You are on page 1of 10

On the mechanism of tissue-specific mRNA delivery

by selective organ targeting nanoparticles


Sean A. Dilliarda , Qiang Chenga,1 , and Daniel J. Siegwarta,1
a
Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390

Edited by Joseph DeSimone, Department of Chemical Engineering, Stanford University, Stanford, CA; received May 19, 2021; accepted November 8, 2021

Lipid nanoparticles (LNPs) are a clinically mature technology for extrahepatic delivery of mRNA. The SORT strategy was gener-
the delivery of genetic medicines but have limited therapeutic alizable to multiple classes of LNPs and SORT molecules.
applications due to liver accumulation. Recently, our laboratory Lung-, liver-, and spleen-targeting SORT LNPs could deliver
developed selective organ targeting (SORT) nanoparticles that diverse cargoes (nucleic acids and proteins) to achieve gene
expand the therapeutic applications of genetic medicines by expression and CRISPR/Cas-based gene editing in therapeuti-
enabling delivery of messenger RNA (mRNA) and gene editing sys- cally relevant cell types, including epithelial cells, endothelial
tems to non-liver tissues. SORT nanoparticles include a supplemen- cells, B cells, T cells, and hepatocytes (21–24). However, the
tal SORT molecule whose chemical structure determines the LNP’s mechanism of action remains undefined. Understanding the
tissue-specific activity. To understand how SORT nanoparticles sur- mechanism which enables SORT is important for optimizing
pass the delivery barrier of liver hepatocyte accumulation, we delivery to currently targetable organs as well as extending the
studied the mechanistic factors which define their organ-targeting SORT concept to other tissue and cell types. Moreover, mecha-
properties. We discovered that the chemical nature of the added nistic understanding would establish a biological rationale for
SORT molecule controlled biodistribution, global/apparent pKa, overcoming the delivery barrier of liver accumulation that
and serum protein interactions of SORT nanoparticles. Addition- currently hampers IV administered nanoparticles (27).
ally, we provide evidence for an endogenous targeting mechanism Herein, we identify and study mechanistic factors which could

ENGINEERING
whereby organ targeting occurs via 1) desorption of poly(ethylene explain the organ-targeting properties of SORT LNPs based on
glycol) lipids from the LNP surface, 2) binding of distinct proteins three established principles that define the efficacy of liver-
to the nanoparticle surface because of recognition of exposed targeting LNPs: biodistribution to the liver, an acid dissociation
SORT molecules, and 3) subsequent interactions between surface- constant (pKa) near 6.4, and the adsorption of apolipoprotein E
bound proteins and cognate receptors highly expressed in specific (ApoE) to the LNP surface (12, 28). We discovered that each of
tissues. These findings establish a crucial link between the molecu- these three factors (organ level biodistribution, apparent pKa,

MEDICAL SCIENCES
lar composition of SORT nanoparticles and their unique and pre- and serum protein adsorption) are distinct for SORT LNPs and
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

cise organ-targeting properties and suggest that the recruitment correlate with their tissue-targeting properties. Furthermore, we
of specific proteins to a nanoparticle’s surface can enable drug provide evidence of a three-step mechanism for the functional
delivery beyond the liver. role of serum protein adsorption on tissue targeting that extends

lipid nanoparticles j mRNA delivery j gene editing j endogenous targeting


Significance

N ucleic acids that enable gene silencing (1, 2), expression


(3–5), and editing (6–8) possess great potential for use as
genetic medicines in multiple clinical settings including cancer (9,
Liver accumulation represents a significant barrier in the
development of therapeutically efficacious nanoparticle drug
10), inherited genetic disorders (11, 12), and infectious diseases delivery systems. Using a series of lipid nanoparticles with
(13–15). Due to the unfavorable pharmacokinetic properties of distinct organ-targeting properties, we provide evidence for
nucleic acids, viral and nonviral delivery approaches are used to a plausible mechanism of action for nanoparticle delivery to
facilitate nucleic acid delivery to target cells (16). Lipid nanopar- non-liver tissues. Following intravenous injection, specific
ticles (LNPs) represent the most clinically mature nonviral plat- proteins in the blood are recruited to the nanoparticle’s sur-
form for the safe and efficacious delivery of genetic medicines. face based on its molecular composition and they endow it
Indeed, LNPs were an enabling technology for the US Food and with a unique biological identity that governs its ultimate
Drug Administration approval of the first small interfering RNA fate in the body. An innovative paradigm emerges from this
(siRNA) drug, Onpattro, in 2018 (17) and the messenger RNA mechanistic understanding of nanoparticle delivery—endog-
(mRNA) vaccines currently being distributed for immunization enous targeting—wherein the molecular composition of a
against the SARS-CoV-2 virus, the causative agent of the nanoparticle is rationally engineered to interact with specific
proteins in the blood to overcome liver accumulation and to
COVID-19 pandemic (18, 19). Despite this progress, intrave-
target specific organs.
nously (IV) administered LNPs typically accumulate in the liver
and are internalized by liver hepatocytes, thereby greatly limiting
Author contributions: S.A.D., Q.C., and D.J.S. designed research; S.A.D. and Q.C.
the scope of their therapeutic applications (20). performed research; S.A.D., Q.C., and D.J.S. analyzed data; D.J.S. supervised research;
Recently, our laboratory overcame this challenge through and S.A.D. and D.J.S. wrote the paper.
the discovery of selective organ targeting (SORT), a strategy to Competing interest statement: D.J.S., Q.C., and the Regents of the University of Texas
rationally design nanoparticles for the extrahepatic delivery of System have filed patent applications related to this technology. D.J.S. is a cofounder
of/consultant with ReCode Therapeutics, which has licensed intellectual property from
mRNA and gene editing systems following IV administration the University of Texas Southwestern.
(Fig. 1A and SI Appendix, Table S1) (21–24). Conventional This article is a PNAS Direct Submission.
LNPs are composed of four components: ionizable cationic lip- Published under the PNAS license.
ids, amphipathic phospholipids, cholesterol, and poly(ethylene 1
To whom correspondence may be addressed. Email: Qiang.Cheng@UTSouthwestern.
glycol) (PEG) lipids (25, 26). We found that augmenting con- edu or Daniel.Siegwart@UTSouthwestern.edu.
ventional four-component LNPs for mRNA delivery to the liver This article contains supporting information online at http://www.pnas.org/lookup/
with a fifth component (termed a SORT molecule) can alter suppl/doi:10.1073/pnas.2109256118/-/DCSupplemental.
the LNPs’ in vivo organ-targeting properties and lead to the Published December 21, 2021.

PNAS 2021 Vol. 118 No. 52 e2109256118 https://doi.org/10.1073/pnas.2109256118 j 1 of 10


LNP biodistribution readout:
A LNP functional readout:
Luc mRNA translation to protein B Tracking of Cy5-mRNA C

Relative Fluorescence (%) Relative Fluorescence (%) Relative Fluorescence (%)


PBS 10% DODAP 20% DODAP 50% DODAP
Addition of 5th SORT molecule 80
Liver
Heart 60 Lung
Lung
40 Spleen

Conventional 4-Component LNP Liver Liver 20


18:1 DAP (DODAP) Spleen
SORT 0
5A2-SC8/DOPE/Chol/C14PEG/DODAP = Kidney Kidney 0 10 20 50
19/19/38/4/20 (mol. %) PBS 10% 18PA 20% 18PA 30% 18PA DODAP (%)

80
5A2-SC8
Liver
60 Lung
Spleen
Spleen 40
DOPE
18:1 PA (18PA)
SORT 20
Cholesterol 5A2-SC8/DOPE/Chol/C14PEG/18PA =
16.7/16.7/33.3/3.3/30 (mol. %) PBS 0% DOTAP 15% DOTAP 50% DOTAP 0
0 10 20 30
C14-PEG2K 18PA (%)
80
5A2-SC8/DOPE/Chol/C14PEG = Liver
23.8/23.8/47.6/4.8 (mol. %) 60 Lung
Lung Spleen
18:1 TAP (DOTAP) 40
SORT
5A2-SC8/DOPE/Chol/C14PEG/DOTAP = 20
11.9/11.9/23.8/2.4/50 (mol. %) ˜107 p/sec/cm2/sr
0
4.0 5.0 6.0 7.0 8.0 µW/cm2 0 15 50
DOTAP (%)

D DODAP SORT LNPs 18PA SORT LNPs DOTAP SORT LNPs


E
0%(mDLNP) 0%(mDLNP)
0%(mDLNP)
1.0 Lungs only
TNS fluorescence (%)

5%

TNS fluorescence (%)


TNS Fluorescence (%)

1.0 10% 1.0 5% Lungs + spleen


10%
20% 10% 15%
50% 20% 20% Spleen + lungs
30% Spleen only
80% 30% 0.5
0.5 0.5 40%
40% 50% Spleen + liver
60%
70%
Liver + spleen
0.0 0.0
80% Liver only
0.0 90%
3 5 7 9 11 3 5 7 9 11 3 5 7 9 11
pH pH pH

Fig. 1. SORT nanoparticles for tissue-specific mRNA delivery have unique biodistribution and ionization behavior. (A) By adding a fifth, supplemental
SORT molecule to a conventional, four-component LNP (mDLNP: 23.8 mol % 5A2-SC8, 23.8 mol % DOPE, 47.6 mol % cholesterol, and 4.8 mol % C14-
PEG2K), the tissue-specific activity of delivered mRNA changes based on the chemical structure of the included SORT molecule. An ionizable cationic lipid
(DODAP) enhances liver-specific mRNA translation (liver SORT: 19 mol % 5A2-SC8, 19 mol % DOPE, 38 mol % cholesterol, 4 mol % C14-PEG2K, and 20
mol % DODAP), an anionic lipid (18PA) results in spleen-specific mRNA translation (spleen SORT: 16.7 mol % 5A2-SC8, 16.7 mol % DOPE, 33.3 mol % cho-
lesterol, 3.3 mol % C14-PEG2K, and 30 mol % 18PA), and a cationic quaternary ammonium lipid (DOTAP) results in lung-specific mRNA translation (lung
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

SORT: 11.9 mol % 5A2-SC8, 11.9 mol % DOPE, 23.8 mol % cholesterol, 2.4 mol % C14-PEG2K, and 50 mol % DOTAP). (B) Ex vivo fluorescence of Cy5-
labeled mRNA in major organs extracted from C57BL/6 mice IV injected with SORT LNPs that incorporate increasing percentages of different SORT mole-
cules (0.5 mg/kg mRNA/body weight, 6 h). (C) Relative average Cy5 fluorescence measured in the liver, lung, and spleen as a function of SORT molecule
percent inclusion (0.5 mg/kg mRNA/body weight, n = 2). SORT molecules promote mRNA biodistribution to target organs. Data are shown as mean ±
SEM. (D) Representative TNS assay curves for determining the apparent pKa of SORT LNPs incorporating increasing percentages of ionizable cationic,
anionic, or permanently cationic lipid SORT molecules. Apparent pKa was defined as the point at which 50% of TNS fluorescence was achieved. (E) LNPs
were assigned a tissue specificity index based on the tissues in which functional luciferase mRNA was detected. Intermediate tissue specificity indexes rep-
resent LNPs in which mRNA activity was detected in multiple organs; these LNPs are plotted in the region of the organ for which higher activity was mea-
sured. For the 67 LNPs tested with the TNS assay, LNP apparent pKa was correlated with the specificity of luciferase mRNA tissue delivery.

the scope and application of the endogenous targeting mecha- they do not readily diffuse through anionic cellular membranes
nism defined for four-component LNP delivery to liver hepato- and are susceptible to degradation by ribonucleases (RNases)
cytes (12, 28). First, desorption of PEG lipids on the LNP surface in the blood (5). Ideally, LNPs will encapsulate and protect
exposes underlying SORT molecules in the LNP. Next, distinct mRNA from enzymatic degradation, enable accumulation in
serum proteins recognize the exposed SORT molecules and the target organ, facilitate receptor-mediated endocytosis into
adsorb to the LNP surface. Finally, surface-adsorbed proteins cells, and release mRNA from endosomes into the cytosol to
interact with cognate receptors expressed by cells in the target undergo translation into a functional protein (12). Through the
organs to facilitate functional mRNA delivery to those tissues. development of SORT LNPs, we showed that the chemical
The results indicate that the choice of SORT molecule gov- identity and amount of SORT molecule added to a four-
erns which proteins most avidly adsorb to the LNP surface, component LNP systematically alters tissue-specific protein
impacting the ultimate biological fate of the LNP (29). We envi- expression following mRNA delivery (21). The inclusion of
sion that this mechanistic understanding of SORT LNPs will ionizable cationic lipids enhanced liver targeting, anionic lipids
enable their optimization for therapeutic applications in the resulted in retargeting of delivery to the spleen, and per-
lung, liver, and spleen and lay the foundation for extending the manently cationic lipids bearing a quaternary ammonium
SORT platform to additional nanoparticle types, physiological headgroup retargeted delivery to the lungs (Fig. 1A and SI
tissues, and cell types. Furthermore, our findings suggest that Appendix, Table S1) (21). These experiments revealed how the
endogenous targeting—that is, tuning a nanoparticle’s molecu- choice of SORT molecule served as a design parameter to pre-
lar composition such that it binds specific proteins in the serum dictably dictate where mRNA would be translated to functional
to enable delivery to the target site—could be a generally useful proteins in vivo (21) but did not elucidate the organs where
strategy for engineering a wide array of nanomaterials capable SORT LNPs accumulated and the mechanism of how SORT
of extrahepatic delivery. molecules function. Biodistribution to the target organ should,
in principle, be a requisite step for targeted delivery.
Results Therefore, we first investigated the biodistribution of mRNA
SORT Molecules Alter LNP Biodistribution. mRNA therapeutics encapsulated in SORT LNPs to determine if there was a corre-
must overcome multiple barriers for intracellular delivery, as lation between the organs in which SORT LNPs accumulated

2 of 10 j PNAS Dilliard et al.


https://doi.org/10.1073/pnas.2109256118 On the mechanism of tissue-specific mRNA delivery by
selective organ targeting nanoparticles
and their tissue-specific mRNA activity. We formulated liver, endogenous mechanism in which the desorption of PEG lipids
lung, and spleen SORT LNPs as well as a representative four- from the LNP surface enables ApoE to bind the LNP; this sub-
component dendrimer LNP for mRNA delivery to the liver sequently enables receptor-mediated binding and uptake of the
(mDLNP) (SI Appendix, Tables S1 and S7) to encapsulate Cy5- LNPs by the low-density lipoprotein receptor (LDL-R) (12).
labeled mRNA following the identical protocols and formula- We hypothesized that SORT LNPs might operate by a similar
tion parameters used in our previous publications (11, 21). We mechanism in which 1) PEG lipid desorbs from the LNP to
administered each LNP IV to C57BL/6 mice and tracked the expose underlying SORT molecules, 2) distinct serum proteins
in vivo biodistribution of the Cy5 mRNA using fluorescence recognize the exposed SORT molecules and adsorb to the LNP
imaging. The organs were excised and imaged ex vivo 6 h post- surface, and 3) these surface-adsorbed proteins interact with
injection (Fig. 1B). The average fluorescence produced by the cognate receptors which mediate uptake of the LNPs by cells in
liver, spleen, and lungs was quantified (Fig. 1C). The incorpora- target tissues (Fig. 2A). First, we established the role of the
tion of the ionizable cationic lipid 1,2-dioleoyl-3-dimethylam- PEG–lipid component on mRNA delivery efficacy in vivo.
monium-propane (DODAP) to mDLNP resulted in an increase LNPs incorporate a PEG lipid on the surface to promote colloi-
in average Cy5 mRNA signal in the liver and a decrease of dal stability (26). Because the PEG–lipid molecules are nonco-
average Cy5 mRNA signal in the spleen, with optimal liver bio- valently incorporated into the LNP, they spontaneously desorb
distribution at 20% inclusion of DODAP (Fig. 1 B and C). at a rate inversely proportional to the length of the PEG lipid’s
Meanwhile, increasing the percentage of anionic SORT mole- hydrophobic anchor (31, 32). PEG lipids on the LNP surface
cule 18PA added to the LNP increased average Cy5 mRNA sig- can impair serum protein adsorption, resulting in reduced cel-
nal in the spleen (Fig. 1 B and C). Similarly, as the permanently lular targeting and delivery efficacy (31). The shedding of PEG
cationic SORT molecule 1,2-dioleoyl-3-trimethylammonium- lipid would be expected to expose the underlying SORT mole-
propane (DOTAP) was included in the LNP in increasing pro- cules for recognition by serum proteins, promoting their bind-
portions, there was a progressive increase in Cy5 mRNA signal ing of the SORT LNP in the blood.
in the lungs (Fig. 1 B and C). Importantly, these tracking stud- To functionally characterize the consequences of PEG lipid
ies quantify where the SORT LNPs biodistribute but not where desorption on mRNA delivery, we measured the effects of
mRNA is translated to protein (functional delivery). These substituting DMG-PEG2000 (C14-PEG2K), a methoxy PEG

ENGINEERING
studies demonstrate that inclusion of a SORT molecule in (mPEG) glyceride with 14-carbon-long alkyl tails, with DSG-
mDLNP facilitates mRNA biodistribution to the target organ PEG2000 (C18-PEG2K), an mPEG glyceride with 18-carbon-
but is not sufficient to explain tissue-specific mRNA activity. long alkyl tails, on the in vivo potency of SORT LNPs.
C18-PEG2K is expected to be less sheddable from the LNP
SORT Molecules Alter Apparent LNP pKa. It is established that compared to C14-PEG2K due to its longer hydrophobic anchor
global/apparent pKa plays an important role in determining the (31, 32). First, we measured the effects of increasing the PEG

MEDICAL SCIENCES
potency of RNA delivery by LNPs. Indeed, siRNA delivery to lipid anchor length on the delivery of luciferase mRNA by
liver hepatocytes strongly depends on LNP pKa, with a pKa of injecting C57BL/6 mice IV at a dosage of 0.1 mg/kg mRNA and
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

6.2 to 6.4 being optimal for gene silencing; LNPs with a pKa imaging organ luminescence ex vivo at a timepoint of 6 h postin-
outside of that narrow range were unable to functionally deliver jection (Fig. 2B). Switching to C18-PEG2K significantly
siRNA to the liver (30). Based on the link between pKa and cel- reduced the total luminescence produced by luciferase mRNA
lular delivery, we analyzed the apparent pKa of 67 different translated into a functional protein in target organs when com-
SORT LNPs (SI Appendix, Table S2), all of which were effica- pared to C14-PEG2K for all SORT LNPs (Fig. 2C). To confirm
cious for tissue-specific mRNA delivery in vivo (21), using the these findings, we delivered human erythropoietin (hEPO)
6-(p-toluidino)-2-naphthalenesulfonic acid (TNS) assay (Fig. 1 D mRNA to target tissues in vivo using liver, spleen, or lung
and E and SI Appendix, Figs. S2 and S3). Because SORT involves SORT LNPs incorporating either C14-PEG2K or C18-PEG2K.
the inclusion of additional charged lipids, the resulting TNS titra- Because hEPO is a secreted protein, delivery efficacy can be
tion curves capture the ionization behavior of more complex readily quantified by measuring hEPO levels in the serum (21).
mixed species LNPs. As the lower limit TNS fluorescence of 0% We verified that serum hEPO concentration was lower in mice
was not measured for some SORT LNPs over the pH range of treated with C18-PEG2K SORT LNPs compared to those
buffers used in the TNS assay, we defined the apparent pKa as treated with C14-PEG2K SORT LNPs (Fig. 2D). Thus, we
the pH at which 50% normalized TNS fluorescence signal is mea- quantitatively confirmed that SORT LNPs which incorporate a
sured (Fig. 1D). PEG lipid with a longer hydrophobic anchor are less effective
When plotting relative pKa with respect to tissue-specific for mRNA delivery to target organs. These studies suggest that
activity, SORT LNPs grouped into defined ranges based on PEG lipid desorption is a necessary process for efficacious
their organ-targeting properties (Fig. 1E). Confirming the liter- mRNA delivery to target tissues by SORT LNPs.
ature precedent, all liver-targeting SORT LNPs had an appar-
ent pKa within the well-established 6 to 7 range (30) (Fig. 1E). SORT Molecule Choice Influences LNP–Protein Interactions in the
Surprisingly, all lung-targeting SORT LNPs had a higher appar- Serum. The second step of the hypothesized endogenous target-
ent pKa (greater than 9) (Fig. 1E), while spleen-targeting LNPs ing mechanism for SORT LNPs involves the adsorption of
had a lower pKa between 2 and 6 (Fig. 1E). This result con- distinct proteins to the LNP surface to form unique protein
trasts with the dogma for conventional LNP delivery to the liver coronas (Fig. 2A). Following PEG lipid desorption, serum pro-
and contributes to the understanding of why SORT LNPs are teins readily adsorb to the surface of IV administered LNPs,
unconventional and can enable extrahepatic delivery. It is forming an interfacial layer known as the “protein corona” that
important to note that the SORT LNPs tested here possess a defines their biological identity (29). Adsorption of ApoE has
similar, near neutral zeta potential surface charge (SI Appendix, been shown to drive liver targeting of conventional four-
Table S1). Thus, the addition of a SORT molecule to a multi- component LNPs through binding of the LDL-R highly
component LNP alters its overall pKa that directly relates to expressed on hepatocytes (28). Given the strikingly different
the organ-targeting properties of the LNP. organ-targeting capabilities of lung and spleen SORT LNPs, we
hypothesized that the set of serum proteins that bind the LNPs
PEG Lipid Desorption Contributes to Effective mRNA Delivery would be distinct. We isolated the plasma proteins which bind
In Vivo. Conventional four-component LNPs are known to mDLNP, liver SORT, spleen SORT, and lung SORT using dif-
enable functional RNA delivery into liver hepatocytes via an ferential centrifugation following ex vivo incubation with mouse

Dilliard et al. PNAS j 3 of 10


On the mechanism of tissue-specific mRNA delivery by https://doi.org/10.1073/pnas.2109256118
selective organ targeting nanoparticles
A E F Functional Classes
of Adsorbed Proteins G Isoelectric Point
H Top 5 Serum
Proteins Adsorbed
mDLNP Liver Spleen Lung
Mw SORT SORT SORT
250 15
150
2

% Abundance
1 2 3 100 ApoE
19 21
75 10 C1qA
16
50 IgκC
42 5
37
mDLNP IgHγ2B

25 C1qB
20 0
(kDa) 0 2 4 6 8 10 12 14
0 5 10 15

B C14-PEG2K C18-PEG2K
3.0
C Luciferase mRNA

D hEpo mRNA

15
ApoE

% Abundance
1.5×10 9 40000
Liver 6 10 C1qA
19

Serum hEPO
Luminescence
SORT 21

(mIU/mL)
30000
1×10 9 IgHµ
x107 20
20000 5
35 C1qB
5×10 8 Liver
10000 Alb
SORT 0
0 0 0 2 4 6 8 10 12 14
0.5 0 5 10 15

1.0 ✱ ✱ 25
1.5×10 8 5000
Spleen 3 β2-GPI

% Abundance
12 17 20
Luminescence

4000
SORT Serum hEPO
IgκC
1×10 8 (mIU/mL) 3000 15
x107 38 30 C1qA
2000 10
5×10 7
Spleen ApoE
1000 5
SORT IgHγ2B
0 0 0
0.05
0 2 4 6 8 10 12 14 0 10 20 30

Lung ✱
1.0 2×10 7 1500
15
SORT ✱
Vtn
Serum hEPO

% Abundance
Luminescence

1.5×10 7 14 18
(mIU/mL)

1000
10 Pon1
1×10 7 9
x106 ApoE
500 46 13
5×10 6 Lung 5 Alb
SORT
0 0 Clu
0.1 C14- C18- C14- C18- 0
PEG2K PEG2K PEG2K PEG2K 0 2 4 6 8 10 12 14 0 5 10 15
pI % Abundance

Fig. 2. Multiple steps are involved in the mechanism of SORT LNP tissue targeting, including formation of unique protein coronas. (A) A proposed three-
step endogenous targeting mechanism for tissue-specific mRNA delivery by SORT LNPs in which 1) PEG lipid desorption 2) enables distinct plasma proteins
to bind SORT LNPs, 3) resulting in cellular internalization in the target tissues by receptor-mediated uptake. (B) Ex vivo bioluminescence of major organs
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

excised from C57BL/6 mice IV injected with liver, spleen, and lung SORT LNPs incorporating either sheddable PEG lipids (C14-PEG2K) or less sheddable PEG
lipids (C18-PEG2K) (0.1 mg FLuc mRNA/kg body weight, 6 h). Total luminescence produced by each organ is reduced when less sheddable PEG lipid is
used, suggesting that PEG lipid desorption is a key process for efficacious mRNA delivery by SORT LNPs. (C) Quantification of total luminescence produced
by functional protein translated from FLuc mRNA in target organs of C57BL/6 mice IV injected with liver, spleen, and lung SORT LNPs incorporating either
C14- or C18-PEG2K (0.1 mg FLuc mRNA/kg body weight, 6 h). (D) ELISA quantification of serum hEPO in C57BL/6 mice treated with liver, spleen, or lung
SORT LNPs encapsulating hEPO mRNA (0.1 mg hEPO mRNA/kg body weight, 6 h). Using a less-sheddable PEG reduces SORT LNP potency. (E) SDS–PAGE of
the plasma proteins adsorbed to the surface of mDLNP, liver SORT, spleen SORT, and lung SORT LNPs. LNPs with different organ-targeting properties
bind distinct plasma proteins. (F) The average abundance of proteins with distinct biological functions in the protein coronas of mDLNP and liver, spleen,
and lung SORT LNPs. The choice of SORT molecule leads to large-scale differences in the functional ensemble of plasma proteins which bind the LNP. (G)
Isoelectric point distribution for the most enriched proteins which constitute 80% of the protein corona of the LNPs. A SORT molecule’s headgroup struc-
ture influences the pI distribution of the protein corona. (H) The top five most abundant plasma proteins that bind different SORT LNPs (n = 3). The
chemical structure of SORT molecule affects the number one plasma protein that is most highly enriched on the surface of SORT LNPs. Data are shown as
mean ± SEM. Statistical significance was determined using an unpaired two-tailed Student’s t test (*P < 0.05).

plasma (33). Treating four-component mDLNP as the base abundance) for each LNP (SI Appendix, Tables S3–S6). First, we
liver-targeting LNP composition, we used sodium dodecyl sul- assessed the biological function of these highly abundant pro-
fate (SDS)–polyacrylamide gel electrophoresis (PAGE) (SDS- teins. By clustering plasma proteins into the physiological classes
PAGE) to qualitatively study how the choice of SORT molecule of apolipoproteins, coagulation proteins, complement proteins,
impacted which proteins adsorbed to five-component SORT immune proteins, and other proteins (34), we discovered that the
LNPs (Fig. 2E). While the set of plasma proteins that adsorb to proportion of proteins in each class changed based on the choice
liver SORT looks qualitatively similar to that of mDLNP, the of SORT molecule, suggesting that the inclusion of a SORT mol-
plasma proteins that bind SORT LNPs targeting extrahepatic ecule results in large-scale differences in the ensemble of proteins
organs are markedly distinct (Fig. 2E). In particular, a key which bind an LNP (Fig. 2F). The distinct functions of these pro-
band is highly enriched for spleen SORT at an Mw of 54 kDa, teins may play a role in shaping an LNP’s endogenous identity
while a band near 65 kDa is highly enriched for lung SORT and subsequent fate in vivo.
(Fig. 2E). Thus, adding a SORT molecule to mDLNP alters the To better understand what drives these large-scale differ-
composition of the protein corona in accordance with the ences in the functional composition of the protein corona, we
SORT molecule’s chemical structure. studied how a SORT molecule’s chemical structure might affect
Unbiased mass spectrometry proteomics enabled identification which proteins bind to LNPs. While each SORT molecule
and quantification of which proteins bind SORT LNPs in the shares a common hydrophobic scaffold that enables the mole-
plasma. We found that over 900 different proteins adsorbed to cule to self-assemble into the LNP, they are distinguished by
SORT LNPs, but nearly 98% of these proteins are present at an the chemical structure and charge state of the headgroup.
abundance of less than 0.1%. We anticipate that the most abun- These molecular features may play a role in the differential
dant proteins are the ones most likely to be functionally impor- enrichment of proteins with distinct characteristics, possibly
tant; thus, we focused our subsequent analysis on the proteins through electrostatic forces which bring specific proteins into
that constitute the majority of the protein corona (80% proximity of the SORT LNP to facilitate further protein–LNP

4 of 10 j PNAS Dilliard et al.


https://doi.org/10.1073/pnas.2109256118 On the mechanism of tissue-specific mRNA delivery by
selective organ targeting nanoparticles
interactions (34). A protein’s isoelectric point (pI) is defined as interact with β2-GPI–bound particles containing anionic phos-
the pH at which the protein molecule bears no net charge, pro- pholipids (37), and found that incubation with β2-GPI
viding an estimate of a protein’s charge state in the physiologi- enhanced Cy5 mRNA uptake of spleen SORT LNPs by a factor
cal milieu. We determined the pI of the major proteins that of 2.1 (Fig. 3 B and C). Finally, we examined A-498 and U-87
bound mDLNP and each SORT LNP using the bioinformatics MG cells, which highly express Vtn’s receptor αvβ3 integrin.
resource ExPASy (35). The inclusion of DODAP, an ionizable Incubation with Vtn increased cellular uptake of lung SORT
cationic lipid, to mDLNP did not greatly alter the pI distribu- LNPs at a factor of 23.2 (Fig. 3 B and C) and 4.2 (SI Appendix,
tion of the major proteins which bound the LNP (Fig. 2G). In Fig. S4) times greater than the uncoated LNPs. Thus, the
contrast, the inclusion of 18PA, a lipid with an anionic head- choice of SORT molecule impacts which plasma proteins
group, to mDLNP promoted the adsorption of plasma proteins adsorb to the LNP surface, and these proteins likely interact
with a pI greater than physiological pH (Fig. 2G), while the with cognate receptors expressed by target cells to enhance cel-
inclusion of DOTAP, a lipid with a cationic quaternary ammo- lular uptake, promoting intracellular mRNA delivery.
nium headgroup, in mDLNP favored the enrichment of pro- Having determined that key proteins can enhance cellular
teins with a pI below physiological pH (Fig. 2G). Although all uptake by distinct receptors, we verified that these proteins
SORT LNPs bound proteins across a broad range of pIs, the improve functional mRNA delivery by measuring the activity
nature of the headgroup of a chosen SORT molecule does of luciferase enzyme translated from mRNA delivered by
affect which proteins adsorb to the LNP. uncoated and coated SORT LNPs in vitro. Additionally, we
We further analyzed the five most abundant proteins in the aimed to determine whether single protein coatings specifically
protein corona of each LNP. It was discovered that the most enhanced the delivery of the individual formulations which
highly enriched protein in each protein corona was unique based most avidly bound those proteins. Cells were treated with liver,
on the choice of SORT molecule (Fig. 2H). ApoE was the most spleen, and lung SORT LNPs, encapsulating luciferase mRNA,
highly enriched plasma protein for mDLNP, a conventional four- that were incubated with increasing amounts of ApoE, β2-GPI,
component LNP which targets the liver, on average composing or Vtn (Fig. 3D). Luciferase activity was enhanced in HuH-7
13.9% of mDLNP’s protein corona (Fig. 2H). This result agrees and Hep G2 cells by ApoE-coated liver SORT LNPs, whereas
with the established role of ApoE in RNA delivery to the liver luciferase activity in these cells was not improved by spleen or

ENGINEERING
(28), supporting the validity of the experimental approach. Addi- lung SORT LNPs preincubated with ApoE (Fig. 3D and SI
tionally, liver SORT most avidly bound ApoE at an average Appendix, Fig. S3), suggesting that spleen and lung SORT
abundance of 13.3%, representing a 55-fold enrichment com- LNPs do not efficiently bind ApoE and enter LDL-R–
pared to native mouse plasma (Fig. 2H). In contrast, spleen expressing cells. Thus, ApoE exclusively enhanced functional
SORT was most highly enriched in β2-glycoprotein I (β2-GPI) at mRNA delivery by liver SORT LNPs in LDL-R–expressing
an average abundance of 20.1% (125-fold higher than native cells. Similarly, incubating spleen SORT LNPs with β2-GPI

MEDICAL SCIENCES
mouse plasma) (Fig. 2H), while lung SORT was most highly exclusively enhanced functional mRNA delivery to THP-1 mac-
enriched in vitronectin (Vtn) at an average abundance of 12.2% rophages (Fig. 3D), while incubating lung SORT LNPs with
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

(108-fold higher than native mouse plasma) (Fig. 2H). Thus, Vtn exclusively enhanced functional mRNA delivery to A-498
SORT LNPs for extrahepatic mRNA most avidly bind proteins and U-87 MG cells (Fig. 3D and SI Appendix, Fig. S4). Interest-
distinct from ApoE compared to mDLNP. Furthermore, there ingly, incubating lung SORT LNPs at a ratio of 1.0 g Vtn/g total
were additional distinctions in the top five plasma proteins bound lipid resulted in diminished luciferase activity, suggesting that
to SORT LNPs in terms of individual molecular species as well excess (free in media) Vtn may be inhibiting αvβ3 integrin to
as their physiological function (Fig. 2H). To further validate our limit mRNA delivery. Collectively, these studies reveal that
findings, we examined the protein coronas formed around SORT unique interactions between specific SORT molecules and indi-
LNPs in the plasma of an additional mouse strain and identified vidual serum proteins selectively promote SORT LNP targeting
the same key proteins highly enriched in the protein coronas of to distinct cell types by enhancing cellular uptake.
SORT LNPs (SI Appendix, Tables S3–S6). Ultimately, SORT
LNPs bind low-abundance serum proteins to form unique protein Extrahepatic mRNA Delivery by SORT LNPs Occurs through an ApoE-
coronas with quantitatively distinct composition based on the Independent Mechanism. The adsorption of ApoE to the surface
nature of the incorporated SORT molecule, which may impact of conventional four-component LNPs is a crucial process neces-
tissue-specific mRNA delivery. sary for highly efficacious RNA delivery to liver hepatocytes (28).
mDLNP, which is a four-component LNP for mRNA delivery to
Key Serum Proteins Define SORT LNP Cellular Targeting. One the liver, avidly binds to ApoE in the serum. However, the addi-
mechanism by which surface-adsorbed proteins can endo- tion of a SORT molecule to mDLNP alters which serum proteins
genously target specific tissues is through interactions with adsorb to the LNP surface. The set of proteins which bind the
cognate cellular receptors, resulting in receptor-mediated endo- LNP might play a role in defining the organ-targeting properties
cytosis of the LNP (36) (Fig. 2A). Having identified the serum of SORT LNPs via an endogenous mechanism whereby surface-
proteins which most avidly bind SORT LNPs, we functionally adsorbed proteins interact with cognate receptors expressed by
characterized how these single proteins affect intracellular cells in the target organ. Since lung and spleen SORT LNPs
delivery of mRNA by SORT LNPs. We incubated all SORT include molecules (DOTAP and 18PA, respectively) that are not
LNPs with either ApoE, β2-GPI, or Vtn, the most highly common to conventional LNPs, we reasoned that differences in
enriched serum proteins discovered to associate with SORT organ targeting could be driven by differences in protein corona
LNPs in our proteomics studies and measured the effects of composition. Genetically modified mice can be used to deplete
these single proteins on LNP uptake and mRNA delivery effi- key proteins from the serum, preventing their adsorption to the
cacy in vitro (Fig. 3A). First, we incubated SORT LNPs encap- LNP surface and possibly impairing expected organ targeting. To
sulating Cy5 mRNA with ApoE and measured cellular uptake evaluate the plausibility of an endogenous targeting mechanism
in HuH-7 and Hep G2 cells, two cell lines which highly express in vivo, we investigated how the addition of a SORT molecule to
ApoE’s receptor, LDL-R. Intracellular accumulation of Cy5 a conventional four-component LNP impacts the functional role
mRNA was increased in HuH-7 and Hep G2 cells by a factor of ApoE on tissue-specific mRNA delivery using knockout mice.
of 2.4 (Fig. 3 B and C) and 6.5 (SI Appendix, Fig. S3), respec- We compared the delivery of luciferase mRNA by SORT
tively, when liver SORT LNPs were incubated with ApoE. LNPs to target organs in B6.129P2-Apoetm1Unc/J (ApoE/)
Next, we examined THP-1 macrophages, a cell line known to mice, a genetic knockout model lacking ApoE expression, to

Dilliard et al. PNAS j 5 of 10


On the mechanism of tissue-specific mRNA delivery by https://doi.org/10.1073/pnas.2109256118
selective organ targeting nanoparticles
A
Cy5-mRNA tracking (cellular uptake)

Bioluminescence (mRNA translation)

SORT LNP Protein Cell Treatment


Coating

B C ✱✱✱✱
D 3×10 6
✱✱✱

Cy5 mRNA Fluorescence


1500000

Luminescence
Liver SORT
2×10 6

ApoE coating
HuH-7 Cells
uncoated

1000000

1×10 6
500000

0
0 g ApoE/g lipid

0. 0

0. 0
25
5
0

25

0. 0
5
0

25
5
0
0.
1.

0.
1.

0.
1.
uncoated ApoE

15000

Cy5 mRNA Fluorescence


100000 ✱

Spleen SORT

Luminescence
80000
β2-GPI coating

THP-1 Cells 10000


uncoated

60000 ✱

40000
5000
20000

0 0
uncoated β2-GPI g β2-GPI/g lipid

0. 0
25

0. 0
5
0

25

0. 0
5
0

25
5
0
0.
1.

0.
1.

0.
1.
400000 ✱✱✱✱ ✱✱✱✱
Cy5 mRNA Fluorescence

Lung SORT 1×10 7


A-498 Cells 300000 8×10 6

Luminescence
Vtn coating
uncoated

200000 6×10 6

4×10 6
100000
2×10 6
0
uncoated Vtn 0
g Vtn/g lipid

0. 0
25

0. 0
25

0. 0
5
0

5
0

25
5
0
0.
1.

0.
1.

0.
1.
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

Fig. 3. Distinct plasma proteins regulate SORT LNP uptake and efficacy in vitro. (A) SORT LNPs were preincubated with either ApoE, β2-GPI, or Vtn prior
to treating relevant cell lines to measure cellular uptake (Cy5 mRNA tracking) or functional mRNA delivery (bioluminescence). (B) Representative images
of cellular uptake of uncoated and coated SORT LNPs taken up by relevant cell types. Incubating a SORT LNP with the protein most avidly binds increases
mRNA uptake in cell lines expressing the cognate receptor (250 ng mRNA per well, 1.5 h). (Scale bar: 50 μm.) (C) Quantification of Cy5 mRNA fluorescence
in cells treated with uncoated or coated SORT LNPs (250 ng mRNA per well, 1.5 h, n = 10). Statistical significance was determined using an unpaired two-
tailed Student’s t test (****P < 0.0001, *P < 0.05). (D) Activity of functional luciferase protein translated from mRNA delivered by uncoated or protein-
coated SORT LNPs in relevant cell lines (25 ng mRNA, 24 h, n = 4). Statistical significance determined using one-way ANOVA with Brown–Forsythe test
(****P < 0.0001, ***P < 0.001, *P < 0.05). Individual proteins exclusively bind to specific SORT LNPs and enhance mRNA delivery only to cell lines express-
ing the cognate receptor. Data are shown as mean ± SEM.

wild-type (WT) C57BL/6 mice containing normal levels of plausible that other serum proteins, such as those identified in
ApoE in the serum. Luminescence was quantified to measure the proteomics study, may be responsible for endogenous target-
functional luciferase mRNA delivery to the target organs of ing to the spleen and lungs in vivo.
each SORT LNP (Fig. 4). By eliminating ApoE from the
serum, mDLNP exhibited significantly reduced mRNA delivery
to the liver, with an average reduction of luciferase activity by Discussion
78% compared to WT C57BL/6 mice (Fig. 4). Liver SORT Clinical applications of genetic medicines are limited by the
LNPs, which also bind ApoE selectively from the serum, main- availability of efficacious carriers for the intracellular delivery
tain this dependence on ApoE for mRNA delivery to the liver: of nucleic acid biomolecules to target tissues. Since IV adminis-
the knockout of ApoE significantly impaired liver targeting, tered LNPs have been limited to targeting liver hepatocytes to
with an average reduction in luciferase activity by 87% com- date, there is great need to develop drug delivery systems capa-
pared to WT C57BL/6 mice (Fig. 4). Thus, ApoE is indispens- ble of extrahepatic targeting. Here, we identified mechanistic
able for efficacious targeting of the liver by both mDLNP and factors which underly the tissue-targeting properties of SORT
liver SORT LNPs. In stark contrast, mRNA delivery by spleen LNPs for extrahepatic mRNA delivery. We found that a SORT
SORT to the spleen was enhanced by a factor of 2.2 in ApoE/ molecule’s chemical structure uniquely impacts an LNP’s bio-
mice, suggesting that ApoE plays an antagonistic role in effica- distribution, apparent pKa, and serum protein interactions.
cious mRNA delivery to the spleen (Fig. 4). Tissue-specific Furthermore, we provide evidence for a plausible endogenous
mRNA delivery by lung SORT LNPs was not significantly differ- targeting mechanism involving PEG lipid desorption, serum
ent in ApoE/ mice compared to WT mice, indicating that this protein adsorption, and receptor binding followed by cellular
serum protein is not functionally important for modulating lung uptake to possibly explain the organ-targeting profiles of SORT
targeting (Fig. 4). Together, these results indicate that mRNA LNPs (Fig. 2A). Preferential uptake and activity of SORT LNPs
delivery is no longer an ApoE-dependent process upon inclusion likely occurs in the organs which highly express the relevant cel-
of either an anionic lipid or cationic lipid to mDLNP. Rather, lular receptors that favorably interact with serum proteins
spleen SORT and lung SORT LNPs can enable extrahepatic enriched at the surface of SORT LNPs. Although we cannot
mRNA delivery via an ApoE-independent mechanism. It is rule out additional mechanisms, we have identified multiple

6 of 10 j PNAS Dilliard et al.


https://doi.org/10.1073/pnas.2109256118 On the mechanism of tissue-specific mRNA delivery by
selective organ targeting nanoparticles
A B C Target Organ
Wild-type (WT) ApoE-/-
Luminescence
5.0 ✱✱
mDLNP 2×10 8

Luminescence
1.5×10 8
x106
1×10 8

5×10 7

0.3
0

Liver SORT 2.0 1.5×10 9

Luminescence
1×10 9
x107
5×10 8

0.2 0


Spleen SORT 3.0 2.5×10 7

2×10 7

Luminescence
1.5×10 7
x106
1×10 7

5×10 6

0.3 0

1.0 2.5×10 7 ns

ENGINEERING
Lung SORT

Luminescence
2×10 7

1.5×10 7
x106
1×10 7

5×10 6

0.1 0
WT ApoE-/-

MEDICAL SCIENCES
Fig. 4. Extrahepatic mRNA delivery occurs via an ApoE-independent mechanism. (A) Ex vivo bioluminescence produced by functional protein translated
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

from FLuc mRNA in major organs excised from WT C57BL/6 mice IV injected with mDLNP or liver, spleen, or lung SORT LNPs (0.1 mg/kg FLuc mRNA, 6 h).
The role of ApoE on SORT LNP efficacy varies based on the chemical structure of the included SORT molecule. (B) Ex vivo bioluminescence produced by
functional protein translated from FLuc mRNA in major organs excised from ApoE/ mice IV injected with mDLNP or liver, spleen, or lung SORT LNPs (0.1
mg/kg FLuc mRNA, 6 h). (C) Quantification of total bioluminescence produced by target organs excised from WT and ApoE/ mice treated with mDLNP
or liver, spleen, or lung SORT LNPs (0.1 mg/kg FLuc mRNA, 6 h, n = 3). Data are shown as mean ± SEM. Statistical significance was determined using an
unpaired two-tailed Student’s t test (**P < 0.01, *P < 0.05, ns, P > 0.05). Elimination of ApoE from the serum using genetic knockout results in a marked
reduction of hepatic mRNA delivery by mDLNP and liver SORT LNPs. In contrast, spleen SORT LNPs have enhanced spleen targeting when ApoE is depleted
from the serum, while the efficacy of lung SORT LNPs is unaffected by ApoE elimination.

key factors which define the organ-targeting properties of the key proteins we discovered to associate with SORT LNPs
SORT LNPs. during a static incubation versus a dynamic incubation (39–41).
The proposed mechanism may have similarity with that of It is known that the composition of the protein corona is
lipoproteins, which can be considered as a natural class of influenced by a nanoparticle’s surface chemistry (29, 42, 43).
nanoparticles for physiological cholesterol transport through Since SORT LNPs incorporate SORT molecules with distinct
the blood. Acquisition of ApoE in the serum results in uptake chemical structures, SORT LNPs likely have different surface
of lipoproteins by liver hepatocytes through receptor-mediated chemistries underneath the PEG layer. The desorption of PEG
endocytosis by LDL-R (38). This physiological function of lipid from the LNP unveils this surface, enabling specific
ApoE is leveraged by DLin-MC3-DMA LNPs for siRNA deliv- plasma protein adsorption. It was recently shown that binding
ery to the liver; by binding ApoE in the serum, these nanopar- of ApoE induces rearrangement of lipids in DLin-MC3-DMA
ticles can endogenously target LDL-R highly expressed by liver LNPs, promoting the migration of certain lipids from the core
hepatocytes (12, 28). The hypothesized mechanism we provide to the shell (44). These observations suggest that protein
evidence for herein builds upon and extends the scope of this adsorption can alter the surface composition of SORT LNPs,
endogenous targeting concept. Importantly, SORT LNPs possibly generating unique nanodomains which further pro-
include classes of “out-of-the-box” supplemental molecules that mote the adsorption of distinct proteins to the LNP surface.
can tune an LNP’s molecular composition to promote the bind- The distinct surface chemistry of SORT LNPs can explain
ing of distinct protein species to the LNP, which are not which specific proteins bind to the LNP. β2-GPI is known to
typically observed in the protein corona, and enable mRNA interact with anionic phospholipids, including 18PA (45), and
delivery to cells and organs beyond liver hepatocytes. Because Vtn has been associated with DOTAP (46). Furthermore,
SORT LNPs include additional classes of molecules that have ApoE, which facilitates liver targeting of LNPs, is highly
not been included in LNPs before, the SORT platform expands enriched in liver SORT LNPs (28). These results further sup-
the toolbox of molecules available to control the protein corona port the proteomics findings.
without loss of efficacy. Although a static ex vivo incubation Understanding the molecular interactions of these individual
does not fully recapitulate the dynamic flow environment in proteins can illuminate why their enrichment might result in
which the protein corona is formed in vivo, studies have not the observed organ-targeting properties of SORT LNPs. Inter-
identified significant differences in the relative abundance of actions between ApoE and LDL-R drive hepatic accumulation

Dilliard et al. PNAS j 7 of 10


On the mechanism of tissue-specific mRNA delivery by https://doi.org/10.1073/pnas.2109256118
selective organ targeting nanoparticles
of endogenous lipoproteins during cholesterol metabolism (38), The ONE-Glo + Tox Luciferase Reporter assay kit was purchased from Promega
possibly explaining why ApoE adsorption is a hallmark of many Corporation. B6.129P2-Apoetm1Unc/J mice were acquired from the Jack-
son Laboratory.
liver-targeting LNPs, including liver SORT. In a similar manner,
Vtn could bind its cognate receptor, αvβ3 integrin, which is Nanoparticle Preparation. RNA-loaded LNP formulations were prepared using
highly expressed by the pulmonary endothelium but not by liver the ethanol dilution method. The mDLNP and the SORT formulations were
cells or other vascular beds (47–49), providing a plausible developed and reported in our previous papers (11, 21, 22, 25). Unless other-
explanation for why Vtn promotes lung specificity. Finally, β2- wise stated, all lipids with specified molar ratios were dissolved in ethanol,
GPI can bind phosphatidyl serine, an anionic lipid exposed by and RNA was dissolved in 10 mM citrate buffer (pH 4.0). The two solutions
senescent red blood cells to promote their filtration from the were rapidly mixed at a ratio of 3:1 (mRNA:lipids, vol:vol) and then incubated
for 10 min at room temperature. To prepare SORT LNP formulations containing
circulation in the spleen (50), suggesting how β2-GPI could be
anionic lipids, the anionic lipids were first dissolved in tetrahydrofuran and then
implicated in spleen SORT LNP targeting. These findings sup- mixed with other lipid components in ethanol. All formulations were named
port the functional role of individual proteins on the organ- based on the additional lipids. All LNPs were prepared to have a 40:1 weight
targeting properties of SORT LNPs. We acknowledge that the ratio of lipids to mRNA. To prepare mDLNP, a mixture of 23.8 mol % 5A2-SC8,
role of other highly enriched proteins in enhancing organ tar- 23.8 mol % DOPE, 47.6 mol % cholesterol, and 4.8 mol % DMG-PEG2K was dis-
geting, such as albumin for liver SORT (51), should not be solved in ethanol, then mixed with mRNA, and diluted in 10 mM, pH 4.0 citrate
discounted. Clusterin, highly enriched in lung SORT, could buffer at a volume ratio of 3:1 (mRNA:lipids). SORT LNPs were prepared by add-
potentially play a role in evading the mononuclear phagocyte ing the SORT molecule to the lipid solution at a given mole %. To prepare liver
SORT, a mixture of 19 mol % 5A2-SC8, 19 mol % DOPE, 38 mol % cholesterol, 4
system to promote lung targeting (52, 53). Additionally, based
mol % DMG-PEG2K, and 20 mol % DODAP was dissolved in ethanol, then
on the key role of ApoE in driving liver targeting, the reduced mixed with mRNA, and diluted in 10 mM, pH 4.0 citrate buffer at a volume ratio
binding of ApoE to spleen and lung SORT LNPs could pro- of 3:1 (mRNA:lipids). To prepare spleen SORT, a mixture of 16.7 mol % 5A2-SC8,
mote extrahepatic mRNA delivery (54, 55), possibly because of 16.7 mol % DOPE, 33.3 mol % cholesterol, 3.3 mol % C14-PEG2K, and 30 mol %
the displacement of ApoE by apolipoprotein C (53), which is 18PA in ethanol was prepared, then mixed with mRNA, and diluted in 10 mM,
present in the protein coronas of spleen and lung SORT LNPs pH 3.0 citrate buffer at a volume ratio of 3:1 (mRNA:lipids). To prepare lung
but not liver SORT (SI Appendix, Tables S4–S6). SORT, a mixture of 11.9 mol % 5A2-SC8, 11.9 mol % DOPE, 23.8 mol % choles-
Our proposed mechanism suggests that tuning the molecular terol, 2.4 mol % C14-PEG2K, and 50 mol % DOTAP was prepared in ethanol,
composition of an LNP serves as a simple and effective method then mixed with mRNA, and diluted in 10 mM, pH 4.0 citrate buffer at a volume
ratio of 3:1 (mRNA:lipids). After 10 min of mixing the mRNA and lipid solutions,
for manipulating the endogenous ligands which bind the nano-
the LNP formulations were diluted with 1× phosphate buffered saline (PBS) to
particle and govern its subsequent biological fate. Our work 0.5 ng/μL mRNA for in vitro assays and characterization of physicochemical
clarifies that endogenous targeting via serum proteins acts as a properties. For in vivo experiments, the formulations were dialyzed (Pur-A-Lyzer
targeting mechanism that can be generalized to other LNP sys- Midi Dialysis Kits, MWCO 3.5 kDa, Sigma-Aldrich) against 1× PBS for 2 h. After-
tems besides those currently utilized for nucleic acid delivery to ward, LNPs were diluted with PBS to a final volume of 250 μL/mouse for
liver hepatocytes. With the key parameters for organ targeting IV injections.
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

identified, spleen and lung SORT LNPs may be further opti-


Characterization of mRNA Formulations. Size distribution, polydispersity
mized for more efficacious tissue-specific delivery. Moving for-
index, and zeta-potential were measured using DLS (Malvern MicroV model;
ward, the function of highly enriched serum proteins with He-Ne laser, λ = 632 nm). To measure the apparent pKa of mRNA formula-
respect to extrahepatic targeting in vivo should be further eluci- tions, the TNS assay was employed. mRNA formulations (60 μM total lipids)
dated. Additionally, identifying SORT molecules that bind and the TNS probe (2 μM) were incubated for 5 min with a series of buffers
serum proteins distinct from those detailed within this manu- containing 10 mM Hepes, 10 mM MES (4-morpholineethanesulfonic acid),
script may serve as a worthwhile strategy for the discovery of 10 mM ammonium acetate, and 130 mM NaCl (the pH ranged from 2.5 to 11).
new LNPs which target other organs beyond the liver, spleen, The mean fluorescence intensity of each well (black-bottom 96-well plate)
and lungs. We view endogenous targeting, that is, engineering was measured by a Tecan plate reader with λEx = 321 nm and λEm = 445 nm,
and data were normalized to the value of pH 2.5. Typically, the apparent pKa
nanoparticle composition to facilitate interactions with distinct
is defined by the pH at half-maximum fluorescence. Although this method
serum proteins and thereby tissue-specific delivery, as an effec- was useful for estimating LNP global/apparent pKa for most all LNPs, it could
tive and broad paradigm for designing nanoparticles of various not be used for SORT LNPs containing >40% permanently cationic lipid
material compositions to overcome liver accumulation and tar- because these LNPs are always charged. Therefore, we instead estimated the
get extrahepatic organs. relative pKa compared to base LNP formulation (no added SORT lipid) when
50% fluorescence normalized to the lowest fluorescence measurement was
Materials and Methods produced. This alternative calculation did not change the pKa for most LNPs
but did allow the estimation of the pKa of SORT LNPs with >40% cationic lipid
Materials. 5A2-SC8 was synthesized and purified by following published pro-
that agreed with experimental results for tissue-selective RNA delivery. Thus,
tocols (25). DOTAP, 1,2-dioleoyl-sn-glycero-3-phosphate (sodium salt) (18PA),
we suggest that the standard TNS assay be used when LNPs contain a single
DODAP, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and 1,2-Dis-
ionizable cationic lipid and the alternative 50% normalized signal method be
tearoyl-rac-glycerol-methoxy(PEG MW 2000) (DSG-PEG2000) were purchased
used for systems such as SORT that contain complex mixtures of multiple lipids
from Avanti Polar Lipids. Cholesterol, sucrose, SDS, and phorbol 12-myristate
harboring a variety of charge states.
13-acetate were purchased from Sigma-Aldrich. DMG-PEG2000 was purchased
from NOF America Corporation. The ReadyPrep 2-D Cleanup Kit, 12% Mini- In Vivo Luc mRNA Biodistribution. All animal experiments were approved by
PROTEAN TGX Precast Protein Gels, 2× Laemmli Buffer, and 10× Tris/Glycine/ the Institution Animal Care and Use Committees of The University of Texas
SDS were purchased from Bio-Rad. SimplyBlue Safe Stain, Pierce 660 nm Pro- Southwestern Medical Center and were consistent with local, state, and federal
tein Assay Reagent, Ionic Detergent Compatibility Reagent for Pierce 660 nm regulations as applicable. C57BL/6 mice weighing 18 to 20 g were IV injected
Protein Assay Reagent, Hoechst 33342, Gibco VTN Recombinant Human Pro- with various SORT LNPs containing Cy5 FLuc mRNA at a dose of 0.5 mg/kg
tein Truncated, and dynamic light scattering (DLS) Ultramicro cuvettes were mRNA. Ex vivo imaging (Cy5 channel) was performed 6 h postinjection. Average
purchased from Thermo Fisher. Innovative Research CD1 Mouse Plasma fluorescence of the liver, lung, and spleen were measured using Living Image
K2EDTA, Innovative Research C57BL/6 Mouse Plasma K2EDTA, Novus Biologi- Software (PerkinElmer) by drawing regions of interest around each organ. The
cals Recombinant Human ApoE4 Protein, and R&D Systems Recombinant relative fluorescence for each organ was calculated as the following:
Mouse ApoH (β2-GPI) protein were purchased from Fisher Scientific. Pur-A-
Lyzer Midi Dialysis Kits (MWCO, 3.5 kDa) were purchased from Sigma-Aldrich. Faverage, organ  Fbackground
Frelative, organ ¼ × 100:
CleanCap FLuc mRNA (5 moU), CleanCap Cyanine 5 FLuc mRNA (5 moU), and ∑ðFaverage, organ  Fbackground Þ
CleanCap EPO mRNA (5 moU) were purchased from TriLink BioTechnologies.
D-Luciferin (sodium salt) was purchased from Gold Biotechnology. The hEPO PEG Lipid mRNA Delivery Efficacy. C57BL/6 mice weighing 18 to 20 g were IV
enzyme-linked immunosorbent assay (ELISA) kit was purchased from Abcam. injected with liver, lung, and spleen SORT LNPs using either DMG-PEG2K or

8 of 10 j PNAS Dilliard et al.


https://doi.org/10.1073/pnas.2109256118 On the mechanism of tissue-specific mRNA delivery by
selective organ targeting nanoparticles
DSG-PEG2K as the PEG lipid component at a dose of 0.1 mg/kg (n = 3). LNPs into white-bottomed 96-well plates at a density of 2 × 104 cells per well and
encapsulated either FLuc mRNA or hEPO mRNA. For mice treated with treated with 100 nM phorbol 12-myristate 13-acetate overnight (to enable dif-
FLuc mRNA, after 6 h, mice were injected with D-Luciferin (150 mg/kg, intra- ferentiation into macrophages). All cells were cultured in complete medium
peritoneal [IP]) and imaged by an IVIS Lumina system (PerkinElmer). Total as recommended by the suppliers. Then, the old media was replaced with
luminescence of target organs was quantified using Living Image Software 100 μL media. After the formulation and dilution of SORT LNPs in 1× PBS, LNPs
(PerkinElmer). For mice treated with hEPO mRNA, blood was withdrawn were incubated with 0, 0.25, 0.5, or 1.0 g protein/g total lipid of ApoE, β2-GPI,
retro-orbitally 6 h after, and serum was recovered. The serum concentration or Vtn by adding the selected protein directly to the LNP solution and incubat-
of hEPO was quantified using an ELISA kit (Abcam) to quantify mRNA deliv- ing it with the LNPs for 15 min at 37 °C. Following incubation, each well was
ery efficacy. treated with 25 ng FLuc mRNA (n = 4). After 24 h, ONE-Glo + Tox kits were
used for mRNA expression and cytotoxicity detection based on the stan-
Isolation of Plasma Proteins Adsorbed to SORT LNPs. SORT LNPs were pre- dard protocol.
pared according to the previously described method and were diluted to a
final lipid concentration of 1 g/L with 1× PBS. Mouse plasma was added to In Vitro Cellular Uptake. A-498, HuH-7, U-87 MG, or Hep G2 cells were seeded
each LNP solution at a 1:1 volume ratio and incubated for 15 min at 37 °C. A into 12-well plates at a density of 1 × 105 cells per well and incubated at 37 °C
0.7-M sucrose solution was prepared by dissolving solid sucrose in MilliQ overnight while undifferentiated THP-1 cells were seeded into 12-well plates
water. The LNP/plasma mixture was loaded onto a 0.7-M sucrose cushion of at a density of 2 × 105 cells per well and treated with 100 nM of Phorbol
equal volume to the mixture and centrifuged at 15,300 g and 4 °C for 1 h. The 12-myristate 13-acetate overnight (to enable differentiation into macro-
supernatant was removed, and the pellet was washed with 1× PBS. Next, the
phages). All cells were cultured in complete medium as recommended by the
pellet was centrifuged at 15,300 g and 4 °C for 5 min, and the supernatant
suppliers. Then, the old media was refreshed and cells treated with 250 ng
was removed. Washing was performed twice more for a total of three washes.
Cyanine 5 FLuc mRNA encapsulated in SORT LNPs. LNPs were either uncoated,
Following the final wash, the pellet was resuspended in 2 weight % SDS.
preincubated with 1.0 g protein/g total lipid ApoE, 1.0 g protein/g total lipid
Excess lipids were removed from each sample by following the protocol pro-
β2-GPI, or 0.25 g protein/g total lipid Vtn. A total of 90 min after treatment,
vided with the ReadyPrep 2-D Cleanup (Bio-Rad). The resulting pellet from the
cells were washed two times with 1× PBS and stained with Hoechst 33342
cleanup step was resuspended in 2× Laemmli buffer. The concentration of
(0.1 mg  mL1) for 10 min at 37 °C. Cells were washed twice more with PBS
protein in each sample was quantified by Bradford Assay using the Pierce 660
and then imaged by fluorescence microscopy (Keyence BZ-X800). Images were
nm Protein Assay Reagent mixed with Ionic Detergent Compatibility Reagent.
captured using a 10× magnification. All image settings were kept consistent
SDS–PAGE Characterization of Plasma Proteins. Plasma proteins isolated from for a single experiment. Cy5 fluorescence intensity was quantified using

ENGINEERING
the surface of SORT LNPs were loaded onto a 10% Mini-PROTEAN TGX Precast ImageJ software version 1.53c (NIH). Each treatment group was performed in
Protein Gel at a volume of 10 μL and separated at 200 V. Proteins were visual- duplicates, and five images were taken for each well.
ized by staining the gel for 1 h with SimplyBlue Safe Stain. The gel was
destained using deionized (DI) water overnight and imaged with a Licor Scan- ApoE Knockout Mouse Experiments. SORT LNPs were prepared according to
ner the following day. the previously described method. B6.129P2-Apoetm1Unc/J mice weighing 18 to
20 g were IV injected with mDLNP and liver, lung, and spleen SORT LNPs at a
Preparation of Plasma Protein Samples for Mass Spectrometry. Plasma pro- dosage of 0.1 mg/kg FLuc mRNA (n = 3). As a comparison, C57BL/6 mice weigh-

MEDICAL SCIENCES
teins isolated from the surface of SORT LNPs were loaded onto a 12% ing 18 to 20 g were IV injected with mDLNP and liver, lung, and spleen SORT
Mini-PROTEAN TGX Precast Protein Gel at a volume of 10 μL and run into LNPs at a dosage of 0.1 mg/kg FLuc mRNA (n = 3). After 6 h, mice were
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

the gel 1 cm at 90 V. The gel was stained with SimplyBlue Safe Stain for 1 injected with D-Luciferin (150 mg/kg, IP) and imaged by an IVIS Lumina system
h to fix and visualize the proteins. After destaining for 1 h, the protein (PerkinElmer). Total luminescence of target organs was quantified using Liv-
bands were excised using a sterile razor blade and sliced into 1-mm3 ing Image Software (PerkinElmer).
cubes. The cubes were added to a 1.5-mL tube that had been rinsed with
1:1 MilliQ water:Ethanol and stored at 4 °C until being submitted to the Data Availability. All study data are included in the article and/or SI Appendix.
University of Texas Southwestern Proteomics Core for mass spectrometry
analysis. For the CD-1 plasma samples, a Thermo Fusion Lumos mass spec- ACKNOWLEDGMENTS. D.J.S. acknowledges grant support from the Cystic
trometer was used to identify the constituents of the protein corona. For Fibrosis Foundation (SIEGWA18XX0), the NIH National Institute of Biomedical
the C57BL/6 samples, a Thermo QExactive HF mass spectrometer was used Imaging and Bioengineering (R01 EB025192-01A1), the Cancer Prevention
to identify the constituents of the protein corona. The identified proteins and Research Institute of Texas (RP190251), and the American Cancer Society
were ordered based on their abundance/Mw. For the proteins constitut- (RSG-17-012-01). We acknowledge the Southwestern Small Animal Imaging
Shared Resource, which is supported in part by the Harold C. Simmons Cancer
ing 80% abundance of each protein corona, the proteins were grouped
Center through a National Cancer Institute Cancer Center Support Grant (P30
into functional classes based on information provided by the UniProt
CA142543). We acknowledge the University of Texas Southwestern Proteo-
database. The pI of these proteins was computed using the bioinformatic mics Core for its assistance with the mass spectrometry proteomics experi-
resource ExPASy (35). ments. We especially thank Prof. Andrew Lemoff for insightful advice
regarding mass spectrometry experiments. S.A.D. acknowledges financial sup-
In Vitro Luciferase Delivery. A-498, HuH-7, U-87 MG, or Hep G2 cells were port from the NIH Pharmacological Sciences Training Grant (GM007062) and
seeded into white-bottom 96-well plates at a density of 1 × 104 cells per well the NIH Molecular Medicine Training Grant (GM109776). Cartoons for the fig-
and incubated at 37 °C overnight. Undifferentiated THP-1 cells were seeded ure were created using Biorender.com.

1. K. A. Whitehead, R. Langer, D. G. Anderson, Knocking down barriers: Advances in 10. N. Kong et al., Synthetic mRNA nanoparticle-mediated restoration of p53 tumor sup-
siRNA delivery. Nat. Rev. Drug Discov. 8, 129–138 (2009). pressor sensitizes p53-deficient cancers to mTOR inhibition. Sci. Transl. Med. 11,
2. J. B. Miller, D. J. Siegwart, Design of synthetic materials for intracellular delivery of eaaw1565 (2019).
RNAs: From siRNA-mediated gene silencing to CRISPR/Cas gene editing. Nano Res. 11. Q. Cheng et al., Dendrimer-based lipid nanoparticles deliver therapeutic FAH mRNA
11, 5310–5337 (2018). to normalize liver function and extend survival in a mouse model of hepatorenal
3. U. Sahin, K. Kariko € T€
 , O. ureci, mRNA-based therapeutics—Developing a new class of tyrosinemia type I. Adv. Mater. 30, e1805308 (2018).
drugs. Nat. Rev. Drug Discov. 13, 759–780 (2014). 12. A. Akinc et al., The Onpattro story and the clinical translation of nanomedicines con-
4. K. A. Hajj, K. A. Whitehead, Tools for translation: Non-viral materials for therapeutic taining nucleic acid-based drugs. Nat. Nanotechnol. 14, 1084–1087 (2019).
mRNA delivery. Nat. Rev. Mater. 2, 17056 (2017). 13. N. Pardi, M. J. Hogan, F. W. Porter, D. Weissman, mRNA vaccines—A new era in vacci-
5. P. S. Kowalski, A. Rudra, L. Miao, D. G. Anderson, Delivering the messenger: Advances nology. Nat. Rev. Drug Discov. 17, 261–279 (2018).
in technologies for therapeutic mRNA delivery. Mol. Ther. 27, 710–728 (2019). 14. R. A. Feldman et al., mRNA vaccines against H10N8 and H7N9 influenza viruses of
6. J. A. Doudna, E. Charpentier, Genome editing. The new frontier of genome engi- pandemic potential are immunogenic and well tolerated in healthy adults in phase 1
neering with CRISPR-Cas9. Science 346, 1258096 (2014). randomized clinical trials. Vaccine 37, 3326–3334 (2019).
7. J. D. Sander, J. K. Joung, CRISPR-Cas systems for editing, regulating and targeting 15. N. Kose et al., A lipid-encapsulated mRNA encoding a potently neutralizing human
genomes. Nat. Biotechnol. 32, 347–355 (2014). monoclonal antibody protects against chikungunya infection. Sci. Immunol. 4,
8. H. X. Wang et al., CRISPR/Cas9-based genome editing for disease modeling and ther- eaaw6647 (2019).
apy: Challenges and opportunities for nonviral delivery. Chem. Rev. 117, 9874–9906 16. T. Wei et al., Delivery of tissue-targeted scalpels: Opportunities and challenges for
(2017). in vivo CRISPR/Cas-based genome editing. ACS Nano 14, 9243–9262 (2020).
9. A. Ventura, T. Jacks, MicroRNAs and cancer: Short RNAs go a long way. Cell 136, 17. D. Adams et al., Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloid-
586–591 (2009). osis. N. Engl. J. Med. 379, 11–21 (2018).

Dilliard et al. PNAS j 9 of 10


On the mechanism of tissue-specific mRNA delivery by https://doi.org/10.1073/pnas.2109256118
selective organ targeting nanoparticles
18. L. R. Baden et al., COVE Study Group, Efficacy and safety of the mRNA-1273 SARS- 38. R. W. Mahley, Apolipoprotein E: Cholesterol transport protein with expanding role
CoV-2 vaccine. N. Engl. J. Med. 384, 403–416 (2021). in cell biology. Science 240, 622–630 (1988).
19. F. P. Polack et al., C4591001 Clinical Trial Group, Safety and efficacy of the BNT162b2 39. A. Amici et al., In vivo protein corona patterns of lipid nanoparticles. RSC Adv. 7,
mRNA COVID-19 vaccine. N. Engl. J. Med. 383, 2603–2615 (2020). 1137–1145 (2017).
20. J. A. Kulkarni, P. R. Cullis, R. van der Meel, Lipid nanoparticles enabling gene thera- 40. D. Pozzi et al., The biomolecular corona of nanoparticles in circulating biological
pies: From concepts to clinical utility. Nucleic Acid Ther. 28, 146–157 (2018). media. Nanoscale 7, 13958–13966 (2015).
21. Q. Cheng et al., Selective organ targeting (SORT) nanoparticles for tissue-specific 41. S. Palchetti et al., The protein corona of circulating PEGylated liposomes. Biochim.
mRNA delivery and CRISPR-Cas gene editing. Nat. Nanotechnol. 15, 313–320 (2020). Biophys. Acta 1858, 189–196 (2016).
22. T. Wei, Q. Cheng, Y. L. Min, E. N. Olson, D. J. Siegwart, Systemic nanoparticle delivery 42. C. D. Walkey, W. C. Chan, Understanding and controlling the interaction of nanoma-
of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editing. Nat. terials with proteins in a physiological environment. Chem. Soc. Rev. 41, 2780–2799
Commun. 11, 3232 (2020). (2012).
23. S. M. Lee et al., A systematic study of unsaturation in lipid nanoparticles leads to 43. Y. Zhang, J. L. Y. Wu, J. Lazarovits, W. C. W. Chan, An analysis of the binding function
improved mRNA transfection in vivo. Angew. Chem. Int. Ed. Engl. 60, 5848–5853 and structural organization of the protein corona. J. Am. Chem. Soc. 142, 8827–8836
(2021). (2020).
24. S. Liu et al., Membrane-destabilizing ionizable phospholipids for organ-selective 44. F. Sebastiani et al., Apolipoprotein E binding drives structural and compositional
mRNA delivery and CRISPR-Cas gene editing. Nat. Mater. 20, 701–710 (2021). rearrangement of mRNA-containing lipid nanoparticles. ACS Nano 15, 6709–6722
25. K. Zhou et al., Modular degradable dendrimers enable small RNAs to extend survival (2021).
in an aggressive liver cancer model. Proc. Natl. Acad. Sci. U.S.A. 113, 520–525 (2016). 45. M. Hammel et al., Mechanism of the interaction of β(2)-glycoprotein I with nega-
26. X. Cheng, R. J. Lee, The role of helper lipids in lipid nanoparticles (LNPs) designed for tively charged phospholipid membranes. Biochemistry 40, 14173–14181 (2001).
oligonucleotide delivery. Adv. Drug Deliv. Rev. 99, 129–137 (2016). 46. G. Caracciolo, D. Pozzi, A. L. Capriotti, C. Cavaliere, A. Lagana , Effect of DOPE and
27. Y.-N. Zhang, W. Poon, A. J. Tavares, I. D. McGilvray, W. C. W. Chan, Nanoparticle-liver cholesterol on the protein adsorption onto lipid nanoparticles. J. Nanopart. Res. 15,
interactions: Cellular uptake and hepatobiliary elimination. J. Control. Release 240, 1498 (2013).
332–348 (2016). 47. B. Singh, C. Fu, J. Bhattacharya, Vascular expression of the alpha(v)beta(3)-integrin in
28. A. Akinc et al., Targeted delivery of RNAi therapeutics with endogenous and exoge- lung and other organs. Am. J. Physiol. Lung Cell. Mol. Physiol. 278, L217–L226 (2000).
nous ligand-based mechanisms. Mol. Ther. 18, 1357–1364 (2010). 48. A. Couvelard et al., Expression of integrins during liver organogenesis in humans.
29. M. P. Monopoli, C. Aberg, A. Salvati, K. A. Dawson, Biomolecular coronas provide the Hepatology 27, 839–847 (1998).
biological identity of nanosized materials. Nat. Nanotechnol. 7, 779–786 (2012). 49. A. Simpson, M. A. Horton, Expression of the vitronectin receptor during embryonic
30. M. Jayaraman et al., Maximizing the potency of siRNA lipid nanoparticles for hepatic development: An immunohistological study of the ontogeny of the osteoclast in the
gene silencing in vivo. Angew. Chem. Int. Ed. Engl. 51, 8529–8533 (2012). rabbit. Br. J. Exp. Pathol. 70, 257–265 (1989).
31. B. L. Mui et al., Influence of polyethylene glycol lipid desorption rates on pharmaco- 50. S. Dinkla et al., Phosphatidylserine exposure on stored red blood cells as a parameter
kinetics and pharmacodynamics of siRNA lipid nanoparticles. Mol. Ther. Nucleic Acids for donor-dependent variation in product quality. Blood Transfus. 12, 204–209
2, e139 (2013). (2014).
32. S. C. Wilson et al., Real time measurement of PEG shedding from lipid nanoparticles 51. L. Miao et al., Synergistic lipid compositions for albumin receptor mediated delivery
in serum via NMR spectroscopy. Mol. Pharm. 12, 386–392 (2015). of mRNA to the liver. Nat. Commun. 11, 2424 (2020).
33. D. Docter et al., Quantitative profiling of the protein coronas that form around nano- 52. S. Scho€ ttler et al., Protein adsorption is required for stealth effect of poly(ethylene
particles. Nat. Protoc. 9, 2030–2044 (2014). glycol)- and poly(phosphoester)-coated nanocarriers. Nat. Nanotechnol. 11, 372–377
34. Z. Ban et al., Machine learning predicts the functional composition of the protein (2016).
corona and the cellular recognition of nanoparticles. Proc. Natl. Acad. Sci. U.S.A. 117, 53. R. C. Kowal et al., Opposing effects of apolipoproteins E and C on lipoprotein binding
10492–10499 (2020). to low density lipoprotein receptor-related protein. J. Biol. Chem. 265, 10771–10779
Downloaded from https://www.pnas.org by 124.219.109.112 on May 3, 2023 from IP address 124.219.109.112.

35. P. Artimo et al., ExPASy: SIB bioinformatics resource portal. Nucleic Acids Res. 40, (1990).
W597–W603 (2012). 54. R. Zhang et al., Helper lipid structure influences protein adsorption and delivery of
36. S. Lara et al., Identification of receptor binding to the biomolecular corona of nano- lipid nanoparticles to spleen and liver. Biomater. Sci. 9, 1449–1463 (2021).
particles. ACS Nano 11, 1884–1893 (2017). 55. H. Parhiz et al., PECAM-1 directed re-targeting of exogenous mRNA providing two
37. P. Thiagarajan, A. Le, C. R. Benedict, β(2)-glycoprotein I promotes the binding of orders of magnitude enhancement of vascular delivery and expression in lungs inde-
anionic phospholipid vesicles by macrophages. Arterioscler. Thromb. Vasc. Biol. 19, pendent of apolipoprotein E-mediated uptake. J. Control. Release 291, 106–115
2807–2811 (1999). (2018).

10 of 10 j PNAS Dilliard et al.


https://doi.org/10.1073/pnas.2109256118 On the mechanism of tissue-specific mRNA delivery by
selective organ targeting nanoparticles

You might also like