ArticlePDF AvailableLiterature Review

siRNA therapeutics: a clinical reality

Authors:

Abstract and Figures

Since the revolutionary discovery of RNA interference (RNAi), a remarkable progress has been achieved in understanding and harnessing gene silencing mechanism; especially in small interfering RNA (siRNA) therapeutics. Despite its tremendous potential benefits, major challenges in most siRNA therapeutics remains unchanged—safe, efficient and target oriented delivery of siRNA. Twenty years after the discovery of RNAi, siRNA therapeutics finally charts its way into clinics. As we journey through the decades, we reminisce the history of siRNA discovery and its application in a myriad of disease treatments. Herein, we highlight the breakthroughs in siRNA therapeutics, with special feature on the first FDA approved RNAi therapeutics Onpattro (Patisiran) and the consideration of effective siRNA delivery system focusing on current siRNA nanocarrier in clinical trials. Lastly, we present some challenges and multiple barriers that are yet to be fully overcome in siRNA therapeutics.
Content may be subject to copyright.
SPECIAL TOPIC: Noncoding RNA: from dark matter to bright star April 2020 Vol.63 No.4: 485–500
REVIEWhttps://doi.org/10.1007/s11427-018-9438-y
siRNA therapeutics: a clinical reality
Phei Er Saw1& Er-Wei Song1,2,3*
1Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen
University, Guangzhou 510120, China;
2Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China;
3Zhongshan School of Medicine, Breast Surgery, Guangzhou 510080, China
Received September 20, 2018; accepted October 14, 2018; published online April 30, 2019
Since the revolutionary discovery of RNA interference (RNAi), a remarkable progress has been achieved in understanding and
harnessing gene silencing mechanism; especially in small interfering RNA (siRNA) therapeutics. Despite its tremendous
potential benefits, major challenges in most siRNA therapeutics remains unchanged—safe, efficient and target oriented delivery
of siRNA. Twenty years after the discovery of RNAi, siRNA therapeutics finally charts its way into clinics. As we journey
through the decades, we reminisce the history of siRNA discovery and its application in a myriad of disease treatments. Herein,
we highlight the breakthroughs in siRNA therapeutics, with special feature on the first FDA approved RNAi therapeutics
Onpattro (Patisiran) and the consideration of effective siRNA delivery system focusing on current siRNA nanocarrier in clinical
trials. Lastly, we present some challenges and multiple barriers that are yet to be fully overcome in siRNA therapeutics.
siRNA therapeutic, disease treatment, systemic siRNA delivery, clinical application
Citation: Saw, P.E., and Song, E.W. (2020). siRNA therapeutics: a clinical reality. Sci China Life Sci 63, 485–500. https://doi.org/10.1007/s11427-018-9438-y
In 1998, Fire et al. (1998) uncovered the mechanism of
RNAi and have since revolutionized the understanding of
gene regulation when they discovered that the silencing ef-
fectors in Caenorhabditis elegans were double stranded
RNAs. By using an RNA sequence that could interfere with
muscular functions, they treated C. elegans with either sin-
gle-stranded RNA (ssRNA) or double-stranded RNA
(dsRNA) and consistently found that ssRNAs were 10- to
100-fold less effective than dsRNA in silencing the same
mRNA. They observed that the occurrence of effective
knockdown led to phenotypical muscle twitching of C. ele-
gans that was observable. Importantly, the only way to in-
crease the effectiveness of ssRNA was to co-inject the
ssRNAs with the antisense strand, therefore suggesting that
ssRNA hybridization into dsRNA is a prerequisite for gene
silencing (Figure 1) (Sen and Blau, 2006).
siRNA: the “FIRST” of interventions
In mammalian cells. In 2001, Elbashir et al. (2001) first
demonstrated the RNAi mechanism in mammalian cell lines.
They used siRNA to specifically silence the expression of
different genes: siRNA against reporter genes coding for sea
pansy (Renilla reniformis, RL) and two sequence variants of
firefly (Photinus pyralis, GL2 and GL3) luciferases. After
transfection with the siRNAs, there was a significant re-
duction in gene expression level of all the silenced genes, and
the silencing effects were directly proportional to the level of
their endogenous gene expression. In animal models. In
2003, we demonstrated that siRNA targeting Fas protected
mice from fulminant hepatitis; and this work became the first
proof-of-principle that siRNAs could be utilized for the
treatment of a disease in an in vivo model. We first injected
Fas siRNAs into mice before sending Fas into hyperdrive to
mimic the liver failure disease model by hepatitis infections.
© Science China Press and Springer-Verlag GmbH Germany, part of Springer Nature 2019 life.scichina.com link.springer.com
SCIENCE CHINA
Life Sciences
*Corresponding author (email: songew@mail.sysu.edu.cn)
All mice without siRNA intervention died of acute liver
failure within a few days, compared to an 82% survival rate
in siRNA-treated mice. Among these mice that survived,
their liver cells incorporated 80%–90% of the injected siR-
NAs (Song et al., 2003). The New England Journal of
Medicine (NEJM) highlighted this work and stated that “the
ability of Fas-specific siRNA to temper the destructive role
of Fas could find widespread clinical application in acute and
chronic liver diseases” (Davidson, 2003). Subsequently,
Hamar et al. (2004) continued to demonstrate the feasibility
of delivering Fas-specific siRNA in protecting mice against
renal ischemic reperfusion injury, verifying the effectiveness
of siRNA therapeutics in the treatment of a myriad of human
diseases (Lieberman et al., 2003). In non-human primates. In
2006, by encapsulating ApoB siRNAs in SNALP for-
mulation and intravenously delivered into non-human
primates, Zimmerman and colleagues showed that apoli-
poprotein B (ApoB) could be specifically silenced with
>90% silencing of ApoB mRNA in the liver even after 48-
h post-administration. Herein, they also proved the spe-
cificity of ApoB siRNA as the cleavage of apoB mRNA
occurred precisely on the site predicted for the RNAi
mechanism. Moreover, within 24 h after treatment, the
level of ApoB protein, serum cholesterol and low-density
lipoprotein levels were significantly reduced. Most im-
portantly, this effect is long-lasting with the highest dose
of siRNA treatment lasted for 11 d; demonstrating a potent
and lasting biological effect of siRNA treatment modality
(Zimmermann et al., 2006). In human. The first in-human
evidence of siRNA silencing was published by Davis and
colleagues in 2011. This data was obtained in conjunction
with the Phase 1 CALAA-1clinical trial (NCT00689065).
CALAA-1 is a nanoparticle delivery system carrying M2
subunit of ribonucleotide reductase (RRM2) siRNA and
was injected systematically into patients of solid cancers
via intravenous injection. Tumor biopsies from patients
obtained after the completion of treatment revealed two
major observations: (i) the delivery carrier was found
abundantly localized in the cytoplasm of cancer cells, and
these amounts correlate directly with the dose levels of
administered nanoparticles and (ii) significant reduction in
both RRM2 mRNA and protein level were detected in
patients. Similar with Zimmermann, they also detected
that the specific mRNA cleavage site corresponded to the
site of siRNA-specific mediated mRNA cleavage (Davis,
2009). In clinics. In August 2018, the Food and Drug
Administration (FDA) has approved the first ever RNA
interference drug–Alnylam’s Onpattro (Patisiran) for the
nerve damage caused by a rare disease hereditary trans-
thyretin-mediated amyloidosis (hATTR) in adults after the
encouraging result in the Phase 3 clinical trial, “Apollo”
(NCT01960348). In the largest cohort study of hATTR
amyloidosis, treatment with Onpattro improved poly-
neuropathy in a majority of patients by reversal of neu-
ropathy impairment of neuropathy impairment, showing a
significant improvement in the Quality of Life (QOL) of
patients as well (Adams et al., 2018).
siRNA: the REAL breakthrough
To circumvent the barriers in systemic siRNA delivery, non-
invasive delivery strategies for directing siRNAs specifically
to target cells needed to be devised (Dykxhoorn and Lie-
berman, 2005;Shankar et al., 2005). Therefore, we designed
a protamine-antibody fusion protein that could deliver siR-
NA specifically to HIV-infected or envelope-transfected
cells by conveniently mixing siRNA with the fusion protein
via electrostatic charge interactions (Song et al., 2005).
NEJM gave high commendations for this study as it pos-
sessed high targeted gene silencing efficacy in vivo despite
its beauty in simplicity (Williams, 2005) and pronounced this
as the magic bullet that fully demonstrated the potential for
successful systemic siRNA delivery. Subsequently, Morris-
sey et al. (2005a) developed a stable nucleic acid-lipid par-
ticle (famously known as SNALPs). As a strategy for siRNA
encapsulation and a delivery vehicle, they demonstrated that
SNALPs could protect and enhance the half-life of siRNAs
in circulation, while effectively reducing hepatitis B re-
plicons specifically in mice liver. Similar results were ob-
served when Zimmermann et al. (2006) demonstrated the
first gene silencing mechanism in non-human primates by
using ApoB-specific siRNA encapsulated in SNALPs to
downregulate ApoB protein, resulting in significant reduc-
tion of ApoB protein, serum cholesterol and low-density li-
poprotein levels.
Taken together, we and many others have proven that
Figure 1 Single stranded (sense or antisense) RNA or double-stranded
RNA encoding for a muscle protein was injected into C. elegans. Both
single-stranded RNA had no effect when injected separately, but started to
twitch when treated with double-stranded RNA, showing similar pheno-
types to C. elegans with defective muscle proteins. This indication denoted
that the gene coding for normal muscle movement had been silenced by the
dsRNA.
486 Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
siRNA therapeutics is indeed potent and effective against
various human diseases. Most importantly, our publications
also resulted in the intellectual property licensing of a potent
in vivo siRNA delivery system to Alnylam- the company that
owns Onpattro and several other siRNA therapeutics pipe-
line in clinical trials, therefore serving as a bridge between
academic and translational research. With on-going colla-
borations between our institution and Harvard Medical
School, we are creating a global avenue for international
clinical siRNA research that could accelerate bench-to-bed-
side transition and thus benefiting more patients in gene si-
lencing therapeutics. The timeline of RNAi discovery and
the progress of siRNA in clinical application is summarized
in Figure 2.
siRNA: the mechanism
siRNAs are short synthetic RNA duplexes commonly con-
sisted of two 21-mer oligonucleotides strand with 19 nu-
cleotides (nt) of complementary bases and a 2-nt overhang at
each 3′-end (de Fougerolles et al., 2005). Once in cell cy-
toplasm, this siRNA molecule will be incorporated into a
RNA-induced silencing complex (RISC), a nuclease-con-
taining multi-protein complex (Hammond et al., 2000). The
siRNA duplexes will be separated within the RISC complex.
The strand with the more stable 5′-end is integrated to the
active RISC complex. This siRNA then guides the RISC
complex to seek and cleave through the action of catalytic
RISC protein, a member of the argonaute family (Ago2).
(Figure 3) (Nair et al., 2014;Ryther et al., 2005).
siRNA: when silence is golden
RNAi therapeutics represent a paradigm shift in treating
human disease. The ability of siRNA to target and silence
virtually any gene of interest provides powerful new tools for
biomedical research and drug discovery (de Fougerolles et
al., 2007). In the development of small molecule drugs, many
oncogenic targets (i.e., RAS and MYC) fall into “un-
druggable” category since there is no “active binding site”
amenable by conventional small drug-like molecules (Sou-
cek and Evan, 2010). Therefore, the development of small
molecule inhibitors targeting these intractable proteins posed
major challenges (Dang et al., 2017;Soucek and Evan,
2010). The use of siRNA therapeutics could turn the tables
around for many devastating diseases, as siRNA could easily
be designed and the identification of a potent siRNA could
be done within weeks as compared to years for a drug de-
velopment (Petrocca and Lieberman, 2011). Besides, potent
siRNA sequences are usually active at extremely low (pi-
comolar) concentrations; and this could be designed for any
gene of interest with appropriate tools (Petrocca and Lie-
berman, 2011). Notably in cancer treatment, siRNA ther-
apeutics promise negligible side effects as compared to
conventional chemotherapeutics. Many have tried to silence
k-RAS or c-MYC siRNA in mouse models, hoping to de-
velop RNAi-based therapeutics by targeting these “un-
druggable” oncogenes (Bäumer et al., 2015;Wang et al.,
2005). By addressing targets that are otherwise untreatable
with existing medicines, RNAi not only promises an efficient
therapeutic modality, but also diminish the possibility of
drug resistance, a side effect so often crippled a che-
Figure 2 Timeline of RNAi discovery and the progress of siRNA in clinical application.
487
Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
motherapeutic regime. Table 1 below summarized the de-
tailed comparison between siRNA therapeutics and small
molecule drugs.
siRNA success story: Patisiran (Onpattro)
On August 10, 2018, the world is euphoric at the ground-
breaking announcement of the FDA approval of Patisiran
(Onpattro) or known as ALN-TTR02 for the treatment of
Hereditary Transthyretin Amyloidosis (hATTR). hATTR,
also referred to as transthyretin familial amyloid poly-
neuropathy (TTR-FAP) or familial amyloid cardiomyopathy
(TTR-FAC), is a rare, progressive, and fatal disease (Ando et
al., 2013;Conceição et al., 2016;Gertz, 2017). Unlike non-
hereditary, wild-type ATTR amyloidosis which primarily
affects only the heart, hATTR amyloidosis affects multiple
organs, such as the heart, nervous system, gastrointestinal
tract, and kidney, leading to various complications (Ando et
al., 2013;Coelho et al., 2013b;Hawkins et al., 2015). hATTR
amyloidosis is characterized by the extracellular deposition
of misfolded transthyretin (TTR) protein. In a normal folding
condition, TTR tetramer is made up of 4 single-chain
monomers. A mutation in TTR gene would destabilize the
tetramer and causes its dissociation into monomers, which
would then aggregate into amyloid fibrils and accumulate in
multiple organs throughout the body causing fatality (Figure
4) (Ando et al., 2013;Gertz, 2017;Johnson et al., 2012).
Since this TTR gene mutation occurs in the liver, silencing
the TTR gene promises to reverse the mutation effect of TTR
gene, therefore resolving the disease.
Earlier this year, Alnylam first presented the APOLLO
Phase 3 study of Onpattro (NCT01960348) at the 16th In-
ternational Symposium on Amyloidosis. These results were
later published in NEJM in July 2018, a month shy of its
FDA approval (Adams et al., 2018). In a series of clinical
assessment, Alnylam presented 87.8% mean serum TTR
knockdown for over 18 months, improvement in all primary
and secondary endpoints, met all exploratory endpoints in
cardiac subpopulations, 50% reduction in all-cause hospita-
lization and mortality, and 45% reduction in cardiac hospi-
talization and all-cause mortality. With these promising
results, Alnylam concluded that Patisiran is capable of sig-
nificantly improving multiple clinical manifestations of
hereditary transthyretin amyloidosis (Adams et al., 2018).
While the approval was historic, the fact remains that the
treatment is expensive as the drug is currently priced at
$450,000. Considering that siRNA treatment is transient and
that repetitive treatment might be needed, this could turn out
Figure 3 Schematic representation of siRNA silencing mechanism.
Table 1 Comparison between siRNA therapeutics and small molecule drug. Adapted from Bumcrot et al., 2006;Lam et al., 2015;Vaishnaw et al., 2010
siRNA Small molecule drug
Specificity High, sequence-driven Low-medium, conformation driven
Potency Typically pM Varies
Number of accessible targets >> 1000 500–1000
Nature of action Inhibition of target Inhibition/activation of target
Lead optimization Rapid, 4 to 8 weeks Slow, 2 to 4 years
Selectivity High Variable
Manufacturing Common, rapid, scalable Varies, can be complex
Serum stability Low (half-life of minutes) High (half-life of hours)
Toxicity Low High
Delivery Difficult Easy
Serum stability Stable Unstable
488 Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
to be a massive burden to patients. According to Alnylam’s
chief executive officer John Maraganore, this price-tag is no
surprise as the drug approval is the culmination of 16 years
of research and the investment of almost $2.5 billion. On top
of that, currently there are only about 3000 diagnosed
hATTR patients in US, thus the drug will not likely bring
much profit margin to Alnylam. Despite being the first RNAi
therapeutic approved, Patisiran will not be the only drug
available for hATTR. In July 2018, Tegsedi (Inotersen) a 2′-
O-methoxyethyl-modified antisense oligonucleotide devel-
oped by Akcea. Therapeutics was approved in Europe and is
currently being reviewed by the FDA while Vyndaqel (Ta-
famidis), also a small molecule drug owned by Pfizer re-
ceived Breakthrough Therapy designation from the FDA for
the treatment of patients with transthyretin cardiomyopathy.
With only an estimated 50,000 cases of hATTR worldwide,
some would argue that the approval of Onpattro is only a
drop in the ocean as compared to the drugs targeting major
diseases such as cancer. According to a statistic given by the
National Cancer Institute, in 2018 alone, an estimated
1,735,350 new cases of cancer will be diagnosed and
609,640 people will die from the disease in the United States.
Nevertheless, this is definitely a milestone in RNAi research
and we now see a glimpse of hope that could pave the way
for more RNAi therapeutics to be approved in the near fu-
ture. In an excerpt from FDA Commissioner Scott Gottlieb,
M.D., on the approval of Onpattro: “This approval is part of a
broader wave of advances that allow us to treat disease by
actually targeting the root cause, enabling us to arrest or
reverse a condition, rather than only being able to slow its
progression or treat its symptoms. In this case, the effects of
the disease cause a degeneration of the nerves, which can
manifest in pain, weakness and loss of mobility.”
siRNA: route of administration
The biggest hurdle in achieving potent siRNA therapeutics
did not lie on the design of potent siRNA, but rather on how
to administer biologically active siRNAs to the target tissue.
Various siRNA delivery strategies are being developed and
examined; and had been reviewed elsewhere (de Fougerolles
et al., 2007;de Fougerolles, 2008). Although there are var-
ious routes to administer siRNA, they are generally divided
into two major pathways: local and systemic delivery
(Whitehead et al., 2009). The choice of administration is
dependent on the accessibility of target site and desired ef-
fects, which would in turn determine the total dose of siRNA
required, the amount of siRNA successfully arriving at target
tissue, potential side effects and to consider the need of an
agent that facilitates delivery (Wittrup and Lieberman, 2015;
Zuckerman and Davis, 2015). One should always be in
check-and-balance and weigh their pros and cons when de-
ciding on the choice of siRNA administration route.
For local siRNA delivery, high concentration of siRNA
can be delivered to the target tissue via local injection, in-
travitreal, intranasal, topical absorption and many other
methods as described in Table 2. This proved to be useful in
the treatment of “locally-treatable” diseases such as age-re-
lated macular degeneration (AMD) affecting only the eyes
and therefore therapeutics should be given through in-
travitreal administration. Apart from higher bioavailability
of siRNA, local siRNA delivery method also promises re-
duction in adverse effects, simple formulation and ease of
administration among others (Akhtar and Benter, 2007).
Herein, we highlight some siRNA drug candidates currently
in clinical trials; targeting different diseases via local deliv-
ery (Table 3).
Nevertheless, systemic delivery of siRNA is vital for
Figure 4 The proper folding of TTR tetramer into protein and the misfolding of the tetramer which then translates into a misfolded protein, resulting in the
formation of amyloid fibril. Patisiran (Onpattro) is a 100 nm multi-lipid component nanoparticle (SNALP) encapsulating siRNA to silence the mutant TTR
gene responsible for tetramer misfolding in the liver of hATTR patients.
489
Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
siRNA to reach a specific target, or multiple organs, for
example diseases like cancer (Dunning et al., 2016; Kol-
dehoff and Elmaagacli, 2009; Koldehoff et al., 2010), hy-
percholesterolemia (Kosmas et al., 2018;Tadin-Strapps et
al., 2011), amyloidosis (Adams et al., 2018;Coelho et al.,
2013a), viral infections (Morrissey et al., 2005b), and others.
Also, siRNA should retain its biological activity upon
reaching target site. Therefore, it is vital for siRNAs to be
formulated into different carrier systems so that they may be
protected from nuclease digestion and have improved phar-
macokinetics and bioactivity (Morrissey et al., 2005a). Im-
proved nuclease-stability is especially important for siRNA
duplexes that are exposed to nuclease-rich environments
during systemic circulation as naked siRNAs will degrade in
serum within minutes (Layzer et al., 2004). Designing sys-
temic delivery carrier for siRNA do possess significantly
larger hurdles than local delivery and therefore causing some
drug candidates to be withdrawn or terminated at various
stages of trials (Kanasty et al., 2013). siRNA drug candidates
targeting different diseases via systemic delivery are listed
below in Table 4.
siRNA delivery: the hurdles
Theoretically, siRNA could be designed to target and silence
virtually all mRNA. However, the major obstacle in clinical
application of siRNA lies in developing safe and effective
method of delivering siRNA to target cells, and not in de-
signing potent siRNAs (Burnett and Rossi, 2012;Whitehead
et al., 2009). siRNA is a large (~13 kD) polyanionic mac-
romolecule and could not cross the cell membrane unaided
due to negative charge repulsion of cell membranes (de
Fougerolles et al., 2007;Robbins et al., 2008). siRNA re-
quires specific vehicles that could facilitate its intracellular
uptake and cytosolic delivery for bioactivity. Apart from that,
these delivery systems should circumvent all barriers faced
by siRNA; these include reducing its susceptibility to serum
nucleases, renal filtration, and uptake by the mononuclear
phagocyte system (depicted in Figure 5)(Jain and Styliano-
poulos, 2010;Kumari et al., 2011;Whitehead et al., 2009;
Wittrup and Lieberman, 2015). Consequently, much attempts
have been made to develop an efficient siRNA carrier which
would mitigate the barriers and toxicity issues. Generally,
delivery system is divided into viral or non-viral techniques.
Although virus-like nanoparticles proved to be highly effi-
cient in delivering siRNA, safety concerns surrounding the
long-term infusion of viral vectors rendered it less attractive
as compared to non-viral vectors, namely lipid-based nano-
particles (Saw et al., 2017;Zatsepin et al., 2016;Zimmer-
mann et al., 2006) and polymeric nanoparticles (Xu et al.,
2017;Xu et al., 2016). Besides encapsulation, siRNA could
also be adsorbed onto nanoparticle surface (Elbakry et al.,
2009;Steinbacher and Landry, 2014), complexed with po-
sitive-charged condensation agent (Beloor et al., 2015;Ta-
galakis et al., 2014), conjugation with other
biomacromolecules (Jeong et al., 2009;Springer and Dowdy,
2018), or being modified to increase siRNA stability
(Choung et al., 2006;Kenski et al., 2012;Terrazas and Kool,
2009). Multiple approaches to overcome siRNA delivery
hurdles are summarized in Table 5.
siRNA delivery systems in current clinical trials
There are innumerable publications on siRNA delivery
system every year, yet interestingly only few made it to the
clinical trial stage. Currently only three siRNA delivery
systems are in active clinical trials; namely lipid-based na-
noparticles (LNPs), N-acetylgalactosamine conjugated siR-
Table 2 The pros and cons of siRNA delivery routes
Route of administration Purpose Advantages Disadvantages
Intravenous injection Systemic delivery Broad distribution of siRNA, high localization
in liver
Non-specific, higher dose needed,
clearance by RES and renal
Subcutaneous injection Systemic delivery Broad distribution of siRNA, high localization
in liver, avoid RES and renal clearance Non-specific, skin toxicity
Local injection Localized delivery High localized concentration of siRNA, lower
dose needed, reduce systemic side effect
Not applicable to all organs
and tissues
Topical application Transepithelial absorption (oral,
rectal, vaginal mucosa)
Higher patient compliance, non-invasive, high
local concentration of siRNA, lower dose
needed, reduce systemic side effect
The need to bypass thick
mucosal layer
Intravitreal injection Localized delivery High local concentration, bypass systemic
barriers
Lower patient compliance, eye
irritation
Intrathecal/intraventricular
injection
Delivery to central nervous
system (CNS)
Bypass the dense blood-brain barrier, high
local concentration, reduce systemic side
effects
Low patient compliance, direct
toxicity to CNS
Inhalation/intranasal/
intratracheal administration Pulmonary delivery High local concentration, reduce systemic
side effects
Higher loss of drug in aerosol,
low patience compliance, especially
for intratracheal administration
490 Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
Table 3 Local delivery of siRNA-based therapeutics and their current status in clinical trialsa)
Drug
Method of
delivery/
Delivery vehicle
Disease Clinical Trial No. Phase Status Year
completed Ref.
Bevasiranib
Opko Health
Target: VEGF
Intravitreal/NC Age-related macular degeneration
NCT 00722384 Phase I Completed 2007 (Dejneka et al., 2008;
Garba and Mousa,
2010;Landa et al.,
2009;Singerman,
2009;Stepien et al.,
2009)
NCT 00259753 Phase II Completed 2007
NCT 00499590 Phase III Terminated 2009
NCT 00557791 Phase III Withdrawn N/A
Diabetic macular edema NCT 00306904 Phase II Completed 2007
AGN-745
Allergan
Target: VEGFR
Intravitreal/NC Age-related macular degeneration,
Choroidal neovascularization
NCT 00363714 Phase I/II Completed 2007 (Cho et al., 2009;
Kleinman et al., 2008)
NCT 00395057 Phase II Terminated 2009
ALN-RSV01
Alnylam
Target: RSV-N
Intranasal/NC Respiratory Syncytial Virus Infection
NCT 00496821 Phase II Completed 2007 (Alvarez et al., 2009;
DeVincenzo et al.,
2010;Zamora et al.,
2011)
NCT 00658086 Phase II Completed 2009
NCT 01065935 Phase IIb Completed 2012
TD101
Transderm
Target: k6a
Transdermal/NC Pachyonychia Congenita NCT 00716014 Phase I Completed 2008 (Leachman et al., 2010)
Excellair
Zabecor
Target: Syk
Intranasal/NC Asthma
Phase I Completed 2009
(Burnett et al., 2011)
Phase II On-going N/A
PF-655
Quark/Pfizer
Target: RTP801
Intravitreal/NC
Age-related macular degeneration NCT 00725686 Phase I Completed 2010
Choroidal neovascularization NCT 00713518 Phase II Completed 2011
Diabetic macular edema NCT01445899 Phase II Completed 2013
Diabetic retinopathy, diabetes
complications NCT 00701181 Phase II Terminated 2011
SYL1001
Sylentis
Target: TRPV1
Intravitreal/NC Ocular pain, dry eye syndrome
NCT 01438281 Phase I Completed 2012
NCT 01776658 Phase I/II Completed 2015
NCT 02455999 Phase II Completed 2016
NCT 03108664 Phase III Recruiting Est: 2018
SYL040012
(Bamosiran)
Sylentis
Target: β2-AR
Intravitreal/NC
Ocular hypertension, glaucoma NCT 00990743 Phase I Completed 2010
(Vaishnaw et al., 2010)
NCT 01227291 Phase I/II Completed 2012
Ocular hypertension, open angle
glaucoma NCT 01739244 Phase II Completed 2013
Open angle glaucoma NCT 02250612 Phase II Completed 2016
SIG12D
Silenseed
Target: KRAS
Implant/LODER
polymer
Pancreatic Ductal Adenocarcinoma,
Pancreatic cancer
NCT 01188785 Phase I Completed 2013
(Golan et al., 2015)
NCT 01676259 Phase II Recruiting Est: 2020
QPI-1007
Quark
Target: Caspase-2
Intravitreal/NC
Optic Atrophy, Non-arteritic Anterior
Ischemic Optic Neuropathy NCT 01064505 Phase I Completed 2013
Glaucoma, Angle closure, Primary,
Acute NCT 01965106 Phase II Completed 2015
Non-arteritic Anterior Ischemic Optic
Neuropathy NCT 02341560 Phase II/III Recruiting Est: 2020
RXI-109
Rxi
Target: CTGF
Transdermal/NC
Cicatrix, scar prevention NCT 01640912 Phase I Completed 2014
Hypertrophic scars NCT 02246465 Phase I Completed 2014
NCT 02030275 Phase II Completed 2016
Keloid NCT 02079168 Phase II Completed 2016
AMD, CN, Subretinal scarring,
subretinal fibrosis NCT 02599064 Phase I/II On-going Est: 2018
OLX-10010
OliX
Target: CTGF
Transdermal/NC Cicatrix, hypertrophic NCT 03569267 Phase I Recruiting Est: 2018
STP705
(Cotsiranib®)
Sirnanomics
Target: TGF-β and
Cox-2
Subcutaneous
injection/
polypeptide
nanoparticle
(PNP)
Hypertrophic scars NCT 02950317 Phase I Recruiting Est: 2018
a) VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; RSV-N, human respiratory syncytial virus neuclo-
protein; k6a, keratin 6a; syk, spleen tyrosine kinase; RTP801 (also known as REDD1/Dig2/DDIT4), DNA damage inducible transcript 4; TRPV1, the
transient receptor potential cation channel subfamily V member 1; β2-AR, Adrenoreceptor Beta-2; KRAS, kirsten rat sarcoma; CTGF (also known as CCN2),
connective tissue growth factor; TGF-β, transforming growth factor-beta; COX2 (also known as PTGS2), prostaglandin-endoperoxide synthase 2.
491
Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
Table 4 Systemic delivery of siRNA-based therapeutics and their current status in clinical trials
Drug Method of
delivery Disease Clinical Trial No. Phase Status Year
completed Ref.
QPI-1002
Quark
Target: p53
Intravenous/NC
Acute renal failure, acute kidney injury NCT 00683553 Phase I Terminated 2010
(Demirjian et al.,
2017)
Delayed graft function, complication of
kidney transplant NCT 00802347 Phase I/II Completed 2014
Acute kidney injury NCT 02610283 Phase II Completed 2018
Delayed graft function NCT 02610296 Phase III On-going Est: 2019
Cardiac surgery NCT 03510897 Phase III On-going Est: 2021
TKM-ApoB
Tekmira
Target: ApoB
Intravenous/
SNALP Hypercholesterolemia NCT 00927459 Phase I Terminated 2010
(Jeffs et al., 2005;
Morrissey et al.,
2005b)
ALN-VSP02
Alnylam
Target: KSP and VEGF
Intravenous/LNP Solid tumors
NCT 00882180 Phase I Completed 2011 (Cervantes et al.,
2011;Tabernero et
al., 2013)
NCT01158079 Phase I Completed 2012
CALAA-01
Calandro
Target: RRM2
Intravenous/
Cyclodextrin
polymer-based
nanoparticles
Solid tumor, cancer NCT 00689065 Phase I Terminated 2012 (Davis, 2009;
Davis et al., 2010)
ALN-TTR01
Alnylam
Target: TTR
Intravenous/first
generation LNP
Transthyretin mediated amyloidosis
(ATTR) NCT 01148953 Phase I Completed 2012 (Coelho et al.,
2013a)
ALN-PCS02
Alnylam
Target: PCSK9
Intravenous/
Second
generation LNP
Elevated LDL-Cholesterol (LDL-C) NCT 01437059 Phase I Completed 2012 (Fitzgerald et al.,
2014)
TKM-PLK1
Tekmira
Target: PLK-1
Intravenous
/LNP
Colorectal cancer with hepatic
metastases, pancreas vancer eith
hepatic metastases, gastric cancer with
hepatic metastases, breast cancer with
hepatic metastases, ovarian cancer with
hepatic metastases
NCT 01437007 Phase I Completed 2012
(Northfelt et al.,
2013;Semple et
al., 2011)
Cancer, neuroendocrine tumors, NET,
adrenocortical carcinoma, ACC NCT 01262235 Phase I Completed 2015
TKM-100201
Tekmira
Target: VP24 and VP35
Intravenous/LNP Ebola virus infection NCT 01518881 Phase I Terminated 2012 (Dunning et al.,
2016)
BCR-ABL
University of
Duisburg-Essen
Target: BCR-ABL gene
Intravenous/
Anionic
liposome
Chronic myeloid leukemia Phase I
(Koldehoff and
Elmaagacli, 2009;
Koldehoff et al.,
2010)
Atu-027
Silence
Target: PKN3
Intravenous/
cationic lipoplex
Advanced solid tumors NCT 00938574 Phase I Completed 2012 (Aleku et al., 2008;
Strumberg et al.,
2012a;Strumberg
et al., 2012b)
Carcinoma, pancreatic ductal NCT 01808638 Phase I/II Completed 2016
ND-L02-S0201
Nitto Biopharma
Target: HSP 47
Intravenous/
Vitamin A
coupled liposome
Fibrosis NCT 01858935 Phase I Completed 2014 (Sato et al., 2008;
Soule et al., 2018)
Moderate to extensive hepatic
fibrosis (METAVIR F3-4) NCT 02227459 Phase I Completed 2017
ARC AAT
Arrowhead
Target: AAT gene
Intravenous/
Dynamic
PolyConjugates
Alpha 1-antitrypsin dDeficiency
NCT 02363946 Phase I Terminated 2016
NCT 02900183 Phase II Withdrawn
ARC 520
Arrowhead
Pharmaceutical
Target: coagulation factor
VII (F7)
Intravenous/
Dynamic
PolyConjugates
Hepatitis B
NCT 01872065 Phase I Terminated 2014
(Lanford R. E.,
2013;Schluep et
al., 2017;Yuen,
2014)
NCT 02535416 Phase I Completed 2016
NCT 02738008 Phase II Terminated 2016
NCT 02604199 Phase II Terminated 2017
Chronic hepatitis B
NCT 02349126 Phase II Terminated 2016
NCT 20604212 Phase II Terminated 2016
NCT 02452528 Phase II Terminated 2016
NCT 02065336 Phase II Terminated 2017
Hepatitis B, Hepatitis D NCT 02577029 Phase II Terminated 2016
ALN-PCSsc (Inclisiran)
Alnylam Target: PCSK9
Subcutaneous/
GalNAc
conjugate
Hypercholesterolemia NCT 02314442 Phase I Completed 2014
(Kosmas et al.,
2018)
Atherosclerotic cardiovascular disease
(ASCVD), familial
hypercholesterolemia (FH), diabetes
NCT 02597127 Phase II Completed 2017
Renal impairment NCT 03159416 Phase I Active, not
recruiting Est: 2018
(To be continued on the next page)
492 Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
(Continued)
Drug Method of
delivery Disease Clinical Trial No. Phase Status Year
completed Ref.
ALN-PCSsc (Inclisiran)
Alnylam Target: PCSK9
Subcutaneous/
GalNAc
conjugate
Homozygous FH NCT 02963311 Phase II Active, not
recruiting Est:2018
(Kosmas et al.,
2018)
Heterozygous FH, elevated cholesterol NCT 03397121 Phase III Active, not
recruiting Est: 2019
ASCVD, elevated cholesterol NCT 03400800 Phase III Active, not
recruiting Est: 2019
NCT 03399370 Phase III
ASCVD, Symptomatic atherosclerosis,
Type 2 Diabetes NCT 03060577 Phase II Active, not
recruiting Est: 2022
ASCVD NCT 03705234 Phase III Not yet
recruiting Est: 2049
ALN-TTRSC (Revusiran)
Alnylam
Target: TTR
Subcutaneous/
GalNAc
conjugate
Transthyretin-mediated amyloidosis
(ATTR)
NCT 01814839 Phase I Completed 2015
(Zimmermann et
al., 2013)
NCT 02797847 Phase I Completed 2018
NCT 01981837 Phase II Completed 2015
NCT 02292186 Phase II Completed 2017
TTR mediated familial amyloidotic
cardiomyopathy (FAC), amyloidosis,
hereditary, amyloid neuropathies,
familial amyloid neuropathies,
transthyretin cardiac amyloidosis
NCT 02319005 Phase III Completed 2017
DCR-MYC
Dicerna
Target: MYC
Intravenous/
LNP-formulated
Dicer substrate
siRNA
(DsiRNA)
Solid tumors, multiple myeloma,
non-hodgkins lymphoma, pancreatic,
neuroendocrine tumors, PNET, NHL
NCT 02110563 Phase I Terminated 2016
(Chipumuro et al.,
2016;Tolcher et
al., 2015)
ALN-AT3SC
(Fitusiran)
Alnylam
Target: AT3
Subcutaneous/
GalNAc
conjugate
Hemophilia A, B
NCT 02035605 Phase I Completed 2017
(Sorensen B et al.,
2015;Sorensen et
al., 2014)
NCT 02554773 Phase II On-going Est: 2021
NCT 03417245 Phase III Recruiting Est: 2019
NCT 03417102 Phase III Recruiting Est: 2019
Hemophilia A, B; inhibitors NCT 03549871 Phase III Recruiting Est: 2021
ALN-AS 1
(Givosiran)
Alnylam
Target: ALAS 1
Subcutaneous/
GalNAc
conjugate
Acute intermittent porphyria NCT 02452372 Phase I Completed 2017
NCT 02949830 Phase I/II On-going Est: 2020
AIP, acute hepatic porphyria (AHP),
acute porphyria (AP) NCT 03505853 Phase I Recruiting Est: 2018
AHP, AIP, AP, hereditary copropor-
phyria (HCP), variegate porphyria
(VP), ALA dehydratas, deficient por-
phyria (ADP)
NCT 03338816 Phase III On-going Est: 2021
ARB-1467
(TKM HBV)
Arbutus
Target: HbsAg
Intravenous/LNP Hepatitis B, Chronic NCT 02631096 Phase II Completed 2018
DCR-PHXC-101
Dicerna
Target: LDHA
Subcutaneous/
GalNAc
conjugate
Primary hyperoxaluria NCT 02956317 Phase I Recruiting Est: 2018
ARO-HBV
Arrowhead
Pharmaceutical
Target: HBV gene product
Subcutaneous/
TRiM platform Hepatitis B NCT 03365947 Phase I/II Recruiting Est: 2019
ARO-AAT
Arrowhead
Pharmaceutical
Target: AAT gene
Subcutaneous/
TRiM platform Alpha 1-antitrypsin deficiency NCT 03362242 Phase I On-going Est: 2019
AMG 890
(formerly ARO-LPA)
Amgen
Target: undisclosed
Subcutaneous/
TRiM platform Cardiovascular diseases NCT 03626662 Phase I Recruiting Est: 2019
siRNA-EphA2
MD Anderson
Target: EphA2
Intravenous/LNP Advanced cancers NCT 01591356 Phase I Recruiting Est: 2021 (Landen et al.,
2005)
a) ApoB, apolipoprotein B; KSP, kinesin-like protein; RRM2, ribonucleoside-diphosphate reductase subunit M2; TTR, transthyretin; PCSK9, Proprotein
convertase subtilisin/kexin type 9; PLK-1, Polo-like Kinase 1; VP24, Membrane-associated protein VP24; VP35, Membrane-associated protein VP35; BCR-
ABL, The ABL gene from chromosome 9 joins to the BCR gene on chromosome 22, to form the BCR-ABL fusion gene; PKN3, Serine/threonine-protein
kinase N3; HSP47, Heat Shock Protein 47; AT3, Antithrombin 3; ALAS1, Delta-aminolevulinate synthase 1; AAT, alpha-1 antitrypsin; EphA2, Ephrin type-
A receptor 2; LDHA, lactate dehydrogenase A.
493
Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
NA system (GalNAc-siRNA), and Targeted RNAi Molecule
(TRiM) platform. LNPs have several advantages compared
to other nanocarriers such as high in vivo compatibility, re-
latively low toxicity, ability to encapsulate large amount of
siRNA, biodegradable, stable in storage, straightforward
synthesis and scale-up procedures. Stable nucleic acid lipid
particles (SNALPs, 70–120 nm) are the second generation of
LNP formulation used in the development of Patisiran
(Onpattro) (Figure 6A). At present, many delivery systems in
clinical trials from Tekmira Pharmaceuticals and Alnylam
Pharmaceuticals are formulated using SNALPs and they are
currently in different stages of clinical trial for various dis-
Figure 5 The mechanism of siRNA and the barriers of siRNA delivery. Naked siRNA faces more biological barriers as compared to siRNA encapsulated,
adsorbed or attached to nanoparticles. Biological barriers include low serum stability, failure to extravasate due to poor tissue penetration, poor diffusion
through extra-cellular matrix, ineffective release from endosomes and failure to dissociate and release the siRNA from the carrier.
Table 5 Multiple approaches in circumventing siRNA delivery hurdles. Adapted from de Fougerolles et al., 2007;Dykxhoorn and Lieberman, 2006a,b;
Lieberman and Sharp, 2015;Petrocca and Lieberman, 2011;Wittrup and Lieberman, 2015
Avoiding excretion and improve
circulation half-life
PEGylation-increases molecular weight of siRNA to avoid renal clearance, simultaneously decreasing
anti-biofouling effect and opsonization by RES.
Nanoparticle formulation–usually around 10–100 nm, above the molecular weight cut-off for renal clearance.
Cholesterol conjugation of siRNA-cholesterol bounds to endogenous apolipoprotein resulting in longer
circulation time.
Avoiding nuclease degradation
Chemically modified siRNA-phosphothioate linkage to siRNA backbones confers nuclease resistance.
Nanoparticle formulation-protecting siRNA inside the core of delivery vehicle ensures that siRNA is biologically
active upon reaching target site.
Extravasation
Targets tissues with leaky vessels (i.e., liver, spleen or tumor tissues) that allow nanoparticles carrying siRNA to
extravasate out of blood vessels into target tissues.
Endothelial transcytosis-applicable to all tissues if carriers can be made to bind endothelial specific receptors to
induce transcytosis.
Specifically targets endothelial cells (i.e., endothelial cells in tumor tissues overexpressed VEGF and EDB) for
entry.
Binding to target cells and
intracellular transport
Targeting ligand-conjugates siRNA or delivery vehicle to cell specific receptor targeting moiety (i.e., peptide,
aptamer, antibody) for specific binding and subsequent endocytosis.
Hijacking endogenous ligand-cholesterol-conjugated siRNA and nanocarriers bind to apolipoproteins in
circulation therefore enhancing their liver homing properties.
Endosomal escape
Increases endosomal accumulation-even if siRNA or carriers do not possess endosomal escape mechanism,
sufficient amount of siRNA present could compensate for poor endosomal release.
Endosomal membrane destabilizing peptide/polymer/lipid-masked charge reversal peptide/polymer could be
activated in acidic pH resulting in unmasking positive charged particle therefore destabilizing the endosomal
membrane. Lipid nanoparticles are also activated by the low pH in endosome, resulting in reversed hexagonal
membrane formation, therefore destabilizing the membrane.
494 Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
eases (Singh and Peer, 2016). MD Anderson Cancer Center
developed a neutral-charged DOPC-based LNPs system to
encapsulate EphA2 siRNA (Landen et al., 2005) for the
treatment of advanced cancers, and have just started re-
cruitment for a Phase I clinical trial (NCT01591356).
GalNAc-conjugated siRNA system chemically links the
siRNA to a trimer of N-acetylgalactosamine (GalNAc)
(Foster et al., 2018;Nair et al., 2017;Springer and Dowdy,
2018). In preclinical rodent and non-human primate, it has
been proven that GalNAc binds with high avidity to the
Asialoglycoprotein receptor (ASGPR) which is pre-
dominantly expressed on liver hepatocytes (Morell et al.,
1971;Nair et al., 2014). Through subcutaneous administra-
tion, the GalNAc-conjugated siRNA then enters the blood
stream and localizes in the liver leading to the binding of
GalNAc ligand towards hepatocyte-restricted asialoglyo-
protein receptor (ASGPR). This specific binding causes he-
patocyte specific uptake of GalNAc-siRNA conjugate,
leading to durable gene silencing in the whole liver with
minimal toxicity. Interestingly, the GalNAc-siRNA con-
jugate system was predicted to eventually replace most
therapeutic LNPs for liver targets as the conjugates are
simpler in structure, cheaper to manufacture, better tolerated
and can be subcutaneously administered (Wittrup and Lie-
berman, 2015). Alnylam is currently running four clinical
trials based on GalNAc-siRNA conjugate system (with three
of them in Phase 3 clinical trial) (Figure 6B).
Recently, Arrowhead Pharmaceutics unveiled their new
targeted RNAi molecule (TRiM) platform which uses li-
gand-mediated tissue-specific targeted delivery of siRNA.
The uniqueness of this platform lies in its ability to screen for
potent siRNA, high affinity targeting ligands and linkers to
optimize the pharmacokinetics of each drug candidate,
leading to a more personalized siRNA delivery platform
(Figure 6C). This simple yet robust siRNA delivery system
could pave the way for next generation siRNA delivery as
simple manufacturing promises a reduction in production
cost combined with the selection of multiple administration
routes (i.e., subcutaneous injection and inhalation). The ad-
vantage of being small in size reduces potential safety and
possibility of inflammatory responses. Arrowhead Pharma-
ceutics currently have three TRiM mediated siRNA delivery
in clinical trials and others still under development.
siRNA: concurrent challenges
To date, almost 100,000 siRNA-related publications can be
found in PubMed. The first decade after siRNA discovery
(1998–2008) saw a steady exponential increase in siRNA-
related publications (Figure 7A). During this period of time,
RNAi became the “go-to” techniques for all research, and
this hype has led major pharmaceutic companies to invest
billions in RNAi therapeutics in hopes to treat major diseases
without speculating the hurdles and technical obstacles,
leading to the initiation of 14 clinical trials. Not too sur-
prisingly, most clinical trials in the early times failed to meet
expectations. In the most striking case, OPKO Health an-
nounced the shutdown of its Phase III trial of an RNAi
treatment for wet macular degeneration even after successful
Phase I and II trials. Other RNAi-based drugs provoked
strong innate immune reactions, failed to produce an im-
provement on the disease, or both (see full references in
Tables 4 and 5). In the latter decade (2008–2018), although
publications were maintained at about 7,000 publications per
year, there is an increase of clinical trials to 44 studies in-
itiated (Figure 7B), indicating that there is an improvement
in the clinical development of RNAi therapeutics. Never-
theless, this is roughly 0.06% successful translation rate from
laboratory to clinics. This seemingly unparallel translation
Figure 6 siRNA delivery system currently in clinical trials. A, Stable nucleic acid lipid particle (SNALP) developed by Alnylam, also the delivery vehicle
of Patisiran (Onpattro). B, siRNA conjugated to triantennary GalNac to form siRNA-GalNAc conjugate for specific hepatocyte targeting. C, Targeted RNAi
Molecule (TRIM platform) consisted of targeting ligand, linker and chemically stabilized siRNA for simple yet robust siRNA delivery.
495
Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
success of siRNA might suggest a direct evidence of diffi-
culties in developing clinically-translatable siRNA ther-
apeutics. Herein, we highlight some of the challenges that
still need to be addressed in siRNA delivery.
Long-term safety of siRNA. RNAi has just emerged in
clinical tests since early 2000. There is no long-term and
comprehensive patient safety data to prove that RNAi ther-
apeutic is absolutely safe and tolerable in human (Dana et al.,
2017). Since siRNA is a potent gene silencing machinery,
there is a high probability of off-target effects in which an
unrelated gene similar to the target gene can also be silenced
(Jackson and Linsley, 2010). Although this could be cir-
cumvented by lowering siRNA administration dose or in-
troducing modifications on the siRNA (i.e., circular siRNA,
Dicer-siRNA complex) (Zhang et al., 2018), such potential
can cause adverse effects on the patients’ health.
Off-target effect of siRNA. Although siRNA is generally
known to be specific with high probability of achieving the
desired on-target silencing, they could generate several types
of off-target effects. When an imperfect pairing of siRNA
and mRNA occurs (usually with sequence motifs in the
3′URT regions of mRNAs), it sets off microRNA-like off-
target silencing effect (Birmingham et al., 2006;Jackson et
al., 2003;Jackson et al., 2006). Interestingly, the delivery
vehicles used to deliver siRNA could be non-specifically
taken up by myeloid or dendritic cells, activating the Toll-
like receptors (TLR) and thus triggering an innate immune
response in these cells (Hornung et al., 2005;Judge et al.,
2005;Kariko et al., 2004;Robbins et al., 2009;Sioud, 2005;
Sledz et al., 2003)
Immunogenicity of siRNA. Based on the few published
data confirming siRNA’s non-immunogenicity, researchers
took it for granted that siRNA is generally non-immunogenic
and had not repeated the immunogenetic assay with all
siRNA sequences. Since all siRNA sequence is unique and
specific, they should all be tested individually to rule out the
possibility of becoming immunogenic especially after few
cycles of administration. Few published studies actually
measured serum cytokine levels after siRNA administration.
Most would simply employ control siRNAs to ensure that
immune stimulation or other unintended events did not oc-
cur.
Long-term safety of nanocarrier. Although numerous for-
mulations are being devised for siRNA delivery, focus
should remain on biological stability, specificity and safety
of these nanocarriers, which would ease bench-to-bedside
transition. Complex formulations involving multiple com-
ponents are not only difficult to scale-up but could also be
costly and challenging from a production regulatory stand-
point.
Immunogenicity of nanocarrier. Naked RNA may be poor
immunogen by itself, but siRNA conjugated to peptides,
polymers (i.e., PEGylation), antibodies could easily become
immunogenic. PEGylation is one of the most common way
to increase systemic stability of siRNA. However, the Phase
III trial of PEGylated RNA aptamer against factor IX (Re-
gado Biosciences) had to be discontinued due to severe
anaphylactic reaction to the PEG part of the conjugate
leading to death in some severe cases, raising significant
concerns in nanocarrier’s immunogenicity (Wittrup and
Lieberman, 2015).
Non-specific liver uptake in systemic siRNA delivery. As
the liver is the main blood-filtering organ, it is undoubtedly
easier to deliver RNAi-based therapeutics to liver cells than
to any other organs. Accordingly, development of siRNA
drugs has mostly focused on targets in the liver (Lieberman
and Sharp, 2015). However, long-term non-specific uptake
could lead to long-term liver toxicity. One should be focus-
ing on strategies that will bring about high target tissue (i.e.,
solid tumor) to liver ratio or increase siRNA bioavailability
at the target tissue, especially for non-liver targeted delivery,
thereby increasing therapeutic efficacy. If a suitable method
of siRNA delivery could be developed to minimize non-
specific liver uptake, the potential applications of RNAi-
based therapies can be greatly expanded.
Heterogeneity of disease (cancer). In a solid tumor, mul-
tiple mutations could emerge from different pathways at
various time-points. In other words, a single siRNA silencing
Figure 7 siRNA related publications and number of clinical trials in the first decade of RNAi discovery (A) and the latter decade (B).
496 Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
one specific oncogene might not be sufficient to effectively
cause long-term tumor inhibition. Several siRNAs can be
used in combination for targeting different survival pathways
to provide a synergistic silencing effect (also known as
cocktail siRNA). In addition, delivering siRNA in combi-
nation with currently FDA-approved anti-cancer drugs may
sensitize the treatment of a tumor (especially for drug re-
sistance tumors) and may become novel therapeutic mod-
alities (Behlke, 2006).
Long process of bench to bedside transition. The process
of a new drug development is excruciatingly long. With the
transition of siRNA drug in mind, one should consider in-
itiating early discussions with FDA to obtain expert re-
commendations that could speed up the development
process. Currently, it is estimated to take about 10 years from
target discovery to IND approval. Since RNAi therapeutics
are considered new as compared to conventional drug, the
hope is to obtain expedited IND approval for siRNA-based
drug to move these promising technologies towards clinical
application in a shorter period of time (Ozcan et al., 2015).
Conclusion
The FDA approval of Patisiran is timely and is seen as an
acknowledgement of the immense potential that siRNA
possess for treating a myriad of diseases. With many other
promising candidates currently at Phase 3 clinical trials, we
can predict more FDA approval of siRNA-based therapeutics
in the years to come. Combining meticulous design of the
target gene and siRNA delivery system, we envision that one
day we would be able to achieve a “knock-out” of all disease-
causing genes to eliminate incurable diseases such as pro-
geria, systemic lupus, Alzheimer, malignant brain tumors
and many more. siRNA therapeutics is now a clinical reality.
Acknowledgements This work was supported by the National Key Re-
search and Development Program of China (2016YFC1302300), the Na-
tional Natural Science Foundation of China (81720108029, 81621004, and
81490750), Guangdong Science and Technology Department
(2016B030229004), Guangzhou Science Technology and Innovation Com-
mission (201803040015), and the Fountain-Valley Life Sciences Fund of
University of Chinese Academy of Sciences Education Foundation.
References
Adams, D., Gonzalez-Duarte, A., O’Riordan, W.D., Yang, C.C., Ueda, M.,
Kristen, A.V., Tournev, I., Schmidt, H.H., Coelho, T., Berk, J.L., et al.
(2018). Patisiran, an RNAi therapeutic, for hereditary transthyretin
amyloidosis. N Engl J Med 379, 11–21.
Akhtar, S., and Benter, I.F. (2007). Nonviral delivery of synthetic siRNAs
in vivo.J Clin Invest 117, 3623–3632.
Aleku, M., Schulz, P., Keil, O., Santel, A., Schaeper, U., Dieckhoff, B.,
Janke, O., Endruschat, J., Durieux, B., Röder, N., et al. (2008). Atu027,
a liposomal small interfering RNA formulation targeting protein kinase
N3, inhibits cancer progression. Cancer Res 68, 9788–9798.
Alvarez, R., Elbashir, S., Borland, T., Toudjarska, I., Hadwiger, P., John,
M., Roehl, I., Morskaya, S.S., Martinello, R., Kahn, J., et al. (2009).
RNA interference-mediated silencing of the respiratory syncytial virus
nucleocapsid defines a potent antiviral strategy. Antimicrob Agents
Chemother 53, 3952–3962.
Ando, Y., Coelho, T., Berk, J.L., Cruz, M.W., Ericzon, B.G., Ikeda, S.,
Lewis, W.D., Obici, L., Planté-Bordeneuve, V., Rapezzi, C., et al.
(2013). Guideline of transthyretin-related hereditary amyloidosis for
clinicians. Orphanet J Rare Dis 8, 31.
Bäumer, S., Bäumer, N., Appel, N., Terheyden, L., Fremerey, J., Schelhaas,
S., Wardelmann, E., Buchholz, F., Berdel, W.E., and Müller-Tidow, C.
(2015). Antibody-mediated delivery of anti-KRAS-siRNA in vivo
overcomes therapy resistance in colon cancer. Clin Cancer Res 21,
1383–1394.
Behlke, M.A. (2006). Progress towards in vivo use of siRNAs. Mol Ther
13, 644–670.
Beloor, J., Zeller, S., Choi, C.S., Lee, S.K., and Kumar, P. (2015). Cationic
cell-penetrating peptides as vehicles for siRNA delivery. Therap Deliv
6, 491–507.
Birmingham, A., Anderson, E.M., Reynolds, A., Ilsley-Tyree, D., Leake,
D., Fedorov, Y., Baskerville, S., Maksimova, E., Robinson, K.,
Karpilow, J., et al. (2006). 3′ UTR seed matches, but not overall
identity, are associated with RNAi off-targets. Nat Meth 3, 199–204.
Bumcrot, D., Manoharan, M., Koteliansky, V., and Sah, D.W.Y. (2006).
RNAi therapeutics: a potential new class of pharmaceutical drugs. Nat
Chem Biol 2, 711–719.
Burnett, J.C., and Rossi, J.J. (2012). RNA-based therapeutics: current
progress and future prospects. Chem Biol 19, 60–71.
Burnett, J.C., Rossi, J.J., and Tiemann, K. (2011). Current progress of
siRNA/shRNA therapeutics in clinical trials. Biotech J 6, 1130–1146.
Cervantes, A., Alsina, M., Tabernero, J., Infante, J.R., LoRusso, P., Shapiro,
G., Paz-Ares, L.G., Falzone, R., Hill, J., Cehelsky, J., et al. (2011).
Phase I dose-escalation study of ALN-VSP02, a novel RNAi
therapeutic for solid tumors with liver involvement. J Clin Oncol 29,
3025.
Chipumuro, E., Siddiquee, Z., Ganesh, S., Shui, S., Shah, A., Kim, B.,
Chen, D., Pandya, P., Storr, R., Wang, W., et al. (2016). Abstract 2925:
anti-tumor activity of a MYC-targeting dicer substrate siRNA in
combination with BRD4/CDK7 inhibitors. Cancer Res 76, 2925.
Cho, W.G., Albuquerque, R.J.C., Kleinman, M.E., Tarallo, V., Greco, A.,
Nozaki, M., Green, M.G., Baffi, J.Z., Ambati, B.K., De Falco, M., et al.
(2009). Small interfering RNA-induced TLR3 activation inhibits blood
and lymphatic vessel growth. Proc Natl Acad Sci USA 106, 7137–7142.
Choung, S., Kim, Y.J., Kim, S., Park, H.O., and Choi, Y.C. (2006).
Chemical modification of siRNAs to improve serum stability without
loss of efficacy. Biochem Biophys Res Commun 342, 919–927.
Coelho, T., Adams, D., Silva, A., Lozeron, P., Hawkins, P.N., Mant, T.,
Perez, J., Chiesa, J., Warrington, S., Tranter, E., et al. (2013a). Safety
and efficacy of RNAi therapy for transthyretin amyloidosis. N Engl J
Med 369, 819–829.
Coelho, T., Maurer, M.S., and Suhr, O.B. (2013b). THAOS–the
transthyretin amyloidosis outcomes survey: initial report on clinical
manifestations in patients with hereditary and wild-type transthyretin
amyloidosis. Curr Med Res Opin 29, 63–76.
Conceição, I., González-Duarte, A., Obici, L., Schmidt, H.H.J., Simoneau,
D., Ong, M.L., and Amass, L. (2016). “Red-flag” symptom clusters in
transthyretin familial amyloid polyneuropathy. J Peripher Nerv Syst 21,
5–9.
Dana, H., Chalbatani, G.M., Mahmoodzadeh, H., Karimloo, R., Rezaiean,
O., Moradzadeh, A., Mehmandoost, N., Moazzen, F., Mazraeh, A.,
Marmari, V., et al. (2017). Molecular Mechanisms and biological
functions of siRNA. Inter J Biomed Sci 13, 48–57.
Dang, C.V., Reddy, E.P., Shokat, K.M., and Soucek, L. (2017). Drugging
the “undruggable” cancer targets. Nat Rev Cancer 17, 502–508.
Davidson, B.L. (2003). Hepatic diseases—hitting the target with inhibitory
RNAs. N Engl J Med 349, 2357–2359.
497
Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
Davis, M.E. (2009). The first targeted delivery of siRNA in humans via a
self-assembling, cyclodextrin polymer-based nanoparticle: from
concept to clinic. Mol Pharm 6, 659–668.
Davis, M.E., Zuckerman, J.E., Choi, C.H.J., Seligson, D., Tolcher, A.,
Alabi, C.A., Yen, Y., Heidel, J.D., and Ribas, A. (2010). Evidence of
RNAi in humans from systemically administered siRNA via targeted
nanoparticles. Nature 464, 1067–1070.
de Fougerolles, A., Manoharan, M., Meyers, R., and Vornlocher, H.P.
(2005). RNA interference in vivo: toward synthetic small inhibitory
RNA-based therapeutics. Methods Enzymol 392, 278–296.
de Fougerolles, A., Vornlocher, H.P., Maraganore, J., and Lieberman, J.
(2007). Interfering with disease: a progress report on siRNA-based
therapeutics. Nat Rev Drug Discov 6, 443–453.
de Fougerolles, A.R. (2008). Delivery vehicles for small interfering RNA
in vivo.Human Gene Ther 19, 125–132.
Dejneka, N.S., Wan, S., Bond, O.S., Kornbrust, D.J., and Reich, S.J.
(2008). Ocular biodistribution of bevasiranib following a single in-
travitreal injection to rabbit eyes. Mol Vision 14, 997–1005.
Demirjian, S., Ailawadi, G., Polinsky, M., Bitran, D., Silberman, S.,
Shernan, S.K., Burnier, M., Hamilton, M., Squiers, E., Erlich, S., et al.
(2017). Safety and tolerability study of an intravenously administered
small interfering ribonucleic acid (siRNA) post on-pump cardiothoracic
surgery in patients at risk of acute kidney injury. Kidney Int Rep 2, 836–
843.
DeVincenzo, J., Lambkin-Williams, R., Wilkinson, T., Cehelsky, J.,
Nochur, S., Walsh, E., Meyers, R., Gollob, J., and Vaishnaw, A.
(2010). A randomized, double-blind, placebo-controlled study of an
RNAi-based therapy directed against respiratory syncytial virus. Proc
Natl Acad Sci USA 107, 8800–8805.
Dunning, J., Sahr, F., Rojek, A., Gannon, F., Carson, G., Idriss, B.,
Massaquoi, T., Gandi, R., Joseph, S., Osman, H.K., et al. (2016).
Experimental treatment of ebola virus disease with TKM-130803: a
single-arm phase 2 clinical trial. PLoS Med 13, e1001997.
Dykxhoorn, D.M., and Lieberman, J. (2005). The silent revolution: RNA
interference as basic biology, research tool, and therapeutic. Annu Rev
Med 56, 401–423.
Dykxhoorn, D.M., and Lieberman, J. (2006a). Knocking down disease with
siRNAs. Cell 126, 231–235.
Dykxhoorn, D.M., and Lieberman, J. (2006b). Running interference:
prospects and obstacles to using small interfering RNAs as small
molecule drugs. Annu Rev Biomed Eng 8, 377–402.
Elbakry, A., Zaky, A., Liebl, R., Rachel, R., Goepferich, A., and Breunig,
M. (2009). Layer-by-layer assembled gold nanoparticles for siRNA
delivery. Nano Lett 9, 2059–2064.
Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., and
Tuschl, T. (2001). Duplexes of 21-nucleotide RNAs mediate RNA
interference in cultured mammalian cells. Nature 411, 494–498.
Fire, A., Xu, S.Q., Montgomery, M.K., Kostas, S.A., Driver, S.E., and
Mello, C.C. (1998). Potent and specific genetic interference by double-
stranded RNA in Caenorhabditis elegans.Nature 391, 806–811.
Fitzgerald, K., Frank-Kamenetsky, M., Shulga-Morskaya, S., Liebow, A.,
Bettencourt, B.R., Sutherland, J.E., Hutabarat, R.M., Clausen, V.A.,
Karsten, V., Cehelsky, J., et al. (2014). Effect of an RNA interference
drug on the synthesis of proprotein convertase subtilisin/kexin type 9
(PCSK9) and the concentration of serum LDL cholesterol in healthy
volunteers: a randomised, single-blind, placebo-controlled, phase 1
trial. Lancet 383, 60–68.
Foster, D.J., Brown, C.R., Shaikh, S., Trapp, C., Schlegel, M.K., Qian, K.,
Sehgal, A., Rajeev, K.G., Jadhav, V., Manoharan, M., et al. (2018).
Advanced siRNA designs further improve in vivo performance of
GalNAc-siRNA conjugates. Mol Ther 26, 708–717.
Garba, A.O., and Mousa, S.A. (2010). Bevasiranib for the treatment of wet,
age-related macular degeneration. Ophthalmol Eye Dis 2, S4878–
S4883.
Gertz, M.A. (2017). Hereditary ATTR amyloidosis: burden of illness and
diagnostic challenges. Am J Managed Care 23, S107–S112.
Golan, T., Khvalevsky, E.Z., Hubert, A., Gabai, R.M., Hen, N., Segal, A.,
Domb, A., Harari, G., David, E.B., Raskin, S., et al. (2015). RNAi
therapy targeting KRAS in combination with chemotherapy for locally
advanced pancreatic cancer patients. Oncotarget 6, 24560–24570.
Hamar, P., Song, E., Kökény, G., Chen, A., Ouyang, N., and Lieberman, J.
(2004). Small interfering RNA targeting Fas protects mice against renal
ischemia-reperfusion injury. Proc Natl Acad Sci USA 101, 14883–
14888.
Hammond, S.M., Bernstein, E., Beach, D., and Hannon, G.J. (2000). An
RNA-directed nuclease mediates post-transcriptional gene silencing in
Drosophila cells. Nature 404, 293–296.
Hawkins, P.N., Ando, Y., Dispenzeri, A., Gonzalez-Duarte, A., Adams, D.,
and Suhr, O.B. (2015). Evolving landscape in the management of
transthyretin amyloidosis. Ann Med 47, 625–638.
Hornung, V., Guenthner-Biller, M., Bourquin, C., Ablasser, A., Schlee, M.,
Uematsu, S., Noronha, A., Manoharan, M., Akira, S., de Fougerolles,
A., et al. (2005). Sequence-specific potent induction of IFN-α by short
interfering RNA in plasmacytoid dendritic cells through TLR7. Nat
Med 11, 263–270.
Jackson, A.L., Bartz, S.R., Schelter, J., Kobayashi, S.V., Burchard, J., Mao,
M., Li, B., Cavet, G., and Linsley, P.S. (2003). Expression profiling
reveals off-target gene regulation by RNAi. Nat Biotechnol 21, 635–
637.
Jackson, A.L., Burchard, J., Schelter, J., Chau, B.N., Cleary, M., Lim, L.,
and Linsley, P.S. (2006). Widespread siRNA “off-target” transcript
silencing mediated by seed region sequence complementarity. RNA 12,
1179–1187.
Jackson, A.L., and Linsley, P.S. (2010). Recognizing and avoiding siRNA
off-target effects for target identification and therapeutic application.
Nat Rev Drug Discov 9, 57–67.
Jain, R.K., and Stylianopoulos, T. (2010). Delivering nanomedicine to solid
tumors. Nat Rev Clin Oncol 7, 653–664.
Jeffs, L.B., Palmer, L.R., Ambegia, E.G., Giesbrecht, C., Ewanick, S., and
MacLachlan, I. (2005). A scalable, extrusion-free method for efficient
liposomal encapsulation of plasmid DNA. Pharm Res 22, 362–372.
Jeong, J.H., Mok, H., Oh, Y.K., and Park, T.G. (2009). siRNA conjugate
delivery systems. Bioconj Chem 20, 5–14.
Johnson, S.M., Connelly, S., Fearns, C., Powers, E.T., and Kelly, J.W.
(2012). The transthyretin amyloidoses: from delineating the molecular
mechanism of aggregation linked to pathology to a regulatory-agency-
approved drug. J Mol Biol 421, 185–203.
Judge, A.D., Sood, V., Shaw, J.R., Fang, D., McClintock, K., and
MacLachlan, I. (2005). Sequence-dependent stimulation of the
mammalian innate immune response by synthetic siRNA. Nat
Biotechnol 23, 457–462.
Kanasty, R., Dorkin, J.R., Vegas, A., and Anderson, D. (2013). Delivery
materials for siRNA therapeutics. Nat Mater 12, 967–977.
Kariko, K., Bhuyan, P., Capodici, J., and Weissman, D. (2004). Small
interfering RNAs mediate sequence-independent gene suppression and
induce immune activation by signaling through toll-like receptor 3. J
Immunol 172, 6545–6549.
Kenski, D.M., Butora, G., Willingham, A.T., Cooper, A.J., Fu, W., Qi, N.,
Soriano, F., Davies, I.W., and Flanagan, W.M. (2012). siRNA-optimized
modifications for enhanced in vivo activity. Mol Ther - Nucleic Acids 1,
e5.
Kleinman, M.E., Yamada, K., Takeda, A., Chandrasekaran, V., Nozaki, M.,
Baffi, J.Z., Albuquerque, R.J.C., Yamasaki, S., Itaya, M., Pan, Y., et al.
(2008). Sequence- and target-independent angiogenesis suppression by
siRNA via TLR3. Nature 452, 591–597.
Koldehoff, M., and Elmaagacli, A.H. (2009). Therapeutic targeting of gene
expression by siRNAs directed against BCR-ABL transcripts in a pa-
tient with imatinib-resistant chronic myeloid leukemia. Methods Mol
Biol 487, 451–466.
Koldehoff, M., Kordelas, L., Beelen, D.W., and Elmaagacli, A.H. (2010).
Small interfering RNA against BCR-ABL transcripts sensitize mutated
T315I cells to nilotinib. Haematologica 95, 388–397.
Kosmas, C.E., Muñoz Estrella, A., Sourlas, A., Silverio, D., Hilario, E.,
Montan, P.D., and Guzman, E. (2018). Inclisiran: a new promising
498 Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
agent in the management of hypercholesterolemia. Diseases 6, 63.
Kumari, A., Kumar, V., and Yadav, S.K. (2011). Nanocarriers: a tool to
overcome biological barriers in siRNA delivery. Expert Opin Biol Ther
11, 1327–1339.
Lam, J.K.W., Chow, M.Y.T., Zhang, Y., and Leung, S.W.S. (2015). siRNA
versus miRNA as therapeutics for gene silencing. Mol Ther-Nucleic
Acids 4, e252.
Landa, G., Amde, W., Doshi, V., Ali, A., McGevna, L., Gentile, R.C.,
Muldoon, T.O., Walsh, J.B., and Rosen, R.B. (2009). Comparative study
of intravitreal bevacizumab (Avastin) versus ranibizumab (Lucentis) in
the treatment of neovascular age-related macular degeneration.
Ophthalmologica 223, 370–375.
Landen, C.N. Jr., Chavez-Reyes, A., Bucana, C., Schmandt, R., Deavers,
M.T., Lopez-Berestein, G., and Sood, A.K. (2005). Therapeutic EphA2
gene targetingin vivo using neutral liposomal small interfering RNA
delivery. Cancer Res 65, 6910–6918.
Lanford R.E., W.C.I., Chavez D, Oropeza C, Chu Q, Hamilton H.L,
McLach-lan A, Given B, Anzalone C.R, Lewis D L (2013). ARC-520
RNAi therapeutic reduces hepatitis B virus DNA, Santigen and eantigen
in a chimpanzee with a very high viral titer: 1035. Hepatology 58.
Layzer, J.M., McCaffrey, A.P., Tanner, A.K., Huang, Z., Kay, M.A., and
Sullenger, B.A. (2004). In vivo activity of nuclease-resistant siRNAs.
RNA 10, 766–771.
Leachman, S.A., Hickerson, R.P., Schwartz, M.E., Bullough, E.E.,
Hutcherson, S.L., Boucher, K.M., Hansen, C.D., Eliason, M.J.,
Srivatsa, G.S., Kornbrust, D.J., et al. (2010). First-in-human mutation-
targeted siRNA phase Ib trial of an inherited skin disorder. Mol Ther 18,
442–446.
Lieberman, J., and Sharp, P.A. (2015). Harnessing RNA interference for
therapy. JAMA 313, 1207–1208.
Lieberman, J., Song, E., Lee, S.K., and Shankar, P. (2003). Interfering with
disease: opportunities and roadblocks to harnessing RNA interference.
Trends Mol Med 9, 397–403.
Morell, A.G., Gregoriadis, G., Scheinberg, I.H., Hickman, J., and Ashwell,
G. (1971). The role of sialic acid in determining the survival of gly-
coproteins in the circulation. J Biol Chem 246, 1461–1467.
Morrissey, D.V., Blanchard, K., Shaw, L., Jensen, K., Lockridge, J.A.,
Dickinson, B., McSwiggen, J.A., Vargeese, C., Bowman, K., Shaffer,
C.S., et al. (2005a). Activity of stabilized short interfering RNA in a
mouse model of hepatitis B virus replication. Hepatology 41, 1349–
1356.
Morrissey, D.V., Lockridge, J.A., Shaw, L., Blanchard, K., Jensen, K.,
Breen, W., Hartsough, K., Machemer, L., Radka, S., Jadhav, V., et al.
(2005b). Potent and persistent in vivo anti-HBV activity of chemically
modified siRNAs. Nat Biotechnol 23, 1002–1007.
Nair, J.K., Attarwala, H., Sehgal, A., Wang, Q., Aluri, K., Zhang, X., Gao,
M., Liu, J., Indrakanti, R., Schofield, S., et al. (2017). Impact of
enhanced metabolic stability on pharmacokinetics and
pharmacodynamics of GalNAc-siRNA conjugates. Nucl Acids Res
45, 10969–10977.
Nair, J.K., Willoughby, J.L.S., Chan, A., Charisse, K., Alam, M.R., Wang,
Q., Hoekstra, M., Kandasamy, P., Kel’in, A.V., Milstein, S., et al.
(2014). Multivalent N-acetylgalactosamine-conjugated siRNA
Localizes in hepatocytes and elicits robust RNAi-mediated gene
silencing. J Am Chem Soc 136, 16958–16961.
Northfelt, D.W., Hamburg, S.I., Borad, M.J., Seetharam, M., Curtis, K.K.,
Lee, P., Crowell, B., Vocila, L., Fredlund, P., Gilbert, M.J., et al. (2013).
A phase I dose-escalation study of TKM-080301, a RNAi therapeutic
directed against polo-like kinase 1 (PLK1), in patients with advanced
solid tumors: expansion cohort evaluation of biopsy samples for evi-
dence of pharmacodynamic effects of PLK1 inhibition. J Clin Oncol 31,
TPS2621.
Ozcan, G., Ozpolat, B., Coleman, R.L., Sood, A.K., and Lopez-Berestein,
G. (2015). Preclinical and clinical development of siRNA-based
therapeutics. Adv Drug Deliver Rev 87, 108–119.
Petrocca, F., and Lieberman, J. (2011). Promise and challenge of RNA
Interference-based therapy for cancer. J Clin Oncol 29, 747–754.
Robbins, M., Judge, A., Ambegia, E., Choi, C., Yaworski, E., Palmer, L.,
McClintock, K., and MacLachlan, I. (2008). Misinterpreting the
therapeutic effects of small interfering RNA caused by immune
stimulation. Human Gene Ther 19, 991–999.
Robbins, M., Judge, A., and MacLachlan, I. (2009). siRNA and innate
immunity. Oligonucleotides 19, 89–102.
Ryther, R.C.C., Flynt, A.S., Phillips, J.A., and Patton, J.G. (2005). siRNA
therapeutics: big potential from small RNAs. Gene Ther 12, 5–11.
Sato, Y., Murase, K., Kato, J., Kobune, M., Sato, T., Kawano, Y., Takimoto,
R., Takada, K., Miyanishi, K., Matsunaga, T., et al. (2008). Resolution
of liver cirrhosis using vitamin A–coupled liposomes to deliver siRNA
against a collagen-specific chaperone. Nat Biotechnol 26, 431–442.
Saw, P.E., Park, J., Jon, S., and Farokhzad, O.C. (2017). A drug-delivery
strategy for overcoming drug resistance in breast cancer through
targeting of oncofetal fibronectin. Nanomed-Nanotechnol Biol Med 13,
713–722.
Schluep, T., Lickliter, J., Hamilton, J., Lewis, D.L., Lai, C.L., Lau, J.Y.,
Locarnini, S.A., Gish, R.G., and Given, B.D. (2017). Safety, tolerability,
and pharmacokinetics of ARC-520 injection, an RNA interference-
based therapeutic for the treatment of chronic hepatitis B virus
infection, in healthy volunteers. Clinl Pharm Drug Dev 6, 350–362.
Semple, S.C., Judge, A.D., Robbins, M., Klimuk, S., Eisenhardt, M.,
Crosley, E., Leung, A., Kwok, R., Ambegia, E., McClintock, K., et al.
(2011). Abstract 2829: preclinical characterization of TKM-080301, a
lipid nanoparticle formulation of a small interfering RNA directed
against polo-like kinase 1. Cancer Res 71, 2829.
Sen, G.L., and Blau, H.M. (2006). A brief history of RNAi: the silence of
the genes. FASEB J 20, 1293–1299.
Shankar, P., Manjunath, N., and Lieberman, J. (2005). The prospect of
silencing disease using RNA interference. JAMA 293, 1367–1373.
Singerman, L. (2009). Combination therapy using the small interfering
RNA bevasiranib. Retina 29, S49–S50.
Singh, M.S., and Peer, D. (2016). siRNA delivery: current trends and future
perspectives. Therap Deliv 7, 51–53.
Sioud, M. (2005). Induction of inflammatory cytokines and interferon
responses by double-stranded and single-stranded siRNAs is sequence-
dependent and requires endosomal localization. J Mol Biol 348, 1079–
1090.
Sledz, C.A., Holko, M., de Veer, M.J., Silverman, R.H., and Williams, B.R.
G. (2003). Activation of the interferon system by short-interfering
RNAs. Nat Cell Biol 5, 834–839.
Song, E., Lee, S.K., Wang, J., Ince, N., Ouyang, N., Min, J., Chen, J.,
Shankar, P., and Lieberman, J. (2003). RNA interference targeting Fas
protects mice from fulminant hepatitis. Nat Med 9, 347–351.
Song, E., Zhu, P., Lee, S.K., Chowdhury, D., Kussman, S., Dykxhoorn, D.
M., Feng, Y., Palliser, D., Weiner, D.B., Shankar, P., et al. (2005).
Antibody mediated in vivo delivery of small interfering RNAs via cell-
surface receptors. Nat Biotechnol 23, 709–717.
Sorensen, B., Mant, T., Georgiev, P., Rangarajan, S., Pasi, K. J., Creagh, D.,
Bevan, D.H., Austin, S., Hay, C., Brand, B., et al., (2015). A sub-
cutaneously administered investigational RNAi therapeutic (ALN-AT3)
targeting antithrombin for treatment of hemophilia: phase 1 study re-
sults in subjects with hemophilia A or B ISTH 2015.
Sorensen, B., Mant, T., Akinc, A., Simon, A., Melton, L., Lynam, C.,
Strahs, A., Sehgal, A., Hutabarat, R., Chaturvedi, P., et al. (2014). A
subcutaneously administered RNAi therapeutic (ALN-AT3) targeting
antithrombin for treatment of hemophilia: interim phase 1 study results
in healthy volunteers and patients with hemophilia A or B. Blood 124,
693.
Soucek, L., and Evan, G.I. (2010). The ups and downs of Myc biology.
Curr Opin Genets Dev 20, 91–95.
Soule, B., Tirucherai, G., Kavita, U., Kundu, S., and Christian, R. (2018).
Safety, tolerability, and pharmacokinetics of BMS-986263/ND-L02-
s0201, a novel targeted lipid nanoparticle delivering HSP47 siRNA, in
healthy participants: a randomised, placebo-controlled, double-blind,
phase 1 study. J Hepatol 68, S112.
Springer, A.D., and Dowdy, S.F. (2018). GalNAc-siRNA conjugates:
499
Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
leading the way for delivery of RNAi therapeutics. Nucl Acid Therap
28, 109–118.
Steinbacher, J.L., and Landry, C.C. (2014). Adsorption and release of
siRNA from porous silica. Langmuir 30, 4396–4405.
Stepien, K.E., Rosenfeld, P.J., Puliafito, C.A., Feuer, W., Shi, W., Al-Attar,
L., Dubovy, S.R., Murray, T.G., Davis, J.L., Lee, W.H., et al. (2009).
Comparison of intravitreal bevacizumab followed by ranibizumab for
the treatment of neovascular age-related macular degeneration. Retina
29, 1067–1073.
Strumberg, D., Schultheis, B., Meyer-Sabellek, W., Vank, C., Gebhardt, F.,
Santel, A., Keil, O., Giese, K., Kaufmann, J., and Drevs, J. (2012a).
Antimetastatic activity of Atu027, a liposomal small interfering RNA
formulation, targeting protein kinase N3 (PKN3): final results of a
phase I study in patients with advanced solid tumors. J Clin Oncol 30,
e13597.
Strumberg, D., Schultheis, B., Traugott, U., Vank, C., Santel, A., Keil, O.,
Giese, K., Kaufmann, J., and Drevs, J. (2012b). Phase I clinical
development of Atu027, a siRNA formulation targeting PKN3 in
patients with advanced solid tumors. Int J Clin Pharmacol Ther 50, 76–
78.
Tabernero, J., Shapiro, G.I., LoRusso, P.M., Cervantes, A., Schwartz, G.K.,
Weiss, G.J., Paz-Ares, L., Cho, D.C., Infante, J.R., Alsina, M., et al.
(2013). First-in-humans trial of an RNA interference therapeutic
targeting VEGF and KSP in cancer patients with liver involvement.
Cancer Discov 3, 406–417.
Tadin-Strapps, M., Peterson, L.B., Cumiskey, A.M., Rosa, R.L., Mendoza,
V.H., Castro-Perez, J., Puig, O., Zhang, L., Strapps, W.R., Yendluri, S.,
et al. (2011). siRNA-induced liver ApoB knockdown lowers serum
LDL-cholesterol in a mouse model with human-like serum lipids. J
Lipid Res 52, 1084–1097.
Tagalakis, A.D., Lee, D.H.D., Bienemann, A.S., Zhou, H., Munye, M.M.,
Saraiva, L., McCarthy, D., Du, Z., Vink, C.A., Maeshima, R., et al.
(2014). Multifunctional, self-assembling anionic peptide-lipid
nanocomplexes for targeted siRNA delivery. Biomaterials 35, 8406–
8415.
Terrazas, M., and Kool, E.T. (2009). RNA major groove modifications
improve siRNA stability and biological activity. Nucl Acids Res 37,
346–353.
Tolcher, A.W., Papadopoulos, K.P., Patnaik, A., Rasco, D.W., Martinez, D.,
Wood, D.L., Fielman, B., Sharma, M., Janisch, L.A., Brown, B.D., et al.
(2015). Safety and activity of DCR-MYC, a first-in-class Dicer-sub-
strate small interfering RNA (DsiRNA) targeting MYC, in a phase I
study in patients with advanced solid tumors. J Clin Oncol 33, 11006–
11006.
Vaishnaw, A.K., Gollob, J., Gamba-Vitalo, C., Hutabarat, R., Sah, D.,
Meyers, R., de Fougerolles, T., and Maraganore, J. (2010). A status
report on RNAi therapeutics. Silence 1, 14.
Wang, Y., Liu, S., Zhang, G., Zhou, C., Zhu, H., Zhou, X., Quan, L., Bai, J.,
and Xu, N. (2005). Knockdown of c-Myc expression by RNAi inhibits
MCF-7 breast tumor cells growth in vitro and in vivo.Breast Cancer
Res 7, R220–R228.
Whitehead, K.A., Langer, R., and Anderson, D.G. (2009). Knocking down
barriers: advances in siRNA delivery. Nat Rev Drug Discov 8, 129–138.
Williams, B.R. (2005). Targeting specific cell types with silencing RNA. N
Engl J Med 353, 1410–1411.
Wittrup, A., and Lieberman, J. (2015). Knocking down disease: a progress
report on siRNA therapeutics. Nat Rev Genet 16, 543–552.
Xu, X., Saw, P.E., Tao, W., Li, Y., Ji, X., Yu, M., Mahmoudi, M.,
Rasmussen, J., Ayyash, D., Zhou, Y., et al. (2017). Tumor
microenvironment-responsive multistaged nanoplatform for systemic
RNAi and cancer therapy. Nano Lett 17, 4427–4435.
Xu, X., Wu, J., Liu, Y., Yu, M., Zhao, L., Zhu, X., Bhasin, S., Li, Q., Ha, E.,
Shi, J., et al. (2016). Ultra-pH-responsive and tumor-penetrating
nanoplatform for targeted siRNA delivery with robust anti-cancer
efficacy. Angew Chem Int Ed 55, 7091–7094.
Yuen, M.F., Chan, H.L.Y., Given, B., Hamilton, J., Schluep, T., Lewis, D.
L., Lai, C.L., Locarnini, S., Lau, J.Y., Gish, R.G. (2014). Phase II, dose
ranging study of ARC-520, a siRNA-based therapeutic, in patients with
chronic hepatitis B virus infection. Hepatology 60, 1280A.
Zamora, M.R., Budev, M., Rolfe, M., Gottlieb, J., Humar, A., Devincenzo,
J., Vaishnaw, A., Cehelsky, J., Albert, G., Nochur, S., et al. (2011). RNA
interference therapy in lung transplant patients infected with respiratory
syncytial virus. Am J Respir Crit Care Med 183, 531–538.
Zatsepin, T.S., Kotelevtsev, Y.V., and Koteliansky, V. (2016). Lipid
nanoparticles for targeted siRNA delivery-going from bench to
bedside. IJN Volume 11, 3077–3086.
Zhang, L., Liang, D., Chen, C., Wang, Y., Amu, G., Yang, J., Yu, L.,
Dmochowski, I.J., and Tang, X. (2018). Circular siRNAs for reducing
off-target effects and enhancing long-term gene silencing in cells and
mice. Mol Ther - Nucleic Acids 10, 237–244.
Zimmermann, T., Karsten, V., Harrop, J., Chan, A., Chiesa, J., Peters, G.,
Falzone, R., Cehelsky, J., Nochur, S., Vaishnaw, A., et al. (2013). Phase
I first-in-humans trial of ALN-TTRsc, a novel RNA interference
therapeutic for the treatment of familial amyloidotic cardiomyopathy
(FAC). J Cardiac Failure 19, S66.
Zimmermann, T.S., Lee, A.C.H., Akinc, A., Bramlage, B., Bumcrot, D.,
Fedoruk, M.N., Harborth, J., Heyes, J.A., Jeffs, L.B., John, M., et al.
(2006). RNAi-mediated gene silencing in non-human primates. Nature
441, 111–114.
Zuckerman, J.E., and Davis, M.E. (2015). Clinical experiences with
systemically administered siRNA-based therapeutics in cancer. Nat Rev
Drug Discov 14, 843–856.
500 Saw, P.E., et al. Sci China Life Sci April (2020) Vol.63 No.4
... These drugs are classified as antisense oligonucleotides (ASO), small interfering RNA (siRNA), and miRNAs. [63][64][65][66][67][68][69][70]85 ASOs target miRNAs and block their functions, such as antagomiRs, and lock nucleic acid (LNA)based anti-miRs, whereas siRNA acts as a mediator of the complementary mRNA degradation and miRNA imitates native double-stranded RNAs to invert the protective elements within cells. 85 Some examples of ASOs are MRG-110, IONIS-ANGPTL3-LRx, AKCEA-APOCIIILR, mipomersen, ISIS APO(a)-Rx, IONIS APO(a)-LRx, and volanesorsen, while the example of siRNA is inclisiran (Table 1). ...
... [63][64][65][66][67][68][69][70]85 ASOs target miRNAs and block their functions, such as antagomiRs, and lock nucleic acid (LNA)based anti-miRs, whereas siRNA acts as a mediator of the complementary mRNA degradation and miRNA imitates native double-stranded RNAs to invert the protective elements within cells. 85 Some examples of ASOs are MRG-110, IONIS-ANGPTL3-LRx, AKCEA-APOCIIILR, mipomersen, ISIS APO(a)-Rx, IONIS APO(a)-LRx, and volanesorsen, while the example of siRNA is inclisiran (Table 1). [63][64][65][66][67][68][69][70] MRG-110 was found to target miR-92a-3p and to bind to 3′UTR of KLF2 and KLF4 which resulted in the inhibition of endothelial inflammation stimulated by shear stress and oxLDL. ...
Article
Full-text available
Atherosclerosis is one of the most dominant pathological processes responsible in cardiovascular diseases (CVD) caused by cholesterol accumulation accompanied by inflammation in the arteries which will subsequently lead to further complications, including myocardial infarction and stroke. Although the incidence of atherosclerosis is decreasing in some countries, it is still considered the leading cause of death worldwide. Atherosclerosis is a vascular pathological process that is chronically inflammatory and is characterized by the invasion of inflammatory cells and cytokines. Many reports have unraveled the pivotal roles of epigenetics such as DNA methylation, post-translational histone modifications, and non-coding RNAs (ncRNAs) in atherogenesis, which regulate the expression of numerous genes related to various responsible pathways. Many studies have been conducted to develop new therapeutical approaches based on epigenetic changes for combating atherosclerosis. This review elaborates on recent updates on the development of new atherosclerosis drugs whose mechanism of action is associated with the modulation of DNA methylation, posttranslational histone modifications, and ncRNA-based gene regulation.
... In the following years, siRNAs were successively used in mammalian cells and mice to specifically silence the expression of different genes which strongly proved the potential of siRNA-therapeutics [19,20]. In 2018, FDA approve the first siRNA therapeutics (Onpattro) or known as ALN-TTR02 for the treatment of Hereditary Transthyretin Amyloidosis (hATTR) [21]. Additional, HSP47 siRNA designed for moderate-tosevere liver fibers was undergoing a phase I clinical trial to evaluate the safety, tolerability, and pharmacokin-etics (PK) of fixed dose in healthy participant in 2018 [22]. ...
Article
Full-text available
Objective Duchenne muscular dystrophy (DMD) is a devastating X-linked neuromuscular disorder caused by various defects in the dystrophin gene and still no universal therapy. This study aims to identify the hub genes unrelated to excessive immune response but responsible for DMD progression and explore therapeutic siRNAs, thereby providing a novel treatment. Methods Top ten hub genes for DMD were identified from GSE38417 dataset by using GEO2R and PPI networks based on Cytoscape analysis. The hub genes unrelated to excessive immune response were identified by GeneCards, and their expression was further verified in mdx and C57 mice at 2 and 4 months (M) by (RT-q) PCR and western blotting. Therapeutic siRNAs were deemed as those that could normalize the expression of the validated hub genes in transfected C2C12 cells. Results 855 up-regulated and 324 down-regulated DEGs were screened from GSE38417 dataset. Five of the top 10 hub genes were considered as the candidate genes unrelated to excessive immune response, and three of these candidates were consistently and significantly up-regulated in mdx mice at 2 M and 4 M when compared with age-matched C57 mice, including Col1a2, Fbn1 and Fn1. Furthermore, the three validated up-regulated candidate genes can be significantly down-regulated by three rational designed siRNA (p < 0.0001), respectively. Conclusion COL1A2, FBN1 and FN1 may be novel biomarkers for DMD, and the siRNAs designed in our study were help to develop adjunctive therapy for Duchenne muscular dystrophy.
... The successful application of Onpattro (Patisiran), the first RNAi therapeutic drug approved by the FDA, is viewed as an acknowledgment of the immense potential of siRNA in treating numerous diseases. 10 In December 2021, the FDA approved inclisiran for the treatment of arteriosclerotic cardiovascular disease (ASCVD), characterized by elevated LDL-C levels or heterozygous familial hypercholesterolemia (HeFH). Research has confirmed that inclisiran effectively reduces LDL-C levels in patients for whom statin therapy alone is inadequate. ...
Article
Full-text available
Psoriasis is a chronic inflammatory skin disease characterized by the excessive proliferation of keratinocytes and heightened immune activation. Targeting pathogenic genes through small interfering RNA (siRNA) therapy represents a promising strategy for the treatment of psoriasis. This mini-review provides a comprehensive summary of siRNA research targeting the pathogenesis of psoriasis, covering aspects such as keratinocyte function, inflammatory cell roles, preclinical animal studies, and siRNA delivery mechanisms. It details recent advancements in RNA interference that modulate key factors including keratinocyte proliferation (Fibroblast Growth Factor Receptor 2, FGFR2), apoptosis (Interferon Alpha Inducible Protein 6, G1P3), differentiation (Grainyhead Like Transcription Factor 2, GRHL2), and angiogenesis (Vascular Endothelial Growth Factor, VEGF); immune cell infiltration and inflammation (Tumor Necrosis Factor-Alpha, TNF-α; Interleukin-17, IL-17); and signaling pathways (JAK-STAT, Nuclear Factor Kappa B, NF-κB) that govern immunopathology. Despite significant advances in siRNA-targeted treatments for psoriasis, several challenges persist. Continued scientific developments promise the creation of more effective and safer siRNA medications, potentially enhancing the quality of life for psoriasis patients and revolutionizing treatments for other diseases. This article focuses on the most recent research advancements in targeting the pathogenesis of psoriasis with siRNA and explores its future therapeutic prospects.
... However, a significant hurdle exists in delivering siRNA molecules to the cancer location since they are chemically unstable and rapidly degraded by nucleases present in the bloodstream. Additionally, effectively and directly delivering siRNA to cancer cells while avoiding off-target effects on normal cells remains a substantial challenge [6,7]. Various types of nanocarriers, such as liposomes [8,9], polymer nanoparticles [10,11], metal nanoparticles [12,13], and peptide nanoparticles [14,15], have been investigated for delivering siRNA. ...
Article
Full-text available
Small interfering RNA (siRNA) has significant potential as a treatment for cancer by targeting specific genes or molecular pathways involved in cancer development and progression. The addition of siRNA to other therapeutic strategies, like photodynamic therapy (PDT), can enhance the anticancer effects, providing synergistic benefits. Nevertheless, the effective delivery of siRNA into target cells remains an obstacle in cancer therapy. Herein, supramolecular nanoparticles were fabricated via the co-assembly of natural histone and hyaluronic acid for the co-delivery of HMGB1-siRNA and the photosensitizer chlorin e6 (Ce6) into the MCF-7 cell. The produced siRNA-Ce6 nanoparticles (siRNA-Ce6 NPs) have a spherical morphology and exhibit uniform distribution. In vitro experiments demonstrate that the siRNA-Ce6 NPs display good biocompatibility, enhanced cellular uptake, and improved cytotoxicity. These outcomes indicate that the nanoparticles constructed by the co-assembly of histone and hyaluronic acid hold enormous promise as a means of siRNA and photosensitizer co-delivery towards synergetic therapy.
... The consecutive FDA approvals of siRNA drugs, Patisiran and Givosiran, have translated the promise of siRNA into clinical reality. 16 With numerous siRNA and mRNA-based therapeutics in the pipeline, the clinical translation of RNA therapeutics has transitioned from mere ''hype'' to a tangible ''hope. '' In the first part of this review, we delve into the role of RNA in diseases and explore the technological advances in RNA therapeutic development. ...
Article
Full-text available
RNA molecules have emerged as promising clinical therapeutics due to their ability to target “undruggable” proteins or molecules with high precision and minimal side effects. Nevertheless, the primary challenge in RNA therapeutics lies in rapid degradation and clearance from systemic circulation, the inability to traverse cell membranes, and the efficient intracellular delivery of bioactive RNA molecules. In this review, we explore the implications of RNAs in diseases and provide a chronological overview of the development of RNA therapeutics. Additionally, we summarize the technological advances in RNA-screening design, encompassing various RNA databases and design platforms. The paper then presents an update on FDA-approved RNA therapeutics and those currently undergoing clinical trials for various diseases, with a specific emphasis on RNA medicine and RNA vaccines.
Article
Full-text available
Glioblastoma multiforme (GBM) is the most aggressive and common adult brain tumor. Current therapies primarily involve surgical resection, followed by chemotherapy and radiotherapy. However, the low survival rate due to high toxicity, side effects, and poor targeting necessitates novel treatments. Post‐transcriptional regulation pathways, particularly the regulation of alternative polyadenylation based on the mRNA 3′UTR, play a critical role in cell growth and development and the progression of tumors. Targeting tumor cells and reprogramming the 3′UTR of tumor suppressor genes to achieve post‐transcriptional regulation is expected to be a new channel for GBM therapy. Herein, a novel siRNA delivery system is developed based on mesoporous silica nanoparticles: siRNA composite nanoparticles (siRNA CNP). Encapsulation in MSNs overcomes siRNA degradation and cellular entry issues, while lipid bilayer coating improves biocompatibility and stability. Surface‐modified nucleic acid aptamer SL1 (Apt‐SL1) significantly enhances GBM targeting, offering therapeutic potential. The findings show that siRNA CNP effectively silences the promoter CFIm25 of distal poly(A) in tumor cells, inducing gene reprogramming, inhibiting tumor growth, and promoting apoptosis in nude mice. The siRNA CNP demonstrates a significant effect in reprogramming tumor suppressor genes and treating GBM. It offers a novel and promising therapeutic avenue for GBM.
Article
Full-text available
Pulmonary fibrosis represents the final alteration seen in a wide variety of lung disorders characterized by increased fibroblast activity and the accumulation of substantial amounts of extracellular matrix, along with inflammatory damage and the breakdown of tissue architecture. This condition is marked by a significant mortality rate and a lack of effective treatments. The depositing of an excessive quantity of extracellular matrix protein follows the damage to lung capillaries and alveolar epithelial cells, leading to pulmonary fibrosis and irreversible damage to lung function. It has been proposed that the connective tissue growth factor (CTGF) plays a critical role in the advancement of pulmonary fibrosis by enhancing the accumulation of the extracellular matrix and exacerbating fibrosis. In this context, the significance of CTGF in pulmonary fibrosis is examined, and a summary of the development of drugs targeting CTGF for the treatment of pulmonary fibrosis is provided.
Article
The mitochondrial enzyme arginase‐2 (Arg‐2) is implicated in the pathophysiology of contrast‐induced acute kidney injury (CI‐AKI). Therefore, Arg‐2 represents a candid target for CI‐AKI prevention. Here, we developed layer‐by‐layer (LbL) assembled renal‐targeting polymeric nanoparticles to efficiently deliver small interfering RNA (siRNA), knockdown Arg‐2 expression in renal tubules, and evaluated prevention of CI‐AKI. Firstly, near‐infrared dye‐loaded poly(lactic‐co‐glycolic acid) (PLGA) anionic cores were electrostatically‐coated with cationic chitosan (CS) to facilitate the adsorption and stabilization of Arg‐2 siRNA. Next, nanoparticles were coated with anionic hyaluronan (HA) to provide protection against siRNA leakage and shielding against early clearance. Sequential layering of electrostatically adsorbed CS and HA improved loading capacity of Arg‐2 siRNA and yielded LbL‐assembled nanoparticles. Renal targeting and accumulation was enhanced by modifying the outermost layer of HA with a kidney targeting peptide (HA‐KTP). The resultant PLGA/CS/HA‐KTP siRNA nanoparticles exhibited proprietary accumulation in kidneys and proximal tubular cells at 24 hours post‐tail vein injection. In iohexol‐induced in vitro and in vivo CI‐AKI models, PLGA/CS/HA‐KTP siRNA nanoparticles alleviated oxidative and nitrification stress, and rescued mitochondrial dysfunction while reducing apoptosis, thereby demonstrating a robust and satisfactory therapeutic effect. Thus, PLGA/CS/HA‐KTP siRNA nanoparticles offer a promising candidate therapy to protect against CI‐AKI. This article is protected by copyright. All rights reserved
Article
Full-text available
The discovery of proprotein convertase subtilisin-kexin type 9 (PCSK9), a serine protease which binds to the low-density lipoprotein (LDL) receptors and targets the receptors for lysosomal degradation, offered an additional route through which plasma LDL-cholesterol (LDL-C) levels can be controlled. Initially, the therapeutic approaches to reduce circulating levels of PCSK9 were focused on the use of monoclonal antibodies. To that effect, evolocumab and alirocumab, two human monoclonal antibodies directed against PCSK9, given on a background of statin therapy, have been shown to markedly decrease LDL-C levels and significantly reduce cardiovascular risk. The small interfering RNA (siRNA) molecules have been used recently to target the hepatic production of PCSK9. siRNA interferes with the expression of specific genes with complementary nucleotide sequences by affecting the degradation of mRNA post-transcription, thus preventing translation. Inclisiran is a long-acting, synthetic siRNA directed against PCSK9 and it has been shown to significantly decrease hepatic production of PCSK9 and cause a marked reduction in LDL-C levels. This review aims to present and discuss the current clinical and scientific evidence pertaining to inclisiran, which is a new promising agent in the management of hypercholesterolemia.
Article
Full-text available
BACKGROUND Patisiran, an investigational RNA interference therapeutic agent, specifically inhibits hepatic synthesis of transthyretin. METHODS In this phase 3 trial, we randomly assigned patients with hereditary transthyretin amyloidosis with polyneuropathy, in a 2:1 ratio, to receive intravenous patisiran (0.3 mg per kilogram of body weight) or placebo once every 3 weeks. The primary end point was the change from baseline in the modified Neuropathy Impairment Score+7 (mNIS+7; range, 0 to 304, with higher scores indicating more impairment) at 18 months. Other assessments included the Norfolk Quality of Life–Diabetic Neuropathy (Norfolk QOL-DN) questionnaire (range, −4 to 136, with higher scores indicating worse quality of life), 10-m walk test (with gait speed measured in meters per second), and modified body-mass index (modified BMI, defined as [weight in kilograms divided by square of height in meters] × albumin level in grams per liter; lower values indicated worse nutritional status). RESULTS A total of 225 patients underwent randomization (148 to the patisiran group and 77 to the placebo group). The mean (±SD) mNIS+7 at baseline was 80.9±41.5 in the patisiran group and 74.6±37.0 in the placebo group; the least-squares mean (±SE) change from baseline was −6.0±1.7 versus 28.0±2.6 (difference, −34.0 points; P<0.001) at 18 months. The mean (±SD) baseline Norfolk QOL-DN score was 59.6±28.2 in the patisiran group and 55.5±24.3 in the placebo group; the least-squares mean (±SE) change from baseline was −6.7±1.8 versus 14.4±2.7 (difference, −21.1 points; P<0.001) at 18 months. Patisiran also showed an effect on gait speed and modified BMI. At 18 months, the least-squares mean change from baseline in gait speed was 0.08±0.02 m per second with patisiran versus −0.24±0.04 m per second with placebo (difference, 0.31 m per second; P<0.001), and the least-squares mean change from baseline in the modified BMI was −3.7±9.6 versus −119.4±14.5 (difference, 115.7; P<0.001). Approximately 20% of the patients who received patisiran and 10% of those who received placebo had mild or moderate infusion-related reactions; the overall incidence and types of adverse events were similar in the two groups. CONCLUSIONS In this trial, patisiran improved multiple clinical manifestations of hereditary transthyretin amyloidosis. (Funded by Alnylam Pharmaceuticals; APOLLO ClinicalTrials .gov number, NCT01960348.)
Article
e13597 Background: Atu027 contains siRNA-lipoplexes, which elicits RNAi mediated suppression on PKN3 in vascular endothelial cells. In various xenograft mouse models, silencing of PKN3 expression and significant inhibition of invasive growth, lymph node and pulmonary metastasis formation was shown. Methods: Atu027 was applied to patients (pts) as a single 4h-infusion with subsequent follow-up for 3 wks. Thereafter pts were treated twice weekly for 4 weeks. In case of SD, pts were treated until PD. Dose escalation was associated with assessment of toxicity, pharmacokinetics (PK), and multiplex biomarker analyses in plasma from treated pts. Results: A total of 33 pts have received Atu027 of 11 dose levels (DL) up to 0.336 mg/kg. No pre-medication was required. No cytokine activation (TNF-α, IL-1β, IFN-γ, IL-6) was observed. In some subjects transient activation of the complement system (C3a, Bb, sC5b-9) was found, but without any clinical relevance. PK-data showed dose-dependent increase in plasma siRNA as well as lipid levels. Among various biomarkers tested, sVEGFR-1 plasma levels decreased significantly upon treatment. Across all dose levels, Atu027 was well-tolerated. Adverse events possibly related to Atu027 were fatigue grade G1 (6pts), hair loss G1 (2pts), sweating G1 (1pt), and abdominal pain G2 (1pt). G3 AEs not considered as DLTs were elevated lipase (2 pts, DL2+DL10) and diarrhea (1 pt, DL5). So far, no DLTs were seen in the last DL. Stable disease after 3 and 6 months was observed in 10 and 3 pts, respectively. Two pts with neuroendocrine cancer had disease stabilization for 9 and 12 months, respectively, including partial regression of pulmonary metastases in 1 pt. Another patient with breast cancer had regression of liver metastases. Conclusions: Atu027 is well-tolerated and anti-metastatic activity has been observed. Soluble VEGFR-1 might serve as a biomarker. So far, 0,336 mg/kg is the recommended dose for further phase II trials.
Article
TPS2621 Background: TKM-080301 is a lipid nanoparticle formulation of a small interfering RNA (siRNA) directed against PLK1, a serine/threonine kinase that regulates multiple critical aspects of cell cycle progression and mitosis. Anti-tumor activity, RNA interference and pharmacodynamic effects of PLK1 inhibition have been conclusively demonstrated in preclinical models. Demonstration of pharmacodynamic effects of PLK1 inhibition in patient biopsy samples is an exploratory objective of this first-in-human study. Methods: TKME080301 is being evaluated in an open-label, non-randomized, dose-escalation study in patients with advanced solid tumors or lymphoma. Sequential cohorts of 3 to 6 patients receive TKME080301 as a 30-minute intravenous infusion on Days 1, 8 and 15 of a 28-day cycle. Treatment can continue until disease progression, based on overall clinical benefit. Tumor response is determined according to RECIST criteria. Primary study objectives include determination of safety, maximum tolerated dose and dose limiting toxicities. Secondary objectives include characterization of pharmacokinetics and the preliminary assessment of anti-tumor activity. Five cohorts have been enrolled and a tentative Phase 2 dose has been identified. An expansion cohort of 10 patients began enrolling in February, 2013. The focus of the expansion cohort will be to collect additional safety and pharmacokinetic data at the tentative Phase 2 dose, as well as pharmacodynamic data from mandatory biopsy samples. Pre- and post-dose biopsy samples will be evaluated for potential evidence of PLK1 inhibition using 5’ RACE (rapid amplification of cDNA ends) polymerase chain reaction (to identify the predicted PLK1 mRNA cleavage product), histology (to assess for the presence of aberrant mitotic figures) and immunohistochemistry. An update on enrollment and pharmacodynamic evaluations will be presented. Clinical trial information: NCT01262235.
Article
Background: Hemophilia A and B are bleeding disorders characterized by a profound defect in thrombin generation (TG). Furthermore, in the presence of normal levels of endogenous anticoagulants a deficiency in factor VIII and IX results in major hemostatic imbalance and a bleeding phenotype. ALN-AT3 is a subcutaneously administered investigational RNAi therapeutic targeting the endogenous anticoagulant antithrombin (AT) that aims to restore the hemostatic balance by increasing TG. Methods: We are conducting a phase 1 multi-center study (NCT02035605) in healthy volunteers and patients with moderate to severe hemophilia A or B. Part A of this study has been completed and assessed a single ascending dose study in healthy volunteers. Parts B and C are multiple ascending dose studies in patients with hemophilia who are receiving weekly or monthly dosing, respectively. Primary endpoints are safety and tolerability. Secondary endpoints include PK, AT knockdown; change in thrombin generation and whole blood clot formation as measured by Calibrated Automated Thrombin generation and ROTEM thromboelastometry. Exploratory endpoints include evaluations of bleed pattern and control. Results: Part A enrolled 4 healthy volunteers, randomized (3:1) to 30 mcg/kg ALN-AT3 or placebo; no serious adverse events (SAE) or injection site reactions were observed. A total of 12 patients with severe hemophilia (10 hemohilia A; 2 hemophilia B) were enrolled in Part B and received 3 weekly subcutaneous doses of ALN-AT3 at 15 (n=3), 45 (n=6), and 75 (n=3) mcg/kg. Similar to part A, weekly administration of ALT-AT3 was generally safe and well tolerated in patients with hemophilia; no SAEs, discontinuations, clinical thromboembolic events or clinically significant D-dimer increases were reported. In the 75 mcg/kg dosing cohort, the mean maximum AT knockdown was 59% (p<0.05, relative to baseline), with nadir levels achieved between days 28 and 42. Maximum plasma AT knockdown of 86% was achieved, resulting in thrombin generation increases that correlated with AT knockdown and a bleed-free period of 114 days in the patient achieving the highest level of AT knockdown. The association between AT KD and TG was assessed in a post hoc exploratory analysis in which AT KD was categorized into tertiles. Part C aims to enroll several cohorts (n=3 per cohort) and will assess a monthly dosing schedule (x3 doses) of ALN-AT3. Patients in cohort 1 and 2 were dosed at 225 and 450 mcg/kg, respectively. Up to 4 additional cohorts may be enrolled within Part C. Updated safety, PK, AT knockdown, TG results as well as bleed patterns from Parts B and C will be presented. Conclusions: Emerging clinical data suggest that targeting AT could be a promising approach for restoring hemostatic balance in hemophilia. The potential for low volume subcutaneous administration, monthly dosing, and applicability to patients with hemophilia A and B with and without inhibitors make ALN-AT3 a potentially encouraging investigational therapy. Disclosures Pasi: Octapharma: Research Funding; Biogen, Octapharma, Genzyme, and Pfizer: Consultancy, Honoraria. Off Label Use: ALN-AT3 is an investigational RNAi therapeutic targeting the endogenous anticoagulant antithrombin.. Mant:Quintiles: Employment, Equity Ownership. Creagh:Bayer Healthcare UK: Honoraria. Austin:SOBI: Other: member of advisory board and received educational support; Pfizer: Other: member of advisory board and received educational support; Novo Nordisk: Other: member of advisory board and received educational support; CSL Behring: Other: member of advisory board and received educational support; Bio Products Laboratory: Other: member of advisory board and received educational support; Bayer: Other: member of advisory board and received educational support; Baxter: Other: member of advisory board and received educational support. Brand:Alnylam: Honoraria. Chowdary:Bayer: Consultancy; Biogen Idec: Consultancy; Baxter: Consultancy; CSL Behring: Consultancy, Research Funding; Novo Nordisk: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; SOBI: Consultancy. Ragni:Tacere Benitec: Membership on an entity's Board of Directors or advisory committees; Alnylam: Research Funding; Bristol Myers Squibb: Research Funding; Biogen: Research Funding; Shire: Membership on an entity's Board of Directors or advisory committees, Research Funding; Dimension: Research Funding; Bayer: Research Funding; SPARK: Research Funding; Genentech Roche: Research Funding; Pfizer: Research Funding; Vascular Medicine Institute: Research Funding; Baxalta: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Biomarin: Research Funding; CSL Behring: Research Funding. Chen:Alnylam Pharmaceuticals: Employment, Equity Ownership. Akinc:Alnylam Pharmaceuticals: Employment, Equity Ownership. Sorensen:Alnylam Pharmaceuticals: Employment, Equity Ownership. Rangarajan:Octapharma: Other: Investigator.
Article
Hereditary transthyretin-mediated (hATTR) amyloidosis is a progressive disease characterized by deposition of amyloid fibrils in various organs and tissues of the body. There are a wide variety of clinical presentations for this multisystemic disorder, so it is often misdiagnosed or subject to delayed diagnosis. Although the exact prevalence is difficult to determine, existing estimates suggest a worldwide prevalence of 50,000 individuals, with varying phenotypic presentations of disease. Due to the heterogeneous nature of its presentation, incorrect or delayed diagnosis can severely impact quality of life for these patients. hATTR amyloidosis can lead to significant disability and mortality. After an accurate diagnosis of hATTR amyloidosis is established, new patients should undergo appropriate therapy as soon as possible. Current treatment options for hATTR amyloidosis are limited, but orthotopic liver transplant serves as an established option for patients with early-stage disease. Consequently, there is a need for new, effective, and safe therapies.
Article
Short-interfering RNA (siRNA)-induced RNAi responses have great potential to treat a wide variety of human diseases from cancer to pandemic viral outbreaks to Parkinson's Disease. However, before siRNAs can become drugs, they must overcome a billion years of evolutionary defenses designed to keep invading RNAs on the outside cells from getting to the inside of cells. Not surprisingly, significant effort has been placed in developing a wide array of delivery technologies. Foremost of these has been the development of N-acetylgalactosamine (GalNAc) siRNA conjugates for delivery to liver. Tris-GalNAc binds to the Asialoglycoprotein receptor that is highly expressed on hepatocytes resulting in rapid endocytosis. While the exact mechanism of escape across the endosomal lipid bilayer membrane remains unknown, sufficient amounts of siRNAs enter the cytoplasm to induce robust, target selective RNAi responses in vivo. Multiple GalNAc-siRNA conjugate clinical trials, including two phase III trials, are currently underway by three biotech companies to treat a wide variety of diseases. GalNAc-siRNA conjugates are a simple solution to the siRNA delivery problem for liver hepatocytes and have shown the RNAi (and antisense oligonucleotide) field the path forward for targeting other tissue types.
Article
Significant progress has been made in the advancement of RNAi therapeutics by combining a synthetic triantennary N-acetylgalactosamine ligand targeting the asialoglycoprotein receptor with chemically modified siRNA designs, including the recently described Enhanced Stabilization Chemistry. This strategy has demonstrated robust RNAi-mediated gene silencing in liver after subcutaneous administration across species, including human. Here we demonstrate that substantial efficacy improvements can be achieved through further refinement of siRNA chemistry, optimizing the positioning of 2′-deoxy-2′-fluoro and 2′-O-methyl ribosugar modifications across both strands of the double stranded siRNA duplex to enhance stability without compromising intrinsic RNAi activity. To achieve this, we employed an iterative screening approach across multiple siRNAs to arrive at advanced designs with low 2′-deoxy-2′-fluoro content that yield significantly improved potency and duration in preclinical species, including non-human primate. Liver exposure data indicate that the improvement in potency is predominantly due to increased metabolic stability of the siRNA conjugates.