Here, a protocol for formulating lipid nanoparticles (LNPs) that encapsulate mRNA encoding firefly luciferase is presented. These LNPs were tested for their potency in vitro in HepG2 cells and in vivo in C57BL/6 mice.
Lipid nanoparticles (LNPs) have attracted widespread attention recently with the successful development of the COVID-19 mRNA vaccines by Moderna and Pfizer/BioNTech. These vaccines have demonstrated the efficacy of mRNA-LNP therapeutics and opened the door for future clinical applications. In mRNA-LNP systems, the LNPs serve as delivery platforms that protect the mRNA cargo from degradation by nucleases and mediate their intracellular delivery. The LNPs are typically composed of four components: an ionizable lipid, a phospholipid, cholesterol, and a lipid-anchored polyethylene glycol (PEG) conjugate (lipid-PEG). Here, LNPs encapsulating mRNA encoding firefly luciferase are formulated by microfluidic mixing of the organic phase containing LNP lipid components and the aqueous phase containing mRNA. These mRNA-LNPs are then tested in vitro to evaluate their transfection efficiency in HepG2 cells using a bioluminescent plate-based assay. Additionally, mRNA-LNPs are evaluated in vivo in C57BL/6 mice following an intravenous injection via the lateral tail vein. Whole-body bioluminescence imaging is performed by using an in vivo imaging system. Representative results are shown for the mRNA-LNP characteristics, their transfection efficiency in HepG2 cells, and the total luminescent flux in C57BL/6 mice.
Lipid nanoparticles (LNPs) have demonstrated great promise in recent years in the field of non-viral gene therapy. In 2018, the United States Food and Drug Administration (FDA) approved the first-ever RNA interference (RNAi) therapeutic, Onpattro by Alnylam, for the treatment of hereditary transthyretin amyloidosis1,2,3,4. This was an important step forward for lipid nanoparticles and RNA-based therapies. More recently, Moderna and Pfizer/BioNTech received FDA approvals for their mRNA-LNP vaccines against SARS-CoV-24,5. In each of these LNP-based nucleic acid therapies, the LNP serves to protect its cargo from degradation by nucleases and facilitate potent intracellular delivery6,7. While LNPs have seen success in RNAi therapies and vaccine applications, mRNA-LNPs have also been explored for use in protein replacement therapies8 as well as for the co-delivery of Cas9 mRNA and guide RNA for the delivery of the CRISPR-Cas9 system for gene editing9. However, there is no one specific formulation that is well-suited for all applications, and subtle changes in the LNP formulation parameters can greatly affect the potency and biodistribution in vivo8,10,11. Thus, individual mRNA-LNPs must be developed and evaluated to determine the optimal formulation for each LNP-based therapy.
LNPs are commonly formulated with four lipid components: an ionizable lipid, a phospholipid, cholesterol, and a lipid-anchored polyethylene-glycol (PEG) conjugate (lipid-PEG)11,12,13. The potent intracellular delivery facilitated by LNPs relies, in part, on the ionizable lipid component12. This component is neutral at physiological pH but becomes positively charged in the acidic environment of the endosome11. This change in ionic charge is thought to be a key contributor to endosomal escape12,14,15. In addition to the ionizable lipid, the phospholipid (helper lipid) component improves the encapsulation of the cargo and aids in endosomal escape, the cholesterol offers stability and enhances membrane fusion, and the lipid-PEG minimizes LNP aggregation and opsonization in circulation10,11,14,16. To formulate the LNP, these lipid components are combined in an organic phase, typically ethanol, and mixed with an aqueous phase containing the nucleic acid cargo. The LNP formulation process is very versatile in that it allows for different components to be easily substituted and combined at different molar ratios in order to formulate many LNP formulations with a multitude of physicochemical properties10,17. However, when exploring this vast variety of LNPs, it is crucial that each formulation is evaluated using a standardized procedure to accurately measure the differences in characterization and performance.
Here, the complete workflow for the formulation of mRNA-LNPs and the assessment of their performance in cells and animals is outlined.
NOTE: Always maintain RNase-free conditions when formulating mRNA-LNPs by wiping the surfaces and equipment with a surface decontaminant for RNases and DNA. Use only RNase-free tips and reagents.
All the animal procedures were performed in accordance with the Guidelines for the Care and Use of Laboratory Animals at the University of Pennsylvania and a protocol approved by the Institutional Animal Care and Use Committee (IACUC) at the University of Pennsylvania.
1. Pre-formulation preparation
2. Preparation of the lipid and nucleic acid mixes
3. Microfluidic formulation of mRNA-LNPs
4. Post-formulation processing and characterization of the mRNA-LNPs
5. In vitro transfection of HepG2 cells
NOTE: Various other cell lines, such as HeLa cells or HEK-293T cells, may be used for assessing the transfection efficiency of LNPs in vitro. All cells should test negative for mycoplasma prior to the LNP transfection studies.
6. In vivo evaluation of mRNA-LNPs in mice following tail vein injection
mRNA-LNPs were formulated using a microfluidic instrument that possessed an average hydrodynamic diameter of 76.16 nm and a polydispersity index of 0.098. The pKa of the mRNA-LNPs was found to be 5.75 by performing a TNS assay18. The encapsulation efficiency for these mRNA-LNPs was calculated to be 92.3% by using the modified fluorescence assay and equation 4.4. The overall RNA concentration that was used for the cell treatment and animal dosing was 40.24 ng/µL. This value was obtained from the modified fluorescence assay19, specifically from converting the fluorescence obtained by diluting the LNPs with 1% Triton X-100 and 1x TE buffer to certain concentrations and calculating the amount of encapsulated mRNA.
Equation 4.4
CTX = Concentration of mRNA from LNPs diluted in 1% Triton X-100
CTE = Concentration of mRNA from LNPs diluted in 1x TE buffer
Increasing bioluminescence was observed with increasing doses upon the treatment of 5,000 HepG2 cells per well with different doses of mRNA-LNP. Luciferase expression was easily detected at the lowest dose (5 ng/well) used in this study. However, other cell lines may require different amounts of mRNA-LNP to readily see differences in luminescence across doses.
Mice were treated with 2 µg of mRNA-LNP and assessed 6 h later for whole-body bioluminescence. As C12-200 mRNA-LNPs predominantly transfect the liver20, a strong bioluminescent signal was observed in the upper abdomen of the mice treated with our formulated LNPs. Using the imaging software, regions of interest were drawn around the liver signals to calculate the total luminescent flux. In this experiment, the total luminescent flux was approximately 5 x 109, which is consistent with other studies conducted with this LNP formulation4,21.
Table 1: Organic phase composition. Concentrations of the individual lipid stock solutions and the volumes that each component contributes to the 1.3 mL organic phase are described. Lipids are combined at a molar ratio of 35/16/46.5/2.5 (C12-200:DOPE:Cholesterol:C14-PEG2000). A 10% extra volume was prepared to account for syringe dead volumes during formulation. Please click here to download this Table.
Table 2: Microfluidic instrument settings for mRNA-LNP formulation at a 3:1 aqueous to organic flow rate ratio. The 5 mL syringe containing mRNA diluted in 10 mM citric acid buffer was inserted into the center channel, while the 3 mL syringe containing lipids was inserted into the right channel. LNPs were formulated at a 10:1 weight ratio of ionizable lipid:mRNA. Please click here to download this Table.
Table 3: mRNA-LNP characterization data determined by dynamic light scattering, a modified fluorescenceassay, and a 2-(p-toluidino) naphthalene-6-sulfonic acid (TNS) assay. Please click here to download this Table.
Figure 1: HepG2 cells treated with firefly luciferase mRNA-LNP. Overall, 5,000 HepG2 cells were seeded per well in a 96-well plate and allowed to adhere overnight. The cells were cultured in DMEM supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin. The cell culture medium was removed before cells were treated with 5 ng, 10 ng, and 20 ng doses of a C12-200-containing LNP formulation encapsulating firefly luciferase-encoding mRNA (in 100 µL of cell culture medium). At 24 h post treatment, the cell culture medium was removed, and 20 µL of 1x lysis buffer was added onto the cells before the addition of 100 µL of luciferase assay reagent. The bioluminescence from each well was measured using a plate reader following a 5 min incubation period. The fold increases are plotted relative to control wells that consisted of untreated HepG2 cells. The statistical significance was evaluated using a one-way ANOVA with post-hoc Tukey's test. ns, not significant; ****, p < 0.0001. Please click here to view a larger version of this figure.
Figure 2: C57BL/6 mice treated with firefly luciferase mRNA-LNP. In this work, 2 µg of total mRNA-LNP in 100 µL or 100 µL of 1x PBS was administered to C57BL/6 mice via the lateral tail vein. (A) Whole-body bioluminescence of the mice treated with either mRNA-LNP or 1X PBS was measured using the in vivo imaging system (IVIS) 6 h post administration. The IVIS Spectrum Living Image software was used to analyze and acquire the whole-body images. N = 3. (B) The total luminescent flux was quantified and plotted. The statistical significance was evaluated using a two-tailed Student's t-test. **, p < 0.01. Please click here to view a larger version of this figure.
With this workflow, a variety of mRNA-LNPs can be formulated and tested for their in vitro and in vivo efficiency. Ionizable lipids and excipients can be swapped out and combined at different molar ratios and different ionizable lipid to mRNA weight ratios to produce mRNA-LNPs with differing physicochemical properties22. Here, we formulated C12-200 mRNA-LNPs with a molar ratio of 35/16/46.5/2.5 (ionizable lipid:helper lipid:cholesterol:lipid-PEG) at a 10:1 ionizable lipid to mRNA weight ratio. These LNPs were tested for their transfection efficiency in vitro in HepG2 cells and in vivo in C57BL/6 mice. The C12-200 mRNA-LNPs were synthesized by microfluidic mixing using a commercially available microfluidic instrument. Though other formulation methods can be used to formulate LNPs, such as pipette mixing or T-junction mixing4, microfluidic mixing formulates mRNA-LNPs that are generally smaller, less polydisperse, and have higher encapsulation efficiencies4,13,23,24.
The formulation of mRNA-LNPs with the commercially available microfluidic instrument allows for the simple and flexible formulation of LNPs. The flow rate at the beginning of a run varies from the target flow rate set as the syringe pushers accelerate to full speed. This can lead to variable particle characteristics and inconsistent results. To overcome this, the microfluidic instrument was designed to collect the LNPs formulated at the start and end of a formulation as waste separately from the LNPs formulated at steady-state conditions. This can decrease the polydispersity and lead to more consistent particle properties. The start waste can be calculated based on the combination of syringes being used for formulation as per the manufacturer's manual. However, the end waste does not experience the same degree of non-steady-state flow conditions, so the volume is often set to 0.05 mL for all syringe combinations as per the manufacturer's recommendation.
It is critical to account for dead volume in syringes and tubes when preparing the correct volumes of the organic and aqueous phases. Preparing 10% extra volume of each phase ensures that the minimum volume of LNP desired can be formulated. In this protocol, a total formulation volume of 4.7 mL was inputted with 0.7 mL of waste, resulting in 4 mL of LNP collected in steady-state conditions. However, 1.3 mL of the organic phase and 3.9 mL of the aqueous phase were prepared for formulation. This ensures that the 5 mL syringe can be filled up to the 3.6 mL mark and the 3 mL syringe can be filled up to the 1.2 mL mark. If the volume reaches the marks on the syringes, then the volume specifications used on the microfluidic instrument are safe inputs. The final screen before pressing the "START" button is crucial for ensuring that all the parameters are correct. It is important to do a final check on the syringe types, the volumes in the syringes, the volumes to be dispensed, the syringe placement, and the flow rates.
After the formulation of mRNA-LNPs, the dialysis step has a few important considerations to keep in mind. The dialysis cassettes used require a minimum hydration time of 2 min before the LNPs can be loaded. This increases the membrane flexibility and allows the membrane to adjust more readily as the sample is added. Additionally, care must be taken not to damage or puncture the membranes of the dialysis cassettes while loading them with mRNA-LNPs. If the membranes become punctured or damaged, the formulated mRNA-LNPs can be easily lost upon loading.
In this protocol, we tested the in vitro efficiency of mRNA-LNPs in HepG2 cells. However, other cell types can be tested for mRNA-LNP transfection efficiency. If non-adherent cells are being used, the 96-well plate requires centrifugation at 500 g for 5 min prior to any removal of the cell culture medium25. This minimizes any loss of cells during the medium removal. The cells should also be tested for the presence of mycoplasma prior to any LNP transfection studies. It will be difficult to obtain consistent results and compare different LNP formulations if the cells test positive for mycoplasma. Additionally, primary cells may require higher doses of LNP to measure transfection without toxicity compared with cell lines. A luciferase-based viability assay can be used to evaluate the mRNA-LNP toxicity in vitro at varying doses in both primary cells and cell lines18.
One of the most important steps in obtaining consistent bioluminescence measurements across different mice following treatment with the same mRNA-LNP formulation is ensuring that the time between the intraperitoneal injection of d-luciferin and the IVIS measurement is consistent26. This time interval influences the stability of the bioluminescent signal obtained. The time interval needs to be long enough so that any fluctuations in the signal are minimized. A preliminary experiment can be run in which the mice are imaged every minute following the d-luciferin injection. The time interval when the signal starts to plateau is the one most suitable for the transfection experiment. In this protocol, a 10 min interval allowed for stable luminescence signals following the 150 mg d-luciferin/kg body weight injection, which is commonly performed for imaging27,28.
Additionally, the mRNA-LNP dose used in this protocol (0.1 mg mRNA/kg body weight) is small compared to the doses used to assess toxicity and therapeutic efficacy29,30. These smaller doses help to evaluate differences in the potencies of unique mRNA-LNP formulations while not oversaturating the luminescent signal obtained. However, this dose can be increased to measure the potential mRNA-LNP toxicity. For example, liver toxicity can be evaluated by quantifying the levels of alanine transaminase, aspartate transaminase, and alkaline phosphatase in the serum at different time points following mRNA-LNP injection31,32,33. These enzymes are normally found at low levels in the serum but are increased as a result of liver damage. Commercially available kits can be used to quantify the amount of these enzymes in the serum.
In this example, we used firefly luciferase-encoding mRNA, but other reporter mRNAs can be formulated into LNPs to assess potency. mRNAs encoding for green fluorescent protein (GFP) or mCherry can be employed to investigate cell-specific delivery following mRNA-LNP treatment34,35. A single-cell suspension can be produced from tissues of interest, and GFP+ or mCherry+ cells can be assessed by flow cytometry analysis. Additionally, mRNA-LNPs can be formulated with mRNA encoding for erythropoietin in order to measure liver transfection through the secretion of erythropoietin in the serum8,36,37. In Ai9 mice, the delivery of mRNA encoding Cre recombinase induces robust tdTomato fluorescence, which can serve as another method for investigating mRNA-LNP delivery to different cell populations38,39,40. Through the demonstration of this workflow, it is our hope that mRNA-LNP formulation ceases to be a barrier for investigators as they explore new ideas and possibilities for mRNA therapeutics.
The authors have nothing to disclose.
M.J.M. acknowledges support from a US National Institutes of Health (NIH) Director’s New Innovator Award (DP2 TR002776), a Burroughs Wellcome Fund Career Award at the Scientific Interface (CASI), a US National Science Foundation CAREER award (CBET-2145491), and additional funding from the National Institutes of Health (NCI R01 CA241661, NCI R37 CA244911, and NIDDK R01 DK123049).
0.1 M Hydrochloric Acid | Sigma | 7647-01-0 | |
0.22 μm Syringe Filters | Genesee | 25-243 | |
1 mL BD Slip Tip Syringe | BD | 309659 | |
1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (ammonium salt) (C14-PEG2000) | Avanti Polar Lipids | 880150P | |
1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) | Avanti Polar Lipids | 850725P | |
1.5 mL Eppendorf Tubes | Fisher Scientific | 05-408-129 | |
15 mL Conical Tubes | Fisher Scientific | 14-959-70C | |
200 proof Ethanol | Decon Labs | 2716 | |
23G Needles | Fisher Scientific | 14-826-6C | |
3 mL BD Disposable Syringes with Luer-Lok tips | Fisher Scientific | 14-823-435 | |
3 mL Dialysis Cassettes | Thermo Scientific | A52976 | |
96 Well Black Wall Black Bottom Plate | Fisher Scientific | 07-000-135 | |
96 Well White/Clear Bottom Plate, TC Surface | Thermo Scientific | 165306 | |
Ammonium Acetate, 1 Kilogram | Research Products International | 631-61-8 | |
Ammonium Citrate dibasic | SIgma | 3012-65-5 | |
BD Luer-Lok Syringe sterile, single use, 5 mL | BD | 309646 | |
C12-200 Ionizable Lipid | Cayman Chemical | 36699 | |
C57BL/6 Mice | Jackson Laboratory | 000664 | |
Cholesterol | Sigma | 57-88-5 | |
CleanCap FLuc mRNA (5moU) | TriLink Biotechnologies | L-7202 | |
Disposable cuvettes | Fisher Scientific | 14955129 | |
D-Luciferin, Potassium Salt | Thermo Scientific | L2916 | |
DMEM, high glucose | Thermofisher Scientific | 11965-084 | |
Exel Insulin Syringes – 0.5 mL | Fisher Scientific | 1484132 | |
Fetal Bovine Serum | Corning | 35-010-CV | |
Hep G2 [HEPG2] | ATCC | HB-8065 | |
HyPure Molecular Biology Grade Water | Cytiva | SH30538.03 | |
Infinite 200 PRO Plate Reader | Tecan | N/A | |
IVIS Spectrum In Vivo Imaging System | Perkin Elmer | N/A | |
Large Kimwipes | Fisher Scientific | 06-666-11D | |
Luciferase Assay Kit | Promega | E4550 | |
NanoAssemblr Ignite Cartridges – Classic – 100 Pack | Precision Nanosystems | NIN0065 | |
NanoAssemblr Ignite Instrument | Precision Nanosystems | NIN0001 | |
PBS – Phosphate-Buffered Saline (10x) pH 7.4, RNase-free | Thermo Scientific | AM9624 | |
Penicillin-Streptomycin | Thermofisher Scientific | 15140122 | |
QB Citrate Buffer, (Citrate 100 mM) pH 3.0 | Teknova | Q2442 | |
Quant-it RiboGreen RNA Assay Kit | Thermo Scientific | R11490 | |
Reporter Lysis 5x Buffer | Promega | E3971 | |
RNase Away Surface Decontaminant | Thermofisher Scientific | 7000TS1 | |
Sodium Chloride | Sigma | 7647-14-5 | |
Sodium Hydroxide | Sigma | 1310-73-2 | |
Sodium Phosphate | Sigma | 7601-54-9 | |
TNS reagent (6-(p-Toluidino)-2-naphthalenesulfonic acid sodium salt) | Sigma | T9792 | |
Triton X-100 | Sigma | 9036-19-5 | |
Zetasizer | Malvern Panalytical | NanoZS |