Jerusalem 91120, Israel.
| |
Collapse
|
35
|
Systemic peptide-mediated oligonucleotide therapy improves long-term survival in spinal muscular atrophy. Proc Natl Acad Sci U S A 2016; 113:10962-7. [PMID: 27621445 DOI: 10.1073/pnas.1605731113] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The development of antisense oligonucleotide therapy is an important advance in the identification of corrective therapy for neuromuscular diseases, such as spinal muscular atrophy (SMA). Because of difficulties of delivering single-stranded oligonucleotides to the CNS, current approaches have been restricted to using invasive intrathecal single-stranded oligonucleotide delivery. Here, we report an advanced peptide-oligonucleotide, Pip6a-morpholino phosphorodiamidate oligomer (PMO), which demonstrates potent efficacy in both the CNS and peripheral tissues in severe SMA mice following systemic administration. SMA results from reduced levels of the ubiquitously expressed survival motor neuron (SMN) protein because of loss-of-function mutations in the SMN1 gene. Therapeutic splice-switching oligonucleotides (SSOs) modulate exon 7 splicing of the nearly identical SMN2 gene to generate functional SMN protein. Pip6a-PMO yields SMN expression at high efficiency in peripheral and CNS tissues, resulting in profound phenotypic correction at doses an order-of-magnitude lower than required by standard naked SSOs. Survival is dramatically extended from 12 d to a mean of 456 d, with improvement in neuromuscular junction morphology, down-regulation of transcripts related to programmed cell death in the spinal cord, and normalization of circulating insulin-like growth factor 1. The potent systemic efficacy of Pip6a-PMO, targeting both peripheral as well as CNS tissues, demonstrates the high clinical potential of peptide-PMO therapy for SMA.
Collapse
|
36
|
Pendergraff HM, Debacker AJ, Watts JK. Single-Stranded Silencing RNAs: Hit Rate and Chemical Modification. Nucleic Acid Ther 2016; 26:216-22. [PMID: 27123752 DOI: 10.1089/nat.2015.0557] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Single-stranded silencing RNAs (ss-siRNAs) are chemically modified single-stranded oligomers that engage the RNA interference machinery normally used by duplex RNAs to silence gene expression. ss-siRNAs have the potential to combine advantages of antisense oligonucleotides and siRNAs. Previous work has explored the chemistry of the phosphate and the oligonucleotide body. We now describe the process of attempting to develop and optimize ss-siRNAs based on five active siRNA duplexes. Three of the sequences failed to show any activity as ss-siRNAs, and in two of those cases the ss-siRNAs showed significantly increased toxicity relative to the parent duplexes. For the two sequences that did work well as ss-siRNAs, we show that the chemistry of the 3'-terminal dinucleotide also has a significant effect on the potency of ss-siRNAs. Previously published work on ss-siRNAs has been based on a 2'-O-methoxyethyl-RNA (MOE) dinucleotide at the 3'-terminus. To our surprise, oligomers containing 2'-O-Me-RNA modifications at the 3'-terminus showed significantly improved potency and activity relative to those modified with MOE at the same sites. Oligonucleotides with two locked nucleic acid units at the 3'-terminus showed improved activity over the MOE-modified analog for one sequence. Importantly, the fact that 2'-O-Me-RNA works so well makes the ss-siRNA approach accessible to a wider range of researchers since it can be achieved with inexpensive commercially available modifications.
Collapse
Affiliation(s)
- Hannah M Pendergraff
- 1 Department of Chemistry and Institute for Life Sciences, University of Southampton , United Kingdom
| | - Alexandre J Debacker
- 1 Department of Chemistry and Institute for Life Sciences, University of Southampton , United Kingdom .,2 RNA Therapeutics Institute and Department of Biochemistry and Molecular Pharmacology, UMass Medical School, Worcester, Massachusetts
| | - Jonathan K Watts
- 1 Department of Chemistry and Institute for Life Sciences, University of Southampton , United Kingdom .,2 RNA Therapeutics Institute and Department of Biochemistry and Molecular Pharmacology, UMass Medical School, Worcester, Massachusetts
| |
Collapse
|
37
|
Delayed Time-to-Treatment of an Antisense Morpholino Oligomer Is Effective against Lethal Marburg Virus Infection in Cynomolgus Macaques. PLoS Negl Trop Dis 2016; 10:e0004456. [PMID: 26901785 PMCID: PMC4764691 DOI: 10.1371/journal.pntd.0004456] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/22/2016] [Indexed: 11/19/2022] Open
Abstract
Marburg virus (MARV) is an Ebola-like virus in the family Filovirdae that causes sporadic outbreaks of severe hemorrhagic fever with a case fatality rate as high as 90%. AVI-7288, a positively charged antisense phosphorodiamidate morpholino oligomer (PMOplus) targeting the viral nucleoprotein gene, was evaluated as a potential therapeutic intervention for MARV infection following delayed treatment of 1, 24, 48, and 96 h post-infection (PI) in a nonhuman primate lethal challenge model. A total of 30 cynomolgus macaques were divided into 5 groups of 6 and infected with 1,830 plaque forming units of MARV subcutaneously. AVI-7288 was administered by bolus infusion daily for 14 days at 15 mg/kg body weight. Survival was the primary endpoint of the study. While none (0 of 6) of the saline group survived, 83–100% of infected monkeys survived when treatment was initiated 1, 24, 48, or 96 h post-infection (PI). The antisense treatment also reduced serum viremia and inflammatory cytokines in all treatment groups compared to vehicle controls. The antibody immune response to virus was preserved and tissue viral antigen was cleared in AVI-7288 treated animals. These data show that AVI-7288 protects NHPs against an otherwise lethal MARV infection when treatment is initiated up to 96 h PI. Marburg virus (MARV) is a filovirus closely related to Ebola virus and similarly causes hemorrhagic fever in humans. MARV is endemic throughout parts of tropical Africa. Severe outbreaks of Marburg virus disease (MVD) have occurred involving hundreds of human cases. No effective MARV antiviral therapies are available. In this study, we used a positive charged phosphorodiamidate morpholino oligomer (PMOplus) targeting the mRNA of the MARV nucleoprotein gene as a medical countermeasure to treat disease in a lethal nonhuman primate model of MVD. The intravenous treatment regimen was well tolerated with no treatment related adverse effects. We showed that when using this antisense treatment, serum virus levels decreased and 83–100% of the animals survived, even when the treatment was delayed as much as 96 hours after infection. None of the untreated animals survived the viral challenge in this model. Our results suggest that antisense therapies, such as PMOs, hold great promise for the treatment of severe viral diseases such as MVD.
Collapse
|
38
|
Margus H, Arukuusk P, Langel Ü, Pooga M. Characteristics of Cell-Penetrating Peptide/Nucleic Acid Nanoparticles. Mol Pharm 2015; 13:172-9. [PMID: 26561739 DOI: 10.1021/acs.molpharmaceut.5b00598] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nucleic acids are highly promising candidates for the treatment of various genetic diseases. However, due to the large size and negative charge, nucleic acids are not efficiently taken up by cells, and thus, their clinical potential remains limited so far. Therefore, various delivery vehicles have been designed to assist the cellular uptake of nucleic acids. Among these, cell-penetrating peptides (CPPs) have gained increasing popularity as efficient and nontoxic delivery vectors. CPPs can be coupled to nucleic acids either by covalent or noncovalent association. Noncovalent coupling, which is based on the formation of nanoparticle-like nanocomplexes (NP), has received much attention in recent years, and the number of studies employing the strategy is explosively increasing due to the high therapeutic potential. However, the properties of CPP/nucleic acid NPs have not been characterized in sufficient detail yet. We performed a comprehensive analysis of the size and morphology of nucleic acid nanoparticles with novel transfection peptides, PepFects (PFs) and NickFects (NFs), using negative staining transmission electron microscopy (TEM). In addition, we examined whether the attachment of fluorescence or (nano)gold label to nucleic acid affects the nanocomplex formation or its morphology. We demonstrated that transportan-10-based new generation CPPs from PF and NF families condense nucleic acids to NPs of homogeneous size and shape. The size and shape of assembled nanoparticles depend on the type of the complexed nucleic acid and the sequence of the used peptide, whereas the label on the nucleic acid does not influence the gross characteristics of formed NPs.
Collapse
Affiliation(s)
- Helerin Margus
- Institute of Molecular and Cell Biology, University of Tartu , Tartu 51010, Estonia
| | - Piret Arukuusk
- Institute of Technology, University of Tartu , Tartu 50411, Estonia
| | - Ülo Langel
- Institute of Technology, University of Tartu , Tartu 50411, Estonia.,Department of Neurochemistry, Stockholm University , S-10691 Stockholm, Sweden
| | - Margus Pooga
- Institute of Molecular and Cell Biology, University of Tartu , Tartu 51010, Estonia
| |
Collapse
|
39
|
Oligonucleotide therapeutics: chemistry, delivery and clinical progress. Future Med Chem 2015; 7:2221-42. [PMID: 26510815 DOI: 10.4155/fmc.15.144] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oligonucleotide therapeutics have the potential to become a third pillar of drug development after small molecules and protein therapeutics. However, the three approved oligonucleotide drugs over the past 17 years have not proven to be highly successful in a commercial sense. These trailblazer drugs have nonetheless laid the foundations for entire classes of drug candidates to follow. This review will examine further advances in chemistry that are earlier in the pipeline of oligonucleotide drug candidates. Finally, we consider the possible effect of delivery systems that may provide extra footholds to improve the potency and specificity of oligonucleotide drugs. Our overview focuses on strategies to imbue antisense oligonucleotides with more drug-like properties and their applicability to other nucleic acid therapeutics.
Collapse
|
40
|
Lehto T, Wagner E. Sequence-defined polymers for the delivery of oligonucleotides. Nanomedicine (Lond) 2015; 9:2843-59. [PMID: 25535686 DOI: 10.2217/nnm.14.166] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Short synthetic oligonucleotides (ONs) are a group of therapeutic molecules with enormous clinical potential owing to their high specificity and ability to target the expression of virtually any single or group of genes. Clinical translation of ONs is hampered by the inadequate bioavailability in the target cells due to the substantial extracellular and intracellular barriers exposed to these molecules. Different cationic polymers have been successfully deployed for the delivery of ONs. However, heterogeneous nature of these classical polymers is not suitable for clinical applications and hence vectors with completely defined structure are required. In this review, we discuss recent advances with sequence-defined polymers and their application for the delivery of short ONs.
Collapse
Affiliation(s)
- Taavi Lehto
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Munich, Germany
| | | |
Collapse
|
41
|
Raucher D, Ryu JS. Cell-penetrating peptides: strategies for anticancer treatment. Trends Mol Med 2015; 21:560-70. [PMID: 26186888 DOI: 10.1016/j.molmed.2015.06.005] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/12/2015] [Accepted: 06/16/2015] [Indexed: 01/09/2023]
Abstract
Cell-penetrating peptides (CPP) provide an efficient strategy for the intracellular delivery of bioactive molecules in various biomedical applications. This review focuses on recent advances in the use of CPPs to deliver anticancer therapeutics and imaging reagents to cancer cells, along with CPP contributions to novel tumor-targeting techniques. CPPs are now used extensively to deliver a variety of therapeutics, despite lacking cell specificity and having a short duration of action. Resolution of these shortcomings to enable increased cancer cell and/or tumor specificity could improve CPP-based drug delivery strategies, expand combined drug delivery possibilities, and strengthen future clinical applications of these peptides.
Collapse
Affiliation(s)
- Drazen Raucher
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Jung Su Ryu
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
42
|
Banerjee A, Bagmare S, Varada M, Kumar VA. Glycine-Linked Nucleoside-β-Amino Acids: Polyamide Analogues of Nucleic Acids. Bioconjug Chem 2015; 26:1737-42. [PMID: 26076350 DOI: 10.1021/acs.bioconjchem.5b00296] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
3'-5'-Deoxyribose-sugar-phoshate backbone in DNA is completely replaced by 2'-deoxyribonucleoside-based β-amino acids interlinked by glycine to create uncharged polyamide DNA with 3'-5'-directionality. These oligomers as conjugates of α-amino acids and nucleoside-β-amino acids bind strongly and sequence-specifically only to the antiparallel complementary RNA and DNA.
Collapse
Affiliation(s)
- Anjan Banerjee
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Pune, India 411008
| | - Seema Bagmare
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Pune, India 411008
| | - Manojkumar Varada
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Pune, India 411008
| | - Vaijayanti A Kumar
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Pune, India 411008
| |
Collapse
|
43
|
siRNA suppression of hTERT using activatable cell-penetrating peptides in hepatoma cells. Biosci Rep 2015; 35:BSR20140145. [PMID: 25671640 PMCID: PMC4370094 DOI: 10.1042/bsr20140145] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Activatable cell-penetrating peptides (aCPPs) allow non-viral, low cytotoxic and selective delivery of compounds into target cells for cancer therapy. In tumour cells, up-regulation of human telomerase reverse transcriptase (hTERT) frequently occurs and is being considered as a target in cancer diagnosis and treatment. siRNA sequence that target hTERT mRNA can silence the gene and reduce hTERT protein expression to reduce cell proliferation and inhibit cell growth. In our study, we tested a matrix metalloproteinase-2 (MPP2) aCPP in delivering hTERT siRNA into hepatocellular carcinoma cells (SMMC-7721) to silence the hTERT gene. Cultured SMMC-7721 cells were transfected with a complex of aCPPs and hTERT-specific siRNA-encoding or control plasmids. Compared with cells treated with the complex of control plasmid–CPPs, cells treated with the hTERT-specific siRNA-encoding plasmid–CPP complex had a prolonged G1-phase, but a shorter G2/S-phase, indicating a G1-arrest. Treatment with the hTERT-specific siRNA resulted in a significant decrease (by 26%; P<0.05) in hTERT mRNA levels. The aCPPs tested in this study provides a non-viral delivery of siRNA into cancer cells to silence target genes in cancer therapy. In the present study, we delivered human telomerase reverse transcriptase (hTERT) siRNA into SMMC-7721 hepatoma cells using a matrix metalloproteinase-2 (MMP2)-activatable cell-penetrating peptide (aCPP). The siRNA subsequently induced down-regulation of the hTERT gene and G1-arrest, implicating the utility of this delivery system in cancer therapy.
Collapse
|
44
|
Nablo BJ, Panchal RG, Bavari S, Nguyen TL, Gussio R, Ribot W, Friedlander A, Chabot D, Reiner JE, Robertson JWF, Balijepalli A, Halverson KM, Kasianowicz JJ. Anthrax toxin-induced rupture of artificial lipid bilayer membranes. J Chem Phys 2014; 139:065101. [PMID: 23947891 DOI: 10.1063/1.4816467] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We demonstrate experimentally that anthrax toxin complexes rupture artificial lipid bilayer membranes when isolated from the blood of infected animals. When the solution pH is temporally acidified to mimic that process in endosomes, recombinant anthrax toxin forms an irreversibly bound complex, which also destabilizes membranes. The results suggest an alternative mechanism for the translocation of anthrax toxin into the cytoplasm.
Collapse
Affiliation(s)
- Brian J Nablo
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899-8120, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
A survey on "Trojan Horse" peptides: opportunities, issues and controlled entry to "Troy". J Control Release 2014; 194:53-70. [PMID: 25151981 DOI: 10.1016/j.jconrel.2014.08.014] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 12/31/2022]
Abstract
Cell-penetrating peptides (CPPs), often vividly termed as the "Trojan Horse" peptides, have attracted considerable interest for the intracellular delivery of a wide range of cargoes, such as small molecules, peptides, proteins, nucleic acids, contrast agents, nanocarriers and so on. Some preclinical and clinical developments of CPP conjugates demonstrate their promise as therapeutic agents for drug discovery. There is increasing evidence to suggest that CPPs have the potential to cross several bio-barriers (e.g., blood-brain barriers, intestinal mucosa, nasal mucosa and skin barriers). Despite revolutionary process in many aspects, there are a lot of basic issues unclear for these entities, such as internalization mechanisms, translocation efficiency, translocation kinetics, metabolic degradation, toxicity, side effect, distribution and non-specificity. Among them, non-specificity remains a major drawback for the in vivo application of CPPs in the targeted delivery of cargoes. So far, diverse organelle-specific CPPs or controlled delivery strategies have emerged and improved their specificity. In this review, we will look at the opportunities of CPPs in clinical development, bio-barriers penetration and nanocarriers delivery. Then, a series of basic problems of CPPs will be discussed. Finally, this paper will highlight the use of various controlled strategies in the organelle-specific delivery and targeted delivery of CPPs. The purpose of this review will be to emphasize most influential advance in this field and present a fundamental understanding for challenges and utilizations of CPPs. This will accelerate their translation as efficient vectors from the in vitro setting into the clinic arena, and retrieve the entry art to "Troy".
Collapse
|
46
|
Cordier C, Boutimah F, Bourdeloux M, Dupuy F, Met E, Alberti P, Loll F, Chassaing G, Burlina F, Saison-Behmoaras TE. Delivery of antisense peptide nucleic acids to cells by conjugation with small arginine-rich cell-penetrating peptide (R/W)9. PLoS One 2014; 9:e104999. [PMID: 25127364 PMCID: PMC4134252 DOI: 10.1371/journal.pone.0104999] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/14/2014] [Indexed: 12/28/2022] Open
Abstract
Peptide nucleic acids (PNAs) are very attractive antisense and antigene agents, but these molecules are not passively taken into cells. Here, using a functional cell assay and fluorescent-based methods, we investigated cell uptake and antisense activity of a tridecamer PNA that targets the HIV-1 polypurine tract sequence delivered using the arginine-rich (R/W)9 peptide (RRWWRRWRR). At micromolar concentrations, without use of any transfection agents, almost 80% inhibition of the target gene expression was obtained with the conjugate in the presence of the endosomolytic agent chloroquine. We show that chloroquine not only induced escape from endosomes but also enhanced the cellular uptake of the conjugate. Mechanistic studies revealed that (R/W)9-PNA conjugates were internalized via pinocytosis. Replacement of arginines with lysines reduced the uptake of the conjugate by six-fold, resulting in the abolition of intracellular target inhibition. Our results show that the arginines play a crucial role in the conjugate uptake and antisense activity. To determine whether specificity of the interactions of arginines with cell surface proteoglycans result in the internalization, we used flow cytometry to examine uptake of arginine- and lysine-rich conjugates in wild-type CHO-K1 and proteoglycan-deficient A745 cells. The uptake of both conjugates was decreased by four fold in CHO-745 cells; therefore proteoglycans promote internalization of cationic peptides, irrespective of the chemical nature of their positive charges. Our results show that arginine-rich cell-penetrating peptides, especially (R/W)9, are a promising tool for PNA internalization.
Collapse
Affiliation(s)
- Céline Cordier
- Muséum National d’Histoire Naturelle, Structure et Instabilité des Génomes, Paris, France
- INSERM, U1154, Paris, France
- CNRS, UMR 7196, Paris, France
| | - Fatima Boutimah
- Muséum National d’Histoire Naturelle, Structure et Instabilité des Génomes, Paris, France
- INSERM, U1154, Paris, France
- CNRS, UMR 7196, Paris, France
| | - Mathilde Bourdeloux
- Muséum National d’Histoire Naturelle, Structure et Instabilité des Génomes, Paris, France
- INSERM, U1154, Paris, France
- CNRS, UMR 7196, Paris, France
| | - Florian Dupuy
- Muséum National d’Histoire Naturelle, Structure et Instabilité des Génomes, Paris, France
- INSERM, U1154, Paris, France
- CNRS, UMR 7196, Paris, France
| | - Elisabeth Met
- Muséum National d’Histoire Naturelle, Structure et Instabilité des Génomes, Paris, France
- INSERM, U1154, Paris, France
- CNRS, UMR 7196, Paris, France
| | - Patrizia Alberti
- Muséum National d’Histoire Naturelle, Structure et Instabilité des Génomes, Paris, France
- INSERM, U1154, Paris, France
- CNRS, UMR 7196, Paris, France
| | - François Loll
- Muséum National d’Histoire Naturelle, Structure et Instabilité des Génomes, Paris, France
- INSERM, U1154, Paris, France
- CNRS, UMR 7196, Paris, France
| | - Gérard Chassaing
- UPMC-Univ Paris 06, Laboratoire des BioMolécules, Université P. et M. Curie, Paris, France
- CNRS, UMR 7203, Paris, France
- ENS, UMR 7203, Département de Chimie, Ecole Normale Supérieure, Paris, France
| | - Fabienne Burlina
- UPMC-Univ Paris 06, Laboratoire des BioMolécules, Université P. et M. Curie, Paris, France
- CNRS, UMR 7203, Paris, France
- ENS, UMR 7203, Département de Chimie, Ecole Normale Supérieure, Paris, France
| | - Tula Ester Saison-Behmoaras
- Muséum National d’Histoire Naturelle, Structure et Instabilité des Génomes, Paris, France
- INSERM, U1154, Paris, France
- CNRS, UMR 7196, Paris, France
- * E-mail:
| |
Collapse
|
47
|
Deuss PJ, Arzumanov AA, Williams DL, Gait MJ. Parallel synthesis and splicing redirection activity of cell-penetrating peptide conjugate libraries of a PNA cargo. Org Biomol Chem 2013; 11:7621-30. [PMID: 24105028 PMCID: PMC4002126 DOI: 10.1039/c3ob41659c] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/01/2013] [Indexed: 11/21/2022]
Abstract
A novel method for the parallel synthesis of peptide-biocargo conjugates was developed that utilizes affinity purification for fast isolation of the conjugates in order to avoid time consuming HPLC purification. The methodology was applied to create two libraries of cell-penetrating peptide (CPP)-PNA705 conjugates from parallel-synthesized peptide libraries. The conjugates were tested for their ability to induce splicing redirection in HeLa pLuc705 cells. The results demonstrate how the novel methodology can be applied for screening purposes in order to find suitable CPP-biocargo combinations and further optimization of CPPs.
Collapse
Affiliation(s)
- Peter J. Deuss
- Medical Research Council , Laboratory of Molecular Biology , Cambridge Biomedical Campus , Francis Crick Avenue , Cambridge , CB2 0QH , UK .
| | - Andrey A. Arzumanov
- Medical Research Council , Laboratory of Molecular Biology , Cambridge Biomedical Campus , Francis Crick Avenue , Cambridge , CB2 0QH , UK .
| | - Donna L. Williams
- Medical Research Council , Laboratory of Molecular Biology , Cambridge Biomedical Campus , Francis Crick Avenue , Cambridge , CB2 0QH , UK .
| | - Michael J. Gait
- Medical Research Council , Laboratory of Molecular Biology , Cambridge Biomedical Campus , Francis Crick Avenue , Cambridge , CB2 0QH , UK .
| |
Collapse
|
48
|
Järver P, O'Donovan L, Gait MJ. A chemical view of oligonucleotides for exon skipping and related drug applications. Nucleic Acid Ther 2013; 24:37-47. [PMID: 24171481 DOI: 10.1089/nat.2013.0454] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Peter Järver
- Medical Research Council , Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | | |
Collapse
|
49
|
Rodrigues M, de la Torre BG, Andreu D, Santos NC. Kinetic uptake profiles of cell penetrating peptides in lymphocytes and monocytes. Biochim Biophys Acta Gen Subj 2013; 1830:4554-63. [DOI: 10.1016/j.bbagen.2013.05.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 05/07/2013] [Accepted: 05/14/2013] [Indexed: 12/26/2022]
|
50
|
Influence of the insertion of a cationic peptide on the size and shape of nanoliposomes: A light scattering investigation. Int J Pharm 2013; 454:621-4. [DOI: 10.1016/j.ijpharm.2013.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/30/2013] [Accepted: 05/03/2013] [Indexed: 01/17/2023]
|