The Skin You Are In: Design-of-Experiments Optimization of Lipid Nanoparticle Self-Amplifying RNA Formulations in Human Skin ExplantsClick to copy article linkArticle link copied!
- Anna K. BlakneyAnna K. BlakneyDepartment of Medicine, Imperial College London, London, W21PG, United KingdomMore by Anna K. Blakney
- Paul F. McKayPaul F. McKayDepartment of Medicine, Imperial College London, London, W21PG, United KingdomMore by Paul F. McKay
- Bárbara Ibarzo YusBárbara Ibarzo YusDepartment of Medicine, Imperial College London, London, W21PG, United KingdomMore by Bárbara Ibarzo Yus
- Judith E. HunterJudith E. HunterDepartment of Plastic Surgery, Imperial NHS Trust, London, W68RF, United KingdomMore by Judith E. Hunter
- Elizabeth A. DexElizabeth A. DexDepartment of Plastic Surgery, Imperial NHS Trust, London, W68RF, United KingdomMore by Elizabeth A. Dex
- Robin J. Shattock*Robin J. Shattock*E-mail: [email protected]Department of Medicine, Imperial College London, London, W21PG, United KingdomMore by Robin J. Shattock
Abstract
Messenger RNA (mRNA) is a promising tool for biotherapeutics, and self-amplifying mRNA (saRNA) is particularly advantageous, because it results in abundant protein expression and production is easily scalable. While mRNA therapeutics have been shown to be highly effective in small animals, the outcomes do not scale linearly when these formulations are translated to dose-escalation studies in humans. Here, we utilize a design of experiments (DoE) approach to optimize the formulation of saRNA lipid nanoparticles in human skin explants. We first observed that luciferase expression from saRNA peaked after 11 days in human skin. Using DoE inputs of complexing lipid identity, lipid nanoparticle dose, lipid concentration, particle concentration, and ratio of zwitterionic to cationic lipids, we optimized the saRNA-induced luciferase expression in skin explants. Lipid identity and lipid concentration were found to be significant parameters in the DoE model, and the optimized formulation resulted in ∼7-fold increase in luciferase expression, relative to initial 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) formulation. Using flow cytometry, we observed that optimized formulations delivered the saRNA to ∼2% of the resident cells in the human skin explants. Although immune cells comprise only 7% of the total population of cells in skin, immune cells were found to express ∼50% of the RNA. This study demonstrates the powerful combination of using a DoE approach paired with clinically relevant human skin explants to optimize nucleic acid formulations. We expect that this system will be useful for optimizing both formulation and molecular designs of clinically translational nucleic acid vaccines and therapeutics.
Results and Discussion
Scheme 1
saRNA Luciferase Expression Kinetics in Human Skin Explants
Figure 1
Figure 1. Firefly luciferase expression in human skin explants over the course of 21 days after ID injection of three separate, simultaneous injections of 10 μg of saRNA with a mass ratio of lipid to RNA of 4:1 (w/w): (a) time course of ex vivo imaging of luciferase expressed by replicon RNA delivered with the initial formulation of DOTAP LNPs; and (b) quantification of luciferase expression, expressed as the mean total flux (p/s) ± standard deviation for n = 3.
Effects of Complexing Lipid Identity and Lipid-to-RNA Ratio on Luciferase Expression
Figure 2
Figure 2. Firefly luciferase expression in human skin explants injected intradermally with LNP formulations with varying lipid identity and LNP dose containing 2 μg of saRNA with medium particle concentration (108 particles/mL) at varying ratios of lipid to RNA (w/w): (a) ex vivo imaging of explants after 11 days and (b) quantification of luciferase imagine expressed as the mean total flux (p/s) ± standard deviation for n = 5.
Effects of Lipid and Particle Concentration on Luciferase Expression
Figure 3
Figure 3. Firefly luciferase expression in human skin explants injected intradermally with LNP formulations with varying lipid and particle concentrations containing 2 μg of saRNA with a ratio of total lipid to RNA of 90:1 (w/w): (a) ex vivo imaging of explants after 11 days and (b) quantification of luciferase imagine expressed as the mean total flux (p/s) ± standard deviation for n = 5. High and low particle concentrations are defined as 109 and 107 particles/mL, respectively, while high lipid concentration is defined as 7.5 mg/mL. H[L]/H[P] denotes high lipid concentration and high particle concentration; H[L]/L[P] denotes high lipid concentration and low particle concentration.
Effects of Combining Zwitterionic and Cationic Complexing Lipids on Luciferase Expression
Figure 4
Figure 4. Firefly luciferase expression in human skin explants injected intradermally with LNP formulations with varying ratios of cationic and zwitterionic lipids containing 2 μg of saRNA with a total lipid to RNA ratio of 18:1 (w/w) and medium particle concentration (108 particles/mL): (a) ex vivo imaging of explants after 11 days and (b) quantification of luciferase imagine expressed as mean total flux (p/s) ± standard deviation for n = 5.
Design of Experiments (DoE) Analysis
Figure 5
Figure 5. Standard least-squares effect screening DoE analysis of lipid nanoparticle formulation in human skin explants.
Figure 6
Figure 6. Comparison of fold change luciferase expression of tested LNP formulations normalized to original DOTAP formulation (blue bar). Values are expressed fold change total flux (p/s) ± standard deviation.
LNP Delivery and Expression of eGFP saRNA in Human Skin Cells
Figure 7
Figure 7. GFP expression in human skin cells after intradermal injection with LNP formulations, as determined using flow cytometry: (a) histogram of number of cells expressing GFP for each formulation, and (b) percentage of GFP-positive cells of total live cells for each sample. Bar represents the average ± standard deviation, with a significance of α = 0.05 indicated by an asterisk (*).
Identification of Cells Expressing eGFP in Human Skin Cells
Figure 8
Figure 8. Identity of cells present in human skin explants and GFP+ cells after ID injection of LNP formulations, as determined by flow cytometry: (a) identity of cells in the population of cells extracted from human skin explants, and (b) identity of GFP-expressing skin cells from explants treated with LNP-formulated RNA. The blue sections of each of the small pie charts indicate the total percentage of immune cells in the GFP+ cell population.
Conclusions
Methods
RNA Synthesis and Purification
Production of Lipid Nanoparticles
Particle Characterization
Human Skin Explant Injection, Culture, and Imaging
Flow Cytometry
Design of Experiment and Statistical Analysis
lipid identity | ratio of total lipid to RNA (w/w) | lipid concentration | particle concentration | ratio cationic to zwitterionic lipids(mol/mol) | ID # |
---|---|---|---|---|---|
C12–200 | 18:1 | medium | medium | – | 1 |
4:1 | medium | medium | – | 2 | |
1:1 | medium | medium | – | 3 | |
90:1 | high | high | 4 | ||
90:1 | high | low | 5 | ||
cephalin | 18:1 | medium | medium | – | 6 |
4:1 | medium | medium | – | 7 | |
1:1 | medium | medium | – | 8 | |
90:1 | high | high | 9 | ||
90:1 | high | low | 10 | ||
DDA | 18:1 | medium | medium | – | 11 |
4:1 | medium | medium | – | 12 | |
1:1 | medium | medium | – | 13 | |
18:1 | medium | medium | 10:1 | 14 | |
18:1 | medium | medium | 1:1 | 15 | |
0.1:1 | 16 | ||||
90:1 | high | high | 17 | ||
90:1 | high | low | 18 | ||
DOTAP | 18:1 | high/low | high/low | – | 19 |
4:1 | low | low | – | 20 | |
1:1 | low | low | – | 21 | |
18:1 | low | low | 10:1 | 22 | |
18:1 | low | low | 1:1 | 23 | |
18:1 | low | low | 0.1:1 | 24 | |
90:1 | high | high | 25 | ||
90:1 | high | low | 26 |
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsnano.9b01774.
LNP characterization of particle size and surface charge (Figure S1); eGFP expression gating strategy (Figure S2); skin explant cell viability (Figure S3); identity of cells present in total population and GFP+ cells (Figure S4); cell viability of human skin explants after LNP-formulation saRNA injection (Figure S5); antibodies used for flow cytometry (Table S1) (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We gratefully acknowledge the nursing team and patients at Charing Cross Hospital and Dormeur Investment Service Ltd. for providing funds to purchase equipment used in these studies.
References
This article references 37 other publications.
- 1Guan, S.; Rosenecker, J. Nanotechnologies in Delivery of mRNA Therapeutics Using Nonviral Vector-Based Delivery Systems. Gene Ther. 2017, 24, 133, DOI: 10.1038/gt.2017.5Google Scholar1Nanotechnologies in delivery of mRNA therapeutics using nonviral vector-based delivery systemsGuan, S.; Rosenecker, J.Gene Therapy (2017), 24 (3), 133-143CODEN: GETHEC; ISSN:0969-7128. (Nature Publishing Group)A review. Because of its safe and effective protein expression profile, in vitro transcribed mRNA (IVT-mRNA) represents a promising candidate in the development of novel therapeutics for genetic diseases, vaccines or gene editing strategies, esp. when its inherent shortcomings (for example, instability and immunogenicity) have been partially addressed via structural modifications. However, numerous unsolved tech. difficulties in successful in vivo delivery of IVT-mRNA have greatly hindered the applications of IVT-mRNA in clin. development. Recent advances in nanotechnol. and material science have yielded many promising nonviral delivery systems, some of which were able to efficiently facilitate targeted in vivo delivery of IVT-mRNA in safe and noninvasive manners. The diversity and flexibility of these delivery systems highlight the recent progress of IVT-mRNA-based therapy using nonviral vectors. In this review, we summarize recent advances of existing and emerging nonviral vector-based nanotechnologies for IVT-mRNA delivery and briefly summarize the interesting but rarely discussed applications on simultaneous delivery of IVT-mRNA with DNA.
- 2Karikó, K.; Muramatsu, H.; Welsh, F. A.; Ludwig, J.; Kato, H.; Akira, S.; Weissman, D. Incorporation of Pseudouridine into mRNA Yields Superior Nonimmunogenic Vector with Increased Translational Capacity and Biological Stability. Mol. Ther. 2008, 16, 1833– 1840, DOI: 10.1038/mt.2008.200Google Scholar2Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stabilityKariko, Katalin; Muramatsu, Hiromi; Welsh, Frank A.; Ludwig, Janos; Kato, Hiroki; Akira, Shizuo; Weissman, DrewMolecular Therapy (2008), 16 (11), 1833-1840CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)In vitro-transcribed mRNAs encoding physiol. important proteins have considerable potential for therapeutic applications. However, in its present form, mRNA is unfeasible for clin. use because of its labile and immunogenic nature. Here, we investigated whether incorporation of naturally modified nucleotides into transcripts would confer enhanced biol. properties to mRNA. We found that mRNAs contg. pseudouridines have a higher translational capacity than unmodified mRNAs when tested in mammalian cells and lysates or administered i.v. into mice at 0.015-0.15 mg/kg doses. The delivered mRNA and the encoded protein could be detected in the spleen at 1, 4, and 24 h after the injection, where both products were at significantly higher levels when pseudouridine-contg. mRNA was administered. Even at higher doses, only the unmodified mRNA was immunogenic, inducing high serum levels of interferon-α (IFN-α). These findings indicate that nucleoside modification is an effective approach to enhance stability and translational capacity of mRNA while diminishing its immunogenicity in vivo. Improved properties conferred by pseudouridine make such mRNA a promising tool for both gene replacement and vaccination.
- 3Thess, A.; Grund, S.; Mui, B. L.; Hope, M. J.; Baumhof, P.; Fotin-Mleczek, M.; Schlake, T. Sequence-Engineered mRNA without Chemical Nucleoside Modifications Enables an Effective Protein Therapy in Large Animals. Mol. Ther. 2015, 23, 1456– 1464, DOI: 10.1038/mt.2015.103Google Scholar3Sequence-engineered mRNA Without Chemical Nucleoside Modifications Enables an Effective Protein Therapy in Large AnimalsThess, Andreas; Grund, Stefanie; Mui, Barbara L.; Hope, Michael J.; Baumhof, Patrick; Fotin-Mleczek, Mariola; Schlake, ThomasMolecular Therapy (2015), 23 (9), 1456-1464CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)Being a transient carrier of genetic information, mRNA could be a versatile, flexible, and safe means for protein therapies. While recent findings highlight the enormous therapeutic potential of mRNA, evidence that mRNA-based protein therapies are feasible beyond small animals such as mice is still lacking. Previous studies imply that mRNA therapeutics require chem. nucleoside modifications to obtain sufficient protein expression and avoid activation of the innate immune system. Here we show that chem. unmodified mRNA can achieve those goals as well by applying sequence-engineered mols. Using erythropoietin (EPO) driven prodn. of red blood cells as the biol. model, engineered Epo mRNA elicited meaningful physiol. responses from mice to nonhuman primates. Even in pigs of about 20 kg in wt., a single adequate dose of engineered mRNA encapsulated in lipid nanoparticles (LNPs) induced high systemic Epo levels and strong physiol. effects. Our results demonstrate that sequence-engineered mRNA has the potential to revolutionize human protein therapies.
- 4Kauffman, K. J.; Webber, M. J.; Anderson, D. G. Materials for Non-Viral Intracellular Delivery of Messenger RNA Therapeutics. J. Controlled Release 2016, 240, 227– 234, DOI: 10.1016/j.jconrel.2015.12.032Google Scholar4Materials for non-viral intracellular delivery of messenger RNA therapeuticsKauffman, Kevin J.; Webber, Matthew J.; Anderson, Daniel G.Journal of Controlled Release (2016), 240 (), 227-234CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Though therapeutics based on mRNA (mRNA) have broad potential in applications such as protein replacement therapy, cancer immunotherapy, and genomic engineering, their effective intracellular delivery remains a challenge. A chem. diverse suite of delivery materials with origins as materials for cellular transfection of DNA and small interfering RNAs (siRNAs) has recently been reported to have promise as non-viral delivery agents for mRNA. These materials include covalent conjugates, protamine complexes, nanoparticles based on lipids or polymers, and hybrid formulations. This review will highlight the use of delivery materials for mRNA, with a specific focus on their mechanisms of action, routes of administration, and dosages. Addnl., strategies in which these materials can be adapted and optimized to address challenges specific to mRNA delivery are also discussed. The technologies included have shown varying promise for therapeutic use, specifically having been used to deliver mRNA in vivo or exhibiting characteristics that could make in vivo use a possibility. In so doing, it is the intention of this review to provide a comprehensive look at the progress and possibilities in applying nucleic acid delivery technol. specifically toward the emerging area of mRNA therapeutics.
- 5Jensen, S.; Thomsen, A. R. Sensing of RNA Viruses: A Review of Innate Immune Receptors Involved in Recognizing RNA Virus Invasion. J. Virol. 2012, 86, 2900, DOI: 10.1128/JVI.05738-11Google Scholar5Sensing of RNA viruses: a review of innate immune receptors involved in recognizing RNA virus invasionJensen, Soeren; Thomsen, Allan RandrupJournal of Virology (2012), 86 (6), 2900-2910CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)A review. Our knowledge regarding the contribution of the innate immune system in recognizing and subsequently initiating a host response to an invasion of RNA virus has been rapidly growing over the last decade. Descriptions of the receptors involved and the mol. mechanisms they employ to sense viral pathogen-assocd. mol. patterns have emerged in great detail. This review presents an overview of our current knowledge regarding the receptors used to detect RNA virus invasion, the mol. structures these receptors sense, and the involved downstream signaling pathways.
- 6Kawai, T.; Akira, S. The Role of Pattern-Recognition Receptors in Innate Immunity: Update on Toll-Like Receptors. Nat. Immunol. 2010, 11, 373, DOI: 10.1038/ni.1863Google Scholar6The role of pattern-recognition receptors in innate immunity: update on Toll-like receptorsKawai, Taro; Akira, ShizuoNature Immunology (2010), 11 (5), 373-384CODEN: NIAMCZ; ISSN:1529-2908. (Nature Publishing Group)A review. The discovery of Toll-like receptors (TLRs) as components that recognize conserved structures in pathogens has greatly advanced understanding of how the body senses pathogen invasion, triggers innate immune responses and primes antigen-specific adaptive immunity. Although TLRs are crit. for host defense, it has become apparent that loss of neg. regulation of TLR signaling, as well as recognition of self mols. by TLRs, are strongly assocd. with the pathogenesis of inflammatory and autoimmune diseases. Furthermore, it is now clear that the interaction between TLRs and recently identified cytosolic innate immune sensors is crucial for mounting effective immune responses. Here we describe the recent advances that have been made by research into the role of TLR biol. in host defense and disease.
- 7Perri, S.; Greer, C. E.; Thudium, K.; Doe, B.; Legg, H.; Liu, H.; Romero, R. E.; Tang, Z.; Bin, Q.; Dubensky, T. W.; Vajdy, M.; Otten, G. R.; Polo, J. M. An Alphavirus Replicon Particle Chimera Derived from Venezuelan Equine Encephalitis and Sindbis Viruses Is a Potent Gene-Based Vaccine Delivery Vector. J. Virol. 2003, 77, 10394– 10403, DOI: 10.1128/JVI.77.19.10394-10403.2003Google Scholar7An alphavirus replicon particle chimera derived from Venezuelan equine encephalitis and Sindbis viruses is a potent gene-based vaccine delivery vectorPerri, Silvia; Greer, Catherine E.; Thudium, Kent; Doe, Barbara; Legg, Harold; Liu, Hong; Romero, Raul E.; Tang, Zequn; Bin, Qian; Dubensky, Thomas W., Jr.; Vajdy, Michael; Otten, Gillis R.; Polo, John M.Journal of Virology (2003), 77 (19), 10394-10403CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Alphavirus replicon particle-based vaccine vectors derived from Sindbis virus (SIN), Semliki Forest virus, and Venezuelan equine encephalitis virus (VEE) have been shown to induce robust antigen-specific cellular, humoral, and mucosal immune responses in many animal models of infectious disease and cancer. However, since little is known about the relative potencies among these different vectors, we compared the immunogenicity of replicon particle vectors derived from two very different parental alphaviruses, VEE and SIN, expressing a human immunodeficiency virus type 1 p55Gag antigen. Moreover, to explore the potential benefits of combining elements from different alphaviruses, we generated replicon particle chimeras of SIN and VEE. Two distinct strategies were used to produce particles with VEE-p55gag replicon RNA packaged within SIN envelope glycoproteins and SIN-p55gag replicon RNA within VEE envelope glycoproteins. Each replicon particle configuration induced Gag-specific CD8+ T-cell responses in murine models when administered alone or after priming with DNA. However, Gag-specific responses varied dramatically, with the strongest responses to this particular antigen correlating with the VEE replicon RNA, irresp. of the source of envelope glycoproteins. Comparing the replicons with respect to heterologous gene expression levels and sensitivity to alpha/beta interferon in cultured cells indicated that each might contribute to potency differences. This work shows that combining desirable elements from VEE and SIN into a replicon particle chimera may be a valuable approach toward the goal of developing vaccine vectors with optimal in vivo potency, ease of prodn., and safety.
- 8Chahal, J. S.; Khan, O. F.; Cooper, C. L.; McPartlan, J. S.; Tsosie, J. K.; Tilley, L. D.; Sidik, S. M.; Lourido, S.; Langer, R.; Bavari, S.; Ploegh, H. L.; Anderson, D. G. Dendrimer-RNA Nanoparticles Generate Protective Immunity against Lethal Ebola, H1N1 Influenza, and Toxoplasma Gondii Challenges with a Single Dose. Proc. Natl. Acad. Sci. U. S. A. 2016, 113, E4133– E4142, DOI: 10.1073/pnas.1600299113Google Scholar8Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and Toxoplasma gondii challenges with a single doseChahal, Jasdave S.; Khan, Omar F.; Cooper, Christopher L.; McPartlan, Justine S.; Tsosie, Jonathan K.; Tilley, Lucas D.; Sidik, Saima M.; Lourido, Sebastian; Langer, Robert; Bavari, Sina; Ploegh, Hidde L.; Anderson, Daniel G.Proceedings of the National Academy of Sciences of the United States of America (2016), 113 (29), E4133-E4142CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Vaccines have had broad medical impact, but existing vaccine technologies and prodn. methods are limited in their ability to respond rapidly to evolving and emerging pathogens, or sudden outbreaks. Here, the authors develop a rapid-response, fully synthetic, single-dose, adjuvant-free dendrimer nanoparticle vaccine platform wherein antigens are encoded by encapsulated mRNA replicons. To the authors' knowledge, this system is the first capable of generating protective immunity against a broad spectrum of lethal pathogen challenges, including H1N1 influenza, Toxoplasma gondii, and Ebola virus. The vaccine can be formed with multiple antigen-expressing replicons, and is capable of eliciting both CD8+ T-cell and antibody responses. The ability to generate viable, contaminant-free vaccines within days, to single or multiple antigens, may have broad utility for a range of diseases.
- 9Geall, A. J.; Verma, A.; Otten, G. R.; Shaw, C. A.; Hekele, A.; Banerjee, K.; Cu, Y.; Beard, C. W.; Brito, L. A.; Krucker, T.; O’Hagan, D. T.; Singh, M.; Mason, P. W.; Valiante, N. M.; Dormitzer, P. R.; Barnett, S. W.; Rappuoli, R.; Ulmer, J. B.; Mandl, C. W. Nonviral Delivery of Self-Amplifying RNA Vaccines. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 14604, DOI: 10.1073/pnas.1209367109Google Scholar9Nonviral delivery of self-amplifying RNA vaccinesGeall, Andrew J.; Verma, Ayush; Otten, Gillis R.; Shaw, Christine A.; Hekele, Armin; Banerjee, Kaustuv; Cu, Yen; Beard, Clayton W.; Brito, Luis A.; Krucker, Thomas; O'Hagan, Derek T.; Singh, Manmohan; Mason, Peter W.; Valiante, Nicholas M.; Dormitzer, Philip R.; Barnett, Susan W.; Rappuoli, Rino; Ulmer, Jeffrey B.; Mandl, Christian W.Proceedings of the National Academy of Sciences of the United States of America (2012), 109 (36), 14604-14609, S14604/1-S14604/7CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Despite more than two decades of research and development on nucleic acid vaccines, there is still no com. product for human use. Taking advantage of the recent innovations in systemic delivery of short interfering RNA (siRNA) using lipid nanoparticles (LNPs), we developed a self-amplifying RNA vaccine. Here we show that nonviral delivery of a 9-kb self-amplifying RNA encapsulated within an LNP substantially increased immunogenicity compared with delivery of unformulated RNA. This unique vaccine technol. was found to elicit broad, potent, and protective immune responses, that were comparable to a viral delivery technol., but without the inherent limitations of viral vectors. Given the many pos. attributes of nucleic acid vaccines, our results suggest that a comprehensive evaluation of nonviral technologies to deliver self-amplifying RNA vaccines is warranted.
- 10Bogers, W. M.; Oostermeijer, H.; Mooij, P.; Koopman, G.; Verschoor, E. J.; Davis, D.; Ulmer, J. B.; Brito, L. A.; Cu, Y.; Banerjee, K.; Otten, G. R.; Burke, B.; Dey, A.; Heeney, J. L.; Shen, X.; Tomaras, G. D.; Labranche, C.; Montefiori, D. C.; Liao, H.-X.; Haynes, B.; Geall, A. J.; Barnett, S. W. Potent Immune Responses in Rhesus Macaques Induced by Nonviral Delivery of a Self-Amplifying RNA Vaccine Expressing HIV Type 1 Envelope with a Cationic Nanoemulsion. J. Infect. Dis. 2015, 211, 947– 955, DOI: 10.1093/infdis/jiu522Google Scholar10Potent immune responses in rhesus macaques induced by nonviral delivery of a selfamplifying RNA vaccine expressing HIV type 1 envelope with a cationic nanoemulsionBogers, Willy M.; Oostermeijer, Herman; Mooij, Petra; Koopman, Gerrit; Verschoor, Ernst J.; Davis, David; Ulmer, Jeffrey B.; Brito, Luis A.; Cu, Yen; Banerjee, Kaustuv; Otten, Gillis R.; Burke, Brian; Dey, Antu; Heeney, Jonathan L.; Shen, Xiaoying; Tomaras, Georgia D.; Labranche, Celia; Montefiori, David C.; Liao, Hua-Xin; Haynes, Barton; Geall, Andrew J.; Barnett, Susan W.Journal of Infectious Diseases (2015), 211 (6), 947-955CODEN: JIDIAQ; ISSN:0022-1899. (Oxford University Press)Self-amplifying mRNA (mRNA) of pos.-strand RNA viruses are effective vectors for in situ expression of vaccine antigens and have potential as a new vaccine technol. platform well suited for global health applications. The SAM vaccine platform is based on a synthetic, self-amplifying mRNA delivered by a nonviral delivery system. The safety and immunogenicity of an HIV SAM vaccine encoding a clade C envelope glycoprotein formulated with a cationic nanoemulsion (CNE) delivery system was evaluated in rhesus macaques. The HIV SAM vaccine induced potent cellular immune responses that were greater in magnitude than those induced by self-amplifying mRNA packaged in a viral replicon particle (VRP) or by a recombinant HIV envelope protein formulated with MF59 adjuvant, anti-envelope binding (including anti-V1V2), and neutralizing antibody responses that exceeded those induced by the VRP vaccine. These studies provide the first evidence in nonhuman primates that HIV vaccination with a relatively low dose (50 μg) of formulated self-amplifying mRNA is safe and immunogenic.
- 11Perche, F.; Benvegnu, T.; Berchel, M.; Lebegue, L.; Pichon, C.; Jaffrès, P.-A.; 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 (N. Y., NY, U. S.) 2011, 7, 445– 453, DOI: 10.1016/j.nano.2010.12.010Google ScholarThere is no corresponding record for this reference.
- 12Uchida, S.; Kinoh, H.; Ishii, T.; Matsui, A.; Tockary, T. A.; Takeda, K. M.; Uchida, H.; Osada, K.; Itaka, K.; Kataoka, K. Systemic Delivery of Messenger RNA for the Treatment of Pancreatic Cancer Using Polyplex Nanomicelles with a Cholesterol Moiety. Biomaterials 2016, 82, 221– 228, DOI: 10.1016/j.biomaterials.2015.12.031Google Scholar12Systemic delivery of messenger RNA for the treatment of pancreatic cancer using polyplex nanomicelles with a cholesterol moietyUchida, Satoshi; Kinoh, Hiroaki; Ishii, Takehiko; Matsui, Akitsugu; Tockary, Theofilus Agrios; Takeda, Kaori Machitani; Uchida, Hirokuni; Osada, Kensuke; Itaka, Keiji; Kataoka, KazunoriBiomaterials (2016), 82 (), 221-228CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)Systemic delivery of mRNA (mRNA) is tech. challenging because mRNA is highly susceptible to enzymic degrdn. in the blood circulation. In this study, we used a nanomicelle-based platform, prepd. from mRNA and poly(ethylene glycol) (PEG)-polycation block copolymers. A cholesterol (Chol) moiety was attached to the ω-terminus of the block copolymer to increase the stability of the nanomicelle by hydrophobic interaction. After in vitro screening, polyaspartamide with four aminoethylene repeats in its side chain (PAsp(TEP)) was selected as the cationic segment of the block copolymer, because it contributes to enhance nuclease resistance and high protein expression from the mRNA. After i.v. injection, PEG-PAsp(TEP)-Chol nanomicelles showed significantly enhanced blood retention of mRNA in comparison to nanomicelles without Chol. We used the nanomicelles for treating intractable pancreatic cancer in a s.c. inoculation mouse model through the delivery of mRNA encoding an anti-angiogenic protein (sFlt-1). PEG-PAsp(TEP)-Chol nanomicelles generated efficient protein expression from the delivered mRNA in tumor tissue, resulting in remarkable inhibition of the tumor growth, whereas nanomicelles without Chol failed to show a detectable therapeutic effect. In conclusion, the stabilized nanomicelle system led to the successful systemic delivery of mRNA in therapeutic application, holding great promise for the treatment of various diseases.
- 13Fotin-Mleczek, M.; Zanzinger, K.; Heidenreich, R.; Lorenz, C.; Kowalczyk, A.; Kallen, K.-J.; Huber, S. M. mRNA-Based Vaccines Synergize with Radiation Therapy to Eradicate Established Tumors. Radiat. Oncol. 2014, 9, 180, DOI: 10.1186/1748-717X-9-180Google Scholar13mRNA-based vaccines synergize with radiation therapy to eradicate established tumorsFotin-Mleczek, Mariola; Zanzinger, Kai; Heidenreich, Regina; Lorenz, Christina; Kowalczyk, Aleksandra; Kallen, Karl-Josef; Huber, Stephan M.Radiation Oncology (2014), 9 (), 180/1-180/23CODEN: ROANCM; ISSN:1748-717X. (BioMed Central Ltd.)Background: The eradication of large, established tumors by active immunotherapy is a major challenge because of the numerous cancer evasion mechanisms that exist. This study aimed to establish a novel combination therapy consisting of mRNA (mRNA)-based cancer vaccines and radiation, which would facilitate the effective treatment of established tumors with aggressive growth kinetics. Methods: The combination of a tumor-specific mRNA-based vaccination with radiation was tested in two syngeneic tumor models, a highly immunogenic E.G7-OVA and a low immunogenic Lewis lung cancer (LLC). The mol. mechanism induced by the combination therapy was evaluated via gene expression arrays as well as flow cytometry analyses of tumor infiltrating cells. Results: In both tumor models we demonstrated that a combination of mRNA-based immunotherapy with radiation results in a strong synergistic anti-tumor effect. This was manifested as either complete tumor eradication or delay in tumor growth. Gene expression anal. of mouse tumors revealed a variety of substantial changes at the tumor site following radiation. Genes assocd. with antigen presentation, infiltration of immune cells, adhesion, and activation of the innate immune system were upregulated. A combination of radiation and immunotherapy induced significant downregulation of tumor assocd. factors and upregulation of tumor suppressors. Moreover, combination therapy significantly increased CD4+, CD8+ and NKT cell infiltration of mouse tumors. Conclusion: Our data provide a scientific rationale for combining immunotherapy with radiation and provide a basis for the development of more potent anti-cancer therapies.
- 14McCullough, K. C.; 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. Mol. Ther.--Nucleic Acids 2014, 3, e173 DOI: 10.1038/mtna.2014.24Google Scholar14Self-replicating Replicon-RNA Delivery to Dendritic Cells by Chitosan-nanoparticles for Translation In Vitro and In VivoMcCullough, Kenneth C.; Bassi, Isabelle; Milona, Panagiota; Suter, Rolf; Thomann-Harwood, Lisa; Englezou, Pavlos; Demoulins, Thomas; Ruggli, NicolasMolecular Therapy--Nucleic Acids (2014), 3 (7), e173CODEN: MTAOC5; ISSN:2162-2531. (Nature Publishing Group)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 assocn. 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 concn.-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 assocd. with chitosan-nanoparticle delivery of self-amplifying RepRNA to DCs, leading to translation of encoded foreign genes, namely influenza virus hemagglutinin and nucleoprotein.
- 15Hekele, A.; Bertholet, S.; Archer, J.; Gibson, D. G.; Palladino, G.; Brito, L. A.; Otten, G. R.; Brazzoli, M.; Buccato, S.; Bonci, A.; Casini, D.; Maione, D.; Qi, Z.-Q.; Gill, J. E.; Caiazza, N. C.; Urano, J.; Hubby, B.; Gao, G. F.; Shu, Y.; De Gregorio, E.; Mandl, C. W.; Mason, P. W.; Settembre, E. C.; Ulmer, J. B.; Craig Venter, J.; Dormitzer, P. R.; Rappuoli, R.; Geall, A. J. Rapidly Produced SAM(®) Vaccine against H7N9 Influenza Is Immunogenic in Mice. Emerging Microbes Infect. 2013, 2, 1– 7, DOI: 10.1038/emi.2013.54Google ScholarThere is no corresponding record for this reference.
- 16Brazzoli, M.; Magini, D.; Bonci, A.; Buccato, S.; Giovani, C.; Kratzer, R.; Zurli, V.; Mangiavacchi, S.; Casini, D.; Brito, L. M.; De Gregorio, E.; Mason, P. W.; Ulmer, J. B.; Geall, A. J.; Bertholet, S. Induction of Broad-Based Immunity and Protective Efficacy by Self-Amplifying mRNA Vaccines Encoding Influenza Virus Hemagglutinin. J. Virol. 2016, 90, 332, DOI: 10.1128/JVI.01786-15Google Scholar16Induction of broad-based immunity and protective efficacy by selfamplifying mRNA vaccines encoding influenza virus hemagglutininBrazzoli, Michela; Magini, Diletta; Bonci, Alessandra; Buccato, Scilla; Giovani, Cinzia; Kratzer, Roland; Zurli, Vanessa; Mangiavacchi, Simona; Casini, Daniele; Brito, Luis M.; De Gregorio, Ennio; Mason, Peter W.; Ulmer, Jeffrey B.; Geall, Andrew J.; Bertholet, SylvieJournal of Virology (2016), 90 (1), 332-344CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)Seasonal influenza is a vaccine-preventable disease that remains a major health problem worldwide, esp. in immunocompromised populations. The impact of influenza disease is even greater when strains drift, and influenza pandemics can result when animal-derived influenza virus strains combine with seasonal strains. In this study, we used the SAM technol. and characterized the immunogenicity and efficacy of a self-amplifying mRNA expressing influenza virus hemagglutinin (HA) antigen [SAM(HA)] formulated with a novel oil-in-water cationic nanoemulsion. We demonstrated that SAM(HA) was immunogenic in ferrets and facilitated containment of viral replication in the upper respiratory tract of influenza virus-infected animals. In mice, SAM(HA) induced potent functional neutralizing antibody and cellular immune responses, characterized by HA-specific CD4 T helper 1 and CD8 cytotoxic T cells. Furthermore, mice immunized with SAM(HA) derived from the influenza A virus A/California/7/2009 (H1N1) strain (Cal) were protected from a lethal challenge with the heterologous mouse-adapted A/PR/8/1934 (H1N1) virus strain (PR8). Sera derived from SAM(H1-Cal)-immunized animals were not cross-reactive with the PR8 virus, whereas cross-reactivity was obsd. for HA-specific CD4 and CD8 T cells. Finally, depletion of T cells demonstrated that T-cell responses were essential in mediating heterologous protection. If the SAM vaccine platform proves safe, well tolerated, and effective in humans, the fully synthetic SAM vaccine technol. could provide a rapid response platform to control pandemic influenza.
- 17Kauffman, K. J.; Dorkin, J. R.; Yang, J. H.; Heartlein, M. W.; DeRosa, F.; Mir, F. F.; Fenton, O. S.; Anderson, D. G. Optimization of Lipid Nanoparticle Formulations for mRNA Delivery In Vivo with Fractional Factorial and Definitive Screening Designs. Nano Lett. 2015, 15, 7300– 7306, DOI: 10.1021/acs.nanolett.5b02497Google Scholar17Optimization of Lipid Nanoparticle Formulations for mRNA Delivery in Vivo with Fractional Factorial and Definitive Screening DesignsKauffman, Kevin J.; Dorkin, J. Robert; Yang, Jung H.; Heartlein, Michael W.; De Rosa, Frank; Mir, Faryal F.; Fenton, Owen S.; Anderson, Daniel G.Nano Letters (2015), 15 (11), 7300-7306CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)Intracellular delivery of mRNA (mRNA) has the potential to induce protein prodn. for many therapeutic applications. Although lipid nanoparticles have shown considerable promise for the delivery of small interfering RNAs (siRNA), their utility as agents for mRNA delivery has only recently been investigated. The most common siRNA formulations contain four components: an amine-contg. lipid or lipid-like material, phospholipid, cholesterol, and lipid-anchored polyethylene glycol, the relative ratios of which can have profound effects on the formulation potency. Here, we develop a generalized strategy to optimize lipid nanoparticle formulations for mRNA delivery to the liver in vivo using Design of Expt. (DOE) methodologies including Definitive Screening and Fractional Factorial Designs. By simultaneously varying lipid ratios and structures, we developed an optimized formulation which increased the potency of erythropoietin-mRNA-loaded C12-200 lipid nanoparticles 7-fold relative to formulations previously used for siRNA delivery. Key features of this optimized formulation were the incorporation of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and increased ionizable lipid:mRNA wt. ratios. Interestingly, the optimized lipid nanoparticle formulation did not improve siRNA delivery, indicating differences in optimized formulation parameter design spaces for siRNA and mRNA. We believe the general method described here can accelerate in vivo screening and optimization of nanoparticle formulations with large multidimensional design spaces.
- 18Bahl, K.; Senn, J. J.; Yuzhakov, O.; Bulychev, A.; Brito, L. A.; Hassett, K. J.; Laska, M. E.; Smith, M.; Almarsson, Ö.; Thompson, J.; Ribeiro, A.; Watson, M.; Zaks, T.; Ciaramella, G. Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza Viruses. Mol. Ther. 2017, 25, 1316– 1327, DOI: 10.1016/j.ymthe.2017.03.035Google Scholar18Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza VirusesBahl, Kapil; Senn, Joe J.; Yuzhakov, Olga; Bulychev, Alex; Brito, Luis A.; Hassett, Kimberly J.; Laska, Michael E.; Smith, Mike; Almarsson, Orn; Thompson, James; Ribeiro, Amilcar; Watson, Mike; Zaks, Tal; Ciaramella, GiuseppeMolecular Therapy (2017), 25 (6), 1316-1327CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)Recently, the World Health Organization confirmed 120 new human cases of avian H7N9 influenza in China resulting in 37 deaths, highlighting the concern for a potential pandemic and the need for an effective, safe, and high-speed vaccine prodn. platform. Prodn. speed and scale of mRNA-based vaccines make them ideally suited to impede potential pandemic threats. Here we show that lipid nanoparticle (LNP)-formulated, modified mRNA vaccines, encoding hemagglutinin (HA) proteins of H10N8 (A/Jiangxi-Donghu/346/2013) or H7N9 (A/Anhui/1/2013), generated rapid and robust immune responses in mice, ferrets, and nonhuman primates, as measured by hemagglutination inhibition (HAI) and microneutralization (MN) assays. A single dose of H7N9 mRNA protected mice from a lethal challenge and reduced lung viral titers in ferrets. Interim results from a first-in-human, escalating-dose, phase 1 H10N8 study show very high seroconversion rates, demonstrating robust prophylactic immunity in humans. Adverse events (AEs) were mild or moderate with only a few severe and no serious events. These data show that LNP-formulated, modified mRNA vaccines can induce protective immunogenicity with acceptable tolerability profiles.
- 19van den Berg, J. H.; Nuijen, B.; Beijnen, J. H.; Vincent, A.; van Tinteren, H.; Kluge, J.; Woerdeman, L. A. E.; Hennink, W. E.; Storm, G.; Schumacher, T. N.; Haanen, J. B. A. G. Optimization of Intradermal Vaccination by DNA Tattooing in Human Skin. Hum. Gene Ther. 2009, 20, 181– 189, DOI: 10.1089/hum.2008.073Google Scholar19Optimization of Intradermal Vaccination by DNA Tattooing in Human Skinvan den Berg, Joost H.; Nuijen, Bastiaan; Beijnen, Jos H.; Vincent, Andrew; van Tinteren, Harm; Kluge, Joern; Woerdeman, Leonie A. E.; Hennink, Wim E.; Storm, Gert; Schumacher, Ton N.; Haanen, John B. A. G.Human Gene Therapy (2009), 20 (3), 181-189CODEN: HGTHE3; ISSN:1043-0342. (Mary Ann Liebert, Inc.)The intradermal administration of DNA vaccines by tattooing is a promising delivery technique for genetic immunization, with proven high immunogenicity in mice and in nonhuman primates. However, the parameters that result in optimal expression of DNA vaccines that are applied by this strategy to human skin are currently unknown. To address this issue we set up an ex vivo human skin model in which DNA vaccine-induced expression of reporter proteins could be monitored longitudinally. Using this model we demonstrate the following: First, the vast majority of cells that express DNA vaccine-encoded antigen in human skin are formed by epidermal keratinocytes, with only a small fraction (about 1%) of antigen-pos. epidermal Langerhans cells. Second, using full randomization of DNA tattoo variables we show that an increase in DNA concn., needle depth, and tattoo time all significantly increase antigen expression (p < 0.001), with DNA concn. forming the most crit. variable influencing the level of antigen expression. Finally, in spite of the marked immunogenicity of this vaccination method in animal models, transfection efficiency of the technique is shown to be extremely low, estd. at approx. 2 to 2000 out of 1 × 1010 copies of plasmid applied. This finding, coupled with the obsd. dependency of antigen expression on DNA concn., suggests that the development of strategies that can enhance in vivo transfection efficacy would be highly valuable. Collectively, this study shows that an ex vivo human skin model can be used to det. the factors that control vaccine-induced antigen expression and define the optimal parameters for the evaluation of DNA tattoo or other dermal delivery techniques in phase 1 clin. trials.
- 20Gelfant, S. Of Mice and Men” the Cell Cycle in Human Epidermis In Vivo. J. Invest. Dermatol. 1982, 78, 296– 299, DOI: 10.1111/1523-1747.ep12507367Google Scholar20"Of mice and men" the cell cycle in human epidermis in vivoGelfant SThe Journal of investigative dermatology (1982), 78 (4), 296-9 ISSN:0022-202X.This article deals with and compares cell cycle information obtained in mouse and in human epidermis in vivo. In order to compare data in mouse and in man, DNA labeling and mitotic index experiments were performed to obtain cell cycle information in normal human epidermis in vivo. Experiments were also performed on genetically inbred and outbred strains of mice--to provide a clue to the differences observed between mouse and man. The article makes the following points: 1. In contrast to mouse epidermis, there are no consistent diurnal fluctuations in and there is no cell kinetic relationship between mitotic and DNA-labeling indices in normal human epidermis in vivo. The variability from individual to individual in human subjects and the lack of cell cycle-related circadian fluctuations, preclude the use of statistical analysis and the use of conventional cell kinetic principles in understanding epidermal cell proliferation in man and may preclude the use of circadian rhythmicity for therapy scheduling. 2. The consistent and intelligible cell cycle information obtained in laboratory mice (as compared to man) is not due to the genetically inbred condition of mice. 3. This report introduces the use of ambient temperature as a potential nontoxic cell cycle tool for manipulating epidermal cell proliferation in the therapy of human proliferative skin diseases.
- 21Love, K. T.; Mahon, K. P.; Levins, C. G.; Whitehead, K. A.; Querbes, W.; Dorkin, J. R.; Qin, J.; Cantley, W.; Qin, L. L.; Racie, T.; Frank-Kamenetsky, M.; Yip, K. N.; Alvarez, R.; Sah, D. W. Y.; de Fougerolles, A.; Fitzgerald, K.; Koteliansky, V.; Akinc, A.; Langer, R.; Anderson, D. G. Lipid-Like Materials for Low-Dose, In Vivo Gene Silencing. Proc. Natl. Acad. Sci. U. S. A. 2010, 107, 1864– 1869, DOI: 10.1073/pnas.0910603106Google Scholar21Lipid-like materials for low-dose, in vivo gene silencingLove, Kevin T.; Mahon, Kerry P.; Levins, Christopher G.; Whitehead, Kathryn A.; Querbes, William; Dorkin, J. Robert; Qin, June; Cantley, William; Qin, Liu Liang; Racie, Timothy; Frank-Kamenetsky, Maria; Yip, Ka Ning; Alvarez, Rene; Sah, Dinah W. Y.; de Fougerolles, Antonin; Fitzgerald, Kevin; Koteliansky, Victor; Akinc, Akin; Langer, Robert; Anderson, Daniel G.Proceedings of the National Academy of Sciences of the United States of America (2010), 107 (5), 1864-1869, S1864/1-S1864/6CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Significant effort has been applied to discover and develop vehicles which can guide small interfering RNAs (siRNA) through the many barriers guarding the interior of target cells. While studies have demonstrated the potential of gene silencing in vivo, improvements in delivery efficacy are required to fulfill the broadest potential of RNA interference therapeutics. Through the combinatorial synthesis and screening of a different class of materials, a formulation has been identified that enables siRNA-directed liver gene silencing in mice at doses below 0.01 mg/kg. This formulation was also shown to specifically inhibit expression of five hepatic genes simultaneously, after a single injection. The potential of this formulation was further validated in nonhuman primates, where high levels of knockdown of the clin. relevant gene transthyretin was obsd. at doses as low as 0.03 mg/kg. To our knowledge, this formulation facilitates gene silencing at orders-of-magnitude lower doses than required by any previously described siRNA liver delivery system.
- 22Ball, R. L.; Hajj, K. A.; Vizelman, J.; Bajaj, P.; Whitehead, K. A. Lipid Nanoparticle Formulations for Enhanced Co-Delivery of siRNA and mRNA. Nano Lett. 2018, 18, 3814– 3822, DOI: 10.1021/acs.nanolett.8b01101Google Scholar22Lipid nanoparticle formulations for enhanced co-delivery of siRNA and mRNABall, Rebecca L.; Hajj, Khalid A.; Vizelman, Jamie; Bajaj, Palak; Whitehead, Kathryn A.Nano Letters (2018), 18 (6), 3814-3822CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)Although mRNA and siRNA have significant therapeutic potential, their simultaneous delivery has not been previously explored. To facilitate the treatment of diseases assocd. with aberrant gene upregulation and downregulation, we sought to co-formulate siRNA and mRNA in a single lipidoid nanoparticle (LNP) formulation. We accommodated the distinct mol. characteristics of mRNA and siRNA in a formulation consisting of an ionizable and biodegradable amine-contg. lipidoid, cholesterol, DSPC, DOPE, and PEG-lipid. Surprisingly, the co-formulation of siRNA and mRNA in the same LNP enhanced the efficacy of both drugs in vitro and in vivo. Compared to LNPs encapsulating siRNA only, co-formulated LNPs improved Factor VII gene silencing in mice from 44 to 87% at an siRNA dose of 0.03 mg/kg. Co-formulation also improved mRNA delivery, as a 0.5 mg/kg dose of mRNA co-formulated with siRNA induced three times the luciferase protein expression compared to when siRNA was not included. As not all gene therapy applications require both RNA drugs, we sought to extend the benefit of co-formulated LNPs to formulations encapsulating only a single type of RNA. We accomplished this by substituting the "helper" RNA with a neg. charged polymer, polystyrenesulfonate (PSS). LNPs contg. PSS mediated the same level of protein silencing or expression as std. LNPs using 2-3-fold less RNA. For example, LNPs formulated with and without PSS induced 50% Factor VII silencing at siRNA doses of 0.01 and 0.03 mg/kg, resp. Together, these studies demonstrate potent co-delivery of siRNA and mRNA and show that inclusion of a neg. charged "helper polymer" enhances the efficacy of LNP delivery systems.
- 23Henriksen-Lacey, M.; Christensen, D.; Bramwell, V. W.; Lindenstrøm, T.; Agger, E. M.; Andersen, P.; Perrie, Y. Comparison of the Depot Effect and Immunogenicity of Liposomes Based on Dimethyldioctadecylammonium (DDA), 3β-[N-(N′,N′-Dimethylaminoethane)Carbomyl] Cholesterol (DC-Chol), and 1,2-Dioleoyl-3-Trimethylammonium Propane (DOTAP): Prolonged Liposome Retention Mediates Stronger Th1 Responses. Mol. Pharmaceutics 2011, 8, 153– 161, DOI: 10.1021/mp100208fGoogle Scholar23Comparison of the Depot Effect and Immunogenicity of Liposomes Based on Dimethyldioctadecylammonium (DDA), 3β-[N-(N',N'-Dimethylaminoethane)carbomyl] Cholesterol (DC-Chol), and 1,2-Dioleoyl-3-trimethylammonium Propane (DOTAP): Prolonged Liposome Retention Mediates Stronger Th1 ResponsesHenriksen-Lacey, Malou; Christensen, Dennis; Bramwell, Vincent W.; Lindenstrom, Thomas; Agger, Else Marie; Andersen, Peter; Perrie, YvonneMolecular Pharmaceutics (2011), 8 (1), 153-161CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The immunostimulatory capacities of cationic liposomes are well-documented and are attributed both to inherent immunogenicity of the cationic lipid and more phys. capacities such as the formation of antigen depots and antigen delivery. Very few studies have however been conducted comparing the immunostimulatory capacities of different cationic lipids. In the present study we therefore chose to investigate three of the most well-known cationic liposome-forming lipids as potential adjuvants for protein subunit vaccines. The ability of 3β-[N-(N',N'-dimethylaminoethane)carbomyl] cholesterol (DC-Chol), 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), and dimethyldioctadecylammonium (DDA) liposomes incorporating immunomodulating trehalose dibehenate (TDB) to form an antigen depot at the site of injection (SOI) and to induce immunol. recall responses against coadministered tuberculosis vaccine antigen Ag85B-ESAT-6 are reported. Furthermore, phys. characterization of the liposomes is presented. Our results suggest that liposome compn. plays an important role in vaccine retention at the SOI and the ability to enable the immune system to induce a vaccine specific recall response. While all three cationic liposomes facilitated increased antigen presentation by antigen presenting cells, the monocyte infiltration to the SOI and the prodn. of IFN-γ upon antigen recall was markedly higher for DDA and DC-Chol based liposomes which exhibited a longer retention profile at the SOI. A long-term retention and slow release of liposome and vaccine antigen from the injection site hence appears to favor a stronger Th1 immune response.
- 24De Serrano, L. O.; Burkhart, D. J. Liposomal Vaccine Formulations as Prophylactic Agents: Design Considerations for Modern Vaccines. J. Nanobiotechnol. 2017, 15, 83, DOI: 10.1186/s12951-017-0319-9Google Scholar24Liposomal vaccine formulations as prophylactic agents: design considerations for modern vaccinesDe Serrano, Luis O.; Burkhart, David J.Journal of Nanobiotechnology (2017), 15 (), 83/1-83/23CODEN: JNOAAO; ISSN:1477-3155. (BioMed Central Ltd.)A review. Vaccinol. is one of the most important cornerstones in modern medicine, providing better quality of life. The human immune system is composed of innate and adaptive immune processes that interplay when infection occurs. Innate immunity relies on pathogen-assocd. mol. patterns which are recognized by pathogen recognition receptors localized in antigen presenting cells. After antigen processing and presentation, CD4+ T cell polarization occurs, further leading to B cell and CD8+ activation and humoral and cell-mediated adaptive immune responses. Liposomes are being employed as vaccine technologies and their design is of importance to ensure proper immune responses. Physicochem. parameters like liposome size, charge, lamellarity and bilayer fluidity must be completely understood to ensure optimal vaccine stability and efficacy. Liposomal vaccines can be developed to target specific immune cell types for the induction of certain immune responses. In this review, we will present promising liposomal vaccine approaches for the treatment of important viral, bacterial, fungal and parasitic infections (including tuberculosis, TB). Cationic liposomes are the most studied liposome types due to their enhanced interaction with the neg. charged immune cells. Thus, a special section on the cationic lipid dimethyldioctadecylammonium and TB is also presented.
- 25Martino, S.; di Girolamo, I.; Tiribuzi, R.; D’Angelo, F.; Datti, A.; Orlacchio, A. Efficient siRNA Delivery by the Cationic Liposome DOTAP in Human Hematopoietic Stem Cells Differentiating into Dendritic Cells. J. Biomed. Biotechnol. 2009, 2009, 410260, DOI: 10.1155/2009/410260Google Scholar25Efficient siRNA delivery by the cationic liposome DOTAP in human hematopoietic stem cells differentiating into dendritic cellsMartino Sabata; di Girolamo Ilaria; Tiribuzi Roberto; D'Angelo Francesco; Datti Alessandro; Orlacchio AldoJournal of biomedicine & biotechnology (2009), 2009 (), 410260 ISSN:.RNA interference technology is an ideal strategy to elucidate the mechanisms associated with human CD34(+) hematopoietic stem cell differentiation into dendritic cells. Simple manipulations in vitro can unequivocally yield alloreactive or tolerogenic populations, suggesting key implications of biochemical players that might emerge as therapeutic targets for cancer or graft-versus-host disease. To knockdown proteins typically involved in the biology of dendritic cells, we employed an siRNA delivery system based on the cationic liposome DOTAP as the carrier. Freshly-isolated CD34(+) cells were transfected with siRNA for cathepsin S with negligible cytotoxicity and transfection rates (>60%) comparable to the efficiency shown by lentiviral vectors. Further, cathepsin S knockdown was performed during both cell commitment and through the entire 14-day differentiation process with repeated transfection rounds that had no effect per se on cell development. Tested in parallel, other commercially-available chemical reagents failed to meet acceptable standards. In addition to safe and practical handling, a direct advantage of DOTAP over viral-mediated techniques is that transient silencing effects can be dynamically appraised through the recovery of targeted proteins. Thus, our findings identify DOTAP as an excellent reagent for gene silencing in resting and differentiating CD34(+) cells, suggesting a potential for applications in related preclinical models.
- 26Kaneda, M. M.; Sasaki, Y.; Lanza, G. M.; Milbrandt, J.; Wickline, S. A. Mechanisms of Nucleotide Trafficking During siRNA Delivery to Endothelial Cells Using Perfluorocarbon Nanoemulsions. Biomaterials 2010, 31, 3079– 3086, DOI: 10.1016/j.biomaterials.2010.01.006Google Scholar26Mechanisms of nucleotide trafficking during siRNA delivery to endothelial cells using perfluorocarbon nanoemulsionsKaneda, Megan M.; Sasaki, Yo; Lanza, Gregory M.; Milbrandt, Jeffrey; Wickline, Samuel A.Biomaterials (2010), 31 (11), 3079-3086CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)RNA interference (RNAi) is a useful in vitro research tool, but its application as a safe and effective therapeutic agent may benefit from improved understanding of mechanisms of exogenous siRNA delivery, including cell trafficking and sorting patterns. We report the development of a transfection reagent for siRNA delivery which employs a distinctive non-digestive mode of particle-cell membrane interaction through the formation of a hemifusion complex resulting in lipid raft transport of cargo to the cytosol, bypassing the usual endosomal nanoparticle uptake pathway. We further demonstrate markedly enhanced efficacy over conventional transfection agents for suppressing endothelial cell expression of upregulated vascular adhesion mols.
- 27Zhang, X.; Li, B.; Luo, X.; Zhao, W.; Jiang, J.; Zhang, C.; Gao, M.; Chen, X.; Dong, Y. Biodegradable Amino-Ester Nanomaterials for Cas9 mRNA Delivery In Vitro and In Vivo. ACS Appl. Mater. Interfaces 2017, 9, 25481– 25487, DOI: 10.1021/acsami.7b08163Google Scholar27Biodegradable Amino-Ester Nanomaterials for Cas9 mRNA Delivery in Vitro and in VivoZhang, Xinfu; Li, Bin; Luo, Xiao; Zhao, Weiyu; Jiang, Justin; Zhang, Chengxiang; Gao, Min; Chen, Xiaofang; Dong, YizhouACS Applied Materials & Interfaces (2017), 9 (30), 25481-25487CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)Efficient and safe delivery of the CRISPR/Cas system is one of the key challenges for genome-editing applications in humans. Herein, we designed and synthesized a series of biodegradable lipidlike compds. contg. ester groups for the delivery of mRNA-encoding Cas9. Two lead materials, termed N-methyl-1,3-propanediamine (MPA)-A and MPA-Ab, showed a tunable rate of biodegrdn. MPA-A with linear ester chains was degraded dramatically faster than MPA-Ab with branched ester chains in the presence of esterase or in wild-type mice. Most importantly, MPA-A and MPA-Ab demonstrated efficient delivery of Cas9 mRNA both in vitro and in vivo. Consequently, these biodegradable lipidlike nanomaterials merit further development as genome-editing delivery tools for biol. and therapeutic applications.
- 28Kim, B.-K.; Hwang, G.-B.; Seu, Y.-B.; Choi, J.-S.; Jin, K. S.; Doh, K.-O. DOTAP/DOPE Ratio and Cell Type Determine Transfection Efficiency with DOTAP-Liposomes. Biochim. Biophys. Acta, Biomembr. 2015, 1848, 1996– 2001, DOI: 10.1016/j.bbamem.2015.06.020Google Scholar28DOTAP/DOPE ratio and cell type determine transfection efficiency with DOTAP-liposomesKim, Bieong-Kil; Hwang, Guen-Bae; Seu, Young-Bae; Choi, Jong-Soo; Jin, Kyeong Sik; Doh, Kyung-OhBiochimica et Biophysica Acta, Biomembranes (2015), 1848 (10_Part_A), 1996-2001CODEN: BBBMBS; ISSN:0005-2736. (Elsevier B.V.)The effects of lipid compns. on their physicochem. properties and transfection efficiencies were investigated. Four liposome formulations with different 1,2-dioleoyl-3-trimethylammoniumpropane (DOTAP) to dioleoylphosphatidylethanolamine (DOPE) wt. ratios were investigated, i.e., wt. ratios 1:0 (T1P0), 3:1 (T3P1), 1:1 (T1P1), and 1:3 (T1P3). Mean sizes of liposomes were influenced by their lipid compn. and the prepn. concn. at the time of sonication. Zeta potentials of liposomes were inversely correlated with their liposome sizes. However, neither liposome sizes nor zeta potentials were correlated with transfection efficiency. The optimum compn. of liposomes was cell-line dependent (T1P0 and T3P1 for Huh7 and AGS, T3P1 and T1P1 for COS7, and T1P1 and T1P3 for A549). The shape of lipoplexes was changed from lamellar to inverted hexagonal structure according to the increased ratio of DOPE, but there was no definite advantage of specific structure in transfection efficiency throughout all used cell lines. However, cellular internalization was consistently faster in T1P0, T3P1, T1P1 compared to T1P3 in all cell lines, suggesting the importance of endosomal escape. Our findings show that the transfection efficiency of DOTAP liposomes is mainly influenced by lipid compn. and cell type, and not by size or zeta potential.
- 29Mével, M.; Haudebourg, T.; Colombani, T.; Peuziat, P.; Dallet, L.; Chatin, B.; Lambert, O.; Berchel, M.; Montier, T.; Jaffrès, P.-A.; Lehn, P.; Pitard, B. Important Role of Phosphoramido Linkage in Imidazole-Based Dioleyl Helper Lipids for Liposome Stability and Primary Cell Transfection. J. Gene Med. 2016, 18, 3– 15, DOI: 10.1002/jgm.2869Google Scholar29Important role of phosphoramido linkage in imidazole-based dioleyl helper lipids for liposome stability and primary cell transfectionMevel, Mathieu; Haudebourg, Thomas; Colombani, Thibault; Peuziat, Pauline; Dallet, Laurence; Chatin, Benoit; Lambert, Olivier; Berchel, Mathieu; Montier, Tristan; Jaffres, Paul-Alain; Lehn, Pierre; Pitard, BrunoJournal of Gene Medicine (2016), 18 (1-3), 3-15CODEN: JGMEFG; ISSN:1521-2254. (Wiley-Blackwell)Background : To optimize synthetic gene delivery systems, there is a need to develop more efficient lipid formulations. Most cationic lipid formulations contain 'helper' neutral lipids because of their ability to increase DNA delivery, in particular by improving endosomal escape of DNA mols. via the pH-buffering effect of protonatable groups and/or fusion with the lipid bilayer of endosomes. Methods : We evaluated the influence of the linker structure between the two oleyl chains in the helper lipid on transfection efficiency in cell lines, as well as in primary cells (hepatocytes/cardiomyocytes). We reported the synthesis of two new pH-buffering imidazole helper lipids characterized by a polar headgroup contg. one (compd. 6) or two (compd. 5) imidazole groups and two oleyl chains linked by an amide group. We studied their assocn. with the aminoglycoside lipidic deriv. dioleylsuccinylparomomycin (DOSP), which contains two oleyl chains linked to the aminoglycoside polar headgroup via an amide function. We compared the morphol. and transfection properties of such binary liposomes of DOSP/5 and DOSP/6 with those of liposomes combining DOSP with another imidazole-based dioleyl helper lipid (MM27) in which a phosphoramido group acts as a linker between the two oleyl chains and imidazole function. Results : The phosphoramido linker in the helper lipid induces a major difference in terms of morphol. and resistance to decomplexation at phys. pH for DOSP/helper lipid complexes. Conclusions : This hybrid dioleyl linker compn. of DOSP/MM27 led to higher transfection efficiency in cell lines and in primary cells compared to complexes with homogeneous dioleyl linker. Copyright © 2015 John Wiley & Sons, Ltd.
- 30Cavalcanti, R. R. M.; Lira, R. B.; Riske, K. A. Study of the Fusion Mechanism of Fusogenic Cationic Liposomes with Anionic Model Membranes. Biophys. J. 2018, 114, 606a, DOI: 10.1016/j.bpj.2017.11.3314Google ScholarThere is no corresponding record for this reference.
- 31Démoulins, T.; Milona, P.; Englezou, P. C.; Ebensen, T.; Schulze, K.; Suter, R.; Pichon, C.; Midoux, P.; Guzmán, C. A.; Ruggli, N.; McCullough, K. C. Polyethylenimine-Based Polyplex Delivery of Self-Replicating RNA Vaccines. Nanomedicine (N. Y., NY, U. S.) 2016, 12, 711– 722, DOI: 10.1016/j.nano.2015.11.001Google Scholar31Polyethylenimine-based polyplex delivery of self-replicating RNA vaccinesDemoulins, Thomas; Milona, Panagiota; Englezou, Pavlos C.; Ebensen, Thomas; Schulze, Kai; Suter, Rolf; Pichon, Chantal; Midoux, Patrick; Guzman, Carlos A.; Ruggli, Nicolas; McCullough, Kenneth C.Nanomedicine (New York, NY, United States) (2016), 12 (3), 711-722CODEN: NANOBF; ISSN:1549-9634. (Elsevier)Self-amplifying replicon RNA (RepRNA) are large mols. (12-14 kb); their self-replication amplifies mRNA template nos., affording several rounds of antigen prodn., effectively increasing vaccine antigen payloads. Their sensitivity to RNase-sensitivity and inefficient uptake by dendritic cells (DCs) - abs. requirements for vaccine design - were tackled by condensing RepRNA into synthetic, nanoparticulate, polyethylenimine (PEI)-polyplex delivery vehicles. Polyplex-delivery formulations for small RNA mols. cannot be transferred to RepRNA due to its greater size and complexity; the N:P charge ratio and impact of RepRNA folding would influence polyplex condensation, post-delivery decompaction and the cytosolic release essential for RepRNA translation. Polyplex-formulations proved successful for delivery of RepRNA encoding influenza virus hemagglutinin and nucleocapsid to DCs. Cytosolic translocation was facilitated, leading to RepRNA translation. This efficacy was confirmed in vivo, inducing both humoral and cellular immune responses. Accordingly, this paper describes the first PEI-polyplexes providing efficient delivery of the complex and large, self-amplifying RepRNA vaccines.
- 32Pardi, N.; Parkhouse, K.; Kirkpatrick, E.; McMahon, M.; Zost, S. J.; Mui, B. L.; Tam, Y. K.; Karikó, K.; Barbosa, C. J.; Madden, T. D.; Hope, M. J.; Krammer, F.; Hensley, S. E.; Weissman, D. Nucleoside-Modified mRNA Immunization Elicits Influenza Virus Hemagglutinin Stalk-Specific Antibodies. Nat. Commun. 2018, 9, 3361, DOI: 10.1038/s41467-018-05482-0Google Scholar32Nucleoside-modified mRNA immunization elicits influenza virus hemagglutinin stalk-specific antibodiesPardi Norbert; Weissman Drew; Parkhouse Kaela; Zost Seth J; Hensley Scott E; Kirkpatrick Ericka; McMahon Meagan; Krammer Florian; Kirkpatrick Ericka; Mui Barbara L; Tam Ying K; Barbosa Christopher J; Madden Thomas D; Hope Michael J; Kariko KatalinNature communications (2018), 9 (1), 3361 ISSN:.Currently available influenza virus vaccines have inadequate effectiveness and are reformulated annually due to viral antigenic drift. Thus, development of a vaccine that confers long-term protective immunity against antigenically distant influenza virus strains is urgently needed. The highly conserved influenza virus hemagglutinin (HA) stalk represents one of the potential targets of broadly protective/universal influenza virus vaccines. Here, we evaluate a potent broadly protective influenza virus vaccine candidate that uses nucleoside-modified and purified mRNA encoding full-length influenza virus HA formulated in lipid nanoparticles (LNPs). We demonstrate that immunization with HA mRNA-LNPs induces antibody responses against the HA stalk domain of influenza virus in mice, rabbits, and ferrets. The HA stalk-specific antibody response is associated with protection from homologous, heterologous, and heterosubtypic influenza virus infection in mice.
- 33Reichmuth, A. M.; Oberli, M. A.; Jaklenec, A.; Langer, R.; Blankschtein, D. mRNA Vaccine Delivery Using Lipid Nanoparticles. Ther. Delivery 2016, 7, 319– 334, DOI: 10.4155/tde-2016-0006Google Scholar33mRNA vaccine delivery using lipid nanoparticlesReichmuth, Andreas M.; Oberli, Matthias A.; Jeklenec, Ana; Langer, Robert; Blankschtein, DanielTherapeutic Delivery (2016), 7 (5), 319-334CODEN: TDHEA7; ISSN:2041-5990. (Future Science Ltd.)MRNA vaccines elicit a potent immune response including antibodies and cytotoxic T cells. MRNA vaccines are currently evaluated in clin. 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 degrdn., facilitate endosomal escape, can be targeted to the desired cell type by surface decoration with ligands, and as needed, can be codelivered with adjuvants.
- 34Albanese, A.; Tang, P. S.; Chan, W. C. W. The Effect of Nanoparticle Size, Shape, and Surface Chemistry on Biological Systems. Annu. Rev. Biomed. Eng. 2012, 14, 1– 16, DOI: 10.1146/annurev-bioeng-071811-150124Google Scholar34The effect of nanoparticle size, shape, and surface chemistry on biological systemsAlbanese, Alexandre; Tang, Peter S.; Chan, Warren C. W.Annual Review of Biomedical Engineering (2012), 14 (), 1-16CODEN: ARBEF7; ISSN:1523-9829. (Annual Reviews Inc.)A review. An understanding of the interactions between nanoparticles and biol. systems is of significant interest. Studies aimed at correlating the properties of nanomaterials such as size, shape, chem. functionality, surface charge, and compn. with biomol. signaling, biol. kinetics, transportation, and toxicity in both cell culture and animal expts. are under way. These fundamental studies will provide a foundation for engineering the next generation of nanoscale devices. Here, the authors provide rationales for these studies, review the current progress in studies of the interactions of nanomaterials with biol. systems, and provide a perspective on the long-term implications of these findings.
- 35Golestani, R.; Pourfathollah, A. A.; Moazzeni, S. M. An Extreme Strategy for the Production of Hybridoma. Hybridoma 2009, 28, 139– 144, DOI: 10.1089/hyb.2008.0076Google Scholar35An Extreme Strategy for the Production of HybridomaGolestani, Reza; Pourfathollah, Ali Akbar; Moazzeni, Seyed MohammadHybridoma (2009), 28 (2), 139-144CODEN: HYBRAV; ISSN:1554-0014. (Mary Ann Liebert, Inc.)The ethical issues surrounding human immunization hamper the prodn. of human monoclonal antibody through scarcity of immunized B cells in peripheral blood. This defect can be compensated in part by improvement of hybridoma prodn. techniques. We have developed a new strategy to bypass the toxic effects of polyethylene glycol (PEG) as fusogenic reagent and hypoxanthine aminoptrin thymidine (HAT) as selective medium on newly fused cells. The Epstein-Barr virus (EBV) transformed peripheral blood mononuclear cells (PBMC) of accidentally Rh antigen sensitized persons were fused using cephalin as fugenic reagent, with emetine and actinomycin D pretreated heteromyeloma cells. Our results showed that 19-34% of EBV-transformed B cells were grown as hybridoma clones following selection. This extreme improvement in hybridoma prodn. rate may end the fusion efficiency problem and make hybridoma prodn. a plug-and-play technol.
- 36Phua, K. K. L.; Leong, K. W.; Nair, S. K. Transfection Efficiency and Transgene Expression Kinetics of mRNA Delivered in Naked and Nanoparticle Format. J. Controlled Release 2013, 166, 227– 233, DOI: 10.1016/j.jconrel.2012.12.029Google Scholar36Transfection efficiency and transgene expression kinetics of mRNA delivered in naked and nanoparticle formatPhua, Kyle K. L.; Leong, Kam W.; Nair, Smita K.Journal of Controlled Release (2013), 166 (3), 227-233CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Transfection efficiencies and transgene expression kinetics of mRNA, an emerging class of nucleic acid-based therapeutics, were poorly characterized. In this study, the authors evaluated transfection efficiencies of mRNA delivered in naked and nanoparticle format in vitro and in vivo using GFP and luciferase as reporters. While mRNA nanoparticles transfect primary human and mouse dendritic cells (DCs) efficiently in vitro, naked mRNA could not produce any detectable gene product. The protein expression of nanoparticle-mediated transfection in vitro peaks rapidly within 5-7 h and decays in a biphasic manner. In vivo, naked mRNA is more efficient than mRNA nanoparticles when administered s.c. In contrast, mRNA nanoparticle performs better when administered intranasally and i.v. Gene expression is most transient when delivered i.v. in nanoparticle format with an apparent half-life of 1.4 h and lasts less than 24 h, and most sustained when delivered in the naked format s.c. at the base of tail with an apparent half-life of 18 h and persists for at least 6 days. Notably, exponential decreases in protein expression are consistently obsd. post-delivery of mRNA in vivo regardless of the mode of delivery (naked or nanoparticle) or the site of administration. This study elucidates the performance of mRNA transfection and suggests a niche for mRNA therapeutics when predictable in vivo transgene expression kinetics is imperative.
- 37Liang, F.; Lindgren, G.; Lin, A.; Thompson, E. A.; Ols, S.; Röhss, J.; John, S.; Hassett, K.; Yuzhakov, O.; Bahl, K.; Brito, L. A.; Salter, H.; Ciaramella, G.; Loré, K. Efficient Targeting and Activation of Antigen-Presenting Cells In Vivo after Modified mRNA Vaccine Administration in Rhesus Macaques. Mol. Ther. 2017, 25, 2635– 2647, DOI: 10.1016/j.ymthe.2017.08.006Google Scholar37Efficient Targeting and Activation of Antigen-Presenting Cells In Vivo after Modified mRNA Vaccine Administration in Rhesus MacaquesLiang, Frank; Lindgren, Gustaf; Lin, Ang; Thompson, Elizabeth A.; Ols, Sebastian; Roehss, Josefine; John, Shinu; Hassett, Kimberly; Yuzhakov, Olga; Bahl, Kapil; Brito, Luis A.; Salter, Hugh; Ciaramella, Giuseppe; Lore, KarinMolecular Therapy (2017), 25 (12), 2635-2647CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)MRNA vaccines are rapidly emerging as a powerful platform for infectious diseases because they are well tolerated, immunogenic, and scalable and are built on precise but adaptable antigen design. We show that two immunizations of modified non-replicating mRNA encoding influenza H10 hemagglutinin (HA) and encapsulated in lipid nanoparticles (LNP) induce protective HA inhibition titers and H10-specific CD4+ T cell responses after i.m. or intradermal delivery in rhesus macaques. Administration of LNP/mRNA induced rapid and local infiltration of neutrophils, monocytes, and dendritic cells (DCs) to the site of administration and the draining lymph nodes (LNs). While these cells efficiently internalized LNP, mainly monocytes and DCs translated the mRNA and upregulated key co-stimulatory receptors (CD80 and CD86). This coincided with upregulation of type I IFN-inducible genes, including MX1 and CXCL10. The innate immune activation was transient and resulted in priming of H10-specific CD4+ T cells exclusively in the vaccine-draining LNs. Collectively, this demonstrates that mRNA-based vaccines induce type-I IFN-polarized innate immunity and, when combined with antigen prodn. by antigen-presenting cells, lead to generation of potent vaccine-specific responses.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 54 publications.
- Adrian P. E. Kromer, Felix Sieber-Schäfer, Johan Farfan Benito, Olivia M. Merkel. Design of Experiments Grants Mechanistic Insights into the Synthesis of Spermine-Containing PBAE Copolymers. ACS Applied Materials & Interfaces 2024, 16
(29)
, 37545-37554. https://doi.org/10.1021/acsami.4c06079
- Anne-Gaëlle Reinhart, Anja Osterwald, Philippe Ringler, Yael Leiser, Matthias E. Lauer, Rainer E. Martin, Christoph Ullmer, Felix Schumacher, Claudia Korn, Michael Keller. Investigations into mRNA Lipid Nanoparticles Shelf-Life Stability under Nonfrozen Conditions. Molecular Pharmaceutics 2023, 20
(12)
, 6492-6503. https://doi.org/10.1021/acs.molpharmaceut.3c00956
- Ramya Kumar. Materiomically Designed Polymeric Vehicles for Nucleic Acids: Quo Vadis?. ACS Applied Bio Materials 2022, 5
(6)
, 2507-2535. https://doi.org/10.1021/acsabm.2c00346
- Alexander B. Cook, Paolo Decuzzi. Harnessing Endogenous Stimuli for Responsive Materials in Theranostics. ACS Nano 2021, 15
(2)
, 2068-2098. https://doi.org/10.1021/acsnano.0c09115
- Pratik Gurnani, Anna K. Blakney, Roberto Terracciano, Joshua E. Petch, Andrew J. Blok, Clément R. Bouton, Paul F. McKay, Robin J. Shattock, Cameron Alexander. The In Vitro, Ex Vivo, and In Vivo Effect of Polymer Hydrophobicity on Charge-Reversible Vectors for Self-Amplifying RNA. Biomacromolecules 2020, 21
(8)
, 3242-3253. https://doi.org/10.1021/acs.biomac.0c00698
- Anna K. Blakney, Yamin Abdouni, Gokhan Yilmaz, Renjie Liu, Paul F. McKay, Clément R. Bouton, Robin J. Shattock, C. Remzi Becer. Mannosylated Poly(ethylene imine) Copolymers Enhance saRNA Uptake and Expression in Human Skin Explants. Biomacromolecules 2020, 21
(6)
, 2482-2492. https://doi.org/10.1021/acs.biomac.0c00445
- Anna K. Blakney, Yunqing Zhu, Paul F. McKay, Clément R. Bouton, Jonathan Yeow, Jiaqing Tang, Kai Hu, Karnyart Samnuan, Christopher L. Grigsby, Robin J. Shattock, Molly M. Stevens. Big Is Beautiful: Enhanced saRNA Delivery and Immunogenicity by a Higher Molecular Weight, Bioreducible, Cationic Polymer. ACS Nano 2020, 14
(5)
, 5711-5727. https://doi.org/10.1021/acsnano.0c00326
- Lei Wang, Yanhong Pan, Yunfei Liu, Zhaowei Sun, Yue Huang, Jinlong Li, Jie Yang, Yang Xiang, Genxi Li. Fabrication of an Aptamer-Coated Liposome Complex for the Detection and Profiling of Exosomes Based on Terminal Deoxynucleotidyl Transferase-Mediated Signal Amplification. ACS Applied Materials & Interfaces 2020, 12
(1)
, 322-329. https://doi.org/10.1021/acsami.9b18869
- Junli Li, Dong Liu, Xiaochi Li, Jiazheng Wei, Weixin Du, Aihua Zhao, Miao Xu. RNA vaccines: The dawn of a new age for tuberculosis?. Human Vaccines & Immunotherapeutics 2025, 21
(1)
https://doi.org/10.1080/21645515.2025.2469333
- Irafasha C. Casmil, Nuthan V. Bathula, Cynthia Huang, Christopher J. Wayne, Evan S. Cairns, Josh J. Friesen, Shekinah K. Soriano, Suiyang Liao, Chia H. Ho, Kristen Y.S. Kong, Anna K. Blakney. Alphaviral backbone of self-amplifying RNA enhances protein expression and immunogenicity against SARS-CoV-2 antigen. Molecular Therapy 2025, 33
(2)
, 514-528. https://doi.org/10.1016/j.ymthe.2024.12.055
- Ruby Sedgwick, John P. Goertz, Molly M. Stevens, Ruth Misener, Mark van der Wilk. Transfer learning Bayesian optimization for competitor DNA molecule design for use in diagnostic assays. Biotechnology and Bioengineering 2025, 122
(1)
, 189-210. https://doi.org/10.1002/bit.28854
- Yunhao Wu, Jin Shang, Xinyue Zhang, Nu Li. Advances in molecular imaging and targeted therapeutics for lymph node metastasis in cancer: a comprehensive review. Journal of Nanobiotechnology 2024, 22
(1)
https://doi.org/10.1186/s12951-024-02940-4
- Elahe Haghighi, Samira Sadat Abolmaali, Ali Dehshahri, Seyed Ali Mousavi Shaegh, Negar Azarpira, Ali Mohammad Tamaddon. Navigating the intricate in-vivo journey of lipid nanoparticles tailored for the targeted delivery of RNA therapeutics: a quality-by-design approach. Journal of Nanobiotechnology 2024, 22
(1)
https://doi.org/10.1186/s12951-024-02972-w
- Olga Vasileva, Olga Zaborova, Bogdan Shmykov, Roman Ivanov, Vasiliy Reshetnikov. Composition of lipid nanoparticles for targeted delivery: application to mRNA therapeutics. Frontiers in Pharmacology 2024, 15 https://doi.org/10.3389/fphar.2024.1466337
- Beatriz D. Barbieri, David J. Peeler, Karnyart Samnuan, Suzanne Day, Kai Hu, Hadijatou J. Sallah, John S. Tregoning, Paul F. McKay, Robin J. Shattock. The role of helper lipids in optimising nanoparticle formulations of self-amplifying RNA. Journal of Controlled Release 2024, 374 , 280-292. https://doi.org/10.1016/j.jconrel.2024.08.016
- Eliana B. Souto, Cristina Blanco-Llamero, Karolline Krambeck, Neelakanta Sarvashiva Kiran, Chandrashekar Yashaswini, Humzah Postwala, Patricia Severino, Ronny Priefer, Bhupendra Gopalbhai Prajapati, Rahul Maheshwari. Regulatory insights into nanomedicine and gene vaccine innovation: Safety assessment, challenges, and regulatory perspectives. Acta Biomaterialia 2024, 180 , 1-17. https://doi.org/10.1016/j.actbio.2024.04.010
- Ibrahim Shaw, George Frimpong Boafo, Yimer Seid Ali, Yang Liu, Ronald Mlambo, Songwen Tan, Chuanpin Chen. Advancements and prospects of lipid-based nanoparticles: dual frontiers in cancer treatment and vaccine development. Journal of Microencapsulation 2024, 41
(3)
, 226-254. https://doi.org/10.1080/02652048.2024.2326091
- Lei Huang, Shuyun Yang, Xiuqin Yu, Fumin Fang, Liping Zhu, Lu Wang, Xiaoping Zhang, Changzhi Yang, Qihong Qian, Tingting Zhu. Association of different cell types and inflammation in early acne vulgaris. Frontiers in Immunology 2024, 15 https://doi.org/10.3389/fimmu.2024.1275269
- Xiaonan Huang, Yishan Ma, Guanghui Ma, Yufei Xia. Unlocking the Therapeutic Applicability of LNP-mRNA: Chemistry, Formulation, and Clinical Strategies. Research 2024, 7 https://doi.org/10.34133/research.0370
- S. Demartis, G. Rassu, V. Mazzarello, E. Larrañeta, A. Hutton, R.F. Donnelly, A. Dalpiaz, M. Roldo, A.J. Guillot, A. Melero, P. Giunchedi, E. Gavini. Delivering hydrosoluble compounds through the skin: what are the chances?. International Journal of Pharmaceutics 2023, 646 , 123457. https://doi.org/10.1016/j.ijpharm.2023.123457
- Lidia Gurba-Bryśkiewicz, Wioleta Maruszak, Damian A. Smuga, Krzysztof Dubiel, Maciej Wieczorek. Quality by Design (QbD) and Design of Experiments (DOE) as a Strategy for Tuning Lipid Nanoparticle Formulations for RNA Delivery. Biomedicines 2023, 11
(10)
, 2752. https://doi.org/10.3390/biomedicines11102752
- Sarah J. Shepherd, Xuexiang Han, Alvin J. Mukalel, Rakan El-Mayta, Ajay S. Thatte, Jingyu Wu, Marshall S. Padilla, Mohamad-Gabriel Alameh, Neha Srikumar, Daeyeon Lee, Drew Weissman, David Issadore, Michael J. Mitchell. Throughput-scalable manufacturing of SARS-CoV-2 mRNA lipid nanoparticle vaccines. Proceedings of the National Academy of Sciences 2023, 120
(33)
https://doi.org/10.1073/pnas.2303567120
- Yasaman Esmaeili, Ashkan Farazin, Ilnaz Rahimmanesh, Ashkan Bigham. Gene Delivery. 2023, 170-192. https://doi.org/10.1039/9781837671540-00170
- Azadeh Rahimi, Yasaman Esmaeili, Nasim Dana, Arezou Dabiri, Ilnaz Rahimmanesh, Setareh Jandaghian, Golnaz Vaseghi, Laleh Shariati, Ali Zarrabi, Shaghayegh Haghjooy Javanmard, Marco Cordani. A comprehensive review on novel targeted therapy methods and nanotechnology-based gene delivery systems in melanoma. European Journal of Pharmaceutical Sciences 2023, 187 , 106476. https://doi.org/10.1016/j.ejps.2023.106476
- Konstantinos N. Kafetzis, Natalia Papalamprou, Elisha McNulty, Kai X. Thong, Yusuke Sato, Aleksandr Mironov, Atul Purohit, Philip J. Welsby, Hideyoshi Harashima, Cynthia Yu‐Wai‐Man, Aristides D. Tagalakis. The Effect of Cryoprotectants and Storage Conditions on the Transfection Efficiency, Stability, and Safety of Lipid‐Based Nanoparticles for mRNA and DNA Delivery. Advanced Healthcare Materials 2023, 12
(18)
https://doi.org/10.1002/adhm.202203022
- Riccardo Rampado, Dan Peer. Design of experiments in the optimization of nanoparticle-based drug delivery systems. Journal of Controlled Release 2023, 358 , 398-419. https://doi.org/10.1016/j.jconrel.2023.05.001
- Dylan Kairuz, Nazia Samudh, Abdullah Ely, Patrick Arbuthnot, Kristie Bloom. Production, Characterization, and Assessment of Permanently Cationic and Ionizable Lipid Nanoparticles for Use in the Delivery of Self-Amplifying RNA Vaccines. Pharmaceutics 2023, 15
(4)
, 1173. https://doi.org/10.3390/pharmaceutics15041173
- Tomokazu Amano, Hong Yu, Misa Amano, Erica Leyder, Maria Badiola, Priyanka Ray, Jiyoung Kim, Akihiro C. Ko, Achouak Achour, Nan-ping Weng, Efrat Kochba, Yotam Levin, Minoru S.H. Ko. Controllable self-replicating RNA vaccine delivered intradermally elicits predominantly cellular immunity. iScience 2023, 26
(4)
, 106335. https://doi.org/10.1016/j.isci.2023.106335
- Monique C.P. Mendonça, Ayse Kont, Piotr S. Kowalski, Caitriona M. O'Driscoll. Design of lipid-based nanoparticles for delivery of therapeutic nucleic acids. Drug Discovery Today 2023, 28
(3)
, 103505. https://doi.org/10.1016/j.drudis.2023.103505
- Artur Filipe Rodrigues, Catarina Rebelo, Susana Simões, Cristiana Paulo, Sónia Pinho, Vítor Francisco, Lino Ferreira. A Polymeric Nanoparticle Formulation for Targeted mRNA Delivery to Fibroblasts. Advanced Science 2023, 10
(5)
https://doi.org/10.1002/advs.202205475
- Patrizia Stoitzner, Nikolaus Romani, Christoph Rademacher, Hans Christian Probst, Karsten Mahnke. Antigen targeting to dendritic cells: Still a place in future immunotherapy?. European Journal of Immunology 2022, 52
(12)
, 1909-1924. https://doi.org/10.1002/eji.202149515
- Dimitri Papukashvili, Nino Rcheulishvili, Cong Liu, Yang Ji, Yunjiao He, Peng George Wang. Self-Amplifying RNA Approach for Protein Replacement Therapy. International Journal of Molecular Sciences 2022, 23
(21)
, 12884. https://doi.org/10.3390/ijms232112884
- Bryant C Nelson, Sven Even Borgos. High-Throughput Synthesis and Characterization of Next-Generation Lipid Nanoparticles for Enhanced
In Vivo
Performance. Nanomedicine 2022, 17
(9)
, 573-576. https://doi.org/10.2217/nnm-2022-0024
- Katrina M. Pollock, Hannah M. Cheeseman, Alexander J. Szubert, Vincenzo Libri, Marta Boffito, David Owen, Henry Bern, Leon R. McFarlane, Jessica O'Hara, Nana-Marie Lemm, Paul McKay, Tommy Rampling, Yee Ting N. Yim, Ana Milinkovic, Cherry Kingsley, Tom Cole, Susanne Fagerbrink, Marites Aban, Maniola Tanaka, Savviz Mehdipour, Alexander Robbins, William Budd, Saul Faust, Hana Hassanin, Catherine A. Cosgrove, Alan Winston, Sarah Fidler, David Dunn, Sheena McCormack, Robin J. Shattock, Kirsty Adams, Fahimah Amini, Nafisah B Atako, Amalina Bakri, Wendy Barclay, Elizabeth Brodnicki, Jonathan C Brown, Ruth Byrne, Rowena Chilvers, Sofia Coelho, Suzanne Day, Monica Desai, Eleanor Dorman, Tamara Elliott, Katie E Flight, James Fletcher, John Galang, Jagruti Gohil, Aneta Gupta, Chris Harlow, Kai Hu, Mohini Kalyan, Dominic Lagrue, Ely Liscano, Cecilia Njenga, Krunal Polra, Derecia A Powlette, Paul Randell, Mary Rauchenberger, Ianto Redknap, Maravic Ricamara, Paul Rogers, Hadijatou Sallah, Karnyart Samnuan, Michael Schumacher, Zareena Shah, Rachel Shaw, Thomas Shaw, Stefan Sivapatham, Susie Slater, Kim Sorley, Regina Storch, Elizabeth Tan, Tricia Tan, Lieze Thielemans, Sarah Whitely, Charlotte Valentine, Jeeva Varghese, Asha Vikraman, Martin Wilkins. Safety and immunogenicity of a self-amplifying RNA vaccine against COVID-19: COVAC1, a phase I, dose-ranging trial. eClinicalMedicine 2022, 44 , 101262. https://doi.org/10.1016/j.eclinm.2021.101262
- Neha Tiwari, Ernesto Rafael Osorio‐Blanco, Ana Sonzogni, David Esporrín‐Ubieto, Huiyi Wang, Marcelo Calderón. Nanocarriers for Skin Applications: Where Do We Stand?. Angewandte Chemie 2022, 134
(3)
https://doi.org/10.1002/ange.202107960
- Neha Tiwari, Ernesto Rafael Osorio‐Blanco, Ana Sonzogni, David Esporrín‐Ubieto, Huiyi Wang, Marcelo Calderón. Nanocarriers for Skin Applications: Where Do We Stand?. Angewandte Chemie International Edition 2022, 61
(3)
https://doi.org/10.1002/anie.202107960
- Nuthan Vikas Bathula, Petya Popova, Anna Blakney. Delivery Vehicles for Self-amplifying RNA. 2022, 355-370. https://doi.org/10.1007/978-3-031-08415-7_16
- Heidrun Steinle, Josefin Weber, Sandra Stoppelkamp, Katharina Große-Berkenbusch, Sonia Golombek, Marbod Weber, Tuba Canak-Ipek, Sarah-Maria Trenz, Christian Schlensak, Meltem Avci-Adali. Delivery of synthetic mRNAs for tissue regeneration. Advanced Drug Delivery Reviews 2021, 179 , 114007. https://doi.org/10.1016/j.addr.2021.114007
- Xucheng Hou, Tal Zaks, Robert Langer, Yizhou Dong. Lipid nanoparticles for mRNA delivery. Nature Reviews Materials 2021, 6
(12)
, 1078-1094. https://doi.org/10.1038/s41578-021-00358-0
- Anna K. Blakney, Paul F. McKay, Kai Hu, Karnyart Samnuan, Nikita Jain, Andrew Brown, Anitha Thomas, Paul Rogers, Krunal Polra, Hadijatou Sallah, Jonathan Yeow, Yunqing Zhu, Molly M. Stevens, Andrew Geall, Robin J. Shattock. Polymeric and lipid nanoparticles for delivery of self-amplifying RNA vaccines. Journal of Controlled Release 2021, 338 , 201-210. https://doi.org/10.1016/j.jconrel.2021.08.029
- Marwa A. Sallam, Supriya Prakash, Ninad Kumbhojkar, Charles Wyatt Shields, Samir Mitragotri. Formulation‐based approaches for dermal delivery of vaccines and therapeutic nucleic acids: Recent advances and future perspectives. Bioengineering & Translational Medicine 2021, 6
(3)
https://doi.org/10.1002/btm2.10215
- David J. Peeler, Albert Yen, Nicholas Luera, Patrick S. Stayton, Suzie H. Pun. Lytic Polyplex Vaccines Enhance Antigen‐Specific Cytotoxic T Cell Response through Induction of Local Cell Death. Advanced Therapeutics 2021, 4
(8)
https://doi.org/10.1002/adtp.202100005
- Kiara Fairman, Miao Li, Baitang Ning, Annie Lumen. Physiologically based pharmacokinetic (PBPK) modeling of RNAi therapeutics: Opportunities and challenges. Biochemical Pharmacology 2021, 189 , 114468. https://doi.org/10.1016/j.bcp.2021.114468
- Qurrat Ul Ain, Estefania V.R. Campos, Ariel Huynh, Dominik Witzigmann, Sarah Hedtrich. Gene Delivery to the Skin – How Far Have We Come?. Trends in Biotechnology 2021, 39
(5)
, 474-487. https://doi.org/10.1016/j.tibtech.2020.07.012
- Lu Yang, Yan-Hong Shou, Yong-Sheng Yang, Jin-Hua Xu, . Elucidating the immune infiltration in acne and its comparison with rosacea by integrated bioinformatics analysis. PLOS ONE 2021, 16
(3)
, e0248650. https://doi.org/10.1371/journal.pone.0248650
- Anna K. Blakney, Paul F. McKay, Clément R. Bouton, Kai Hu, Karnyart Samnuan, Robin J. Shattock. Innate Inhibiting Proteins Enhance Expression and Immunogenicity of Self-Amplifying RNA. Molecular Therapy 2021, 29
(3)
, 1174-1185. https://doi.org/10.1016/j.ymthe.2020.11.011
- Anna K. Blakney, Polina Deletic, Paul F. McKay, Clément R. Bouton, Marianne Ashford, Robin J. Shattock, Alan Sabirsh. Effect of complexing lipids on cellular uptake and expression of messenger RNA in human skin explants. Journal of Controlled Release 2021, 330 , 1250-1261. https://doi.org/10.1016/j.jconrel.2020.11.033
- Giulietta Maruggi, Jeffrey B. Ulmer, Rino Rappuoli, Dong Yu. Self-amplifying mRNA-Based Vaccine Technology and Its Mode of Action. 2021, 31-70. https://doi.org/10.1007/82_2021_233
- Katrina M. Pollock, Hannah M. Cheeseman, Alexander J. Szubert, Vincenzo Libri, Marta Boffito, David Owen, Henry Bern, Jessica O’Hara, Leon R. McFarlane, Nana-Marie Lemm, Paul F. McKay, Tommy Rampling, Yee Ting N. Yim, Ana Milinkovic, Cherry Kingsley, Tom Cole, Susanne Fagerbrink, Marites Aban, Maniola Tanaka, Savviz Mehdipour, Alexander Robbins, William Budd, Saul N. Faust, Hana Hassanin, Catherine A. Cosgrove, Alan Winston, Sarah Fidler, David T. Dunn, Sheena McCormack, Robin John Shattock. Safety and Immunogenicity of a Self-Amplifying RNA Vaccine Against COVID-19: COVAC1, a Phase I, Dose-Ranging Trial. SSRN Electronic Journal 2021, 397 https://doi.org/10.2139/ssrn.3859294
- Tingting Ye, Zifu Zhong, Adolfo García‐Sastre, Michael Schotsaert, Bruno G. De Geest. Current Status of COVID‐19 (Pre)Clinical Vaccine Development. Angewandte Chemie 2020, 132
(43)
, 19045-19057. https://doi.org/10.1002/ange.202008319
- Tingting Ye, Zifu Zhong, Adolfo García‐Sastre, Michael Schotsaert, Bruno G. De Geest. Current Status of COVID‐19 (Pre)Clinical Vaccine Development. Angewandte Chemie International Edition 2020, 59
(43)
, 18885-18897. https://doi.org/10.1002/anie.202008319
- María Cristina Ballesteros-Briones, Noelia Silva-Pilipich, Guillermo Herrador-Cañete, Lucia Vanrell, Cristian Smerdou. A new generation of vaccines based on alphavirus self-amplifying RNA. Current Opinion in Virology 2020, 44 , 145-153. https://doi.org/10.1016/j.coviro.2020.08.003
- Gustavo Lou, Giulia Anderluzzi, Signe Tandrup Schmidt, Stuart Woods, Simona Gallorini, Michela Brazzoli, Fabiola Giusti, Ilaria Ferlenghi, Russell N. Johnson, Craig W. Roberts, Derek T. O'Hagan, Barbara C. Baudner, Yvonne Perrie. Delivery of self-amplifying mRNA vaccines by cationic lipid nanoparticles: The impact of cationic lipid selection. Journal of Controlled Release 2020, 325 , 370-379. https://doi.org/10.1016/j.jconrel.2020.06.027
- Francesca Saviano, Tatiana Lovato, Annapina Russo, Giulia Russo, Clément R. Bouton, Robin J Shattock, Cameron Alexander, Fabiana Quaglia, Anna K. Blakney, Pratik Gurnani, Claudia Conte. Ornithine-derived oligomers and dendrimers for
in vitro
delivery of DNA and
ex vivo
transfection of skin cells
via
saRNA. Journal of Materials Chemistry B 2020, 8
(22)
, 4940-4949. https://doi.org/10.1039/D0TB00942C
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Scheme 1
Scheme 1. Schematic of Lipid Nanoparticle (LNP) Formulations Used for DoE Analysis: (a) Lipid Nanoparticles Containing a Complexing Lipid, DOPE, and Cholesterol; and (b) Complexing Lipids Used in the DoE LibraryFigure 1
Figure 1. Firefly luciferase expression in human skin explants over the course of 21 days after ID injection of three separate, simultaneous injections of 10 μg of saRNA with a mass ratio of lipid to RNA of 4:1 (w/w): (a) time course of ex vivo imaging of luciferase expressed by replicon RNA delivered with the initial formulation of DOTAP LNPs; and (b) quantification of luciferase expression, expressed as the mean total flux (p/s) ± standard deviation for n = 3.
Figure 2
Figure 2. Firefly luciferase expression in human skin explants injected intradermally with LNP formulations with varying lipid identity and LNP dose containing 2 μg of saRNA with medium particle concentration (108 particles/mL) at varying ratios of lipid to RNA (w/w): (a) ex vivo imaging of explants after 11 days and (b) quantification of luciferase imagine expressed as the mean total flux (p/s) ± standard deviation for n = 5.
Figure 3
Figure 3. Firefly luciferase expression in human skin explants injected intradermally with LNP formulations with varying lipid and particle concentrations containing 2 μg of saRNA with a ratio of total lipid to RNA of 90:1 (w/w): (a) ex vivo imaging of explants after 11 days and (b) quantification of luciferase imagine expressed as the mean total flux (p/s) ± standard deviation for n = 5. High and low particle concentrations are defined as 109 and 107 particles/mL, respectively, while high lipid concentration is defined as 7.5 mg/mL. H[L]/H[P] denotes high lipid concentration and high particle concentration; H[L]/L[P] denotes high lipid concentration and low particle concentration.
Figure 4
Figure 4. Firefly luciferase expression in human skin explants injected intradermally with LNP formulations with varying ratios of cationic and zwitterionic lipids containing 2 μg of saRNA with a total lipid to RNA ratio of 18:1 (w/w) and medium particle concentration (108 particles/mL): (a) ex vivo imaging of explants after 11 days and (b) quantification of luciferase imagine expressed as mean total flux (p/s) ± standard deviation for n = 5.
Figure 5
Figure 5. Standard least-squares effect screening DoE analysis of lipid nanoparticle formulation in human skin explants.
Figure 6
Figure 6. Comparison of fold change luciferase expression of tested LNP formulations normalized to original DOTAP formulation (blue bar). Values are expressed fold change total flux (p/s) ± standard deviation.
Figure 7
Figure 7. GFP expression in human skin cells after intradermal injection with LNP formulations, as determined using flow cytometry: (a) histogram of number of cells expressing GFP for each formulation, and (b) percentage of GFP-positive cells of total live cells for each sample. Bar represents the average ± standard deviation, with a significance of α = 0.05 indicated by an asterisk (*).
Figure 8
Figure 8. Identity of cells present in human skin explants and GFP+ cells after ID injection of LNP formulations, as determined by flow cytometry: (a) identity of cells in the population of cells extracted from human skin explants, and (b) identity of GFP-expressing skin cells from explants treated with LNP-formulated RNA. The blue sections of each of the small pie charts indicate the total percentage of immune cells in the GFP+ cell population.
References
This article references 37 other publications.
- 1Guan, S.; Rosenecker, J. Nanotechnologies in Delivery of mRNA Therapeutics Using Nonviral Vector-Based Delivery Systems. Gene Ther. 2017, 24, 133, DOI: 10.1038/gt.2017.51Nanotechnologies in delivery of mRNA therapeutics using nonviral vector-based delivery systemsGuan, S.; Rosenecker, J.Gene Therapy (2017), 24 (3), 133-143CODEN: GETHEC; ISSN:0969-7128. (Nature Publishing Group)A review. Because of its safe and effective protein expression profile, in vitro transcribed mRNA (IVT-mRNA) represents a promising candidate in the development of novel therapeutics for genetic diseases, vaccines or gene editing strategies, esp. when its inherent shortcomings (for example, instability and immunogenicity) have been partially addressed via structural modifications. However, numerous unsolved tech. difficulties in successful in vivo delivery of IVT-mRNA have greatly hindered the applications of IVT-mRNA in clin. development. Recent advances in nanotechnol. and material science have yielded many promising nonviral delivery systems, some of which were able to efficiently facilitate targeted in vivo delivery of IVT-mRNA in safe and noninvasive manners. The diversity and flexibility of these delivery systems highlight the recent progress of IVT-mRNA-based therapy using nonviral vectors. In this review, we summarize recent advances of existing and emerging nonviral vector-based nanotechnologies for IVT-mRNA delivery and briefly summarize the interesting but rarely discussed applications on simultaneous delivery of IVT-mRNA with DNA.
- 2Karikó, K.; Muramatsu, H.; Welsh, F. A.; Ludwig, J.; Kato, H.; Akira, S.; Weissman, D. Incorporation of Pseudouridine into mRNA Yields Superior Nonimmunogenic Vector with Increased Translational Capacity and Biological Stability. Mol. Ther. 2008, 16, 1833– 1840, DOI: 10.1038/mt.2008.2002Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stabilityKariko, Katalin; Muramatsu, Hiromi; Welsh, Frank A.; Ludwig, Janos; Kato, Hiroki; Akira, Shizuo; Weissman, DrewMolecular Therapy (2008), 16 (11), 1833-1840CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)In vitro-transcribed mRNAs encoding physiol. important proteins have considerable potential for therapeutic applications. However, in its present form, mRNA is unfeasible for clin. use because of its labile and immunogenic nature. Here, we investigated whether incorporation of naturally modified nucleotides into transcripts would confer enhanced biol. properties to mRNA. We found that mRNAs contg. pseudouridines have a higher translational capacity than unmodified mRNAs when tested in mammalian cells and lysates or administered i.v. into mice at 0.015-0.15 mg/kg doses. The delivered mRNA and the encoded protein could be detected in the spleen at 1, 4, and 24 h after the injection, where both products were at significantly higher levels when pseudouridine-contg. mRNA was administered. Even at higher doses, only the unmodified mRNA was immunogenic, inducing high serum levels of interferon-α (IFN-α). These findings indicate that nucleoside modification is an effective approach to enhance stability and translational capacity of mRNA while diminishing its immunogenicity in vivo. Improved properties conferred by pseudouridine make such mRNA a promising tool for both gene replacement and vaccination.
- 3Thess, A.; Grund, S.; Mui, B. L.; Hope, M. J.; Baumhof, P.; Fotin-Mleczek, M.; Schlake, T. Sequence-Engineered mRNA without Chemical Nucleoside Modifications Enables an Effective Protein Therapy in Large Animals. Mol. Ther. 2015, 23, 1456– 1464, DOI: 10.1038/mt.2015.1033Sequence-engineered mRNA Without Chemical Nucleoside Modifications Enables an Effective Protein Therapy in Large AnimalsThess, Andreas; Grund, Stefanie; Mui, Barbara L.; Hope, Michael J.; Baumhof, Patrick; Fotin-Mleczek, Mariola; Schlake, ThomasMolecular Therapy (2015), 23 (9), 1456-1464CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)Being a transient carrier of genetic information, mRNA could be a versatile, flexible, and safe means for protein therapies. While recent findings highlight the enormous therapeutic potential of mRNA, evidence that mRNA-based protein therapies are feasible beyond small animals such as mice is still lacking. Previous studies imply that mRNA therapeutics require chem. nucleoside modifications to obtain sufficient protein expression and avoid activation of the innate immune system. Here we show that chem. unmodified mRNA can achieve those goals as well by applying sequence-engineered mols. Using erythropoietin (EPO) driven prodn. of red blood cells as the biol. model, engineered Epo mRNA elicited meaningful physiol. responses from mice to nonhuman primates. Even in pigs of about 20 kg in wt., a single adequate dose of engineered mRNA encapsulated in lipid nanoparticles (LNPs) induced high systemic Epo levels and strong physiol. effects. Our results demonstrate that sequence-engineered mRNA has the potential to revolutionize human protein therapies.
- 4Kauffman, K. J.; Webber, M. J.; Anderson, D. G. Materials for Non-Viral Intracellular Delivery of Messenger RNA Therapeutics. J. Controlled Release 2016, 240, 227– 234, DOI: 10.1016/j.jconrel.2015.12.0324Materials for non-viral intracellular delivery of messenger RNA therapeuticsKauffman, Kevin J.; Webber, Matthew J.; Anderson, Daniel G.Journal of Controlled Release (2016), 240 (), 227-234CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Though therapeutics based on mRNA (mRNA) have broad potential in applications such as protein replacement therapy, cancer immunotherapy, and genomic engineering, their effective intracellular delivery remains a challenge. A chem. diverse suite of delivery materials with origins as materials for cellular transfection of DNA and small interfering RNAs (siRNAs) has recently been reported to have promise as non-viral delivery agents for mRNA. These materials include covalent conjugates, protamine complexes, nanoparticles based on lipids or polymers, and hybrid formulations. This review will highlight the use of delivery materials for mRNA, with a specific focus on their mechanisms of action, routes of administration, and dosages. Addnl., strategies in which these materials can be adapted and optimized to address challenges specific to mRNA delivery are also discussed. The technologies included have shown varying promise for therapeutic use, specifically having been used to deliver mRNA in vivo or exhibiting characteristics that could make in vivo use a possibility. In so doing, it is the intention of this review to provide a comprehensive look at the progress and possibilities in applying nucleic acid delivery technol. specifically toward the emerging area of mRNA therapeutics.
- 5Jensen, S.; Thomsen, A. R. Sensing of RNA Viruses: A Review of Innate Immune Receptors Involved in Recognizing RNA Virus Invasion. J. Virol. 2012, 86, 2900, DOI: 10.1128/JVI.05738-115Sensing of RNA viruses: a review of innate immune receptors involved in recognizing RNA virus invasionJensen, Soeren; Thomsen, Allan RandrupJournal of Virology (2012), 86 (6), 2900-2910CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)A review. Our knowledge regarding the contribution of the innate immune system in recognizing and subsequently initiating a host response to an invasion of RNA virus has been rapidly growing over the last decade. Descriptions of the receptors involved and the mol. mechanisms they employ to sense viral pathogen-assocd. mol. patterns have emerged in great detail. This review presents an overview of our current knowledge regarding the receptors used to detect RNA virus invasion, the mol. structures these receptors sense, and the involved downstream signaling pathways.
- 6Kawai, T.; Akira, S. The Role of Pattern-Recognition Receptors in Innate Immunity: Update on Toll-Like Receptors. Nat. Immunol. 2010, 11, 373, DOI: 10.1038/ni.18636The role of pattern-recognition receptors in innate immunity: update on Toll-like receptorsKawai, Taro; Akira, ShizuoNature Immunology (2010), 11 (5), 373-384CODEN: NIAMCZ; ISSN:1529-2908. (Nature Publishing Group)A review. The discovery of Toll-like receptors (TLRs) as components that recognize conserved structures in pathogens has greatly advanced understanding of how the body senses pathogen invasion, triggers innate immune responses and primes antigen-specific adaptive immunity. Although TLRs are crit. for host defense, it has become apparent that loss of neg. regulation of TLR signaling, as well as recognition of self mols. by TLRs, are strongly assocd. with the pathogenesis of inflammatory and autoimmune diseases. Furthermore, it is now clear that the interaction between TLRs and recently identified cytosolic innate immune sensors is crucial for mounting effective immune responses. Here we describe the recent advances that have been made by research into the role of TLR biol. in host defense and disease.
- 7Perri, S.; Greer, C. E.; Thudium, K.; Doe, B.; Legg, H.; Liu, H.; Romero, R. E.; Tang, Z.; Bin, Q.; Dubensky, T. W.; Vajdy, M.; Otten, G. R.; Polo, J. M. An Alphavirus Replicon Particle Chimera Derived from Venezuelan Equine Encephalitis and Sindbis Viruses Is a Potent Gene-Based Vaccine Delivery Vector. J. Virol. 2003, 77, 10394– 10403, DOI: 10.1128/JVI.77.19.10394-10403.20037An alphavirus replicon particle chimera derived from Venezuelan equine encephalitis and Sindbis viruses is a potent gene-based vaccine delivery vectorPerri, Silvia; Greer, Catherine E.; Thudium, Kent; Doe, Barbara; Legg, Harold; Liu, Hong; Romero, Raul E.; Tang, Zequn; Bin, Qian; Dubensky, Thomas W., Jr.; Vajdy, Michael; Otten, Gillis R.; Polo, John M.Journal of Virology (2003), 77 (19), 10394-10403CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Alphavirus replicon particle-based vaccine vectors derived from Sindbis virus (SIN), Semliki Forest virus, and Venezuelan equine encephalitis virus (VEE) have been shown to induce robust antigen-specific cellular, humoral, and mucosal immune responses in many animal models of infectious disease and cancer. However, since little is known about the relative potencies among these different vectors, we compared the immunogenicity of replicon particle vectors derived from two very different parental alphaviruses, VEE and SIN, expressing a human immunodeficiency virus type 1 p55Gag antigen. Moreover, to explore the potential benefits of combining elements from different alphaviruses, we generated replicon particle chimeras of SIN and VEE. Two distinct strategies were used to produce particles with VEE-p55gag replicon RNA packaged within SIN envelope glycoproteins and SIN-p55gag replicon RNA within VEE envelope glycoproteins. Each replicon particle configuration induced Gag-specific CD8+ T-cell responses in murine models when administered alone or after priming with DNA. However, Gag-specific responses varied dramatically, with the strongest responses to this particular antigen correlating with the VEE replicon RNA, irresp. of the source of envelope glycoproteins. Comparing the replicons with respect to heterologous gene expression levels and sensitivity to alpha/beta interferon in cultured cells indicated that each might contribute to potency differences. This work shows that combining desirable elements from VEE and SIN into a replicon particle chimera may be a valuable approach toward the goal of developing vaccine vectors with optimal in vivo potency, ease of prodn., and safety.
- 8Chahal, J. S.; Khan, O. F.; Cooper, C. L.; McPartlan, J. S.; Tsosie, J. K.; Tilley, L. D.; Sidik, S. M.; Lourido, S.; Langer, R.; Bavari, S.; Ploegh, H. L.; Anderson, D. G. Dendrimer-RNA Nanoparticles Generate Protective Immunity against Lethal Ebola, H1N1 Influenza, and Toxoplasma Gondii Challenges with a Single Dose. Proc. Natl. Acad. Sci. U. S. A. 2016, 113, E4133– E4142, DOI: 10.1073/pnas.16002991138Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and Toxoplasma gondii challenges with a single doseChahal, Jasdave S.; Khan, Omar F.; Cooper, Christopher L.; McPartlan, Justine S.; Tsosie, Jonathan K.; Tilley, Lucas D.; Sidik, Saima M.; Lourido, Sebastian; Langer, Robert; Bavari, Sina; Ploegh, Hidde L.; Anderson, Daniel G.Proceedings of the National Academy of Sciences of the United States of America (2016), 113 (29), E4133-E4142CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Vaccines have had broad medical impact, but existing vaccine technologies and prodn. methods are limited in their ability to respond rapidly to evolving and emerging pathogens, or sudden outbreaks. Here, the authors develop a rapid-response, fully synthetic, single-dose, adjuvant-free dendrimer nanoparticle vaccine platform wherein antigens are encoded by encapsulated mRNA replicons. To the authors' knowledge, this system is the first capable of generating protective immunity against a broad spectrum of lethal pathogen challenges, including H1N1 influenza, Toxoplasma gondii, and Ebola virus. The vaccine can be formed with multiple antigen-expressing replicons, and is capable of eliciting both CD8+ T-cell and antibody responses. The ability to generate viable, contaminant-free vaccines within days, to single or multiple antigens, may have broad utility for a range of diseases.
- 9Geall, A. J.; Verma, A.; Otten, G. R.; Shaw, C. A.; Hekele, A.; Banerjee, K.; Cu, Y.; Beard, C. W.; Brito, L. A.; Krucker, T.; O’Hagan, D. T.; Singh, M.; Mason, P. W.; Valiante, N. M.; Dormitzer, P. R.; Barnett, S. W.; Rappuoli, R.; Ulmer, J. B.; Mandl, C. W. Nonviral Delivery of Self-Amplifying RNA Vaccines. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 14604, DOI: 10.1073/pnas.12093671099Nonviral delivery of self-amplifying RNA vaccinesGeall, Andrew J.; Verma, Ayush; Otten, Gillis R.; Shaw, Christine A.; Hekele, Armin; Banerjee, Kaustuv; Cu, Yen; Beard, Clayton W.; Brito, Luis A.; Krucker, Thomas; O'Hagan, Derek T.; Singh, Manmohan; Mason, Peter W.; Valiante, Nicholas M.; Dormitzer, Philip R.; Barnett, Susan W.; Rappuoli, Rino; Ulmer, Jeffrey B.; Mandl, Christian W.Proceedings of the National Academy of Sciences of the United States of America (2012), 109 (36), 14604-14609, S14604/1-S14604/7CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Despite more than two decades of research and development on nucleic acid vaccines, there is still no com. product for human use. Taking advantage of the recent innovations in systemic delivery of short interfering RNA (siRNA) using lipid nanoparticles (LNPs), we developed a self-amplifying RNA vaccine. Here we show that nonviral delivery of a 9-kb self-amplifying RNA encapsulated within an LNP substantially increased immunogenicity compared with delivery of unformulated RNA. This unique vaccine technol. was found to elicit broad, potent, and protective immune responses, that were comparable to a viral delivery technol., but without the inherent limitations of viral vectors. Given the many pos. attributes of nucleic acid vaccines, our results suggest that a comprehensive evaluation of nonviral technologies to deliver self-amplifying RNA vaccines is warranted.
- 10Bogers, W. M.; Oostermeijer, H.; Mooij, P.; Koopman, G.; Verschoor, E. J.; Davis, D.; Ulmer, J. B.; Brito, L. A.; Cu, Y.; Banerjee, K.; Otten, G. R.; Burke, B.; Dey, A.; Heeney, J. L.; Shen, X.; Tomaras, G. D.; Labranche, C.; Montefiori, D. C.; Liao, H.-X.; Haynes, B.; Geall, A. J.; Barnett, S. W. Potent Immune Responses in Rhesus Macaques Induced by Nonviral Delivery of a Self-Amplifying RNA Vaccine Expressing HIV Type 1 Envelope with a Cationic Nanoemulsion. J. Infect. Dis. 2015, 211, 947– 955, DOI: 10.1093/infdis/jiu52210Potent immune responses in rhesus macaques induced by nonviral delivery of a selfamplifying RNA vaccine expressing HIV type 1 envelope with a cationic nanoemulsionBogers, Willy M.; Oostermeijer, Herman; Mooij, Petra; Koopman, Gerrit; Verschoor, Ernst J.; Davis, David; Ulmer, Jeffrey B.; Brito, Luis A.; Cu, Yen; Banerjee, Kaustuv; Otten, Gillis R.; Burke, Brian; Dey, Antu; Heeney, Jonathan L.; Shen, Xiaoying; Tomaras, Georgia D.; Labranche, Celia; Montefiori, David C.; Liao, Hua-Xin; Haynes, Barton; Geall, Andrew J.; Barnett, Susan W.Journal of Infectious Diseases (2015), 211 (6), 947-955CODEN: JIDIAQ; ISSN:0022-1899. (Oxford University Press)Self-amplifying mRNA (mRNA) of pos.-strand RNA viruses are effective vectors for in situ expression of vaccine antigens and have potential as a new vaccine technol. platform well suited for global health applications. The SAM vaccine platform is based on a synthetic, self-amplifying mRNA delivered by a nonviral delivery system. The safety and immunogenicity of an HIV SAM vaccine encoding a clade C envelope glycoprotein formulated with a cationic nanoemulsion (CNE) delivery system was evaluated in rhesus macaques. The HIV SAM vaccine induced potent cellular immune responses that were greater in magnitude than those induced by self-amplifying mRNA packaged in a viral replicon particle (VRP) or by a recombinant HIV envelope protein formulated with MF59 adjuvant, anti-envelope binding (including anti-V1V2), and neutralizing antibody responses that exceeded those induced by the VRP vaccine. These studies provide the first evidence in nonhuman primates that HIV vaccination with a relatively low dose (50 μg) of formulated self-amplifying mRNA is safe and immunogenic.
- 11Perche, F.; Benvegnu, T.; Berchel, M.; Lebegue, L.; Pichon, C.; Jaffrès, P.-A.; 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 (N. Y., NY, U. S.) 2011, 7, 445– 453, DOI: 10.1016/j.nano.2010.12.010There is no corresponding record for this reference.
- 12Uchida, S.; Kinoh, H.; Ishii, T.; Matsui, A.; Tockary, T. A.; Takeda, K. M.; Uchida, H.; Osada, K.; Itaka, K.; Kataoka, K. Systemic Delivery of Messenger RNA for the Treatment of Pancreatic Cancer Using Polyplex Nanomicelles with a Cholesterol Moiety. Biomaterials 2016, 82, 221– 228, DOI: 10.1016/j.biomaterials.2015.12.03112Systemic delivery of messenger RNA for the treatment of pancreatic cancer using polyplex nanomicelles with a cholesterol moietyUchida, Satoshi; Kinoh, Hiroaki; Ishii, Takehiko; Matsui, Akitsugu; Tockary, Theofilus Agrios; Takeda, Kaori Machitani; Uchida, Hirokuni; Osada, Kensuke; Itaka, Keiji; Kataoka, KazunoriBiomaterials (2016), 82 (), 221-228CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)Systemic delivery of mRNA (mRNA) is tech. challenging because mRNA is highly susceptible to enzymic degrdn. in the blood circulation. In this study, we used a nanomicelle-based platform, prepd. from mRNA and poly(ethylene glycol) (PEG)-polycation block copolymers. A cholesterol (Chol) moiety was attached to the ω-terminus of the block copolymer to increase the stability of the nanomicelle by hydrophobic interaction. After in vitro screening, polyaspartamide with four aminoethylene repeats in its side chain (PAsp(TEP)) was selected as the cationic segment of the block copolymer, because it contributes to enhance nuclease resistance and high protein expression from the mRNA. After i.v. injection, PEG-PAsp(TEP)-Chol nanomicelles showed significantly enhanced blood retention of mRNA in comparison to nanomicelles without Chol. We used the nanomicelles for treating intractable pancreatic cancer in a s.c. inoculation mouse model through the delivery of mRNA encoding an anti-angiogenic protein (sFlt-1). PEG-PAsp(TEP)-Chol nanomicelles generated efficient protein expression from the delivered mRNA in tumor tissue, resulting in remarkable inhibition of the tumor growth, whereas nanomicelles without Chol failed to show a detectable therapeutic effect. In conclusion, the stabilized nanomicelle system led to the successful systemic delivery of mRNA in therapeutic application, holding great promise for the treatment of various diseases.
- 13Fotin-Mleczek, M.; Zanzinger, K.; Heidenreich, R.; Lorenz, C.; Kowalczyk, A.; Kallen, K.-J.; Huber, S. M. mRNA-Based Vaccines Synergize with Radiation Therapy to Eradicate Established Tumors. Radiat. Oncol. 2014, 9, 180, DOI: 10.1186/1748-717X-9-18013mRNA-based vaccines synergize with radiation therapy to eradicate established tumorsFotin-Mleczek, Mariola; Zanzinger, Kai; Heidenreich, Regina; Lorenz, Christina; Kowalczyk, Aleksandra; Kallen, Karl-Josef; Huber, Stephan M.Radiation Oncology (2014), 9 (), 180/1-180/23CODEN: ROANCM; ISSN:1748-717X. (BioMed Central Ltd.)Background: The eradication of large, established tumors by active immunotherapy is a major challenge because of the numerous cancer evasion mechanisms that exist. This study aimed to establish a novel combination therapy consisting of mRNA (mRNA)-based cancer vaccines and radiation, which would facilitate the effective treatment of established tumors with aggressive growth kinetics. Methods: The combination of a tumor-specific mRNA-based vaccination with radiation was tested in two syngeneic tumor models, a highly immunogenic E.G7-OVA and a low immunogenic Lewis lung cancer (LLC). The mol. mechanism induced by the combination therapy was evaluated via gene expression arrays as well as flow cytometry analyses of tumor infiltrating cells. Results: In both tumor models we demonstrated that a combination of mRNA-based immunotherapy with radiation results in a strong synergistic anti-tumor effect. This was manifested as either complete tumor eradication or delay in tumor growth. Gene expression anal. of mouse tumors revealed a variety of substantial changes at the tumor site following radiation. Genes assocd. with antigen presentation, infiltration of immune cells, adhesion, and activation of the innate immune system were upregulated. A combination of radiation and immunotherapy induced significant downregulation of tumor assocd. factors and upregulation of tumor suppressors. Moreover, combination therapy significantly increased CD4+, CD8+ and NKT cell infiltration of mouse tumors. Conclusion: Our data provide a scientific rationale for combining immunotherapy with radiation and provide a basis for the development of more potent anti-cancer therapies.
- 14McCullough, K. C.; 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. Mol. Ther.--Nucleic Acids 2014, 3, e173 DOI: 10.1038/mtna.2014.2414Self-replicating Replicon-RNA Delivery to Dendritic Cells by Chitosan-nanoparticles for Translation In Vitro and In VivoMcCullough, Kenneth C.; Bassi, Isabelle; Milona, Panagiota; Suter, Rolf; Thomann-Harwood, Lisa; Englezou, Pavlos; Demoulins, Thomas; Ruggli, NicolasMolecular Therapy--Nucleic Acids (2014), 3 (7), e173CODEN: MTAOC5; ISSN:2162-2531. (Nature Publishing Group)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 assocn. 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 concn.-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 assocd. with chitosan-nanoparticle delivery of self-amplifying RepRNA to DCs, leading to translation of encoded foreign genes, namely influenza virus hemagglutinin and nucleoprotein.
- 15Hekele, A.; Bertholet, S.; Archer, J.; Gibson, D. G.; Palladino, G.; Brito, L. A.; Otten, G. R.; Brazzoli, M.; Buccato, S.; Bonci, A.; Casini, D.; Maione, D.; Qi, Z.-Q.; Gill, J. E.; Caiazza, N. C.; Urano, J.; Hubby, B.; Gao, G. F.; Shu, Y.; De Gregorio, E.; Mandl, C. W.; Mason, P. W.; Settembre, E. C.; Ulmer, J. B.; Craig Venter, J.; Dormitzer, P. R.; Rappuoli, R.; Geall, A. J. Rapidly Produced SAM(®) Vaccine against H7N9 Influenza Is Immunogenic in Mice. Emerging Microbes Infect. 2013, 2, 1– 7, DOI: 10.1038/emi.2013.54There is no corresponding record for this reference.
- 16Brazzoli, M.; Magini, D.; Bonci, A.; Buccato, S.; Giovani, C.; Kratzer, R.; Zurli, V.; Mangiavacchi, S.; Casini, D.; Brito, L. M.; De Gregorio, E.; Mason, P. W.; Ulmer, J. B.; Geall, A. J.; Bertholet, S. Induction of Broad-Based Immunity and Protective Efficacy by Self-Amplifying mRNA Vaccines Encoding Influenza Virus Hemagglutinin. J. Virol. 2016, 90, 332, DOI: 10.1128/JVI.01786-1516Induction of broad-based immunity and protective efficacy by selfamplifying mRNA vaccines encoding influenza virus hemagglutininBrazzoli, Michela; Magini, Diletta; Bonci, Alessandra; Buccato, Scilla; Giovani, Cinzia; Kratzer, Roland; Zurli, Vanessa; Mangiavacchi, Simona; Casini, Daniele; Brito, Luis M.; De Gregorio, Ennio; Mason, Peter W.; Ulmer, Jeffrey B.; Geall, Andrew J.; Bertholet, SylvieJournal of Virology (2016), 90 (1), 332-344CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)Seasonal influenza is a vaccine-preventable disease that remains a major health problem worldwide, esp. in immunocompromised populations. The impact of influenza disease is even greater when strains drift, and influenza pandemics can result when animal-derived influenza virus strains combine with seasonal strains. In this study, we used the SAM technol. and characterized the immunogenicity and efficacy of a self-amplifying mRNA expressing influenza virus hemagglutinin (HA) antigen [SAM(HA)] formulated with a novel oil-in-water cationic nanoemulsion. We demonstrated that SAM(HA) was immunogenic in ferrets and facilitated containment of viral replication in the upper respiratory tract of influenza virus-infected animals. In mice, SAM(HA) induced potent functional neutralizing antibody and cellular immune responses, characterized by HA-specific CD4 T helper 1 and CD8 cytotoxic T cells. Furthermore, mice immunized with SAM(HA) derived from the influenza A virus A/California/7/2009 (H1N1) strain (Cal) were protected from a lethal challenge with the heterologous mouse-adapted A/PR/8/1934 (H1N1) virus strain (PR8). Sera derived from SAM(H1-Cal)-immunized animals were not cross-reactive with the PR8 virus, whereas cross-reactivity was obsd. for HA-specific CD4 and CD8 T cells. Finally, depletion of T cells demonstrated that T-cell responses were essential in mediating heterologous protection. If the SAM vaccine platform proves safe, well tolerated, and effective in humans, the fully synthetic SAM vaccine technol. could provide a rapid response platform to control pandemic influenza.
- 17Kauffman, K. J.; Dorkin, J. R.; Yang, J. H.; Heartlein, M. W.; DeRosa, F.; Mir, F. F.; Fenton, O. S.; Anderson, D. G. Optimization of Lipid Nanoparticle Formulations for mRNA Delivery In Vivo with Fractional Factorial and Definitive Screening Designs. Nano Lett. 2015, 15, 7300– 7306, DOI: 10.1021/acs.nanolett.5b0249717Optimization of Lipid Nanoparticle Formulations for mRNA Delivery in Vivo with Fractional Factorial and Definitive Screening DesignsKauffman, Kevin J.; Dorkin, J. Robert; Yang, Jung H.; Heartlein, Michael W.; De Rosa, Frank; Mir, Faryal F.; Fenton, Owen S.; Anderson, Daniel G.Nano Letters (2015), 15 (11), 7300-7306CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)Intracellular delivery of mRNA (mRNA) has the potential to induce protein prodn. for many therapeutic applications. Although lipid nanoparticles have shown considerable promise for the delivery of small interfering RNAs (siRNA), their utility as agents for mRNA delivery has only recently been investigated. The most common siRNA formulations contain four components: an amine-contg. lipid or lipid-like material, phospholipid, cholesterol, and lipid-anchored polyethylene glycol, the relative ratios of which can have profound effects on the formulation potency. Here, we develop a generalized strategy to optimize lipid nanoparticle formulations for mRNA delivery to the liver in vivo using Design of Expt. (DOE) methodologies including Definitive Screening and Fractional Factorial Designs. By simultaneously varying lipid ratios and structures, we developed an optimized formulation which increased the potency of erythropoietin-mRNA-loaded C12-200 lipid nanoparticles 7-fold relative to formulations previously used for siRNA delivery. Key features of this optimized formulation were the incorporation of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and increased ionizable lipid:mRNA wt. ratios. Interestingly, the optimized lipid nanoparticle formulation did not improve siRNA delivery, indicating differences in optimized formulation parameter design spaces for siRNA and mRNA. We believe the general method described here can accelerate in vivo screening and optimization of nanoparticle formulations with large multidimensional design spaces.
- 18Bahl, K.; Senn, J. J.; Yuzhakov, O.; Bulychev, A.; Brito, L. A.; Hassett, K. J.; Laska, M. E.; Smith, M.; Almarsson, Ö.; Thompson, J.; Ribeiro, A.; Watson, M.; Zaks, T.; Ciaramella, G. Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza Viruses. Mol. Ther. 2017, 25, 1316– 1327, DOI: 10.1016/j.ymthe.2017.03.03518Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza VirusesBahl, Kapil; Senn, Joe J.; Yuzhakov, Olga; Bulychev, Alex; Brito, Luis A.; Hassett, Kimberly J.; Laska, Michael E.; Smith, Mike; Almarsson, Orn; Thompson, James; Ribeiro, Amilcar; Watson, Mike; Zaks, Tal; Ciaramella, GiuseppeMolecular Therapy (2017), 25 (6), 1316-1327CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)Recently, the World Health Organization confirmed 120 new human cases of avian H7N9 influenza in China resulting in 37 deaths, highlighting the concern for a potential pandemic and the need for an effective, safe, and high-speed vaccine prodn. platform. Prodn. speed and scale of mRNA-based vaccines make them ideally suited to impede potential pandemic threats. Here we show that lipid nanoparticle (LNP)-formulated, modified mRNA vaccines, encoding hemagglutinin (HA) proteins of H10N8 (A/Jiangxi-Donghu/346/2013) or H7N9 (A/Anhui/1/2013), generated rapid and robust immune responses in mice, ferrets, and nonhuman primates, as measured by hemagglutination inhibition (HAI) and microneutralization (MN) assays. A single dose of H7N9 mRNA protected mice from a lethal challenge and reduced lung viral titers in ferrets. Interim results from a first-in-human, escalating-dose, phase 1 H10N8 study show very high seroconversion rates, demonstrating robust prophylactic immunity in humans. Adverse events (AEs) were mild or moderate with only a few severe and no serious events. These data show that LNP-formulated, modified mRNA vaccines can induce protective immunogenicity with acceptable tolerability profiles.
- 19van den Berg, J. H.; Nuijen, B.; Beijnen, J. H.; Vincent, A.; van Tinteren, H.; Kluge, J.; Woerdeman, L. A. E.; Hennink, W. E.; Storm, G.; Schumacher, T. N.; Haanen, J. B. A. G. Optimization of Intradermal Vaccination by DNA Tattooing in Human Skin. Hum. Gene Ther. 2009, 20, 181– 189, DOI: 10.1089/hum.2008.07319Optimization of Intradermal Vaccination by DNA Tattooing in Human Skinvan den Berg, Joost H.; Nuijen, Bastiaan; Beijnen, Jos H.; Vincent, Andrew; van Tinteren, Harm; Kluge, Joern; Woerdeman, Leonie A. E.; Hennink, Wim E.; Storm, Gert; Schumacher, Ton N.; Haanen, John B. A. G.Human Gene Therapy (2009), 20 (3), 181-189CODEN: HGTHE3; ISSN:1043-0342. (Mary Ann Liebert, Inc.)The intradermal administration of DNA vaccines by tattooing is a promising delivery technique for genetic immunization, with proven high immunogenicity in mice and in nonhuman primates. However, the parameters that result in optimal expression of DNA vaccines that are applied by this strategy to human skin are currently unknown. To address this issue we set up an ex vivo human skin model in which DNA vaccine-induced expression of reporter proteins could be monitored longitudinally. Using this model we demonstrate the following: First, the vast majority of cells that express DNA vaccine-encoded antigen in human skin are formed by epidermal keratinocytes, with only a small fraction (about 1%) of antigen-pos. epidermal Langerhans cells. Second, using full randomization of DNA tattoo variables we show that an increase in DNA concn., needle depth, and tattoo time all significantly increase antigen expression (p < 0.001), with DNA concn. forming the most crit. variable influencing the level of antigen expression. Finally, in spite of the marked immunogenicity of this vaccination method in animal models, transfection efficiency of the technique is shown to be extremely low, estd. at approx. 2 to 2000 out of 1 × 1010 copies of plasmid applied. This finding, coupled with the obsd. dependency of antigen expression on DNA concn., suggests that the development of strategies that can enhance in vivo transfection efficacy would be highly valuable. Collectively, this study shows that an ex vivo human skin model can be used to det. the factors that control vaccine-induced antigen expression and define the optimal parameters for the evaluation of DNA tattoo or other dermal delivery techniques in phase 1 clin. trials.
- 20Gelfant, S. Of Mice and Men” the Cell Cycle in Human Epidermis In Vivo. J. Invest. Dermatol. 1982, 78, 296– 299, DOI: 10.1111/1523-1747.ep1250736720"Of mice and men" the cell cycle in human epidermis in vivoGelfant SThe Journal of investigative dermatology (1982), 78 (4), 296-9 ISSN:0022-202X.This article deals with and compares cell cycle information obtained in mouse and in human epidermis in vivo. In order to compare data in mouse and in man, DNA labeling and mitotic index experiments were performed to obtain cell cycle information in normal human epidermis in vivo. Experiments were also performed on genetically inbred and outbred strains of mice--to provide a clue to the differences observed between mouse and man. The article makes the following points: 1. In contrast to mouse epidermis, there are no consistent diurnal fluctuations in and there is no cell kinetic relationship between mitotic and DNA-labeling indices in normal human epidermis in vivo. The variability from individual to individual in human subjects and the lack of cell cycle-related circadian fluctuations, preclude the use of statistical analysis and the use of conventional cell kinetic principles in understanding epidermal cell proliferation in man and may preclude the use of circadian rhythmicity for therapy scheduling. 2. The consistent and intelligible cell cycle information obtained in laboratory mice (as compared to man) is not due to the genetically inbred condition of mice. 3. This report introduces the use of ambient temperature as a potential nontoxic cell cycle tool for manipulating epidermal cell proliferation in the therapy of human proliferative skin diseases.
- 21Love, K. T.; Mahon, K. P.; Levins, C. G.; Whitehead, K. A.; Querbes, W.; Dorkin, J. R.; Qin, J.; Cantley, W.; Qin, L. L.; Racie, T.; Frank-Kamenetsky, M.; Yip, K. N.; Alvarez, R.; Sah, D. W. Y.; de Fougerolles, A.; Fitzgerald, K.; Koteliansky, V.; Akinc, A.; Langer, R.; Anderson, D. G. Lipid-Like Materials for Low-Dose, In Vivo Gene Silencing. Proc. Natl. Acad. Sci. U. S. A. 2010, 107, 1864– 1869, DOI: 10.1073/pnas.091060310621Lipid-like materials for low-dose, in vivo gene silencingLove, Kevin T.; Mahon, Kerry P.; Levins, Christopher G.; Whitehead, Kathryn A.; Querbes, William; Dorkin, J. Robert; Qin, June; Cantley, William; Qin, Liu Liang; Racie, Timothy; Frank-Kamenetsky, Maria; Yip, Ka Ning; Alvarez, Rene; Sah, Dinah W. Y.; de Fougerolles, Antonin; Fitzgerald, Kevin; Koteliansky, Victor; Akinc, Akin; Langer, Robert; Anderson, Daniel G.Proceedings of the National Academy of Sciences of the United States of America (2010), 107 (5), 1864-1869, S1864/1-S1864/6CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Significant effort has been applied to discover and develop vehicles which can guide small interfering RNAs (siRNA) through the many barriers guarding the interior of target cells. While studies have demonstrated the potential of gene silencing in vivo, improvements in delivery efficacy are required to fulfill the broadest potential of RNA interference therapeutics. Through the combinatorial synthesis and screening of a different class of materials, a formulation has been identified that enables siRNA-directed liver gene silencing in mice at doses below 0.01 mg/kg. This formulation was also shown to specifically inhibit expression of five hepatic genes simultaneously, after a single injection. The potential of this formulation was further validated in nonhuman primates, where high levels of knockdown of the clin. relevant gene transthyretin was obsd. at doses as low as 0.03 mg/kg. To our knowledge, this formulation facilitates gene silencing at orders-of-magnitude lower doses than required by any previously described siRNA liver delivery system.
- 22Ball, R. L.; Hajj, K. A.; Vizelman, J.; Bajaj, P.; Whitehead, K. A. Lipid Nanoparticle Formulations for Enhanced Co-Delivery of siRNA and mRNA. Nano Lett. 2018, 18, 3814– 3822, DOI: 10.1021/acs.nanolett.8b0110122Lipid nanoparticle formulations for enhanced co-delivery of siRNA and mRNABall, Rebecca L.; Hajj, Khalid A.; Vizelman, Jamie; Bajaj, Palak; Whitehead, Kathryn A.Nano Letters (2018), 18 (6), 3814-3822CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)Although mRNA and siRNA have significant therapeutic potential, their simultaneous delivery has not been previously explored. To facilitate the treatment of diseases assocd. with aberrant gene upregulation and downregulation, we sought to co-formulate siRNA and mRNA in a single lipidoid nanoparticle (LNP) formulation. We accommodated the distinct mol. characteristics of mRNA and siRNA in a formulation consisting of an ionizable and biodegradable amine-contg. lipidoid, cholesterol, DSPC, DOPE, and PEG-lipid. Surprisingly, the co-formulation of siRNA and mRNA in the same LNP enhanced the efficacy of both drugs in vitro and in vivo. Compared to LNPs encapsulating siRNA only, co-formulated LNPs improved Factor VII gene silencing in mice from 44 to 87% at an siRNA dose of 0.03 mg/kg. Co-formulation also improved mRNA delivery, as a 0.5 mg/kg dose of mRNA co-formulated with siRNA induced three times the luciferase protein expression compared to when siRNA was not included. As not all gene therapy applications require both RNA drugs, we sought to extend the benefit of co-formulated LNPs to formulations encapsulating only a single type of RNA. We accomplished this by substituting the "helper" RNA with a neg. charged polymer, polystyrenesulfonate (PSS). LNPs contg. PSS mediated the same level of protein silencing or expression as std. LNPs using 2-3-fold less RNA. For example, LNPs formulated with and without PSS induced 50% Factor VII silencing at siRNA doses of 0.01 and 0.03 mg/kg, resp. Together, these studies demonstrate potent co-delivery of siRNA and mRNA and show that inclusion of a neg. charged "helper polymer" enhances the efficacy of LNP delivery systems.
- 23Henriksen-Lacey, M.; Christensen, D.; Bramwell, V. W.; Lindenstrøm, T.; Agger, E. M.; Andersen, P.; Perrie, Y. Comparison of the Depot Effect and Immunogenicity of Liposomes Based on Dimethyldioctadecylammonium (DDA), 3β-[N-(N′,N′-Dimethylaminoethane)Carbomyl] Cholesterol (DC-Chol), and 1,2-Dioleoyl-3-Trimethylammonium Propane (DOTAP): Prolonged Liposome Retention Mediates Stronger Th1 Responses. Mol. Pharmaceutics 2011, 8, 153– 161, DOI: 10.1021/mp100208f23Comparison of the Depot Effect and Immunogenicity of Liposomes Based on Dimethyldioctadecylammonium (DDA), 3β-[N-(N',N'-Dimethylaminoethane)carbomyl] Cholesterol (DC-Chol), and 1,2-Dioleoyl-3-trimethylammonium Propane (DOTAP): Prolonged Liposome Retention Mediates Stronger Th1 ResponsesHenriksen-Lacey, Malou; Christensen, Dennis; Bramwell, Vincent W.; Lindenstrom, Thomas; Agger, Else Marie; Andersen, Peter; Perrie, YvonneMolecular Pharmaceutics (2011), 8 (1), 153-161CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The immunostimulatory capacities of cationic liposomes are well-documented and are attributed both to inherent immunogenicity of the cationic lipid and more phys. capacities such as the formation of antigen depots and antigen delivery. Very few studies have however been conducted comparing the immunostimulatory capacities of different cationic lipids. In the present study we therefore chose to investigate three of the most well-known cationic liposome-forming lipids as potential adjuvants for protein subunit vaccines. The ability of 3β-[N-(N',N'-dimethylaminoethane)carbomyl] cholesterol (DC-Chol), 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), and dimethyldioctadecylammonium (DDA) liposomes incorporating immunomodulating trehalose dibehenate (TDB) to form an antigen depot at the site of injection (SOI) and to induce immunol. recall responses against coadministered tuberculosis vaccine antigen Ag85B-ESAT-6 are reported. Furthermore, phys. characterization of the liposomes is presented. Our results suggest that liposome compn. plays an important role in vaccine retention at the SOI and the ability to enable the immune system to induce a vaccine specific recall response. While all three cationic liposomes facilitated increased antigen presentation by antigen presenting cells, the monocyte infiltration to the SOI and the prodn. of IFN-γ upon antigen recall was markedly higher for DDA and DC-Chol based liposomes which exhibited a longer retention profile at the SOI. A long-term retention and slow release of liposome and vaccine antigen from the injection site hence appears to favor a stronger Th1 immune response.
- 24De Serrano, L. O.; Burkhart, D. J. Liposomal Vaccine Formulations as Prophylactic Agents: Design Considerations for Modern Vaccines. J. Nanobiotechnol. 2017, 15, 83, DOI: 10.1186/s12951-017-0319-924Liposomal vaccine formulations as prophylactic agents: design considerations for modern vaccinesDe Serrano, Luis O.; Burkhart, David J.Journal of Nanobiotechnology (2017), 15 (), 83/1-83/23CODEN: JNOAAO; ISSN:1477-3155. (BioMed Central Ltd.)A review. Vaccinol. is one of the most important cornerstones in modern medicine, providing better quality of life. The human immune system is composed of innate and adaptive immune processes that interplay when infection occurs. Innate immunity relies on pathogen-assocd. mol. patterns which are recognized by pathogen recognition receptors localized in antigen presenting cells. After antigen processing and presentation, CD4+ T cell polarization occurs, further leading to B cell and CD8+ activation and humoral and cell-mediated adaptive immune responses. Liposomes are being employed as vaccine technologies and their design is of importance to ensure proper immune responses. Physicochem. parameters like liposome size, charge, lamellarity and bilayer fluidity must be completely understood to ensure optimal vaccine stability and efficacy. Liposomal vaccines can be developed to target specific immune cell types for the induction of certain immune responses. In this review, we will present promising liposomal vaccine approaches for the treatment of important viral, bacterial, fungal and parasitic infections (including tuberculosis, TB). Cationic liposomes are the most studied liposome types due to their enhanced interaction with the neg. charged immune cells. Thus, a special section on the cationic lipid dimethyldioctadecylammonium and TB is also presented.
- 25Martino, S.; di Girolamo, I.; Tiribuzi, R.; D’Angelo, F.; Datti, A.; Orlacchio, A. Efficient siRNA Delivery by the Cationic Liposome DOTAP in Human Hematopoietic Stem Cells Differentiating into Dendritic Cells. J. Biomed. Biotechnol. 2009, 2009, 410260, DOI: 10.1155/2009/41026025Efficient siRNA delivery by the cationic liposome DOTAP in human hematopoietic stem cells differentiating into dendritic cellsMartino Sabata; di Girolamo Ilaria; Tiribuzi Roberto; D'Angelo Francesco; Datti Alessandro; Orlacchio AldoJournal of biomedicine & biotechnology (2009), 2009 (), 410260 ISSN:.RNA interference technology is an ideal strategy to elucidate the mechanisms associated with human CD34(+) hematopoietic stem cell differentiation into dendritic cells. Simple manipulations in vitro can unequivocally yield alloreactive or tolerogenic populations, suggesting key implications of biochemical players that might emerge as therapeutic targets for cancer or graft-versus-host disease. To knockdown proteins typically involved in the biology of dendritic cells, we employed an siRNA delivery system based on the cationic liposome DOTAP as the carrier. Freshly-isolated CD34(+) cells were transfected with siRNA for cathepsin S with negligible cytotoxicity and transfection rates (>60%) comparable to the efficiency shown by lentiviral vectors. Further, cathepsin S knockdown was performed during both cell commitment and through the entire 14-day differentiation process with repeated transfection rounds that had no effect per se on cell development. Tested in parallel, other commercially-available chemical reagents failed to meet acceptable standards. In addition to safe and practical handling, a direct advantage of DOTAP over viral-mediated techniques is that transient silencing effects can be dynamically appraised through the recovery of targeted proteins. Thus, our findings identify DOTAP as an excellent reagent for gene silencing in resting and differentiating CD34(+) cells, suggesting a potential for applications in related preclinical models.
- 26Kaneda, M. M.; Sasaki, Y.; Lanza, G. M.; Milbrandt, J.; Wickline, S. A. Mechanisms of Nucleotide Trafficking During siRNA Delivery to Endothelial Cells Using Perfluorocarbon Nanoemulsions. Biomaterials 2010, 31, 3079– 3086, DOI: 10.1016/j.biomaterials.2010.01.00626Mechanisms of nucleotide trafficking during siRNA delivery to endothelial cells using perfluorocarbon nanoemulsionsKaneda, Megan M.; Sasaki, Yo; Lanza, Gregory M.; Milbrandt, Jeffrey; Wickline, Samuel A.Biomaterials (2010), 31 (11), 3079-3086CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)RNA interference (RNAi) is a useful in vitro research tool, but its application as a safe and effective therapeutic agent may benefit from improved understanding of mechanisms of exogenous siRNA delivery, including cell trafficking and sorting patterns. We report the development of a transfection reagent for siRNA delivery which employs a distinctive non-digestive mode of particle-cell membrane interaction through the formation of a hemifusion complex resulting in lipid raft transport of cargo to the cytosol, bypassing the usual endosomal nanoparticle uptake pathway. We further demonstrate markedly enhanced efficacy over conventional transfection agents for suppressing endothelial cell expression of upregulated vascular adhesion mols.
- 27Zhang, X.; Li, B.; Luo, X.; Zhao, W.; Jiang, J.; Zhang, C.; Gao, M.; Chen, X.; Dong, Y. Biodegradable Amino-Ester Nanomaterials for Cas9 mRNA Delivery In Vitro and In Vivo. ACS Appl. Mater. Interfaces 2017, 9, 25481– 25487, DOI: 10.1021/acsami.7b0816327Biodegradable Amino-Ester Nanomaterials for Cas9 mRNA Delivery in Vitro and in VivoZhang, Xinfu; Li, Bin; Luo, Xiao; Zhao, Weiyu; Jiang, Justin; Zhang, Chengxiang; Gao, Min; Chen, Xiaofang; Dong, YizhouACS Applied Materials & Interfaces (2017), 9 (30), 25481-25487CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)Efficient and safe delivery of the CRISPR/Cas system is one of the key challenges for genome-editing applications in humans. Herein, we designed and synthesized a series of biodegradable lipidlike compds. contg. ester groups for the delivery of mRNA-encoding Cas9. Two lead materials, termed N-methyl-1,3-propanediamine (MPA)-A and MPA-Ab, showed a tunable rate of biodegrdn. MPA-A with linear ester chains was degraded dramatically faster than MPA-Ab with branched ester chains in the presence of esterase or in wild-type mice. Most importantly, MPA-A and MPA-Ab demonstrated efficient delivery of Cas9 mRNA both in vitro and in vivo. Consequently, these biodegradable lipidlike nanomaterials merit further development as genome-editing delivery tools for biol. and therapeutic applications.
- 28Kim, B.-K.; Hwang, G.-B.; Seu, Y.-B.; Choi, J.-S.; Jin, K. S.; Doh, K.-O. DOTAP/DOPE Ratio and Cell Type Determine Transfection Efficiency with DOTAP-Liposomes. Biochim. Biophys. Acta, Biomembr. 2015, 1848, 1996– 2001, DOI: 10.1016/j.bbamem.2015.06.02028DOTAP/DOPE ratio and cell type determine transfection efficiency with DOTAP-liposomesKim, Bieong-Kil; Hwang, Guen-Bae; Seu, Young-Bae; Choi, Jong-Soo; Jin, Kyeong Sik; Doh, Kyung-OhBiochimica et Biophysica Acta, Biomembranes (2015), 1848 (10_Part_A), 1996-2001CODEN: BBBMBS; ISSN:0005-2736. (Elsevier B.V.)The effects of lipid compns. on their physicochem. properties and transfection efficiencies were investigated. Four liposome formulations with different 1,2-dioleoyl-3-trimethylammoniumpropane (DOTAP) to dioleoylphosphatidylethanolamine (DOPE) wt. ratios were investigated, i.e., wt. ratios 1:0 (T1P0), 3:1 (T3P1), 1:1 (T1P1), and 1:3 (T1P3). Mean sizes of liposomes were influenced by their lipid compn. and the prepn. concn. at the time of sonication. Zeta potentials of liposomes were inversely correlated with their liposome sizes. However, neither liposome sizes nor zeta potentials were correlated with transfection efficiency. The optimum compn. of liposomes was cell-line dependent (T1P0 and T3P1 for Huh7 and AGS, T3P1 and T1P1 for COS7, and T1P1 and T1P3 for A549). The shape of lipoplexes was changed from lamellar to inverted hexagonal structure according to the increased ratio of DOPE, but there was no definite advantage of specific structure in transfection efficiency throughout all used cell lines. However, cellular internalization was consistently faster in T1P0, T3P1, T1P1 compared to T1P3 in all cell lines, suggesting the importance of endosomal escape. Our findings show that the transfection efficiency of DOTAP liposomes is mainly influenced by lipid compn. and cell type, and not by size or zeta potential.
- 29Mével, M.; Haudebourg, T.; Colombani, T.; Peuziat, P.; Dallet, L.; Chatin, B.; Lambert, O.; Berchel, M.; Montier, T.; Jaffrès, P.-A.; Lehn, P.; Pitard, B. Important Role of Phosphoramido Linkage in Imidazole-Based Dioleyl Helper Lipids for Liposome Stability and Primary Cell Transfection. J. Gene Med. 2016, 18, 3– 15, DOI: 10.1002/jgm.286929Important role of phosphoramido linkage in imidazole-based dioleyl helper lipids for liposome stability and primary cell transfectionMevel, Mathieu; Haudebourg, Thomas; Colombani, Thibault; Peuziat, Pauline; Dallet, Laurence; Chatin, Benoit; Lambert, Olivier; Berchel, Mathieu; Montier, Tristan; Jaffres, Paul-Alain; Lehn, Pierre; Pitard, BrunoJournal of Gene Medicine (2016), 18 (1-3), 3-15CODEN: JGMEFG; ISSN:1521-2254. (Wiley-Blackwell)Background : To optimize synthetic gene delivery systems, there is a need to develop more efficient lipid formulations. Most cationic lipid formulations contain 'helper' neutral lipids because of their ability to increase DNA delivery, in particular by improving endosomal escape of DNA mols. via the pH-buffering effect of protonatable groups and/or fusion with the lipid bilayer of endosomes. Methods : We evaluated the influence of the linker structure between the two oleyl chains in the helper lipid on transfection efficiency in cell lines, as well as in primary cells (hepatocytes/cardiomyocytes). We reported the synthesis of two new pH-buffering imidazole helper lipids characterized by a polar headgroup contg. one (compd. 6) or two (compd. 5) imidazole groups and two oleyl chains linked by an amide group. We studied their assocn. with the aminoglycoside lipidic deriv. dioleylsuccinylparomomycin (DOSP), which contains two oleyl chains linked to the aminoglycoside polar headgroup via an amide function. We compared the morphol. and transfection properties of such binary liposomes of DOSP/5 and DOSP/6 with those of liposomes combining DOSP with another imidazole-based dioleyl helper lipid (MM27) in which a phosphoramido group acts as a linker between the two oleyl chains and imidazole function. Results : The phosphoramido linker in the helper lipid induces a major difference in terms of morphol. and resistance to decomplexation at phys. pH for DOSP/helper lipid complexes. Conclusions : This hybrid dioleyl linker compn. of DOSP/MM27 led to higher transfection efficiency in cell lines and in primary cells compared to complexes with homogeneous dioleyl linker. Copyright © 2015 John Wiley & Sons, Ltd.
- 30Cavalcanti, R. R. M.; Lira, R. B.; Riske, K. A. Study of the Fusion Mechanism of Fusogenic Cationic Liposomes with Anionic Model Membranes. Biophys. J. 2018, 114, 606a, DOI: 10.1016/j.bpj.2017.11.3314There is no corresponding record for this reference.
- 31Démoulins, T.; Milona, P.; Englezou, P. C.; Ebensen, T.; Schulze, K.; Suter, R.; Pichon, C.; Midoux, P.; Guzmán, C. A.; Ruggli, N.; McCullough, K. C. Polyethylenimine-Based Polyplex Delivery of Self-Replicating RNA Vaccines. Nanomedicine (N. Y., NY, U. S.) 2016, 12, 711– 722, DOI: 10.1016/j.nano.2015.11.00131Polyethylenimine-based polyplex delivery of self-replicating RNA vaccinesDemoulins, Thomas; Milona, Panagiota; Englezou, Pavlos C.; Ebensen, Thomas; Schulze, Kai; Suter, Rolf; Pichon, Chantal; Midoux, Patrick; Guzman, Carlos A.; Ruggli, Nicolas; McCullough, Kenneth C.Nanomedicine (New York, NY, United States) (2016), 12 (3), 711-722CODEN: NANOBF; ISSN:1549-9634. (Elsevier)Self-amplifying replicon RNA (RepRNA) are large mols. (12-14 kb); their self-replication amplifies mRNA template nos., affording several rounds of antigen prodn., effectively increasing vaccine antigen payloads. Their sensitivity to RNase-sensitivity and inefficient uptake by dendritic cells (DCs) - abs. requirements for vaccine design - were tackled by condensing RepRNA into synthetic, nanoparticulate, polyethylenimine (PEI)-polyplex delivery vehicles. Polyplex-delivery formulations for small RNA mols. cannot be transferred to RepRNA due to its greater size and complexity; the N:P charge ratio and impact of RepRNA folding would influence polyplex condensation, post-delivery decompaction and the cytosolic release essential for RepRNA translation. Polyplex-formulations proved successful for delivery of RepRNA encoding influenza virus hemagglutinin and nucleocapsid to DCs. Cytosolic translocation was facilitated, leading to RepRNA translation. This efficacy was confirmed in vivo, inducing both humoral and cellular immune responses. Accordingly, this paper describes the first PEI-polyplexes providing efficient delivery of the complex and large, self-amplifying RepRNA vaccines.
- 32Pardi, N.; Parkhouse, K.; Kirkpatrick, E.; McMahon, M.; Zost, S. J.; Mui, B. L.; Tam, Y. K.; Karikó, K.; Barbosa, C. J.; Madden, T. D.; Hope, M. J.; Krammer, F.; Hensley, S. E.; Weissman, D. Nucleoside-Modified mRNA Immunization Elicits Influenza Virus Hemagglutinin Stalk-Specific Antibodies. Nat. Commun. 2018, 9, 3361, DOI: 10.1038/s41467-018-05482-032Nucleoside-modified mRNA immunization elicits influenza virus hemagglutinin stalk-specific antibodiesPardi Norbert; Weissman Drew; Parkhouse Kaela; Zost Seth J; Hensley Scott E; Kirkpatrick Ericka; McMahon Meagan; Krammer Florian; Kirkpatrick Ericka; Mui Barbara L; Tam Ying K; Barbosa Christopher J; Madden Thomas D; Hope Michael J; Kariko KatalinNature communications (2018), 9 (1), 3361 ISSN:.Currently available influenza virus vaccines have inadequate effectiveness and are reformulated annually due to viral antigenic drift. Thus, development of a vaccine that confers long-term protective immunity against antigenically distant influenza virus strains is urgently needed. The highly conserved influenza virus hemagglutinin (HA) stalk represents one of the potential targets of broadly protective/universal influenza virus vaccines. Here, we evaluate a potent broadly protective influenza virus vaccine candidate that uses nucleoside-modified and purified mRNA encoding full-length influenza virus HA formulated in lipid nanoparticles (LNPs). We demonstrate that immunization with HA mRNA-LNPs induces antibody responses against the HA stalk domain of influenza virus in mice, rabbits, and ferrets. The HA stalk-specific antibody response is associated with protection from homologous, heterologous, and heterosubtypic influenza virus infection in mice.
- 33Reichmuth, A. M.; Oberli, M. A.; Jaklenec, A.; Langer, R.; Blankschtein, D. mRNA Vaccine Delivery Using Lipid Nanoparticles. Ther. Delivery 2016, 7, 319– 334, DOI: 10.4155/tde-2016-000633mRNA vaccine delivery using lipid nanoparticlesReichmuth, Andreas M.; Oberli, Matthias A.; Jeklenec, Ana; Langer, Robert; Blankschtein, DanielTherapeutic Delivery (2016), 7 (5), 319-334CODEN: TDHEA7; ISSN:2041-5990. (Future Science Ltd.)MRNA vaccines elicit a potent immune response including antibodies and cytotoxic T cells. MRNA vaccines are currently evaluated in clin. 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 degrdn., facilitate endosomal escape, can be targeted to the desired cell type by surface decoration with ligands, and as needed, can be codelivered with adjuvants.
- 34Albanese, A.; Tang, P. S.; Chan, W. C. W. The Effect of Nanoparticle Size, Shape, and Surface Chemistry on Biological Systems. Annu. Rev. Biomed. Eng. 2012, 14, 1– 16, DOI: 10.1146/annurev-bioeng-071811-15012434The effect of nanoparticle size, shape, and surface chemistry on biological systemsAlbanese, Alexandre; Tang, Peter S.; Chan, Warren C. W.Annual Review of Biomedical Engineering (2012), 14 (), 1-16CODEN: ARBEF7; ISSN:1523-9829. (Annual Reviews Inc.)A review. An understanding of the interactions between nanoparticles and biol. systems is of significant interest. Studies aimed at correlating the properties of nanomaterials such as size, shape, chem. functionality, surface charge, and compn. with biomol. signaling, biol. kinetics, transportation, and toxicity in both cell culture and animal expts. are under way. These fundamental studies will provide a foundation for engineering the next generation of nanoscale devices. Here, the authors provide rationales for these studies, review the current progress in studies of the interactions of nanomaterials with biol. systems, and provide a perspective on the long-term implications of these findings.
- 35Golestani, R.; Pourfathollah, A. A.; Moazzeni, S. M. An Extreme Strategy for the Production of Hybridoma. Hybridoma 2009, 28, 139– 144, DOI: 10.1089/hyb.2008.007635An Extreme Strategy for the Production of HybridomaGolestani, Reza; Pourfathollah, Ali Akbar; Moazzeni, Seyed MohammadHybridoma (2009), 28 (2), 139-144CODEN: HYBRAV; ISSN:1554-0014. (Mary Ann Liebert, Inc.)The ethical issues surrounding human immunization hamper the prodn. of human monoclonal antibody through scarcity of immunized B cells in peripheral blood. This defect can be compensated in part by improvement of hybridoma prodn. techniques. We have developed a new strategy to bypass the toxic effects of polyethylene glycol (PEG) as fusogenic reagent and hypoxanthine aminoptrin thymidine (HAT) as selective medium on newly fused cells. The Epstein-Barr virus (EBV) transformed peripheral blood mononuclear cells (PBMC) of accidentally Rh antigen sensitized persons were fused using cephalin as fugenic reagent, with emetine and actinomycin D pretreated heteromyeloma cells. Our results showed that 19-34% of EBV-transformed B cells were grown as hybridoma clones following selection. This extreme improvement in hybridoma prodn. rate may end the fusion efficiency problem and make hybridoma prodn. a plug-and-play technol.
- 36Phua, K. K. L.; Leong, K. W.; Nair, S. K. Transfection Efficiency and Transgene Expression Kinetics of mRNA Delivered in Naked and Nanoparticle Format. J. Controlled Release 2013, 166, 227– 233, DOI: 10.1016/j.jconrel.2012.12.02936Transfection efficiency and transgene expression kinetics of mRNA delivered in naked and nanoparticle formatPhua, Kyle K. L.; Leong, Kam W.; Nair, Smita K.Journal of Controlled Release (2013), 166 (3), 227-233CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Transfection efficiencies and transgene expression kinetics of mRNA, an emerging class of nucleic acid-based therapeutics, were poorly characterized. In this study, the authors evaluated transfection efficiencies of mRNA delivered in naked and nanoparticle format in vitro and in vivo using GFP and luciferase as reporters. While mRNA nanoparticles transfect primary human and mouse dendritic cells (DCs) efficiently in vitro, naked mRNA could not produce any detectable gene product. The protein expression of nanoparticle-mediated transfection in vitro peaks rapidly within 5-7 h and decays in a biphasic manner. In vivo, naked mRNA is more efficient than mRNA nanoparticles when administered s.c. In contrast, mRNA nanoparticle performs better when administered intranasally and i.v. Gene expression is most transient when delivered i.v. in nanoparticle format with an apparent half-life of 1.4 h and lasts less than 24 h, and most sustained when delivered in the naked format s.c. at the base of tail with an apparent half-life of 18 h and persists for at least 6 days. Notably, exponential decreases in protein expression are consistently obsd. post-delivery of mRNA in vivo regardless of the mode of delivery (naked or nanoparticle) or the site of administration. This study elucidates the performance of mRNA transfection and suggests a niche for mRNA therapeutics when predictable in vivo transgene expression kinetics is imperative.
- 37Liang, F.; Lindgren, G.; Lin, A.; Thompson, E. A.; Ols, S.; Röhss, J.; John, S.; Hassett, K.; Yuzhakov, O.; Bahl, K.; Brito, L. A.; Salter, H.; Ciaramella, G.; Loré, K. Efficient Targeting and Activation of Antigen-Presenting Cells In Vivo after Modified mRNA Vaccine Administration in Rhesus Macaques. Mol. Ther. 2017, 25, 2635– 2647, DOI: 10.1016/j.ymthe.2017.08.00637Efficient Targeting and Activation of Antigen-Presenting Cells In Vivo after Modified mRNA Vaccine Administration in Rhesus MacaquesLiang, Frank; Lindgren, Gustaf; Lin, Ang; Thompson, Elizabeth A.; Ols, Sebastian; Roehss, Josefine; John, Shinu; Hassett, Kimberly; Yuzhakov, Olga; Bahl, Kapil; Brito, Luis A.; Salter, Hugh; Ciaramella, Giuseppe; Lore, KarinMolecular Therapy (2017), 25 (12), 2635-2647CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)MRNA vaccines are rapidly emerging as a powerful platform for infectious diseases because they are well tolerated, immunogenic, and scalable and are built on precise but adaptable antigen design. We show that two immunizations of modified non-replicating mRNA encoding influenza H10 hemagglutinin (HA) and encapsulated in lipid nanoparticles (LNP) induce protective HA inhibition titers and H10-specific CD4+ T cell responses after i.m. or intradermal delivery in rhesus macaques. Administration of LNP/mRNA induced rapid and local infiltration of neutrophils, monocytes, and dendritic cells (DCs) to the site of administration and the draining lymph nodes (LNs). While these cells efficiently internalized LNP, mainly monocytes and DCs translated the mRNA and upregulated key co-stimulatory receptors (CD80 and CD86). This coincided with upregulation of type I IFN-inducible genes, including MX1 and CXCL10. The innate immune activation was transient and resulted in priming of H10-specific CD4+ T cells exclusively in the vaccine-draining LNs. Collectively, this demonstrates that mRNA-based vaccines induce type-I IFN-polarized innate immunity and, when combined with antigen prodn. by antigen-presenting cells, lead to generation of potent vaccine-specific responses.
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsnano.9b01774.
LNP characterization of particle size and surface charge (Figure S1); eGFP expression gating strategy (Figure S2); skin explant cell viability (Figure S3); identity of cells present in total population and GFP+ cells (Figure S4); cell viability of human skin explants after LNP-formulation saRNA injection (Figure S5); antibodies used for flow cytometry (Table S1) (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.