top of page

Challenge of Delivering mRNA and the Delivery via LNP 

Lipid nanoparticles have made notable strides in clinical applications, particularly in delivering mRNA. Notably, lipid nanoparticle-mRNA vaccines have been deployed in clinical settings to combat coronavirus disease 2019 (COVID-19), representing a significant milestone in mRNA therapeutics. 

  

The successfully design of the lipid nanoparticles ("LNPs") for mRNA delivery has overcome the physiological barriers - extracellular and intracellular ones (Ref.1,2)

  • mRNA needs to be protected from nuclease degradation in physiological fluids (Ref.1,2); 

  • The formulation should evade the interception by the MPS and clearance by renal glomerular filtration post systemic administration (Ref.1,2); 

  • Lipid nanoparticle–mRNA systems need to reach target tissues, followed by internalization by target cells (Ref. 1,2); 

  • mRNA molecules must escape endosomes to reach the cytoplasm, where translation occurs (Ref.1,2.) 

  

Lipid nanoparticle-mRNA formulations, produced through rapid mixing techniques, demonstrate a stable nanostructure (Ref. 3,4,5). Within this framework, mRNA molecules can be encapsulated within the core interior via electrostatic interactions with the lipids (Ref. 3,5). This structural attribute shields mRNA molecules from degradation by nucleases and enhances nanoparticle stability in physiological fluids (Ref. 3,6). The incorporation of PEG-lipids additionally reduces recognition by the mononuclear phagocyte system (MPS) and clearance via renal filtration (Ref. 3,7). Furthermore, targeted biodistribution of lipid nanoparticle-mRNA formulations can be enhanced through additional modification and optimization of the nanoparticle structure (Ref. 8,9,10,11,12,13,14,15). For instance, nanoparticles can be coated with antibodies (Ref.12) to facilitate the delivery of mRNA molecules into inflammatory leukocytes and epidermal growth factor receptor (EGFR)-positive tumor cells. This approach holds promise for treating inflammatory bowel disease (Ref.9) and cancer (Ref.13), respectively. Organ selectivity can also be attained by adjusting the ratios of lipid components. For instance, this can involve designing spleen-targeted mRNA vaccines (Ref. 8,9) or lung-targeted genome editing delivery systems (Ref. 11,14). 

GetImage.jpeg
GetImage (2).png

Upon reaching target cells, lipid nanoparticles can be internalized through various mechanisms, including macropinocytosis, clathrin-mediated endocytosis, and caveolae-mediated endocytosis (Ref. 10,3). The specific endocytic pathway employed depends on both the properties of the nanoparticle and the type of cell (Ref.16,17,18). Once internalized, lipid nanoparticles are typically sequestered within endosomal compartments (Ref. 19,20,21), with only a fraction escaping into the cytoplasm (Ref. 19,20,21). This process, known as endosomal escape, is critical for effective mRNA delivery. While the precise mechanism is not fully understood, positively charged lipids may facilitate electrostatic interaction and fusion with negatively charged endosomal membranes, leading to mRNA release into the cytoplasm (Ref. 1,2,14,3,16). Optimization of ionizable lipid pKa values (Ref. 22,23,24,25,26,16,27) and the properties of lipid tails can enhance endosomal escape (Ref. 28,29,11,141). For instance, lipids with branched tails may exhibit superior endosomal escape compared to those with linear tails due to stronger protonation at endosomal pH29. Additionally, adjusting the type and ratio of lipids, such as DSPC and DOPE, can further improve endosomal escape efficiency (Ref. 30,31,32,33,18,34). 

  

  

  1. Hajj, K. A. & Whitehead, K. A. Tools for translation: non-viral materials for therapeutic mRNA delivery. Nat. Rev. Mater. 2, 17056 (2017).  

  2. Kowalski, P. S., Rudra, A., Miao, L. & Anderson, D. G. Delivering the messenger: advances in technologies for therapeutic mRNA delivery. Mol. Ther. 27, 710–728 (2019).  

  3. Kim, J., Eygeris, Y., Gupta, M. & Sahay, G. Self-assembled mRNA vaccines. Adv. Drug Deliv. Rev. 170, 83–112 (2021).  

  4. Karnik, R. et al. Microfluidic platform for controlled synthesis of polymeric nanoparticles. Nano Lett. 8, 2906–2912 (2008).  

  5. Leung, A. K., Tam, Y. Y. C., Chen, S., Hafez, I. M. & Cullis, P. R. Microfluidic mixing: a general method for encapsulating macromolecules in lipid nanoparticle systems. J. Phys. Chem. B 119, 8698–8706 (2015). 

  6. Blakney, A. K., McKay, P. F., Yus, B. I., Aldon, Y. & Shattock, R. J. Inside out: optimization of lipid nanoparticle formulations for exterior complexation and in vivo delivery of saRNA. Gene Ther. 26, 363–372 (2019). 

  7. Knop, K., Hoogenboom, R., Fischer, D. & Schubert, U. S. Poly(ethylene glycol) in drug delivery: pros and cons as well as potential alternatives. Angew. Chem. Int. Ed. 49, 6288–6308 (2010). 

  8. Kranz, L. M. et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 534, 396–401 (2016). 

  9. Krienke, C. et al. A noninflammatory mRNA vaccine for treatment of experimental autoimmune encephalomyelitis. Science 371, 145–153 (2021) 

  10. Lee, S. M. et al. A systematic study of unsaturation in lipid nanoparticles leads to improved mRNA transfection in vivo. Angew. Chem. Int. Ed. 60, 5848–5853 (2021). 

  11. Veiga, N. et al. Cell specific delivery of modified mRNA expressing therapeutic proteins to leukocytes. Nat. Commun. 9, 4493 (2018). 

  12. Kedmi, R. et al. A modular platform for targeted RNAi therapeutics. Nat. Nanotechnol. 13, 214–219 (2018). 

  13. Rosenblum, D. et al. CRISPR-Cas9 genome editing using targeted lipid nanoparticles for cancer therapy. Sci. Adv. 6, eabc9450 (2020). 

  14. Cheng, Q. et al. Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR–Cas gene editing. Nat. Nanotechnol. 15, 313–320 (2020). 

  15. Kim, M. et al. Engineered ionizable lipid nanoparticles for targeted delivery of RNA therapeutics into different types of cells in the liver. Sci. Adv. 7, eabf4398 (2021). 

  16. Miao, L. et al. Synergistic lipid compositions for albumin receptor mediated delivery of mRNA to the liver. Nat. Commun. 11, 2424 (2020). 

  17. Zhang, X. et al. Functionalized lipid-like nanoparticles for in vivo mRNA delivery and base editing. Sci. Adv. 6, eabc2315 (2020). 

  18. Hou, X. et al. Vitamin lipid nanoparticles enable adoptive macrophage transfer for the treatment of multidrug-resistant bacterial sepsis. Nat. Nanotechnol. 15, 41–46 (2020). 

  19. Sahay, G. et al. Efficiency of siRNA delivery by lipid nanoparticles is limited by endocytic recycling. Nat. Biotechnol. 31, 653–658 (2013). 

  20. Gilleron, J. et al. Image-based analysis of lipid nanoparticle–mediated siRNA delivery, intracellular trafficking and endosomal escape. Nat. Biotechnol. 31, 638–646 (2013). 

  21. Wittrup, A. et al. Visualizing lipid-formulated siRNA release from endosomes and target gene knockdown. Nat. Biotechnol. 33, 870–876 (2015). 

  22. Jayaraman, M. et al. Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Angew. Chem. 124, 8657–8661 (2012). 

  23. Maier, M. A. et al. Biodegradable lipids enabling rapidly eliminated lipid nanoparticles for systemic delivery of RNAi therapeutics. Mol. Ther. 21, 1570–1578 (2013). 

  24. Sabnis, S. et al. A novel amino lipid series for mRNA delivery: improved endosomal escape and sustained pharmacology and safety in non-human primates. Mol. Ther. 26, 1509–1519 (2018). 

  25. Hassett, K. J. et al. Optimization of lipid nanoparticles for intramuscular administration of mRNA vaccines. Mol. Ther. Nucleic Acids 15, 1–11 (2019). 

  26. Whitehead, K. A. et al. Degradable lipid nanoparticles with predictable in vivo siRNA delivery activity. Nat. Commun. 5, 4277 (2014). 

  27. Alabi, C. A. et al. Multiparametric approach for the evaluation of lipid nanoparticles for siRNA delivery. Proc. Natl Acad. Sci. USA 110, 12881–12886 (2013). 

  28. Heyes, J., Palmer, L., Bremner, K. & MacLachlan, I. Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids. J. Control. Release 107, 276–287 (2005). 

  29. Hajj, K. A. et al. Branched-tail lipid nanoparticles potently deliver mRNA in vivo due to enhanced ionization at endosomal pH. Small 15, 1805097 (2019). 

  30. Kauffman, K. J. et al. Optimization of lipid nanoparticle formulations for mRNA delivery in vivo with fractional factorial and definitive screening designs. Nano Lett. 15, 7300–7306 (2015). 

  31. Li, B. et al. An orthogonal array optimization of lipid-like nanoparticles for mRNA delivery in vivo. Nano Lett. 15, 8099–8107 (2015). 

  32. Kim, J., Jozic, A. & Sahay, G. Naturally derived membrane lipids impact nanoparticle-based messenger RNA delivery. Cell. Mol. Bioeng. 13, 463–474 (2020). 

  33. Patel, S. et al. Naturally-occurring cholesterol analogues in lipid nanoparticles induce polymorphic shape and enhance intracellular delivery of mRNA. Nat. Commun. 11, 983 (2020). 

  34. Cheng, Q. et al. Dendrimer-based lipid nanoparticles deliver therapeutic

bottom of page