Open Access Article
Sin A
Park‡
a,
Dajeong
Hwang‡
a,
Jae Hoon
Kim
a,
Seung-Yeul
Lee
a,
Jaebeom
Lee
bc,
Han Sang
Kim
de,
Kyung-A
Kim
e,
Bumhee
Lim
f,
Jae-Eon
Lee
g,
Yong Hyun
Jeon
g,
Tae Jeong
Oh
a,
Jaewook
Lee
*a and
Sungwhan
An
*a
aGenomictree Inc., Yuseong-gu, Daejeon, 34027, Republic of Korea. E-mail: jwlee@genomictree.com; sungwhan@genomictree.com
bDepartment of Chemistry, Chungnam National University, Yuseong-gu, Daejeon, 34134, Republic of Korea
cDepartment of Chemical Engineering and Applied Chemistry, Chungnam National University, Yuseong-gu, Daejeon, 34134, Republic of Korea
dYonsei Cancer Center, Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
eDepartment of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
fNew Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), Dong-gu, Daegu 41061, Republic of Korea
gPreclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), Dong-gu, Daegu 41061, Republic of Korea
First published on 29th October 2024
Lipid nanoparticles (LNPs) are widely recognized as crucial carriers of mRNA in therapeutic and vaccine development. The typical lipid composition of mRNA-LNP systems includes an ionizable lipid, a helper lipid, a polyethylene glycol (PEG)-lipid, and cholesterol. Concerns arise regarding cholesterol's susceptibility to oxidation, potentially leading to undesired immunological responses and toxicity. In this study, we formulated novel LNPs by replacing cholesterol with phytochemical-derived compounds, specifically ginsenoside Rg2 and its derivative phytosterol protopanaxadiol (PPD), and validated their efficacy as mRNA delivery systems. The mRNA–LNP complexes were manually prepared through a simple mixing process. The biocompatibility of these Rg2-based LNPs (Rg2-LNP) and PPD-based LNPs (PPD-LNP) was assessed through cell viability assays, while the protective function of LNPs for mRNA was demonstrated by RNase treatment. Enhanced green fluorescent protein (EGFP) mRNA delivery and expression in A549 and HeLa cells were analyzed using optical microscopy and flow cytometry. The expression efficiency of Rg2-LNP and PPD-LNP was compared with that of commercially available LNPs, with both novel formulations demonstrating superior transfection and EGFP expression. Furthermore, in vivo tests following intramuscular (I.M.) injection in hairless mice demonstrated efficient luciferase (Luc) mRNA delivery and effective Luc expression using Rg2-LNP and PPD-LNP compared to commercial LNPs. Results indicated that the efficiency of EGFP and Luc expression in Rg2-LNP and PPD-LNP surpassed that of the cholesterol-based LNP formulation. These findings suggest that Rg2-LNP and PPD-LNP are promising candidates for future drug and gene delivery systems.
Lipid nanoparticles (LNPs) stand out as notable vehicles that have significantly advanced mRNA applications in humans, with several mRNA-based therapies either approved or in clinical trials.10 Various mRNA delivery systems have been developed, including viral vectors like adeno-associated virus (AAV), lentivirus (LV) and non-viral carriers such as lipid nanoparticles (LNPs), liposomes, exosomes, polyol-based NPs, and inorganic NPs like gold or magnetic NPs.11–16 In most cases, LNP-based mRNA delivery systems have been widely used, and the FDA has approved those complex structure-based COVID-19 vaccines from Moderna and Pfizer/BioNTech.17–20 Typically, LNPs are composed of four essential components: an ionic lipid, helper lipid, PEG lipid, and sterol, and they have a specific role in structural formation. Among these, sterol is important in maintaining the lipid bilayer structure, enhancing the nanostructure stability and mRNA protection.21–24 Although cholesterol is commonly used as the sterol component, its application poses certain risks.25–28 Because cholesterol is easily oxidated and converted to oxidative derivatives, including oxysterols like 25-hydroxycholesterol and 7β-hydroxycholesterol, it exhibits potential toxicity.29–31 Moreover, it is possible that there is a risk of unexpected virus contamination because of animal-derived cholesterol.32
In this study, we have developed an alternative LNP formulation that replaces cholesterol with ginsenoside, Rg2, or its derivative phytosterol, protopanaxadiol (PPD), to address such issues. These new formulations, abundant in ginseng, mitigate the risks associated with cholesterol and leverage the significant bioactivity and biocompatibility of these phytochemicals, including anti-cancer and antioxidant effects.33–36 These beneficial properties make them suitable components for LNP formulation, with potentially synergistic effects alongside mRNA therapeutics.37–39 For therapeutic efficacy, mRNA needs to penetrate target cells and translate proteins adequately. In the present study, we have evaluated new LNPs that were Rg2-based LNPs (Rg2-LNP) and PPD-based LNPs (PPD-LNP) regarding their low toxicity, high biocompatibility, and mRNA protection ability against RNase. Also, we have demonstrated their effectiveness as a carrier for mRNA delivery and expression in vitro using enhanced green fluorescent protein (EGFP) mRNA as well as in vivo using luciferase (Luc) mRNA. Overall, the results described here show that ginsenoside and phytosterol-based LNPs could be alternatives to cholesterol-based LNPs, and consequently, Rg2-LNP and PPD-LNP exhibited considerable potential as delivery systems for mRNA therapeutics and vaccines.
The size and morphology of Rg2-LNP and PPD-LNP were characterized using a nanoparticle tracking analyzer (NTA, NanoSight NS300, Malvern Panalytical, Malvern, UK), a zetasizer (Nano ZS, Malvern Panalytical, Malvern, UK) for measurement of dynamic light scattering (DLS) and a transmission electron microscope (TEM, H-7600, Hitachi, Tokyo, Japan). The cell morphology and expression of EGFP after EGFP mRNA delivery into the cell were monitored using a fluorescence optical microscope (Olympus, CKX553, Tokyo, Japan). The cell viability was measured using cell counting kit-8 (CCK-8) from Dojindo (Kumamoto, Kyushu, Japan), and the absorbance was measured using a microplate spectrophotometer (Mobi, MicroDigital, Seongnam, South Korea). The fluorescence (FL) intensity was monitored using a microplate multi-mode reader (Bio Tek Synergy HTX, Agilent, Santa Clara, CA, USA). The EGFP positive cells and the mean of FL intensities were characterized by flow cytometry (BD FACSLyric™, BD Biosciences, Franklin Lakes, NJ, USA). Male Balb/c nude mice were purchased (Orient Bio Inc, Seongnam, South Korea), and live animal imaging was performed using an IVIS Lumina III imaging system (PerkinElmer, Waltham, MA, USA).
:
10
:
1.5
:
78 or 50
:
10
:
1.5
:
39 (molar ratio = ionizable lipid
:
phospholipid
:
PEG lipid
:
Rg2). Subsequently, Rg2-LNP was softly mixed with mRNA to construct the mRNA-Rg2-LNP complex for the delivery and expression test under various weight ratios (wmRNA
:
wLNP), 1
:
5, 1
:
10 and 1
:
20, and that the mixture was incubated for 15 min at room temperature (R.T.). In this case, the solvent volume ratio (VmRNA
:
VLNP) between aqueous solution (mRNA) and ethanol (LNP) was 3
:
1. After incubation, the mRNA-Rg2-LNP was sonicated for 1 min and then treated with the cell to evaluate the delivery and expression efficiency.
To synthesize the PPD-LNP, components for the LNP formulation were mixed under varying concentrations: 50
:
10
:
1.5
:
45.8 and 50
:
10
:
1.5
:
22.9 (molar ratio = ionizable lipid
:
phospholipid
:
PEG lipid
:
PPD). The mRNA
:
PPD-LNP mixing ratio for further study was the same as for Rg2-LNP.
To evaluate the encapsulation efficiency of Rg2-LNP and PPD-LNP using the Ribogreen RNA assay kit, the FL intensity was measured with a microplate multi-mode reader at an excitation wavelength of 480 nm and an emission wavelength of 530 nm.
:
phospholipid
:
PEG lipid
:
Rg2 or PPD) were further optimized to enhance the expression efficiency, resulting in a ratio of 50
:
10
:
0.75
:
39 for Rg2-LNP and 50
:
10
:
0.75
:
22.9 for PPD-LNP.
In addition, after 24 h of transfection, both cell lines were harvested and resuspended in PBS with 1% FBS and then the expression efficiency was evaluated by flow cytometry.
000g for 10 min at 4 °C. The supernatant was then collected to measure relative luciferase activity using the luciferase assay system.
Male Balb/c nude mice were sacrificed to assess the efficiency of delivery and expression in vivo. Each mouse was dosed with 10 μg of Luc mRNA encapsulated in Rg2-LNP, PPD-LNP, SM-102-LNP, or modified SM-102-LNP through an intramuscular (I.M.) injection. The molar ratios of Rg2-LNP and PPD-LNP formulation used for the Luc mRNA in vivo delivery and expression tests were 50
:
10
:
0.75
:
39 and 50
:
10
:
0.75
:
22.9, respectively. The molar ratios of SM-102-LNP and modified SM-102-LNP formulations followed the manufacturer's protocol. For bioluminescence imaging (BLI), mice were administered D-luciferin via intraperitoneal injection. During imaging, anesthesia using 1–2% isoflurane gas was applied to all mice. BLI was conducted 5 min after substrate injection using the IVIS Lumina III imaging system, from 1 h to 54 h post-injection of Luc mRNA-encapsulated Rg2-LNP and PPD-LNP as well as SM-102-LNP and modified SM-102-LNP as the positive control. LIVING-IMAGE software (version 3.0, PerkinElmer) was utilized to overlay grayscale photographic images and bioluminescent color images. BLI signals were quantified in units of photons per cm2 per second per steradian (P cm−2 s−1 sr−1).
![]() | ||
| Fig. 1 TEM images of (A) Rg2-LNP, (B) EGFP mRNA-Rg2-LNP, (D) PPD-LNP, and (E) EGFP mRNA-PPD-LNP and NTA results of (C) EGFP mRNA-Rg2-LNP and (F) EGFP mRNA-PPD-LNP. | ||
:
10 and 1
:
20, indicating complete encapsulation. In contrast, PPD-LNP exhibited unencapsulated mRNA bands at all tested ratios (1
:
5, 1
:
10, and 1
:
20), though the band intensity decreased with increasing LNP mass. The encapsulation efficiency of Rg2-LNP and PPD-LNP was evaluated using the Ribogreen RNA assay, showing 81.9% and 68.7%, respectively. These mRNA-LNPs were prepared through simple hand mixing, which might result in a lower encapsulation efficiency compared to those prepared using a microfluidics system. Nevertheless, the Rg2-LNP and PPD-LNP produced by this mixing process were sufficient mRNA carriers for expressing EGFP and Luc for in vitro or in vivo experiments. However, we recognized the limitations of manual preparation, including challenges in achieving precise size uniformity and optimal mRNA encapsulation per LNP. To address these limitations, we plan to utilize microfluidic devices in future studies to enhance the development of mRNA-based therapeutics.
On the other hand, the higher encapsulation efficiency of Rg2-LNP can be attributed to the glucose component forming hydrogen bonds with mRNA and the ionizable lipid binding through electrostatic interactions, resulting in tighter mRNA capture, as mentioned above. mRNA stability within the LNPs was assessed by analyzing mRNA integrity after RNase treatment via electrophoresis (Fig. 2B and D). Naked mRNA was completely degraded by RNase exposure, whereas mRNA within Rg2-LNP and PPD-LNP remained intact even at increased RNase concentrations (red box in Fig. 2B and D). The unencapsulated mRNA band in PPD-LNP disappeared after RNase treatment, while the encapsulated mRNA remained protected, demonstrating that both Rg2-LNP and PPD-LNP effectively shield mRNA from degradation.
:
10
:
1.5
:
39 for Rg2-LNP and 50
:
10
:
1.5
:
22.9 for PPD-LNP. Firstly, in evaluating the delivery efficacy of LNPs, 250 ng of EGFP mRNA encapsulated by Rg2-LNP or PPD-LNP at a 1
:
10 weight ratio (mRNA
:
LNP) was transfected into A549 cells. After 24 h of treatment, optical microscopy revealed a clear signal of green fluorescence in all treated cells, indicating successful delivery and translation of EGFP mRNA (Fig. 4B and C). Additionally, the delivery effectiveness and transfection efficiency of Rg2-LNP and PPD-LNP were compared with those of commercially available LNPs SM-102-LNP (Moderna) and ALC-0315-LNP (Pfizer/BioNTech), which retain the cholesterol component. After delivering EGFP mRNA with these LNPs, EGFP expression in A549 cells was carefully observed. According to the fluorescence image, the fluorescence brightness was significantly higher in cells transfected with Rg2-LNP and PPD-LNP compared to those of SM-102-LNP and ALC-0315-LNP (Fig. S5A†). In addition, the efficiency of EGFP expression was compared by flow cytometry with various LNP carriers (Fig. S5B–D†). In this case, the population of EGFP-positive A549 cells was over 95% after treatment with EGFP mRNA encapsulated Rg2-LNP, PPD-LNP, and SM-102-LNP, but it was only 25% in ALC-0315-LNP. Interestingly, the mean fluorescence intensity (MFI) was the highest in the case of PPD-LNP, followed by Rg2-LNP, which was higher than SM-102-LNP. In addition, Luc mRNA transfection yields were compared between commercial LNPs and Rg2-LNP and PPD-LNP in A549 cell lines using a bioluminescence assay (Fig. S6†). The results were similar to those of the EGFP expression trends in that relative light units (RLU) were higher for Rg2-LNP and PPD-LNP compared to SM-102-LNP and ALC-0315-LNP. Additionally, the GFP mRNA delivery and expression efficiency of modified SM-102-LNP, where the helper lipid was switched from DSPC to DOPE, was compared with Rg2-LNP and PPD-LNP, and the results are presented in Fig. S7.† In this case, brighter green fluorescence was observed in LNPs based on SM-102/DOPE/Rg2/PEG lipid (Rg2-LNP) or SM-102/DOPE/PPD/PEG lipid (PPD-LNP) than in LNPs formulated with SM-102/DOPE/cholesterol/PEG lipid (modified SM-102 LNP).
![]() | ||
| Fig. 4 Bright field images and fluorescence images of (A) non-treated, (B) EGFP mRNA-Rg2-LNP treated and (C) EGFP mRNA-PPD-LNP treated A549 cells (scale bar: 100 μm). | ||
:
LNP) at 1
:
10 and 1
:
20, and a higher signal intensity of EGFP was observed at a 1
:
20 weight ratio (mRNA
:
LNP) instead of 1
:
10 (Fig. 6). Interestingly, when assessing the encapsulation efficiency of LNPs by gel electrophoresis, results showed that nearly all mRNA was encapsulated in Rg2-LNP at 1
:
10 and 1
:
20 ratios, and for PPD-LNP, unencapsulated mRNA bands were similar at both ratios, so it was expected that the tendency of EGFP expression might be the same for both weight ratios in HeLa cells as in A549 cells. This indicates that the tendency of expression efficiency did not directly correspond to the encapsulation efficiency. The expression efficiency and MFI of EGFP in HeLa cells were also evaluated after transfection of EGFP mRNA using commercial LNPs, Rg2-LNP, and PPD-LNP, similar to the tests conducted in A549 cells. Flow cytometry analysis results showed that although the expression efficiency and MFI were lower in HeLa cells compared to A549 cells, PPD-LNP had the highest MFI, demonstrating a similar pattern in HeLa cells (Fig. S8†). This finding indicates that the newly developed LNPs have excellent transfection efficiency. Also, the results highlight the importance of tuning and optimizing formulation conditions for effective protein expression in target cell lines.
![]() | ||
| Fig. 5 Observation of EGFP expression in HeLa cells. (A) Non-treated conditions, (B) EGFP mRNA-Rg2-LNP conditions and (C) EGFP mRNA-PPD-LNP conditions (scale bar: 100 μm). | ||
Additional optimization involved reducing the PEG lipid component in the LNP formulation, which enhanced the EGFP translation efficiency (Fig. 7B and D). It was reported that a lower surface density of PEG has been associated with increased uptake into cancer cells, including HeLa cells.49–51 In our case, also a lower PEG density improved lipid nanoparticle uptake by HeLa cells, thereby increasing EGFP translation. Furthermore, high levels of EGFP expression remained after 48 h post-treatment (Fig. S9†). These findings emphasize optimizing the mRNA
:
LNP weight ratio and the PEG lipid content based on the target cell type to achieve delivery and optimal translation efficiency.
Potential of in vivo mRNA delivery and expression
The potential for in vivo applications of Rg2-LNP and PPD-LNP was assessed using bioluminescence imaging in mice. Mice were intramuscularly injected with Luc mRNA encapsulated in Rg2-LNP and PPD-LNP. The bioluminescence imaging revealed a signal of Luc mRNA translation at 6 hours post-injection, which persisted until 54 h (Fig. 8B, Fig. S10 and S11†), while no evident signal from empty vehicles was present. To evaluate the efficiency of in vivo mRNA delivery and expression in cells of mRNA encapsulated in LNPs, Rg2-LNP, and PPD-LNP, mRNA-Luc expression was examined in hairless mice. Commercially available cholesterol-retaining SM-102-LNP (with DSPC as the helper lipid) and modified SM-102-LNP (with DOPE as the helper lipid) were used as positive controls because the population of EGFP-positive cells was comparable between SM-102-LNP, Rg2-LNP, and PPD-LNP, SM-102-LNP. According to IVIS imaging and total flux profiles after I.M. injection, the total flux of the SM-102-LNP series decreased rapidly after 6 hours (Fig. S10 and S11†).
In contrast, the expression pattern of Luc in Rg2-LNP and PPD-LNP showed a slightly delayed initial expression compared to that of SM-102-LNP, but the decrease in Luc expression was slower. Notably, Rg2-LNP had the highest flux value after 12 hours post-injection across all LNP conditions according to the profile of total flux. Additionally, the total flux of PPD-LNP was higher than that of SM-102-LNP at 12 hours post-injection (Fig. S11†). These results unequivocally demonstrate the effectiveness of Rg2-LNP and PPD-LNP in delivering mRNA and facilitating translation in vivo, instilling confidence in their potential for gene therapy applications.
In conclusion, the mRNA complexes with Rg2-LNP or PPD-LNP, synthesized through a straightforward manual mixing process, demonstrated highly effective mRNA delivery and expression, highlighting their potential as robust mRNA delivery systems in this study.
Footnotes |
| † Electronic supplementary information (ESI) available. See DOI: https://doi.org/10.1039/d4bm01070a |
| ‡ Both contributed equally. |
| This journal is © The Royal Society of Chemistry 2024 |