ArticlePDF AvailableLiterature Review

Lipid-Based Nanoparticles in The Systemic Delivery of siRNA

Taylor & Francis
Nanomedicine
Authors:

Abstract and Figures

RNAi therapeutics are believed to be the future of personalized medicine and have shown promise in early clinical trials. However, many physiological barriers exist in the systemic delivery of siRNAs to the cytoplasm of targeted cells to perform their function. To overcome these barriers, many siRNA delivery systems have been developed. Among these, lipid-based nanoparticles have great potential owing to their biocompatibility and low toxicity in comparison with inorganic nanoparticles and viral systems. This review discusses the hurdles of systemic siRNA delivery and highlights the recent progress made in lipid-based nanoparticles, which are categorized based on their key lipid components, including cationic lipid, lipoprotein, lipidoid, neutral lipid and anionic lipid-based nanoparticles. It is expected that these lipid nanoparticle-based siRNA delivery systems will have an enabling role for personalized cancer medicine, where siRNA delivery will join forces with genetic profiling of individual patients to achieve the best treatment outcome.
Content may be subject to copyright.
105
Review
IS SN 174 3- 58 8910.2217/NNM.13.192 © 2014 Future Med icin e Ltd Nanomedici ne (2 014) 9(1), 10 5 –120
Since the discovery of the RNAi in 1998 by Fire
et al. [1], the first validation of specific gene knock-
down in mammalian cells [2 ] and the first clinical
trial of siRNA for age-related macular degenera-
tion in 2004 [3], RNAi therapeutics gained the
world’s attention and became an attractive and
promising technique in personalized treatment
of a broad range of diseases, including cancer,
liver and immune-related diseases.
RNAi is found in the cytoplasm [4]. As illus-
trated in Figur e 1, siRNA, a double strand of RNA,
incorporates into the RNA-induced silencing
complex (RISC), causing unwinding of its dou-
ble strand. The sense strand of siRNA is then
removed from RISC, and the activated RISC
with the antisense strand serves as a template for
the binding of complementary mRNA, inducing
mRNA degradation. As siRNA only functions
when it reaches the cytoplasm of cells that pro-
duce the targeted gene, systemic siRNA delivery
encounters many barriers from its administration
all the way to reaching the target gene to be fully
functional.
Barriers in the systemic delivery
of siRNA
Stability in the blood stream
After intravenous injection, the first aim is to
keep the siRNA stable in the bloodstream. Naked
siRNA is easily degraded by many endogenous
enzymes and aggregated by serum proteins in the
blood, which requires the siRNA delivery system
to be ‘cohesive’ enough to minimize degradation
or disintegration. The delivery system must also
be effective at minimizing nonspecific opsoniza-
tion, phagocytosis and immune activation, while
also presenting surface properties that promote
interaction with the desired cellular targets [5 ].
In addition, prolonging the blood circulation of
siRNA is also important for its effective delivery
as naked siRNA is eliminated from blood just
5 min after intravenous injection [4–8].
Transport across the vascular
endothelium
The endothelium acts as a semiselective barrier
between the vessel lumen and surrounding tis-
sue, controlling the passage of materials into
tissues. As listed in Table 1, the normal capillary
endothelium can be divided into three types:
continuous endothelium, fenestrated capillaries
and discontinuous capillaries [9]. The size limita-
tion of normal endothelium structure suggests
that siRNA delivery systems should have small
sizes (150 nm) to readily cross the vascular
endothelial barrier. It is also known that the
regional structure of blood vessels changes in
the inflammation sites and solid tumors, where
the enhanced permeation and retention effect on
drug delivery has been widely observed. Small
nanoparticles (NPs) with diameter of less than
500 nm, usually less than 150 nm, showed signif-
icant enhanced permeation and retention effects
in ‘leaky’ tumor blood vessels [1 0] .
RNAi therapeutics are believed to be the future of personalized medicine and have shown promise in
early clinical trials. However, many physiological barriers exist in the systemic delivery of siRNAs to the
cytoplasm of targeted cells to perform their function. To overcome these barriers, many siRNA delivery
systems have been developed. Among these, lipid-based nanoparticles have great potential owing to
their biocompatibility and low toxicity in comparison with inorganic nanoparticles and viral systems.
This review discusses the hurdles of systemic siRNA delivery and highlights the recent progress made in
lipid-based nanoparticles, which are categorized based on their key lipid components, including cationic
lipid, lipoprotein, lipidoid, neutral lipid and anionic lipid-based nanoparticles. It is expected that these
lipid nanoparticle-based siRNA delivery systems will have an enabling role for personalized cancer
medicine, where siRNA delivery will join forces with genetic profiling of individual patients to achieve
the best treatment outcome.
KEYWORDS: lipid lipoprotein liposome nanoparticle RNAi siRNA delivery
systemic delivery barrier
Lipid-based nanoparticles in the systemic delivery
of siRNA
Qiaoya Lin1,2,3,
Juan Chen1,
Zhihong Zhang3
& Gang Zheng*1,2
1Ontario Can cer Instute & Techn a
Instute, Uni versity H ealth Netwo rk,
Toronto, ON, Cana da
2Department of Medic al Biophysi cs,
Universit y of Toronto, Toronto Medic al
Discover y Tower 5-363, 101 Col lege
Street, Toronto, ON, M 5G 1L7, Canada
3Brion Cha nce Center for B iomedical
Photonics, Wuhan Naona l Laborator y
for Optoelectronics, Huazhong
Universit y of Science & Technology,
Wuhan, China
*Author for correspondence:
gang.zheng@uhnres.utoronto.ca
part of
For reprint orders, please contact: reprints@futuremedicine.com
Nanomedicine (20 14) 9(1)
106 future science group
Review Lin, Chen, Zhang & Zheng
Diffusion through the extracellular
matrix
After crossing the vascular endothelium, the
siRNA delivery vehicle has to diffuse through
the extracellular matrix (ECM), which is com-
posed of gels of polysaccharides and fibrous
proteins, and acts as the structural support for
the animal cells. The tight structure of the ECM
hinders the diffusion of larger NPs, especially
in poorly permeable tumor tissue. For example,
Cabral et al. demonstrated that 30-nm micelles
could effectively penetrate deep tumor tissue of
Endosome
Nucleus
RNAi
Ta rget mRNA cleavage
RISC
siRNA
Cytoplasm
i
ii a
b
Nanomedicine © Future Medicine Ltd (2014)
Figure 1. In vivo siRNA systemic delivery barriers and the mechanism of RNAi. (A) Stability in
the blood stream; (B) transport across the vascular endothelial barrier; (C) diffusion through the
extracellular matrix; (D) delivery into the cy toplasm by (Di) endosomal escape and (Dii) direct
cytosolic delivery. (Dia) The siRNAs or siRNA nanoparticles were trapped in the endosome and
(Dib) the siRNAs were released from the endosome into the cytoplasm.
www.futuremedicine.com 107
future science group
Lipid-based nanoparticles in the systemic delivery of siRNA Review
pancreatic adenocarcinoma while the 70-nm
micelles were retained around the vasculature,
indicating that only NPs smaller than 50 nm
can penetrate poorly permeable tumors [1 1].
In human glioblastoma (U87) and melanoma
(Mu89), it was demonstrated that an increase
of the particles’ molecular size decreased their
interstitial diffusion [1 2] . In addition, the trans-
mission electron microscopy results showed the
interfibrillar spacing between bundles of aligned
and compact fibrils in U87 mouse tumors was
20–42 nm [1 2] . Altogether, the small size of
nanocarriers is superior for effective delivery of
siRNA to poorly permeable tissues. In addition,
the charge of the NPs could also influence parti-
cle diffusion. Stylianopoulos et al. demonstrated
that neutral particles diffuse faster than charged
particles [13] .
Delivery into the cytoplasm
After diffusion through the ECM, the siRNA
delivery system needs to transport through the
cell membrane, reach the cytoplasm and release
the siRNA payload. The naked siRNA with a
negative charge cannot readily cross the nega-
tively charged cell membrane. The siRNAs fer-
ried by nanocarriers usually enter into cells via
endocytosis pathways, such as macropinocytosis,
clathrin-mediated endocytosis (CME) and cave-
olae-/lipid raft-mediated endocytosis [14 , 15] . Mac-
ropinocytosis and CME usually drive the siRNA
carrier into the endosome (Fig ur e 1Dia), where
mature endosomes fuse easily with lysosomal
vesicles, resulting in enzymatic destruction of
siRNA. Therefore, escaping from the endosome
is further required for effective siRNA delivery
(Fig ur e 1 Dib ). The caveolae-/lipid raft-mediated
endocytosis is a lesser characterized pathway
compared with macropincytosis and CME [1 6 ,17].
However, it is has been demonstrated that some
lipid raft-related delivery could bypass the endo-
somal route to achieve direct cytosolic delivery
of drugs ( Figur e 1Dii) [18–20 ], opening a new avenue
for enhanced cytosolic siRNA delivery.
To overcome the barriers in the systemic
delivery of siRNA, variable nonviral siRNA
delivery vehicles have been developed, includ-
ing lipid-based NPs [21 –24 ], polymers [25] or lipid
polymer hybrid NPs [26 ], hydrogels [27], micro-
bubbles [2 8], inorganic NPs, such as silica [29],
gold [30], quantum dot [31], iron oxide [32 ,33]
and carbon nanotubes [3 4 ,35] , and other materi-
als, such as oligonucleotide NPs [3 6] and exo-
somes [3 7]. Among them, the lipid-based NPs
are in the most advanced stage of development
and have shown favorable biocompatible and
biodegradable properties in comparison with
inorganic carriers and viral vectors. This review
will focus on the development of lipid-based NPs
for the systemic delivery of siRNAs.
The lipid-based NPs reviewed here are divided
into the following types, based on their key
component lipids: cationic lipid-based NPs;
lipoprotein-related NPs; and other lipid NPs,
including lipid-like material-, neutral lipid- and
anionic lipid-based NPs.
Cationic lipid-based siRNA delivery
Cationic liposomes are the most common lipid-
based nanocarrier used for siRNA delivery.
siRNA loading onto the cationic liposome is
mainly attributed to the electrostatic interac-
tion between the anionic siRNA and cationic
lipids. The cationic lipids facilitate the par-
ticles’ intracellular uptake and further endo-
somal escape. Some cationic liposomes have
been widely used for siRNA delivery, such as
1,2-dioleoyl-3-trimethylammonium-propane
(DOTAP)–siR NA, N-(1-[2,3-dioleyloxy]-
propyl)-N,N,N-trimethylammonium chloride
siRNA and Lipofectamine® 2000 (Life Tech-
nologies, MD, USA)–siRNA complexes [3 8 – 41] ,
which are simply formulated by cationic lipids
and siRNAs. However, these complexes gener-
ally have a high electric charge density on their
surface that readily induce nonspecific interac-
tions with serum proteins and immunogenic
response, resulting in their rapid removal from
the blood stream [4,42,43].
Development of advanced cationic
lipid-based siRNA delivery system
Stable loading of siRNA
To stabilize the cationic lipid-based siRNA
delivery system, many neutral lipids, such as
cholesterol (Chol) or 1,2-dioleoyl-sn-glycerol-
3-phosphoethanolamine (DOPE) [4 4], have been
added to the complex formulation. These neutral
lipids not only stabilize the formation but also
enhance the cellular uptake of particles, which
will be discussed later (see the ‘Improving the
cellular uptake & enhancing siRNA release into
the cytoplasm’ section).
Table 1. Normal capillary endothelium.
Typ e Organs Size (nm)
Continuous Brain, skeletal, cardiac and smooth muscles, lung,
skin, subcutaneous and mucous membranes
1.8 2.0
Fenestrated Kidney, small intestine and salivary glands 11
Discontinuous Liver, spleen and bone marrow Up to 150
Nanomedicine (20 14) 9(1)
108 future science group
Review Lin, Chen, Zhang & Zheng
In comparison with surface loading, siRNA
loading in the NP core should provide better
delivery stability. To improve the siRNA core
loading efficiency, many helper cationic poly-
mers, such as protamine, were introduced to pre-
condense siRNA into the core of a liposome. A
good example is the liposome–polycation–DNA
(LPD) NPs developed by Li et al. [45]. In this
NP, protamine interacts with the nucleic acid
to form a negatively charged compact core,
cationic liposomes composed of DOTAP/Chol
collapse onto the core via charge–charge inter-
action and 1,2-distearoyl-sn-glycero-3-phos-
phoethanolamine–PEG is then coated onto the
outer surface of the particles. Thus, the siRNA
delivery is supported and shielded by both the
cationic lipid bilayer and PEG coating [45].
To improve siRNA loading efficacy, many
efforts have been undertaken to optimize the
key component of liposome cationic lipids. San-
tel et al. synthesized a series of b-l-argi nyl-2,
3-l-diaminopropionic acid-N-palmityl-N-ole-
ylamide trihydrochloride (AtuFECT) cationic
lipids and validated that AtuFECT01 enables
enhancement of the siR NA-binding ability
when compared with commercial cationic lip-
ids, such as DOTAP or N-(1-[2,3-dioleyloxy]-
propyl)-N,N,N-trimethylammonium chloride
[7]. The ability of the AtuFECT-formulated
liposome to deliver siRNA (the resultant Atu-
FECT–liposome–siRNA complex is termed
as AtuPLEX) that inhibits CD31 and Tie2
in the vasculature of mice has also been dem-
onstrated [7] . Later, the same group extended
the application of the optimized AtuPLEX to
treat advanced solid tumor by targeting PKN3
(termed Atu027) and achieved significant
inhibition of tumor growth [46 ], lymph node
metastasis [46] and lung metastasis formation
[47]. Notably, a Phase I clinical trial of Atu027
for the treatment of advanced solid cancers has
been completed, and a further clinical test using
a combination of Atu027 and gemcitabine is
currently ongoing [201,202].
The stable loading of siRNA depends not
only on the formulation components but also on
siRNA-integrating methods. Buyens et al. pub-
lished a comprehensive review on this aspect,
which covers variable methods for siRNA load-
ing, such as simple mixing, direct hydration
of a lipid film by a siRNA-contained solution
and the ethanol dilution method [48]. Among
them, the simple mixing method usually gave
the poorest siRNA encapsulation efficacy [4 8].
Furthermore, the stable systemic deliv-
ery of siRNA can be improved by chemical
modifications of the siRNAs backbone, such
as 2´-O-methyl and 2´-fluoro RNA. Chol con-
jugation to the siRNA sequence also enhanced
siRNA loading efficacy on lipid-based NPs, and
exhibited higher biologic activity compared with
unmodified siRNA [49].
Improving siRNA delivery
pharmacokinetics
To improve the blood stability and the in vivo
pharmacokinetics properties, many other helper
lipids were introduced into the siRNA delivery
system, such as PEGylated lipid (PEG lipid) [48].
The PEGylation effect of prolonging the circula-
tion time of many carriers in the blood has been
reviewed by many groups [48,50,51], including,
but not limited to, the influence of acyl chain
length in PEG lipids and their molar percentage
in the liposome composition [52 ]. Sonoke et al.
found that the PEG lipids with long acyl chains
showed better siRNA delivery pharmacokinet-
ics and efficiency when compared with those
with the short or unsaturated chains [5 3]. The
percentage of PEG lipids in the siRNA delivery
system should consider both blood circulation
time and cell uptake efficiency. A higher percent-
age of PEG lipids usually gives a longer blood
circulation time but weakens the cellular uptake
and subsequent endosomal escape. Therefore, an
optimal density of PEG lipids is required [ 48 , 51] .
Yagi et al. developed a ‘wrapsome’ NP, com-
prised of a core of siRNA/cationic DOTAP that
was fully enveloped by a neutral lipid bilayer
containing egg phosphatidylcholine and PEG
lipid in a weight ratio of 24:14.8. They found
that the wrapsome could improve the stability
and systemic circulation of siRNA, thus result-
ing in enhanced specific gene knockdown and
significant anti-tumor activity in vivo [54 ].
To overcome the PEGylation-induced prob-
lem of cell uptake and endosomal escaping, Car-
mona et al. developed a pH-sensitive PEGylated
liposome by postcoupling a PEG-2000 dialde-
hyde on the surface of particles composed of
cationic cholesteryl polyamine–N1-cholester-
yloxycarbonyl-3, 7-diazanonane-1, 9-diamine,
neutral lipids (DOPE) and Chol–PEG350 ami-
noxy lipid via oxime linkage. The oxime link-
age of the NPs is stable at pH 7, but is prone to
decomposition in an acidic environment (pH 5.5
and below), which induces the release of PEG
coating from particles, thus granting acidic pH-
triggered cell uptake and endosome release of
nucleic acids. This formulation demonstrated
effective RNAi delivery for on the control of
hepatitis B virus (HBV) virus replication [55] .
www.futuremedicine.com 109
future science group
Lipid-based nanoparticles in the systemic delivery of siRNA Review
Hatakeyama et al. developed another PEG-
cleavable siRNA delivery system, termed a
‘multifunctional envelope-type nanodevice’
(MEND), composed of DOTAP, DOPE, Chol,
1,2-distearoyl-sn-glycero-3-phosphoethanol-
amine–PEG and a PEG–peptide–DOPE (PPD).
PPD has a peptide sequence-linked PEG and
DOPE lipid moiety together, so that the PEG is
removed when the peptide linker is cleaved by
the target molecule, such as MMP. It has been
demonstrated that PPD-formulated NPs acceler-
ated both cellular uptake and endosomal escape
in MMP-rich tumor environments, compared
with a conventional PEG fomulation, resulting
in potent silencing (~70%) activity in vivo with
no observed hepatotoxicity and innate immune
stimulation [56].
Notably, the PEGylation methods also influ-
ence the stability of siRNA delivery. For exam-
ple, simply mixing siRNAs with PEGylated lipo-
somes results in poor loading of siRNAs onto the
outer surface of particles, causing the premature
release of siRNA, whereas post-PEG-coating on
the siRNA liposome could enable more stable
siRNA delivery. This aspect has been discussed
and reviewed in detail by Buyens et al. [48].
Although the beneficial PEGylation effect on
siRNA delivery is well acknowledged, it has been
found that repeated injection of PEGylated NPs
could induce the loss of their long circulation
characteristics owing to the production of the
anti-PEG IgM [57]. The phenomenon of acceler-
ated blood clearance serves as a reminder of the
limitation of PEG-coated NPs.
In addition, other challenges in the context of
the pharmaceutical development of lipid-based
siRNA therapeutics have been reviewed exten-
sively by Gindy et al., including the development
of a robust manufacturing process, the setting of
appropriate product specifications and controls,
development of strategies to assess and ensure
product stability, and the evaluation of product
comparability throughout development [5 8].
Improving biocompatibility & reducing
immunotoxicity
To reduce the immunotoxicity of siRNA deliv-
ery, some biogenic materials were included in the
cationic lipid-based siRNA delivery system. For
example, hyaluronic acid, a biogenic component
distributed widely in the ECM, was introduced
into LPD-NPs by Chono et al. to develop a lipo-
some–protamine–hyaluronic acid (LPH)-NP
[59]. Hyaluronic acid provides LPH-NP multi-
valent charges to enhance the particle conden-
sation while containing no immunostimulatory
CpG motifs. This LPH-NP delivery of siRNA
induced a 80% silence of luciferase activity in
the metastatic B16F10 tumor in the lungs at a
low, single, intravenous injection dose of 0.15 mg
siRNA/kg [59 ], while not causing obvious
immunotoxicity at a dose range of 0.15–1.2 mg
siRNA /kg. Recently, they employed LPH-NP in
the delivery of siRNA for silencing of CD47, a
‘self-marker’ that is usually overexpressed on the
surface of cancer cells to enable them to escape
from immunosurveillance, and achieved effec-
tive inhibition of melanoma tumor growth and
lung metastasis [6 0].
To improve the biocompatibility of cationic
lipid delivery systems, Yang et al. developed a
lipid–polymer hybrid NP prepared by a single-
step nanoprecipitation of a formulation of cat-
ionic lipids, BHEM– Chol and amphiphilic
polymers. Such hybrid NPs exhibited excel-
lent stability in serum and showed significant
improvement on biocompatibility compared
with the pure BHEM–Chol particles [26 ]. A
similar toxicity reduction effect has also been
observed for other hybrid NPs with involvement
of polyglutamate [61– 63] .
Improving the cellular uptake
& enhancing siRNA release into the
cytoplasm
To enhance the cellular uptake and further
endosomal escaping of siRNA, some neutral
helper lipids, such as DOPE and 1,2-distear-
oyl-sn-glycero-3-phosphocholine were added
to the cationic lipid-based siRNA delivery sys-
tem. The role of such fusogenic lipids has been
reviewed by Wasungu et al. [4 4]. DOPE pro-
foundly affects the polymorphic features of the
liposome–siRNA complex (termed lipoplexes)
in that it promotes the transition from a lamel-
lar to a hexagonal phase, thus inducing fusion
and disruption of the membrane. The opti-
mized 3-b-(N-[N´,N´-dimethylaminoethane]
carbamoyl) Chol/DOPE-based lipoplexes have
been found to improve the transfection ef-
ciency of the lipoplex [64 ]. Khoury et al. pre-
pared a NP using the cationic lipid RPR209120
in combination with DOPE. This NP enables
effective systemic delivery of DNA, resulting
in successful silencing of TNF-a in collagen-
induced arthritis of mice, and complete cure of
collagen-induced arthritis via weekly intrave-
nous administration [65] . Since then, this for-
mulation for siRNA delivery to myeloid cells
has been successfully applied [66,67].
By replacing the protamine/DNA core of
the LPD NPs with a pH-sensitive calcium
Nanomedicine (20 14) 9(1)
110 future science group
Review Lin, Chen, Zhang & Zheng
phosphate (CaP) core, Yang et al. recently
developed a biodegradable lipid/calcium/phos-
phate NP (LCP-NP). CaP is biocompatible, bio-
degradable material and native to the body, as
it is the principle mineral component of teeth
and bones. It is hypothesized that the CaP core
could be rapidly dissolved at acidic endosome
pH, which will increase the osmotic pressure and
cause NP disassembly, endosome swelling and,
finally, siRNA release into the cytoplasm. The
study showed that the LCP-NP releases more
cargo to the cytoplasm compared with the LPD
formulation, leading to a significant (~40-fold
in vitro and ~fourfold in vivo) improvement in
siRNA delivery. More recently, they coformu-
lated three siRNAs against MDM2, c-myc and
VEGF in a LCP-NP. Such a formulation caused
simultaneous silencing of the three oncogenes in
metastatic nodules, and resulted in significant
inhibition of lung metastases (~70–80%) at a
relatively low dose (0.36 mg/kg) without any
observed toxicity [68].
Recently, to enhance endosomal escape,
Harashima et al. synthesized a pH-sensitive
cationic lipid, YSK05, which contains a tertiary
amine group for pH sensitivity. They incor-
porated this lipid into the above mentioned
MEND siRNA delivery system, and found
that the YSK05–MEND combination had a
better ability for endosomal escape than other
MEND-containing conventional cationic lip-
ids [69, 70]. Some other new cationic lipids have
also been reported for improving siRNA deliv-
ery, such as N´,N´-dioctadecyl-N-4, 8-diaza-
10-aminodecanoylglycine amide [7 1] a nd
1,2-dilinoleyloxy-3-dimethylaminopropane,
which will be the subject of later discussion [2 3].
Active targeting delivery
Incorporating an active target motif, such as
transferrin, RGD, EGF and various cell-penetra-
tion peptides, to the cationic lipid-based siRNA
delivery system could significantly improve the
intracellular uptake and endosomal release of
siRNA. Daka et al. [7 2] and Ogris et al. [73] have
reviewed this aspect extensively.
Example of an advanced cationic
lipid-based siRNA delivery system:
stable nucleic acid lipid particles
& their application
Among the cationic lipid-based siRNA vehicles,
several advanced delivery systems have gained
worldwide attention owing to their high ef-
ciency in systemic siRNA delivery methods,
such as LPD, AtuPLEX, stable nucleic acid lipid
particle (SNALP) systems, LPD and AtuPLEX,
and have been well reviewed previously [74, 75].
Here, we take SNA LP as an example to highlight
its approach in overcoming barriers of siRNA
systemic delivery.
SNALP & its application
SNALP is constructed by three basic lipids:
an ionizable cationic lipid (1,2-dilinoleyloxy-
3-dimethylaminopropane), a neutral helper
lipid, including Chol and fusogenic lipids, and
a PEG lipid (Table 2). In SNALPs, the backbone-
modified siR NA is encapsulated within a closed
shell of a cationic-zwitterionic lipid bilayer, fur-
nished with an outer PEG shield (Figu re 2a). The
lipid bilayer contains a mixture of cationic and
fusogenic lipids to enable cellular uptake and
further endosomal escape owing to electrostatic
interactions between the negatively charge cell
Table 2. Stable nucleic acid lipid particle formulations.
Stable nucleic acid lipid particle formulations in systemic
delivery
Target siRNA and
modification (yes/no)
Animal model Ref.
DSPC:Chol:PEG–C–DMA:DLin– DMA (20:48:2:30) siRNA Hepatitis B virus (yes) Mouse model of hepatitis B
virus replication
[23]
DSPC:Chol:PEG–C–DMA:DLin– DMA (10:48:2:40) siRNA ApoB (yes) Cynomolgus monkeys [76]
DSPC:Chol:PEG–C–DMA:DLin– DMA (10:48:2:40) siRNA Cell cycle proteins PLK1 (yes) Intrahepatic mouse tumor
models and s.c. tumor
models
[78–80]
DSPC:Chol:PEG–C–DMA:cationic lipid (10:40:10:40)–siRNA Factor VII (yes) C57BL / 6 mice [24]
DPPC:PEG–C–DMA:Chol:DLin–KC2–DMA (7.1:1.4:34.3:57.1)–siRNA ApoB, transthyretin (yes) Cynomolgus monkeys [24]
PEG–DMG:Chol:Octyl–CLinDMA (2:48:50) –siRNA Luciferase (yes) Rosa26-LSL-Luc mouse strain,
Adeno-liver-Luc mice
[77]
Chol: Cholesterol; DLin– DMA: 1,2-dilinoleyloxy-3-dimethylaminopropane; DLin–KC2–DMA: 2,2-dilinoleyl- 4-dimethylaminoethyl- (1,3)-
dioxolane; DMG: Dimyristoylglycerol; DPPC: Dipalmitoylphosphatidylcholine; DSPC: 1,2-distearoyl-sn- glycero-3-phosphocholine; Octyl–CLinDMA: 2-( [8- ([3b]-
cholest-5-en-3-yloxy)octyloxy) -N,N-dimethyl-3- ([9Z,12Z] -octadeca-9,12-dien-1-yloxy)propan-1-amine; PEG–C–DMA: 3-N- ([-methoxy PEG 2000] carbamoyl) -1,2-
dimyristyloxypropylamine; s.c.: Subcutaneous.
www.futuremedicine.com 111
future science group
Lipid-based nanoparticles in the systemic delivery of siRNA Review
membrane and cationic lipids. The coated PEG
provides a neutral, hydrophilic exterior to shield
the cationic bilayer, protect the nucleic acid core
against degradation by nucleases, sterically sta-
bilize the particles against disassembly in col-
lagen networks and prevent nonspecific bind-
ing to cells, thus making NPs ‘cohesive’ and
‘stealthy’ to prevent siRNA degradation and
rapid clearance in the bloodstream.
The SNALPs are approximately 70–150 nm
in diameter (Figure 2b), and their major accumula-
tion (±standard deviation) was found in mouse
liver (28 ± 1.7% of injected dose administered)
and spleen (8.2 ± 2.8% of injected dose admin-
istratered) [2 3]. Their biodistribution is related
to the property of NPs, such as size, lipid and
PEGylation features. In general, the PEGylated
NPs are readily navigated to the mononuclear
phagocyte systems, such as the liver and spleen.
The therapeutic efficacy of SNALP–siRNA
against HBV was validated in a HBV replication
mouse model by Morrissey et al. [23]. They found
that SNALP delivery prolonged the circulation
time of siRNA and achieved efficient inhibition
of serum HBV by daily intravenous injections
of 3 mg/kg/day of siRNA [23]. Zimmermann
et al. further demonstrated the efficient systemic
delivery of SNALP–siRNA in nonrodent spe-
cies. They administered intravenous injections of
SNALP–siRNA against the ApoB gene at single
doses of 1 or 2.5 mg/kg on cynomolgus monkeys
and achieved potent gene knockdown of ApoB
mRNA (>90%). Significant downregulation of
ApoB protein, serum Chol and low-density lipo-
protein (LDL) levels was also observed as early as
24 h after treatment and lasted for 11 days with
a 2.5-mg/kg siRNA dose [76].
More recently, Semple et al. found a high-
performance lipid, 2,2-dilinoleyl-4-dimethyl-
aminoethyl-(1,3)-dioxolane by screening vari-
ous ionizable cationic lipids, and demonstrated
the excellent activity of the 2,2-dilinoleyl-
4-dimethylaminoethyl-(1,3)-dioxolane-based
SNALP in effectively silencing the endogenous
Cytosolic siRNA–gold (%)
Hepatocytes HeLa
Time (h)
1. 51. 0
0.5
1. 5
1. 0
2.0 3.0 4.0 5.0 6.0
Neutral lipids Cationic lipids
siRNA PEG lipids
pH 5.5–6.5
Trapped in the
EE compartment
Transport through
the PM
Trapped in the
LE compartment
pH >5.5
Trapped in
the Lys
pH >4.5
Cytosolic release of siRNAs
M
LE
Golgi
500 nm 50 nm
In vitro HeLa cells
200 nm
200 nm
200 nm
200 nm
50 nm
50 nm
50 nm
50 nm
PM
PM
EE
EE
LE
LE
Lys
Golgi 0.5
50 nm
Figure 2. Stable nucleic acid lipid particle siRNA delivery system. (A) Stable nucleic acid lipid particle (SNALP) siRNA delivery
system. (B) Visualization of SNALP–siRNA –gold by electron microscopy. (C) Ultrastructural analysis of SNALP in vitro trafficking.
SNALP–siRNA–gold detected in HeLa cells in vitro, by electron microscopy. SNALP–siRNA–gold was found in the extracellular matrix
close to the PM and inside the EE compartment, LE compartment and Lys within cells. Magnified images (right panels) permit
appreciation of the subcellular localization of siRNA–gold. (D & E) Cytosolic release of siRNA. (D) siRNA–gold concentrates within the LE
compartment of HeLa cells in vitro and (E) the quantification of cytosolic siRNA– gold release kinetics in a liver section in vivo (solid line)
and in HeLa cells in vitro (dashed line). The error bars represent the standard error.
EE: Early endocytic; LE: Late endocytic; Lys: Lysosome; PM: Plasma membrane.
Adapted with permission from [82] .
Nanomedicine (20 14) 9(1)
112 future science group
Review Lin, Chen, Zhang & Zheng
hepatic gene at doses of siRNA as low as
0.01 mg/kg in rodents and 0.1 mg/kg in nonhu-
man primates in vivo [24] . Meanwhile, Tao et al.
developed another optimized SNALP system by
screening cationic lipids and adjusting PEG lipid
density, and achieved potent gene knockdown in
the liver (90%) of mice [77] .
Besides the application of SNALP in liver
diseases, Judge et al. demonstrated the success-
ful delivery of siRNA to solid tumors in mice
targeting the PLK1 gene, resulting in signifi-
cant inhibition of subcutaneous Hep3B tumor
growth [78].
Notably, the SNALP system encapsulated
with self-amplifying RNA has been reported as
a new vaccine platform by Geall et al., which
substantially increases immunogenicity by
eliciting broad, potent and protective immune
responses [79]. Altogether, it is not surprising that
the SNALP system has entered or completed
multiple clinical trials for systemic siRNA deliv-
ery, such as VEGF and KSP in liver cancer, ApoB
in liver disease, TTR in transthyretin amyloi-
dosis, PLK1 in cancer and Elola in Ebola virus
disease [75] .
The impact of physiological constrains, such
as tumor vasculature, on the efficiency of siRNA
delivery has also been investigated. Li et al. found
that SNALP predominantly delivered siRNA to
areas adjacent to functional tumor blood vessels
by analyzing the spatial distribution of local-
ized target knockdown within tumor sections
relative to tumor hypoxia [80] . This pheno-
menon suggests that it is probably not easy for
the SNALP–siRNA complex to cross the ECM
with a size of 70–150 nm; therefore, most of
them are trapped in the areas adjacent to blood
vessels [80]. To improve the transport of SNALP
across the ECM barrier, Rudorf et al. devel-
oped a small SNALP, called a ‘mono-NALPs’,
which was self-assembled by solvent exchange
from solution containing siRNA mixed with
the four lipid components DOTAP, DOPE,
1,2-dioleoyl-sn-glycero-3-phosphatidylcholine
and 1,2-d istearoyl-sn-glycero-3-phosphoetha-
nolamine-PEG-2000. The mono-NALP has a
similar core–shell structure as SNALP, but is
only 30 nm in diameter, which should facilitate
its transport in vivo across the ECM to achieve
deeper tissue penetration [8 1].
Recently, the mechanism of cellular uptake,
intracellular transport and the endosomal escape
of siRNA by SNALP delivery had been investi-
gated. It has been shown that SNALPs entered
cells by both constitutive and inducible pathways
in a cell type-specific manner using CME and
macropinocytosis (F igur e 2C) [82]. However, the
escape of siRNAs from the endosome into the
cytosol occurred at low efficiency (<5%), sug-
gesting further optimization of the SNALP for-
mulation is required to improve the endosomal
release of siRNA (Fi gure 2D ).
Lipoprotein-related siRNA delivery
Lipoproteins are naturally existing endogenous
serum proteins that play critical roles in lipid
transport in humans. Lipoproteins share a com-
mon structure consisting of an apolar core sur-
rounded by a shell composed of a phospholipid
monolayer containing unesterified Chol and
one or more apolipoproteins (Figu re 3a). Apoli-
poproteins function as structural components
of lipoprotein particles, cofactors for enzymes
and ligands for cell surface receptors. Accord-
ing to their size, density, lipid and apolipoprotein
components, lipoproteins are divided into high-
density lipoprotein (HDL; 8–12 nm), LDL
(18–25 nm), very LDL (30–80 nm) and chylomi-
cron (75–1200 nm). Being endogenous carriers,
lipoproteins escape recognition as foreign entities
by the human immune system and escape absorp-
tion by the reticuloendothelial system [83]. Their
favorable blood circulation characteristics pres-
ent them as viable alternate drug delivery systems
without the need for PEGylation. Thus, lipopro-
tein NPs may provide a solution to the biocom-
patibility problem associated with most synthetic
nanodevices. Among the lipoproteins, HDL and
LDL have been widely used for systemic delivery
of imaging agents and chemotherapeutics [8 4 ,85] .
With the controlled ultra-small size (<30 nm)
and suitable blood circulation (half-life in blood
is 13.5 and 15 h for native HDL and LDL, respec-
tively) they hold great potential for effective
siRNA systemic delivery [84].
Natural lipoproteins for siRNA
delivery
In nature, both HDL and LDL play a vital role
in Chol transport. LDL functions as an extra-
cellular carrier for delivery of Chol to different
tissues via LDL receptor-mediated endocytosis,
while HDL is responsible for the reverse Chol
transport from peripheral tissues back to the
liver for catabolism [86]. Although the mecha-
nism of selective transport of lipids by HDL is
still unclear, it is generally accepted that HDLs
actively offload Chol esters and other lipids into
cells through SR-BI, via a nonendocytic pathway.
Therefore, LDL and HDL are attractive drug
delivery vehicles for potential cardio vascular
disease treatment. For example, Wolfrum et al.
www.futuremedicine.com 113
future science group
Lipid-based nanoparticles in the systemic delivery of siRNA Review
utilized HDL and LDL for siRNA delivery.
Unlike most of the cationic lipid-based NPs car-
rying siRNA by electrostatic interaction, HDL
or LDL stably load with siR NAs at a stoichio-
metry of 0.94 and 1.26 (molar proportion),
respectively, via interaction between lipoproteins
and lipophilic molecules on siRNA sequences,
such as Chol, long-chain fatty acids or bile acids
[22]. They found that LDL delivered most of the
Chol-labeled siRNA (Chol–siRNA) into the
liver, while HDL delivered Chol–siRNA into the
adrenal glands, ovaries, kidneys and liver. When
compared with naked Chol–siRNA–ApoB
(Chol–siRNA targeted to ApoB mRNA) treat-
ment, higher ApoB protein downregulation
(~8–15-fold more effective in the liver, gut
and blood) has been achieved by HDL deliv-
ery [2 2]. Interestingly, they also found that the
lipoprotein-associated Chol–siR NAs were taken
up by hepatocytes through a mechanism that
was dependent on lipoprotein-associated recep-
tors but independent of internalization of the
lipoprotein particles, suggesting the rapid uptake
of Chol–siRNAs compared with a much slower
cellular uptake of lipoprotein NPs.
Jin et al. has systematically studied the abil-
ity of LDL for Chol–siR NA delivery and dem-
onstrated that over 25 Chol–siRNAs could be
stably incorporated onto each LDL without
changing NP morphology [8 7]. The resulting
LDL–Chol–siRNA NPs were selectively taken
up by cells via LDL receptor-mediated endocy-
tosis, resulting in enhanced gene silencing com-
pared with naked Chol–siRNA (38 vs 0% gene
knockdown at 100 nM). However, most of the
Chol–siRNA was validated by being tracked in
Figure 3. Lipoprotein-based nanoparticle delivery system. (A) Lipoprotein or
lipoprotein-mimetic nanoparticle delivery system. (B) Transmission electron microscopy of
HPPS–siRNA nanoparticles. (C) Visualization of intracellular delivery of FITC-labeled siRNA in KB cells
(SR-BI receptor positive) by confocal imaging. (D) Quantitative measurement of the uptake of
FITC–Chol–siRNA –bcl-2 in different cell organelles via different delivery approaches (n = 3). The
results of (C & D) suggest the direct cytosolic delivery of siRNA by HPPS. Error bars represent the
standard deviation.
*p < 0.05; **p < 0.01.
Chol: Cholesterol; FITC: Fluorescein isothiocyanate; FITC–Chol–siRNA–bcl-2: FITC-labeled Chol-siRNA
targeted to bcl-2; HPPS: High-density lipoprotein-mimicking peptide–phospholipid scaffold.
Adapted with permission from [94 ].
FITC–Chol–
siRNA–bcl-2
Lipofectamine–
FITC–Chol–
siRNA–bcl-2
HPPS–FITC–
Chol–siRNA–
bcl-2
Intracellular uptake (%)
0
20
40
60
80
100
120
Cytosol
Nucleus and membrane
Other organelles
*
**
Phospholipid
siRNA
FITC–Chol–
siRNA–bcl-2
Alexa-Fluor®
transferrin Merged
FITC–Chol–
siRNA–bcl-2
Lipofectamine®
FITC–Chol–
siRNA–bcl-2
HPPS–FITC–
Chol–siRNA–
bcl-2
20 µm 20 µm
20 µm20 µm
20 µm 20 µm
20 µm
20 µm
20 µm
100 nm
Hydrophobic cargo
Apolipoprotein/mimetic
peptide
Nanomedicine (20 14) 9(1)
114 future science group
Review Lin, Chen, Zhang & Zheng
the endolysosome. By using a photochemical
internalization strategy to trigger endosomal
escape of LDLChol–siRNA, further improved
gene knockdown (78%) was achieved.
Kuwahara et al. extended the utility of endog-
enous lipoprotein delivery of siRNA for neuro-
logical diseases treatment [88] . They found that
HDL successfully delivered the Chol–siRNA
targeting OAT3 mRNA (CholsiOAT3) into
brain capillary endothelial cells and induced a
significant reduction of OAT3 mRNA expression,
whereas the naked Chol–siOAT3 showed poor
delivery efficacy to brain capillary endothelial
cells and had a negligible gene silencing effect.
They also validated that the efficient delivery of
Chol–siOAT3 by HDL was mainly mediated by
ApoE targeted to LDL receptor. In addition to
Chol modification, another lipophilic molecule,
vitamin E or a-tocopherol, has been used to mod-
ify siRNA for improved brain disease treatment.
The combination of vitamin E (a-tocopherol)-
conjugated siRNA with HDL caused dramatic
improvement of siRNA delivery to neurons,
resulting in specific knockdown of the targeted
BACE1 mRNA at both the mRNA and protein
level, especially in the parietal cortex [8 9].
Lipoprotein-mimetic NPs
Beyond the naturally existing lipoproteins,
many reconstituted lipoproteins and lipoprotein-
mimetic NPs have been developed for siRNA
delivery, such as reconstituted HDL (rHDL) and
HDL-mimicking peptide–phospholipid scaf-
fold (HPPS) NPs. Their application has been
extended to RNAi cancer therapeutics, as many
cancer cells, particularly ovarian, colon, lung,
prostate and breast cancers, highly express the
SR-BI receptor [9 0 –92 ].
In 2009, Zhang et al. developed HPPS, com-
posed of cholesteryl oleate, phospholipid and an
18-amino acid ApoA-1-mimetic peptide (Figur e 3a)
[20], which closely mimics the structural and
functional properties of plasma-derived HDL.
The HPPS NP has a small particle size (<30 nm)
(Figu re 3b), long circulation half-life (15 h) and
excellent biocompatibility, with high tolerability
of systemic administration doses (>2000 mg/kg)
[93]. HPPS could stably carry a high payload of
Chol–siRNA (ten siRNAs per particle) and
deliver them directly into the cytosol of the tar-
get cells via the SR-BI receptor (Fig ure 3 C & D),
thereby bypassing the detrimental endosomal
trapping, resulting in enhanced gene silencing
efficacy [94] . Further studies demonstrated that
SR-BI-driven cytosolic delivery is predominately
mediated through a lipid raft/caveolae-like
pathway [95]. Furthermore, the successful sys-
temic delivery of bcl-2-targeting Chol–siRNA
by HPPS has been demonstrated in a xenograft
tumor mouse model, which resulted in bcl-2
mRNA knockdown, tumor cell apoptosis and
tumor growth inhibition, with no observable
adverse effects, thus demonstrating the ability
of HPPS as a safe and efficient nanocarrier for
RNAi therapeutics [90] .
McMahon et al. developed a rHDL by using a
gold NP (~5 nm) core as a template to assemble
a mixed phospholipid layer and adsorb ApoA-1,
forming a core–shell spherical structure [3 0].
These synthesized structures have the general
size (~16 nm) and surface composition of natural
HDL, and importantly, have shown high bind-
ing affinity to Chol. It has been further demon-
strated that the rHDL is capable of carrying a
high payload of Chol–DNA (over ten DNA pay-
load per particle) and mediated efficient cellular
nucleic acid delivery [3 0].
Recently, Ding et al. reported a rHDL-
based siRNA delivery system by integrat-
ing Chol–siRNA loading into rHDL particle
assembly. Briefly, a lipid film comprised of
soya bean phospholipids, Chol and choles-
teryl esters was prepared and hydrated with
an aqueous solution containing Chol–siRNA
to form lipoplexes/Chol–siRNA complexes,
then ApoA-1 protein was added to assemble
rHDL/Chol–siR NA NPs. Compared with
unmodified siRNA, Chol–siRNA showed sig-
nificantly higher loading efficiency in rHDL.
Although the size of rHDL/Chol–siRNA NPs
is approximately 90 nm, the cytosolic delivery of
Chol–siRNA was demonstrated using confocal
microscopy. In addition, enhanced tumor uptake
of rHDL/Chol–siRNA was observed in compari-
son with the lipoplexes/Chol–siRNA complex
(without ApoA-1 protein), resulting in improved
of anti-tumor efficacy in vivo [96] .
As well as using lipophilic modified siR NA
(e.g., with Chol) to help siRNA stably incorpo-
rate into lipoprotein NPs, other helper materials,
such as cationic polymer, were also introduced to
facilitate siRNA loading into the rHDL–siRNA
delivery system. Shahzad et al. developed an
rHDL–siRNA delivery system using unmodi-
fied siRNA/oligolysine to hydrate a lipid film
composed of Chol, cholesteryl oleate and egg
yolk phospholipids, followed by the ApoA-1 pro-
tein to assemble rHDL particles. The resulting
rHDL/siRNA NPs have a sma ll size of 12–18 nm,
which facilitates systemic delivery of siRNA
in vivo, evidenced by the therapeutic silencing
of two proteins that are key to cancer growth
www.futuremedicine.com 115
future science group
Lipid-based nanoparticles in the systemic delivery of siRNA Review
and metastasis (STAT3 and FAK) in orthotropic
mouse models of ovarian and colorectal cancer
[97]. Recently, Rui et al. prepared complexes of
siRNA with various cationic polymers including
protamine, polyethyleneimines or poly-l-lysine,
and then coated them with anionic lipid layers
containing ApoA-1 protein, lipid and sodium
cholate to form a rHDL–siRNA delivery sys-
tem. This formulation has shown minimum
cytotoxicity with improved gene silencing ef-
ciency in the hepatocellular carcinoma cell line
SMMC-7721 when compared with cationic poly-
mer delivery [9 8]. The confocal studies revealed
that intracellular uptake of the rHDL–iRNA
occurs via the HDL-receptor-mediated pathway.
Overall, HDL-based siRNA delivery possesses
many attractive features, including ultra-small
size, favorable surface properties (no PEGylation
is required for achieving suitable blood circula-
tion) and direct delivery of siRNA into the cyto-
plasm of cells via the SR-BI-mediated pathway,
thus offering a new avenue for effective siRNA
delivery.
Apolipoprotein-incorporated
liposomes
In addition to lipoprotein-based nanoplat-
forms, many groups take the apolipoprotein
components of HDL or LDL, such as ApoA-1,
Apo-E and ApoB-100, as targeting motifs for
siRNA delivery. In 2007, Kim et al. incorpo-
rated ApoA-1 protein in cationic liposomes
formulation for siRNA delivery against HBV
and achieved a significant reduction of HBV
protein expression with high liver-targeting
specificity [99]. Nakayama et al. reported the use
of ApoA-1 or ApoE to enhance the targeting
efficacy of phosphatidylcholine-based liposomes
for Chol–siRNAs delivery [1 00] . The ApoE–lipo-
some also demonstrated higher efficiency in the
functional delivery of Chol–siRNA to mouse
liver than ApoA–liposomes. The delivery of
ApoE–liposomes was not significantly affected
by high endogenous levels of plasma LDL.
Apolipoprotein-free lipoprotein
Recently, a cationic solid lipid NP derived from
apolipoprotein-free reconstituted LDLs was
reported for target-specific systemic treatment
of liver fibrosis [10 1].
Other siRNA delivery systems
Lipid-like material lipidoid & its
application
As well as the use of cationic lipid-based NPs and
lipoprotein, another promising siR NA delivery
vehicle is lipid-like material (termed ‘lipidoids’)-
based NPs. Such NPs are structurally similar to
the SNALP, formulated with Chol and PEG
lipids, but with lipidoids instead of cationic lipids.
Akinc et al. synthesized a large library of over
1200 structurally diverse lipidoids. Among these,
the lipidoid 98N12-5 (five-tail) was identified as
performing optimally for in vivo siRNA deliv-
ery, as it could stably deliver siRNA to achieve
potent and specific gene therapeutic efficacy in
three different animal species: mice, rats and
nonhuman primates. Lipidoid 98N12-5–siRNA
delivery not only induced successful gene knock-
down of the Factor VII and ApoB gene in the
rodent liver, but also resulted in therapeutic ef-
cacy in lung tissue targeting respiratory syncytial
virus and in peritoneal macrophages against the
CD45 gene [21] . The same group further demon-
strated that in vivo biodistribution of the lipidoid
98N12-5 system was influenced by many factors,
including the formulation composition, nature
of PEG lipid, degree of drug loading and particle
size [10 2 ]. Akinc et al. finally developed a liver-
targeted lipidoid formulation with over 90% of
injected dose distribution in the liver [1 02].
In 2010, Love et al. developed a new generation
of lipidoid formulations using lipidoid C12-200,
1,2-distearoyl-sn-glycero-3-phosphocholine,
Chol and PEG lipid. Using this lipidoid for-
mulation, potent liver gene silencing has been
achieved in mice at a much lower injection dose
of siRNA (<0.01 mg/kg) compared with the
original lipidoid formulation (>1 mg/kg) [102 ,103].
The lipidoid formulation with a pool of five siR-
NAs also induced gene silencing of five specific
hepatic genes simultaneously via a single injec-
tion [10 3] . The formulation was further validated
in nonhuman primates, where significant knock-
down of the clinically relevant gene transthyretin
was observed at doses as low as 0.03 mg/kg, pro-
viding good potential for low-dose in vivo gene
silencing [10 3] .
The lipidoids system has also been applied in
ovarian tumor therapy by Huang et al. Follow-
ing intraperitoneal injection of lipidoid–siRNA
against the CLDN3 gene, a tight junction pro-
tein overexpressed in 90% of ovarian tumors,
obvious tumor growth inhibition and metastasis
suppression were observed in MISIIR/Tag trans-
genic mice and tumor-bearing mice derived from
mouse ovarian surface epithelial cells [1 0 4] .
Recently, the application of RNAi in the
immune system has been rapidly developed,
including using the lipidoids as a delivery sys-
tem [10 5] . The induction of innate immunity by
in vivo administration of siRNA was considered
Nanomedicine (20 14) 9(1)
116 future science group
Review Lin, Chen, Zhang & Zheng
as a major undesirable side effect of RNAi ther-
apy owing to the toxicities associated with exces-
sive cytokine release, such as TNF-a, IL-6 and
IFN release [42 ,106 –108]. Thus, it is essential to
control the undesirable immune activation dur-
ing application of the siR NA complex in treat-
ment of immune-related diseases. It has been
reported that certain chemical modifications to
the siRNA backbone, such as 2´-O-methyl- and
-fluoro-modified siRNA, could completely
abrogate its immunostimulatory activity [10 7] .
Nguyen et al. demonstrated that effective anti-
viral activity could be achieved without the
undesirable immune activation when lipidoid
98N12-5(1) was used to deliver 2´-O-methyl-
modified siRNA to the influenza ucleoprotein
gene [10 9] . After that, the same group optimized
the lipidoid system and demonstrated that the
lipidoid could be used as a vaccine adjuvant
to deliver immunostimulatory RNA to TLR-
expressing cells, inducing robust antiviral activ-
ity [105] . Recently, the lipidoid NPs were also
found to be able to deliver modified PD-L1
siRNA into Kupffer cells in vivo and enhance
natural killer and CD8+ T-cell-mediated hepatic
antiviral immunity [1 10] .
The intracellular uptake of siRNA by lip-
idoid-based NPs has been validated through
a macropinocytosis-mediated pathway [10 3] .
Recently, Sahay et al. further confirmed that
lipidoid C12-200 siRNA delivery is medi-
ated by Cdc42-dependent macropinocytosis,
and its internalization remains largely within
endosomes with little escape to the cytosol [111].
Neutral lipid-based liposomes
Although cationic NPs offer many advantages
in systemic delivery of siRNAs, including
protecting siRNAs from enzymatic degrada-
tion and facilitating cellular uptake and ef-
cient endosomal escape, their positive charges
undergo nonspecific interactions with negatively
charged cellular components, often causing side
effects and toxicity in drug delivery. To over-
come this, Landen et al. developed a neutral
liposome composed of 1,2-dioleoyl-sn-glycero-
3-phosphatidylcholine for systemic delivery of
siRNA against the oncoprotein EphA2, and
demonstrated the successful knockdown of
EphA2 gene expression in vivo and significant
tumor growth inhibition in an orthotropic
mouse model of ovarian cancer [1 12 ]. This neutral
liposomal system was further applied for deliv-
ery of various siRNAs, such as FAK [11 3], TG2
[114 ], PAR-1 [115] , ATP7B [116] , PELP1 [117] and
survivin splice variant 2B [118], among others.
Among them, the system’s utility for EphA2
gene therapy is scheduled to be investigated in
a Phase I clinical trial according to the NIH
clinical trials database [20 3].
Anionic lipid-based liposomes
Anionic liposomes offer another safer alternative
to cationic-based NPs. Koldehoff et al. utilized
an anionic liposome, composed of soya bean oil,
glycerol and phospholipids from egg, in delivery of
bcr–abl siRNA for treatment of chronic myeloid
leukemia and observed negligible side effects
[119]. However, anionic liposomes often have poor
siRNA encapsulation capability owing to the
electrostatic repulsion between negatively charged
liposomes and anionic siRNAs. To overcome this
hurdle a new formulation of siRNA–anionic lipo-
some has been developed based on physiologically
occurring anionic lipids, 1,2-dioleoyl-sn-glycero-
3-phospho-(1´-rac-glycerol), in which the anionic
liposomes integrate with siRNA through stable
calcium ion bridges. The optimal siRNA–anionic
liposomes showed good intracellular uptake,
efficient endosomal escape, and induced compa-
rable silencing efficiency with much lower toxic-
ity when compared with Lipofectamine® 2000
delivery [1 20] .
Conclusion
To overcome the obstacles in systemic siRNA
delivery, a number of NPs have been developed
with enhanced blood stability, decreased toxicity
and improved siRNA intracellular uptake to the
cytoplasm. This review covers recent progress
made in lipid-based NPs, which are categorized
according to their key lipid components, includ-
ing cationic lipid-, lipoprotein-, lipidoid-, neutral
lipid- and anionic lipid-based NPs. Compared
with inorganic and viral delivery systems, lipid
NPs in general have favorable biocompatible
and biodegradable properties. The advanced
cationic lipid NPs in combination with various
advantageous effects provided by different lipid
components, including the use of PEG lipids for
optimizing pharmacokinetics, cationic lipids
for improving cellular uptake and endosomal
escaping and neutral lipids for enhancing par-
ticle stability, enable achievement of significant
RNAi levels in the liver at a very low adminis-
tration doses (~0.01 mg/kg). The lipoprotein-
based NPs have shown superior advantages over
siRNA delivery owing to their excellent biocom-
patibility, favorable pharmacokinetics, nonim-
munogenicity, suitable small size and active tar-
geting features (built-in or redirected targeting)
[121,122]. Among them, the ultra-small HDL-like
www.futuremedicine.com 117
future science group
Lipid-based nanoparticles in the systemic delivery of siRNA Review
particles (<30 nm) with SR-BI-mediated direct
cytosolic delivery features hold great promise for
effective siRNA delivery to fulfill the vision of
personalized medicine.
Future perspective
Moving forward, we expect that lipid-based
NPs will have a key role in systemic applica-
tion of siRNA in the clinic. In particular, it will
have an enabling role for personalized cancer
medicine, where siRNA delivery will join forces
with genetic profiling of individual patients to
achieve the best treatment outcome. We expect
major advancements in efficient delivery of
siRNA into the tissue beyond the liver/spleen
by optimizing the particle size, charge and sur-
face properties. For poorly permeable tumors
(e.g., pancreatic cancer), decreasing particle
size (<30 nm) and including active targeting
will be beneficial. Future research is expected to
improve particle biocompatibility and address
the immuno stimulatory effects to improve
the therapeutic index. Finally, lipid NP-based
siRNA delivery systems have the potential to
achieve multi target RNAi or synergism with
other therapeutic modalities, leading to a more
effective treatment efficacy.
Financial & competing interests disclosure
The authors would like to thank the China– Canada Joint
Health Research Initiative (NSFC-30911120489, CIHR
CCI-102936), DLVR Therapeutics, Canadian Institutes
of Health Research, Ontario Institute for Cancer Research,
Natural Sciences and Engineering Research Council of
Canada, Canada Foundation for Innovation and the Joey
and Toby Tanenbaum/Brazilian Ball Chair in Prostate
Cancer Research for funding support. G Zheng is a scientific
cofounder of DLVR Therapeutics. The authors have no
other relevant affiliations or financial involvement with
any organization or entity with a financial interest in or
financial conflict with the subject matter or materials
discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of
this manuscript.
Executive summary
siRNA systemic delivery barriers
The ability to deliver siRNA systemically to its intended site of action is a challenge for the current field of RNAi therapy.
Lipid-based nanoparticles for siRNA delivery
Nanoplatforms have higher biocompatibility and lower toxicity in comparison with inorganic nanoparticles and viral vectors.
Function-engineerable nanoparticles in combination with the advantageous effects of various functional lipid components enable
achievement of enhanced blood stability, decreased toxicity and improved siRNA delivery to the cytoplasm.
Lipoprotein-based nanoparticles with excellent biocompatibility, favorable pharmacokinetics, nonimmunogenicity, suitably small size
(<30 nm) and active targeting features demonstrate great potential for siRNA delivery.
Lipid nanoparticle-based siRNA delivery enables us to fulfill the vision of personalized medicine.
References
Papers of special note have been highlighted as:
of interest
1 Fire A, Xu S, Montgomery MK, Kostas SA,
Driver SE, Mello CC. Potent and specific
genetic interference by double-stranded R NA
in Caenorhabditis elegans. Nature 391(6669),
80 6– 811 (1998 ).
 Describes the discover y of RNAi.
2 Elbashir SM, Harborth J, Lendeckel W,
Yalcin A, Weber K, Tuschl T.
Duplexes of 21-nucleotide R NAs
mediate R NA interference in cultured
mammalian cells. Nature 411(6836),
494– 498 (2001).
 First validation of specific gene knockdow n
in mammalian cells.
3 Whelan J. First clinica l data on R NAi. Drug
Discov. Today 10(15), 1014–1015 (20 05).
 First clinica l trial of siR NA.
4 Whitehead KA, Langer R, A nderson DG.
Knock ing down barriers: advances in siRNA
delivery. Nat. Rev. Drug Discov. 8(2),
129–138 (2009).
5 Alexis F, Pridgen E, Molnar LK, Farokhzad
OC. Factors affecting the clearance and
biodistribution of polymeric nanoparticles.
Mol. Pharm. 5(4), 505–515 (2008).
6 Bumcrot D, Manoharan M, Koteliansky V,
Sah DW. RNAi therapeutics: a potential new
class of pharmaceutical drugs. Nat. Chem.
Biol. 2(12), 711–719 (200 6).
7 Santel A, Aleku M, Keil O et al. A novel
siRNA-lipoplex technology for RNA
interference in the mouse vascular
endothelium. Gene Ther. 13(16), 1222–1234
(2006 ).
8 van de Water FM, Boerman OC, Wouterse
AC, Peters JG, Russel FG, Ma sereeuw R.
Intravenously administered short interfering
RNA accumulates in the kidney and
selectively suppresses gene function in renal
proximal tubules. Drug Metab. Dispos. 34(8),
1393–1397 (2006).
9 Takakura Y, Mahato R I, Hashida M.
Extravasation of macromolecules. Adv. Drug
Deliv. Rev. 34(1), 93 –108 (19 98).
10 Li W, Szoka FC Jr. Lipid-based nanoparticles
for nucleic acid delivery. Pharm. Res. 24(3),
438–449 (2007).
11 Cabral H, Matsumoto Y, Mizuno K et al.
Accumulation of sub-100 nm polymeric
micelles in poorly permeable tumours
depends on size. Nat. Nanotechnol. 6(12),
815 –823 (2011).
12 Pluen A, Boucher Y, Ramanujan S et al. Role
of tumor-host interactions in interstitial
diff usion of macromolecules: cranial vs.
subcutaneous tumors. Proc. Natl Acad. Sci.
USA 98(8), 4628 4633 (2001).
13 Stylianopoulos T, Poh MZ, Insin N et al.
Diffusion of particles in the extracellular
Nanomedicine (20 14) 9(1)
118 future science group
Review Lin, Chen, Zhang & Zheng
matrix: the effect of repulsive electrostatic
interactions. Biophys. J. 99(5), 1342–1349
(2010).
14 Petros RA, Desimone JM. Strategies in the
design of nanoparticles for therapeutic
applications. Nat. Rev. Drug Discov. 9(8),
615 – 627 (2010 ).
15 Conner SD, Schmid SL. Regulated portals of
entry into the cell. Nature 422(6927), 37–4 4
(2003).
16 Swanson JA, Watts C. Macropinocytosis.
Trends Cell. Biol. 5(11), 424 428 (1995).
17 Bareford LM, Swaan PW. Endocytic
mechanisms for targeted drug delivery. Adv.
Drug Deliv. Rev. 59(8), 748–758 (2007).
18 Partlow KC, Lanza GM, Wickline SA.
Exploiting lipid raf t transport with
membrane targeted nanoparticles: a strategy
for cytosolic drug delivery. Biomaterials
29(23), 3367–3375 (2008).
19 Kunisawa J, Masuda T, Katayama K et al.
Fusogenic liposome delivers encapsulated
nanoparticles for cytosolic controlled gene
release. J. Control . Release 105(3), 3 44– 353
(2005).
20 Zhang Z , Cao W, Jin H et al. Biomimetic
nanocarrier for direct c ytosolic drug deliver y.
Angew. Chem. Int. Ed . Engl. 48(48),
9171– 9175 (2 0 09 ).
21 Ak inc A, Zumbuehl A, Goldberg M et al.
A combinatorial library of lipid-like materials
for deliver y of RNAi therapeutics. Nat.
Biotechnol. 26(5), 561–569 (2008).
22 Wolfrum C, Shi S, Jayapra kash KN et al .
Mechanisms and optimization of in vivo
delivery of lipophilic siRNAs. Nat.
Biotechnol. 25(10), 1149–1157 (2 007 ).
 First paper on the in vi vo delivery of siR NA
using natural lipoproteins.
23 Morrissey DV, L ockridge JA, Shaw L et al.
Potent and persistent in vivo anti-HBV
activity of chemically modified siRNAs. Nat .
Biotechnol. 23(8), 1002–1007 (2005).
24 Semple SC, A kinc A, Chen J et al. Rational
design of cationic lipids for siRNA delivery.
Nat. Biotechnol. 28(2), 172 –176 (2 010).
25 Truong NP, Gu W, Prasada m I et al.
An influenza virus-inspired polymer system
for the timed release of siRNA. Nat.
Commun. 4, 1902 (2013).
26 Yang XZ, Dou S, Wang YC et al. Single-step
assembly of cationic lipid–polymer hybrid
nanoparticles for systemic delivery of siR NA.
ACS Nano 6(6), 4955– 4965 (2012).
27 Dunn SS, Tian S, Bla ke S et al.
Reductively responsive siR NA-conjugated
hydrogel nanoparticles for gene silencing.
J. Am. Chem. Soc. 134 (17), 742 3 –74 30
(2012).
28 Endo-Takahashi Y, Negishi Y, Nakamura A
et al. pDNA-loaded Bubble liposomes as
potential ultrasound imaging and gene delivery
agents. Biomaterials 34(11), 2807–2813 (2013).
29 Suma T, Miyata K, A nraku Y et al. Smart
multilayered assembly for biocompatible
siRNA delivery featuring dissolvable silica,
endosome-disrupting polycation, and
detachable PEG. ACS Nano 6(8), 6 693 –6705
(2012).
30 McMahon K M, Mutha rasan RK, Tripathy S
et al. Biomimetic high densit y lipoprotein
nanoparticles for nucleic acid delivery. Nano
Lett. 11( 3), 1208 –1214 (2 011).
31 Qi L, Gao X. Quantum dot-amphipol
nanocomplex for intracellular delivery and
real-time imaging of siR NA. ACS Nano 2(7),
1403–1410 (2 008).
32 Lee JH, L ee K, Moon SH, Lee Y, Park TG,
Cheon J. All-in-one target-cell-specific
magnetic nanopa rticles for simultaneous
molecular imaging and siRNA deliver y.
Angew. Chem. Int. Ed . Engl. 48(23),
4174 4179 (20 09).
33 Jiang S, Eltoukhy AA, Love KT, Langer R,
Anderson DG. Lipidoid-coated iron oxide
nanoparticles for efficient DNA and siRNA
delivery. Nano Lett. 13(3), 1059 –1064
(2013).
34 Posadas I, Guerra FJ, Cena V. Nonviral
vectors for the deliver y of small interfering
RNAs to the CNS. Nanomedicine (Lond.)
5(8), 1219–123 6 (2 010).
35 Al-Jamal KT, Gherardini L, Bardi G et al.
Functiona l motor recovery from brain
ischemic insult by carbon nanotube-mediated
siRNA silencing. Proc. Natl Acad. Sci. USA
108(27), 109 52–10957 (2 011).
36 Lee H, Lytton-Jean AK, Chen Y et al.
Molecula rly self-assembled nucleic acid
nanoparticles for targeted in vivo siRNA
delivery. Nat. Nanotechnol. 7(6), 389–393
(2012).
37 Alvarez-Erviti L, Seow Y, Yin H, Betts C,
Lak hal S, Wood MJ. Delivery of siRNA to
the mouse brain by systemic injection of
targeted exosomes. Nat. Biotechnol. 29(4),
341–345 (2011).
38 Sorensen DR, Leirdal M, Sioud M. Gene
silencing by systemic deliver y of synthetic
siRNAs in adu lt mice. J. Mol. Biol. 327(4 ),
761–766 (2003).
39 Sioud M, Sorensen DR. Cationic
liposome-mediated delivery of siRNAs in
adult mice. Biochem. Biophys. Res. Commun.
312(4), 1220–1225 (2003).
40 Ren T, Song YK, Zhang G, Liu D. Structural
basis of DOTMA for its high intravenous
transfection activity in mouse. Gene Ther.
7(9), 76 4–768 (2000).
41 Zhang S, Zhao B, Jiang H, Wang B, Ma B.
Cationic lipids and poly mers mediated vectors
for deliver y of siRNA. J. Control. Release
123(1), 1–10 (2007).
42 Kedmi R, Ben-Arie N, Peer D. The systemic
toxicity of positively charged lipid
nanoparticles and the role of Toll-like
receptor 4 in immune activation. Biomaterials
31(26), 6867–6875 (2010).
43 Tao W, Mao X, Davide JP et al.
Mechanistically probing lipid-siRNA
nanoparticle-associated toxicities identifies
Jak inhibitors effective in mitigating
multifaceted toxic responses. Mol. Ther.
19(3), 567–575 (2011).
44 Wasungu L, Hoekstra D. Cationic lipids,
lipoplexes and intracellular delivery of genes.
J. Control . Release 116(2), 255–264 (2006).
45 Li SD, Chen YC, Hackett MJ, Huang L .
Tumor-targeted delivery of siRNA by
self-assembled nanopa rticles. Mol. Ther.
16(1), 163–169 (20 08).
 Good example of the improvement of stable
loading siRNA via precondensing into the
core of the liposome.
46 Aleku M, Schulz P, Keil O et al. Atu027, a
liposoma l small interfering RNA formulation
targeting protein kinase N3, inhibits cancer
progression. Cancer Res. 68(23), 9788 –9798
(2008).
47 Santel A, Aleku M, Roder N et al. Atu027
prevents pulmonar y metastasis in
experimental and spontaneous mouse
metastasis models. Clin. Cancer Res. 16(2 2),
5469–5480 (2010).
48 Buyens K, De Smedt SC, Braeckma ns K et al.
Liposome based systems for systemic siRNA
delivery: stability in blood sets the
requirements for optimal carrier design.
J. Control . Release 158(3), 362–370 (2012).
49 Soutschek J, Akinc A, Bramlage B et al.
Therapeutic silencing of an endogenous gene
by systemic administration of modified
siRNAs. Nature 432(7014), 173–178 (2004).
50 Immordino ML, Dosio F, Cattel L. Stealth
liposomes: review of t he basic science,
rationale, and clinica l applications, existing
and potential. Int. J. Nanomed. 1(3), 297–315
(2006 ).
51 Li SD, Huang L. Stealth nanoparticles: high
density but sheddable PEG is a key for tumor
targeting. J. Control. Release 145(3), 178–181
(2010).
52 Gomes-Da-Silva LC, Fonseca NA, Moura V,
Pedroso de Lima MC, Simoes S, Moreira JN.
Lipid-based nanoparticles for siRNA delivery
in cancer therapy: paradigms and cha llenges.
Acc. Chem. Res. 45(7), 1163–1171 (2012).
53 Sonoke S, Ueda T, Fujiwara K et al. Tumor
regression in mice by delivery of Bcl-2 small
www.futuremedicine.com 119
future science group
Lipid-based nanoparticles in the systemic delivery of siRNA Review
119
www.futuremedicine.com
interfering RNA with PEGylated cationic
liposomes. Cancer Res. 68(21), 8843–8851
(2008).
54 Yagi N, Manabe I, Tottori T et al.
A nanoparticle system specifically designed to
deliver short interfering RNA inhibits tumor
growth in vivo. Cancer Res. 69(16),
6531–6538 (20 09).
55 Carmona S, Jorgensen MR, Kolli S et al .
Controlling HBV replication in vivo by
intravenous administration of triggered
PEGylated siRNA-nanoparticles. Mol. Pharm.
6(3), 706 –717 (2009).
56 Hatakeyama H, Akita H, Ito E et al. Sy stemic
delivery of siRNA to tumors using a lipid
nanoparticle containing a tumor-specific
cleavable PEG-lipid. Biomaterials 32 (18),
4306–4316 (2011).
57 Ishida T, Wang X, Shimizu T, Nawata K,
Kiwada H. PEGylated liposomes elicit an
anti-PEG IgM response in a
T cell-independent manner. J. Control. Release
122(3), 349–355 (2007).
58 Gindy ME, Leone AM, Cunningha m JJ.
Challenges in the pharmaceutical
development of lipid-based short interfering
ribonucleic acid therapeutics. Expert Opin.
Drug Deliv. 9(2), 171–182 (2012).
59 Chono S, Li SD, Conwell CC, Huang L.
An efficient and low immunostimulator y
nanoparticle formulation for systemic siRNA
delivery to the tumor. J. Control . Release
131(1), 64– 69 (2008).
60 Wang Y, Xu Z, Guo S et al. Intravenous
delivery of siRNA targeting CD47 effectively
inhibits melanoma tumor growth and lung
metastasis. Mol. Ther. 21(10), 1919 –19 29
(2013).
61 Schlegel A, Bigey P, Dhotel H, Scherman D,
Escriou V. Reduced in vitro and in vivo
toxicity of siRNA-lipoplexes with addition of
polyglutamate. J. Control. Release 165(1), 1–8
(2013).
62 Asai T, Matsushita S, Kenjo E et al. Dicetyl
phosphate-tetraethylenepentamine-based
liposomes for systemic siRNA delivery.
Bioconjug. Chem. 22(3), 429– 435 (2011).
63 Schlegel A, Largeau C, Bigey P et al. Anionic
polymers for decreased toxicity and enhanced
in vivo delivery of siR NA complexed with
cationic liposomes. J. Control. Release 152( 3),
393 401 (2011).
64 Zhang Y, Li H, Sun J et al. DC-Chol/DOPE
cationic liposomes: a comparative study of the
influence factors on plasmid pDNA and
siRNA gene delivery. Int. J. Pharm. 390(2),
198–207 (2010).
65 Khoury M, Louis-Plence P, Escriou V et al.
Efficient new cationic liposome formulation
for systemic delivery of small interfering R NA
silencing tumor necrosis factor a in
experimental arthritis. Arthritis Rheum.
54(6), 1867–18 77 (200 6 ).
66 Courties G, Seiffart V, Presumey J et al.
In vivo R NAi-mediated silencing of TAK1
decreases inflammatory Th1 and Th17 cells
through targeting of myeloid cells. Blood
116(18), 3505–3516 (2010 ).
67 Courties G, Ba ron M, Presumey J et al.
Cytosolic phospholipase A 2a gene silencing
in the myeloid lineage alters development of
Th1 responses and reduces disease severity in
collagen-induced arthritis. Arthritis Rheum.
63(3), 681–690 (2011).
68 Yang Y, Li J, Liu F, Huang L. Systemic
delivery of siRNA via LCP nanoparticle
efficiently inhibits lung metastasis. Mol. Ther.
20(3), 609– 615 (2012).
69 Sato Y, Hatakeyama H, Sakurai Y, Hyodo M,
Akita H, Harashima H. A pH-sensitive
cationic lipid facilitates the delivery of
liposoma l siRNA and gene silencing activit y
in vitro and in vivo. J. Control. Release 163(3),
267–276 (2012).
70 Sakurai Y, Hatakeyama H, Sato Y, Hyodo M,
Akita H, Harashima H. Gene silencing via
RNA i and siR NA qua ntification in tumor
tissue using MEND, a liposomal siR NA
delivery system. Mol . Ther. 21(6), 1195 –1203
(2013).
71 Mevel M, Kamaly N, Carmona S et al.
DODAG; a versatile new cationic lipid that
mediates efficient delivery of pDNA and
siRNA. J. Control . Release 143(2), 222–232
(2010).
72 Daka A, Peer D. RNAi-based nanomedicines
for targeted personalized therapy. Adv. Drug
Deliv. Rev. 64(13 ), 15 08 –15 21 (2012).
73 Ogris M, Wagner E. To be targeted: is the
magic bullet concept a viable option for
synthetic nucleic acid therapeutics? Hum.
Gene Ther. 22(7), 799–807 (2011).
74 Tseng YC, Mozumdar S, Huang L .
Lipid-based systemic delivery of siR NA. Adv.
Drug Deliv. Rev. 61(9), 721–731 (2009).
75 Barros SA, Gollob JA. Safety profi le of RNAi
nanomedicines. Adv. Drug Deliv. Rev. 6 4(15) ,
1730–1737 (2012).
76 Zimmermann TS, Lee AC, Akinc A et al.
RNA i-mediated gene silencing in non-human
primates. Nature 441(70 89 ), 111–114 ( 2006 ).
 First report on RNAi-mediated gene
silencing in nonhuman primates by systemic
deliver y of siR NA.
77 Tao W, Davide JP, Cai M et al. Noninvasive
imaging of lipid nanoparticle-mediated
systemic delivery of small-interfering RNA to
the liver. Mol. Ther. 18(9), 1657–16 66
(2010).
78 Judge AD, Robbins M, Tavakoli I et al.
Confirming the RNAi-mediated mechanism
of action of siRNA-based cancer therapeutics
in mice. J. Clin. Invest. 119(3), 661–673
(2009).
79 Geall AJ, Verma A, Otten GR et al. Nonviral
delivery of self-amplifying R NA vaccines.
Proc. Natl Acad. Sci . USA 109(3 6),
146 04–1460 9 ( 2012).
80 Li L, Wang R, Wilcox D et al. Tumor
vasculature is a key determinant for the
efficiency of nanoparticle-mediated siRNA
delivery. Gene Ther. 19(7), 775–780 (2012).
81 Rudorf S, Radler JO. Self-assembly of stable
monomolecular nucleic acid lipid particles
with a size of 30 nm. J. Am. Chem. Soc.
13 4( 28), 11652 –11658 (2 012).
82 Gilleron J, Querbes W, Zeigerer A et al.
Image-based a nalysis of lipid
nanoparticle-mediated siRNA delivery,
intracellular trafficking and endosomal escape.
Nat. Biotechnol. 31(7), 638– 646 (2013).
83 Rensen PC , De Vrueh RL, Kuiper J,
Bijsterbosch MK, Biessen EA, Van Berkel TJ.
Recombinant lipoproteins: lipoprotein-like
lipid particles for drug ta rgeting. Adv. Drug
Deliv. Rev. 47(2–3), 251–276 (2001).
84 Ng KK, Lovell JF, Zheng G.
Lipoprotein-inspired nanoparticles for cancer
theranostics. Acc. Chem . Res. 44(10),
1105 –1113 (2011).
85 Jin HL, Chen J, Lovell JF, Zhang ZH, Zheng
G. Synthesis and development of
lipoprotein-based na nocarriers for
light-activated theranostics. Israel J. Chem.
52(8–9), 715–727 (2012).
86 Krieger M. Charting the fate of the ‘good
cholesterol’: identification and characterization
of the high-density lipoprotein receptor SR-BI.
Annu. Rev. Biochem. 68, 523–558 (1999).
87 Jin H, Lovell JF, Chen J et al. Mechanistic
insights into LDL nanoparticle-mediated
siRNA delivery. Bioconjug. Chem. 23(1),
3341 (2 012).
88 Kuwahara H, Nishina K, Yoshida K et al.
Efficient in vivo delivery of siRNA into brain
capillary endothelial cells along with
endogenous lipoprotein. Mol. Ther. 19(12),
2213–2221 (2011).
89 Uno Y, Piao W, Miyata K, Nishina K,
Mizusawa H, Yokota T. High-density
lipoprotein facilitates in vivo delivery of
a-tocopherol-conjugated short-interfering
RNA to the brain. Hum. Gene Ther. 22(6 ),
711–719 (2011).
90 Lin Q, Chen J, Jin H et al . Efficient systemic
delivery of siRNA by using high-density
lipoprotein-mimicking peptide lipid
nanoparticles. Nanomedicine (Lond.) 7(12),
1813 –1825 (2 012).
91 Mcconathy WJ, Nair MP, Paranjape S,
Mooberry L, Lacko AG. Evaluation of
Nanomedicine (20 14) 9(1)
120 future science group
Review Lin, Chen, Zhang & Zheng
synthetic/reconstituted high-density
lipoproteins as delivery vehicles for paclitaxel.
Anticancer Drugs 19(2), 183 –188 (2 008).
92 Kader A, Pater A. Loading anticancer drugs
into HDL as well as LDL has little affect on
properties of complexes and enhances
cytotox icity to human ca rcinoma cells.
J. Control . Release 80(1–3), 29–44 (2002).
93 Yang M, Chen J, Cao W et al. At tenuation of
nontargeted cell-kill using a high-density
lipoprotein-mimicking peptide –phospholipid
nanoscaffold. Nanomedicine (Lond.) 6(4),
631–641 (2011).
94 Yang M, Jin H, Chen J et al. Efficient cytosolic
delivery of siRNA using HDL-mimicking
nanoparticles. Small 7(5), 568–573 (2011).
95 Lin Q, Chen J, Ng KK, Cao W, Zhang Z,
Zheng G. Imaging the cytosolic drug delivery
mechanism of HDL-like nanoparticles.
Pharm. Res. doi:10.1007/s11095 -013-1046- z
(2013) (Epub ahead of print).
96 Ding Y, Wang W, Feng M et al. A
biomimetic nanovector-mediated targeted
cholesterol-conjugated siRNA deliver y for
tumor gene therapy. Biomaterials 33(34),
8893–8905 (2012).
97 Shahzad MM, Mangala LS, Han HD et al .
Targeted delivery of small interfering RNA
using reconstituted high-density lipoprotein
nanoparticles. Neoplasia 13(4), 309–319
(2 011).
98 Rui M, Tang H, Li Y, Wei X, Xu Y.
Recombinant high density lipoprotein
nanoparticles for target-specific deliver y of
siRNA. Pharm. Res. 30(5), 1203–1214
(2013).
99 Kim SI, Shin D, Choi TH et al . Systemic and
specific delivery of small interfering RNAs to
the liver mediated by apolipoprotein A-I. Mol.
Ther. 15(6 ), 1145 1152 (2 007).
100 Nakayama T, Butler JS, Sehgal A et al.
Harnessing a physiologic mechanism for
siRNA delivery with mimetic lipoprotein
particles. Mol. Ther. 20(8), 1582–1589
(2012).
101 Kong WH, Park K, Lee MY, Lee H, Sung
DK, Hahn SK. Cationic solid lipid
nanoparticles derived from apolipoprotein-
free LDLs for target specific systemic
treatment of liver fibrosis. Biomaterials 34(2),
542–551 (2013).
102 Akinc A, Goldberg M, Qin J et al.
Development of lipidoid-siRNA formulations
for systemic delivery to the liver. Mol. Ther.
17(5), 872–879 (2009).
103 Love KT, Mahon KP, Levins CG et al.
Lipid-like materials for low-dose, in vivo gene
silencing. Proc. Natl Acad. Sci. USA 107(5),
1864–1869 (2010).
 Good example of low-dose RNAi.
104 Huang YH, Bao Y, Peng W et al. Claudin-3
gene silencing with siRNA suppresses ovarian
tumor grow th and metastasis. Proc. Natl
Acad. Sci. USA 106(9), 3426–3430 (2009).
105 Nguyen DN, Mahon KP, Chikh G et al .
Lipid-derived nanoparticles for
immunostimulatory RNA adjuvant delivery.
Proc. Natl Acad. Sci . USA 10 9(14),
E797–E803 (2012).
106 Kleinman ME, Yamada K, Takeda A et al.
Sequence- and target-independent
angiogenesis suppression by siR NA via TLR3.
Nature 452(7187), 591–597 (2008).
107 Judge A, Maclachlan I. Overcoming the
innate immune response to small interfering
RNA. Hum. Gene Ther. 19(2), 111–12 4
(2008).
108 Robbins M, Judge A, Maclachlan I. siRNA
and innate immunity. Oligonucleotides 19 (2 ),
89–102 (2009).
109 Nguyen DN, Chen SC, Lu J et al. Drug
delivery-mediated control of RNA
immunostimulation. Mol. Ther. 17( 9),
1555 –156 2 (2 009).
110 Dolina JS, Sung SS, Novobrantseva TI,
Nguyen TM, Hahn YS. Lipidoid
nanoparticles containing PD-L1 siRNA
delivered in vivo enter Kupffer cells and
enhance NK and CD8+ T cell-mediated
hepatic antiviral immunity. Mol. Ther.
Nucleic Acids 2, e72 (2013).
111 Sahay G, Querbes W, Alabi C et al. Efficiency
of siRNA delivery by lipid nanoparticles is
limited by endocytic recycling. Nat.
Biotechnol. 31(7), 653– 658 (2013).
112 Landen CN Jr, Chavez-Reyes A,
Bucana C et al. Therapeutic EphA2 gene
targeting in vivo using neutra l liposomal small
interfering RNA delivery. Cancer Res. 65(15),
6910 6918 (20 05).
113 Halder J, K amat A A, Landen CN Jr et al .
Focal adhesion kinase targeting using in vivo
short interfering R NA delivery in neutral
liposomes for ovaria n carcinoma therapy.
Clin. Cancer Res. 12(16 ), 4916 4924 (20 06).
114 Verma A, Guha S, Diagaradjane P et al.
Therapeutic significance of elevated tissue
transglutaminase expression in pancreatic
cancer. Clin. Cancer Res. 14(8), 2476–2483
(2008).
115 Villares GJ, Zigler M, Wang H et al. Targeting
melanoma growth and metastasis with
systemic deliver y of liposome-incorporated
protease-activated receptor-1 small interfering
RNA. Cancer Res. 68(21), 9078–9086 (2008).
116 Mangala LS, Zuzel V, Schmandt R et al.
Therapeutic targeting of ATP7B in ovarian
carcinoma. Clin. Cancer Res. 15(11),
3770–3780 (2009).
117 Chak ravarty D, Roy SS, Babu CR et al .
Therapeutic targeting of PELP1 prevents
ovarian cancer growth and metastasis. Clin.
Cancer Res. 17(8), 2250 –2259 (2011).
118 Vivas-Mejia PE, Rodriguez-Ag uayo C, Han
HD et al. Silencing sur vivin splice variant 2B
leads to antitumor activity in taxane-resista nt
ovarian cancer. Clin. Cancer Res. 17(11),
3716–3726 (2011).
119 Koldehoff M, Steckel NK, Beelen DW,
Elmaagacli AH. Therapeutic application of
small interfering RNA directed against
bcr–abl transcripts to a patient with
imatinib-resistant chronic myeloid leukaemia.
Clin. Exp. Med. 7(2), 47–55 (2007).
120 Kapoor M, Burgess DJ. Efficient and safe
delivery of siRNA using a nionic lipids:
formulation optimization studies. Int.
J. Pharm. 432(1–2), 80 –90 (2012).
121 Mckee TD, Chen J, Corbin I, Zheng G,
Khokha R. Quantif ying nanoparticle
transport in vivo using hyperspectral imaging
with a dorsal skinfold window chamber.
J. Innovat. Opt. Health Sci. 5(4), (2012).
122 Zheng G, Chen J, Li H, Glickson JD.
Rerouting lipoprotein nanoparticles to selected
alternate receptors for the targeted delivery of
cancer diagnostic and therapeutic agents. Proc.
Natl Acad. Sci. USA 102 (49), 17757–1776 2
(2005).
Websites
201 Clinica lTrials.gov. Study With Atu027 in
Patients With Advanced Solid Cancer.
http://clinicaltrials.gov/ct2/show/
NCT00938574?term=At u027&rank=2
202 ClinicalTrials.gov. Atu027 Plus Gemcitabine
in Advanced or Metastatic Pancreatic Cancer
(Atu027-I-02).
http://clinicaltrials.gov/ct2/show/
NCT01808638?term=A tu027&rank=1
203 ClinicalTrials.gov. EphA2 Gene Targeting
Using Neutral Liposomal Sma ll Interfering
RNA De liver y.
http://clinicaltrials.gov/ct2/show/
NCT01591356?term=EphA2&rank=1
... 22,23 In addition, thirdgeneration cationic lipids have been exploited to complex oligonucleotides and transfect cells for gene therapy purposes with low toxicity and immunogenicity. 20,24,25 As a result of the combination of novel polymeric and lipidic materials, lipid− polymer hybrid nanoparticles (LPNs) have been proven to be an efficient nanoplatform for the coloading of drugs with different physicochemical properties. 26−29 Recently, we demonstrated that newly optimized LPNs were successfully coloaded with an anti-inflammatory drug in the polymeric core and a model siRNA in the lipid shell using a newly developed microfluidics approach. ...
Article
Full-text available
Musculoskeletal diseases involving tissue injury comprise tendon, ligament, and muscle injury. Recently, macro-phages have been identified as key players in the tendon repair process, but no therapeutic strategy involving dual drug delivery and gene delivery to macrophages has been developed for targeting the two main dysregulated aspects of macrophages in tendinop-athy, i.e., inflammation and fibrosis. Herein, the anti-inflammatory and antifibrotic effects of dual-loaded budesonide and serpine1 siRNA lipid−polymer hybrid nanoparticles (LPNs) are evaluated in murine and human macrophage cells. The modulation of the gene and protein expression of factors associated with inflammation and fibrosis in tendinopathy is demonstrated by real time polymerase chain reaction and Western blot. Macrophage polarization to the M2 phenotype and a decrease in the production of pro-inflammatory cytokines are confirmed in macrophage cell lines and primary cells. The increase in the activity of a matrix metalloproteinase involved in tissue remodelling is proven, and studies evaluating the interactions of LPNs with T cells proved that dual-loaded LPNs act specifically on macrophages and do not induce any collateral effects on T cells. Overall, these dual-loaded LPNs are a promising combinatorial therapeutic strategy with immunomodulatory and antifibrotic effects in dysregulated macrophages in the context of tendinopathy.
... [5][6][7][8]. Lower pH helps quick release of the cargo from endosomal compartment and protects mRNA from acid hydrolysis and lysosomal digestion in intracellular milieu [29]. Together, numbers of minute changes in the design context likely playing in concert and produced quick, balanced, stable Th1-IgG2-biased antibody response. ...
Article
D614G genotype of SARS-CoV-2 virus is highly infectious and responsible for almost all infection for 2nd wave. However, there are currently no reports with D614G as vaccine candidate. Here we report the development of an mRNA-LNP vaccine with D614G variant and characterization in animal model. We have used special mRNA-architecture and formulation that provides suitable response of the product. The surface plasmon resonance (SPR) data with spike protein (S) revealed that immunization generated specific antibody pools against the whole extracellular domain (RBD and S2) of the spike protein. The anti-sera and purified IgGs from immunized mice neutralized SARS-CoV-2-pseudoviruses in ACE2-expressing HEK293 cells in a dose dependent manner. Importantly, single-dose immunization protected mice-lungs from homotypic-pseudovirus entry and cytopathy. The immunologic responses have been implicated by a balanced and stable population of CD4+ cells with a Th1 bias. The data suggested great promise for immediate translation of the technology to the clinic.
... Identifier is NCT04592692). Lipid nanoparticles (LNPs) have been exploited as well-organized, biocompatible, and scalable carriers to transfer mRNA to hepatocytes, thereby providing increased stability of mRNA and a high amount of protein expression [109][110][111]. mRNA therapy has numerous advantages over gene therapy, such as the impossibility of integration mutations and the possibility of modifications that cause increased mRNA post-translational activity and stability [112][113][114]. ...
Article
Full-text available
Recent advancements have focused on enhancing factor VIII half-life and refining its delivery methods, despite the well-established knowledge that factor VIII deficiency is the main clotting protein lacking in hemophilia. Consequently, both viral and non-viral delivery systems play a crucial role in enhancing the quality of life for hemophilia patients. The utilization of viral vectors and the manipulation of non-viral vectors through targeted delivery are significant advancements in the field of cellular and molecular therapies for hemophilia. These developments contribute to the progression of treatment strategies and hold great promise for improving the overall well-being of individuals with hemophilia. This review study comprehensively explores the application of viral and non-viral vectors in cellular (specifically T cell) and molecular therapy approaches, such as RNA, monoclonal antibody (mAb), and CRISPR therapeutics, with the aim of addressing the challenges in hemophilia treatment. By examining these innovative strategies, the study aims to shed light on potential solutions to enhance the efficacy and outcomes of hemophilia therapy.
Article
Small interfering RNA (siRNA) has emerged as a valuable tool to address RNA interference (RNAi) to modulate gene expression also in therapy. However, challenges such as inefficient cell targeting and rapid degradation in biological systems have limited its success. To address these issues, the development of a receptor‐specific shuttle system represents a promising solution. [F ⁷ ,P ³⁴ ]‐NPY analogues were modified by solid‐phase peptide synthesis, enabling non‐covalent conjugation with siRNA. This modification yielded an efficient siRNA vehicle capable of binding and transporting its cargo into target cells without adversely affecting receptor activation or cell viability. Mass spectrometry and gel shift assays confirmed successful and stable siRNA binding under various conditions. Microscopy experiments further demonstrated the co‐internalization of labeled peptides and siRNA in Hepa1c1 cells, highlighting the stability of the complex. In vitro quantitative RT‐PCR experiments, targeting the TSC22D4 gene to normalize systemic glucose homeostasis and insulin resistance, revealed a functional peptide‐based siRNA shuttle system with the ability to decrease mRNA expression to approximately 40%. These findings strengthen the potential of receptor‐specific siRNA shuttle systems as efficient tools for gene therapy that offer a possibility for reducing side effects.
Article
Alzheimer's disease (AD) is a neurodegenerative condition that exhibits a gradual decline in cognitive function and is prevalent among a significant number of individuals globally. The use of small interfering RNA (siRNA) molecules in RNA interference (RNAi) presents a promising therapeutic strategy for AD. Lipid nanoparticles (LNPs) have been developed as a delivery vehicle for siRNA, which can selectively suppress target genes, by enhancing cellular uptake and safeguarding siRNA from degradation. Numerous research studies have exhibited the effectiveness of LNP‐mediated siRNA delivery in reducing amyloid beta (Aβ) levels and enhancing cognitive function in animal models of AD. The feasibility of employing LNP‐mediated siRNA delivery as a therapeutic approach for AD is emphasized by the encouraging outcomes reported in clinical studies for other medical conditions. The use of LNP‐mediated siRNA delivery has emerged as a promising strategy to slow down or even reverse the progression of AD by targeting the synthesis of tau phosphorylation and other genes linked to the condition. Improvement of the delivery mechanism and determination of the most suitable siRNA targets are crucial for the efficacious management of AD. This review focuses on the delivery of siRNA through LNPs as a promising therapeutic strategy for AD, based on the available literature.
Article
Cancer remains a major global health challenge, with increasing incidence and mortality rates projected for the coming years. Lung cancer, in particular, poses significant obstacles due to late-stage diagnosis and limited treatment options. While advancements in molecular diagnostics have been made, there is a critical need to connect the dots between laboratory and hospital for better lung cancer treatment. Systemic therapy plays a crucial role in treating advanced-stage lung cancer, and recent efforts have focused on developing innovative drug delivery techniques. Nanoparticles (NPs) have emerged as a promising approach to lung cancer treatment, offering enhanced drug delivery, active targeting, and reduced toxicity. Organic-based nanomaterials, like polymeric nanoparticles, solid lipid nanoparticles, and liposomes hold great potential in this field. This review examines the application of NPs in lung cancer treatment, highlights current therapies, explores organic nanoparticle-based approaches, and discusses limitations and future perspectives in clinical translation.
Article
Five small interfering RNA (siRNA)-based therapeutics have been approved by the Food and Drug Administration (FDA), namely patisiran, givosiran, lumasiran, inclisiran, and vutrisiran. Besides, siRNA delivery to the target site without toxicity is a big challenge for researchers, and naked-siRNA delivery possesses several challenges, including membrane impermeability, enzymatic degradation, mononuclear phagocyte system (MPS) entrapment, fast renal excretion, endosomal escape, and off-target effects. The siRNA therapeutics can silence any disease-specific gene, but their intracellular and extracellular barriers limit their clinical applications. For this purpose, several modifications have been employed to siRNA for better transfection efficiency. Still, there is a quest for better delivery systems for siRNA delivery to the target site. In recent years, nanoparticles have shown promising results in siRNA delivery with minimum toxicity and off-target effects. Patisiran is a lipid nanoparticle (LNP)-based siRNA formulation for treating hereditary transthyretin-mediated amyloidosis that ultimately warrants the use of nanoparticles from different classes, especially lipid-based nanoparticles. These nanoparticles may belong to different categories, including lipid-based, polymer-based, and inorganic nanoparticles. This review briefly discusses the lipid, polymer, and inorganic nanoparticles and their sub-types for siRNA delivery. Finally, several clinical trials related to siRNA therapeutics are addressed, followed by the future prospects and conclusions.
Article
Full-text available
Lipid nanovehicles are currently the most advanced vehicles used for RNA delivery, as demonstrated by the approval of patisiran for amyloidosis therapy in 2018. To illuminate the unique superiority of lipid nanovehicles in RNA delivery, in this review, we first introduce various RNA therapeutics, describe systemic delivery barriers, and explain the lipid components and methods used for lipid nanovehicle preparation. Then, we emphasize crucial advances in lipid nanovehicle design for overcoming barriers to systemic RNA delivery. Finally, the current status and challenges of lipid nanovehicle-based RNA therapeutics in clinical applications are also discussed. Our objective is to provide a comprehensive overview showing how to utilize lipid nanovehicles to overcome multiple barriers to systemic RNA delivery, inspiring the development of more high-performance RNA lipid nanovesicles in the future.
Article
Full-text available
We have developed a noninvasive imaging method to quantify in vivo drug delivery pharmacokinetics without the need for blood or tissue collection to determine drug concentration. By combining the techniques of hyperspectral imaging and a dorsal skinfold window chamber, this method enabled the real-time monitoring of vascular transport and tissue deposition of nanoparticles labeled with near-infrared (NIR) dye. Using this imaging method, we quantified the delivery pharmacokinetics of the native high-density lipoprotein (HDL) and epidermal growth factor receptor (EGFR)-targeted HDL nanoparticles and demonstrated these HDLs had long circulation time in blood stream (half-life >12 h). These HDL nanoparticles could efficiently carry cargo DiR-BOA to extravasate from blood vessels, diffuse through extracellular matrix, and penetrate and be retained in the tumor site. The EGFR targeting specificity of EGFR-targeted HDL (EGFR-specific peptide conjugated HDL) was also visualized in vivo by competitive inhibition with excess EGFR-specific peptide. In summary, this imaging technology may help point the way toward the development of novel imaging-based pharmacokinetic assays for preclinical drugs and evaluation of drug delivery efficiency, providing a dynamic window into the development and application of novel drug delivery systems.
Article
Full-text available
Delivery of short interfering RNAs (siRNAs) remains a key challenge in the development of RNA interference (RNAi) therapeutics. A better understanding of the mechanisms of siRNA cellular uptake, intracellular transport and endosomal release could critically contribute to the improvement of delivery methods. Here we monitored the uptake of lipid nanoparticles (LNPs) loaded with traceable siRNAs in different cell types in vitro and in mouse liver by quantitative fluorescence imaging and electron microscopy. We found that LNPs enter cells by both constitutive and inducible pathways in a cell type-specific manner using clathrin-mediated endocytosis as well as macropinocytosis. By directly detecting colloidal-gold particles conjugated to siRNAs, we estimated that escape of siRNAs from endosomes into the cytosol occurs at low efficiency (1-2%) and only during a limited window of time when the LNPs reside in a specific compartment sharing early and late endosomal characteristics. Our results provide insights into LNP-mediated siRNA delivery that can guide development of the next generation of delivery systems for RNAi therapeutics.
Article
Full-text available
Despite efforts to understand the interactions between nanoparticles and cells, the cellular processes that determine the efficiency of intracellular drug delivery remain unclear. Here we examine cellular uptake of short interfering RNA (siRNA) delivered in lipid nanoparticles (LNPs) using cellular trafficking probes in combination with automated high-throughput confocal microscopy. We also employed defined perturbations of cellular pathways paired with systems biology approaches to uncover protein-protein and protein-small molecule interactions. We show that multiple cell signaling effectors are required for initial cellular entry of LNPs through macropinocytosis, including proton pumps, mTOR and cathepsins. siRNA delivery is substantially reduced as ≅70% of the internalized siRNA undergoes exocytosis through egress of LNPs from late endosomes/lysosomes. Niemann-Pick type C1 (NPC1) is shown to be an important regulator of the major recycling pathways of LNP-delivered siRNAs. NPC1-deficient cells show enhanced cellular retention of LNPs inside late endosomes and lysosomes, and increased gene silencing of the target gene. Our data suggest that siRNA delivery efficiency might be improved by designing delivery vehicles that can escape the recycling pathways.
Article
Full-text available
CD47 is a "self marker" that is usually overexpressed on the surface of cancer cells to enable them to escape immunosurveillance. Recognition of CD47 by its receptor, signal regulatory protein α (SIRPα), which is expressed in the macrophages, inhibits phagocytic destruction of cancer cells by the macrophages. In this study, we have first shown that clinical isolates of human melanoma significantly upregulate CD47, possibly as a mechanism to defend themselves against the macrophages. We then exploited RNA interference (RNAi) technology to test the hypothesis that knocking down CD47 in the tumor cells will render them targets for macrophage destruction; hence, creating a novel anti-cancer therapy. Anti-CD47 siRNA was encapsulated in a liposome-protamine-hyaluronic acid (LPH) nanoparticle (NP) formulation to address the challenge of targeted delivery of siRNA-based therapeutics in vivo. Efficient silencing of CD47 in tumor tissues with systemic administration of LPH(CD47) also significantly inhibited the growth of melanoma tumors. In a lung metastasis model, LPH(CD47) efficiently inhibited lung metastasis to about 27% of the untreated control. Moreover, no hematopoietic toxicity was observed in the animals that received multiple doses of LPH(CD47). Our findings indicate CD47 as a potential prognostic marker for melanoma development as well as a target for therapeutic intervention with RNAi-based nanomedicines.Molecular Therapy (2013); doi:10.1038/mt.2013.135.
Article
Full-text available
Small interfering RNA silences specific genes by interfering with mRNA translation, and acts to modulate or inhibit specific biological pathways; a therapy that holds great promise in the cure of many diseases. However, the naked small interfering RNA is susceptible to degradation by plasma and tissue nucleases and due to its negative charge unable to cross the cell membrane. Here we report a new polymer carrier designed to mimic the influenza virus escape mechanism from the endosome, followed by a timed release of the small interfering RNA in the cytosol through a self-catalyzed polymer degradation process. Our polymer changes to a negatively charged and non-toxic polymer after the release of small interfering RNA, presenting potential for multiple repeat doses and long-term treatment of diseases.
Article
Full-text available
Purpose: Molecular therapeutics often require an effective nanoparticle-based delivery strategy to transport them to cytosolic organelles to be functional. Recently, a cytosolic delivery strategy based on the scavenger receptor class B type I (SR-BI) mediated pathway has shown great potential for the effective delivery of theranostics agents into the cytoplasm of cells without detrimental endosomal entrapment. This study elucidates this unique delivery mechanism for improving cytosolic drug delivery. Methods: Multifluorophore-labeled HDL-mimicking peptide phospholipid scaffold (HPPS) nanoparticles were developed. Fluorescence imaging was utilized to examine HPPS transporting payloads into cells step by step through sequential inhibition studies. Results: HPPS specifically recognizes and binds to SR-BI, then interacts with SR-BI, which results in direct transport of payload molecules into the cell cytoplasm without entire particles internalization. The cytosolic transport of payloads occurred through a temperature- and energy-independent pathway, and was also different from actin- and clathrin-mediated endocytosis. Furthermore, this transport was significantly inhibited by disruption of lipid rafts using filipin or methyl-β-cyclodextrin. Conclusions: The cytosolic delivery of payloads by HPPS via SR-BI targeting is predominately mediated through a lipid rafts/caveolae-like pathway. This cytosolic delivery strategy can be utilized for transporting molecular therapeutics that require their action sites to be within cytosolic organelles to enhance therapeutic effect.
Article
Full-text available
Small interfering RNA (siRNA) would be predicted to function as a cancer drug, but an efficient siRNA delivery system is required for clinical development. To address this issue, we developed a liposomal siRNA carrier, a multifunctional envelope-type nanodevice (MEND). We previously reported that a MEND composed of a pH-sensitive cationic lipid, YSK05, showed significant knockdown in both in vitro and in tumor tissue by intratumoral injection. Here, we report on the development of an in vivo siRNA delivery system that is delivered by systemic injection and an analysis of the pharmacokinetics of an intravenously administered siRNA molecule in tumor tissue. Tumor delivery of siRNA was quantified by means of stem-loop primer quantitative reverse transcriptase PCR (qRT-PCR) method. PEGylation of the YSK-MEND results in the increase in the accumulation of siRNA in tumor tissue from 0.0079% ID/g tumor to 1.9% ID/g tumor. The Administration of the MEND (3 mg siRNA/kg body weight) showed about a 50% reduction in the target gene mRNA and protein. Moreover, we verified the induction of RNA interference by 5' RACE-PCR method. The collective results reported here indicate that an siRNA carrier was developed that can deliver siRNA to a target cell in tumor tissue through an improved siRNA bioavailability.Molecular Therapy (2013); doi:10.1038/mt.2013.57.
Article
Nature Reviews Drug Discovery 8, 129–138 (2009) | doi:10.1038/nrd2742 On page 136, in the glossary term and in the main text, age-related macular degeneration (AMD) has been incorrectly attributed to “excessive growth and rupture of blood vessels within the cornea”. The correct version should state that this occurs within the retina.
Article
The design and synthesis of new light-activated contrast agents for theranostics (therapy/diagnosis) has the potential to facilitate multifunctional and improved personalized medicine. The use of light as a remote activation strategy provides spatial and temporal control of drug effect and nanotechnology can play a key role in this process. Lipoproteins (LDL and HDL), which transport water-insoluble cholesteryl esters and triacylglycerols in nature, have evolved to efficiently ferry exogenous hydrophobic compounds in vivo. They are naturally biocompatible, maneuverable due to their small size (
Article
Keywords: Multifunctional envelope-type nano device (MEND) siRNA delivery pH-sensitive cationic lipid Intracellular trafficking Endosomal escape Modification of liposomal siRNA carriers with polyethylene glycol, i.e., PEGylation, is a generally accepted strategy for achieving in vivo stability and delivery to tumor tissue. However, PEGylation significantly inhibits both cellular uptake and the endosomal escape process of the carriers. In a previous study, we reported on the development of a multifunctional envelope-type nano device (MEND) for siRNA delivery and peptide-based functional devices for overcoming the limitations and succeeded in the efficient delivery of siRNA to tumors. In this study, we synthesized a pH-sensitive cationic lipid, YSK05, to overcome the limitations. The YSK05-MEND had a higher ability for endosomal escape than other MENDs containing conventional cationic lipids. The PEGylated YSK05-MEND induced efficient gene silencing and overcame the limitations followed by optimization of the lipid composition. Furthermore, the intratumoral administration of the YSK05-MEND resulted in a more efficient gene silencing compared with MENDs containing conventional cationic lipids. Collectively, these data confirm that YSK05 facilitates the endosomal escape of the MEND and thereby enhances the efficacy of siRNA delivery into cytosol and gene silencing.