You are on page 1of 19

Chem. Pharm. Bull.

69, 1141–1159 (2021)


Vol. 69, No. 121141

Review

Development of Lipid Nanoparticles for the Delivery


of Macromolecules Based on the Molecular Design
of pH-Sensitive Cationic Lipids
Yusuke Sato
Faculty of Pharmaceutical Sciences, Hokkaido University; Kita-12, Nishi-6, Kita-ku,
Sapporo 060–0812, Japan.
Received August 18, 2021

Considerable efforts have been made on the development of lipid nanoparticles (LNPs) for delivering of
nucleic acids in LNP-based medicines, including a first-ever short interfering RNA (siRNA) medicine, Onpat-
tro, and the mRNA vaccines against the coronavirus disease 2019 (COVID-19), which have been approved
and are currently in use worldwide. The successful rational design of ionizable cationic lipids was a major
breakthrough that dramatically increased delivery efficiency in this field. The LNPs would be expected to
be useful as a platform technology for the delivery of various therapeutic modalities for genome editing and
even for undiscovered therapeutic mechanisms. In this review, the current progress of my research, including
the molecular design of pH-sensitive cationic lipids, their applications for various tissues and cell types, and
for delivering various macromolecules, including siRNA, antisense oligonucleotide, mRNA, and the clustered
regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) system will be described. Mecha-
nistic studies regarding relationships between the physicochemical properties of LNPs, drug delivery, and
biosafety are also summarized. Furthermore, current issues that need to be addressed for next generation
drug delivery systems are discussed.
Key words lipid nanoparticle; pH-sensitive cationic lipid; endosomal escape; macromolecule delivery;
nucleic acid; ribonucleoprotein

1. Introduction loid polyneuropathy caused by mutations in the transthyletin


Abnormal genetic status, including mutations and dele- gene.6) In addition, both Comirnaty® (Pfizer/BionTech) and
tions in genomes and the mis regulation of gene expressions, Spikevax® (Moderna) are the first mRNA vaccines against the
causes a variety of diseases.1–4) These therapeutic targets are corona virus diseases 2019 (COVID-19) that were developed
sometimes difficult to treat using conventional small molecule for treatment of infectins caused by the severe acute respira-
drugs due to limitations regarding their selectivity, affin- tory syndrome coronavirus 2 (SARS-CoV-2).7) Both siRNA
ity, and untraslated RNAs.5) Therefore, alternative approaches medicines and mRNA vaccines are lipid nanoparticle (LNP)
using (modified) DNAs/RNAs, including short interfering preparations. The LNP can encapsulate RNAs in its core thus
RNAs (siRNAs), antisense oligonucleotides (ASOs), aptamers, protecting the cargo from enzymatic degradation by ribo-
decoys, and mRNAs, and genome editing, including clustered nucleases (RNases) present in biological environments and to
regularly palindromic repeat (CRISPR)/Cas, have been consid- efficiently deliver the cargo to the cytosol of target cells (Fig.
ered for use as novel therapeutics against various refractory 1A). The LNPs are typically composed of 4 types of lipids, in-
diseases. Among these, some nucleic acid therapeutics have cluding a cationic lipid (CL), a phospholipid (PL), cholesterol,
already been approved by the Ministry of Health, Labour and and a pegylated (PEG)-lipid (Fig. 1A). Among them, CL is
Welfare (MHLW) in Japan, including pegaptanib (Macugen®; the major component (approx. 50 mol% of the total lipid) and
aptamer), nusinersen (Spinraza®; ASO), patisiran (Onpattro®; is essential for successfully encapsulating negatively charged
siRNA), givosiran (Givlaari®; siRNA), viltolarsen (Viltepso®; nucleic acids through electrostatic interactions and to achieve
ASO), BNT162b2 (Comirnaty®; mRNA), and mRNA-1273 the efficient cytosolic delivery of nucleic acids through mem-
(Spikevax®; mRNA) as of the writing of this review. In par- brane fusion-mediated endosomal escape. Therefore, as of this
ticular, Onpattro® is a first-ever RNA interference (RNAi) writing, numerous attempts have been made to develop origi-
medicine that can be used in the treatment of familial amy- nal potent CLs. Quaternary ammonium-based permanently
CLs were classically used for nucleic acid delivery because
This review of the author’s work was written by the author upon receiving their highly positive charge facilitates the loading of nucleic
the 2021 Pharmaceutical Society of Japan Award for Young Scientists. acids on LNPs as well as uptake by cells in vitro.8,9) However,
 e-mail: y_sato@pharm.hokudai.ac.jp
© 2021 The Pharmaceutical Society of Japan
1142 Chem. Pharm. Bull. Vol. 69, No. 12 (2021)

Fig. 1. Typical Structures of RNA-Loaded LNPs and pH-Sensitive CLs


(A) RNA-loaded LNPs are typically composed of CL, PL, cholesterol and PEG-lipid. The LNPs composed of permanently CLs show highly cationic properties in the
blood stream causing poor biodistribution. The LNPs composed of pH-sensitive CLs shows electrostatically near neutral in blood stream, leading to improved biodistribu-
tion, and convert to cationic property in acidic endosomes, facilitating endosomal escape. (B–F) Chemical structures of DLinDMA (B), DLin-KC2-DMA (C), DLin-MC3-
DMA (D), ALC-0315, and SM-102 (F). (Color figure can be accessed in the online version.)

the positive charge greatly hinders the control of the blood SNALP) at a clinically relevant dose (50% effective dose; ED50
circulation, biodistribution, and activation of complement sys- of approx. 1 mg siRNA/kg.18) In 2010, Semple et al. reported
tems due to non-specific interactions with biocomponents10,11) that the linker structure between the tertiary amino group and
(Fig. 1A). Therefore, tertiary amine-based pH-sensitive CLs, the scaffolds was critical for maximizing fusogenic activity.19)
which are near neutral at physiological pH (e.g. blood stream) Specifically, the efficacy of a ketal ring linker was found to
but develop cationic properties in a weakly acidic environment be superior to a 1,2-diol-derived linker, which was adopted
(e.g. endosomes/lysosomes), were first reported by Bailey and in DLinDMA. The best performing CL, DLin-KC2-DMA,
Cullis in 199412) (Fig. 1A). After reports on the efficient en- showed an ED50 of approx. 0.02 mg siRNA/kg in a mouse
capsulation of ASOs in LNPs composed of the pH-sensitive factor VII (FVII) model, which was a 50-fold improvement
CLs,13,14) these pH-sensitive CLs have now become the current in gene silencing efficiency in hepatocytes (Fig. 1C). Further
gold standard.15,16) Hayes et al. clearly demonstrated that un- structural optimizations resulted in the identification of the
saturation in the scaffolds in CLs is critical for inducing mem- optimal CL, DLin-MC3-DMA (MC3)20) (Fig. 1D), which ex-
brane fusion for endosomal escape, and found that DLinDMA, hibited an ED50 of approx. 0.005 mg siRNA/kg in the FVII
which is composed of 2 scaffolds derived from linoleic acid model and is used as a main ingredient in Onpattro®. Based
(C18:2), showed the best endosomal escape and cytosolic de- on this structure, numerous potent CLs, including ALC-0315
livery of siRNA17) (Fig. 1B). In 2006, Zimmermann et al. first and SM-102 used for COVID-19 mRNA vaccines, have been
demonstrated successful induction of hepatic apolipoprotein developed so far21–31) (Figs. 1E, F). Based on the above find-
B (ApoB) gene silencing in mice and in non-human primates ings, the development of potent CLs is one of the major break-
after the intravenous injection of siApoB-loaded DLinDMA- throughs in the field of LNPs.
LNPs (referred to as a stable nucleic acid lipid particle; I began work on developing LNPs for the in vivo delivery

Biography
Yusuke SATO is an Assistant Professor in the Faculty of Pharmaceutical Sciences, Hokkaido Uni-
versity. He joined Laboratory for Molecular Design of Pharmaceutics organized by Prof. Hideyoshi
HARASHIMA when he was a 4th degree of undergraduate, and received the B. S., M. S. and Ph.
D. degrees from Hokkaido University in 2009, 11 and 14, respectively. After completing a Research
Fellowship for Young Scientists from Japan Society for Promotion of Science (JSPS), he was ap-
pointed as an Assistant Professor in the Faculty of Pharmaceutical Sciences, Hokkaido University
in 2014. His main research interest is the development of potent lipid nanoparticles for the in vivo
delivery of macromolecules, including short interfering RNAs (siRNAs), mRNAs, and clustered regu-
Yusuke Sato
larly interspaced short palindromic repeats (CRISPR)-associated (Cas) ribonucleoproteins (RNPs),
based on the molecular design of functional lipids. He promotes industry-academia collaboration activities with the aim
of implementing the use of his lipids and formulation technologies in collaboration with the Institute for the Promotion of
Business-Regional Collaboration at Hokkaido University and private sector companies.
Vol. 69, No. 12 (2021)1143
Chem. Pharm. Bull.

of macromolecules based on the molecular design of origi- in weakly acidified endosomes, bind to anionic lipids in the
nal pH-sensitive cationic lipids in 2010 in the laboratory for endosomal membranes and form ion pairs (Fig. 2B). When the
the molecular design of pharmaceutics organized by Prof. ion pairs adopt a cone shape that is sufficient to destabilize
Hideyoshi Harashima. In this review, current progress of my lipid bilayers and to induce a phase transition into non-bilayer
research, including the molecular design of pH-sensitive cat- structures (e.g. HII phase), efficient endosomal escape can be
ionic lipids, their applications for liver, extra-hepatic tissues achieved. 31 Phosphorus (31P) NMR can be utilized to under-
and cell types, and for the delivery of various macromol- stand lipid polymorphism of lipid dispersion. Due to the an-
ecules, including siRNA, antisense oligonucleotides, mRNA, isotropy of the chemical shift of the 31P of PLs that is derived
and clustered regularly interspaced short palindromic repeats from different limitations in molecular motion between lipid
(CRISPR)-associated (Cas) system are described. Mechanistic polymorphism, it is known that L and HII phases show a dis-
studies regarding the relationship between the physicochemi- criminative broad signal with a high-field peak and a low-field
cal properties of LNPs, drug delivery, and biosafety will be peak, respectively35) (Fig. 2B). A 31P-NMR analysis using lipid
also summarized. Current limitations that should be addressed dispersions composed of equimolar amounts of a pH-sensitive
for next generation drug delivery systems are also discussed. CL, zwitterionic 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphol-
choline (POPC), and negatively charged 1,2-distearoyl-sn-
2. Development of pH-Sensitive CLs and Their Appli- glycero-3-phosphatidylserine (DSPS) suspended in an acidic
cations for Liver buffer revealed that the phase transition temperature from L to
In 2012, a pH-sensitive CL, YSK05, was first synthesized HII phase (TH) for YSK05 was significantly lower (21–25 °C)
as the 1st generation CL32,33) (Fig. 2A). YSK05 can be syn- than that for a conventional pH-sensitive CL, 1,2-dioleoyloxy-
thesized in a one-step condensation reaction of N-methyl- 3-dimethylaminopropane (DODAP) (35–40 °C) (Fig. 2C). This
4-piperidone and linoleyl alcohols. The structure that two result indicates that YSK05 is more fusogenic than DODAP.
linoleyl alcohol-derived scaffolds that are attached to one A hemolysis assay which can be used to examine the fusoge-
carbon atom leads to an increase in the angle between the two nicity of LNPs using red blood cells as model biomembranes
scaffolds (109.5°) (Fig. 2A). Based on theory of critical pack- also revealed that YSK05-containing LNPs (YSK05-LNPs)
ing parameter (CPP),34) a framework for determining the type are more fusogenic. Indeed, the siRNA-loaded YSK05-LNPs
of aggregates formed by surfactants, this design emphasizes showed much higher gene silencing activity compared to
the formation of a CL with a cone shape. The cone shape of DODAP or 1,2-dioleoyloxy-3-trimethylammonium propane
the CLs is important for inducing membrane fusion-mediated (DOTAP)-LNPs in HeLa cells despite the fact that the cellular
endosomal escape.35,36) Membrane fusion can be achieved uptake of the YSK05-LNPs was consistent or lower than the
through the phase transition of the lipid bilayer of biologi- counterparts. Treatment with chloroquine or ammonium chlo-
cal membranes from a lamellar (L) to an inverted hexagonal ride, endosome buffering reagents,37–40) revealed that the acidi-
(HII) phase. The pH-sensitive CLs acquire a cationic charge fication of endosomes is an essential process for the successful

Fig. 2. Structure and Properties of YSK05


(A) Chemical structure of YSK05. A ketal linkage increases the angle between the two scaffolds and emphasizes a cone shape. (B) Schematic illustration of L to H II
phase transition by ion-pairing between CLs and anionic lipids. Both L and H II phases can be determined by 31P-NMR due to the anisotropy of chemical shifts which are
derived from the specific limitation in molecular motion in each phase. (C) 31P-NMR spectra of a lipid dispersion composed of pH-sensitive CL, DODAP or YSK05 at the
indicated temperature. Blue arrow indicates a peak derived from H II phase. (Color figure can be accessed in the online version.)
1144 Chem. Pharm. Bull. Vol. 69, No. 12 (2021)

cytosolic delivery of siRNAs for YSK05-LNPs. The YSK05-LNPs showed ED50 values of 0.06 mg
The YSK05-LNPs were applied to liver tissue. It is known siRNA/kg and a durable gene silencing for up to 2 weeks in
that size-controlled LNPs with diameters of 100 nm or less can a mouse FVII model through optimizing the lipid composi-
reach hepatocytes due to the structural features of the liver, tion.54) To apply this to the treatment of hepatitis C virus
including the presence of fenestrae which are pores with diam- (HCV), mice with chronic HCV infections were produced by
eters in the range of 100 to 150 nm in liver sinusoidal endothe- the inoculation of the HCV genotype 1b in chimeric mice with
lial cells (LSECs) and a lack of basement membranes between humanized liver cells and the siHCV-loaded YSK05-LNPs
LSECs and hepatocytes.41,42) In addition to this, the presence of were then intravenously administered. HCV genomic RNA
the apolipoprotein E (ApoE)-low density lipoprotein receptor levels in serum were suppressed up to 90% for periods of up
(LDLR) pathway as an endogenous uptake mechanism in hepa- to 2 weeks (Fig. 3C). A significant decrease in the levels of
tocytes greatly contributes to the development of liver-targeting HCV core proteins in liver tissue was also observed (Fig. 3D).
LNPs.43,44) It was found that the bare YSK05-LNPs were cleared The optimized YSK05-LNPs were also applied to the in
from blood stream very quickly (t1/2 of approx. 1.8 min) after vivo validation of candidate genes as therapeutic targets.55) A
intravenous administration through the ApoE-LDLR pathway45) comparative transcriptome analysis in liver between diabetic
(Fig. 3A). This rate was over 10-fold faster than that for N- and normal mice identified an elevated expression of mono-
acetyl-D -galactosamine (GalNAc)-modified YSK05-LNPs which acylglycerol O-acyltransferase 1 (Mogat-1), an enzyme that
were cleared through the GalNAc-asialoglycoprotein receptor is involved in triglyceride synthesis and its storage.56–58) Long
(ASGPR) pathway (Fig. 3A). It is well known that GalNAc term Mogat-1 gene silencing by the repeated administration
functions as a ligand of ASGPRs, which are highly and spe- of siMogat-1-loaded YSK05-LNPs in diabetic mice resulted in
cifically expressed on hepatocytes, and are used for active preventive effects of type 2 diabetes, including reduced blood
targeting to hepatocytes.46–50) Intrahepatic observations revealed glucose levels, triglycerides and cholesterol, and increased lev-
that bare YSK05-LNPs are adsorbed to microvillar surface of els of adiponectin. Related to this study, the durable silencing
hepatocytes in wild-type mice but not in ApoE-deficient mice, of Lipin1, which regulates fatty acid utilization in the triglyc-
indicating that the presence of the ApoE protein is essential eride biosynthesis pathway,59) by YSK05-LNPs successfully
for adsorption to occur. This adsorption was competitively revealed that Lipin1 silencing in liver tissue led to a marked
inhibited by treatment with heparin or heparinases, suggesting decrease in adipose tissue mass and adipocyte diameters.60)
that involvement of heparan sulfate proteoglycans (HSPGs),51,52) These results suggest that the established LNPs could be ap-
which are abundantly expressed on the surface of hepatocytes plied not only for the treatment of a specific disease but also
and are known to be a receptor of the ApoE protein.53) These for the validation and investigation of in vivo functions of
findings that liver accumulation is a complex process in which genes of interest.
the bare LNPs rapidly adsorb to the microvillar surface of he- PEGylation of the LNP surface is a widely used strategy for
patocytes through ApoE-HSPG interactions following the bind- inhibiting non-specific interactions with biocomponents and to
ing of ApoE proteins to the LNPs in the blood circulation after maximize the efficiency of active targeting.61–63) However, the
intravenous administration (Fig. 3B). PEGylation is known to severely inhibit the endosomal escape

Fig. 3. Application of YSK05-LNPs for the Delivery of siRNAs to Hepatocytes


(A) Comparison of blood clearance rate between bare and GalNAc-LNPs which can be cleared via the ApoE/LDLR and GalNAc/ASGPR pathways, respectively. (B)
Schematic illustration of the process of hepatic accumulation of the LNPs. ApoE bind rapidly to bare LNPs trapped by HSPGs on hepatocytes followed by LDLR-mediated
endocytosis. The GalNAc-LNPs gradually accumulate to hepatocytes via GalNAc-ASGPR interactions. (C) Inhibition of HCV infection by intravenous administration of
siHCV-loaded YSK05-LNPs. (D) Microscoppic observation of intrahepatic HCV core proteins after treatment with the YSK05-LNPs. These figures are reproduced, in
part, with permission from Elsevier and Springer Nature. (Color figure can be accessed in the online version.)
Vol. 69, No. 12 (2021)1145
Chem. Pharm. Bull.

process, a phenomenon known as the ‘PEG dilemma.’64,65) modulated by the substituents on the cis-double bond of the
Although pH-sensitive CLs with an acid dissociation constant maleic anhydride71–74) (Fig. 4A). The siRNA-loaded GalNAc-
(pKa) around 6.4 can be used to reduce the concentration of modified YSK05-LNPs were PEGylated though carboxylated
PEG on the surface of LNPs needed for long blood circulation dimethylmaleic anhydride (CDM) or carboxylated ethylmaleic
compared to permanently CLs, even such a lower concentra- anhydride (CEM), a maleic anhydride derivative, linker.75) The
tion of surface PEG can inhibit both cellular uptake and endo- use of a maleic anhydride linker promoted the detachment of
somal escape.32) PEGylation of a LNP surface via a pH-labile the modified PEG and the re-activation of the fusogenisity of
linkage is one of the more useful strategies for overcoming the LNPs under endosomal pH mimetic conditions. The ef-
this dilemma.66–70) Maleic anhydride reacts with amino groups ficiency of hepatocyte-targeting and gene silencing activity
and forms a labile amide bond in an acidic environment, were observed to be enhanced in vivo. Taken together, the pH-
and the pH-sensitive degradability of the amide bond can be labile PEGylation of GalNAc-modified LNPs through a maleic
anhydride linkage successfully shielded the LNPs, enhanced
active targeting to hepatocytes, and re-activated the LNPs by
the detachment of PEG in response to the acidic environment
in endosomes (Fig. 4B).
MicroRNAs (miRNAs) are a class of small non-coding
RNAs that are related to homeostasis and diseases and
function by regulating gene expression by binding to the
3′-untranslated region (3′-UTR) of target mRNAs, thus trig-
gering the degradation of the mRNAs or the suppression of
translation.76) miRNA-122 (miR-122) is a liver-specific miRNA
and is known to play important roles in lipid metabolism and
to facilitate the replication of HCV RNA.77–79) Therefore, the
inhibition of miR-122 by the anti-miRNA oligonucleotide
(AMO) is a promising strategy for the treatment of liver
diseases.80) Consistent with siRNAs, LNPs can increase the
bioavailability of AMOs. Three separate intravenous ad-
ministrations of AMO122-loaded YSK05-LNPs at doses of
1 mg AMO/kg in mice resulted in a nearly 90% inhibition
of miR-122, a value that was significantly higher than free
AMO122 (<40% inhibition).81) This resulted in an elevated ex-
pression of miR-122 target genes (AldoA, Bckdk, and Ndrg3)
in liver tissue and a reduced plasma cholesterol concentration.
The YSK05-LNPs also succeeded in inhibiting miR-122 by
Fig. 4. Improvement of Active Targeting Using pH-Labile PEGylation delivering 2′-OMe-4′-thioribonucleoside-modified AMO122.82)
(A) Maleic anhydride molecules react with primary amino groups and form an Taken together, these results demonstrate that the use of drug
amide bond. The amino bond can be cleaved under weakly acidic conditions. The
cleavage rate can be modulated by the substituents on the cis-double bond of the delivery technologies presents a practical and valuable alterna-
maleic anhydride. CDM shows more rapid cleavage rate than CEM. (B) Schematic tive to anti-miRNA therapeutics.
illustration of improved active targeting using pH-labile PEGylation. The pH-labile
PEGylation of the GalNAc-LNPs inhibits non-specific interaction in blood stream, In 2016, the 2nd generation pH-sensitive CLs, YSK13 and
facilitating GalNAc-mediated accumulation to hepatocytes. After being endocy- YSK15, were designed with the objective of overcoming the
tosed, the pH-labile linkages are rapidly cleaved and the LNPs are re-activated,
promoting endosomal escape. These figures are reproduced, in part, with permis- limitations of YSK05, including insufficient fusogenisity and
sion from Elsevier. (Color figure can be accessed in the online version.) potential chemical instability under acidic conditions due to

Fig. 5. Development of the 2nd Generation pH-Sensitive CLs, YSK13 and YSK15
(A) Chemical structures of the YSK13 and YSK15. Apparent pKa value can be modulated by carbon length between a tertiary amine and an ester bond. (B) Inhibition
of HBV infection by an intravenous administration of siHBV-loaded YSK05-LNPs. Robust and durable suppression of HBV antigens and HBV DNA was observed. (Color
figure can be accessed in the online version.)
1146 Chem. Pharm. Bull. Vol. 69, No. 12 (2021)

an acid-labile ketal linker.83) The YSK13 and YSK15 contain the treatment of chronic hepatitis B virus (HBV) infections.84)
α,β-unsaturated ester and allyl ester linkers, respectively, and Mice that are persistently infected with HBV were intrave-
two linoleic acid-derived scaffolds are attached to one sp2 car- nously administered with the siHBV-loaded YSK13-C3-LNPs
bon atom of the linker (Fig. 5A). This design makes the angle at a dose of 5 mg siRNA/kg. An approximately 80–90% sup-
between the two scaffolds be 120°, which is higher than that of pression of HBV antigens (HBsAg and HBeAg) and HBV
YSK05, and therefore would be expected to emphasize a cone genomic DNA was observed for periods of up to 10 d (Fig.
shape and to increase fusogenicity based on CPP theory.34) 5B). The oral administration of a clinically used nucleoside
A 31P-NMR analysis demonstrated that YSK13 show clearly analogue of the reverse transcription (RT) inhibitor, enteca-
a lower TH compared to that of YSK05, suggesting a higher vir,85) strongly suppressed HBV DNA in sera but failed to
fusogenicity. Both the YSK13 and YSK15 contain some de- reduce the levels of HBV antigens in the sera. The elimination
rivatives with different spacer lengths between an ester bond or suppression of HBV antigens is important for the treatment
and a tertiary amino group. A longer spacer length resulted of hepatitis because several HBV antigens downregulate the
in LNPs with an increased apparent pKa value, which can be adaptive immune response to HBV in patients.86–88) Therefore,
determined by a 6-(p-toluidino)-2-naphthalenesulfonic acid the above findings indicate that a gene silencing approach
(TNS) assay. It is known that the optimal pKa range for de- could be the basis for a novel class of anti-HBV drugs which
livering siRNA to hepatocytes is 6.2–6.5.20) A YSK13 deriva- would be superior to RT inhibitors.
tive, YSK13-C3, with a pKa of 6.45 and ED50 of 0.015 mg/kg More recently, a pH-sensitive CL library was developed
in a mouse FVII model, was selected as the optimal CL for from a benchmark CL, YSK12-C4.89) The YSK12-C4, which
hepatocytes. The impact of the apparent pKa value on cell- shows a high fusogenicity and a high pKa (approx. 8.2),
specificity is described below. The YSK13-C3 was used in has been used for treating hard-to-transfect immune cells

Fig. 6. Development of an Original pH-Sensitive CL Library


(A) Overall picture of the pH-sensitive CL library. (B) A plot of calculated pKa by ChemDraw versus the apparent pKa measured by a TNS assay. (C) Structure of one
of the best pH-sensitive CLs, CL4H6, for the delivery of siRNAs to hepatocytes. (D) Dose-dependent FVII silencing activity of the siFVII-loaded CL4H6-LNPs. (E) Cy-
tosolic delivery of siRNAs by the CL4H6-LNPs. The siRNAs are observed as diffused patterns in the cytosol of hepatocytes. These figures are reproduced, in part, with
permission from Elsevier. (Color figure can be accessed in the online version.)
Vol. 69, No. 12 (2021)1147
Chem. Pharm. Bull.

(this subject is described below). To develop the library, the reduced number of experiments, is a useful methodology for
YSK12-C4 was divided into 3 parts, including the distal and determining the optimal formulation from a large number of
proximal sides of the scaffolds and the hydrophobic region, manufacturing factors, including the choice of lipids, lipid
and each region was derivatized independently to develop a composition, nitrogen per phosphate (N/P) ratio, and appropri-
systematic library of pH-sensitive CLs (Fig. 6A). The pH- ate conditions for synthesis (e.g. buffer pH, flow rate ratio and
sensitive CLs with different structures around tertiary amine total flow rate during microfluidic synthesis). However, the
showed various apparent pKa values. A plot of the cLog p conventional DOE used for formulation optimization involves
value of the hydrophilic region versus the experimentally the analysis of only one response such as gene expression in
determined apparent pKa value revealed that the apparent pKa a target tissue.24,99,100) Therefore, a detailed understanding of
value decreased with increasing hydrophobicity of the hydro- the relationship between a main response and physicochemi-
philic region. On the other hand, the pKa values estimated cal properties was very limited. Therefore, DOE with mul-
by the ChemDraw software were significantly lower than the tiple responses were conducted to optimize liver-targeting
apparent pKa values and predicting the precise apparent pKa mRNA-loaded LNPs and to identify the important properties
value clearly failed (Fig. 6B). This could be explained by the (e.g. physicochemical properties) needed for gene expression.
fact that a tertiary amino group, a proton donor, is present on In addition to the factors examined by DOE (e.g. a type of
the interface of the LNPs where the proton is relatively hard to lipid and lipid composition), it was found that LNP size and
access and an increased cLog p value leads to the preferential PEG-to-PL ratio were other key factors for liver-specific gene
localization of the tertiary amine in the hydrophobic layer expression. The optimized mRNA-loaded CL4H6-LNPs de-
of scaffolds, resulting in resistance to protonation. Moreover, livered over 8000 ng/mL of human erythropoietin protein in
extremely high concentrations of the pH-sensitive CLs (ap- serum at 6 h after the intravenous administration of a 0.5 mg
prox. 50 mol% of total lipid) in the LNPs results in reduced mRNA/kg dose. The optimized mRNA-loaded CL4H6-LNPs
pKa values due to an increased charge repulsion between the also showed 2.5-fold higher nanoluciferase expression in liver
CLs that are in close proximity with each other.90) In contrast tissue compared to MC3-LNPs with no observable toxicity.
to the hydrophilic region, the structures of hydrophobic scaf- Since the discovery of the CRISPR/Cas system,101) targeted
folds had relatively minor effects on apparent pKa values and genome editing through engineered nucleases has become
unexpected interactions between the hydrophobic tails and the a major focus of attention in both biological research and
hydrophilic head group on apparent pKa values was limited. therapeutics.102,103) Intended genome editing can be achieved
After several examinations regarding structure–activity re- through several approaches, including gene disruption by non-
lationships, a novel pH-sensitive CL, CL4H6, was identified homologous end joining and the insertion of a target sequence
as one of the best CLs for delivering siRNA into hepatocytes by homology-directed repair. Moreover, the rapid generation
and was determined to be a 3rd generation CL (Fig. 6C). The of re-engineered Cas nucleases, including high-fidelity Cas9,
lipid composition-optimized CL4H6-LNPs delivered a dose protospacer adjacent motif (PAM)-modified Cas9, base edi-
of 0.0025 mg siRNA/kg in a mouse FVII model, which was tor, transcription activator or repressor (CRISPRa/CRISPRi),
a 6- and 2-fold higher efficiency compared to 2nd generation a programmable epigenetic memory writer (CRISPRoff),
YSK13-C3-LNPs and MC3-LNPs, respectively (Fig. 6D). Flu- a prime editor, and type I Cas system, has dramatically
orescent microscopic observations in liver tissue revealed that amplified application diversity and the significance of the
AlexaFluor647-labeled siRNAs had escaped from endosomes CRISPR/Cas system for therapeutics.104–111) The introduction
and diffused into the cytosol (Fig. 6E). A quantitative PCR of CRISPR/Cas machinery as a ribonucleoprotein (RNP; a
analysis revealed that 4.2% of the siRNAs that had accumu- complex of Cas nuclease and guide RNA) is known to be
lated in liver tissue was successfully loaded in RNA-induced preferable for both the efficiency of genome editing and for
silencing complex (RISC), which indicates a higher bioavail- achieving a lower off-target effect.112,113) However, the technol-
ability of siRNAs compared to previous reports regarding ogy for formulating RNP delivery has been not established
MC3 and its biodegradable analogue, L319.91,92) yet. To overcome this limitation, RNP-loaded LNPs were
Introducing exogenous mRNAs into target cells is a promis- synthesized by means of a clinically relevant manufactur-
ing strategy for expressing therapeutic proteins of interest. For ing process using a micromixer-equipped microfluidic mixer,
vaccinations, the introduction of antigen-encoding mRNAs iLiNP114,115) (Fig. 7A). A detailed examination of manufactur-
leads to the expression of antigen proteins and the activation ing conditions revealed that both a higher pH (5.5 or over)
of antigen presenting cells, resulting in the activation of both and a higher flow rate ratio (7 or over) compared to standard
innate and adoptive immunity in attempts to eliminate patho- conditions for the synthesis of nucleic acid-loaded LNPs were
gens or cancer cells.93–95) Current progress in mRNA optimi- required to avoid aggregation and the loss of DNA cleavage
zation (including cap, codon, UTRs, poly(A) tail, chemically activity of the RNPs.116) Severe aggregation and the loss of
modified nucleosides, purification) have substantially improved DNA cleavage activity of RNPs were observed under standard
the translatability of the RNAs with reduced immunogenicity, conditions used for the synthesis of nucleic acid-loaded LNPs.
resulted in realization of and successful development of the Furthermore, by using an iLiNP device with 3 inlets, the
current COVID-19 mRNA vaccines.96,97) LNP technology was introduction of a buffer solution from a center inlet between
also naturally critical for this realization.7,98) However, the two inlets used for RNP and lipid solutions resulted in the ag-
composition of the optimal formulation and the physicochemi- gregation of RNPs at the junction to be completely inhibited
cal properties of the LNPs for mRNA remain unclear. The by preventing the RNPs from coming into direct contact with
design of experiment (DOE), an experimental design method high concentrations of ethanol (Fig. 7A). These optimizations
that rationally allocates experiments by using mathematical in manufacturing parameters and the structure of the micro-
statistics and reaches reliable conclusions with a considerably fluidic device enabled us to synthesize RNP-loaded LNPs
1148 Chem. Pharm. Bull. Vol. 69, No. 12 (2021)

Fig. 7. Application of the CL4H6-LNPs for Cas RNP Delivery


(A) Strategy for the synthesis of the Cas RNP-loaded LNPs using a microfluidic device, iLiNP. The use of the iLiNP device with 3 inlets resulted in the complete inhibi-
tion of aggregation of RNPs at the junction by avoiding the direct contact of RNPs with high concentrations of ethanol. (B, C) Impact of complexation of ssONs and length
of crRNA on functional delivery of Cpf1 RNPs. KO activity was maximized by combination of ssRNA with 120 nt and elongated crRNA (B) and the KO activity was
well correlated with the total RNA length (C). (D) Inhibition of HBV infection by Cas9 RNP-loaded CL4H6-LNPs in HBV-infected cells. The RNP-loaded LNPs showed
a superior inhibition of HBV infection compared to the AAV2 vector. These figures are reproduced, in part, with permission from Elsevier. (Color figure can be accessed
in the online version.)

reproducibly. The optimization of Streptococcus pyogenes dramatically increased EGFP KO activity (Fig. 7B). A combi-
Cas9 (spCas9)-loaded LNPs was conducted through 2 cycles nation of the two approaches further increased the EGFP KO,
of DOE using a flowcytometric analysis of enhanced green providing an IC50 less than 0.5 nM RNP (Fig. 7B). The total
fluorescent protein (EGFP) knockout (KO) in HeLa stably RNA length (crRNA + ssON) was well correlated with the KO
expressing EGFP (HeLa-GFP) cells. The optimized CL4H6- activity (Fig. 7C), indicating the significance of the density of
LNPs showed a IC50 of approx. 0.2 nM RNP, which was a 10- negative charges of RNPs on functional delivery. Although an
to 100-fold lower concentration compared to previous reports increase in EGFP KO activity was also observed for spCas9
regarding non-viral spCas9 RNP delivery systems. To expand RNP, the impact of ssON complexation was much less than
the applicability of the optimized LNPs, Acidaminococcussp. that for Cpf1 RNP due to the longer guide RNA for spCas9
BV3L6 Cas12a (Cpf1) RNP delivery was examined. The Cpf1 RNP. To confirm that the optimized RNP-loaded LNPs have
has a unique PAM sequence that is different from that of therapeutic potential, the inhibitory effect of HBV in HBV-
spCas9, exhibits robust genome editing activity in mammalian infected cells was examined. HBV-infected HepG2-hNTCP-30
cells and potentially shows lower off-target effects compared cells were consecutively treated with spCas9 RNP-loaded
to spCas9.117) The Cpf1 utilizes a shorter single crispr RNA LNPs for 12 d, resulting in 60 and 80% inhibition of HBV
(crRNA) compared to the guide RNA (approx. 100 nt length) DNA and covalently closed circular DNA, respectively, a sig-
of spCas9, making the loading of this cargo in LNPs dif- nificantly higher inhibitory effect than the adeno-associated
ficult due to the poor electrostatic interactions.118) Therefore, virus 2 (AAV2) vectors used for comparison (Fig. 7D). These
both the complexation of single strand oligonucleotide (ssON) data suggest that using RNP-loaded LNPs for gene editing
with RNPs and the elongation of crRNA were examined in would be a novel strategy for the treatment of chronic HBV
attempts to increase the negative charge density in the Cpf1 infections. Optimization of RNP-loaded LNPs targeting liver
RNPs. Interestingly, while the original Cpf1 RNP-loaded tissue for gene KO and knock-in is currently an on-going
LNPs failed to induce a significant EGFP KO (<10% at 2 nM effort.
RNP), the elongation of crRNA or the complexation of ssON
Vol. 69, No. 12 (2021)1149
Chem. Pharm. Bull.

3. Application of LNPs to Extra-Hepatic Tissues and genes simultaneously without any obvious adverse effect.
Cell Types The tumor-microenvironment is known to contain large
Macrophages are one of the most important contributors numbers of macrophages, which are referred to as TAMs and
to homeostasis and various inflammatory diseases.119) Macro- are mostly the pro-tumorous (M2) phenotype.125–128) Modify-
phages in different tissues play various roles in inflammatory ing the functions of TAMs or the manipulation of TAMs from
diseases and are distinguished by different names, including M2 to the anti-tumorous M1 phenotype by siRNAs could be
peritoneal macrophages (PEMs), Kupffer cells, microglia, a novel immunotherapy for the treatment of cancer. To target
and tumor-associated macrophages (TAMs) in the peritoneal TAMs, the CL4H6-LNPs were modified with distearoylg-
cavity, liver, brain, and tumor tissue, respectively.120–123) Ma- lycerol PEG, which has longer scaffolds (C18:0), dissociates
nipulation of macrophages at the genetic level represents a more slowly from the LNPs, and contributes to longer blood
promising approach for the treatment of a number of diseases circulation.129) The CL4H6-LNPs accumulated in subcuta-
in which macrophages are involved. neously inoculated human renal cell carcinoma (OS-RC-2)
Gene silencing in PEMs was examined by focusing on the tumor tissues. Flowcytometric analysis revealed that high
size and route of administration used for the YSK05-LNPs.124) levels of the CL4H6-LNPs were taken up by TAMs (defined
The diameter of the YSK05-LNPs was modulated from 75 to as CD45+CD11b+F4/80+ cells) compared to other cell popula-
460 nm by changing the amount of PEG-lipid. Cellular uptake tions, including neutrophils (defined as CD45+CD11b+F4/80−
in PEMs (defined as F4/80+ cells in the peritoneal cavity) was cells), other leukocytes (defined as CD45+CD11b− cells), and
clearly size-dependent and reached a maximum at a diameter non-leukocytes (defined as CD45− cells, which include tumor
of 340 nm after intravenous administration. An intravenous cells, tumor-associated fibroblasts, and endothelial cells). A
administration of siCD45-loaded YSK05-LNPs in mice at a significant (approx. 70%) gene silencing of CD45 in TAMs
dose of 2 mg siRNA/kg resulted in approx. 80% CD45 silenc- was obtained after the intravenous administrations of the
ing at the protein level for particles with diameters of 200 and CL4H6-LNPs. Motivated by these findings, we examined the
340 nm, which was significantly higher than that for 75 nm manipulation of TAMs from M2 to M1 phenotype by the gene
(approx. 40% silencing). On the other hand, no significant dif- silencing of M2-related genes, signal transducer and activator
ference in gene silencing activity in PEMs was observed after of transcription 3 (STAT3) and hypoxia-inducible factor 1α
an intraperitoneal administration of the same LNPs in mice at (HIF-1α), which are elevated in TAMs and involved in various
a dose of 0.003 mg siRNA/kg, suggesting that impact of size pro-tumorous functions, including immunosuppression, the
on intravenous administration involves translocation from the promotion of tumor growth, and angiogenesis.130–132) The re-
blood circulation to the peritoneal cavity. The YSK05-LNPs peated administration of the siSTAT3/siHIF1α-loaded CL4H6-
showed an ED50 value of 6 µg siRNA/kg for CD45 gene si- LNPs in OS-RC-2 tumor-bearing mice induced a significant
lencing in PEMs after intraperitoneal administration. This anti-tumor effect. An analysis of the gene expression in tumor
single digit µg/kg range enabled us to easily analyze multiple tissues revealed the elevation of CD11b (a macrophage mark-

Fig. 8. Development of a pH-Sensitive CL, YSK12-C4


(A) Chemical structure of the YSK12-C4. (B) Efficient Socs1 silencing by siSOCS1-loaded YSK12-C4-LNPs in hard-to-transfect murine BMDCs. (C) Therapeutic anti-
tumor effect of SOCS1-silenced BMDCs in an E.G7-OVA tumor model. The SOCS1-silenced BMDCs by YSK12-C4-LNPs showed superior anti-tumor effect compared
to that by RNAiMAX. (D) Gene silencing activity of the YSK12-C4-LNPs in hard-to-transfect human immune cell lines, Jurkat, THP-1, KG-1, and NK92 cells. These
figures are reproduced, in part, with permission from Elsevier and Springer Nature. (Color figure can be accessed in the online version.)
1150 Chem. Pharm. Bull. Vol. 69, No. 12 (2021)

er), CD169 (an M1 macrophage marker), and tumor necrosis STING agonists such as CDNs are expected to function as
factor-α (TNF-α) and the suppression of CD31 (an endothelial potent cancer adjuvants. However, the delivery of the CDNs
cell marker) and transforming growth factor-β (TGF-β) in ad- into cytosol where the STING pathway is located is some-
dition to target genes (STAT3 and HIF-1α). These findings what challenging due to hydrophilic nature of the CDNs.140)
suggest that the use of CL4H6-LNPs leads to the infiltration A CDN, cyclic di-GMP (c-di-GMP)-loaded YSK05-LNPs
of M1 macrophages in the tumor microenvironment, the were developed for cancer immunotherapy.141) The optimized
suppression of angiogenesis, and the release of immunosup- c-di-GMP-loaded YSK05-LNPs induced a significantly higher
pression. Importantly, these reactions were derived from gene level of IFN-β production in a murine Raw264.7 macrophage
silencing in TAMs but not in human tumor cells because the cell line compared to Lipofectamine 2000 and DOTAP-based
siRNAs utilized only target murine genes. LNPs. The subcutaneous administration of the c-di-GMP-
The delivery of siRNAs to dendritic cells (DCs) is a prom- loaded YSK05-LNPs enhanced in vivo antigen-specific cyto-
ising strategy for improving the efficacy of DC-based cancer toxic T cell (CTL) activities and showed preventive antitumor
immunotherapy. However, the efficient delivery of siRNAs to effects in major histocompatibility complex class I (MHC-
DCs is somewhat challenging.133–135) A fusogenic CL, YSK12- I)-expressing E.G7-OVA-bearing mice, suggesting that the
C4, was originally designed to overcome this hurdle.136) induction of MHC-I restricted antitumor immunity. The loss
YSK12-C4 contains two linoleic acid-derived scaffolds that or down regulation of the MHC-I in tumors is a well-known
are attached to one carbon atom, which emphasizes the adop- mechanism for escaping from immunosurveillance by CTL,
tion of a cone shape and enhances fusogenisity (Fig. 8A). A which is observed in malignant melanomas. In this situation,
hydrophilic tertiary hydroxy group that is present between the activation of natural killer (NK) cells would be desirable
a tertiary amine and the scaffolds contributes to the higher for inducing an antitumor effect against malignant melano-
apparent pKa (approx. 8.0) (Fig. 8A), which leads to strong mas.142,143) The intravenous administration of c-di-GMP-loaded
cationic properties in the cell culture medium and promotes YSK05-LNPs resulted in the production of TNF-α and IFN-γ,
cellular uptake in DCs. The optimized YSK12-C4-LNPs which are secreted from activated NK cells.144) The activation
showed IC50 values of 1.5 nM siRNA and achieved up to of NK cells was confirmed by the observation of a significant
90% gene silencing at higher concentrations in mouse bone increase in the population of NKG2D- and CD69-positive NK
marrow-derived DCs (BMDCs), while a commercially cells. Significant antitumor effects in a lung metastatic mouse
available transfection reagent, Lipofectamine RNAiMAX, model with a B16-F10 melanoma was observed after the in-
achieved less than 60% gene silencing at a maximum and travenous administration of c-di-GMP-loaded YSK05-LNPs
showed an IC50 of 25 nM, which was much higher than that and was cancelled by the depletion of NK cells, thus confirm-
for YSK12-C4-LNPs (Fig. 8B). The efficient gene silencing ing that the c-di-GMP-loaded YSK05-LNPs are capable of
was expected to enhance the efficacy of DC-based immuno- activating NK cells and inducing antitumor effects against
therapy by targeting the suppressor of cytokine signaling 1 tumors with the loss or downregulation of MHC-I. It was re-
(SOCS1), which blocks the janus kinase (JAK)–STAT sig- cently demonstrated that c-di-GMP-loaded YSK12-C4-LNPs
naling pathways.137) The siSOCS1-loaded YSK12-C4-LNPs overcome anti-programmed cell death 1 (PD-1) resistance and
achieved approx. 80% silencing of the SOCS1 gene, resulting show a synergistic antitumor effect by activating NK cells
in an enhanced production of TNF-α, including interleukin-6 followed by inducing the formation of PD ligand 1 on tumor
(IL-6) in response to interferon (IFN)-γ stimulation. Immuni- cells in the same melanoma model.145)
zation of the SOCS1-silenced DCs by the YSK12-C4-LNPs in A technology for the treatment of brain disorders, including
mice bearing the EL4 murine lymphoma cell line expressing traumatic brain injury, Parkinson’s disease, and Alzheimer’s
chicken ovalbumin (E.G7-OVA) resulted in a significant anti- disease, is still an unmet medical need. Nucleic acid and
tumor effect, while the SOCS1-silenced DCs by Lipofectamine gene therapy is a promising strategy for realizing efficacious
RNAiMAX failed (Fig. 8C). These results clearly suggest that therapeutics for the hard-to-treat brain disorders, as dem-
the YSK12-C4-LNPs are a promising technology for use in onstrated by the clinically approved zolgensma (wild-type
enhancing DC-based immunotherapy. The silencing of the AAV9-based gene therapy) and spinraza (ASO for exon inclu-
indoleamine 2,3-dioxygenase 1 gene by the YSK12-C4-LNPs sion) for the treatment of spinal muscular atrophy, intractable
in BMDCs also led to an enhanced antitumor effect against genetic diseases caused by the deletion of a responsible gene,
E.G7-OVA tumors.138) Motivated by these results, the gene the survival motor neuron 1.146) A disorder of the brain endo-
silencing activity of the YSK12-C4-LNPs in several hard-to- thelium is known to be associated with the pathophysiology
transfect human immune cell lines, including Jurkat, THP-1, of brain disorders.147) Therefore, targeting brain endothelial
KG-1, and NK92, was examined.139) Although substantial cy- cells (BECs) would be a useful approach for the treatment of
totoxicity was observed at higher concentrations in the cases brain disorders. The gene silencing activity of siRNA-loaded
of KG-1 and NK92 cells, the YSK12-C4-LNPs showed an ef- YSK05-LNPs in BECs was examined as a possible approach.
ficient gene silencing activity of up to 80 to 90% (Fig. 8D). It YSK05-LNPs were modified with recombinant ApoE proteins
was suggested that small-size and colloidal stability in the cell as ligands against LDLR family-expressing BECs.148) An
culture medium of the YSK12-C4-LNPs contributed to the ApoE-dependent increase in both cellular uptake and gene si-
enhanced cellular uptake followed by efficient gene silencing lencing activity was clearly obtained. Motivated by the impact
in these cells. of ApoE modification on delivery efficiency, ApoE-modified
The stimulator of the interferon gene (STING) pathway YSK05-LNPs containing pDNA were intracerebroventricularly
recognizes cytosolic DNA and cyclic dinucleotides (CDNs) to administered to mice for transgene expression in brain.149)
induce type I IFN and inflammatory cytokine responses and A significant increase in transgene expression was observed
appears to be essential for the innate sensing of tumor cells.140) after the ApoE modification in vivo. Immunostaining analysis
Vol. 69, No. 12 (2021)1151
Chem. Pharm. Bull.

revealed transgene (mCherry) expression in neural stem cells idic devices that can achieve fast and reproducible mixing of
and/or neural progenitor cells, suggesting usefulness of this two or more miscible solutions (e.g. ethanol and water) have
system for the treatment of neurodegenerative diseases. greatly contributed to overcoming this limitation.114,161,162)
Targeting the lung endothelium is promising approach However, small-sized LNPs show a lower integrity and drug
for the treatment of various diseases, including sepsis, pul- delivery efficiency. Chen et al. reported on the rapid disso-
monary hypertension, and lung cancer.150) Previously, our ciation of lipid components, including CL, PL, and PEG-lipid,
laboratory coincidentally found that a pH-sensitive fusogenic from small-sized LNPs after incubation in mouse plasma
GALA peptide, functions as a specific ligand for the lung, compared to a larger counterpart, resulting in poor gene
and, using it, we were able to successfully induce gene si- silencing activity in hepatocytes.163) However, the detailed
lencing of an endothelial cell marker, CD31, in lungs with an mechanism responsible for this poor delivery that is associated
ED50 of 0.21–0.4 mg siRNA/kg.151,152) Lung-targeting LNPs with downsizing remains unknown. Therefore, siRNA-loaded
composed of YSK05 were recently finely optimized, and an LNPs with different diameters, approx. 35 and approx. 65 nm,
ED50 of 0.01 mg siRNA/kg was achieved, a value that was were synthesized by changing amount of PEG-lipid to 3% and
20- to 40-fold lower than that for previous formulations.153) A 1%, respectively, to examine the impact of particle size164)
clinically relevant dose (0.5 mg siRNA/kg) was sufficient to (Fig. 9A). The 3%PEG-LNPs showed a 4-fold lower FVII gene
eradicate a metastatic lung cancer model without any signs of silencing activity compared to the 1%PEG-LNPs. Fluorescent
toxicity. Development of novel lung targeting formulations for microscopic observations of intrahepatic siRNA distribution
mRNA delivery is currently an on-going project. revealed that most of the siRNAs showed a punctate pattern
in the case of the 3%PEG-LNPs, indicating failure of endo-
4. Elucidation of Structure–Activity Relationships somal escape, while substantial amounts of siRNAs showed
The particle diameter of LNPs is a critical factor that af- a diffused pattern in the case of 1%PEG-LNPs, indicating the
fects their biodistribution and infiltration into deep tissue. It successful cytosolic delivery of siRNAs. The quantification
is possible for small-sized nanoparticles to be translocated of siRNAs also permitted the amount of siRNAs delivered
from the lung to lymph nodes where they then drain into by 3%PEG-LNPs to be measured and the findings indicated
the lymphatic system and penetrate deeply into stromal-rich that the amount was significantly lower than that delivered by
tumor tissues.154–158) It is known that subcutaneously admin- 1%PEG-LNPs, suggesting the premature release of siRNAs
istered small-sized LNPs show an excellent transitivity to and from the 3%PEG-LNPs in the blood stream. An in vitro study
distribution within the lymph nodes.159) Furthermore, the sub- confirmed that the siRNAs released from the 3%PEG-LNPs
cutaneous administration of small-sized siRNA-loaded LNPs showed a lower gene silencing activity only in the presence of
modified with GalNAc successfully reached hepatocytes and fetal bovine serum (FBS) but not in the absence of FBS (Fig.
induced substantial gene silencing while a larger counterpart 9B), suggesting that serum components promote the destabi-
failed.160) Despite the fact that small-sized LNPs are desired lization and inactivation of small-sized LNPs. Measurement
for efficient drug delivery due to its great potential for tissue of the relative surface area using a TNS probe revealed that
penetration, Efforts to develop small-sized LNPs that can be the actual relative surface area of 3%PEG-LNPs was 2.99-fold
used for efficient drug delivery have been limited. The fact higher than that of the 1%PEG-LNPs (Fig. 9C), which was
that it is difficult to reproducibly produce small-sized LNPs significantly higher than the theoretically calculated relative
with low polydispersity represents a fundamental limita- surface area (1.72-fold) (Fig. 9D), indicating that lower lipid
tion.115) The recent development of mixer-equipped microflu- packing on the interface may have occurred, possible due to

Fig. 9. Impact of Particle Size of the LNPs on the Delivery of siRNAs


(A) Diameter of the LNPs decreases depending on the amount of PEG-lipid. (B) Gene silencing activity of the LNPs with varied diameter in the presence or absence
of FBS in HeLa-dluc cells. (C) The measured total surface area of the LNPs using TNS. (D) Theoretically calculated total surface area of the LNPs. (E) Measurement of
hydration level on the interface of the LNPs using laurdan. (F) Schematic illustration of packing stress derived from the substantial difference between the actual positive
curvature of the YSK05 on the interface of the LNPs and the spontaneous negative curvature of the YSK05. These figures are reproduced, in part, with permission from
Elsevier. (Color figure can be accessed in the online version.)
1152 Chem. Pharm. Bull. Vol. 69, No. 12 (2021)

packing stress derived from strain between the actual positive were optimal from the viewpoint of hepatocyte-specificity.
curvature on the interface of the LNPs and the spontaneous The LNPs with optimal pKa (approx. 6.4) values for gene
negative curvature of the fusogenic CLs (Fig. 9E). Measure- silencing activity in hepatocytes naturally accumulated in he-
ment of the hydration level of the interface of the LNPs using patocytes but also were substantially co-localized with LSECs.
laurdan supported the above finding (Fig. 9F). The interface The LNPs with pKa values of 6.8 specifically accumulated
of the small-sized LNPs can cause instability, thus facilitating in LSECs. Gene silencing activity in LSECs was examined
the dissociation of lipid components and the premature release using siRNA targeting CD31, and a plot of CD31 expression
of siRNAs, and the adsorption of large quantities of serum in the liver versus the apparent pKa value of the LNPs clearly
proteins on the interface, resulting in the inhibition of fu- showed that gene silencing activity in LSECs was clearly pKa-
sogenisity, a process that was confirmed by a hemolysis assay dependent and became maximum at pKa values in excess of
and an siRNA release assay established by the Merck research 7.0 (Fig. 10A). A similar experiment was conducted using a
group.165) Cholesterol is known to contribute to the stabil- pH-sensitive CL library with different hydrophilic regions, and
ity of LNPs.166,167) The premature release of siRNAs from it was revealed that the optimal pKa range for gene silencing
small-sized LNPs was successfully inhibited by increasing the activity in LSECs was from 7.0 to 7.5 (Fig. 10B), which was
molar ratio of cholesterol in the lipid composition. However, just 1 unit higher than that in hepatocytes. Although the ap-
the gene silencing activity of the small-sized LNPs was still parent pKa value of the LNPs can be controlled by the design
significantly lower than that of the larger counterparts, sug- of chemical structure of the CL as examples of the 2nd gen-
gesting that reduced endosomal escape needs to be addressed eration CLs and CL library,83,89) precise adjustment of appar-
if small-sized LNPs with efficient nucleic acid delivery charac- ent pKa value is somewhat challenging due to time-consuming
teristics are to be developed. and labor-intensive processes and is sometimes impossible
The apparent pKa value is another key factor that can af- due to limitations in the permissibility of chemical structure.
fect the biodistribution and endosomal escape of LNPs. As Therefore, the manipulation of the apparent pKa value of the
described above, the optimal pKa range for hepatic siRNA LNPs using a combination of two pH-sensitive CLs with dif-
delivery is known to be from 6.0 to 6.5.20) Therefore, the ferent apparent pKa values would be an alternative strategy for
original pH-sensitive CLs, YSK05, YSK13-C3, and CL4H6, precisely adjusting apparent pKa to the target values. LNPs
that can deliver nucleic acids into hepatocytes efficiently composed of both YSK05 and YSK12-C4 (YSK05/12-LNPs)
also show pKa values in the range from 6.35 to 6.45, which were developed in attempts to achieve new pKa values.168) In-
are within the desired optimal range.32,54,83,84,89) However, the terestingly, a plot of pKa of the final LNP versus the YSK12-
impact of the apparent pKa on intrahepatic distribution has C4 ratio in total CLs indicated that contribution of YSK12-C4
not yet been examined. The effect of the apparent pKa on the was higher than that of YSK05 for the final pKa pf the LNPs
intrahepatic distribution of the LNPs was examined using the (Fig. 10C). For example, the actual final pKa of the LNPs com-
2nd generation pH-sensitive CLs, YSK13 and YSK15, with posed of equal molar amount of YSK05 and YSK12-C4 was
varied apparent pKa values ranging from 5.70 to 7.2583) (Fig. clearly higher than the pKa value that was simply calculated
5A). The intrahepatic distribution of the LNPs clearly changed as an average of the apparent pKa value of each lipid (Fig.
from hepatocytes to LSECs. The LNPs with lower pKa values 10C). Similar results were obtained when mixtures of YSK13,

Fig. 10. Manipulation of the Apparent pKa Value for Cell-Specific Delivery of siRNAs
(A, B) The pKa-dependency of gene silencing activity in LSECs. The consistent optimal pKa range for gene silencing in LSECs was observed for the 2nd generation
YSK13/YSK15 (A) and CL library (B). (C) Higher contribution of YSK12-C4 for final pKa was observed in the YSK05/YSK12-C4-LNP system. (D) Higher contribution of
YSK13-C4 for the final pKa was observed in the YSK13-C2/YSK13-C4-LNP system. (E) Mixture of YSK13-C2 and YSK13-C4 resulted in a new pKa that superimposes
with the theoretical curve obtained by the Henderson-Hasselbalch equation. These figures are reproduced, in part, with permission from Elsevier and Dovepress. (Color
figure can be accessed in the online version.)
Vol. 69, No. 12 (2021)1153
Chem. Pharm. Bull.

YSK13-C2 and YSK13-C4 lipids were used (Fig. 10D). Impor- hydrophobic nature of the HII phase. These findings lead to
tantly, the contribution of the pH of LNPs composed of two the hypothesis that PCs (L phase) are segregated from the HII
CLs with different pKa values was superimposed on the theo- structures formed by CH4H6-DSPS ion pairs and cover the
retical curve for the new pKa but not on the averaged curve of HII structure, which leads to a dispersion of small vesicles
original two pKa (Fig. 10E), strongly suggesting that the new that are well-suspended, even at high temperatures (Fig. 11A).
pKa value was contributed by the mixing of two CLs. A small-angle X-ray scattering (SAXS) analysis of the same
Neutral zwitterionic lipids with a bulky phosphochline, ph- lipid dispersion revealed the existence of a mixture of L and
sophatidylcholine (PC) and sphingomyelin (SM) are known to HII phases at higher temperature (Fig. 11B), supporting the hy-
stabilize the L phase and inhibit the phase transition to an HII pothesis that CL4H6 is immiscible with PCs when in the HII
phase due to their cylindrical structure.35) As described above, structure. The immiscible property of the CLs would greatly
a phase transition is essential for achieving membrane fusion- contribute to membrane fusion-mediated endosomal escape
mediated endosomal escape. Therefore, a high amount of PCs because the main lipid component of endosomal membranes is
or SMs in siRNA-loaded LNPs greatly inhibits endosomal PCs that can inhibit phase transition. This hypothesis is also
escape followed by the cytosolic release of siRNAs. Indeed, supported by the fact that mRNA-loaded CL4H6-LNPs with
the YSK05-LNPs showed robust gene silencing activity, but high amounts (25 mol%) of DSPC showed consistent gene ex-
only when phosphatidylethanolamines and not PCs were used pression in liver tissue compared to a lower amount (5 mol%)
as a helper lipid.32) Interestingly, siRNA-loaded LNPs com- in liver tissue.171)
posed of CL15H6, which has much longer scaffolds (C24 + O1)
compared to the conventional linoleic acid-derived scaffolds 5. Improving the Biosafety of LNPs
(C18), and 50 mol% egg-derived SM showed excellent gene While the administration of nucleic acid-loaded LNPs
silencing in HeLa-dluc cells, while that with CL15A6, a coun- sometimes causes toxicity,172–176) our knowledge regarding
terpart with conventional scaffolds, failed.169) Gene silencing LNP-associated inflammatory toxicities remains limited and
activity was also clearly observed when any PCs with C16 or is somewhat controversial. Two reports demonstrated that a
C18 scaffolds were used but not when brain- and milk-derived co-treatment of dexamethasone (Dex), a glucocorticoid recep-
SMs which contain longer scaffolds (C22 to C24) were used. tor agonist, and a JAK 2 inhibitor with siRNA-loaded LNPs
It is known that lipids with different scaffold lengths have a dramatically inhibited LNP-associated toxicities including
tendency to be segregated from each other due to the higher elevated serum chemistry parameters and inflammatory cyto-
Gibbs free energy for mixing.170) The inhibitory effect of PCs kines.177,178) Another study reported on the controversial find-
and SMs on the L-to-HII phase transition should be affected ing that a pro-inflammatory cytokine response was observed
only when fusogenic CLs are miscible with the PCs and SMs after the administration of LNPs even with pre-medication
during the phase transition. Taking the above into consider- with Dex and other drugs.179) Differences in the components,
ation, CLs with much longer scaffolds would be immiscible characteristics, and biodistribution of LNPs, and in the animal
with typical PCs and SMs consisting of C16 to C18 scaffolds model being used likely explain the difficulties associated with
and would be resistant to the inhibitory effect of PCs and SMs the interpretation of these findings. Elucidation of the mecha-
on membrane fusion (Fig. 11A). CL4H6, which is made up of nism regarding LNP-associated toxicity would contribute to
scaffolds with CL15H6, formed well-dispersed lipid suspen- the development of new strategies for improving the safety of
sions composed of equal molar ratios of CL4H6, distearoyl PC LNPs.
(DSPC), and DSPS in an acidic buffer even at higher tempera- To elucidate mechanism responsible for LNP-associated
ture (50 °C), and therefore, a 31P-NMR analysis failed to detect hepatotoxicity, elevated doses of siRNA-loaded YSK13-C3-
a phase transition because the anisotropic 31P spectra from LNPs were intravenously administered to mice.180) Interesting-
the L or HII phases were averaged as the result of the rapid ly, dose-dependent hepatotoxicity was observed with changes
rotation of relatively small-sized dispersed particles (Fig. 11B, in the intrahepatic distribution of the LNPs from hepatocytes
insertion). A lipid suspension consisting of CLs with conven- to LSECs (Figs. 12A, B). Hepatotoxicity was also observed
tional scaffolds, including YSK05, adopts a completely aggre- at all dose range regardless of the siRNA-loading, indicating
gated form at temperatures higher than TH due to the highly that the toxicity is derived from the LNP itself and appears

Fig. 11. Impact of Scaffold Length of CLs on Immiscibility with Neutral PLs
(A) Schematic illustration of the impact of scaffold length of CLs on the segregation of neutral PLs from H II structures of CL-anionic PL complexes. (B) SAXS analysis
of CL4H6/DSPC/DSPS dispersion that the inserted isotropic 31P-NMR spectra were obtained. These figures are reproduced, in part, with permission from Elsevier. (Color
figure can be accessed in the online version.)
1154 Chem. Pharm. Bull. Vol. 69, No. 12 (2021)

Fig. 12. Strategies for Improving the Safety of the LNPs


(A) Dose-dependency of intrahepatic distribution of the LNPs. The LNPs accumulated to LSECs at a higher dose. (B) Dose-dependency of the hepatotoxicity of the
LNPs. The hepatotoxicity was correlated with the change in intrahepatic distribution. (C) Schematic illustration of the estimated mechanism of LNP-associated hepato-
toxicity. The LNPs partially accumulate in LSECs, leading to the expression of adhesion molecules and cytokines followed by neutrophilic inflammation that aggravates
hepato- and systemic toxicity. (D) GalNAc modification significantly decreased the accumulation of the LNPs in LSECs. (E) Impact of surface modification of the LNPs
on hepatotoxicity. GalNAc modification significantly decreased the LNP-associated hepatotoxicity. Further PEG modification completely suppressed the hepatotoxicity.
(F) Schematic illustration of the development of the LLC-NPs. (G) Weight of each component. The amount of lipids for the LLC-NPs is significantly lower than that for
conventional HL-NP formulations. The amount of protamine is much lower than that for lipids or siRNAs. (H) Estimated cleavage of the CL4H6 by endogenous lipases.
(I) Biodegradation of the CL4H6 in both the liver and spleen. These figures are reproduced, in part, with permission from Elsevier. (Color figure can be accessed in the
online version.)

to be associated with changes in intrahepatic distribution. In Neutrophil depletion resulted in the significant suppression
toxic conditions, adhesion molecules, including the vascular of LNP-associated toxicity, suggesting the involvement of
cell adhesion molecule-1, E-selectin, and intercellular adhe- neutrophilic inflammation on hepato- and systemic toxicity. It
sion molecule-1, are upregulated in liver tissue. A multiplex should be noted here that Kupffer cells did not contribute to
suspension assay revealed the production of neutrophil-related the toxicity due to rapid depletion of these cells after the ad-
cytokines, including keratinocyte-derived cytokines and ministration a high dose of the LNP. Taken together, it thought
granulocyte colony stimulating factor, in addition to some that the unintended accumulation of LNPs to LSECs cause
inflammatory cytokines, the IL-6, IFN-γ-induced protein 10. the upregulation in adhesion molecules and pro-inflammatory
Vol. 69, No. 12 (2021)1155
Chem. Pharm. Bull.

cytokines including neutrophil-related cytokines followed by The 3rd generation CL, CL4H6, contains biodegradable ester
the induction of neutrophilic inflammation (Fig. 12C). Based bonds in its hydrophobic scaffolds. Cleavage of the ester bonds
on this supposition, the LNPs were modified with GalNAc to produces a water-soluble alkanol amine and two oleic acids89)
improve hepatocyte-specific accumulation and to avoid the (Fig. 12H). The amount of CL4H6 in both the liver and spleen
unintended accumulation to LSECs. Hepatocyte-specificity tissues rapidly dropped with time (Fig. 12I). The expected
was substantially improved by the GalNAc modification (Fig. alkanol amine was detected at 30 min in both tissues, indicat-
12D). Surprisingly, LNP-associated hepatotoxicity was dra- ing that the CL4H6 was rapidly hydrolyzed after the delivery
matically suppressed by GalNAc modification (Fig. 12E). of siRNAs, as expected. The incubation of CL4H6-LNPs in
Further modification of GalNAc-LNPs with PEG to minimize 90% mouse plasma revealed that the extent of degradation
the ApoE-mediated unintended accumulation to LSECs com- of CL4H6 was negligible for periods of up to 2 h, suggesting
pletely inhibited hepatotoxicity (Fig. 12E). Importantly, gene that CL4H6, when contained within the LNPs is protected
silencing activity in hepatocytes was fully maintained (ED50 from enzymatic degradation and is rapidly degraded once
of 0.015 mg siRNA/kg in a mouse FVII model), even after the integrity of the LNPs is lost by membrane fusion with
surface modification. A single intravenous administration of endosomal membranes. A single dose toxicity test revealed
the modified YSK13-C3-LNPs in mice persistently infected that the CL4H6-LNPs were tolerated for at least up to levels
with HBV resulted in the suppression of HBV antigens by ap- of 5 mg siRNA/kg while the undegradable MC3-LNPs showed
prox. 90% without any signs of toxicity. a dose-dependent elevation in both alanine transaminase
It is known that high doses or high concentrations of CLs and aspartate transaminase and a decrease in body weight.
can cause undesired toxicity through multiple mechanisms, Furthermore, repeated dose toxicity also confirmed that the
including the production of reactive oxygen species, the in- CL4H6-LNPs were well tolerated.
duction of apoptosis and pro-inflammatory cytokines.172–176) These strategies for improving the safety of LNPs, includ-
Therefore, the reduction of CLs for functional delivery would ing maximizing hepatocyte-specificity, reduction of the CL
be a useful and straightforward strategy for improving safety. dose, and the addition of biodegradability to CLs, are not ex-
However, the reduction of CLs by reducing the molar ratio or clusive and could be combined with each other. It is expected
reducing the total lipids against nucleic acids typically causes that these findings will contribute to the further development
a dramatic decrease in delivery efficiency because the CLs of LNPs with excellent safety profiles.
are critical components for endosomal escape, which is a late-
limiting step in nucleic acid delivery181,182) (Fig. 12F). To ad- 6. Perspectives
dress this dilemma, the negative charges of siRNAs were neu- LNP technology has been enjoyed dramatic progress in the
tralized by protamines, cationic proteins, in an effort to reduce last 10 years, which in part, was driven by a breakthrough
the consumption of CLs for siRNA-loading and to reduce the of development of potential CLs. However, the limitations
net dose of CLs183) (Fig. 12F). The siRNA/protamine core- that yet need to be addressed to realize ideal drug delivery
loaded LNPs with low lipid/siRNA charge ratios are referred systems and to overcome widespread refractory diseases still
to as low lipid core-nanoparticles (LLC-NPs) (Fig. 12F). The remain. There is still much room for the development of the
LLC-NPs showed consistent gene silencing activity (IC50 of efficient extra-hepatic targeting of therapeutically important
approx. 1 nM siRNA) when conventional siRNA-loaded LNPs tissues, including heart, muscles, and brain, and cell types,
with high lipid/siRNA charge ratios (high lipid-nanoparticles; including specific subpopulations of immune cells. In addi-
HL-NPs) were used in HeLa cells stably expressing dual- tion, payloads that can be loaded and delivered by LNPs are
luciferase (HeLa-dluc). On the other hand, the gene silencing still limited. Developing general loading/delivery strategies for
activity of the siRNA-loaded LNPs with low lipid/siRNA various classes of payloads (e.g. proteins, peptides, and others)
charge ratios (low lipid-nanoparticles; LL-NPs) were dramati- remains an important issue. It is also important to understand
cally reduced (approx. 10% gene silencing at 30 nM siRNA). the fundamental reasons for in vivo fate of the LNPs (e.g. bio-
These data suggest that the neutralization of the negative distribution, stability, endosomal escape, toxicity, and so on)
charges of the siRNAs by protamines significantly restored at the molecular and particle levels. Therefore, the importance
the gene silencing activity of the LNPs with low lipid/siRNA of related technologies such as synthetic devices (e.g. sophis-
ratios. A liposome fusion assay revealed that the neutraliza- ticated and flexible microfluidic devices) and analytical meth-
tion restored fusogenic activity, indicating that the amount of odologies (e.g. NMR, SAXS, small-angle neutron scattering,
CLs available for endosomal escape was increased by the re- thermal analyses, transmission electron microscopy, robotics,
duced consumption of CLs for siRNA loading by protamines. machine learning and so on) promise to become more and
Importantly, while the total amount of lipid was significantly more important.186–192) I sincerely hope that the LNP technol-
decreased from 152.3 µg (HL-NPs) to 38.1 µg (LLC-NPs), only ogy will continue to develop and contribute to the provision of
7.9 µg of protamines were added against 10 µg siRNA (Fig. advanced and safe medical care in the future.
12G). Moreover, protamines are biodegradable and were elim-
inated from liver tissue with a half time (t1/2) of less than 4 h. Acknowledgments The author wishes to acknowledge the
The restoration of gene silencing activity in liver tissue and financial support by, in part, a Grant-in-Aid for JSPS Fellows,
the reduced hepatotoxicity for LLC-NPs was also confirmed. a Grant-in-Aid for Research Activity Start-up, Grant-in-Aid
Furthermore, a similar strategy was also successful in the case for Young Scientists (B) (Grant Number: 15K20831), a Grant-
of human immune cells.184) in-Aid for Young Scientists (A) (Grant Number: 17H05052),
Because CLs cause toxicity, the addition of biodegradability Grant-in-Aid for Challenging Research (Exploratory) (Grant
into CLs would be also promising strategy in addition to the Number: 18K19889), Grant-in-Aid for Scientific Research
above strategy for reducing the amount of CLs used.27,28,185) (A) (Grant Number: 19H01170) from the Japan Society for
1156 Chem. Pharm. Bull. Vol. 69, No. 12 (2021)

the Promotion of Science (JSPS), the Special Education and W., Dorkin J. R., Qin J., Cantley W., Qin L. L., Racie T., Frank-
Research Expenses from the Ministry of Education, Culture, Kamenetsky M., Yip K. N., Alvarez R., Sah D. W., de Fougerolles
A., Fitzgerald K., Koteliansky V., Akinc A., Langer R., Anderson
Sports, Science and Technology (MEXT), The Uehara Memo-
D. G., Proc. Natl. Acad. Sci. U.S.A., 107, 1864–1869 (2010).
rial Foundation, The Mochida Memorial Foundation for Medi-
22) Dong Y., Love K. T., Dorkin J. R., et al., Proc. Natl. Acad. Sci.
cal and Pharmaceutical Research, TERUMO LIFE SCIENCE U.S.A., 111, 3955–3960 (2014).
FOUNDATION, and Creative Research Institute (CRIS, 23) Lokugamage M. P., Sago C. D., Gan Z., Krupczak B. R., Dahlman
Hokkaido University). I also wish to thank Dr. Milton Feather J. E., Adv. Mater., 31, e1902251 (2019).
for his helpful advice in preparing this manuscript. I would 24) Li B., Luo X., Deng B., Wang J., McComb D. W., Shi Y., Gaensler
like to thank Prof. Hideyoshi Harashima, Prof. Hidetaka K. M., Tan X., Dunn A. L., Kerlin B. A., Dong Y., Nano Lett., 15,
Akita, Dr. Hiroto Hatakeyama, Dr. Mamoru Hyodo, Dr. Yuma 8099–8107 (2015).
Yamada, and Dr. Takashi Nakamura for critical advice, tech- 25) Ramaswamy S., Tonnu N., Tachikawa K., Limphong P., Vega J. B.,
niques and other supports, and all the students who worked Karmali P. P., Chivukula P., Verma I. M., Proc. Natl. Acad. Sci.
diligently with me. U.S.A., 114, E1941–E1950 (2017).
26) Miller J. B., Zhang S., Kos P., Xiong H., Zhou K., Perelman S. S.,
Zhu H., Siegwart D. J., Angew. Chem. Int. Ed. Engl., 56, 1059–
Conflict of Interest The author declares no conflict of 1063 (2017).
interest. 27) Hassett K. J., Benenato K. E., Jacquinet E., Lee A., Woods A.,
Yuzhakov O., Himansu S., Deterling J., Geilich B. M., Ketova T.,
References Mihai C., Lynn A., McFadyen I., Moore M. J., Senn J. J., Stan-
1) Sahin U., Karikó K., Türeci Ö., Nat. Rev. Drug Discov., 13, 759– ton M. G., Almarsson Ö., Ciaramella G., Brito L. A., Mol. Ther.
780 (2014). Nucleic. Acids, 15, 1–11 (2019).
2) Rupaimoole R., Slack F. J., Nat. Rev. Drug Discov., 16, 203–222 28) Suzuki Y., Hyodo K., Suzuki T., Tanaka Y., Kikuchi H., Ishihara
(2017). H., Int. J. Pharm., 519, 34–43 (2017).
3) Tabebordbar M., Zhu K., Cheng J. K. W., Chew W. L., Widrick J. 29) Akita H., Biol. Pharm. Bull., 43, 1617–1625 (2020).
J., Yan W. X., Maesner C., Wu E. Y., Xiao R., Ran F. A., Cong L., 30) Miao L., Li L., Huang Y., Delcassian D., Chahal J., Han J., Shi Y.,
Zhang F., Vandenberghe L. H., Church G. M., Wagers A., J. Sci., Sadtler K., Gao W., Lin J., Doloff J. C., Langer R., Anderson D.
351, 407–411 (2016). G., Nat. Biotechnol., 37, 1174–1185 (2019).
4) Sánchez-Rivera F. J., Papagiannakopoulos T., Romero R., Tammela 31) Lee S. M., Cheng Q., Yu X., Liu S., Johnson L. T., Siegwart D. J.,
T., Bauer M. R., Bhutkar A., Joshi N. S., Subbaraj L., Bronson R. Angew. Chem. Int. Ed. Engl., 60, 5848–5853 (2021).
T., Xue W., Jacks T., Nature (London), 516, 428–431 (2014). 32) Sato Y., Hatakeyama H., Sakurai Y., Hyodo M., Akita H., Hara-
5) Wu S. Y., Lopez-Berestein G., Calin G. A., Sood A. K., Sci. shima H., J. Control. Release, 163, 267–276 (2012).
Transl. Med., 6, 240ps247 (2014). 33) Sato Y., Hatakeyama H., Hyodo M., Akita H., Harashima H.,
6) Hoy S. M., Drugs, 78, 1625–1631 (2018). Yakugaku Zasshi, 132, 1355–1363 (2012).
7) Buschmann M. D., Carrasco M. J., Alishetty S., Paige M., Alameh 34) Israelachvili J., Colloids Surf. A Physicochem. Eng. Asp., 91, 1–8
M. G., Weissman D., Vaccines (Basel), 9, 65 (2021). (1994).
8) Felgner P. L., Gadek T. R., Holm M., Roman R., Chan H. W., 35) Hafez I. M., Maurer N., Cullis P. R., Gene Ther., 8, 1188–1196
Wenz M., Northrop J. P., Ringold G. M., Danielsen M., Proc. Natl. (2001).
Acad. Sci. U.S.A., 84, 7413–7417 (1987). 36) Cullis P. R., de Kruijff B., Biochim. Biophys. Acta, 559, 399–420
9) Hope M. J., Mui B., Ansell S., Ahkong Q. F., Mol. Membr. Biol., (1979).
15, 1–14 (1998). 37) Wolfert M. A., Seymour L. W., Gene Ther., 5, 409–414 (1998).
10) Devine D. V., Wong K., Serrano K., Chonn A., Cullis P. R., Bio- 38) Erbacher P., Roche A. C., Monsigny M., Midoux P., Exp. Cell Res.,
chim. Biophys. Acta, 1191, 43–51 (1994). 225, 186–194 (1996).
11) Chonn A., Cullis P. R., Devine D. V., J. Immunol., 146, 4234–4241 39) Choi Y. H., Liu F., Kim J. S., Choi Y. K., Park J. S., Kim S. W.,
(1991). J. Control. Release, 54, 39–48 (1998).
12) Bailey A. L., Cullis P. R., Biochemistry, 33, 12573–12580 (1994). 40) Akinc A., Thomas M., Klibanov A. M., Langer R., J. Gene Med.,
13) Semple S. C., Klimuk S. K., Harasym T. O., Dos Santos N., Ansell 7, 657–663 (2005).
S. M., Wong K. F., Maurer N., Stark H., Cullis P. R., Hope M. J., 41) Wisse E., Jacobs F., Topal B., Frederik P., De Geest B., Gene
Scherrer P., Biochim. Biophys. Acta, 1510, 152–166 (2001). Ther., 15, 1193–1199 (2008).
14) Maurer N., Wong K. F., Stark H., Louie L., McIntosh D., Wong T., 42) Jacobs F., Wisse E., De Geest B., Am. J. Pathol., 176, 14–21
Scherrer P., Semple S. C., Cullis P. R., Biophys. J., 80, 2310–2326 (2010).
(2001). 43) Yan X., Kuipers F., Havekes L. M., Havinga R., Dontje B., Poel-
15) Sato Y., Nakamura T., Yamada Y., Harashima H., J. Control. Re- stra K., Scherphof G. L., Kamps J. A., Biochem. Biophys. Res.
lease, 330, 305–316 (2021). Commun., 328, 57–62 (2005).
16) Rietwyk S., Peer D., ACS Nano, 11, 7572–7586 (2017). 44) Akinc A., Querbes W., De S., et al., Mol. Ther., 18, 1357–1364
17) Heyes J., Palmer L., Bremner K., MacLachlan I., J. Control. Re- (2010).
lease, 107, 276–287 (2005). 45) Sato Y., Kinami Y., Hashiba K., Harashima H., J. Control. Re-
18) Zimmermann T. S., Lee A. C., Akinc A., et al., Nature (London), lease, 322, 217–226 (2020).
441, 111–114 (2006). 46) D’Souza A. A., Devarajan P. V., J. Control. Release, 203, 126–139
19) Semple S. C., Akinc A., Chen J., et al., Nat. Biotechnol., 28, (2015).
172–176 (2010). 47) Baenziger J. U., Maynard Y., J. Biol. Chem., 255, 4607–4613
20) Jayaraman M., Ansell S. M., Mui B. L., Tam Y. K., Chen J., Du (1980).
X., Butler D., Eltepu L., Matsuda S., Narayanannair J. K., Rajeev 48) Connolly D. T., Townsend R. R., Kawaguchi K., Bell W. R., Lee
K. G., Hafez I. M., Akinc A., Maier M. A., Tracy M. A., Cullis P. Y. C., J. Biol. Chem., 257, 939–945 (1982).
R., Madden T. D., Manoharan M., Hope M. J., Angew. Chem. Int. 49) Lee R. T., Lin P., Lee Y. C., Biochemistry, 23, 4255–4261 (1984).
Ed. Engl., 51, 8529–8533 (2012). 50) Wooddell C. I., Rozema D. B., Hossbach M., John M., Hamilton
21) Love K. T., Mahon K. P., Levins C. G., Whitehead K. A., Querbes
Vol. 69, No. 12 (2021)1157
Chem. Pharm. Bull.

H. L., Chu Q., Hegge J. O., Klein J. J., Wakefield D. H., Oropeza Propp S., Lollo B. A., Freier S., Bennett C. F., Bhanot S., Monia B.
C. E., Deckert J., Roehl I., Jahn-Hofmann K., Hadwiger P., Vorn- P., Cell Metab., 3, 87–98 (2006).
locher H. P., McLachlan A., Lewis D. L., Mol. Ther., 21, 973–985 79) Jopling C. L., Yi M., Lancaster A. M., Lemon S. M., Sarnow P.,
(2013). Science, 309, 1577–1581 (2005).
51) Wells M. J., Blajchman M. A., J. Biol. Chem., 273, 23440–23447 80) Lewis A. P., Jopling C. L., Biochem. Soc. Trans., 38, 1553–1557
(1998). (2010).
52) Narita M., Bu G., Olins G. M., Higuchi D. A., Herz J., Broze G. J. 81) Hatakeyama H., Murata M., Sato Y., Takahashi M., Minakawa N.,
Jr., Schwartz A. L., J. Biol. Chem., 270, 24800–24804 (1995). Matsuda A., Harashima H., J. Control. Release, 173, 43–50 (2014).
53) Libeu C. P., Lund-Katz S., Phillips M. C., Wehrli S., Hernáiz M. J., 82) Takahashi M., Yamada N., Hatakeyama H., Murata M., Sato Y.,
Capila I., Linhardt R. J., Raffaï R. L., Newhouse Y. M., Zhou F., Minakawa N., Harashima H., Matsuda A., Nucleic Acids Res., 41,
Weisgraber K. H., J. Biol. Chem., 276, 39138–39144 (2001). 10659–10667 (2013).
54) Watanabe T., Hatakeyama H., Matsuda-Yasui C., Sato Y., Sudoh 83) Sato Y., Hatakeyama H., Hyodo M., Harashima H., Mol. Ther., 24,
M., Takagi A., Hirata Y., Ohtsuki T., Arai M., Inoue K., Hara- 788–795 (2016).
shima H., Kohara M., Sci. Rep., 4, 4750 (2014). 84) Yamamoto N., Sato Y., Munakata T., Kakuni M., Tateno C.,
55) Hayashi Y., Suemitsu E., Kajimoto K., Sato Y., Akhter A., Sakurai Sanada T., Hirata Y., Murakami S., Tanaka Y., Chayama K.,
Y., Hatakeyama H., Hyodo M., Kaji N., Baba Y., Harashima H., Hatakeyama H., Hyodo M., Harashima H., Kohara M., J. Hepatol.,
Mol. Ther. Nucleic. Acids, 3, e154 (2014). 64, 547–555 (2016).
56) Yen C. L., Stone S. J., Cases S., Zhou P., Farese R. V. Jr., Proc. 85) Seto W. K., Lam Y. F., Fung J., Wong D. K., Huang F. Y., Hung
Natl. Acad. Sci. U.S.A., 99, 8512–8517 (2002). I. F., Lai C. L., Yuen M. F., J. Gastroenterol. Hepatol., 29, 1028–
57) Hall A. M., Kou K., Chen Z., Pietka T. A., Kumar M., Korenblat 1034 (2014).
K. M., Lee K., Ahn K., Fabbrini E., Klein S., Goodwin B., Finck 86) Jochum C., Voth R., Rossol S., Meyer zum Buschenfelde K.
B. N., J. Lipid Res., 53, 990–999 (2012). H., Hess G., Will H., Schroder H. C., Steffen R., Muller W. E.,
58) Lee Y. J., Ko E. H., Kim J. E., Kim E., Lee H., Choi H., Yu J. H., J. Virol., 64, 1956–1963 (1990).
Kim H. J., Seong J. K., Kim K. S., Kim J. W., Proc. Natl. Acad. 87) Nagaraju K., Naik S. R., Naik S., J. Viral Hepat., 4, 221–230
Sci. U.S.A., 109, 13656–13661 (2012). (1997).
59) Phan J., Reue K., Cell Metab., 1, 73–83 (2005). 88) Reignat S., Webster G. J., Brown D., Ogg G. S., King A., Senevi-
60) Kajimoto K., Suemitsu E., Sato Y., Sakurai Y., Harashima H., Biol. ratne S. L., Dusheiko G., Williams R., Maini M. K., Bertoletti A.,
Pharm. Bull., 39, 1653–1661 (2016). J. Exp. Med., 195, 1089–1101 (2002).
61) Klibanov A. L., Maruyama K., Torchilin V. P., Huang L., FEBS 89) Sato Y., Hashiba K., Sasaki K., Maeki M., Tokeshi M., Harashima
Lett., 268, 235–237 (1990). H., J. Control. Release, 295, 140–152 (2019).
62) Allen T. M., Hansen C., Martin F., Redemann C., Yau-Young A., 90) Zhang J., Fan H., Levorse D. A., Crocker L. S., Langmuir, 27,
Biochim. Biophys. Acta, 1066, 29–36 (1991). 1907–1914 (2011).
63) Cullis P. R., Chonn A., Semple S. C., Adv. Drug Deliv. Rev., 32, 91) Gilleron J., Querbes W., Zeigerer A., Borodovsky A., Marsico
3–17 (1998). G., Schubert U., Manygoats K., Seifert S., Andree C., Stöter M.,
64) Hatakeyama H., Akita H., Harashima H., Adv. Drug Deliv. Rev., Epstein-Barash H., Zhang L., Koteliansky V., Fitzgerald K., Fava
63, 152–160 (2011). E., Bickle M., Kalaidzidis Y., Akinc A., Maier M., Zerial M., Nat.
65) Hatakeyama H., Akita H., Harashima H., Biol. Pharm. Bull., 36, Biotechnol., 31, 638–646 (2013).
892–899 (2013). 92) Wittrup A., Ai A., Liu X., Hamar P., Trifonova R., Charisse K.,
66) Kale A. A., Torchilin V. P., Bioconjug. Chem., 18, 363–370 (2007). Manoharan M., Kirchhausen T., Lieberman J., Nat. Biotechnol.,
67) Walker G. F., Fella C., Pelisek J., Fahrmeir J., Boeckle S., Ogris 33, 870–876 (2015).
M., Wagner E., Mol. Ther., 11, 418–425 (2005). 93) Liang F., Lindgren G., Lin A., Thompson E. A., Ols S., Röhss J.,
68) Guo X., MacKay J. A., Szoka F. C. Jr., Biophys. J., 84, 1784–1795 John S., Hassett K., Yuzhakov O., Bahl K., Brito L. A., Salter H.,
(2003). Ciaramella G., Loré K., Mol. Ther., 25, 2635–2647 (2017).
69) Masson C., Garinot M., Mignet N., Wetzer B., Mailhe P., Scher- 94) Xu S., Yang K., Li R., Zhang L., Int. J. Mol. Sci., 21, 6582 (2020).
man D., Bessodes M., J. Control. Release, 99, 423–434 (2004). 95) Grabbe S., Haas H., Diken M., Kranz L. M., Langguth P., Sahin
70) Knorr V., Allmendinger L., Walker G. F., Paintner F. F., Wagner U., Nanomedicine (Lond.), 11, 2723–2734 (2016).
E., Bioconjug. Chem., 18, 1218–1225 (2007). 96) Kwon H., Kim M., Seo Y., Moon Y. S., Lee H. J., Lee K., Lee H.,
71) Rozema D. B., Ekena K., Lewis D. L., Loomis A. G., Wolff J. A., Biomaterials, 156, 172–193 (2018).
Bioconjug. Chem., 14, 51–57 (2003). 97) Topol E. J., Cell, 184, 1401 (2021).
72) Maeda Y., Pittella F., Nomoto T., Takemoto H., Nishiyama N., 98) Schoenmaker L., Witzigmann D., Kulkarni J. A., Verbeke R.,
Miyata K., Kataoka K., Macromol. Rapid Commun., 35, 1211–1215 Kersten G., Jiskoot W., Crommelin D. J. A., Int. J. Pharm., 601,
(2014). 120586 (2021).
73) Wong S. C., Klein J. J., Hamilton H. L., Chu Q., Frey C. L., Tru- 99) Kauffman K. J., Dorkin J. R., Yang J. H., Heartlein M. W., DeRosa
betskoy V. S., Hegge J., Wakefield D., Rozema D. B., Lewis D. L., F., Mir F. F., Fenton O. S., Anderson D. G., Nano Lett., 15, 7300–
Nucleic Acid Ther., 22, 380–390 (2012). 7306 (2015).
74) Rozema D. B., Lewis D. L., Wakefield D. H., Wong S. C., Klein J. 100) Oberli M. A., Reichmuth A. M., Dorkin J. R., Mitchell M. J., Fen-
J., Roesch P. L., Bertin S. L., Reppen T. W., Chu Q., Blokhin A. ton O. S., Jaklenec A., Anderson D. G., Langer R., Blankschtein
V., Hagstrom J. E., Wolff J. A., Proc. Natl. Acad. Sci. U.S.A., 104, D., Nano Lett., 17, 1326–1335 (2017).
12982–12987 (2007). 101) Jinek M., Chylinski K., Fonfara I., Hauer M., Doudna J. A., Char-
75) Hashiba K., Sato Y., Harashima H., J. Control. Release, 262, pentier E., Science, 337, 816–821 (2012).
239–246 (2017). 102) Cho S. W., Kim S., Kim J. M., Kim J. S., Nat. Biotechnol., 31,
76) Ambros V., Nature (London), 431, 350–355 (2004). 230–232 (2013).
77) Krützfeldt J., Rajewsky N., Braich R., Rajeev K. G., Tuschl T., 103) Mali P., Yang L., Esvelt K. M., Aach J., Guell M., DiCarlo J. E.,
Manoharan M., Stoffel M., Nature (London), 438, 685–689 (2005). Norville J. E., Church G. M., Science, 339, 823–826 (2013).
78) Esau C., Davis S., Murray S. F., Yu X. X., Pandey S. K., Pear M., 104) Nishimasu H., Shi X., Ishiguro S., et al., Science, 361, 1259–1262
Watts L., Booten S. L., Graham M., McKay R., Subramaniam A., (2018).
1158 Chem. Pharm. Bull. Vol. 69, No. 12 (2021)

105) Kleinstiver B. P., Pattanayak V., Prew M. S., Tsai S. Q., Nguyen N. 128) Jinushi M., Komohara Y., Biochim. Biophys. Acta, 1855, 123–130
T., Zheng Z., Joung J. K., Nature (London), 529, 490–495 (2016). (2015).
106) Gilbert L. A., Horlbeck M. A., Adamson B., Villalta J. E., Chen 129) Shobaki N., Sato Y., Suzuki Y., Okabe N., Harashima H., J. Con-
Y., Whitehead E. H., Guimaraes C., Panning B., Ploegh H. L., trol. Release, 325, 235–248 (2020).
Bassik M. C., Qi L. S., Kampmann M., Weissman J. S., Cell, 159, 130) Wilson H. M., Front. Immunol., 5, 357 (2014).
647–661 (2014). 131) Goswami K. K., Ghosh T., Ghosh S., Sarkar M., Bose A., Baral R.,
107) Anzalone A. V., Randolph P. B., Davis J. R., Sousa A. A., Koblan Cell. Immunol., 316, 1–10 (2017).
L. W., Levy J. M., Chen P. J., Wilson C., Newby G. A., Raguram 132) Wang Y., Shen Y., Wang S., Shen Q., Zhou X., Cancer Lett., 415,
A., Liu D. R., Nature (London), 576, 149–157 (2019). 117–128 (2018).
108) Nishida K., Arazoe T., Yachie N., Banno S., Kakimoto M., Tabata 133) Jantsch J., Turza N., Volke M., Eckardt K. U., Hensel M.,
M., Mochizuki M., Miyabe A., Araki M., Hara K. Y., Shimatani Steinkasserer A., Willam C., Prechtel A. T., J. Immunol. Methods,
Z., Kondo A., Science, 353, aaf8729 (2016). 337, 71–77 (2008).
109) Komor A. C., Kim Y. B., Packer M. S., Zuris J. A., Liu D. R., Na- 134) Hobo W., Novobrantseva T. I., Fredrix H., Wong J., Milstein S.,
ture (London), 533, 420–424 (2016). Epstein-Barash H., Liu J., Schaap N., van der Voort R., Dolstra H.,
110) Nuñez J. K., Chen J., Pommier G. C., Cogan J. Z., Replogle J. M., Cancer Immunol. Immunother., 62, 285–297 (2013).
Adriaens C., Ramadoss G. N., Shi Q., Hung K. L., Samelson A. J., 135) Zhang Q., Hossain D. M., Nechaev S., Kozlowska A., Zhang W.,
Pogson A. N., Kim J. Y. S., Chung A., Leonetti M. D., Chang H. Liu Y., Kowolik C. M., Swiderski P., Rossi J. J., Forman S., Pal
Y., Kampmann M., Bernstein B. E., Hovestadt V., Gilbert L. A., S., Bhatia R., Raubitschek A., Yu H., Kortylewski M., Blood, 121,
Weissman J. S., Cell, 184, 2503–2519.e2517 (2021). 1304–1315 (2013).
111) Morisaka H., Yoshimi K., Okuzaki Y., Gee P., Kunihiro Y., Son- 136) Warashina S., Nakamura T., Sato Y., Fujiwara Y., Hyodo M.,
pho E., Xu H., Sasakawa N., Naito Y., Nakada S., Yamamoto T., Hatakeyama H., Harashima H., J. Control. Release, 225, 183–191
Sano S., Hotta A., Takeda J., Mashimo T., Nat. Commun., 10, 5302 (2016).
(2019). 137) Naka T., Narazaki M., Hirata M., Matsumoto T., Minamoto S.,
112) Liu J., Gaj T., Yang Y., Wang N., Shui S., Kim S., Kanchiswamy Aono A., Nishimoto N., Kajita T., Taga T., Yoshizaki K., Akira S.,
C. N., Kim J. S., Barbas C. F. 3rd, Nat. Protoc., 10, 1842–1859 Kishimoto T., Nature (London), 387, 924–929 (1997).
(2015). 138) Endo R., Nakamura T., Kawakami K., Sato Y., Harashima H., Sci.
113) Liang X., Potter J., Kumar S., Zou Y., Quintanilla R., Sridharan Rep., 9, 11335 (2019).
M., Carte J., Chen W., Roark N., Ranganathan S., Ravinder N., 139) Nakamura T., Kuroi M., Fujiwara Y., Warashina S., Sato Y., Ha-
Chesnut J. D., J. Biotechnol., 208, 44–53 (2015). rashima H., Sci. Rep., 6, 37849 (2016).
114) Kimura N., Maeki M., Sato Y., Note Y., Ishida A., Tani H., Hara- 140) Woo S. R., Fuertes M. B., Corrales L., Spranger S., Furdyna M. J.,
shima H., Tokeshi M., Acs Omega, 3, 5044–5051 (2018). Leung M. Y., Duggan R., Wang Y., Barber G. N., Fitzgerald K. A.,
115) Maeki M., Kimura N., Sato Y., Harashima H., Tokeshi M., Adv. Alegre M. L., Gajewski T. F., Immunity, 41, 830–842 (2014).
Drug Deliv. Rev., 128, 84–100 (2018). 141) Miyabe H., Hyodo M., Nakamura T., Sato Y., Hayakawa Y., Hara-
116) Suzuki Y., Onuma H., Sato R., Sato Y., Hashiba A., Maeki M., shima H., J. Control. Release, 184, 20–27 (2014).
Tokeshi M., Kayesh M. E. H., Kohara M., Tsukiyama-Kohara K., 142) Chang C. C., Campoli M., Ferrone S., Adv. Cancer Res., 93, 189–
Harashima H., J. Control. Release, 330, 61–71 (2021). 234 (2005).
117) Zetsche B., Gootenberg J. S., Abudayyeh O. O., Slaymaker I. M., 143) Vivier E., Raulet D. H., Moretta A., Caligiuri M. A., Zitvogel L.,
Makarova K. S., Essletzbichler P., Volz S. E., Joung J., van der Lanier L. L., Yokoyama W. M., Ugolini S., Science, 331, 44–49
Oost J., Regev A., Koonin E. V., Zhang F., Cell, 163, 759–771 (2011).
(2015). 144) Nakamura T., Miyabe H., Hyodo M., Sato Y., Hayakawa Y., Hara-
118) Park H. M., Liu H., Wu J., Chong A., Mackley V., Fellmann C., shima H., J. Control. Release, 216, 149–157 (2015).
Rao A., Jiang F., Chu H., Murthy N., Lee K., Nat. Commun., 9, 145) Nakamura T., Sato T., Endo R., Sasaki S., Takahashi N., Sato Y.,
3313 (2018). Hyodo M., Hayakawa Y., Harashima H., J. Immunother. Cancer, 9,
119) van Furth R., Cohn Z. A., J. Exp. Med., 128, 415–435 (1968). e002852 (2021).
120) Borges da Silva H., Fonseca R., Pereira R. M., Cassado A. A., 146) Messina S., Sframeli M., J. Clin. Med., 9, 2222 (2020).
Álvarez J. M., D’Império Lima M. R., Front. Immunol., 6, 480 147) Zlokovic B. V., Nat. Rev. Neurosci., 12, 723–738 (2011).
(2015). 148) Tamaru M., Akita H., Kajimoto K., Sato Y., Hatakeyama H., Hara-
121) Zheng X., Turkowski K., Mora J., Brüne B., Seeger W., Weigert shima H., Int. J. Pharm., 465, 77–82 (2014).
A., Savai R., Oncotarget, 8, 48436–48452 (2017). 149) Tamaru M., Akita H., Nakatani T., Kajimoto K., Sato Y.,
122) Ghosn E. E., Cassado A. A., Govoni G. R., Fukuhara T., Yang Y., Hatakeyama H., Harashima H., Int. J. Nanomedicine, 9, 4267–4276
Monack D. M., Bortoluci K. R., Almeida S. R., Herzenberg L. A., (2014).
Herzenberg L. A., Proc. Natl. Acad. Sci. U.S.A., 107, 2568–2573 150) Huertas A., Guignabert C., Barberà J. A., Bärtsch P., Bhattacharya
(2010). J., Bhattacharya S., Bonsignore M. R., Dewachter L., Dinh-Xuan
123) Triantafyllou E., Woollard K. J., McPhail M. J. W., Antoniades C. A. T., Dorfmüller P., Gladwin M. T., Humbert M., Kotsimbos T.,
G., Possamai L. A., Front. Immunol., 9, 2948 (2018). Vassilakopoulos T., Sanchez O., Savale L., Testa U., Wilkins M.
124) Matsui H., Sato Y., Hatakeyama H., Akita H., Harashima H., Int. R., Eur. Respir. J., 51, 1700745 (2018).
J. Pharm., 495, 171–178 (2015). 151) Kusumoto K., Akita H., Ishitsuka T., Matsumoto Y., Nomoto T.,
125) Komohara Y., Fujiwara Y., Ohnishi K., Takeya M., Adv. Drug Furukawa R., El-Sayed A., Hatakeyama H., Kajimoto K., Yamada
Deliv. Rev., 99 (Pt B), 180–185 (2016). Y., Kataoka K., Harashima H., ACS Nano, 7, 7534–7541 (2013).
126) Sica A., Larghi P., Mancino A., Rubino L., Porta C., Totaro M. 152) Santiwarangkool S., Akita H., Nakatani T., Kusumoto K., Kimura
G., Rimoldi M., Biswas S. K., Allavena P., Mantovani A., Semin. H., Suzuki M., Nishimura M., Sato Y., Harashima H., J. Pharm.
Cancer Biol., 18, 349–355 (2008). Sci., 106, 2420–2427 (2017).
127) DeNardo D. G., Brennan D. J., Rexhepaj E., Ruffell B., Shiao S. 153) Abd Elwakil M., Khalil I., Elewa Y., Kusumoto K., Sato Y., Sho-
L., Madden S. F., Gallagher W. M., Wadhwani N., Keil S. D., baki N., Kon Y., Harashima H., Adv. Funct. Mater., 29, 1807677
Junaid S. A., Rugo H. S., Hwang E. S., Jirström K., West B. L., (2019).
Coussens L. M., Cancer Discov., 1, 54–67 (2011). 154) Choi H. S., Ashitate Y., Lee J. H., Kim S. H., Matsui A., Insin N.,
Vol. 69, No. 12 (2021)1159
Chem. Pharm. Bull.

Bawendi M. G., Semmler-Behnke M., Frangioni J. V., Tsuda A., 175) Aramaki Y., Takano S., Tsuchiya S., Arch. Biochem. Biophys., 392,
Nat. Biotechnol., 28, 1300–1303 (2010). 245–250 (2001).
155) Allen T. M., Hansen C. B., Guo L. S., Biochim. Biophys. Acta, 176) Kusumoto K., Ishikawa T., J. Control. Release, 147, 246–252
1150, 9–16 (1993). (2010).
156) Huo S., Ma H., Huang K., Liu J., Wei T., Jin S., Zhang J., He S., 177) Abrams M. T., Koser M. L., Seitzer J., Williams S. C., DiPietro M.
Liang X. J., Cancer Res., 73, 319–330 (2013). A., Wang W., Shaw A. W., Mao X., Jadhav V., Davide J. P., Burke
157) Cabral H., Matsumoto Y., Mizuno K., Chen Q., Murakami M., P. A., Sachs A. B., Stirdivant S. M., Sepp-Lorenzino L., Mol.
Kimura M., Terada Y., Kano M. R., Miyazono K., Uesaka M., Ther., 18, 171–180 (2010).
Nishiyama N., Kataoka K., Nat. Nanotechnol., 6, 815–823 (2011). 178) Tao W., Mao X., Davide J. P., Ng B., Cai M., Burke P. A., Sachs A.
158) Jain R. K., Stylianopoulos T., Nat. Rev. Clin. Oncol., 7, 653–664 B., Sepp-Lorenzino L., Mol. Ther., 19, 567–575 (2011).
(2010). 179) Barros S. A., Gollob J. A., Adv. Drug Deliv. Rev., 64, 1730–1737
159) Nakamura T., Kawai M., Sato Y., Maeki M., Tokeshi M., Hara- (2012).
shima H., Mol. Pharm., 17, 944–953 (2020). 180) Sato Y., Matsui H., Yamamoto N., Sato R., Munakata T., Kohara
160) Chen S., Tam Y. Y., Lin P. J., Leung A. K., Tam Y. K., Cullis P. R., M., Harashima H., J. Control. Release, 266, 216–225 (2017).
J. Control. Release, 196, 106–112 (2014). 181) Belliveau N. M., Huft J., Lin P. J., Chen S., Leung A. K., Leaver
161) Zhigaltsev I. V., Belliveau N., Hafez I., Leung A. K., Huft J., Han- T. J., Wild A. W., Lee J. B., Taylor R. J., Tam Y. K., Hansen C. L.,
sen C., Cullis P. R., Langmuir, 28, 3633–3640 (2012). Cullis P. R., Mol. Ther. Nucleic. Acids, 1, e37 (2012).
162) Jahn A., Vreeland W. N., DeVoe D. L., Locascio L. E., Gaitan M., 182) Leung A. K., Hafez I. M., Baoukina S., Belliveau N. M., Zhigalt-
Langmuir, 23, 6289–6293 (2007). sev I. V., Afshinmanesh E., Tieleman D. P., Hansen C. L., Hope
163) Chen S., Tam Y. Y. C., Lin P. J. C., Sung M. M. H., Tam Y. K., M. J., Cullis P. R., J. Phys. Chem. C Nanomater. Interfaces, 116,
Cullis P. R., J. Control. Release, 235, 236–244 (2016). 18440–18450 (2012).
164) Sato Y., Note Y., Maeki M., Kaji N., Baba Y., Tokeshi M., Hara- 183) Sato Y., Matsui H., Sato R., Harashima H., J. Control. Release,
shima H., J. Control. Release, 229, 48–57 (2016). 284, 179–187 (2018).
165) Zhang Y., Arrington L., Boardman D., Davis J., Xu Y., DiFelice 184) Nakamura T., Yamada K., Fujiwara Y., Sato Y., Harashima H.,
K., Stirdivant S., Wang W., Budzik B., Bawiec J., Deng J., Beutner Mol. Pharm., 15, 2142–2150 (2018).
G., Seifried D., Stanton M., Gindy M., Leone A., J. Control. Re- 185) Maier M. A., Jayaraman M., Matsuda S., et al., Mol. Ther., 21,
lease, 174, 7–14 (2014). 1570–1578 (2013).
166) Hung W. C., Lee M. T., Chen F. Y., Huang H. W., Biophys. J., 92, 186) Kimura N., Maeki M., Sato Y., Ishida A., Tani H., Harashima H.,
3960–3967 (2007). Tokeshi M., ACS Appl. Mater. Interfaces, 12, 34011–34020 (2020).
167) Alwarawrah M., Dai J., Huang J., J. Phys. Chem. B, 114, 7516– 187) Viger-Gravel J., Schantz A., Pinon A. C., Rossini A. J., Schantz S.,
7523 (2010). Emsley L., J. Phys. Chem. B, 122, 2073–2081 (2018).
168) Shobaki N., Sato Y., Harashima H., Int. J. Nanomedicine, 13, 188) Yanez Arteta M., Kjellman T., Bartesaghi S., Wallin S., Wu X.,
8395–8410 (2018). Kvist A. J., Dabkowska A., Székely N., Radulescu A., Bergenholtz
169) Sato Y., Okabe N., Note Y., Hashiba K., Maeki M., Tokeshi M., J., Lindfors L., Proc. Natl. Acad. Sci. U.S.A., 115, E3351–E3360
Harashima H., Acta Biomater., 102, 341–350 (2020). (2018).
170) Longo G. S., Schick M., Szleifer I., Biophys. J., 96, 3977–3986 189) Sebastiani F., Yanez Arteta M., Lerche M., Porcar L., Lang C.,
(2009). Bragg R. A., Elmore C. S., Krishnamurthy V. R., Russell R. A.,
171) Hashiba A., Toyooka M., Sato Y., Maeki M., Tokeshi M., Hara- Darwish T., Pichler H., Waldie S., Moulin M., Haertlein M., For-
shima H., J. Control. Release, 327, 467–476 (2020). syth V. T., Lindfors L., Cárdenas M., ACS Nano, 15, 6709–6722
172) Yan W., Chen W., Huang L., Mol. Immunol., 44, 3672–3681 (2007). (2021).
173) Kongkaneramit L., Sarisuta N., Azad N., Lu Y., Iyer A. K., Wang 190) Egorov E., Pieters C., Korach-Rechtman H., Shklover J., Schroeder
L., Rojanasakul Y., J. Pharmacol. Exp. Ther., 325, 969–977 A., Drug Deliv. Transl. Res., 11, 345–352 (2021).
(2008). 191) Bannigan P., Aldeghi M., Bao Z., Häse F., Aspuru-Guzik A., Allen
174) Iwaoka S., Nakamura T., Takano S., Tsuchiya S., Aramaki Y., C., Adv. Drug Deliv. Rev., 175, 113806 (2021).
J. Leukoc. Biol., 79, 184–191 (2006). 192) Bunjes H., Unruh T., Adv. Drug Deliv. Rev., 59, 379–402 (2007).

You might also like