Generic placeholder image

Pharmaceutical Nanotechnology

Editor-in-Chief

ISSN (Print): 2211-7385
ISSN (Online): 2211-7393

Research Article

Investigating the Fate of MP1000-LPX In Vivo by Adding Serum to Transfection Medium

Author(s): Siyan He, Shan Xia, Xiangrong Song, Hai Huang, Xueyan Wang, Xuehua Jiang and Zhaohui Jin*

Volume 8, Issue 5, 2020

Page: [399 - 408] Pages: 10

DOI: 10.2174/2211738508666200907105224

Price: $65

Abstract

Background: Cationic liposomes (CLs) based messenger RNA (mRNA) vaccine has been a promising approach for cancer treatment. However, rapid lung accumulation after intraveous injection and significantly decreased transfection efficacy (TE) in serum substantially hamper its application.

Objective: In this study, we attempt to investigate the fate of Mannose-PEG1000-lipoplex (MP1000-LPX) in vivo, a previous reported mRNA vaccine, and potential mechanism in it.

Methods: MP1000-CLs and different type of MP1000-LPX were produced by previous method and characterized by dynamic light scattering (DLS). Organ distribution and Luc-mRNA expression of DiD loaded luciferase (Luc-mRNA)-MP1000-LPX were evaluated by IVIS Spectrum imaging system. Cellular transfection and uptake under serum-free and serum-containing conditions were analysed by flow cytometry and counted by FlowJo software.

Results: MP1000-CLs had an average size of 45.3 ± 0.9 nm, a positive charge of 39.9 ± 0.9 mV. When MP1000-LPX formed, the particle size increased to about 130 nm, and zeta potential decreased to about 30 mV. All formulations were in narrow size distribution with PDI < 0.3. 6 h after intraveous injection, Luc-MP1000-LPX mostly distributed to liver, lung and spleen, while only successfully expressed Luc in lung. DC2.4 cellular transfection assay indicated serum substantially lowered TE of MP1000-LPX. However, the cellular uptake on DC2.4 cells was enhanced in the presence of serum.

Conclusion: MP1000-LPX distributed to spleen but failed to transfect. Because serum dramatically decreased TE of MP1000-LPX on DC2.4 cells, but not by impeding its interaction to cell membrane. Serum resistance and avoidance of lung accumulation might be prerequisites for CLs based intravenous mRNA vaccines.

Lay Summary: mRNA vaccine has been promising immunotherapy to treat cancer by delivering mRNA encoding tumor antigens to APCs and activating immune system against tumor cells. We are investigating the in vivo fate of MP1000-LPX, a CLs based mRNA vaccine. To see if serum causes the fate, we’ll be looking at the influence of serum on transfection and uptake efficacy of MP1000-LPX by DC2.4 cells experiments in vitro. Our findings will imply that serum inhibits transfection but not by decreasing uptake. Thus, we can ultilize serum to enhance transfection if we make intracellular process of MP1000-LPX successful.

Keywords: Cationic liposomes, cellular uptake, DC2.4 cell line, gene delivery, serum, transfection efficacy.

Graphical Abstract
[1]
Miao L, Li L, Huang Y, et al. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation. Nat Biotechnol 2019; 37(10): 1174-85.
[http://dx.doi.org/10.1038/s41587-019-0247-3] [PMID: 31570898]
[2]
Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov 2018; 17(4): 261-79.
[http://dx.doi.org/10.1038/nrd.2017.243 PMID: 29326426]
[3]
Perche F, Benvegnu T, Berchel M, et al. Enhancement of dendritic cells transfection in vivo and of vaccination against B16F10 melanoma with mannosylated histidylated lipopolyplexes loaded with tumor antigen messenger RNA. Nanomedicine (Lond) 2011; 7(4): 445-53.
[http://dx.doi.org/10.1016/j.nano.2010.12.010] [PMID: 21220051]
[4]
Sahin U, Karikó K, Türeci Ö. mRNA-based therapeutics--developing a new class of drugs. Nat Rev Drug Discov 2014; 13(10): 759-80.
[http://dx.doi.org/10.1038/nrd4278] [PMID: 25233993]
[5]
Kowalski PS, Rudra A, Miao L, Anderson DG. Delivering the messenger: advances in technologies for therapeutic mRNA delivery. Mol Ther 2019; 27(4): 710-28.
[http://dx.doi.org/10.1016/j.ymthe.2019.02.012] [PMID: 30846391]
[6]
Reichmuth AM, Oberli MA, Jaklenec A, Langer R, Blankschtein D. mRNA vaccine delivery using lipid nanoparticles. Ther Deliv 2016; 7(5): 319-34.
[http://dx.doi.org/10.4155/tde-2016-0006] [PMID: 27075952]
[7]
Malone RW, Felgner PL, Verma IM. Cationic liposome-mediated RNA transfection. Proc Natl Acad Sci USA 1989; 86(16): 6077-81.
[http://dx.doi.org/10.1073/pnas.86.16.6077 PMID: 2762315]
[8]
Porteous DJ, Dorin JR, McLachlan G, et al. Evidence for safety and efficacy of DOTAP cationic liposome mediated CFTR gene transfer to the nasal epithelium of patients with cystic fibrosis. Gene Ther 1997; 4(3): 210-8.
[http://dx.doi.org/10.1038/sj.gt.3300390 PMID: 9135734]
[9]
Digiacomo L, Pozzi D, Amenitsch H, Caracciolo G. Impact of the biomolecular corona on the structure of PEGylated liposomes. Biomater Sci 2017; 5(9): 1884-8.
[http://dx.doi.org/10.1039/C7BM00387K PMID: 28676869]
[10]
Guan S, Rosenecker J. Nanotechnologies in delivery of mRNA therapeutics using nonviral vector-based delivery systems. Gene Ther 2017; 24(3): 133-43.
[http://dx.doi.org/10.1038/gt.2017.5] [PMID: 28094775]
[11]
Qiu Y, Guo L, Zhang S, et al. DNA-based vaccination against hepatitis B virus using dissolving microneedle arrays adjuvanted by cationic liposomes and CpG ODN. Drug Deliv 2016; 23(7): 2391-8.
[http://dx.doi.org/10.3109/10717544.2014.992497] [PMID: 25625495]
[12]
Diao L, Tao J, Wang Y, Hu Y, He W. Co-delivery of dihydroartemisinin and HMGB1 siRNA By TAT-modified cationic liposomes through the TLR4 signaling pathway for treatment of lupus nephritis. Int J Nanomedicine 2019; 14: 8627-45.
[http://dx.doi.org/10.2147/IJN.S220754 PMID: 31806961]
[13]
Markov OO, Mironova NL, Maslov MA, et al. Novel cationic liposomes provide highly efficient delivery of DNA and RNA into dendritic cell progenitors and their immature offsets. J Control Release 2012; 160(2): 200-10.
[http://dx.doi.org/10.1016/j.jconrel.2011.11.034] [PMID: 22155599]
[14]
Khademi F, Taheri RA, Momtazi-Borojeni AA, Farnoosh G, Johnston TP, Sahebkar A. Potential of cationic liposomes as adjuvants/delivery systems for tuberculosis subunit vaccines. Rev Physiol Biochem Pharmacol 2018; 175: 47-69.
[http://dx.doi.org/10.1007/112_2018_9 PMID: 29700609]
[15]
Chan CL, Ewert KK, Majzoub RN, et al. Optimizing cationic and neutral lipids for efficient gene delivery at high serum content. J Gene Med 2014; 16(3-4): 84-96.
[http://dx.doi.org/10.1002/jgm.2762] [PMID: 24753287]
[16]
Ishiwata H, Suzuki N, Ando S, Kikuchi H, Kitagawa T. Characteristics and biodistribution of cationic liposomes and their DNA complexes. J Control Release 2000; 69(1): 139-48.
[http://dx.doi.org/10.1016/S0168-3659(00)00293-5] [PMID: 11018552]
[17]
Wonder E, Simón-Gracia L, Scodeller P, et al. Competition of charge-mediated and specific binding by peptide-tagged cationic liposome-DNA nanoparticles in vitro and in vivo. Biomaterials 2018; 166: 52-63.
[http://dx.doi.org/10.1016/j.biomaterials.2018.02.052] [PMID: 29544111]
[18]
Martin-Herranz A, Ahmad A, Evans HM, Ewert K, Schulze U, Safinya CR. Surface functionalized cationic lipid-DNA complexes for gene delivery: PEGylated lamellar complexes exhibit distinct DNA-DNA interaction regimes. Biophys J 2004; 86(2): 1160-8.
[http://dx.doi.org/10.1016/S0006-3495(04)74190-9] [PMID: 14747350]
[19]
Majzoub RN, Chan CL, Ewert KK, et al. Uptake and transfection efficiency of PEGylated cationic liposome-DNA complexes with and without RGD-tagging. Biomaterials 2014; 35(18): 4996-5005.
[http://dx.doi.org/10.1016/j.biomaterials.2014.03.007] [PMID: 24661552]
[20]
Wang F, Xiao W, Elbahnasawy MA, et al. Optimization of the linker length of mannose-cholesterol conjugates for enhanced mRNA delivery to dendritic cells by liposomes. Front Pharmacol 2018; 9: 980.
[http://dx.doi.org/10.3389/fphar.2018.00980] [PMID: 30233368]
[21]
Ding GB, Meng X, Yang P, Li B, Stauber RH, Li Z. Integration of polylactide into polyethylenimine facilitates the safe and effective intracellular siRNA delivery. Polymers (Basel) 2020; 12(2): 445.
[http://dx.doi.org/10.3390/polym12020445] [PMID: 32074943]
[22]
Koo BH, Hong D, Hong HD, et al. Arginase II activity regulates cytosolic Ca2+ level in a p32-dependent manner that contributes to Ca2+-dependent vasoconstriction in native low-density lipoprotein-stimulated vascular smooth muscle cells. Exp Mol Med 2019; 51(6): 1-12.
[http://dx.doi.org/10.1038/s12276-019-0262-y] [PMID: 31155612]
[23]
Sternberg B, Sorgi FL, Huang L. New structures in complex formation between DNA and cationic liposomes visualized by freeze-fracture electron microscopy. FEBS Lett 1994; 356(2-3): 361-6.
[http://dx.doi.org/10.1016/0014-5793(94)01315-2] [PMID: 7805873]
[24]
McLean JW, Fox EA, Baluk P, et al. Organ-specific endothelial cell uptake of cationic liposome-DNA complexes in mice. Am J Physiol 1997; 273(1 Pt 2): H387-404.
[http://dx.doi.org/10.1152/ajpheart.1997.273.1.H387] [PMID: 9249514]
[25]
Yang JP, Huang L. Overcoming the inhibitory effect of serum on lipofection by increasing the charge ratio of cationic liposome to DNA. Gene Ther 1997; 4(9): 950-60.
[http://dx.doi.org/10.1038/sj.gt.3300485 PMID: 9349432]
[26]
Marelli-Berg FM, Jarmin SJ. Antigen presentation by the endothelium: a green light for antigen-specific T cell trafficking? Immunol Lett 2004; 93(2-3): 109-13.
[http://dx.doi.org/10.1016/j.imlet.2004.03.014] [PMID: 15158605]
[27]
Arias-Alpizar G, Kong L, Vlieg RC, et al. Light-triggered switching of liposome surface charge directs delivery of membrane impermeable payloads in vivo. Nat Commun 2020; 11(1): 3638.
[http://dx.doi.org/10.1038/s41467-020-17360-9] [PMID: 32686667]
[28]
Hadjidemetriou M, Al-Ahmady Z, Kostarelos K. Time-evolution of in vivo protein corona onto blood-circulating PEGylated liposomal doxorubicin (DOXIL) nanoparticles. Nanoscale 2016; 8(13): 6948-57.
[http://dx.doi.org/10.1039/C5NR09158F PMID: 26961355]
[29]
Chen H, Tang L, Qin Y, et al. Lactoferrin-modified procationic liposomes as a novel drug carrier for brain delivery. Eur J Pharm Sci 2010; 40(2): 94-102.
[http://dx.doi.org/10.1016/j.ejps.2010.03.007] [PMID: 20298779]
[30]
Resina S, Prevot P, Thierry AR. Physico-chemical characteristics of lipoplexes influence cell uptake mechanisms and transfection efficacy. PLoS One 2009; 4(6)e6058
[http://dx.doi.org/10.1371/journal.pone.0006058] [PMID: 19557145]
[31]
Patel HM. Serum opsonins and liposomes: their interaction and opsonophagocytosis. Crit Rev Ther Drug Carrier Syst 1992; 9(1): 39-90.
[PMID: 1544174]
[32]
Prozeller D, Pereira J, Simon J, Mailänder V, Morsbach S, Landfester K. Prevention of dominant IgG adsorption on nanocarriers in IgG-enriched blood plasma by clusterin precoating. Adv Sci (Weinh) 2019; 6(10)1802199
[http://dx.doi.org/10.1002/advs.201802199] [PMID: 31131195]
[33]
Jiang Y, Lu Q, Wang Y, et al. Quantitating endosomal escape of a library of polymers for mRNA delivery. Nano Lett 2020; 20(2): 1117-23.
[http://dx.doi.org/10.1021/acs.nanolett.9b04426] [PMID: 32003222]
[34]
Hafez IM, Maurer N, Cullis PR. On the mechanism whereby cationic lipids promote intracellular delivery of polynucleic acids. Gene Ther 2001; 8(15): 1188-96.
[http://dx.doi.org/10.1038/sj.gt.3301506 PMID: 11509950]
[35]
Francia V, Yang K, Deville S, Reker-Smit C, Nelissen I, Salvati A. Corona composition can affect the mechanisms cells use to internalize nanoparticles. ACS Nano 2019; 13(10): 11107-21.
[http://dx.doi.org/10.1021/acsnano.9b03824] [PMID: 31525954]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy