Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2024 Mar 4;121(11):e2307809121. doi: 10.1073/pnas.2307809121

Electrostatic adsorption of polyanions onto lipid nanoparticles controls uptake, trafficking, and transfection of RNA and DNA therapies

Namita Nabar a,b,c, Tamara G Dacoba a,c, Gil Covarrubias a, Denisse Romero-Cruz d,e, Paula T Hammond a,b,c,1
PMCID: PMC10945854  PMID: 38437543

Significance

Gene therapies, in the form of messenger RNA or plasmid DNA, are promising treatments for genetic conditions. Lipid nanoparticles (LNPs) can effectively package and deliver these therapeutics. However, the efficacy of LNP-mediated gene delivery is challenged by off-target and nonspecific uptake, by clearance organs such as the liver and by the mononuclear phagocytic system. In this work, we developed and characterized layered LNPs (LLNPs) or LNPs electrostatically coated with a charged, biocompatible polyanion. Variations in polyanion chemistry modulated transfection in multiple cell lines and altered organ-level biodistribution and transfection in healthy C57BL/6 mice. Using this layering approach, LNP surface chemistry can be rationally designed to mitigate off-target and nonspecific uptake while boosting transfection in target cells.

Keywords: gene delivery, lipid nanoparticles, messenger RNA, layer-by-layer

Abstract

Rapid advances in nucleic acid therapies highlight the immense therapeutic potential of genetic therapeutics. Lipid nanoparticles (LNPs) are highly potent nonviral transfection agents that can encapsulate and deliver various nucleic acid therapeutics, including but not limited to messenger RNA (mRNA), silencing RNA (siRNA), and plasmid DNA (pDNA). However, a major challenge of targeted LNP-mediated systemic delivery is the nanoparticles’ nonspecific uptake by the liver and the mononuclear phagocytic system, due partly to the adsorption of endogenous serum proteins onto LNP surfaces. Tunable LNP surface chemistries may enable efficacious delivery across a range of organs and cell types. Here, we describe a method to electrostatically adsorb bioactive polyelectrolytes onto LNPs to create layered LNPs (LLNPs). LNP cores varying in nucleic acid cargo and component lipids were stably layered with four biologically relevant polyanions: hyaluronate (HA), poly-L-aspartate (PLD), poly-L-glutamate (PLE), and polyacrylate (PAA). We further investigated the impact of the four surface polyanions on the transfection and uptake of mRNA- and pDNA-loaded LNPs in cell cultures. PLD- and PLE-LLNPs increased mRNA transfection twofold over unlayered LNPs in immune cells. HA-LLNPs increased pDNA transfection rates by more than twofold in epithelial and immune cells. In a healthy C57BL/6 murine model, PLE- and HA-LLNPs increased transfection by 1.8-fold to 2.5-fold over unlayered LNPs in the liver and spleen. These results suggest that LbL assembly is a generalizable, highly tunable platform to modify the targeting specificity, stability, and transfection efficacy of LNPs, as well as incorporate other charged targeting and therapeutic molecules into these systems.


Nucleic acid therapies have successfully been used to treat numerous genetic conditions and infectious diseases (1, 2). Modalities such as messenger RNA (mRNA), small interfering RNA (siRNA), and plasmid DNA (pDNA) can control levels of disease-relevant proteins via gene editing, silencing, or expression (2). Recent advances in gene editing, such as CRISPR/Cas systems, have significantly accelerated the development of curative therapies for hereditary disorders (36). However, translatable and scalable gene therapies require efficacious delivery carriers, since naked genetic vectors minimally penetrate cells and are rapidly degraded by enzymes in the body (7, 8).

Lipid nanoparticles (LNPs) are highly potent gene delivery vehicles that have been clinically translated in multiple applications. Patisiran, the first FDA-approved LNP therapy in 2018, delivers siRNA to the liver to treat transthyretin-mediated amyloidosis (1, 9). The 2020 emergency FDA approval and global deployment of the Moderna and Pfizer/BioNTech mRNA-LNP COVID-19 vaccines further demonstrated LNPs’ therapeutic potential (1). LNPs are composed of four component lipids, each contributing to the nanocarrier’s encapsulation and transfection potency: a) Ionizable cationic lipids, which electrostatically bind anionic nucleic acids and drive transfection by inducing intracellular endosomal escape; b and c) cholesterol and phospholipids, which provide structure and encapsulation; d) lipid-anchored surface polyethylene glycol (PEG)-chains, which provide colloidal stability (7, 10). Delivered intravenously, LNPs preferentially accumulate in the liver, due in part to the adsorption of serum proteins such as apolipoprotein E, that act as natural ligands for receptors on hepatocytes (10, 11). Liver accumulation is further aided by slow local blood flow, organ vasculature, and phagocytic cell phenotypes in hepatic sinusoids (12, 13). While these features greatly facilitate hepatic gene delivery, they challenge delivery for many genetic disorders that require extrahepatic delivery (11, 14).

Certain therapeutic applications require well-defined LNP selectivity as well as efficacy in order to avoid off-target effects or optimize treatments. Selectivity can be gained by controlling surface protein adsorption and introducing more specific targeting interactions. PEG-functionalized lipids ensure serum stability of LNPs but may elicit immune responses in chronic or repeat LNP doses, due to anti-PEG antibodies in humans (11). Furthermore, PEGylated lipids exist in constant equilibrium between their free form and their incorporated state in the LNP; their constant lipid exchange allows for additional serum protein adsorption (11). Prior work has illustrated that the chemistries of the four component lipids can be tuned to drive LNP potency and biodistribution. Combinatorial libraries of ionizable lipid chemistries have identified structures capable of redirecting maximal LNP transfection from the liver to target areas, such as the lungs or spleen (1517). Other studies have demonstrated the impact of ionizable lipid chemistry and charged helper lipids on redirecting LNP biodistribution and transfection, by tuning LNP size, charge, and pKa (18, 19). Although these correlations can be used to leverage the protein corona for targeted delivery where advantageous, identification of hit formulations requires the synthesis and screening of hundreds of lipid structures. Ideally, LNP stabilization and targeting could be accomplished by introducing nonPEGylated LNP modifications, which can provide both the required colloidal stability and a rational methodology toward selectivity and influence on nanoparticle (NP) cellular trafficking.

We hypothesized that LNP trafficking, transfection, and biodistribution could be tuned with layer-by-layer (LbL) self-assembly. In the LbL approach, surfaces such as cationic NPs are coated by polyelectrolyte layers of alternating charge (20, 21). Adsorption of bioactive polyanions that leverage interactions with key cell receptors has modulated NP uptake by various cancer and immune cell types (2224). Furthermore, adsorbed polyelectrolytes adopt dense, loopy conformations that form hydrated “stealthy” layers around NPs that can control cargo release, reduce opsonin adsorption, and target therapeutics to cells of interest (25). LbL self-assembly is highly modular and has been successfully applied to a diverse range of NP cores, including lipidic nanocarriers (20, 22, 24, 26). Here, we describe the formation and behavior of layered LNPs (LLNPs) or LNPs that are electrostatically coated with a biocompatible polyanion. To achieve stable adsorption, LNPs were titrated to a positive charge, then incubated in solutions of polyanions. Layering was generalizable across cores; stable LLNPs were formed from a diverse set of LNP cores varying in component lipids and nucleic acid cargos. Tested polyanions included biologically relevant chemistries, such as polysaccharides, homopolypeptides, and synthetic hydrocarbon backbones (21, 22). We demonstrated that surface polyanion adsorption alters the physicochemical properties of LNPs while maintaining encapsulation of nucleic acid cargos. Further, polyanions modulated uptake and transfection in vitro and in vivo. This method of stable electrostatic adsorption onto LNP surfaces provides a simple and modular platform to control LNP targeting and specificity, as well as incorporate more diverse targeting and therapeutic molecules to benefit on-target nucleic acid delivery.

Results

LNPs Can Be Stably Layered with Biologically Relevant Polyanions.

To test the impact of polyanion structure on the performance of LLNPs, we layered LNPs with a library of four carboxylated polyanions: hyaluronate (HA), poly-L-aspartate (PLD), poly-L-glutamate (PLE), and poly-acrylate (PAA) (Fig. 1A). These biopolymers were chosen for their therapeutic promise, given that they have previously modulated NP uptake by specific cancer and primary cells. HA has facilitated NP tumor targeting, since it binds the CD44 receptor over-expressed in several cancers; PLD has further improved uptake in ovarian cancer lines (2224). PLE and PAA have, respectively, up-regulated and down-regulated NP uptake across panels of ovarian cancer and primary immune cells (22). The polyanions varied in monomer structures, sampling biologically relevant saccharides (HA), uniformly repeating poly(amino acid)s (PLD, PLE), and hydrocarbon-based acrylate backbones (PAA). Polyanion molecular weights were 20, 14, 15, and 15 kDa, respectively. To test whether layering was modular across LNPs, we formulated a set of LNP cores, varying in component lipids, lipid ratios, and nucleic acid cargos (either mRNA or pDNA). Formulations contained commercially available ionizable cationic lipids used in prior applications, with either single valency (DLin-MC3-DMA, DLin-KC2-DMA, and ALC-0315) or multiple valency (C12-200 and cKK-E12) (SI Appendix, Table S1) (2, 2730). LNPs were suspended in acidic conditions to express positive surface charge, then incubated in equal volumes of each of the four polyanions (Fig. 1B). Stable layering was defined as a state achieving complete surface charge conversion from >30 to <−30 mV, with diameters below 200 nm as measured by dynamic light scattering (DLS). Optimal polyanion concentrations were determined empirically through titration (SI Appendix, Fig. S1). Relative to unlayered LNPs, LLNP diameters increased by 5 to 50 nm (Fig. 1C and SI Appendix, Table S2). Variations in sizes are attributed to both polyelectrolyte secondary interactions and hydration effects. For example, HA, which caused the largest diameter increases, can strongly self-associate through hydrogen bonding, leading to a thicker adsorbed layer; its swelling may also be due to high water association (31, 32). Layering LNPs induced complete charge reversal, from unlayered LNP zeta potentials of +35 ± 1 mV to layered zeta potentials of −23 ± 10 to −36 ± 17 mV (Fig. 1D). The magnitude of charge reversal depended on the weight equivalents of polyanion used for layering. Nucleic acid encapsulation efficiency did not change significantly upon the addition of surface layers, suggesting that the polyanions do not displace encapsulated cargo (Fig. 1E). Furthermore, LLNPs retain internal core structure, as confirmed by transmission electron micrography (TEM) (Fig. 1F).

Fig. 1.

Fig. 1.

LNPs can be stably layered with carboxylated polyanions. (A) Schematic depicting a set of carboxylated polyanions with varying chemistries deposited onto positively charged LNP surfaces through electrostatic adsorption. (B) Schematic depicting process of layering LNPs with polyanions under stirring. (C) Z-averages measured via dynamic light scattering (DLS) of unlayered and layered mRNA-LNPs containing ALC-0315. N = 3, three technical replicates. (D) Surface zeta potential measurements of unlayered and layered mRNA-LNPs. N = 3, three technical replicates; N = 1, three technical replicates for UL. (E) mRNA encapsulation efficiency of unlayered and layered mRNA-LNPs. N = 3. (F) Representative negative-stained transmission electron micrography of unlayered and PAA-layered mRNA LNPs. All data represented as mean ± SD. (G) TNS titration curves of unlayered and layered mRNA-LNPs containing ALC-0315, curve visualized using locally weighted scatterplot smoothing. N = 6 per reported value at a given pH; N = 3 for HA-LLNPs. Apparent pKa was determined by conducting a four-parameter logistic regression on each curve and determining pH at 50% normalized TNS fluorescence. R2 of fit = 0.98, 0.96, 0.96, 0.94, 0.94 for UL, HA, PLD, PLE, and PAA respectively. (H) Fold change in total protein adsorbed onto LLNPs, relative to total protein adsorbed onto unlayered LNPs, as measured with a BCA assay. Protein was quantified using a standard curve of baseline-corrected values. Data was fit to a second-order polynomial; R2 = 0.99. Standard curve was used to interpolate each sample value. N = 3; two values in PAA-LLNP group were outside the interpolation range. Data represented as mean ± SD. Ordinary one-way ANOVA, Tukey’s test post hoc. ****P < 0.0001, ***P < 0.001, **P < 0.01, *P < 0.05.

Surface Polyanion Alters Apparent pKa of LNPs.

We hypothesized that the addition of a charged polyanion with acidic carboxylic groups may change the apparent pKa of the LLNP. Prior work optimizing ionizable lipids has demonstrated that the apparent pKa of the LNP impacts its transfection potency, immunogenicity, and targeting in vivo (18, 28, 3335). We therefore quantified the impact of surface polyanions on the apparent pKa of LLNPs. To illustrate trends in pKa, mRNA-LNPs with ionizable lipid ALC-0315, which has a reported pKa of 6.5, were layered with the four polyanions. ALC-0315 was chosen as a representative lipid given its monovalency and use in the Pfizer/BioNTech COVID-19 vaccine (1). Apparent pKa was determined using 2-p-toluidinonaphthalene-6-sulfonic acid (TNS), an anionic fluorophore that emits signal when in hydrophobic environments, such as cationic lipid membranes, through previously established binding assays (35). Briefly, pKa was reported as the pH at which the normalized TNS fluorescence, a proxy for LNP protonation, was 50% of the maximum detected TNS fluorescence. While unlayered LNPs exhibited a pKa of 6.5 (95% CI [6.4, 6.6]), the addition of PLD, PLE, and PAA decreased the apparent pKa to 4.7 (95% CI [4.4, 4.9]), 5.0 (95% CI [4.7, 5.3]), and 4.4 (95% CI [3.3, 4.9]), respectively (Fig. 1G). The acidified pKa of LLNPs suggests that the proximity and concentration of the deposited polyanion’s functional groups—in this case, acidified carboxyl groups—may modulate the pKa of the LNP (Fig. 1G). Notably, HA-LLNPs exhibited a pKa of 6.5 (95% CI [6.3, 6.6]) similar to unlayered LNPs, though HA itself has a reported pKa ranging from 3 to 4 (36, 37). Furthermore, the pKa curves of LLNPs showed a buffering zone, within which TNS fluorescence plateaued, between pH 3.5 to 4, 3.5 to 4.5, and 3.5 to 5 for PLD, PLE, and PAA, respectively. The diffusion of TNS, an anionic molecule, through the outer polyanion layer into the core may be driven by hydrophobicity gradients. The presence of two ionizable species—the polyanion and the ionizable lipid—may create a multi-step equilibrium.

Surface Polyanion Reduces LNP Surface Protein Adsorption.

Given that LNP extrahepatic delivery is hindered by adsorption of liver-targeting endogenous proteins, we aimed to quantify the impact of the surface polyanion on protein adsorption (10). To test the impact of the surface polyanion on LNP stability in biological conditions, both unlayered and LLNPs were incubated in either mouse plasma or water at 37 °C and then washed via centrifugation. DLS measurements indicated that unlayered LNPs in mouse plasma were 131 ± 6 nm larger than LNPs in water, likely due to the adsorption of plasma proteins. By contrast, PLD-LLNPs displayed a size increase of 21 ± 15 nm; PAA-LLNPs displayed a size increase of 9 ± 6 nm (SI Appendix, Fig. S2A). This improved stability may be attributed to the polyanion’s ability to mitigate protein adsorption. In a separate study, unlayered and layered LNPs containing DOPE-Cy5 were similarly incubated in mouse plasma for 1 h at 37 °C and washed via centrifugation. Total adsorbed plasma protein content was quantified with a BCA assay and normalized by Cy5 signal. Within this study, LLNPs adsorbed fourfold less total plasma protein than did unlayered LNPs (Fig. 1H). The zeta potentials of all LLNPs tested were between −29.3 ± 0.9 and −38.7 ± 2.0 mV; therefore, differences in adsorbed protein content are not attributed to any major differences in surface charge (SI Appendix, Table S3). Differences in adsorbed protein profiles have been shown to influence biodistribution and transfection in vivo (38). Preliminary characterization of adsorbed protein profiles was visualized with SDS-PAGE (SI Appendix, Fig. S2B); however, further proteomics analysis of the proteins preferentially bound to each layer will provide greater insights into LLNPs’ cellular interactions in future work.

Surface Polyanion Alters LNP Transfection of mRNA and pDNA.

To quantify the impact of LNP surface chemistry on transfection, different cell lines were treated with unlayered and layered LNPs containing luciferase-encoding mRNA. To isolate the impact of the polyanion, LNP core formulations were held constant in a 25/52.5/16/2.5 ratio of ALC-0315/cholesterol/DOPE/DMG-PEG-2000 and then layered with each polyanion. Cell lines included HEK293T, adherent epithelial cells; RAW 264.7, highly phagocytic macrophages; and EL4 and Jurkats, T lymphoma cells with low uptake rates. To assess the impact of the surface layer on rates of uptake and internalization, cells were treated with LNPs for either 4 h or 24 h. For the shorter treatment, cells were washed at 4 h and incubated for an additional 20 h in fresh media before readout, to allow time for detectable protein translation.

Comparisons of transfection after either 4 h or 24 h treatment suggested that polyanion coatings alter LNP uptake or RNA trafficking and release in a cell-specific manner (Fig. 2 A and B). Perhaps the most apparent trends are selective cell transfection based on the outer layer; for example, HA-LLNPs show significant uptake or association with each cell line examined in this study, but selective transfection in RAW 264.7 macrophages. PLE-LLNPs appear to drive much more transfection in HEK293T and Jurkat cells over EL4s and macrophages at longer timeframes. This behavior is particularly notable because macrophages tend to take up NPs at high rates compared to other cells; however, PLE-LLNPs do not show significant cell uptake or transfection in macrophages. After a 24 h treatment, PLD-LLNPs generated comparable or greater luciferase expression than unlayered LNPs, outperforming other layered formulations and providing strong transfection across these cell lines independent of cell association (Fig. 2B). On the other hand, PAA, which is a synthetic and highly charged polyanion, shows significantly lower uptake and relatively low transfection across each cell line. These observations contrast sharply with the unlayered LNPs, which exhibit entirely different trends in association and transfection. These results indicate that the polyanion layer coats the LNP surface and can meaningfully alter the trafficking, and ultimately the transfection, in different cell types.

Fig. 2.

Fig. 2.

Surface polyanions modulate LNP uptake and transfection across cell lines. (A) Luciferase activity of HEK293T, RAW 264.7, EL4, and Jurkat cells treated with unlayered or layered luciferase mRNA-LNPs for 4 h, then incubated for 20 h. HEK293T cells were dosed at 50 ng mRNA/well; all other cell lines were dosed at 100 ng mRNA/well. Data were normalized by cell viability; z-scores were plotted per cell line. N = 3. (B) Luciferase activity of cell lines treated with unlayered or layered luciferase mRNA-LNPs for 24 h. Data were normalized by cell viability; z-scores were plotted per cell line. N = 3. (C) Associated Cy5 signal of cell lines treated with unlayered or layered luciferase mRNA-LNPs for 4 h, then incubated for 20 h. Z-scores were plotted per cell line. N = 3. (D) Associated Cy5 signal of above-mentioned cell lines treated with unlayered or layered luciferase mRNA-LNPs for 24 h. Z-scores were plotted per cell line. N = 3. (E) GFP-positive cell percentages of HEK293Ts, EL4s, and HepG2s treated with unlayered or layered GFP-encoding pDNA LNPs for 4 h, then incubated for 20 h. HEK293T cells were dosed at 50 ng pDNA/well; all other cell lines were dosed at 100 ng pDNA/well. N = 3. Data represented as mean ± SD. Ordinary one-way ANOVA, Tukey’s test post hoc. ****P < 0.0001, ***P < 0.001, **P < 0.01, *P < 0.05.

There are also meaningful differences in the apparent kinetics of association and transfection of the NPs. In HEK293Ts, for example, PLE-LLNPs treated for 4 h induced significantly lower luciferase levels than did unlayered LNPs, which increased to much higher relative levels after 24 h treatment, suggesting that this polyanion may direct trafficking in a manner that leads to slower mRNA delivery. Conversely, PAA-LLNPs after 4-h treatment generated comparable luciferase expression to unlayered LNPs; however, after 24-h treatment, PAA-LLNPs showed reduced luciferase levels relative to unlayered LNPs, suggesting that this layer may undergo trafficking that leads to either rapid uptake and clearance, degradation, or endosomal trapping within the first hour. mRNA delivery kinetics varied by cell type; in Jurkats, for example, PLD- and PLE-LLNPs treated for 4 h exhibited an increased luciferase expression over unlayered LNPs; these differences among groups decreased after 24 h treatment. In RAW 264.7 s, HA-LLNPs treated for 4 h transfected comparably to unlayered LNPs, but transfection was greatly reduced relative to other groups at 24 h. Trends in LLNP mRNA transfection were also assessed in two cancer cell lines via flow cytometry, using LNP cores of the same lipid composition, containing EGFP-expressing mRNA (SI Appendix, Fig. S3). LOXIMVI melanoma cells exhibited similar trends in LLNP transfection; HepG2 liver carcinoma cells exhibited less specificity for polyanion outer layers.

To assess the effect of layering on a different core, HA, PLE, and PAA were layered onto LNP formulations in a 50/38.5/1.5/10 ratio of DLin-MC3-DMA/cholesterol/DMG-PEG-2000/DSPC. Similar trends as those seen with the original cores were observed at 4-h and 24-h treatments across cell lines (SI Appendix, Fig. S4). These data suggest that the LNP surface chemistry and resulting interactions are influenced by the polyanion outer layer coating, beyond the lipid composition.

To characterize LNP cellular association, cells were treated with LLNPs containing 0.5 mol % Cy5-labeled DOPE for either 4 h or 24 h; cellular Cy5 fluorescence was measured on a Tecan plate reader (Fig. 2 C and D). Notably, HA-LLNPs generated the highest Cy5 signal across cell lines at both incubations. PAA-LLNPs associated least across cell lines that similarly demonstrated reduced luciferase expression. Interestingly, though PLD-LLNPs generated the greatest luciferase expression, they associated moderately across cell lines. The discrepancy between cell association or uptake and transfection trends suggests that polyanions may further modulate the internal trafficking and RNA release.

To test the impact of polyanion adsorption on transfection of other nucleic acid cargos, we formulated ALC-0315 LLNPs containing GFP-encoding pDNA, layered with each of the four polyanions. pDNA delivery requires different intracellular release and trafficking profiles than mRNA delivery, since the former requires nuclear translocation for effective transfection. HEK293T, EL4, and HepG2 liver carcinoma cells were treated with LNPs for 4 h, then washed and cultured for an additional 20 h; GFP-positive cells were assessed via flow cytometry (Fig. 2E). Plasmid delivery across cell lines was enhanced most notably by HA-LLNPs, which more than doubled transfection rates. HA has previously improved lipoplex-mediated pDNA transfection, potentially due to intracellular interactions associated with its glycosaminoglycan structure (25). PLD- and PLE-LLNPs decreased pDNA-LLNP transfection in both EL4s and HEK293Ts. Transfection rates in HepG2s, which divide and intake material rapidly, were not significantly impacted by PAA-, PLD-, or PLE-LLNPs.

Intracellularly, LLNP Polyanions Traffic with pDNA Cargo.

To examine the impact of the polyanion on intracellular trafficking of pDNA LLNPs, confocal microscopy was conducted on EL4s and HEK293Ts treated with unlayered or layered LNPs for 4 h. These cell lines were chosen as representative adherent and suspension cultures. To facilitate co-tracking of lipid, nucleic acid, and polyanion, DOPE-Cy5 labeled LNPs were loaded with MFP488-labeled pDNA and layered with BDP 558/568-labeled polyanions.

After 4 h treatment, EL4s internalized both unlayered and layered LNPs; polyanions were taken up along with core lipids and pDNA (Fig. 3A). Across all groups, MFP488-pDNA signal appeared diffuse throughout the cytoplasm. EL4s treated with unlayered and HA-LLNPs additionally displayed some punctate pDNA signals co-localized with lipid signals; these NPs may be trapped in trafficking endosomes or lysosomes. pDNA nuclear localization was minimal, expected given the low rates of pDNA transfection in these cells. Though all groups exhibited some pDNA colocalization with the cell membrane, colocalization appeared strongest in cells treated with PLE- and PAA-LLNPs. In past studies, PLE has demonstrated localization to extracellular surfaces in cancer cell lines; similar cellular interactions may promote extracellular localization in EL4 lymphoma cells (22, 39). DOPE signal appeared punctate within the cytoplasm and cell membrane. The detected DOPE signal was significantly weaker than that of pDNA or polyanion; this may partially be due to the relative infrequency of labeled DOPE molecules in NP composition. Polyanion signal in LLNPs appeared diffuse in the cytoplasm, similar to pDNA dispersal. The relative positions of all three components suggest that polyanions and pDNA translocate through the cell in more similar patterns than do LNP lipids (Fig. 3 B and C). HEK293Ts exhibited similar internalization patterns; in addition, the three labeled LNP components were visualized trafficking between cells (SI Appendix, Fig. S5).

Fig. 3.

Fig. 3.

LLNP polyanions are internalized and co-traffic with LLNP cargo. (A) Confocal microscopy images of EL4 cells treated with unlayered or layered LNPs containing DOPE-Cy5, MFP488-labeled pDNA, and BDP-labeled polyanions. Cell membranes were visualized by staining cells with wheat germ agglutinin, AF405 conjugate. Images shown are representative z-slices. Imaged on Olympus FV1200, 100× magnification, 4 h after treatment. Size bars represent 10 microns. (B) Manders’ overlap coefficients of LNP DNA co-localization with polyanion. (C) Manders’ overlap coefficients of LNP DNA co-localization with Cy5 DOPE.

To further assess the mechanisms of uptake and intracellular trafficking of LLNPs, EL4s were treated with unlayered or layered LNPs with MFP488-labeled pDNA and BDP555/568-labeled polyanions. In this study, HA was not labeled. After either 4 or 24 h, cells were stained for each of clathrin, caveolin, or lysosomes, then imaged (SI Appendix, Figs. S6–S12). At 4 h, HA-, PLE-, and PAA-LLNP DNA localized more with clathrin markers than with caveolin; minimal localization was observed with caveolin (SI Appendix, Figs. S6, S7, and S9). In comparison, cells treated with unlayered LNPs showed variable degrees of co-localization of LNP DNA with clathrin; co-localization with caveolin was minimal. Prior work has observed clathrin-dependent uptake of HA-layered NPs; by contrast, LNPs are thought to traffic by macropinocytosis (22).

Quantification of co-localization of LNP DNA and LAMP1 at 4 h suggested a trend that LLNP cargo is less associated with lysosomes than unlayered LNP cargo, though statistical significance was not achieved (SI Appendix, Figs. S8 and S9). Interactions between the surface polyanion and cellular surface markers therefore modulate uptake rates and mechanisms.

PLE and HA Moderately Improve Hepatic and Splenic Transfection.

Next, we evaluated the impact of the surface polyanion on LNP biodistribution and transfection in healthy female C57BL/6 mice injected intravenously (i.v.). LNPs with each of the four polyanions induced no toxicity 72 h after dosing, as measured by liver enzymes, serum protein, and weight changes (SI Appendix, Fig. S13) (40, 41). Lower blood urea nitrogen (BUN) levels across groups, relative to literature-determined ranges, may be attributed to variability in vendor strains; however, no significant difference in BUN levels was observed between mice treated with LLNPs and mice treated with saline.

To study biodistribution and transfection, mice were dosed with unlayered and layered ALC-0315 LNPs at 0.3 mg/kg luciferase-encoding mRNA. Four hours after injection, the heart, lungs, liver, kidneys, and spleen were imaged for luciferase expression ex vivo. PLE-LLNPs increased liver and spleen luciferase expression 1.8-fold and 2.5-fold, respectively, over unlayered LNPs (Fig. 4A). HA-LLNPs increased spleen luciferase expression 2.3-fold over unlayered LNPs; however, this formulation did not increase liver transfection (Fig. 4A). In contrast to in vitro trends, PLD-LLNPs showed minimally improved or decreased transfection across organs; PAA-LLNPs performed similarly to unlayered LNPs in all organs studied. No significant differences from unlayered LNPs were detected in the heart or lungs for any polyanion. Comparing the ratio of weight-normalized flux in the spleen and liver, we observed that HA-LLNPs showed higher preferential localization to the spleen than did other layers (Fig. 4 B and E). In comparison, PLE-LLNPs increased expression in both liver and spleen.

Fig. 4.

Fig. 4.

LLNPs modulate transfection and biodistribution in vivo. (A) Luciferase activity in liver, spleen, and lungs of C57BL/6 mice 4 h after treatment with 0.3 mg/kg ALC-0315 mRNA-LLNPs. Flux was normalized by organ weight. N = 5; N = 4 for HA-LLNPs. (B) Ratio of weight-normalized luciferase signals in spleen and liver, 4 h after injection. N = 5; N = 4 for HA-LLNPs. (C) Weight-normalized luciferase activity in liver, spleen, and lungs of C57BL/6 mice 24 h after treatment with 0.3 mg/kg ALC-0315 mRNA-LLNPs. N = 5. (D) Ratio of weight-normalized luciferase signals in spleen and liver, 24 h after injection. N = 5. (E) Ex vivo representative images of luminescence in the heart, lungs, spleen, kidneys, and liver of C57BL/6 mice 4 h after treatment with 0.3 mg/kg ALC-0315 mRNA-LLNPs. (F) Ex vivo representative images of luminescence in the heart, lungs, spleen, kidneys, and liver of C57BL/6 mice 24 h after treatment with 0.3 mg/kg ALC-0315 mRNA-LLNPs. Data represented as mean ± SD. Ordinary one-way ANOVA, Tukey’s test post hoc. ****P < 0.0001, ***P < 0.001, **P < 0.01, *P < 0.05.

For a longer-term biodistribution study, we analyzed organs for luciferase expression 24 h after transfection. At 24 h, HA-LLNPs showed reduced luciferase expression across all measured organs (Fig. 4 C and F). All other LLNPs performed similarly to unlayered LNPs, suggesting that the outer polyanion confers the greatest benefit at earlier times (Fig. 4 C and D and SI Appendix, Fig. S14). Differences in transfection of LLNPs decrease over time, potentially due to stripping or degradation of the outer layer over time.

To compare trends in in vivo transfection across cores, 4-h organ-level transfection studies were conducted with LLNPs in which the core was composed of a 50/38.5/1.5/10 ratio of DLin-MC3-DMA/cholesterol/DMG-PEG-2000/DSPC (SI Appendix, Fig. S7); PLD was excluded given minimal differences between PLD-LLNPs and unlayered LNPs in prior studies. Similar trends were observed by swapping the LLNP core; however, most notably, HA-LLNPs with the alternate ionizable lipid decreased spleen luciferase expression 2.3-fold relative to unlayered LNPs (SI Appendix, Fig. S15). The prominent difference in splenic performance may be attributed to synergistic impacts of both the polyanion and core helper lipids, though additional studies are required to isolate the impact of each component.

To evaluate the pharmacokinetics in vivo, mice were dosed i.v. with Cy5-labeled LNPs. Blood retention times were evaluated over a 24-h period post-administration by quantifying the Cy5 signal in the blood serum (SI Appendix, Fig. S16). Minimal differences in residence time were observed across LLNPs.

Discussion

In this work, we describe a method to stably deposit charged bioactive polymers onto LNP surfaces, forming LLNPs, as a tunable modality to alter the NP’s surface presentation and cellular interactions both in vitro and in vivo. Polyanion chemistries can modulate selectivity in LNP uptake and transfection, helping to address the ongoing challenge of targeted nucleic acid delivery.

The LbL assembly enables the incorporation of a wide variety of charged polymers in a modular fashion; many types of polymers can be stably layered onto a diverse range of LNP cores varying in component lipids and nucleic acid cargos. Relative to lipid-centered LNP development, which requires the synthesis of novel compounds and iterative optimization of LNP formulations (15, 16), the electrostatic adsorption of surface polymers can be easily applied to already-optimized LNP cores. Indeed, an optimized LNP delivery vector may benefit significantly from the combination of a highly efficacious ionizable lipid to boost transfection and a rationally chosen surface polyanion to provide on-target selectivity.

Polyanion chemistry impacts LNP apparent pKa and colloidal stability, while maintaining LNP structure and encapsulation of nucleic acid cargos (Fig. 1). Furthermore, a stably adsorbed layer of polyanion helps ensure colloidal stability without incorporating additional PEG and PEGylated compounds; the polyanion may also shield LNP PEG-chains from generating or reacting with human PEG antibodies (11). While recent works have conjugated active targeting elements, such as antibodies, peptides, or even polymers, to LNP surfaces (4246), the LbL assembly allows the noncovalent attachment of polyanions capable of inherent targeting and specificity. In addition, LbL assembly enables the facile incorporation of diverse chemistries onto LNP surfaces. Rational choice of polyanions, which include many known biomolecules such as polysaccharides and polypeptides, can leverage cell-specific receptor expression to improve the specificity of NP association. In conjunction with this capability to choose a layer with specific cell interactions, the strongly negative charge and hydrated brushy nature of the polyanion outer layers on LLNPs reduce nonspecific protein adsorption that can lead to undesired uptake and transfection of off-target cells and tissues (Fig. 1H). For example, HA is a natural ligand for CD44, a receptor overexpressed by endothelial cells, macrophages, and certain cancer cells (31, 47). LLNPs with adsorbed HA showed higher luciferase signal and Cy5 uptake in RAW 264.7 macrophages, known to express CD44, than other LLNPs (Fig. 2 A and C) (47, 48). Moreover, at 4-h treatment, other cell lines showed significantly reduced transfection by HA-LLNPs. This selectively increased uptake can enable delivery specific to cell phenotypes expressing natural binding sites for HA; for example, HA-conjugated LNPs have been used for targeting of ovarian and glioblastoma multiforme (GBM) cancer cells (43, 44). Both HA and PLD have also been adsorbed onto liposomal NPs to improve uptake by ovarian cancer cells in murine models (22, 23, 39). Given that these polyanions have multiple disease-relevant biological binding partners, LbL assembly may enable future LNP targeting strategies for cancer or immunotherapy applications without the need for bioconjugation of additional ligands. By contrast, the relatively low uptake and transfection that was observed with PAA-LLNPs may arise because PAA is a bioinert polymer that may not benefit from receptor-mediated uptake mechanisms. These properties may make PAA a desirable “stealth” layer to minimize off-target uptake, for cases in which an additional ligand is utilized.

Prior studies have demonstrated that LNPs with acidified apparent pKa negative surface charge preferentially localize to the spleen over the liver; the opposite trends induce preferential localization to the lungs (18, 19, 38). In this study, PLE-, PLD-, and PAA-LLNPs all and exhibited acidified pKa and negative charges of similar magnitudes; notably, only PLE-LLNPs improved splenic transfection. Variability in localization and transfection may be attributed to affinities between the outer polyanion and receptors of cells of particular phenotypes. This discrepancy also suggests that polyanion chemistry and nano-bio interactions are key in governing LLNP cellular interactions. PLE- and HA-LLNPs showed improved LLNP luciferase signal 4 h after injection but did not perform similarly 24 h after injection. Cellular subtype profiling may indicate whether drops in luciferase signal are due to the metabolic rate of the cell subtypes primarily transfected by these NPs. Different polymer coatings have been shown to uniquely modify the protein corona of polymeric NPs, which may impact uptake and cell phenotype selectivity (49). Furthermore, prior work has demonstrated that the polyanion identity impacts intracellular trafficking and lysosomal clearance pathways (22).

The use of electrostatics to decorate NP surfaces is a powerful tool that can be generalized to facilitate incorporation of other therapeutic or targeting moieties into LNP delivery systems. In prior work, the LbL technique has been applied to electrostatically lipid-based nanocarrier surfaces with targeting moieties, such as charged peptides, and therapeutics, such as small molecules or proteins (4951). The four polyanions in this study were chosen for their demonstrated bioactivity and cell specificity (20, 22, 23, 26, 32, 39). However, future LLNP libraries can incorporate different polyanion chemistries and charged groups such as sulfonates or phosphonates, which may vary in impact on transfection and biodistribution (20, 22, 25). We anticipate that the LNP layering method described in this work can be adapted to incorporate additional targeting and therapeutic modalities into these highly potent delivery vehicles, to improve on-target and selective uptake and transfection, deliver combination therapies, and harness receptor-mediated interactions to boost transfection of difficult cell types.

Materials and Methods

Materials.

Dilinoleylmethyl-4-dimethylaminobutyrate (DLin-MC3-DMA), N,N-dimethyl-2,2-di-(9Z,12Z)-9,12-octadecadien-1-yl-1,3-dioxolane-4-ethanamine (DLin-KC2-DMA), and 1,1′-((2-(4-(2-((2-(bis(2-hydroxydodecyl)amino) ethyl) (2-hydroxydodecyl)amino)ethyl)piperazin-1-yl)ethyl)azanediyl)bis(dodecan-2-ol) (C12-200) were purchased from MedChemExpress. 1,2-dimyristoyl-rac-glycero-3-methoxypolyethylene glycol-2000 (DMG-PEG-2000) and 3,6-bis[4-[bis (2-hydroxydodecyl)amino]butyl]-2,5-piperazinedione (cKK-E12) was purchased from Cayman Chemical Company. Cholesterol, 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (18:1 (Δ9-Cis) PC), 1,2-dioleyol-sn-glycero-3-phosphoethanolamine (18:1 (Δ9-Cis) PE), 1,2-dioleyol-sn-glycero-3-phosphoethanolamine-N-(Cyanine-5) (18:1 Cy5 PE), and 6-((2-hexyldecanoyl)oxy)-N-(6-((2-hexyldecanoyl)oxy)hexyl)-N-(4-hydroxybutyl)hexan-1-aminium (ALC-0315) were purchased from Avanti Polar Lipids.

CleanCap Firefly Luciferase mRNA (5-methoxyuridine) was purchased from TriLink Biotechnologies. The Label IT Nucleic Acid Labeling Kit (MFP488) was purchased from Mirus Bio.

Sodium hyaluronate was purchased from Lifecore Technologies. Polyacrylic acid, sodium salt (PAA, 15 kDa) solution, N-hydroxysuccinimide ester (NHS), and N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (EDC) were purchased from Sigma Aldrich. Poly-L-glutamic acid sodium salt (PLE100, 15 kDa) and poly-L-aspartic acid, sodium salt (PLD100, 14 kDa) were purchased from Alamanda Polymers. MWCO dialysis membranes were purchased from Repligen. BDP-558/568-amine was purchased from Lumiprobe.

Methods.

Cell culture.

Cell lines used for this study included: HEK293Ts, EL4s, RAW 264.7, HepG2, LOXIMVI, Jurkat. HEK293T, EL4, and RAW 264.7 cells were purchased from ATCC. LOXIMVI and Jurkat cells were a gift from the Straehla Lab. HepG2 cells were sourced from the Koch Institute’s High Throughout Sciences Facility. HEK293T, EL4, and RAW 264.7 s were cultured in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin (Pen-Strep). HepG2s were cultured in Eagle’s Minimum Essential Medium (EMEM) supplemented with 10% FBS. Jurkats and LOXIMVIs were cultured in Roswell Park Memorial Institute (RPMI) media supplemented with 10% FBS, 1% Pen-Strep. All cells were cultured in a humidified chamber at 37 °C and 5% CO2 and handled in sterile conditions. Cell cultures were tested for mycoplasma contamination using a MycoAlert kit (Lonza), in the High Throughput Sciences core and the ES Cell and Transgenics Facility at MIT’s Koch Institute for Integrative Cancer Research. All cell lines were used up to passage 20.

LNP formation.

DSPC, DOPC, DOPE, and DOPE-Cy5 were dried from chloroform stocks under nitrogen and then dissolved in 100% ethanol. Cholesterol, DLin-MC3-DMA, DLin-KC2-DMA, and DMG-PEG-2000 were directly dissolved in 100% ethanol. LNP lipid stocks were formed at varying molar compositions (SI Appendix, Table S1).

Nucleic acid cargo (either mRNA or pDNA) was dissolved in 25 mM sodium acetate in a 4-mL scintillation vial. To 4 volumes of nucleic acid under stirring, 1 volume of lipid mixture in 100% ethanol was pipetted in rapidly. The solution was rested without stirring for 5 min. Then, under resumed stirring, the solution was diluted with 5 volumes of DNAse/RNAse-free water. Ethanol and sodium acetate were removed via dialysis against either water or 1× PBS for 4 h. LNPs were concentrated using Amicon Ultra-4 ultracentrifugal filter units, MWCO 100 K. LNP solutions were stored at 4 °C and used within 3 d of preparation.

LNP characterization.

LNP hydrodynamic diameter, polydispersity index, and zeta-potential were measured on a Malvern Zetasizer Pro (Malvern Panalytical) with a red laser and a detection angle of 173°.

LNP encapsulation efficiency.

Encapsulation efficiency of mRNA and pDNA was determined with the Quant-it RiboGreen RNA assay kit and PicoGreen dsDNA assay kit, respectively (Fisher Scientific). In a Nunc F96 MicroWell Black polystyrene plate, 5 μL of LNP samples were incubated in 45 μL of either 1× TE or 0.5% (v/v) Triton X-100 solution in 1× TE. Samples were shaken at 130 rpm, at 37 °C, for 10 min. RiboGreen reagent was diluted 200-fold in 1× TE and protected from light. Samples were then mixed with 50 μL of the diluted RiboGreen reagent. Then, samples were shaken at 300 rpm at room temperature (RT) for 5 min, protected from light. Fluorescence intensities were read immediately on a Tecan M1000 plate reader, at an excitation of 485 nm and emission of 525 nm. Encapsulation was calculated as (Fluorescence of Triton X-100 LNPs−Fluorescence of TE LNPs)/(Fluorescence of Triton X-100 LNPs).

LNP layering.

Polyanions were dissolved in nuclease-free water to concentrations of 10 to 20 mg/mL, then sonicated for 10 min. Layering baths were prepared by diluting polyanion stocks into 5 mM HEPES buffer. Stocks of HA, PLD, PLE, and PAA were diluted to 8, 2, 2, and 1.2 respective weight equivalents, relative to LNP lipid concentration. The polyanion bath was added to a 4-mL scintillation vial and stirred at 800 rpm at RT, to which an equal volume of LNPs in water was added. The mixture was stirred for 15 min and then incubated at RT for 1 h. To purify and concentrate layered LNPs, nonbound soluble polyanions were removed using filtration. The layered LNPs were then isolated and concentrated via three water washes with Amicon Ultra-4 ultracentrifugal filter units, MWCO 100 K. Layered LNPs were stored at 4 °C.

Dye-labeled polymer synthesis.

Carboxylated polymers were fluorescently labeled with BDP 558/568-amine by NHS/EDC coupling chemistry. Polymers (HA, PLD, PLE, or PAA) were diluted to 2 mg/mL in MES buffer (0.1 N, pH 6). Then, solutions of BDP 558/568-amine in DMSO and of NHS and EDC in MES buffer, were added at a concentration 6.3-fold (0.63-fold for PLE), 14.3-fold, and 143-fold times higher than the polymer, respectively. The solution was stirred for 4 h at RT, then dialyzed against NaCl 50 mM overnight and against ultra-pure water for up to 72 h to remove free reagents. A 3.5 KDa regenerated cellulose membrane was used for dialysis (Spectrum Labs). The final polymer solution was frozen at −80 °C and lyophilized in a Labconco FreeZone Freeze Dryer System.

Characterization of plasma proteins adsorbed to LLNPs.

Adsorbed serum proteins on LLNPs were characterized using a previously described method (38). Unlayered or layered mRNA-LNPs were diluted to 0.4 mg/mL lipid. Characterized adsorbed serum proteins, unlayered or layered mRNA-LNPs containing DOPE-Cy5 were incubated in an equal volume of 100% mouse plasma for 1 h at 37 °C. The solution was then centrifuged at 25,000 rcf at 4 °C for 2 h. The supernatant was removed, and the pellet was washed with nuclease-free water. The pellet was washed twice in water via centrifugation at 25,000 rcf at 4 °C for 2 h, then resuspended in 50 μL water. A volume of 5 μL of each pellet was diluted into 95 μL DMSO in a black 96-well flat-bottom plate, and Cy5 fluorescence was read on a Tecan M1000 plate reader (Ex/Em 630/670 nm). Pellets were diluted to equal Cy5 concentrations. The concentration of protein in each pellet was determined using the Pierce BCA Protein Assay Kit; standard curves for the BCA assay were prepared using Cy5-mRNA LNPs as background.

Transfection studies.

Adherent cells were seeded in 96-well flat-bottom plates at 10,000 cells/well in 90 μL of supplemented cell media and allowed to grow at 37 °C, 5% CO2, for 24 h prior to treatment with LNPs. Suspension cells were seeded at 25,000 cells/well in 90 μL of complete cell media and were treated immediately with LNPs. For 24 incubations, cells were not washed. For 4-h incubations, cells were washed and re-suspended in fresh media 4 h after transfection, then incubated for an additional 20 h before readout.

Luciferase and cell viability assay.

Adherent cells were washed in 100 μL 1× PBS, treated with 30 μL 0.25% Trypsin-EDTA for 5 min, diluted with 150 μL complete media, washed, and resuspended in complete media. Suspension cells were washed in 100 μL 1× PBS and then resuspended in complete media.

Half of the cell volume was plated in a black 96-well flat-bottom plate and diluted to 100 µL per well with complete media. A volume of 10 µL of PrestoBlue viability reagent was added to each well. Samples were incubated for 30 min at 37 °C, 5% CO2, protected from light. PrestoBlue fluorescence was read at an excitation of 560 nm and emission of 600 nm on a Tecan M1000 plate reader.

The remaining half of the cell volume was plated in a clear 96-well plate, washed in 1× PBS, then lysed with 1× Lysis Buffer for 15 min at room temperature while shaking. Then, 20 µL of cell lysate was plated into a white flat-bottom 96-well plate and mixed with 100 µL of 0.2 mg/mL D-luciferin, diluted in Biotium Firefly Luciferase Assay Buffer 2.0. Immediately after adding D-luciferin, luminescence was read on a Tecan M1000 plate reader, using an integration time of 1,000 ms. Background signal from untreated cells was subtracted from all luminescence values.

Animal experiments.

All animal experiments were approved by the MIT Committee on Animal Care (protocol #221000434). Additional details on animal experimental procedures can be found in SI Appendix.

Statistical analysis.

Statistics were calculated using GraphPad Prism. Multiple comparisons among groups were analyzed using a one-way ANOVA test, with Tukey’s multiple comparisons post hoc test. Differences were considered statistically significant if the calculated P-value was less than 0.05. All data are presented as a mean value with SD (mean ± SD).

Supplementary Material

Appendix 01 (PDF)

Acknowledgments

This work was supported, in part, by the Bill and Melinda Gates Foundation (grant ID INV-021791 and INV-050202). Under the grant conditions of the Foundation, a Creative Commons Attribution 4.0 Generic License has already been assigned to the Author Accepted Manuscript version that might arise from this submission. All data underlying the results are available as part of the article and no additional source data are required. This material is based on work supported by the NSF Graduate Research Fellowship Program under Grant No. 2141064. Any opinions, findings, and conclusions or recommendations expressed in this material are those of the authors and do not necessarily reflect the views of the NSF. G.C. is supported by the VHL Alliance and NIH (F99CA274651). We would like to thank the Koch Institute's Robert A. Swanson (1969) Biotechnology Center for technical support, specifically the Flow Cytometry Core, Microscopy Core, Nanotechnology and Nanomaterials core, High Throughput Sciences core, and the ES Cell and Transgenics Facility. We would also like to thank the Department of Comparative Medicine at the Massachusetts Institute of Technology. Fig. 1 was partially created using https://www.Biorender.com and ChemDraw. Prism software was used for statistical analysis and data visualization. We would additionally like to thank Dr. Margaret Billingsley, Adam Berger, and Victoria Gomerdinger for their help in reviewing this manuscript.

Author contributions

N.N., T.G.D., and G.C. designed research; N.N., T.G.D., G.C., and D.R.-C. performed research; N.N. contributed new reagents/analytic tools; N.N. and T.G.D. analyzed data; and N.N. and P.T.H. wrote the paper.

Competing interests

P.T.H. is the co-founder and member of the Board of LayerBio, Inc., a member of the Board of Alector Therapeutics, Focal Biomedical and the Board of Senda Biosciences, a Flagship company, and a former member of the Scientific Advisory Board of Moderna Therapeutics.

Footnotes

This article is a PNAS Direct Submission. J.D. is a guest editor invited by the Editorial Board.

Data, Materials, and Software Availability

Relevant data has been deposited on Figshare (DOI: 10.6084/m9.figshare.24970335) (52). All other data are included in the article and/or SI Appendix.

Supporting Information

References

  • 1.Kulkarni J. A., et al. , The current landscape of nucleic acid therapeutics. Nat. Nanotechnol. 16, 630–643 (2021). [DOI] [PubMed] [Google Scholar]
  • 2.Cullis P. R., Hope M. J., Lipid nanoparticle systems for enabling gene therapies. Mol. Therapy 25, 1467–1475 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Doudna J. A., Charpentier E., The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014). [DOI] [PubMed] [Google Scholar]
  • 4.Frangoul H., et al. , CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. N Engl. J. Med. 384, 252–260 (2021). [DOI] [PubMed] [Google Scholar]
  • 5.Schwank G., et al. , Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem. Cell 13, 653–658 (2013). [DOI] [PubMed] [Google Scholar]
  • 6.Ohmori T., et al. , CRISPR/Cas9-mediated genome editing via postnatal administration of AAV vector cures haemophilia B mice. Sci. Rep. 7, 4159 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kulkarni J. A., Cullis P. R., Van Der Meel R., Lipid nanoparticles enabling gene therapies: From concepts to clinical utility. Nucleic Acid Ther. 28, 146–157 (2018). [DOI] [PubMed] [Google Scholar]
  • 8.Whitehead K. A., Langer R., Anderson D. G., Knocking down barriers: Advances in siRNA delivery. Nat. Rev. Drug. Discov. 8, 129–138 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hoy S. M., Patisiran: First global approval. Drugs 78, 1625–1631 (2018). [DOI] [PubMed] [Google Scholar]
  • 10.Samaridou E., Heyes J., Lutwyche P., Lipid nanoparticles for nucleic acid delivery: Current perspectives. Adv. Drug. Deliv. Rev. 154–155, 37–63 (2020). [DOI] [PubMed] [Google Scholar]
  • 11.Hald Albertsen C., et al. , The role of lipid components in lipid nanoparticles for vaccines and gene therapy. Adv. Drug. Deliv. Rev. 188, 114416 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Tsoi K. M., et al. , Mechanism of hard-nanomaterial clearance by the liver. Nat. Mater. 15, 1212–1221 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Poon W., et al. , Elimination pathways of nanoparticles. ACS Nano 13, 5785–5798 (2019). [DOI] [PubMed] [Google Scholar]
  • 14.Jayaram D. T., Pustulka S. M., Mannino R. G., Lam W. A., Payne C. K., Protein corona in response to flow: Effect on protein concentration and structure. Biophys. J. 115, 209–216 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Qiu M., et al. , Lung-selective mRNA delivery of synthetic lipid nanoparticles for the treatment of pulmonary lymphangioleiomyomatosis. Proc. Natl. Acad. Sci. U.S.A. 119, e2116271119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.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, 1606944 (2017). [DOI] [PubMed] [Google Scholar]
  • 17.Li B., et al. , Combinatorial design of nanoparticles for pulmonary mRNA delivery and genome editing. Nat. Biotechnol. 41, 1410–1415 (2023), 10.1038/s41587-023-01679-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.LoPresti S. T., Arral M. L., Chaudhary N., Whitehead K. A., The replacement of helper lipids with charged alternatives in lipid nanoparticles facilitates targeted mRNA delivery to the spleen and lungs. J. Controll. Release 345, 819–831 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Morton S. W., Poon Z., Hammond P. T., The architecture and biological performance of drug-loaded LbL nanoparticles. Biomaterials 34, 5328–5335 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Correa S., Boehnke N., Deiss-Yehiely E., Hammond P. T., Solution conditions tune and optimize loading of therapeutic polyelectrolytes into layer-by-layer functionalized liposomes. ACS Nano 13, 5623–5634 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Correa S., et al. , Tuning nanoparticle interactions with ovarian cancer through layer-by-layer modification of surface chemistry. ACS Nano 14, 2224–2237 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kong S., et al. , Synergistic combination therapy delivered via layer-by-layer nanoparticles induces solid tumor regression of ovarian cancer. Bioeng. Transl. Med. 8, e10429 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Choi K. Y., et al. , Binary targeting of siRNA to hematologic cancer cells in vivo using layer-by-layer nanoparticles. Adv. Funct. Mater. 29, 1900018 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ruponen M., et al. , Extracellular glycosaminoglycans modify cellular trafficking of lipoplexes and polyplexes. J. Biol. Chem. 276, 33875–33880 (2001). [DOI] [PubMed] [Google Scholar]
  • 26.Deng Z. J., et al. , Layer-by-layer nanoparticles for systemic codelivery of an anticancer drug and siRNA for potential triple-negative breast cancer treatment. ACS Nano 7, 9571–9584 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.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). [DOI] [PubMed] [Google Scholar]
  • 28.Semple S. C., et al. , Rational design of cationic lipids for siRNA delivery. Nat. Biotechnol. 28, 172–176 (2010). [DOI] [PubMed] [Google Scholar]
  • 29.Dong Y., et al. , Lipopeptide nanoparticles for potent and selective siRNA delivery in rodents and nonhuman primates. Proc. Natl. Acad. Sci. U.S.A. 111, 3955–3960 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kulkarni J. A., et al. , Design of lipid nanoparticles for in vitro and in vivo delivery of plasmid DNA. Nanomedicine 13, 1377–1387 (2017). [DOI] [PubMed] [Google Scholar]
  • 31.Deiss-Yehiely E., et al. , Surface presentation of hyaluronic acid modulates nanoparticle-cell association. Bioconjug. Chem. 33, 2065–2075 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Dreaden E. C., et al. , Bimodal tumor-targeting from microenvironment responsive hyaluronan layer-by-layer (LbL) nanoparticles. ACS Nano 8, 8374–8382 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Patel P., Ibrahim N. M., Cheng K., The importance of apparent pKa in the development of nanoparticles encapsulating siRNA and mRNA. Trends Pharmacol. Sci. 42, 448–460 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.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). [DOI] [PubMed] [Google Scholar]
  • 36.Mero A., Campisi M., Hyaluronic acid bioconjugates for the delivery of bioactive molecules. Polymers (Basel) 6, 346–369 (2014). [Google Scholar]
  • 37.Chen M., Gupta V., Anselmo A. C., Muraski J. A., Mitragotri S., Topical delivery of hyaluronic acid into skin using SPACE-peptide carriers. J. Controll. Release 173, 67–74 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dilliard S. A., Cheng Q., Siegwart D. J., On the mechanism of tissue-specific mRNA delivery by selective organ targeting nanoparticles. Proc. Natl. Acad. Sci. U.S.A. 118, e2109256118 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Barberio A. E., et al. , Cancer cell coating nanoparticles for optimal tumor-specific cytokine delivery. ACS Nano 14, 11238–11253 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Mazzaccara C., et al. , Age-related reference intervals of the main biochemical and hematological parameters in C57BL/6J, 129SV/EV and C3H/HeJ mouse strains. PLoS One 3, e3772 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Otto G. P., et al. , Clinical chemistry reference intervals for C57BL/6J, C57BL/6N, and C3HeB/FeJ mice (Mus musculus). J. Am. Assoc. Lab Anim. Sci. 55, 375–386 (2016). [PMC free article] [PubMed] [Google Scholar]
  • 42.Anthiya S., et al. , Targeted siRNA lipid nanoparticles for the treatment of KRAS-mutant tumors. J. Control. Release 357, 67–83 (2023). [DOI] [PubMed] [Google Scholar]
  • 43.Cohen Z. R., et al. , Localized RNAi therapeutics of chemoresistant grade IV glioma using hyaluronan-grafted lipid-based nanoparticles. ACS Nano 9, 1581–1591 (2015). [DOI] [PubMed] [Google Scholar]
  • 44.Singh M. S., et al. , Therapeutic gene silencing using targeted lipid nanoparticles in metastatic ovarian cancer. Small 17, 2100287 (2021). [DOI] [PubMed] [Google Scholar]
  • 45.Kheirolomoom A., et al. , In situ T-cell transfection by anti-CD3-conjugated lipid nanoparticles leads to T-cell activation, migration, and phenotypic shift. Biomaterials 281, 121339 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Parhiz H., et al. , PECAM-1 directed re-targeting of exogenous mRNA providing two orders of magnitude enhancement of vascular delivery and expression in lungs independent of apolipoprotein E-mediated uptake. J. Control. Release 291, 106–115 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lesley J., Hyaluronan binding by cell surface CD44. J. Biol. Chem. 275, 26967–26975 (2000), 10.1074/jbc.M002527200. [DOI] [PubMed] [Google Scholar]
  • 48.Krejcova D., Pekarova M., Safrankova B., Kubala L., The effect of different molecular weight hyaluronan on macrophage physiology. Neuro. Endocrinol. Lett. 30, 106–111 (2009). [PubMed] [Google Scholar]
  • 49.Boehnke N., Dolph K. J., Juarez V. M., Lanoha J. M., Hammond P. T., Electrostatic conjugation of nanoparticle surfaces with functional peptide motifs. Bioconjug. Chem. 31, 2211–2219 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Boehnke N., et al. , Theranostic layer-by-layer nanoparticles for simultaneous tumor detection and gene silencing. Angew. Chem. 132, 2798–2805 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Straehla J. P., et al. , A predictive microfluidic model of human glioblastoma to assess trafficking of blood–brain barrier-penetrant nanoparticles. Proc. Natl. Acad. Sci. U.S.A. 119, e2118697119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Nabar N., Dacoba T., Covarrubias G., Supplemental data to “Electrostatic adsorption of polyanions onto lipid nanoparticles controls uptake, trafficking, and transfection of RNA and DNA therapies”. FigShare. 10.6084/m9.figshare.24970335. Deposited 9 January 2024. [DOI] [PMC free article] [PubMed]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Appendix 01 (PDF)

Data Availability Statement

Relevant data has been deposited on Figshare (DOI: 10.6084/m9.figshare.24970335) (52). All other data are included in the article and/or SI Appendix.


Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES