nature.com

mRNA lipid nanoparticle formulation, characterization and evaluation

Abstract

mRNA-based therapies have emerged as a cutting-edge approach for diverse therapeutic applications. However, substantial barriers exist that hinder scientists from entering this research field, including the technical complexity and multiple potential workflows available for formulating and evaluating mRNA lipid nanoparticles (LNPs). Here we present an easy-to-follow and step-by-step guide for mRNA LNP formulation, characterization and in vitro and in vivo evaluation that could lower these barriers, facilitating entry for scientists in academia, industry and clinical settings into this research space. In this protocol, we detail steps for formulating representative mRNA LNPs (0.5 d) and characterizing key parameters (1–6 d) such as size, polydispersity index, zeta potential, mRNA concentration, mRNA encapsulation efficiency and stability. Then, we describe in vitro evaluations (3–6 d), such as protein expression, cell uptake and mechanism investigations (3–5 d), including endosomal escape, as well as in vivo delivery evaluation (2–3 d) encompassing intracellular and secreted protein expression levels, biodistribution and additional tolerability studies (1–2 weeks). Unlike some alternative protocols that may focus on discrete aspects of the workflow—such as formulation, characterization or evaluation—our protocol instead aims to integrate each of these aspects into a simplified, singular workflow applicable across multiple types of mRNA LNP formulations. In describing these procedures, we wish to disseminate one potential workflow for mRNA LNP production and evaluation, with the ultimate goal of furthering innovation, collaboration and the translational advancement of mRNA LNPs.

Key points

This protocol represents one possible approach for the formulation, characterization and evaluation of mRNA lipid nanoparticles.

It assesses key parameters such as size, polydispersity index and zeta potential, as well as mRNA concentration, encapsulation efficiency and stability. In vitro evaluations include protein expression, cell uptake and mechanism investigations, while assessment of intracellular and secreted protein expression levels, biodistribution and additional tolerability studies are carried out in vivo.

This is a preview of subscription content, access via your institution

Access options

Access through your institution

Change institution

Buy or subscribe

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

$29.99 / 30 days

cancel any time

Learn more

Subscribe to this journal

Receive 12 print issues and online access

$259.00 per year

only $21.58 per issue

Learn more

Buy this article

Purchase on SpringerLink

Instant access to full article PDF

Buy now

Prices may be subject to local taxes which are calculated during checkout

Additional access options:

Log in

Learn about institutional subscriptions

Read our FAQs

Contact customer support

Fig. 1: Overview of the protocol for formulating and evaluating mRNA LNPs.

Fig. 2: Considerations for the formulation and characterization of mRNA LNPs.

Fig. 3: Outline of in vitro evaluation methods for mRNA LNPs.

Fig. 4: Outline of mechanism investigation methods for mRNA LNPs.

Fig. 5: LNP formulation and characterization methods.

Fig. 6: In vitro evaluation methods of mRNA LNPs.

Fig. 7: Mechanism investigation methods.

Fig. 8: In vivo evaluation methods of mRNA LNPs.

Data availability

The datasets generated and/or analyzed during the current study are provided in Supplementary Information, source data for Figs. 5–8 and statistical source data for Supplementary Figs. 1, 5, 6, 7, 11 and 12. Any additional data required for research purposes are available from the corresponding authors upon request. Source data are provided with this paper.

References

Kon, E., Ad-El, N., Hazan-Halevy, I., Stotsky-Oterin, L. & Peer, D. Targeting cancer with mRNA-lipid nanoparticles: key considerations and future prospects. Nat. Rev. Clin. Oncol. 20, 739–754 (2023).

ArticleCASPubMedGoogle Scholar

Li, F. et al. mRNA lipid nanoparticle-mediated pyroptosis sensitizes immunologically cold tumors to checkpoint immunotherapy. Nat. Commun. 14, 4223 (2023).

ArticleCASPubMedPubMed CentralGoogle Scholar

Neill, B., Romero, A. R. & Fenton, O. S. Advances in nonviral mRNA delivery materials and their application as vaccines for melanoma therapy. ACS Appl. Bio Mater. 7, 4894–4913 (2024).

ArticleCASPubMedGoogle Scholar

Wang, C., Zhang, Y. & Dong, Y. Lipid nanoparticle–mRNA formulations for therapeutic applications. Acc. Chem. Res. 54, 4283–4293 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Jeong, M., Lee, Y., Park, J., Jung, H. & Lee, H. Lipid nanoparticles (LNPs) for in vivo RNA delivery and their breakthrough technology for future applications. Adv. Drug Deliv. Rev. 200, 114990 (2023).

ArticleCASPubMedGoogle Scholar

Sun, B. et al. Engineering nanoparticle toolkits for mRNA delivery. Adv. Drug Deliv. Rev. 200, 115042 (2023).

ArticleCASPubMedGoogle Scholar

Chabanovska, O., Galow, A.-M., David, R. & Lemcke, H. mRNA—a game changer in regenerative medicine, cell-based therapy and reprogramming strategies. Adv. Drug Deliv. Rev. 179, 114002 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Elia, U. et al. Design of SARS-CoV-2 hFc-conjugated receptor-binding domain mRNA vaccine delivered via lipid nanoparticles. ACS Nano 15, 9627–9637 (2021).

ArticleCASPubMedGoogle Scholar

McKay, P. F. et al. Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibody titers in mice. Nat. Commun. 11, 3523 (2020).

ArticleCASPubMedPubMed CentralGoogle Scholar

VanBlargan, L. A. et al. An mRNA vaccine protects mice against multiple tick-transmitted flavivirus infections. Cell Rep. 25, 3382–3392.e3 (2018).

ArticleCASPubMedPubMed CentralGoogle Scholar

Chaudhary, N., Weissman, D. & Whitehead, K. A. mRNA vaccines for infectious diseases: principles, delivery and clinical translation. Nat. Rev. Drug Discov. 20, 817–838 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Wang, X. et al. Preparation of selective organ-targeting (SORT) lipid nanoparticles (LNPs) using multiple technical methods for tissue-specific mRNA delivery. Nat. Protoc. 18, 265–291 (2023).

ArticleCASPubMedGoogle Scholar

Müller, J. A. et al. Kinetics of RNA–LNP delivery and protein expression. Eur. J. Pharm. Biopharm. 197, 114222 (2024).

ArticlePubMedGoogle Scholar

Aliakbarinodehi, N. et al. Interaction kinetics of individual mRNA-containing lipid nanoparticles with an endosomal membrane mimic: dependence on pH, protein corona formation, and lipoprotein depletion. ACS Nano 16, 20163–20173 (2022).

ArticleCASPubMedPubMed CentralGoogle Scholar

Amici, A., Pozzi, D., Marchini, C. & Caracciolo, G. The transformative potential of lipid nanoparticle-protein corona for next-generation vaccines and therapeutics. Mol. Pharm. 20, 5247–5253 (2023).

ArticleCASPubMedPubMed CentralGoogle Scholar

Chen, D., Ganesh, S., Wang, W. & Amiji, M. The role of surface chemistry in serum protein corona-mediated cellular delivery and gene silencing with lipid nanoparticles. Nanoscale 11, 8760–8775 (2019).

ArticleCASPubMedGoogle Scholar

Baden, L. R. et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 384, 403–416 (2021).

ArticleCASPubMedGoogle Scholar

Andresen, J. L. & Fenton, O. S. Nucleic acid delivery and nanoparticle design for COVID vaccines. MRS Bull. 46, 832–839 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Callaway, E. COVID vaccine excitement builds as Moderna reports third positive result. Nature 587, 337–338 (2020).

ArticleCASPubMedGoogle Scholar

Webb, C. et al. Current status and future perspectives on MRNA drug manufacturing. Mol. Pharm. 19, 1047–1058 (2022).

ArticleCASPubMedGoogle Scholar

Zuhorn, I. S. et al. Nonbilayer phase of lipoplex-membrane mixture determines endosomal escape of genetic cargo and transfection efficiency. Mol. Ther. 11, 801–810 (2005).

ArticleCASPubMedGoogle Scholar

Lu, J. J., Langer, R. & Chen, J. A novel mechanism is involved in cationic lipid-mediated functional siRNA delivery. Mol. Pharm. 6, 763–771 (2009).

ArticleCASPubMedPubMed CentralGoogle Scholar

Narasipura, E. A., VanKeulen-Miller, R., Ma, Y. & Fenton, O. S. Ongoing clinical trials of nonviral siRNA therapeutics. Bioconjug. Chem. 34, 1177–1197 (2023).

ArticleCASPubMedGoogle Scholar

Allen, T. M. & Cullis, P. R. Liposomal drug delivery systems: from concept to clinical applications. Adv. Drug Deliv. Rev. 65, 36–48 (2013).

ArticleCASPubMedGoogle Scholar

Fenton, O. S. et al. Customizable lipid nanoparticle materials for the delivery of siRNAs and mRNAs. Angew. Chem. Int. Ed. 57, 13582–13586 (2018).

ArticleCASGoogle Scholar

Mui, B. L. et al. Influence of polyethylene glycol lipid desorption rates on pharmacokinetics and pharmacodynamics of siRNA lipid nanoparticles. Mol. Ther. Nucleic Acids 2, e139 (2013).

ArticleCASPubMedPubMed CentralGoogle Scholar

Malburet, C. et al. Size and charge characterization of lipid nanoparticles for mRNA vaccines. Anal. Chem. 94, 4677–4685 (2022).

ArticleCASPubMedGoogle Scholar

Hassett, K. J. et al. Impact of lipid nanoparticle size on mRNA vaccine immunogenicity. J. Control. Release 335, 237–246 (2021).

ArticleCASPubMedGoogle Scholar

Cullis, P. R. & Hope, M. J. Lipid nanoparticle systems for enabling gene therapies. Mol. Ther. 25, 1467–1475 (2017).

ArticleCASPubMedPubMed CentralGoogle Scholar

Yanez Arteta, M. et al. Successful reprogramming of cellular protein production through mRNA delivered by functionalized lipid nanoparticles. Proc. Natl Acad. Sci. USA 115, E3351–E3360 (2018).

ArticlePubMedPubMed CentralGoogle Scholar

Guéguen, C. et al. Evaluating how cationic lipid affects mRNA–LNP physical properties and biodistribution. Eur. J. Pharm. Biopharm. 195, 114077 (2024).

ArticlePubMedGoogle Scholar

Carrasco, M. J. et al. Ionization and structural properties of mRNA lipid nanoparticles influence expression in intramuscular and intravascular administration. Commun. Biol. 4, 956 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Zhang, H. et al. Fluorinated lipid nanoparticles for enhancing mRNA delivery efficiency. ACS Nano 18, 7825–7836 (2024).

ArticleCASPubMedGoogle Scholar

Tenchov, R., Bird, R., Curtze, A. E. & Zhou, Q. Lipid nanoparticles—from liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano 15, 16982–17015 (2021).

ArticleCASPubMedGoogle Scholar

Zhang, L. et al. Effect of mRNA–LNP components of two globally-marketed COVID-19 vaccines on efficacy and stability. NPJ Vaccines 8, 156 (2023).

ArticleCASPubMedPubMed CentralGoogle Scholar

Rohner, E., Yang, R., Foo, K. S., Goedel, A. & Chien, K. R. Unlocking the promise of mRNA therapeutics. Nat. Biotechnol. 40, 1586–1600 (2022).

ArticleCASPubMedGoogle Scholar

Rui, Y. et al. High-throughput and high-content bioassay enables tuning of polyester nanoparticles for cellular uptake, endosomal escape, and systemic in vivo delivery of mRNA. Sci. Adv. 8, eabk2855 (2022).

ArticleCASPubMedPubMed CentralGoogle Scholar

Pei, D. & Buyanova, M. Overcoming endosomal entrapment in drug delivery. Bioconjug. Chem. 30, 273–283 (2019).

ArticleCASPubMedGoogle Scholar

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

ArticleCASPubMedGoogle Scholar

Herrera, M., Kim, J., Eygeris, Y., Jozic, A. & Sahay, G. Illuminating endosomal escape of polymorphic lipid nanoparticles that boost mRNA delivery. Biomater. Sci. 9, 4289–4300 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Boussif, O. et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc. Natl Acad. Sci. USA 92, 7297–7301 (1995).

ArticleCASPubMedPubMed CentralGoogle Scholar

Vermeulen, L. M. P., De Smedt, S. C., Remaut, K. & Braeckmans, K. The proton sponge hypothesis: fable or fact? Eur. J. Pharm. Biopharm. 129, 184–190 (2018).

ArticleCASPubMedGoogle Scholar

Behr, J.-P. The proton sponge: a trick to enter cells the viruses did not exploit. Int. J. Chem. 51, 34 (1997).

CASGoogle Scholar

Hu, Y. et al. Cytosolic delivery of membrane-impermeable molecules in dendritic cells using pH-responsive core-shell nanoparticles. Nano Lett. 7, 3056–3064 (2007).

ArticleCASPubMedGoogle Scholar

Dröse, S. & Altendorf, K. Bafilomycins and concanamycins as inhibitors of V-ATPases and P-ATPases. J. Exp. Biol. 200, 1–8 (1997).

ArticlePubMedGoogle Scholar

Zhang, W. et al. Lipid carriers for mRNA delivery. Acta Pharm. Sin. B 13, 4105–4126 (2023).

ArticleCASPubMedGoogle Scholar

Yin, H. et al. Non-viral vectors for gene-based therapy. Nat. Rev. Genet. 15, 541–555 (2014).

ArticleCASPubMedGoogle Scholar

Fenton, O. S. et al. Synthesis and biological evaluation of ionizable lipid materials for the in vivo delivery of messenger RNA to B lymphocytes. Adv. Mater. 29, 1606994 (2017).

ArticleGoogle Scholar

Polack, F. P. et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. N. Engl. J. Med. 383, 2603–2615 (2020).

ArticleCASPubMedGoogle Scholar

Riley, R. S. et al. Ionizable lipid nanoparticles for in utero mRNA delivery. Sci. Adv. 7, eaba1028 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Fenton, O. S. et al. Bioinspired alkenyl amino alcohol ionizable lipid materials for highly potent in vivo mRNA delivery. Adv. Mater. 28, 2939–2943 (2016).

ArticleCASPubMedPubMed CentralGoogle Scholar

Vogel, A. B. et al. BNT162b vaccines protect rhesus macaques from SARS-CoV-2. Nature 592, 283–289 (2021).

ArticleCASPubMedGoogle Scholar

Corbett, K. S. et al. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature 586, 567–571 (2020).

ArticleCASPubMedPubMed CentralGoogle Scholar

Fenton, O. S., Olafson, K. N., Pillai, P. S., Mitchell, M. J. & Langer, R. Advances in biomaterials for drug delivery. Adv. Mater. 30, 1705328 (2018).

ArticleGoogle Scholar

Hunter, D. G. & Frisken, B. J. Effect of extrusion pressure and lipid properties on the size and polydispersity of lipid vesicles. Biophys. J. 74, 2996–3002 (1998).

ArticleCASPubMedPubMed CentralGoogle Scholar

Martínez Rivas, C. J. et al. Nanoprecipitation process: from encapsulation to drug delivery. Int. J. Pharm. 532, 66–81 (2017).

ArticlePubMedGoogle Scholar

Fessi, H., Puisieux, F., Devissaguet, J. P., Ammoury, N. & Benita, S. Nanocapsule formation by interfacial polymer deposition following solvent displacement. Int. J. Pharm. 55, R1–R4 (1989).

ArticleCASGoogle Scholar

Bangham, A. D., De Gier, J. & Greville, G. D. Osmotic properties and water permeability of phospholipid liquid crystals. Chem. Phys. Lipids 1, 225–246 (1967).

ArticleCASGoogle Scholar

Umbarkar, M., Thakare, S., Surushe, T., Giri, A. & Chopade, V. Formulation and evaluation of liposome by thin film hydration method. J. Drug Deliv. Ther. 11, 72–76 (2021).

ArticleCASGoogle Scholar

Shepherd, S. J. et al. Scalable mRNA and siRNA lipid nanoparticle production using a parallelized microfluidic device. Nano Lett. 21, 5671–5680 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Thelen, J. L. et al. Morphological characterization of self-amplifying mRNA lipid nanoparticles. ACS Nano 18, 1464–1476 (2024).

ArticleCASPubMedGoogle Scholar

Eygeris, Y., Patel, S., Jozic, A. & Sahay, G. Deconvoluting lipid nanoparticle structure for messenger RNA delivery. Nano Lett. 20, 4543–4549 (2020).

ArticleCASPubMedGoogle Scholar

Cui, L. et al. Development of a high-throughput platform for screening lipid nanoparticles for mRNA delivery. Nanoscale 14, 1480–1491 (2022).

ArticleCASPubMedGoogle Scholar

Hammel, M. et al. Correlating the structure and gene silencing activity of oligonucleotide-loaded lipid nanoparticles using small-angle X-ray scattering. ACS Nano 17, 11454–11465 (2023).

ArticleCASPubMedPubMed CentralGoogle Scholar

Schwamberger, A. et al. Combining SAXS and DLS for simultaneous measurements and time-resolved monitoring of nanoparticle synthesis. Nucl. Instrum. Methods Phys. Res. Sect. B 343, 116–122 (2015).

ArticleCASGoogle Scholar

Bauer, P. S. et al. In-situ aerosol nanoparticle characterization by small angle X-ray scattering at ultra-low volume fraction. Nat. Commun. 10, 1122 (2019).

ArticleCASPubMedPubMed CentralGoogle Scholar

Rungta, R. L. et al. Lipid nanoparticle delivery of siRNA to silence neuronal gene expression in the brain. Mol. Ther. Nucleic Acids 2, e136 (2013).

ArticleCASPubMedPubMed CentralGoogle Scholar

Hsu, F.-F. et al. An efficient approach for SARS-CoV-2 monoclonal antibody production via modified mRNA-LNP immunization. Int. J. Pharm. 627, 122256 (2022).

ArticleCASPubMedPubMed CentralGoogle Scholar

Wang, H. et al. Targeted quantitation of CFTR protein expression in vivo using immunoprecipitation & parallel reaction monitoring tandem mass spectrometry. Transl. Med. Commun. 7, 9 (2022).

ArticleGoogle Scholar

Rafique, A. et al. Targeted lipid nanoparticle delivery of calcitriol to human monocyte-derived macrophages in vitro and in vivo: investigation of the anti-inflammatory effects of calcitriol. Int. J. Nanomed. 14, 2829–2846 (2019).

ArticleCASGoogle Scholar

Yang, T. et al. Efficient hepatic delivery and protein expression enabled by optimized mRNA and ionizable lipid nanoparticle. Bioact. Mater. 5, 1053–1061 (2020).

PubMedPubMed CentralGoogle Scholar

Tiwari, A. et al. Improving endothelial cell retention for single stage seeding of prosthetic grafts: use of polymer sequences of arginine-glycine-aspartate. Eur. J. Vasc. Endovasc. Surg. 25, 325–329 (2003).

ArticleCASPubMedGoogle Scholar

La Peyre, M., Casas, S. & La Peyre, J. Salinity effects on viability, metabolic activity and proliferation of three Perkinsus species. Dis. Aquat. Org. 71, 59–74 (2006).

ArticleGoogle Scholar

Han, X. et al. Validation of an LDH assay for assessing nanoparticle toxicity. Toxicology 287, 99–104 (2011).

ArticleCASPubMedGoogle Scholar

Parot, J. et al. Quality assessment of LNP–RNA therapeutics with orthogonal analytical techniques. J. Control. Release 367, 385–401 (2024).

ArticleCASPubMedGoogle Scholar

Xue, L. et al. High-throughput barcoding of nanoparticles identifies cationic, degradable lipid-like materials for mRNA delivery to the lungs in female preclinical models. Nat. Commun. 15, 1884 (2024).

ArticleCASPubMedPubMed CentralGoogle Scholar

Guimaraes, P. P. G. et al. Ionizable lipid nanoparticles encapsulating barcoded mRNA for accelerated in vivo delivery screening. J. Control. Release 316, 404–417 (2019).

ArticleCASPubMedPubMed CentralGoogle Scholar

Ci, L. et al. Biodistribution of lipid 5, mRNA, and its translated protein following intravenous administration of mRNA-encapsulated lipid nanoparticles in rats. Drug Metab. Dispos. 51, 813–823 (2023).

ArticleCASPubMedGoogle Scholar

Hyldbakk, A., Hansen, T., Hak, S. & Borgos, S. E. F. Polyethylene glycol (PEG) as a broad applicability marker for LC-MS/MS-based biodistribution analysis of nanomedicines. J. Control. Release 366, 611–620 (2024).

ArticleCASPubMedGoogle Scholar

Chen, S. et al. Influence of particle size on the in vivo potency of lipid nanoparticle formulations of siRNA. J. Control. Release 235, 236–244 (2016).

ArticleCASPubMedGoogle Scholar

Goutayer, M. et al. Tumor targeting of functionalized lipid nanoparticles: assessment by in vivo fluorescence imaging. Eur. J. Pharm. Biopharm. 75, 137–147 (2010).

ArticleCASPubMedGoogle Scholar

Ma, Y. & Fenton, O. S. Tannic acid lipid nanoparticles can deliver messenger RNA payloads and improve their endosomal escape. Adv. Ther. 6, 2200305 (2023).

ArticleCASGoogle Scholar

Ma, Y. & Fenton, O. S. A unified strategy to improve lipid nanoparticle mediated mRNA delivery using adenosine triphosphate. J. Am. Chem. Soc. 145, 19800–19811 (2023).

ArticleCASPubMedGoogle Scholar

Ma, Y. & Fenton, O. S. An efficacy and mechanism driven study on the impact of hypoxia on lipid nanoparticle mediated mRNA delivery. J. Am. Chem. Soc. 145, 11375–11386 (2023).

ArticleCASPubMedGoogle Scholar

Ma, Y., Tiwade, P. B., VanKeulen-Miller, R., Narasipura, E. A. & Fenton, O. S. Polyphenolic nanoparticle platforms (PARCELs) for in vitro and in vivo mRNA delivery. Nano Lett. 24, 6092–6101 (2024).

ArticleCASPubMedPubMed CentralGoogle Scholar

Erickson, H. P. Size and shape of protein molecules at the nanometer level determined by sedimentation, gel filtration, and electron microscopy. Biol. Proced. Online 11, 32–51 (2009).

ArticleCASPubMedPubMed CentralGoogle Scholar

Li, S. et al. Payload distribution and capacity of mRNA lipid nanoparticles. Nat. Commun. 13, 5561 (2022).

ArticleCASPubMedPubMed CentralGoogle Scholar

Suzuki, Y. & Ishihara, H. Difference in the lipid nanoparticle technology employed in three approved siRNA (Patisiran) and mRNA (COVID-19 vaccine) drugs. Drug Metab. Pharmacokinet. 41, 100424 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Gujrati, M. et al. Multifunctional cationic lipid-based nanoparticles facilitate endosomal escape and reduction-triggered cytosolic siRNA release. Mol. Pharm. 11, 2734–2744 (2014).

ArticleCASPubMedPubMed CentralGoogle Scholar

Douka, S. et al. Lipid nanoparticle-mediated messenger RNA delivery for ex vivo engineering of natural killer cells. J. Control. Release 361, 455–469 (2023).

ArticleCASPubMedGoogle Scholar

Schoenmaker, L. et al. mRNA-lipid nanoparticle COVID-19 vaccines: structure and stability. Int. J. Pharm. 601, 120586 (2021).

ArticleCASPubMedPubMed CentralGoogle Scholar

Skoglund, S. et al. Difficulties and flaws in performing accurate determinations of zeta potentials of metal nanoparticles in complex solutions-Four case studies. PLoS ONE 12, e0181735 (2017).

ArticlePubMedPubMed CentralGoogle Scholar

Wang, L. et al. Effects of ionic strength and temperature on the aggregation and deposition of multi-walled carbon nanotubes. J. Environ. Sci. 51, 248–255 (2017).

ArticleCASGoogle Scholar

Berg, J. M., Romoser, A., Banerjee, N., Zebda, R. & Sayes, C. M. The relationship between pH and zeta potential of ~30 nm metal oxide nanoparticle suspensions relevant to in vitro toxicological evaluations. Nanotoxicology 3, 276–283 (2009).

ArticleCASGoogle Scholar

Yap, S. L. et al. Cell interactions with lipid nanoparticles possessing different internal nanostructures: Liposomes, bicontinuous cubosomes, hexosomes, and discontinuous micellar cubosomes. J. Colloid Interface Sci. 656, 409–423 (2024).

ArticleCASPubMedGoogle Scholar

Escalona-Rayo, O. et al. In vitro and in vivo evaluation of clinically-approved ionizable cationic lipids shows divergent results between mRNA transfection and vaccine efficacy. Biomed. Pharmacother. 165, 115065 (2023).

ArticleCASPubMedGoogle Scholar

Zhang, H., Leal, J., Soto, M. R., Smyth, H. D. C. & Ghosh, D. Aerosolizable lipid nanoparticles for pulmonary delivery of mRNA through design of experiments. Pharmaceutics 12, 1042 (2020).

ArticleCASPubMedPubMed CentralGoogle Scholar

French, A. P., Mills, S., Swarup, R., Bennett, M. J. & Pridmore, T. P. Colocalization of fluorescent markers in confocal microscope images of plant cells. Nat. Protoc. 3, 619–628 (2008).

ArticleCASPubMedGoogle Scholar

Wong, A. S. M. et al. Self-assembling dual component nanoparticles with endosomal escape capability. Soft Matter 11, 2993–3002 (2015).

ArticleCASPubMedGoogle Scholar

Smith, S. A., Selby, L. I., Johnston, A. P. R. & Such, G. K. The endosomal escape of nanoparticles: toward more efficient cellular delivery. Bioconjug. Chem. 30, 263–272 (2019).

ArticleCASPubMedGoogle Scholar

Chen, J. et al. Metal-phenolic coatings as a platform to trigger endosomal escape of nanoparticles. ACS Nano 13, 11653–11664 (2019).

ArticleCASPubMedGoogle Scholar

Burkholder, T., Foltz, C., Karlsson, E., Linton, C. G. & Smith, J. M. Health evaluation of experimental laboratory mice. Curr. Protoc. Mouse Biol. 2, 145–165 (2012).

ArticlePubMedPubMed CentralGoogle Scholar

Korzun, T. et al. Lipid nanoparticles elicit reactogenicity and sickness behavior in mice via toll-like receptor 4 and myeloid differentiation protein 88 axis. ACS Nano 18, 24842–24859 (2024).

ArticleCASPubMedGoogle Scholar

Parhiz, H. et al. Added to pre-existing inflammation, mRNA-lipid nanoparticles induce inflammation exacerbation (IE). J. Control. Release 344, 50–61 (2022).

ArticleCASPubMedGoogle Scholar

Miller, L. R. et al. Considering sex as a biological variable in preclinical research. FASEB J. 31, 29–34 (2017).

ArticleCASPubMedGoogle Scholar

Binici, B., Rattray, Z., Schroeder, A. & Perrie, Y. The role of biological sex in pre-clinical (Mouse) mRNA vaccine studies. Vaccines 12, 282 (2024).

ArticleCASPubMedPubMed CentralGoogle Scholar

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).

ArticleCASPubMedGoogle Scholar

Mrksich, K. et al. Influence of ionizable lipid tail length on lipid nanoparticle delivery of mRNA of varying length. J. Biomed. Mater. Res. A 112, 1494–1505 (2024).

ArticleCASPubMedGoogle Scholar

Chen, D. et al. Rapid discovery of potent siRNA-containing lipid nanoparticles enabled by controlled microfluidic formulation. J. Am. Chem. Soc. 134, 6948–6951 (2012).

ArticleCASPubMedGoogle Scholar

Wquimby, F. & Hluong, R. in The Mouse in Biomedical Research (eds Fox, J. G. et al.) 171–216 (Elsevier, 2007).

Wei, Q. & Dong, Z. Mouse model of ischemic acute kidney injury: technical notes and tricks. Am. J. Physiol. Ren. Physiol. 303, F1487–F1494 (2012).

ArticleCASGoogle Scholar

Download references

Acknowledgements

This work was supported by an NIH National Institute of Biomedical Imaging and Bioengineering award (1R21EB034942-01). This work was also supported by the NC Translational and Clinical Sciences (NC TraCS) Institute, which is supported by the NIH National Center for Advancing Translational Sciences (NCATS) award 1K12TR004416-01. This work was also supported by the National Institute of General Medicine Sciences (NIGMS) award 1R35GM157060-01. We also thank C. Pham, M. Evangelista and R. Sellers in the Pathology Services Core (PSC) for expert technical assistance with Histopathology and Digital Pathology. The PSC is supported in part by an NCI Center Core Support Grant (5P30CA016080-42). All animal studies were approved by the UNC Institutional Animal Care and Use Committee, were consistent with local, state and federal regulations as applicable and were supported within the UNC Lineberger ASC at the University of North Carolina at Chapel Hill which is supported in part by an NCI Center Core Support Grant (CA16086) to the UNC Lineberger Comprehensive Cancer Center. Flow cytometry was performed at UNC Flow Cytometry Core Facility (RRID: SCR_019170). Microscopy was performed at the UNC Neuroscience Microscopy Core (RRID: SCR_019060), supported, in part, by funding from the NIH-NINDS Neuroscience Center Support Grant P30 NS045892 and the NIH-NICHD Intellectual and Developmental Disabilities Research Center Support Grant P50 HD103573. Research reported in this publication was supported in part by the North Carolina Biotech Center institutional support grant 2017-IDG-1025 and by the National Institutes of Health 1UM2AI30836-01. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA

Yutian Ma & Owen S. Fenton

Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA

Rachel VanKeulen-Miller

Authors

Yutian Ma

View author publications

You can also search for this author in PubMedGoogle Scholar

2. Rachel VanKeulen-Miller

View author publications

You can also search for this author in PubMedGoogle Scholar

3. Owen S. Fenton

View author publications

You can also search for this author in PubMedGoogle Scholar

Contributions

Y.M. designed and conducted experiments, analyzed the data and contributed to writing and editing the manuscript. R.V.-M. contributed to writing and editing the manuscript. O.S.F. supervised all the experiments, wrote and edited the manuscript and managed the project. All authors contributed to the development of the protocol and improved the manuscript.

Corresponding author

Correspondence to Owen S. Fenton.

Ethics declarations

Competing interests

The University of North Carolina at Chapel Hill has filed some patents on data related to this work, with the authors named as inventors.

Peer review

Peer review information

Nature Protocols thanks Yizhou Dong and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Key references

Ma, Y. & Fenton, O. S. J. Am. Chem. Soc. 145, 19800–19811 (2023): https://doi.org/10.1021/jacs.3c05574

Ma, Y. & Fenton, O. S. J. Am. Chem. Soc. 145, 11375–11386 (2023): https://doi.org/10.1021/jacs.3c02584

Ma, Y. & Fenton, O. S. Adv. Therap. 6, 2200305 (2023): https://doi.org/10.1002/adtp.202200305

Supplementary information

Supplementary Information

Supplementary Figs. 1–13 and Tables 1 and 2.

Reporting Summary

Supplementary Data 1

Source data for Supplementary Figs. 1, 5, 6a–c, 7b, 11 and 12.

Source data

Source Data Fig. 5

Statistical source data for Fig. 5.

Source Data Fig. 6

Statistical source data for Fig. 6.

Source Data Fig. 7

Statistical source data for Fig. 7.

Source Data Fig. 8

Statistical source data for Fig. 8.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, Y., VanKeulen-Miller, R. & Fenton, O.S. mRNA lipid nanoparticle formulation, characterization and evaluation. Nat Protoc (2025). https://doi.org/10.1038/s41596-024-01134-4

Download citation

Received:10 May 2024

Accepted:12 December 2024

Published:11 March 2025

DOI:https://doi.org/10.1038/s41596-024-01134-4

Share this article

Anyone you share the following link with will be able to read this content:

Get shareable link

Sorry, a shareable link is not currently available for this article.

Copy to clipboard

Provided by the Springer Nature SharedIt content-sharing initiative

Read full news in source page