1
|
Ngalle Loth A, Maroquenne M, Medjmedj A, Coste F, Bizien T, Pichon C, Logeart-Avramoglou D, Perche F. Structural and functional characterization of a histidylated liposome for mRNA delivery. J Control Release 2025; 379:164-176. [PMID: 39788374 DOI: 10.1016/j.jconrel.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The development of lipid-based mRNA delivery systems has significantly facilitated recent advances in mRNA-based therapeutics. Liposomes, as the pioneering class of mRNA vectors, continue to lead in clinical trials. We previously developed a histidylated liposome that demonstrated efficient nucleic acid delivery. In this study, the liposome preparation process was optimized by freeze-drying followed by extrusion to homogenize size distribution and improve storage stability. A comprehensive characterization of these LYX liposomes was performed, including evaluation in cellular and murine animal models. LYX liposomes can be stored for up to one year at 4 °C, maintaining a stable size (150 ± 10 nm) and polydispersity index (0.10 ± 0.02), while preserving their transfection efficacy. They exhibit high encapsulation efficacy (∼95 %) and protect mRNA from RNase degradation. Lamellar organization was confirmed by Small Angle X-ray Scattering and CryoTEM, and intracellular trafficking was examined using confocal microscopy. LYX-mRNA lipoplexes can transfect both cell lines and primary cells, albeit with a lower transfection efficacy compared to the commercial Lipofectamine MessengerMAX™ vector. Our data suggest that this could be attributed to slower cell uptake and reduced endosomal escape of LYX. LYX liposomes effectively delivered mRNA encoding therapeutic BMP2 and BMP9 molecules, producing significant amounts of functional proteins that successfully induced BMP signaling. In addition, in vivo studies demonstrated the potential of LYX lipoplexes when incorporated into hydrogels and implanted subcutaneously in mice. These findings provided evidence that LYX liposomes are a promising platform for mRNA delivery, offering versatility for multiple applications.
Collapse
Affiliation(s)
| | - Manon Maroquenne
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, 75010, Paris, France
| | - Ayoub Medjmedj
- Centre de Biophysique Moléculaire, CBM, CNRS UPR4301, Orléans, France
| | - Franck Coste
- Centre de Biophysique Moléculaire, CBM, CNRS UPR4301, Orléans, France
| | - Thomas Bizien
- Université Paris-Saclay, Synchrotron Soleil, 91190 Saint-Aubin, France
| | - Chantal Pichon
- Inserm UMS 55 ART ARNm and LI2RSO, Université d'Orléans, F-45100 Orléans, France; Institut Universitaire de France, 1 rue Descartes, F-75035 Paris, France
| | | | - Federico Perche
- Centre de Biophysique Moléculaire, CBM, CNRS UPR4301, Orléans, France.
| |
Collapse
|
2
|
Zhang Y, Béland LC, Roussel S, Bertrand N, Hébert SS, Vallières L. Optimization of a lipid nanoparticle-based protocol for RNA transfection into primary mononuclear phagocytes. J Leukoc Biol 2024; 115:1165-1176. [PMID: 38466819 DOI: 10.1093/jleuko/qiae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/07/2024] [Accepted: 02/01/2024] [Indexed: 03/13/2024] Open
Abstract
The effective delivery of synthetic RNA into mononuclear phagocytes is a prerequisite for experimental research and therapeutic development. However, traditional methods are highly ineffective and toxic for these cells. Here, we aimed to optimize a transfection protocol for primary bone marrow-derived phagocytes, specifically dendritic cells and macrophages, using lipid nanoparticles generated by microfluidics. Our results show that a lipid mixture similar to that used in Moderna's COVID-19 messenger RNA vaccine outperforms the others tested. Improved messenger RNA transfection can be achieved by replacing uridine with methylpseudouridine but not methoxyuridine, which interferes with transfection. The addition of diphenyleneiodonium or apocynin can enhance transfection in a cell type-dependent manner without adverse effects, while apolipoprotein E provides no added value. These optimized transfection conditions can also be used for microRNA agonists and antagonists. In sum, this study offers a straightforward, highly efficient, reproducible, and nontoxic protocol to deliver RNA into different primary mononuclear phagocytes in culture.
Collapse
Affiliation(s)
- Yu Zhang
- Neuroscience Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Louis-Charles Béland
- Neuroscience Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Sabrina Roussel
- Endocrinology and Nephrology Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Nicolas Bertrand
- Endocrinology and Nephrology Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Sébastien S Hébert
- Neuroscience Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| | - Luc Vallières
- Neuroscience Unit, University Hospital Center of Quebec-Laval University, 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada
| |
Collapse
|
3
|
Steffens RC, Folda P, Fendler NL, Höhn M, Bücher-Schossau K, Kempter S, Snyder NL, Hartmann L, Wagner E, Berger S. GalNAc- or Mannose-PEG-Functionalized Polyplexes Enable Effective Lectin-Mediated DNA Delivery. Bioconjug Chem 2024; 35:351-370. [PMID: 38440876 DOI: 10.1021/acs.bioconjchem.3c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
A cationic, dendrimer-like oligo(aminoamide) carrier with four-arm topology based on succinoyl tetraethylene pentamine and histidines, cysteines, and N-terminal azido-lysines was screened for plasmid DNA delivery on various cell lines. The incorporated azides allow modification with various shielding agents of different polyethylene glycol (PEG) lengths and/or different ligands by copper-free click reaction, either before or after polyplex formation. Prefunctionalization was found to be advantageous over postfunctionalization in terms of nanoparticle formation, stability, and efficacy. A length of 24 ethylene oxide repetition units and prefunctionalization of ≥50% of azides per carrier promoted optimal polyplex shielding. PEG shielding resulted in drastically reduced DNA transfer, which could be successfully restored by active lectin targeting via novel GalNAc or mannose ligands, enabling enhanced receptor-mediated endocytosis of the carrier system. The involvement of the asialoglycoprotein receptor (ASGPR) in the uptake of GalNAc-functionalized polyplexes was confirmed in the ASGPR-positive hepatocarcinoma cell lines HepG2 and Huh7. Mannose-modified polyplexes showed superior cellular uptake and transfection efficacy compared to unmodified and shielded polyplexes in mannose-receptor-expressing dendritic cell-like DC2.4 cells.
Collapse
Affiliation(s)
- Ricarda C Steffens
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Paul Folda
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Nikole L Fendler
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Katharina Bücher-Schossau
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Susanne Kempter
- Faculty of Physics, LMU Munich, 80539 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Nicole L Snyder
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Laura Hartmann
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
- Institute for Macromolecular Chemistry, University Freiburg, Stefan-Meier-Str. 31, 79104 Freiburg im Breisgau, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| |
Collapse
|
4
|
Zeyn Y, Hobernik D, Wilk U, Pöhmerer J, Hieber C, Medina-Montano C, Röhrig N, Strähle CF, Thoma-Kress AK, Wagner E, Bros M, Berger S. Transcriptional Targeting of Dendritic Cells Using an Optimized Human Fascin1 Gene Promoter. Int J Mol Sci 2023; 24:16938. [PMID: 38069260 PMCID: PMC10706967 DOI: 10.3390/ijms242316938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Deeper knowledge about the role of the tumor microenvironment (TME) in cancer development and progression has resulted in new strategies such as gene-based cancer immunotherapy. Whereas some approaches focus on the expression of tumoricidal genes within the TME, DNA-based vaccines are intended to be expressed in antigen-presenting cells (e.g., dendritic cells, DCs) in secondary lymphoid organs, which in turn induce anti-tumor T cell responses. Besides effective delivery systems and the requirement of appropriate adjuvants, DNA vaccines themselves need to be optimized regarding efficacy and selectivity. In this work, the concept of DC-focused transcriptional targeting was tested by applying a plasmid encoding for the luciferase reporter gene under the control of a derivative of the human fascin1 gene promoter (pFscnLuc), comprising the proximal core promoter fused to the normally more distantly located DC enhancer region. DC-focused activity of this reporter construct was confirmed in cell culture in comparison to a standard reporter vector encoding for luciferase under the control of the strong ubiquitously active cytomegalovirus promoter and enhancer (pCMVLuc). Both plasmids were also compared upon intravenous administration in mice. The organ- and cell type-specific expression profile of pFscnLuc versus pCMVLuc demonstrated favorable activity especially in the spleen as a central immune organ and within the spleen in DCs.
Collapse
Affiliation(s)
- Yanira Zeyn
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Dominika Hobernik
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Ulrich Wilk
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| | - Jana Pöhmerer
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| | - Christoph Hieber
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Nadine Röhrig
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Caroline F. Strähle
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.F.S.); (A.K.T.-K.)
| | - Andrea K. Thoma-Kress
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.F.S.); (A.K.T.-K.)
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| |
Collapse
|
5
|
Delehedde C, Ciganek I, Rameix N, Laroui N, Gonçalves C, Even L, Midoux P, Pichon C. Impact of net charge, targeting ligand amount and mRNA modification on the uptake, intracellular routing and the transfection efficiency of mRNA lipopolyplexes in dendritic cells. Int J Pharm 2023; 647:123531. [PMID: 37863445 DOI: 10.1016/j.ijpharm.2023.123531] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/08/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
Targeting mRNA formulations to achieve cell specificity is one of the challenges that must be tackled to mettle their therapeutic potential. Here, lipopolyplexes (LPR) bearing tri-mannose-lipid (TM) are used to target mannose receptor on dendritic cells. We investigated the impact of the net charge and percentage of TM units on the binding, uptake, transfection efficiency (TE) and RNA sensors activation. Binding and uptake capacities of naked and targeted LPR increase with the percent of cationic lipid, but the latter are 2-fold more up taken by the cells. Cationic LPR bearing 5 % and 10 % TM were localized in acidic compartments in contrast to naked LPR and 2.5 % TM-LPR. The drawback is the dramatic decrease of TE as the number of TM-units increases. Cationic LPR bearing 5 % and 10 % TM strongly induced NF-κB and PKR phosphorylation at 6 h. Conversely, mTOR is less activated in line with their low TE. Those side effects are overcome by using 5-methoxyuridine mRNA resulting in an improved TE due to non-phosphorylation of NF-κB and PKR and mTOR activation. Our results point out that targeting DC via mannose receptor triggers a higher uptake of cationic LPRs and fast routing to acidic compartments, and that efficient TE requires low number of TM units use or modified mRNA to escape RNA sensors activation to enhance the translation.
Collapse
Affiliation(s)
- Christophe Delehedde
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France; Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Ivan Ciganek
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Nathalie Rameix
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Nabila Laroui
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Cristine Gonçalves
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Luc Even
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France; Inserm UMS 55 ART ARNm and University of Orléans, F-45100 Orléans; Institut Universitaire de France, 1 rue Descartes, F-75035 Paris, France.
| |
Collapse
|
6
|
mRNA Lipoplexes with Cationic and Ionizable α-Amino-lipophosphonates: Membrane Fusion, Transfection, mRNA Translation and Conformation. Pharmaceutics 2022; 14:pharmaceutics14030581. [PMID: 35335957 PMCID: PMC8952827 DOI: 10.3390/pharmaceutics14030581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/24/2022] [Accepted: 03/02/2022] [Indexed: 01/27/2023] Open
Abstract
Cationic liposomes are attractive carriers for mRNA delivery. Here, mRNA lipoplexes (LX) were prepared with the cationic lipids α-aminolipophosphonate (3b) or imidazolium lipophosphoramidate (2) associated with various α-aminolipophosphonates co-lipids comprising protonable groups (imidazole or pyridine) and DOPE. Physicochemical parameters of liposomes and their membrane fusion activity were measured. LXs comprising either 3b- or 2- allowed transfection of ~25% and 40% of dendritic cells with low cytotoxicity, respectively; the efficiency increased up to 80% when 2 was combined with the imidazole-based co-lipid 1. The transfections were high with 3b/1, 3b/DOPE, 2/1 and 2/DOPE LXs. We observed that the transfection level was not well correlated with the acid-mediated membrane fusion activity of liposomes supposed to destabilize endosomes. The mRNA release from LXs and its translation capacity after release were studied for the most efficient LXs. The results showed that the more mRNA was condensed, the poorer the translation efficiency after release was. In contrast to DNA, circular dichroism performed on mRNA complexed with 2/DOPE revealed the presence of denatured mRNA in LXs explaining this lack of translation efficiency. This is an important parameter that should be stressed for the preparation of mRNA LXs with a conserved mRNA translation activity.
Collapse
|
7
|
Tarvirdipour S, Skowicki M, Schoenenberger CA, Palivan CG. Peptide-Assisted Nucleic Acid Delivery Systems on the Rise. Int J Mol Sci 2021; 22:9092. [PMID: 34445799 PMCID: PMC8396486 DOI: 10.3390/ijms22169092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Concerns associated with nanocarriers' therapeutic efficacy and side effects have led to the development of strategies to advance them into targeted and responsive delivery systems. Owing to their bioactivity and biocompatibility, peptides play a key role in these strategies and, thus, have been extensively studied in nanomedicine. Peptide-based nanocarriers, in particular, have burgeoned with advances in purely peptidic structures and in combinations of peptides, both native and modified, with polymers, lipids, and inorganic nanoparticles. In this review, we summarize advances on peptides promoting gene delivery systems. The efficacy of nucleic acid therapies largely depends on cell internalization and the delivery to subcellular organelles. Hence, the review focuses on nanocarriers where peptides are pivotal in ferrying nucleic acids to their site of action, with a special emphasis on peptides that assist anionic, water-soluble nucleic acids in crossing the membrane barriers they encounter on their way to efficient function. In a second part, we address how peptides advance nanoassembly delivery tools, such that they navigate delivery barriers and release their nucleic acid cargo at specific sites in a controlled fashion.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- Department of Biosystem Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Michal Skowicki
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cora-Ann Schoenenberger
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cornelia G. Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| |
Collapse
|
8
|
Delehedde C, Even L, Midoux P, Pichon C, Perche F. Intracellular Routing and Recognition of Lipid-Based mRNA Nanoparticles. Pharmaceutics 2021; 13:pharmaceutics13070945. [PMID: 34202584 PMCID: PMC8308975 DOI: 10.3390/pharmaceutics13070945] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/07/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Messenger RNA (mRNA) is being extensively used in gene therapy and vaccination due to its safety over DNA, in the following ways: its lack of integration risk, cytoplasmic expression, and transient expression compatible with fine regulations. However, clinical applications of mRNA are limited by its fast degradation by nucleases, and the activation of detrimental immune responses. Advances in mRNA applications, with the recent approval of COVID-19 vaccines, were fueled by optimization of the mRNA sequence and the development of mRNA delivery systems. Although delivery systems and mRNA sequence optimization have been abundantly reviewed, understanding of the intracellular processing of mRNA is mandatory to improve its applications. We will focus on lipid nanoparticles (LNPs) as they are the most advanced nanocarriers for the delivery of mRNA. Here, we will review how mRNA therapeutic potency can be affected by its interactions with cellular proteins and intracellular distribution.
Collapse
Affiliation(s)
- Christophe Delehedde
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR4301, Rue Charles Sadron, 45071 Orléans, France; (C.D.); (P.M.)
- Sanofi R&D, Integrated Drug Discovery, 91385 Chilly-Mazarin, France;
| | - Luc Even
- Sanofi R&D, Integrated Drug Discovery, 91385 Chilly-Mazarin, France;
| | - Patrick Midoux
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR4301, Rue Charles Sadron, 45071 Orléans, France; (C.D.); (P.M.)
| | - Chantal Pichon
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR4301, Rue Charles Sadron, 45071 Orléans, France; (C.D.); (P.M.)
- Correspondence: (C.P.); (F.P.); Tel.: +33-2-3825-5595 (C.P.); Tel.: +33-2-3825-5544 (F.P.)
| | - Federico Perche
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR4301, Rue Charles Sadron, 45071 Orléans, France; (C.D.); (P.M.)
- Correspondence: (C.P.); (F.P.); Tel.: +33-2-3825-5595 (C.P.); Tel.: +33-2-3825-5544 (F.P.)
| |
Collapse
|
9
|
Raoufi E, Bahramimeimandi B, Salehi-Shadkami M, Chaosri P, Mozafari MR. Methodical Design of Viral Vaccines Based on Avant-Garde Nanocarriers: A Multi-Domain Narrative Review. Biomedicines 2021; 9:520. [PMID: 34066608 PMCID: PMC8148582 DOI: 10.3390/biomedicines9050520] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/27/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
The current health crisis caused by coronavirus 2019 (COVID-19) and associated pathogens emphasize the urgent need for vaccine systems that can generate protective and long-lasting immune responses. Vaccination, employing peptides, nucleic acids, and other molecules, or using pathogen-based strategies, in fact, is one of the most potent approaches in the management of viral diseases. However, the vaccine candidate requires protection from degradation and precise delivery to the target cells. This can be achieved by employing different types of drug and vaccine delivery strategies, among which, nanotechnology-based systems seem to be more promising. This entry aims to provide insight into major aspects of vaccine design and formulation to address different diseases, including the recent outbreak of SARS-CoV-2. Special emphasis of this review is on the technical and practical aspects of vaccine construction and theranostic approaches to precisely target and localize the active compounds.
Collapse
Affiliation(s)
- Ehsan Raoufi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran; (E.R.); (B.B.)
| | - Bahar Bahramimeimandi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran; (E.R.); (B.B.)
| | - M. Salehi-Shadkami
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran;
| | - Patcharida Chaosri
- Supreme NanoBiotics Co. Ltd. and Supreme Pharmatech Co. Ltd., 399/90-95 Moo 13 Kingkaew Rd. Soi 25/1, T. Rachateva, A. Bangplee, Samutprakan 10540, Thailand;
| | - M. R. Mozafari
- Supreme NanoBiotics Co. Ltd. and Supreme Pharmatech Co. Ltd., 399/90-95 Moo 13 Kingkaew Rd. Soi 25/1, T. Rachateva, A. Bangplee, Samutprakan 10540, Thailand;
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
10
|
Buschmann MD, Carrasco MJ, Alishetty S, Paige M, Alameh MG, Weissman D. Nanomaterial Delivery Systems for mRNA Vaccines. Vaccines (Basel) 2021; 9:65. [PMID: 33478109 PMCID: PMC7836001 DOI: 10.3390/vaccines9010065] [Citation(s) in RCA: 315] [Impact Index Per Article: 78.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
The recent success of mRNA vaccines in SARS-CoV-2 clinical trials is in part due to the development of lipid nanoparticle delivery systems that not only efficiently express the mRNA-encoded immunogen after intramuscular injection, but also play roles as adjuvants and in vaccine reactogenicity. We present an overview of mRNA delivery systems and then focus on the lipid nanoparticles used in the current SARS-CoV-2 vaccine clinical trials. The review concludes with an analysis of the determinants of the performance of lipid nanoparticles in mRNA vaccines.
Collapse
Affiliation(s)
- Michael D. Buschmann
- Department of Bioengineering, George Mason University, 4400 University Drive, MS 1J7, Fairfax, VA 22030, USA; (M.J.C.); (S.A.)
| | - Manuel J. Carrasco
- Department of Bioengineering, George Mason University, 4400 University Drive, MS 1J7, Fairfax, VA 22030, USA; (M.J.C.); (S.A.)
| | - Suman Alishetty
- Department of Bioengineering, George Mason University, 4400 University Drive, MS 1J7, Fairfax, VA 22030, USA; (M.J.C.); (S.A.)
| | - Mikell Paige
- Department of Chemistry & Biochemistry, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA;
| | - Mohamad Gabriel Alameh
- Perelman School of Medicine, University of Pennsylvania, 130 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104, USA;
| | - Drew Weissman
- Perelman School of Medicine, University of Pennsylvania, 410B Hill Pavilion, 380 S. University Ave, Philadelphia, PA 19104, USA;
| |
Collapse
|
11
|
Gu W, Meng F, Haag R, Zhong Z. Actively targeted nanomedicines for precision cancer therapy: Concept, construction, challenges and clinical translation. J Control Release 2021; 329:676-695. [DOI: 10.1016/j.jconrel.2020.10.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/13/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023]
|
12
|
Abstract
Messenger RNA (mRNA) has immense potential for developing a wide range of therapies, including immunotherapy and protein replacement. As mRNA presents no risk of integration into the host genome and does not require nuclear entry for transfection, which allows protein production even in nondividing cells, mRNA-based approaches can be envisioned as safe and practical therapeutic strategies. Nevertheless, mRNA presents unfavorable characteristics, such as large size, immunogenicity, limited cellular uptake, and sensitivity to enzymatic degradation, which hinder its use as a therapeutic agent. While mRNA stability and immunogenicity have been ameliorated by direct modifications on the mRNA structure, further improvements in mRNA delivery are still needed for promoting its activity in biological settings. In this regard, nanomedicine has shown the ability for spatiotemporally controlling the function of a myriad of bioactive agents in vivo. Direct engineering of nanomedicine structures for loading, protecting, and releasing mRNA and navigating in biological environments can then be applied for promoting mRNA translation toward the development of effective treatments. Here, we review recent approaches aimed at enhancing mRNA function and its delivery through nanomedicines, with particular emphasis on their applications and eventual clinical translation.
Collapse
Affiliation(s)
- Satoshi Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki 210-0821, Japan
| | - Federico Perche
- Centre de Biophysique Moléculaire, UPR4301 CNRS Rue Charles Sadron Orléans, Orléans 45071 Cedex 02, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, UPR4301 CNRS Rue Charles Sadron Orléans, Orléans 45071 Cedex 02, France.,Faculty of Sciences and Techniques, University of Orléans, Orléans 45071 Cedex 02, France
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki 210-0821, Japan
| |
Collapse
|
13
|
Simion V, Henriet E, Juric V, Aquino R, Loussouarn C, Laurent Y, Martin F, Midoux P, Garcion E, Pichon C, Baril P. Intracellular trafficking and functional monitoring of miRNA delivery in glioblastoma using lipopolyplexes and the miRNA-ON RILES reporter system. J Control Release 2020; 327:429-443. [PMID: 32853728 DOI: 10.1016/j.jconrel.2020.08.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/11/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022]
Abstract
MicroRNA (miRNA) oligonucleotides therapeutics are potent and attractive drugs for cancer treatment, but the kinetics of their intracellular trafficking, RISC processing and interaction with their mRNA targets in the cells are still not well understood. Moreover, the absence of efficient carriers impairs their translation into the clinic. Here, we compare the kinetics of miRNA-133a activity after transfection of U87MG glioblastoma cells with either a home-made lipopolyplexes (LPRi) or with the RNAiMax transfection reagent. For this purpose, we combined miRNA intracellular trafficking studies by confocal microscopy with our previously described RILES miRNA-ON reporter system subcloned here in a lentivirus expression vector (LentiRILES) for longitudinal analysis of miRNA activity in transfected cells. Using the LentiRILES system, we report significant differences in terms of miRNA delivery kinetics performed by these two transfection regents. We decipher the mechanisms of miRNA delivery by LPRi and investigate the main steps of miRNA internalization and cytosolic processing. We demonstrate that LPRi preferentially uses caveolae-mediated endocytosis as the main internalization pathway, releases miRNA into the cytosol after the first 3 h of incubation, and addresses the cytosolic miRNAs to P-bodies, while a fraction of miRNAs are exported to the extracellular space through exosomes which were found fully capable to re-transfect the cells. We implanted the LentiRILES cells in the brain of mice and infused the tumours with LPRi.miRNA using the convection-enhanced delivery method. Bioluminescence imaging of the live mice revealed efficient delivery of miRNAs in glioblastoma tumours, attesting successful miRNA uptake, internalization and RISC activation in vivo. Overall, our study provides a comprehensive overview of miRNA intracellular trafficking and processing in a glioblastoma context and highlights the potential use of LPRi for miRNA-based therapy.
Collapse
Affiliation(s)
- Viorel Simion
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, France.
| | - Elodie Henriet
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, France
| | - Viktorija Juric
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, France
| | - Ruth Aquino
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, France
| | - Claire Loussouarn
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Yoan Laurent
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, France
| | - Francisco Martin
- GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, France
| | - Emmanuel Garcion
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, France
| | - Patrick Baril
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, France.
| |
Collapse
|
14
|
He J, Xu S, Mixson AJ. The Multifaceted Histidine-Based Carriers for Nucleic Acid Delivery: Advances and Challenges. Pharmaceutics 2020; 12:E774. [PMID: 32823960 PMCID: PMC7465012 DOI: 10.3390/pharmaceutics12080774] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022] Open
Abstract
Histidines incorporated into carriers of nucleic acids may enhance the extracellular stability of the nanoparticle, yet aid in the intracellular disruption of the nanoparticle, enabling the release of the nucleic acid. Moreover, protonation of histidines in the endosomes may result in endosomal swelling with subsequent lysis. These properties of histidine are based on its five-member imidazole ring in which the two nitrogen atoms may form hydrogen bonds or act as a base in acidic environments. A wide variety of carriers have integrated histidines or histidine-rich domains, which include peptides, polyethylenimine, polysaccharides, platform delivery systems, viral phages, mesoporous silica particles, and liposomes. Histidine-rich carriers have played key roles in our understanding of the stability of nanocarriers and the escape of the nucleic acids from endosomes. These carriers show great promise and offer marked potential in delivering plasmids, siRNA, and mRNA to their intracellular targets.
Collapse
Affiliation(s)
| | | | - A. James Mixson
- Department of Pathology, University Maryland School of Medicine, 10 S. Pine St., University of Maryland, Baltimore, MD 21201, USA; (J.H.); (S.X.)
| |
Collapse
|
15
|
Tarvirdipour S, Huang X, Mihali V, Schoenenberger CA, Palivan CG. Peptide-Based Nanoassemblies in Gene Therapy and Diagnosis: Paving the Way for Clinical Application. Molecules 2020; 25:E3482. [PMID: 32751865 PMCID: PMC7435460 DOI: 10.3390/molecules25153482] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/26/2022] Open
Abstract
Nanotechnology approaches play an important role in developing novel and efficient carriers for biomedical applications. Peptides are particularly appealing to generate such nanocarriers because they can be rationally designed to serve as building blocks for self-assembling nanoscale structures with great potential as therapeutic or diagnostic delivery vehicles. In this review, we describe peptide-based nanoassemblies and highlight features that make them particularly attractive for the delivery of nucleic acids to host cells or improve the specificity and sensitivity of probes in diagnostic imaging. We outline the current state in the design of peptides and peptide-conjugates and the paradigms of their self-assembly into well-defined nanostructures, as well as the co-assembly of nucleic acids to form less structured nanoparticles. Various recent examples of engineered peptides and peptide-conjugates promoting self-assembly and providing the structures with wanted functionalities are presented. The advantages of peptides are not only their biocompatibility and biodegradability, but the possibility of sheer limitless combinations and modifications of amino acid residues to induce the assembly of modular, multiplexed delivery systems. Moreover, functions that nature encoded in peptides, such as their ability to target molecular recognition sites, can be emulated repeatedly in nanoassemblies. Finally, we present recent examples where self-assembled peptide-based assemblies with "smart" activity are used in vivo. Gene delivery and diagnostic imaging in mouse tumor models exemplify the great potential of peptide nanoassemblies for future clinical applications.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
- Department of Biosystem Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Xinan Huang
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
| | - Voichita Mihali
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
| | - Cora-Ann Schoenenberger
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
| | - Cornelia G. Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
| |
Collapse
|
16
|
Patil TS, Deshpande AS. Mannosylated nanocarriers mediated site-specific drug delivery for the treatment of cancer and other infectious diseases: A state of the art review. J Control Release 2020; 320:239-252. [PMID: 31991156 DOI: 10.1016/j.jconrel.2020.01.046] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/24/2020] [Accepted: 01/24/2020] [Indexed: 01/06/2023]
Abstract
The non-modified nanocarriers-based therapies for the treatment of cancer and other infectious diseases enhanced the chemical stability of therapeutically active agents, protected them from enzymatic degradation and extended their blood circulation time. However, the lack of specificity and off-target effects limit their applications. Mannose receptors overexpressed on antigen presenting cells such as dendritic cells and macrophages are one of the most desirable targets for treating cancer and other infectious diseases. Therefore, the development of mannosylated nanocarrier formulation is one of the most extensively explored approaches for targeting these mannose receptors. The present manuscript gives readers the background information on C-type lectin receptors followed by the roles, expression, and distribution of the mannose receptors. It further provides a detailed account of different mannosylated nanocarrier formulations. It also gives the tabular information on most relevant and recently granted patents on mannosylated systems. The overview of mannosylated nanocarrier formulations depicted site-specific targeting, enhanced pharmacokinetic/pharmacodynamic profiles, and improved transfection efficiency of the therapeutically active agents. This suggests the bright future ahead for mannosylated nanocarriers in the treatment of cancer and other infectious diseases. Nevertheless, the mechanism behind the enhanced immune response by mannosylated nanocarriers and their thorough clinical and preclinical evaluation need to explore further.
Collapse
Affiliation(s)
- Tulshidas S Patil
- Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India.
| | - Ashwini S Deshpande
- School of Pharmacy & Technology Management, SVKM's NMIMS, Shirpur, Maharashtra, India.
| |
Collapse
|
17
|
Perche F, Clemençon R, Schulze K, Ebensen T, Guzmán CA, Pichon C. Neutral Lipopolyplexes for In Vivo Delivery of Conventional and Replicative RNA Vaccine. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 17:767-775. [PMID: 31446119 PMCID: PMC6716064 DOI: 10.1016/j.omtn.2019.07.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/17/2019] [Accepted: 07/17/2019] [Indexed: 12/27/2022]
Abstract
Nucleic acid vaccination relies on injecting DNA or RNA coding antigen(s) to induce a protective immune response. RNA vaccination is being increasingly used in preclinical and clinical studies. However, few delivery systems have been reported for in vivo delivery of RNA of different sizes. Using a tripartite formulation with RNA, cationic polymer, and anionic liposomes, we were able to encapsulate RNA into neutral lipopolyplexes (LPPs). LPPs were stable in vitro and successfully delivered conventional RNA and replicative RNA to dendritic cells in cellulo. Their injection led to reporter gene expression in mice. Finally, administration of LPP-Replicon RNA (RepRNA) led to an adaptive immune response against the antigen coded by the RepRNA. Accordingly, LPPs may represent a universal formulation for RNA delivery.
Collapse
Affiliation(s)
- Federico Perche
- Centre de Biophysique Moléculaire, UPR4301 CNRS Rue Charles Sadron Orléans, Orléans Cedex 02, France.
| | - Rudy Clemençon
- Centre de Biophysique Moléculaire, UPR4301 CNRS Rue Charles Sadron Orléans, Orléans Cedex 02, France
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, UPR4301 CNRS Rue Charles Sadron Orléans, Orléans Cedex 02, France.
| |
Collapse
|
18
|
Bartheldyová E, Turánek Knotigová P, Zachová K, Mašek J, Kulich P, Effenberg R, Zyka D, Hubatka F, Kotouček J, Čelechovská H, Héžová R, Tomečková A, Mašková E, Fojtíková M, Macaulay S, Bystrický P, Paulovičová L, Paulovičová E, Drož L, Ledvina M, Raška M, Turánek J. N-Oxy lipid-based click chemistry for orthogonal coupling of mannan onto nanoliposomes prepared by microfluidic mixing: Synthesis of lipids, characterisation of mannan-coated nanoliposomes and in vitro stimulation of dendritic cells. Carbohydr Polym 2018; 207:521-532. [PMID: 30600036 DOI: 10.1016/j.carbpol.2018.10.121] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/18/2018] [Accepted: 10/25/2018] [Indexed: 11/26/2022]
Abstract
New synthetic aminooxy lipid was designed and synthesized as a building block for the formulation of functionalised nanoliposomes (presenting onto the outer surface of aminooxy groups) by microfluidic mixing. Orthogonal binding of cellular mannan (Candida glabrata (CCY 26-20-1) onto the outer surface of functionalised nanoliposomes was modified by orthogonal binding of reducing termini of mannans to oxime lipids via a click chemistry reaction based on aminooxy coupling (oxime ligation). The aminooxy lipid was proved as a suitable active component for preparation of functionalised nanoliposomes by the microfluidic mixing method performed with the instrument NanoAssemblr™. This "on-chip technology" can be easily scaled-up. The structure of mannan-liposomes was visualized by transmission and scanning electron microscopy, including immunogold staining of recombinant mannan receptor bound onto mannosylated-liposomes. The observed structures are in a good correlation with data obtained by DLS, NTA, and TPRS methods. In vitro experiments on human and mouse dendritic cells demonstrate selective internalisation of fluorochrome-labelled mannan-liposomes and their ability to stimulate DC comparable to lipopolysaccharide. We describe a potentially new drug delivery platform for mannan receptor-targeted antimicrobial drugs as well as for immunotherapeutics. Furthermore, the platform based on mannans bound orthogonally onto the surface of nanoliposomes represents a self-adjuvanted carrier for construction of liposome-based recombinant vaccines for both systemic and mucosal routes of administration.
Collapse
Affiliation(s)
- Eliška Bartheldyová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Pavlína Turánek Knotigová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Kateřina Zachová
- Department of Immunology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Josef Mašek
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Pavel Kulich
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Roman Effenberg
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Technická 5, 166, 28 Prague 6, Czech Republic
| | - Daniel Zyka
- APIGENEX s.r.o., Poděbradská 173/5, Prague 9, 190 00, Czech Republic
| | - František Hubatka
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Jan Kotouček
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Hana Čelechovská
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Renata Héžová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Andrea Tomečková
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Eliška Mašková
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Martina Fojtíková
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | | | - Peter Bystrický
- Division of Neurosciences, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University, Malá Hora 10701/4A, 036 01 Martin, Slovakia
| | - Lucia Paulovičová
- Department of Immunochemistry of Glycoconjugates, Immunology & Cell Culture Laboratory, Institute of Chemistry, Center for Glycomics Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia
| | - Ema Paulovičová
- Department of Immunochemistry of Glycoconjugates, Immunology & Cell Culture Laboratory, Institute of Chemistry, Center for Glycomics Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia.
| | - Ladislav Drož
- APIGENEX s.r.o., Poděbradská 173/5, Prague 9, 190 00, Czech Republic
| | - Miroslav Ledvina
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Technická 5, 166, 28 Prague 6, Czech Republic.
| | - Milan Raška
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; Department of Immunology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic.
| | - Jaroslav Turánek
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic.
| |
Collapse
|
19
|
Le Moignic A, Malard V, Benvegnu T, Lemiègre L, Berchel M, Jaffrès PA, Baillou C, Delost M, Macedo R, Rochefort J, Lescaille G, Pichon C, Lemoine FM, Midoux P, Mateo V. Preclinical evaluation of mRNA trimannosylated lipopolyplexes as therapeutic cancer vaccines targeting dendritic cells. J Control Release 2018; 278:110-121. [PMID: 29630987 DOI: 10.1016/j.jconrel.2018.03.035] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 12/28/2022]
Abstract
Clinical trials with direct administration of synthetic mRNAs encoding tumor antigens demonstrated safety and induction of tumor-specific immune responses. Their proper delivery to dendritic cells (DCs) requires their protection against RNase degradation and more specificity for dose reduction. Lipid-Polymer-RNA lipopolyplexes (LPR) are attractive mRNA delivery systems and their equipment with mannose containing glycolipid, specific of endocytic receptors present on the membrane of DCs is a valuable strategy. In this present work, we evaluated the capacity of LPR functionalized with a tri-antenna of α-d-mannopyranoside (triMN-LPR) concerning (i) their binding to CD209/DC-SIGN and CD207/Langerin expressing cell lines, human and mouse DCs and other hematopoietic cell populations, (ii) the nature of induced immune response after in vivo immunization and (iii) their therapeutic anti-cancer vaccine efficiency. We demonstrated that triMN-LPR provided high induction of a local inflammatory response two days after intradermal injection to C57BL/6 mice, followed by the recruitment and activation of DCs in the corresponding draining lymph nodes. This was associated with skin production of CCR7 and CXCR4 at vaccination sites driving DC migration. High number of E7-specific T cells was detected after E7-encoded mRNA triMN-LPR vaccination. When evaluated in three therapeutic pre-clinical murine tumor models such as E7-expressing TC1 cells, OVA-expressing EG7 cells and MART-1-expressing B16F0 cells, triMN-LPR carrying mRNA encoding the respective antigens significantly exert curative responses in mice vaccinated seven days after initial tumor inoculation. These results provide evidence that triMN-LPR give rise to an efficient stimulatory immune response allowing for therapeutic anti-cancer vaccination in mice. This mRNA formulation should be considered for anti-cancer vaccination in Humans.
Collapse
Affiliation(s)
- A Le Moignic
- Sorbonne Universite, Paris, France; UMR-S INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et Maladies Infectieuses (CIMI-Paris), Paris, France
| | - V Malard
- Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| | | | | | - M Berchel
- CEMCA, CNRS UMR 6521, SFR148 ScInBioS, Université de Brest, Brest, France
| | - P-A Jaffrès
- CEMCA, CNRS UMR 6521, SFR148 ScInBioS, Université de Brest, Brest, France
| | - C Baillou
- Sorbonne Universite, Paris, France; UMR-S INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et Maladies Infectieuses (CIMI-Paris), Paris, France
| | - M Delost
- UMR-S INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et Maladies Infectieuses (CIMI-Paris), Paris, France
| | - R Macedo
- UMR-S INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et Maladies Infectieuses (CIMI-Paris), Paris, France
| | - J Rochefort
- UMR-S INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et Maladies Infectieuses (CIMI-Paris), Paris, France; Paris Diderot/Paris 07, Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier Pitié-Salpêtrière, Department of Odontology, Paris, France
| | - G Lescaille
- UMR-S INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et Maladies Infectieuses (CIMI-Paris), Paris, France; Paris Diderot/Paris 07, Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier Pitié-Salpêtrière, Department of Odontology, Paris, France
| | - C Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| | - F M Lemoine
- Sorbonne Universite, Paris, France; UMR-S INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et Maladies Infectieuses (CIMI-Paris), Paris, France; AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Cell and Gene Therapy Unit, Paris, France.
| | - P Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France.
| | - V Mateo
- Sorbonne Universite, Paris, France; UMR-S INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et Maladies Infectieuses (CIMI-Paris), Paris, France
| |
Collapse
|
20
|
Khalil IA, Kimura S, Sato Y, Harashima H. Synergism between a cell penetrating peptide and a pH-sensitive cationic lipid in efficient gene delivery based on double-coated nanoparticles. J Control Release 2018; 275:107-116. [PMID: 29452131 DOI: 10.1016/j.jconrel.2018.02.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/21/2018] [Accepted: 02/12/2018] [Indexed: 12/16/2022]
Abstract
We report on the development of a highly efficient gene delivery system based on synergism between octaarginine (R8), a representative cell penetrating peptide, and YSK05, a recently developed pH-sensitive cationic lipid. Attaching a high density of R8 on the surface of YSK05 nanoparticles (NPs) that contained encapsulated plasmid DNA resulted in the formation of positively charged NPs with improved transfection efficiency. To avoid the development of a net positive charge, we controlled the density and topology of the R8 peptide through the use of a two-step coating methodology, in which the inner lipid coat was modified with a low density of R8 which was then covered with an outer neutral YSK05 lipid layer. Although used in low amounts, the R8 peptide improved cellular uptake and endosomal escape of the DNA encapsulated in YSK05 NPs, which resulted in a high transfection efficiency. The two-step coating design was essential for achieving a high degree of transfection, as evidenced by the low activity of NPs modified with the same amount of R8 in a regular single-coated design. In addition, a high transfection efficiency was not observed when R8 or YSK05 were used alone, which confirms the existence of a synergistic effect between both components. The results of this study indicate that cationic cell penetrating peptides have the ability to improve transfection activities without imparting a net positive charge when used in the proper amount and in conjunction with the appropriate design. This is expected to significantly increase the potential applications of these peptides as tools for augmenting the activity of lipid nanoparticles used in gene delivery.
Collapse
Affiliation(s)
- Ikramy A Khalil
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Seigo Kimura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
21
|
Sergeeva OV, Koteliansky VE, Zatsepin TS. mRNA-Based Therapeutics - Advances and Perspectives. BIOCHEMISTRY (MOSCOW) 2017; 81:709-22. [PMID: 27449617 DOI: 10.1134/s0006297916070075] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In this review we discuss features of mRNA synthesis and modifications used to minimize immune response and prolong efficiency of the translation process in vivo. Considerable attention is given to the use of liposomes and nanoparticles containing lipids and polymers for the mRNA delivery. Finally we briefly discuss mRNAs which are currently in the clinical trials for cancer immunotherapy, vaccination against infectious diseases, and replacement therapy.
Collapse
Affiliation(s)
- O V Sergeeva
- Lomonosov Moscow State University, Department of Chemistry, Moscow, 119991, Russia.
| | | | | |
Collapse
|
22
|
Berchel M, Akhter S, Berthe W, Gonçalves C, Dubuisson M, Pichon C, Jaffrès PA, Midoux P. Synthesis of α-amino-lipophosphonates as cationic lipids or co-lipids for DNA transfection in dendritic cells. J Mater Chem B 2017; 5:6869-6881. [DOI: 10.1039/c7tb01080j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cationic lipid/co-lipid combinations have been extensively explored in gene delivery as alternatives to viral vectors.
Collapse
Affiliation(s)
- Mathieu Berchel
- CEMCA
- UMR CNRS 6521
- University of Brest
- IBSAM
- 6 Avenue Victor Le Gorgeu
| | - Sohail Akhter
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
- Le Studium® Loire Valley Institute for Advanced Studies
| | - Wilfried Berthe
- CEMCA
- UMR CNRS 6521
- University of Brest
- IBSAM
- 6 Avenue Victor Le Gorgeu
| | - Cristine Gonçalves
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
| | - Marine Dubuisson
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
| | | | - Patrick Midoux
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
| |
Collapse
|
23
|
Abstract
mRNA vaccines elicit a potent immune response including antibodies and cytotoxic T cells. mRNA vaccines are currently evaluated in clinical trials for cancer immunotherapy applications, but also have great potential as prophylactic vaccines. Efficient delivery of mRNA vaccines will be key for their success and translation to the clinic. Among potential nonviral vectors, lipid nanoparticles are particularly promising. Indeed, lipid nanoparticles can be synthesized with relative ease in a scalable manner, protect the mRNA against degradation, facilitate endosomal escape, can be targeted to the desired cell type by surface decoration with ligands, and as needed, can be codelivered with adjuvants.
Collapse
|
24
|
Berchel M, Lozach O, Berthe W, Hernot S, Couthon-Gourvès H, Mottais A, Le Gall T, Midoux P, Montier T, Jaffrès PA. Synthetic phospholipids and phospho-bola-amphiphiles for nucleic acid delivery. PHOSPHORUS SULFUR 2016. [DOI: 10.1080/10426507.2016.1212049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- M. Berchel
- CEMCA, UMR CNRS 6521, Université de Brest, IBSAM, Brest, France
| | - O. Lozach
- CEMCA, UMR CNRS 6521, Université de Brest, IBSAM, Brest, France
| | - W. Berthe
- CEMCA, UMR CNRS 6521, Université de Brest, IBSAM, Brest, France
| | - S. Hernot
- CEMCA, UMR CNRS 6521, Université de Brest, IBSAM, Brest, France
| | | | - A. Mottais
- U INSERM 1078, CHRU de Brest Hôpital Morvan, Faculté de Médecine, Université de Brest, Brest, France
| | - T. Le Gall
- U INSERM 1078, CHRU de Brest Hôpital Morvan, Faculté de Médecine, Université de Brest, Brest, France
| | - P. Midoux
- Centre de Biophysique Moléculaire, Orléans cedex 2, France
| | - T. Montier
- U INSERM 1078, CHRU de Brest Hôpital Morvan, Faculté de Médecine, Université de Brest, Brest, France
| | - P. A. Jaffrès
- CEMCA, UMR CNRS 6521, Université de Brest, IBSAM, Brest, France
- Centre de Biophysique Moléculaire, Orléans cedex 2, France
| |
Collapse
|
25
|
Barbeau J, Lemiègre L, Quelen A, Malard V, Gao H, Gonçalves C, Berchel M, Jaffrès PA, Pichon C, Midoux P, Benvegnu T. Synthesis of a trimannosylated-equipped archaeal diether lipid for the development of novel glycoliposomes. Carbohydr Res 2016; 435:142-148. [DOI: 10.1016/j.carres.2016.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/10/2016] [Accepted: 10/10/2016] [Indexed: 11/16/2022]
|
26
|
Tuettenberg A, Steinbrink K, Schuppan D. Myeloid cells as orchestrators of the tumor microenvironment: novel targets for nanoparticular cancer therapy. Nanomedicine (Lond) 2016; 11:2735-2751. [DOI: 10.2217/nnm-2016-0208] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Macrophages, myeloid-derived suppressor cells and tolerogenic dendritic cells are central players of a heterogeneous myeloid cell population, with the ability to suppress innate and adaptive immune responses and thus to promote tumor growth. Their influx and local proliferation are mainly induced by the cancers themselves, and their numbers in the tumor microenvironment and the peripheral blood correlate with decreased survival. Therapeutic targeting these innate immune cells, either aiming at their elimination or polarization toward tumor suppressive cells is an attractive novel approach to control tumor progression and block metastasis. We review the current understanding of cancer immunology including immune surveillance and immune editing in the context of these prominent innate suppressor cells, and their targetability by nanoparticular immunotherapy with small molecules or siRNA.
Collapse
Affiliation(s)
- Andrea Tuettenberg
- Department of Dermatology & Research Center for Immunotherapy (FZI) University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology & Research Center for Immunotherapy (FZI) University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology & Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Rezaee M, Oskuee RK, Nassirli H, Malaekeh-Nikouei B. Progress in the development of lipopolyplexes as efficient non-viral gene delivery systems. J Control Release 2016; 236:1-14. [DOI: 10.1016/j.jconrel.2016.06.023] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 06/12/2016] [Accepted: 06/13/2016] [Indexed: 01/05/2023]
|
28
|
Towards Targeted Delivery Systems: Ligand Conjugation Strategies for mRNA Nanoparticle Tumor Vaccines. J Immunol Res 2015; 2015:680620. [PMID: 26819957 PMCID: PMC4706915 DOI: 10.1155/2015/680620] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/18/2015] [Indexed: 12/02/2022] Open
Abstract
The use of nanoparticles encapsulating messenger RNA (mRNA) as a vaccine has recently attracted much attention because of encouraging results achieved in many nonviral genetic antitumor vaccination studies. Notably, in all of these studies, mRNA nanoparticles are passively targeted to dendritic cells (DCs) through careful selection of vaccination sites. Hence, DC-targeted mRNA nanoparticle vaccines may be an imminent next step forward. In this brief report, we will discuss established conjugation strategies that have been successfully applied to both polymeric and liposomal gene delivery systems. We will also briefly describe promising DC surface receptors amenable for targeting mRNA nanoparticles. Practicable conjugation strategies and receptors reviewed in this paper will provide a convenient reference to facilitate future development of targeted mRNA nanoparticle vaccine.
Collapse
|
29
|
Markov OV, Mironova NL, Shmendel EV, Serikov RN, Morozova NG, Maslov MA, Vlassov VV, Zenkova MA. Multicomponent mannose-containing liposomes efficiently deliver RNA in murine immature dendritic cells and provide productive anti-tumour response in murine melanoma model. J Control Release 2015; 213:45-56. [DOI: 10.1016/j.jconrel.2015.06.028] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/15/2015] [Accepted: 06/21/2015] [Indexed: 12/21/2022]
|
30
|
Vallazza B, Petri S, Poleganov MA, Eberle F, Kuhn AN, Sahin U. Recombinant messenger RNA technology and its application in cancer immunotherapy, transcript replacement therapies, pluripotent stem cell induction, and beyond. WILEY INTERDISCIPLINARY REVIEWS-RNA 2015; 6:471-99. [DOI: 10.1002/wrna.1288] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/23/2015] [Accepted: 04/28/2015] [Indexed: 12/24/2022]
Affiliation(s)
| | | | | | | | | | - Ugur Sahin
- BioNTech RNA Pharmaceuticals GmbH; Mainz Germany
- TRON gGmbH; Mainz Germany
| |
Collapse
|
31
|
Majzoub RN, Chan CL, Ewert KK, Silva BFB, Liang KS, Safinya CR. Fluorescence microscopy colocalization of lipid-nucleic acid nanoparticles with wildtype and mutant Rab5-GFP: A platform for investigating early endosomal events. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1308-18. [PMID: 25753113 DOI: 10.1016/j.bbamem.2015.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/26/2015] [Accepted: 03/01/2015] [Indexed: 11/19/2022]
Abstract
Endosomal entrapment is known to be a major bottleneck to successful cytoplasmic delivery of nucleic acids (NAs) using cationic liposome-NA nanoparticles (NPs). Quantitative measurements of distributions of NPs within early endosomes (EEs) have proven difficult due to the sub-resolution size and short lifetime of wildtype EEs. In this study we used Rab5-GFP, a member of the large family of GTPases which cycles between the plasma membrane and early endosomes, to fluorescently label early endosomes. Using fluorescence microscopy and quantitative image analysis of cells expressing Rab5-GFP, we found that at early time points (t<1h), only a fraction (≈35%) of RGD-tagged NPs (which target cell surface integrins) colocalize with wildtype EEs, independent of the NP's membrane charge density. In comparison, a GTP-hydrolysis deficient mutant, Rab5-Q79L, which extends the size and lifetime of EEs yielding giant early endosomes (GEEs), enabled us to resolve and localize individual NPs found within the GEE lumen. Remarkably, nearly all intracellular NPs are found to be trapped within GEEs implying little or no escape at early time points. The observed small degree of colocalization of NPs and wildtype Rab5 is consistent with recycling of Rab5-GDP to the plasma membrane and not indicative of NP escape from EEs. Taken together, our results show that endosomal escape of PEGylated nanoparticles occurs downstream of EEs i.e., from late endosomes/lysosomes. Our studies also suggest that Rab5-Q79L could be used in a robust imaging assay which allows for direct visualization of NP interactions with the luminal membrane of early endosomes.
Collapse
Affiliation(s)
- Ramsey N Majzoub
- Department of Physics, University of California, Santa Barbara, CA 93106, USA; Department of Materials, University of California, Santa Barbara, CA 93106, USA; Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| | - Chia-Ling Chan
- Department of Physics, University of California, Santa Barbara, CA 93106, USA; Department of Materials, University of California, Santa Barbara, CA 93106, USA; Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA; Institute of Physics, Academica Sinica, Taipei 11529, Taiwan; National Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan
| | - Kai K Ewert
- Department of Physics, University of California, Santa Barbara, CA 93106, USA; Department of Materials, University of California, Santa Barbara, CA 93106, USA; Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| | - Bruno F B Silva
- Department of Physics, University of California, Santa Barbara, CA 93106, USA; Department of Materials, University of California, Santa Barbara, CA 93106, USA; Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA; Division of Physical Chemistry, Centre for Chemistry and Chemical Engineering, Lund University, SE-221 00 Lund, Sweden
| | - Keng S Liang
- National Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan; Department of Electrophysics, National Chiao-Tung University, Hsinchu 30010, Taiwan
| | - Cyrus R Safinya
- Department of Physics, University of California, Santa Barbara, CA 93106, USA; Department of Materials, University of California, Santa Barbara, CA 93106, USA; Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
32
|
Heller P, Mohr N, Birke A, Weber B, Reske-Kunz A, Bros M, Barz M. Directed Interactions of Block Copolypept(o)ides with Mannose-binding Receptors: PeptoMicelles Targeted to Cells of the Innate Immune System. Macromol Biosci 2015; 15:63-73. [DOI: 10.1002/mabi.201400417] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 10/23/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Philipp Heller
- Institute of Organic Chemistry; Johannes Gutenberg-University; Duesbergweg 10-14 55099 Mainz Germany
| | - Nicole Mohr
- Institute of Organic Chemistry; Johannes Gutenberg-University; Duesbergweg 10-14 55099 Mainz Germany
| | - Alexander Birke
- Institute of Organic Chemistry; Johannes Gutenberg-University; Duesbergweg 10-14 55099 Mainz Germany
| | - Benjamin Weber
- Institute of Organic Chemistry; Johannes Gutenberg-University; Duesbergweg 10-14 55099 Mainz Germany
| | - Angelika Reske-Kunz
- Department of Dermatology; University Medical Center; Johannes Gutenberg-University Mainz; Obere Zahlbacher Straße 63 55131 Mainz Germany
| | - Matthias Bros
- Department of Dermatology; University Medical Center; Johannes Gutenberg-University Mainz; Obere Zahlbacher Straße 63 55131 Mainz Germany
| | - Matthias Barz
- Institute of Organic Chemistry; Johannes Gutenberg-University; Duesbergweg 10-14 55099 Mainz Germany
| |
Collapse
|
33
|
Abstract
Synthetic mRNAs can become biopharmaceutics allowing vaccination against cancer, bacterial and virus infections. Clinical trials with direct administration of synthetic mRNAs encoding tumor antigens demonstrated safety and induction of tumor-specific immune responses. Although immune responses are generated by naked mRNAs, their formulations with chemical carriers are expected to provide more specificity and internalization in dendritic cells (DCs) for better immune responses and dose reduction. This review reports lipid-based formulations (LBFs) that have proved preclinical efficacy. The selective delivery of mRNA LBFs to favor intracellular accumulation in DCs and reduction of the effective doses is discussed, notably to decorate LBFs with carbohydrates or glycomimetics allowing endocytosis in DCs. We also report how smart intracellular delivery is achieved using pH-sensitive lipids or polymers for an efficient mRNA escape from endosomes and limitations regarding cytosolic mRNA location for translation.
Collapse
Affiliation(s)
- Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and Université d'Orléans, Orléans, 45071, cedex 02, France
| | | |
Collapse
|
34
|
McCullough KC, Milona P, Thomann-Harwood L, Démoulins T, Englezou P, Suter R, Ruggli N. Self-Amplifying Replicon RNA Vaccine Delivery to Dendritic Cells by Synthetic Nanoparticles. Vaccines (Basel) 2014; 2:735-54. [PMID: 26344889 PMCID: PMC4494254 DOI: 10.3390/vaccines2040735] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/29/2014] [Accepted: 09/28/2014] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC) play essential roles determining efficacy of vaccine delivery with respect to immune defence development and regulation. This renders DCs important targets for vaccine delivery, particularly RNA vaccines. While delivery of interfering RNA oligonucleotides to the appropriate intracellular sites for RNA-interference has proven successful, the methodologies are identical for RNA vaccines, which require delivery to RNA translation sites. Delivery of mRNA has benefitted from application of cationic entities; these offer value following endocytosis of RNA, when cationic or amphipathic properties can promote endocytic vesicle membrane perturbation to facilitate cytosolic translocation. The present review presents how such advances are being applied to the delivery of a new form of RNA vaccine, replicons (RepRNA) carrying inserted foreign genes of interest encoding vaccine antigens. Approaches have been developed for delivery to DCs, leading to the translation of the RepRNA and encoded vaccine antigens both in vitro and in vivo. Potential mechanisms favouring efficient delivery leading to translation are discussed with respect to the DC endocytic machinery, showing the importance of cytosolic translocation from acidifying endocytic structures. The review relates the DC endocytic pathways to immune response induction, and the potential advantages for these self-replicating RNA vaccines in the near future.
Collapse
Affiliation(s)
| | - Panagiota Milona
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | | | - Thomas Démoulins
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | - Pavlos Englezou
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | - Rolf Suter
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | - Nicolas Ruggli
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| |
Collapse
|
35
|
Yan CY, Gu JW, Hou DP, Jing HY, Wang J, Guo YZ, Katsumi H, Sakane T, Yamamoto A. Synthesis of Tat tagged and folate modified N-succinyl-chitosan self-assembly nanoparticles as a novel gene vector. Int J Biol Macromol 2014; 72:751-6. [PMID: 25281874 DOI: 10.1016/j.ijbiomac.2014.09.031] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/20/2014] [Accepted: 09/25/2014] [Indexed: 12/11/2022]
Abstract
The purpose of this research was to prepare a novel type of Tat tagged and folate modified N-succinyl-chitosan (Tat-Suc-FA) self-assembly nanoparticles, to provide a new vector for tumor gene therapy. In this study, Tat-Suc-FA polymers was synthesized and characterized using (1)H NMR and FT-IR. The copolymer had a mean diameter of 65 ± 22.6 nm, a zeta potential of 40 ± 0.2 mV. The cytotoxicity assay showed that Tat-Suc-FA polymers were less toxic than chitosan in the tested concentration range (from 2 to 500 μg/ml). Tat-Suc-FA/DNA complexes at various weight ratios were formulated and characterized. Particle sizes of Tat-Suc-FA/DNA complexes were between 54 and 106 nm as determined by dynamic light scattering. Accordingly, Transmission electron microscope photo of Tat-Suc-FA/DNA complexes exhibited a spherical and compact morphology. Zeta potentials of these complexes changed as the weight ratio varied (from 3 to 44 mV). Agarose gel electrophoresis assay showed that Tat-Suc-FA could efficiently condense the DNA, when the weight ratio was above 1.5/1. Together, these results suggest that the low toxic Tat-Suc-FA cationic polymers could be considered for use as a novel type of gene delivery vectors.
Collapse
Affiliation(s)
- Cheng-yun Yan
- College of Pharmacy, Guilin Medical University, Guilin 541004, China; First Affiliated Hospital of Jiamusi University, Jiamusi of University, Jiamusi 154000, China.
| | - Ji-wei Gu
- First Affiliated Hospital of Jiamusi University, Jiamusi of University, Jiamusi 154000, China
| | - Da-ping Hou
- First Affiliated Hospital of Jiamusi University, Jiamusi of University, Jiamusi 154000, China
| | - Hong-ying Jing
- First Affiliated Hospital of Jiamusi University, Jiamusi of University, Jiamusi 154000, China
| | - Jing Wang
- First Affiliated Hospital of Jiamusi University, Jiamusi of University, Jiamusi 154000, China
| | - Yu-zhi Guo
- First Affiliated Hospital of Jiamusi University, Jiamusi of University, Jiamusi 154000, China
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Toshiyasu Sakane
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| |
Collapse
|
36
|
Abstract
Use of mRNA-based vaccines for tumour immunotherapy has gained increasing attention in recent years. A growing number of studies applying nanomedicine concepts to mRNA tumour vaccination show that the mRNA delivered in nanoparticle format can generate a more robust immune response. Advances in the past decade have deepened our understanding of gene delivery barriers, mRNA's biological stability and immunological properties, and support the notion for engineering innovations tailored towards a more efficient mRNA nanoparticle vaccine delivery system. In this review we will first examine the suitability of mRNA for engineering manipulations, followed by discussion of a model framework that highlights the barriers to a robust anti-tumour immunity mediated by mRNA encapsulated in nanoparticles. Finally, by consolidating existing literature on mRNA nanoparticle tumour vaccination within the context of this framework, we aim to identify bottlenecks that can be addressed by future nanoengineering research.
Collapse
Affiliation(s)
- Kyle K L Phua
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| | | | | |
Collapse
|
37
|
McCullough KC, Bassi I, Milona P, Suter R, Thomann-Harwood L, Englezou P, Démoulins T, Ruggli N. Self-replicating Replicon-RNA Delivery to Dendritic Cells by Chitosan-nanoparticles for Translation In Vitro and In Vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e173. [PMID: 25004099 PMCID: PMC4121514 DOI: 10.1038/mtna.2014.24] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 05/20/2014] [Indexed: 02/06/2023]
Abstract
Self-amplifying replicon RNA (RepRNA) possesses high potential for increasing antigen load within dendritic cells (DCs). The major aim of the present work was to define how RepRNA delivered by biodegradable, chitosan-based nanoparticulate delivery vehicles (nanogel-alginate (NGA)) interacts with DCs, and whether this could lead to translation of the RepRNA in the DCs. Although studies employed virus replicon particles (VRPs), there are no reports on biodegradable, nanoparticulate vehicle delivery of RepRNA. VRP studies employed cytopathogenic agents, contrary to DC requirements—slow processing and antigen retention. We employed noncytopathogenic RepRNA with NGA, demonstrating for the first time the efficiency of RepRNA association with nanoparticles, NGA delivery to DCs, and RepRNA internalization by DCs. RepRNA accumulated in vesicular structures, with patterns typifying cytosolic release. This promoted RepRNA translation, in vitro and in vivo. Delivery and translation were RepRNA concentration-dependent, occurring in a kinetic manner. Including cationic lipids with chitosan during nanoparticle formation enhanced delivery and translation kinetics, but was not required for translation of immunogenic levels in vivo. This work describes for the first time the characteristics associated with chitosan-nanoparticle delivery of self-amplifying RepRNA to DCs, leading to translation of encoded foreign genes, namely influenza virus hemagglutinin and nucleoprotein.
Collapse
Affiliation(s)
| | - Isabelle Bassi
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Panagiota Milona
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Rolf Suter
- 1] Institute of Virology and Immunology, Mittelhäusern, Switzerland [2] Current address: Laboratory of Experimental Biophysics, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Pavlos Englezou
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Thomas Démoulins
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Nicolas Ruggli
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| |
Collapse
|
38
|
Le Corre SS, Berchel M, Couthon-Gourvès H, Haelters JP, Jaffrès PA. Atherton-Todd reaction: mechanism, scope and applications. Beilstein J Org Chem 2014; 10:1166-96. [PMID: 24991268 PMCID: PMC4077366 DOI: 10.3762/bjoc.10.117] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 04/09/2014] [Indexed: 12/14/2022] Open
Abstract
Initially, the Atherton-Todd (AT) reaction was applied for the synthesis of phosphoramidates by reacting dialkyl phosphite with a primary amine in the presence of carbon tetrachloride. These reaction conditions were subsequently modified with the aim to optimize them and the reaction was extended to different nucleophiles. The mechanism of this reaction led to controversial reports over the past years and is adequately discussed. We also present the scope of the AT reaction. Finally, we investigate the AT reaction by means of exemplary applications, which mainly concern three topics. First, we discuss the activation of a phenol group as a phosphate which allows for subsequent transformations such as cross coupling and reduction. Next, we examine the AT reaction applied to produce fire retardant compounds. In the last section, we investigate the use of the AT reaction for the production of compounds employed for biological applications. The selected examples to illustrate the applications of the Atherton-Todd reaction mainly cover the past 15 years.
Collapse
Affiliation(s)
- Stéphanie S Le Corre
- Université de Brest, Université Européenne de Bretagne, CEMCA, CNRS UMR 6521, SynNanoVect, IFR 148 ScInBIoS, 6 Avenue Le Gorgeu, 29238 Brest, France
| | - Mathieu Berchel
- Université de Brest, Université Européenne de Bretagne, CEMCA, CNRS UMR 6521, SynNanoVect, IFR 148 ScInBIoS, 6 Avenue Le Gorgeu, 29238 Brest, France
| | - Hélène Couthon-Gourvès
- Université de Brest, Université Européenne de Bretagne, CEMCA, CNRS UMR 6521, SynNanoVect, IFR 148 ScInBIoS, 6 Avenue Le Gorgeu, 29238 Brest, France
| | - Jean-Pierre Haelters
- Université de Brest, Université Européenne de Bretagne, CEMCA, CNRS UMR 6521, SynNanoVect, IFR 148 ScInBIoS, 6 Avenue Le Gorgeu, 29238 Brest, France
| | - Paul-Alain Jaffrès
- Université de Brest, Université Européenne de Bretagne, CEMCA, CNRS UMR 6521, SynNanoVect, IFR 148 ScInBIoS, 6 Avenue Le Gorgeu, 29238 Brest, France
| |
Collapse
|
39
|
Gonçalves C, Berchel M, Gosselin MP, Malard V, Cheradame H, Jaffrès PA, Guégan P, Pichon C, Midoux P. Lipopolyplexes comprising imidazole/imidazolium lipophosphoramidate, histidinylated polyethyleneimine and siRNA as efficient formulation for siRNA transfection. Int J Pharm 2014; 460:264-72. [DOI: 10.1016/j.ijpharm.2013.11.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/01/2013] [Indexed: 02/05/2023]
|
40
|
Gajbhiye V, Gong S. Lectin functionalized nanocarriers for gene delivery. Biotechnol Adv 2013; 31:552-62. [DOI: 10.1016/j.biotechadv.2013.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/03/2013] [Accepted: 01/09/2013] [Indexed: 01/01/2023]
|
41
|
Berchel M, Le Corre S, Le Gall T, Couthon-Gourvès H, Haelters JP, Midoux P, Montier T, Lehn P, Jaffrès PA. Trimethylarsonium-Based Cationic Phospholipids for Gene Delivery. PHOSPHORUS SULFUR 2013. [DOI: 10.1080/10426507.2012.741161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Mathieu Berchel
- a Université de Brest, UEB, CEMCA, UMR CNRS 6521, 6 avenue Le Gorgeu , 29238 , Brest , France
| | - Stéphanie Le Corre
- a Université de Brest, UEB, CEMCA, UMR CNRS 6521, 6 avenue Le Gorgeu , 29238 , Brest , France
| | - Tony Le Gall
- b Université de Brest, IFR148 ScInBioS, INSERM U1078, Bat 2 bis, Hôpital Morvan CHU Brest, 5 avenue du Maréchal Foch , Brest , France
| | - Hélène Couthon-Gourvès
- a Université de Brest, UEB, CEMCA, UMR CNRS 6521, 6 avenue Le Gorgeu , 29238 , Brest , France
| | - Jean-Pierre Haelters
- a Université de Brest, UEB, CEMCA, UMR CNRS 6521, 6 avenue Le Gorgeu , 29238 , Brest , France
| | - Patrick Midoux
- c CBM, CNRS UPR4301, rue Charles Sadron F-45071 Orléans , Cedex , 02 , France
| | - Tristan Montier
- b Université de Brest, IFR148 ScInBioS, INSERM U1078, Bat 2 bis, Hôpital Morvan CHU Brest, 5 avenue du Maréchal Foch , Brest , France
| | - Pierre Lehn
- b Université de Brest, IFR148 ScInBioS, INSERM U1078, Bat 2 bis, Hôpital Morvan CHU Brest, 5 avenue du Maréchal Foch , Brest , France
| | - Paul-Alain Jaffrès
- a Université de Brest, UEB, CEMCA, UMR CNRS 6521, 6 avenue Le Gorgeu , 29238 , Brest , France
| |
Collapse
|
42
|
Pichon C, Midoux P. Mannosylated and histidylated LPR technology for vaccination with tumor antigen mRNA. Methods Mol Biol 2013; 969:247-74. [PMID: 23296939 DOI: 10.1007/978-1-62703-260-5_16] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
mRNA-based vaccines are currently being developed for treating various diseases including cancers. For this purpose, synthetic or in vitro transcribed (IVT) mRNA encoding tumor antigen offers several advantages over plasmid DNA encoding tumor antigen including better delivery and security. In this chapter, we report the preparation of mannosylated mRNA nanoparticles termed mannosylated lipopolyplexes or Man-LPR loaded with mRNA encoding a melanoma antigen. This formulation enhances the transfection of dendritic cells (DCs) in vivo and the anti-B16F10 melanoma vaccination in mice. The mRNA is formulated with histidylated liposomes and a histidylated polymer. Those pH-sensitive vectors promote membrane destabilization in endosomes upon the protonation of their histidine groups, allowing nucleic acid delivery in the cytosol. To favor DCs targeting via the mannose receptor, a mannose lipid is incorporated in the liposomes. Here, we provide protocols for the preparation of mannosylated liposomes, the synthesis of mRNA, mice immunization based on systemic injection, measurement of the cellular immune response and determination of the number of transfected splenic DC.
Collapse
Affiliation(s)
- Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| | | |
Collapse
|
43
|
Sun X, Chen S, Han J, Zhang Z. Mannosylated biodegradable polyethyleneimine for targeted DNA delivery to dendritic cells. Int J Nanomedicine 2012; 7:2929-42. [PMID: 22745554 PMCID: PMC3384368 DOI: 10.2147/ijn.s31760] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND To establish a potential gene-delivery system with the ability to deliver plasmid DNA to dendritic cells (DCs) more efficiently and specifically, we designed and synthesized a low-molecular-weight polyethyleneimine and triethyleneglycol polymer (PEI-TEG) and a series of its mannosylated derivatives. METHODS PEI-TEG was synthesized from PEI2000 and PEI600 with TEG as the cross-linker. PEI-TEG was then linked to mannose via a phenylisothiocyanate bridge to obtain man-PEI-TEG conjugates. The DNA conveyance abilities of PEI-TEG, man-PEI-TEG, as well as control PEI25k were evaluated by measuring their zeta potential, particle size, and DNA-binding abilities. The in vitro cytotoxicity, cell uptake, and transfection efficiency of these PEI/DNA complexes were examined on the DC2.4 cell line. Finally, a maturation experiment evaluated the effect of costimulatory molecules CD40, CD80, and CD86 on murine bone marrow-derived DCs (BMDCs) using flow cytometry. RESULTS PEI-TEG and man-PEI-TEG were successfully synthesized and were shown to retain the excellent properties of PEI25k for condensing DNA. Compared with PEI-TEG as well as PEI25k, the man-PEI-TEG had less cytotoxicity and performed better in both cellular uptake and transfection assays in vitro. The results of the maturation experiment showed that all the PEI/DNA complexes induced an adequate upregulation of surface markers for DC maturation. CONCLUSION These results demonstrated that man-PEI-TEG can be employed as a DC-targeting gene-delivery system.
Collapse
Affiliation(s)
- Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China.
| | | | | | | |
Collapse
|
44
|
Gene transfer by chemical vectors, and endocytosis routes of polyplexes, lipoplexes and lipopolyplexes in a myoblast cell line. Biomaterials 2012; 33:2980-90. [PMID: 22243799 DOI: 10.1016/j.biomaterials.2011.12.027] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 12/13/2011] [Indexed: 11/23/2022]
Abstract
Chemical vectors are widely developed for providing safe DNA delivery systems. It is well admitted that their endocytosis and intracellular trafficking are critical for the transfection efficiency. Here, we have compared the endocytic pathways of lipoplexes, polyplexes and lipopolyplexes formed with carriers of various chemical compositions. Engineered C2C12 mouse myoblast cells expressing Rab5-EGFP, Rab7-EGFP or Cav1-GFP were used to monitor the location of the plasmid DNA into the endocytic compartments by real time fluorescence confocal microscopy. We observed that (i) DNA complexes made with dioleyl succinyl paromomycin:O,O-dioleyl-N-histamine phosphoramidate (DOSP/MM27) liposomes or histidinylated lPEI (His-lPEI) allowing the highest transfection efficiency displayed a positive ζ potential and were internalized by clathrin-mediated endocytosis, (ii) DOSP/MM27 lipoplexes were 6-times more internalized than His-lPEI polyplexes, (iii) all negatively charged DNA complexes lead to less efficient transfection and entered the cells via caveolae and (iv) lipopolyplexes allowing high transfection efficiency were weakly internalized via caveolae. Our results indicate that the transfection efficiency is better correlated with the nature of the endocytic pathway than with the uptake efficacy. This study shows also that engineered cells expressing specific fluorescent compartments are convenient tools to monitor endocytosis of a fluorescent plasmid DNA by real time fluorescence confocal microscopy.
Collapse
|
45
|
Fraix A, Montier T, Le Gall T, Sevrain CM, Carmoy N, Lindberg MF, Lehn P, Jaffrès PA. Lipothiophosphoramidates for gene delivery: critical role of the cationic polar headgroup. Org Biomol Chem 2012; 10:2051-8. [DOI: 10.1039/c2ob06812e] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Berchel M, Le Gall T, Couthon-Gourvès H, Haelters JP, Montier T, Midoux P, Lehn P, Jaffrès PA. Lipophosphonate/lipophosphoramidates: A family of synthetic vectors efficient for gene delivery. Biochimie 2012; 94:33-41. [DOI: 10.1016/j.biochi.2011.07.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 07/20/2011] [Indexed: 01/27/2023]
|
47
|
Perche F, Benvegnu T, Berchel M, Lebegue L, Pichon C, Jaffrès PA, Midoux P. Enhancement of dendritic cells transfection in vivo and of vaccination against B16F10 melanoma with mannosylated histidylated lipopolyplexes loaded with tumor antigen messenger RNA. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 7:445-53. [PMID: 21220051 DOI: 10.1016/j.nano.2010.12.010] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 12/05/2010] [Accepted: 12/29/2010] [Indexed: 01/24/2023]
Abstract
UNLABELLED We report the preparation of mannosylated nanoparticles loaded with messenger RNA (mRNA) that enhance the transfection of dendritic cells (DCs) in vivo and the anti-B16F10 melanoma vaccination in mice. Mannosylated and histidylated lipopolyplexes (Man(11)-LPR100) were obtained by adding mannosylated and histidylated liposomes to mRNA-PEGylated histidylated polylysine polyplexes. Upon intravenous injection, ∼9% of the radioactivity of technetium 99 m-labeled lipopolyplexes measured in the liver, spleen, lungs, and kidneys was found in the spleen. We demonstrate that spleen from mice injected with enhanced green fluorescent protein (EGFP) mRNA-loaded Man(11)-LPR100 contained four times more DCs expressing EGFP than that from mice injected with sugar-free LPR100. This better transfection of DCs is correlated with a better inhibition of B16F10 melanoma growth and an increased survival time when mice were immunized with MART-1 mRNA-loaded Man(11)-LPR100. These results indicate that mannosylated and histidylated LPR is an efficient system for the delivery of tumor antigen mRNA in splenic DCs aiming to induce an anticancer immune response. FROM THE CLINICAL EDITOR This paper discusses the preparation of mannosylated nanoparticles loaded with messenger RNA that enhance the transfection of dendritic cells (DCs) in vivo and the anti-B16F10 melanoma vaccination in mice. The authors describe an efficient system for the delivery of tumor antigen mRNA in splenic DCs aiming to induce an anticancer immune response.
Collapse
Affiliation(s)
- Federico Perche
- Centre de Biophysique Moléculaire CNRS UPR 4301, University and Inserm, Orléans, France
| | | | | | | | | | | | | |
Collapse
|