mRNA-based COVID-19 Vaccines using Lipid Nanoparticles (LNPs): History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , , , ,

Lipid nanoparticles (LNPs) have recently emerged as one of the most advanced technologies for the highly efficient in vivo delivery of exogenous mRNA, particularly for COVID-19 vaccine delivery. LNPs comprise four different lipids: ionizable lipids, helper or neutral lipids, cholesterol, and lipids attached to polyethylene glycol (PEG). Since its first isolation in 1961, mRNA (that encodes the protein of interest) research has taken several paths, which made us understand its diversified functions and modification-mediated potential for therapeutic applications. As a result of the COVID-19 pandemic, nucleic acid therapeutics (NATs), particularly mRNA vaccines, potentials have been enabled for emerging infectious diseases. The translation of host genetic information (DNA) into proteins by ribosomes in the cytoplasm is mediated by mRNA.

  • vaccine
  • mRNA
  • COVID-19
  • lipid nanoparticles

1. Use of mRNA as Prophylactics and Therapeutics

Despite the usage of DNA vaccines, RNA-based vaccines have grown in popularity in the 21st century. Interestingly, mRNA vaccine technology purely depends on the genetic code of the virus; since the SARS-CoV-2 genetic sequence was published [1], virally generated self-amplifying RNA and non-replicating mRNA are being used abundantly for vaccine development. Indeed, during the COVID-19 pandemic, mRNA vaccines have become the only vaccine source available to combat the rapid spread of disease. While multiple pharmaceutical companies have investigated several vaccine technologies against COVID-19, the FDA has approved two vaccines (first for emergency use and then for full approval), Pfizer/BioNTech’s and Moderna’s Spikevax (Table 1) [2]. Both use the complete sequence of spike and receptor-binding domains of SARS-CoV-2 as target antigens. However, mRNA-based preclinical studies have been reported since 1990. Its poor stability as naked mRNA and tremendous capacity to induce an innate immune response (particularly double-stranded RNA) has halted the progress of RNA-based therapeutics. The emergence of advanced nanotechnologies for the protection and targeted delivery of water-soluble molecules have shined a light on developing the LNP-mRNA vaccines. Nucleic acid delivery to the cytosol, without hampering the cell wall integrity, and degradation by nucleases is a rate limiting step for targeted delivery. Despite mechanical and viral particle-mediated targeted delivery, chemical based targeted delivery via LNPs have been the ease of the process and have been less toxic, with less immunogenicity. During the start of the decade, several clinical trials have been initiated on mRNA therapeutics, which cover a range of different types of diseases, such as cancer, infectious diseases, protein replacement therapy, and others. Correspondingly, promising results have been shown in terms of disease alleviation, reduction of pathogenicity, and others. Interestingly, several diseases or disorders are targeted by using mRNA therapeutics. For the proof-of-concept studies, in vitro transcription (IVT)-produced mRNA is more suitable (Figure 1). To characterize the efficiency of mRNA in vitro, several transfection reagents are available to facilitate mRNA translation and targeted protein production, although they are not as efficient as targeted delivery systems. The Figure 1 shows the most successful mRNA vaccines. As explained earlier, a suitable (perfect mix) vehicle is needed for the mRNA vaccine delivery. LNPs have become the one of the most characterized mRNA delivery vehicles.
Figure 1. Immune response by mRNA vaccines. LNPs are prepared by encapsulating mRNA, which encodes the viral protein of interest. Upon injection of vaccines, muscular cells take up the LNPs following the release of mRNA into the cytosol and translation of target protein with the help of host machinery. In parallel, the danger associated signals produced by the LNPs recruit the innate immune cells, including neutrophils, monocytes, macrophages, dendritic cells, and others. The antigen-presenting cells (APC) process and present the antigen to the T cells, which further polarizes into effector T cells and helps in B cell-mediated responses. The cytotoxic T cells produced upon activation kill the infected cells, and antibodies (produced by B cells or plasma cells) neutralize the virus.

2. LNP Role in mRNA Delivery

Using lipid-encapsulated forms of sequence-optimized mRNA vaccines has shown strong immunity (both in preclinical and clinical) against infectious disease targets like the flu virus, Zika virus, rabies virus, and others, especially in the last few years [3]. Of note, the LNPs act as potential vaccine adjuvants [2], which helps in shaping antigen-specific immunity [4]. Despite several components of the LNP, ionizable lipids have a prominent role, including forming LNPs and the targeted delivery of mRNA into the cytosol. The ionizable lipid imparts the neutral charge to the LNPs at physiological pH and ionizes at lower pH in the endosome/lysosome, followed by the delivery of intact mRNA to the cytoplasm, where it can then be translated into the encoded protein. Despite mRNA, LNPs also induce the innate immune signaling pathways that aid in the adaptive immune responses. Similar to other potential adjuvants, LNPs (based on the composition) also induce the activation of several signaling pathways, including nuclear factor kappa B, interferons, inflammasome activation, and other [5].
In the future, it will be essential to compare and understand the immune pathways activated by different mRNA vaccine platforms, improve current approaches based on these mechanisms, and start new clinical trials against more disease targets.

3. Salient Features of LNP-Based Therapeutics

  • Naked mRNA is unsuitable for therapeutic purposes, as it is rapidly degraded by extracellular RNases. Several nanotechnology platforms have been set and optimized for mRNA-targeted delivery.
  • As mRNA is thermolabile, LNPs would enhance its stability and half-life at room temperature and be useful for avoiding vaccine cold chain.
  • Modifications of the type of delivery system (carrier molecules) rule out the organ-specific mRNA delivery (for lung, spleen, liver, etc.) and its in vivo half-life.
  • Composition of LNP could decide the type of immune response induced.
  • LNPs act as adjuvants systems for mRNA vaccines.
  • The stable LNPs make the lower dose of mRNA work.
  • Composition of LNPs also decides the number of booster doses required if it is admixed with a suitable adjuvant.
  • Moreover, for chronic treatments, multiple administration via different routes of administration is possible.

4. Mechanism of mRNA Vaccines

Even though the needle-free route of administration is long awaited [6][7][8], mRNA encapsulated by LNPs is administered mostly by the intramuscular route, and several study results support this route of administration [9][10][11]. Upon injection, local innate immune cells are recruited to the site of injection, followed by the uptake of LNPs by innate immune cells [12]. Once the LNPs reach the cytosol, lower pH enables the LNPs to release mRNA, followed by the translation of targeted antigens (Figure 1). The antigens are presented to the T helper cells (CD4+ T cells) and release several cytokines and chemokines, which further help in the shaping the antigen-specific immune responses, including B cell differentiation and plasma cell development [13]. The two successful vaccines (Pfizer/BioNTech and Moderna’s Spikevax) have confirmed this mechanism in both preclinical and clinical studies (Figure 2) [14].
Table 1. FDA approved COVID-19 mRNA vaccines [15][16].
Figure 2. mRNA vaccine development. The figure illustrates the sequential steps involved in the mRNA vaccine development, from design to preclinical studies. Prior to going on to the next step, quality evaluation is required at each step.

This entry is adapted from the peer-reviewed paper 10.3390/vaccines11030658

References

  1. Oude Munnink, B.B.; Nieuwenhuijse, D.F.; Stein, M.; O’Toole, Á.; Haverkate, M.; Mollers, M.; Kamga, S.K.; Schapendonk, C.; Pronk, M.; Lexmond, P.; et al. The Dutch-Covid-19 response, t., Rapid SARS-CoV-2 whole-genome sequencing and analysis for informed public health decision-making in the Netherlands. Nat. Med. 2020, 26, 1405–1410.
  2. Verbeke, R.; Hogan, M.J.; Loré, K.; Pardi, N. Innate immune mechanisms of mRNA vaccines. Immunity 2022, 55, 1993–2005.
  3. Arevalo, C.P.; Bolton, M.J.; Le Sage, V.; Ye, N.; Furey, C.; Muramatsu, H.; Alameh, M.-G.; Pardi, N.; Drapeau, E.M.; Parkhouse, K.; et al. A multivalent nucleoside-modified mRNA vaccine against all known influenza virus subtypes. Science 2022, 378, 899–904.
  4. Bonam, S.R.; Partidos, C.D.; Halmuthur, S.K.M.; Muller, S. An Overview of Novel Adjuvants Designed for Improving Vaccine Efficacy. Trends Pharmacol. Sci. 2017, 38, 771–793.
  5. Riley, R.S.; Kashyap, M.V.; Billingsley, M.M.; White, B.; Alameh, M.-G.; Bose, S.K.; Zoltick, P.W.; Li, H.; Zhang, R.; Cheng, A.Y.; et al. Ionizable lipid nanoparticles for in utero mRNA delivery. Sci. Adv. 2021, 7, eaba1028.
  6. Vaca, G.B.; Meyer, M.; Cadete, A.; Hsiao, C.J.; Golding, A.; Jeon, A.; Jacquinet, E.; Azcue, E.; Guan, C.M.; Sanchez-Felix, X.; et al. Intranasal mRNA-LNP vaccination protects hamsters from SARS-CoV-2 infection. bioRxiv 2023, 11, 523616.
  7. Jansen, E.M.; Frijlink, H.W.; Hinrichs, W.L.J.; Ruigrok, M.J.R. Are inhaled mRNA vaccines safe and effective? A review of preclinical studies. Expert Opin. Drug Deliv. 2022, 19, 1471–1485.
  8. Phua, K.K.L.; Leong, K.W.; Nair, S.K. Transfection efficiency and transgene expression kinetics of mRNA delivered in naked and nanoparticle format. J. Control. Release 2013, 166, 227–233.
  9. Hassett, K.J.; Benenato, K.E.; Jacquinet, E.; Lee, A.; Woods, A.; Yuzhakov, O.; Himansu, S.; Deterling, J.; Geilich, B.M.; Ketova, T.; et al. Optimization of Lipid Nanoparticles for Intramuscular Administration of mRNA Vaccines. Mol. Ther. Nucleic Acids 2019, 15, 1–11.
  10. Ols, S.; Yang, L.; Thompson, E.A.; Pushparaj, P.; Tran, K.; Liang, F.; Lin, A.; Eriksson, B.; Karlsson Hedestam, G.B.; Wyatt, R.T.; et al. Route of Vaccine Administration Alters Antigen Trafficking but Not Innate or Adaptive Immunity. Cell Rep. 2020, 30, 3964–3971.e7.
  11. Davies, N.; Hovdal, D.; Edmunds, N.; Nordberg, P.; Dahlén, A.; Dabkowska, A.; Arteta, M.Y.; Radulescu, A.; Kjellman, T.; Höijer, A.; et al. Functionalized lipid nanoparticles for subcutaneous administration of mRNA to achieve systemic exposures of a therapeutic protein. Mol. Ther. —Nucleic Acids 2021, 24, 369–384.
  12. Alameh, M.-G.; Tombácz, I.; Bettini, E.; Lederer, K.; Ndeupen, S.; Sittplangkoon, C.; Wilmore, J.R.; Gaudette, B.T.; Soliman, O.Y.; Pine, M.; et al. Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses. Immunity 2021, 54, 2877–2892.e7.
  13. Chen, J.; Chen, J.; Xu, Q. Current Developments and Challenges of mRNA Vaccines. Annu. Rev. Biomed. Eng. 2022, 24, 85–109.
  14. Barbier, A.J.; Jiang, A.Y.; Zhang, P.; Wooster, R.; Anderson, D.G. The clinical progress of mRNA vaccines and immunotherapies. Nat. Biotechnol. 2022, 40, 840–854.
  15. Naveed, Z.; Li, J.; Wilton, J.; Spencer, M.; Naus, M.; Velásquez García, H.A.; Kwong, J.C.; Rose, C.; Otterstatter, M.; Janjua, N.Z. Comparative Risk of Myocarditis/Pericarditis Following Second Doses of BNT162b2 and mRNA-1273 Coronavirus Vaccines. J. Am. Coll. Cardiol. 2022, 80, 1900–1908.
  16. Wagenhäuser, I.; Reusch, J.; Gabel, A.; Krone, L.B.; Kurzai, O.; Petri, N.; Krone, M. Bivalent BNT162b2mRNA original/Omicron BA.4-5 booster vaccination: Adverse reactions and inability to work compared to the monovalent COVID-19 booster. medRxiv 2022, 2022, 22281982.
More
This entry is offline, you can click here to edit this entry!
Video Production Service