Você está na página 1de 14

Advanced Drug Delivery Reviews 61 (2009) 746759

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / a d d r

siRNA vs. shRNA: Similarities and differences


Donald D. Rao a, John S. Vorhies a, Neil Senzer a,b,c,d, John Nemunaitis a,b,c,d,
a
Gradalis, Inc., Dallas, TX, USA
b
Mary Crowley Cancer Research Centers, Dallas, TX, USA
c
Texas Oncology PA, USA
d
Baylor Sammons Cancer Center, Dallas, TX, USA

a r t i c l e i n f o a b s t r a c t

Article history: RNA interference (RNAi) is a natural process through which expression of a targeted gene can be knocked
Received 23 January 2009 down with high specicity and selectivity. Using available technology and bioinformatics investigators will
Accepted 13 April 2009 soon be able to identify relevant bio molecular tumor network hubs as potential key targets for knockdown
Available online 20 April 2009
approaches. Methods of mediating the RNAi effect involve small interfering RNA (siRNA), short hairpin RNA
(shRNA) and bi-functional shRNA. The simplicity of siRNA manufacturing and transient nature of the effect
Keywords:
per dose are optimally suited for certain medical disorders (i.e. viral injections). However, using the
RNA interference
Bi-functional
endogenous processing machinery, optimized shRNA constructs allow for high potency and sustainable
Cancer effects using low copy numbers resulting in less off-target effects, particularly if embedded in a miRNA
Personalized scaffold. Bi-functional design may further enhance potency and safety of RNAi-based therapeutics. Remaining
challenges include tumor selective delivery vehicles and more complete evaluation of the scope and scale of
off-target effects. This review will compare siRNA, shRNA and bi-functional shRNA.
2009 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 746
2. Targeted cancer gene therapy and RNA interference. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 747
2.1. Personalized approach for cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 747
2.2. RNA interference for cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 747
3. Small interfering RNA (siRNA) and short hairpin RNA (shRNA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 748
3.1. siRNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 748
3.2. shRNA. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 750
3.3. Bi-functional shRNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 751
3.4. Summary of si/sh/bi . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 752
4. Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 753
5. Off-target effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 753
5.1. Specic off-target effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 753
5.2. Nonspecic off-target effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 754
6. The future outlook . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 755
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 755
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 756
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 756

1. Introduction
This review is part of the Advanced Drug Delivery Reviews theme issue on Towards
Therapeutic Application of RNA-mediated Gene Regulation. Cancer is a disease of genes, whether based on aberrant changes in
Corresponding author. 1700 Pacic, Suite 1100, Dallas, Texas 75201, USA. Tel.: +1 214 sequence or expression (epigenomics). The constellation of genetic
658 1964; fax: +1 214 658 1992.
E-mail addresses: DRao@gradalisinc.com (D.D. Rao), jsvorhies@gmail.com
and epigenetic abnormalities characterizing cancer cells present new
(J.S. Vorhies), NSenzer@marycrowley.org (N. Senzer), jnemunaitis@marycrowley.org and more specic targets for cancer treatment and, hopefully,
(J. Nemunaitis). prevention. Over the last decade, the mapping of the human genome,

0169-409X/$ see front matter 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2009.04.004
D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759 747

along with improved understanding of signal transduction and the of cancer scale-free networks does not have a threshold for single target
pathways responsible for tumor survival, have been transforming disintegration insofar as random pathway component failure predomi-
therapeutic oncology from a more promiscuously targeted che- nantly affects targets with low connectivity within the network, thereby
motherapeutic approach towards a highly selective targeted ther- having limited functional impact. However, highly connected informa-
apeusis. Indeed, in 1997, antibody-based Herceptin (trastuzumab) tion-transfer targets do allow for attack vulnerability. In other words,
became the rst targeted therapy for breast cancer, specically for the disordered circuitry characteristic of malignancy results in a change,
HER2-positive metastatic breast cancer. In 2001, the small molecule such that the otherwise robust oncogenic process can become, almost
Gleevec (imatinib mesylate), became the rst approved kinase paradoxically, more highly dependent on a specic rewired pathway
inhibitor for cancer targeting bcr-abl in chronic myeloid leukemia (i.e., pathway addiction). Conceptually, knockout of those rewired
(CML), and it has since been approved for the treatment of tumor specic oncogenic pathways should produce a lethal effect on
gastrointestinal stromal tumors (GIST) targeting c-kit. Over the last cancer cells yet not signicantly perturb normal cell functionality.
few years, several other targeted cancer therapies have been Genetic diversity of cancer, pathway addiction and targeted therapy are
approved. Targeted therapeutics directed against amplied genes not the subject of this review and have been extensively reviewed
and/or over-expressed proteins in malignant cells have proven to be elsewhere [68]; here, we discuss how RNAi-based therapeutics can be
powerful tools for cancer treatment. A growing understanding and use best applied in light of these mechanisms.
of proteomic, genetic, and pharmacogenomic tools are actualizing a For example, we harvested tumor and normal cells from the lymph
long desired concept of personalized cancer therapy. Genetic nodes of a melanoma cancer patient for molecular proling by
abnormalities of each patient's tumor can be analyzed through a microarray and proteomic analysis [9]. Expression proles for
variety of established means to quantitatively determine both gene malignant versus normal tissue were compared at the mRNA and
and protein over- and under-expression. Moreover, functional path- protein levels. The goal was to identify a group of gene and protein
ways can be determined and integrated within the cancer network doublets differentially over-expressed in that individual's malignancy.
allowing for the identication of key molecular relays enabling Fig. 1 is a comparative analysis of protein expression prole by the
experimental testing of target specic therapeutics. Such information two-dimensional difference gel electrophoresis (2D-DIGE) analysis.
can potentially allow medical care takers to prioritize, if not yet 2D-DIGE can very effectively identify several over-expressed proteins
optimize, treatment for cancer patients, and to uncover surrogate in the patient's tumor. Correlated DNA/RNA over-expression can then
biomarkers for prognosis, prediction, and therapy assessment. be conrmed with microarray data. The resulting data is further
The recent discovery of RNA interference (RNAi), a natural process analyzed by a modeling and simulation computational system
through which the expression of a targeted gene can be knocked down developed by our team specically for clinical application including,
with high specicity and selectivity, presents an invaluable tool for but not limited to, gene set enrichment analysis and network
personalized cancer therapy. Target specic RNAi agents have the inference modeling platforms (Fig. 2). Grouped gene expression
potential to selectively knockdown key abnormally over- or constitu- patterns that are highly correlated with the pathway phenotype in an
tively expressed molecular targets that are essential for the survival of individual patient allow target genes to be selected, and prioritized
each patient's tumor for effective personalized cancer treatment. based on connectivity and vector-driven criteria developed analytic
Conceptually, target specic RNAi agents can also be applied in network algorithms. Once this individual cancer ngerprint is
combination with immune modulating agents or small molecules to created, it serves as the template for the design, synthesis, and
improve the efcacy of cancer treatment. Like other new therapeutic subsequent validation of individualized therapeutic RNAi molecules
paradigms, there are a multitude of issues which need to be addressed with knockdown activity against these high-degree hub genes for a
in order for us to translate RNA interference technology (siRNA, personalized and targeted cancer gene therapy.
shRNA, bi-functional RNA) from laboratory to bedside and from
concept to reality. These issues include comparison of each of the RNAi 2.2. RNA interference for cancer
technologies with respect to effective delivery, possible off-target
effects and the pharmacokinetics and pharmacodynamics. This review The concept of antisense oligodeoxynucleotides as modulators of
will focus on several issues currently confronting clinical development gene expression and their application in targeted cancer gene therapy
of RNAi therapeutics. We will discuss the role of RNAi technology in was developed more than 25 years ago (for review, [10]). By processes
personalized cancer gene therapy and address clinical considerations still unknown, the antisense nucleic acid (ASNA) strand and the mRNA
of appropriate RNAi-based therapeutic agents for cancer. target come into proximity leading to the destruction of the mRNA
target either by endogenous nucleases, such as RNase H [11,12] that
2. Targeted cancer gene therapy and RNA interference are recruited into the mRNAASNA duplex or by intrinsic enzymatic
activity engineered into the ASNA sequence, as is the case with
2.1. Personalized approach for cancer therapy ribozymes [13,14] and DNAzymes [15,16]. The discovery of an
evolutionarily conserved gene silencing mechanism whereby small
Most human tumors manifest gene expression patterns that differ sequences of extrinsic dsRNA or intrinsic microRNA inhibit comple-
not only from their normal counterparts but, to a lesser extent, even mentary post-transcriptional mRNA (siRNA) or suppress translation
from each other based on both intrinsic gene modications and (miRNA), respectively, ignited strong hope that the natural gene
modulated cancer cellmatrix interactions. Such variability in genetic silencing process would be specic and robust. The silencing process
patterns found between histologically identical tumors arising in occurs following interaction of the RNA effector precursors with the
different patients may well explain the widely divergent responses to RNase III enzymes Drosha (for miRNA) and Dicer (for miRNA and
the standard treatment regimens most often prescribed for a particular siRNA) and subsequent formation of the RNA-interfering silencing
tumor type. This is supported by recent studies demonstrating that complex (RISC) [17]. Endonucleolytic cleavage of the target mRNA
certain patterns of genetic expression (i.e. expression signatures) occurs at a single site ~10 nucleotides from the 5 end of the guide
identied in tumor samples from patients with breast cancer are not (antisense) siRNA sequence [18,19]. RNAi offers several advantages
only strongly correlated with prognosis [14] but can actually be sub- over antisense and ribozyme approaches, including ease of synthesis
classied into differing prognostic categories [5]. The presence of [20] and greater activity [18,2124].
functional redundancy in a robust, predominantly scale-free network Preclinical studies conrm that RNAi techniques can be used to
such as cancer buffers the effect of any single gene/target modication silence cancer-related targets [2535]. In vivo studies have also shown
on the malignant process, with rare exception (e.g., CML). The hierarchy favorable outcomes by RNAi targeting of components critical for
748 D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759

Fig. 1. Analysis of the 2-D DIGE images by DeCyder Software and mass spectrometry protein identication. The upper panel shows on the left the protein expression pattern of a
normal lymph node from patient RW following 2-D gel electrophoresis. In the middle of the upper panel the protein expression pattern of a malignant lymph node from patient RW is
shown and to the right is an overlaid image with proteins from the normal lymph node labeled with Cy3 (green) and proteins from the malignant lymph node labeled with Cy5 (red).
Circles indicate protein spots with signicant expression level changes. The upper right panel shows the fold-of-change distribution curve and the level of change for the spots of
interest. The middle panel shows the 3D view of one protein spot change between the normal and malignant lymph nodes. Mass spectrometry (lower panel) subsequently (following
robotic spot picking) identied this protein as RACK1.

tumor cell growth [26,3639], metastasis [4042], angiogenesis end or the 3 end of the siRNA. Using a uorescence resonance energy
[43,44], and chemoresistance [4547]. transfer (FRET)-based visualization method, the intact siRNA can be
observed to be translocated into the nucleus within 15 min of the
3. Small interfering RNA (siRNA) and short hairpin RNA (shRNA) delivery and then disseminated into the cytoplasm within the next 4 h
both in intact and dissociated form [51]. The initial accumulation of
The applications of RNAi can be mediated through two types of siRNA in the nuclei is similar to observations made on the behavior of
molecules; the chemically synthesized double-stranded small interfer- antisense olignucleotides [52]. The translocation of antisense oligo-
ing RNA (siRNA) or vector based short hairpin RNA (shRNA). Effective nucleotide into the nuclei was not dependent on either the ATP pool or
RNAi was initially demonstrated by the application of synthetic siRNA temperature and thus may not involve the active import transport
[48]; later, siRNA produced in vitro by T7 RNA polymerase was found to system of the nuclear pore [52]; it is not clear whether siRNA
be active and it was soon demonstrated that active siRNA consists of a translocate into the nuclei using the same mechanism as antisense
hairpin structure can be transcribed in cells from an RNA polymerase III olignucleotides. Using HeLa cells and targeting 7SK snRNA which is
promoter on a plasmid construct [49,50]. Although siRNA and shRNA exclusively located in the nucleus, Robb showed the efciency of
can be applied to achieve similar functional outcomes, siRNA and shRNA siRNA mediated silencing to be greater than that effected by antisense
are intrinsically different molecules. Therefore, the molecular mechan- 7SK DNA [53]. Berezhna et al. observed nuclear localization of siRNA
isms of action, the RNA interference pathways, the off-target effects and targeted against small nuclear RNA (snRNA) and cytoplasmic
the applications can also be different. localization of siRNA targeted against viral mRNA suggesting selective
localization and compartmentalization of siRNA based on its intended
3.1. siRNA target [54]. Ago1 and Ago2 containing RISC were found both in the
cytoplasm and nucleus [5557]. A recent study using uorescence
Fluorescent labeled siRNA has been used to trace the fate of correlation spectroscopy and uorescence cross-correlation spectro-
delivered siRNA. A uorescent label can either be tagged onto the 5 scopy (FCS/FCCS) to correlate the presence of siRNA with Ago2
D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759 749

Fig. 2. (A) Nearest neighbor proteinprotein (rst order) interactions of the 6 prioritized proteins in VisualCell. Second order interactions for Stathmin1 (SDCBP) (B) and RACK1
(GNB2L1) (C).

protein, indicated a shuttling of RISC between nucleus and cytoplasm siRNA mediated RNA interference activity which peaks around 24 h
[58]. Importin 8 (Imp8) binds to all Ago proteins in a Ran-dependent post delivery and diminishes within 48 h.
manner, but independently of RNA [59]. Knockdown of Imp8 results in The life-cycle of siRNA inside transfected cells is diagrammatically
a shift of Ago2 from the nucleus to the cytoplasm without affecting the illustrated in Fig. 3. In Drosophila, double-stranded RNA-binding
total quantity of Ago2. However, although Imp8 is not required for proteins (dsRBPs), such as R2D2 and Loquacious (Loqs), function in
target mRNA cleavage it is necessary for Ago2 binding to miRNA tandem with Dicer (Dcr) enzymes in RNA interference (RNAi) [64
targets. In Caenorhabditis elegans, the argonaute protein NRDE-3 is 66]. Dcr-1/Loqs and Dcr-2/R2D2 complexes generate microRNAs
essential for binding nuclear RNAs and appears to interact with (miRNAs) and small interfering RNAs (siRNAs), respectively. Thus,
cytoplasmic siRNAs generated by RNA-dependent RNA polymerase Loqs and R2D2 represent two distinct functional modes for dsRBPs in
(RdRP) followed by redistribution to the nucleus [60]. The nuclear the RNAi pathways [67]. In mammalian cells, only one Dicer gene has
RISC (nRISC) is a complex that is 20 smaller in size than the cyto- thus far been identied [68]. Human Dicer is an integral component of
plasmic RISC (cRISC). The nucleus may be the check point controlling the RNA interference pathway. Dicer processes pre-microRNA and
distribution as either the nuclear acting siRNA or the cytoplasmic double-strand RNA (dsRNA) to mature miRNA and siRNA, respec-
acting siRNA. tively, and transfers the processed products to the RISC [69,70]. Since
Dynamically, siRNA steadily increases its accumulation in cells for there is only one Dicer in the human, the RNA-interfering pathway for
4 h before plateau [61]. The steady-state nuclear distribution of siRNA siRNA and for miRNA may not be as compartmentalized as for
was mainly found in the nucleolus region and was excluded from the Drosophila. Dicer is a multi-domain RNase III-related endonuclease
nucleoplasm [62]. The cytoplasmic distribution of siRNA appears to be responsible for processing dsRNA to siRNAs [71]. Dicer preferentially
in the perinuclear region forming a ring-like pattern around the binds to the 5 phosphate of 2 nt 3 overhang and cleaves dsRNAs into
nucleus [63]. The nucleolus and perinuclear regions are possibly the 21 to 22 nucleotide siRNAs [72,73]. Mammalian Dicer interacts with
main site for RNAi. However, Ohrt et al. labeled siRNA with uorescent the double-stranded TatRNA-binding protein (TRBP) or PACT (PKR
dye at the 3 end of either strand of siRNA and did not nd activating protein) to mediate RNA interference and miRNA proces-
accumulation of siRNA at the perinuclear region, but rather evenly sing. TRBP and PACT are structurally related but exert opposite
distributed throughout the cytoplasm [62]. This discrepancy may be regulatory activities on RNA-dependent protein kinase (PKR). Knock-
the result of the uorescent tagging process. At 48 h post injection, the down of both TRBP and PACT in cultured cells leads to signicant
majority of siRNA appears to have been degraded with only 1% inhibition of gene silencing mediated by short hairpin RNA but not by
uorescence remaining in the cell. The spatial and temporal distribu- siRNA, suggesting that TRBP and PACT function primarily at the step of
tion of siRNA within the cell is in accord with the observed kinetics of siRNA production [74]. Human TRBP and PACT directly interact with
750 D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759

Fig. 3. Schematic of the siRNA mediated RNA interference pathway. After entry into the cytoplasm, siRNA is either loaded onto RISC directly or utilize a Dicer mediated process. After
RISC loading, the passenger strand departs, thereby commencing the RNA interference process via target mRNA cleavage and degradation. siRNA loaded RISCs are also found to be
associated with nucleolus region and maybe shuttled in and out of nucleus through an yet unidentied process.

each other and associate with Dicer to stimulate the cleavage of for the synthesis of shRNA [86], particularly the miR-30 based shRNAs
double-stranded or short hairpin RNA to siRNA [74]. Dicer knockout ES [87].
cells can effectively load processed siRNA onto RISC and carry out RNA shRNA can be transcribed by either RNA polymerase II or III
interference as efciently as Dicer+ ES cells [68]. So, it appears that in through RNA polymerase II or III promoters on the expression cassette.
mammalian cells, a perfectly processed siRNA can be effectively The primary transcript generated from RNA polymerase II promoter
loaded onto RISC for RNAi without the help of the TRBP/PACT/Dicer contains a hairpin like stem-loop structure that is processed in the
complex. The TRBP/PACT/Dicer complex, however, is required to nucleus by a complex containing the RNase III enzyme Drosha and
process either shRNA or long dsRNA to appropriate size and form for the double-stranded RNA-binding domain protein DGCR8 [88]. The
their loading onto RISC. complex measures the hairpin and allows precise processing of
Duplex siRNA in association with holo-RISC, composed of at least the long primary transcripts into individual shRNAs with a 2 nt 3
Ago-2, Dicer and TRBP, is identied as the RISC loading complex (RLC) overhang [89]. The processed primary transcript is the pre-shRNA
[75]. In the RLC, the two strands of the duplex are separated, resulting in molecule. It is transported to the cytoplasm by exportin 5, a Ran-GTP-
the departure of the passenger strand [7678]. The passenger strand is dependent mechanism [90,91]. In the cytoplasm the pre-shRNA is
cleaved by the RNase-H like activity of Ago-2, provided there are loaded onto another RNase III complex containing the RNase III
thermodynamically favorable conditions for passenger strand depar- enzyme Dicer and TRBP/PACT where the loop of the hairpin is
ture. This is referred to as the cleavage-dependent pathway [79]. There is processed off to form a double-stranded siRNA with 2 nt 3 overhangs
also a cleavage-independent by-pass pathway, in which the passenger [9294]. The Dicer containing complex then coordinates loading onto
strand with mismatches is induced to unwind and depart by an ATP the Ago2 protein containing RISC as described earlier for siRNA. Pre-
dependent helicase activity [76,79,80]. The RISC with single-stranded shRNA has been found to be part of the RLC; thus, pre-shRNA may
guide strand siRNA is then able to execute multiple rounds of RNA potentially directly associate with RLC rather than through a two steps
interference. ATP is not required for shRNA processing, RISC assembly, process via a different Dicer/TRBP/PACT complex [95].
cleavage-dependent pathway, or multiple rounds of target-RNA After loading onto RLC and passenger strand departure; both siRNA
cleavage [8183]. Single-stranded siRNA (containing 5-phosphates) and shRNA in the RISC, in principle, should behave the same. The
and pre-miRNA can be loaded on RISC, but not duplex siRNA [84]. argonaute family of proteins is the major component of RISC [96,97].
Within the Argonaute family of proteins, only Ago2 contains the
endonuclease activity necessary to cleave and release the passenger
3.2. shRNA strand of the double-stranded stem [76,77,79]. The remaining three
members of Argonaute family, Ago1, Ago3 and Ago4, which do not
shRNAs, as opposed to siRNAs, are synthesized in the nucleus of have identiable endonuclease activity, are also assembled into RISC
cells, further processed and transported to the cytoplasm, and then and presumably function through a cleavage-independent manner.
incorporated into the RISC for activity [85]. The life-cycle of shRNA Thus, RISC can be further classied as cleavage-dependent and
inside of transfected cells is diagrammatically illustrated in Fig. 4. To cleavage-independent [79].
be effective, the shRNA are designed to follow the rules predicated by The argonaute family of proteins in RISCs are not only involved in the
the specics of the cellular machinery and are presumably processed loading of siRNA or miRNA, but also implicated in both transcriptional
similar to the microRNA maturation pathways. Thus, studies on the (targeting heterochromatin) and post-transcriptional gene silencing.
synthesis and maturation of miRNAs have provided the groundwork Ago protein complexes loaded with passenger strandless siRNA or
D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759 751

Fig. 4. Schematic of the shRNA mediated RNA interference pathway. After delivery of the shRNA expression vector into the cytoplasm, the vector needs to be transported into the
nucleus for transcription. The primary transcripts (pre-shRNA) follow a similar route as discovered for the primary transcripts of microRNA. The primary transcripts are processed by
the Drosha/DGCR8 complex and form pre shRNAs. Pre-shRNAs are transported to the cytoplasm via exportin 5, to be loaded onto the Dicer/TRBP/PACT complex where they are
further processed to mature shRNA. Mature shRNA in the Dicer/TRBP/PACT complex are associated with Argonaute protein containing RISC and provide RNA interference function
either through mRNA cleavage and degradation, or through translational suppression via p-bodies.

miRNA seeks out complementary target sites in mRNAs, where functional shRNA) in cells should achieve a higher level of efcacy,
endonucleolytically active Ago-2 cleaves mRNA to initiate mRNA greater durability compared to siRNA, and a more rapid onset of gene
degradation [98,99]. Other Ago protein containing complexes without expression silencing (the rate dependent on mRNA turnover and
endonucleolytic activity predominantly bind to partially complemen- protein kinetics) compared to shRNA as illustrated on Fig. 5. The bi-
tary target sites located at the 3 UTR for translation repression through functional shRNA, by virtue of loading onto multiple types of RISCs, is
mRNA sequestration in processing bodies (p-bodies) [100102]. The thus able to simultaneously induce degradation of target mRNA and
detailed mechanism of mRNA sequestration in p-bodies and later also inhibit translation through mRNA sequestration. This bi-func-
release from p-bodies is still a debated issue; deadenylation of the target tional design should be, in principal, much more efcient for two
mRNA which leads to destabilization of the mRNA was also observed to reasons; rst, the bi-functional will promote loading the guide strand
occur in p-bodies [103,104]. Coimmunoprecipitation experiments onto at least two types of RISCs to increase activity; second, by loading
determined that RISCs are also strongly associated with polyribosomes onto both cleavage-dependent RISC and cleavage-independent RISC,
or the small subunit ribosomes [95] and Ago-2 (actually identied as target mRNA can be silenced both through mRNA degradation and
elF2c2), strongly suggesting that RISC surveillance is compartmenta- translational inhibition or sequestration.
lized with translational machinery of the cell. Details of the mechanism The design of the bi-functional shRNA expression unit consists of two
involving mRNA scanning and target mRNA identication are still stem-loop shRNA structures; one stem-loop structure composed of fully
largely unknown. Whatever the scanning or surveillance mechanism matched passenger and guide strand for cleavage-dependent RISC
may be, once the target mRNA is identied, the target mRNA is either loading, the second stem-loop structure composed of mis-matched
cleaved or conformationally changed following which both types of passenger strand (at the positions 912) for cleavage-independent RISC
structures are routed to the p-body for either sequestration or loading.
degradation [103,104]. The active siRNA or miRNA loaded complex is There are several experimental observations that support this
then released for additional rounds of gene silencing activity. approach. In Drosophila, Ago1 preferentially binds to miRNAs that
have been excised from imperfectly paired hairpin precursors, whereas
3.3. Bi-functional shRNA those miRNAs that have near-perfectly paired hairpin precursors are
bound by Ago2 [105108]. In HEK293 cells transfected with tagged-Ago
There is, however, a third unique RNAi option in development proteins, coimmunoprecipitation found similar sets of about 600
called bi-functional shRNA. shRNA can potentially be manipulated to transcripts to be bound to Ago1, 2, 3 or 4 [95], suggest all four
take advantage of the gene silencing machinery within the cells to mammalian Ago protein containing RISCs are involved in the RNAi
improve its efciency and durability of action. Conceptually, targeted function. Insofar as most mRNA have multiple miRNA target sites (with
shRNAs can be designed so as to effectively load shRNA onto both the distance constraints) at their 3 UTR, the miRNA mediated RNAi system
cleavage-dependent and the cleavage-independent RISCs. This differ- appears to be redundant for the targeted mRNAs allowing for
ential processing is mediated by two pathways primarily dependent cooperative downregulation to ensure target mRNA knockdown. The
on strand complementarity and/or access to RNase-H cleavage and, bi-functional shRNA approach mimics the natural process by mediating
presumably, for nal target effect, on interaction with Imp8. target mRNA knockdown through multiple RNAi pathways and
Simultaneous expression of both types of shRNAs (i.e. the bi- complexes.
752 D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759

Fig. 5. Schematic of the bi-functional shRNA concept. The bi-functional concept is to design two shRNAs for each targeted mRNA; one with perfect match, one with mismatches at the
central location (bases 912). The purpose of the bi-functional design is to promote loading of mature shRNAs onto both cleavage-dependent and cleavage-independent RISCs, so that
the expression of target mRNA can be more effectively and efciently shut down both through target mRNA degradation and through translational repression.

In C. elegans, structural features of small RNA precursors determine low nM range for most siRNAs, while less than 5 copies of shRNA
Argonaute loading [109]. Recently, Azuma-Mukai et al. observed integrated in the host genome is sufcient to provide continual gene
miRNAs associated with hAgo-2 and hAgo-3 have some overlaps; knockdown effect (Cleary M, personal communication). The higher
however, some are discriminately loaded onto hAgo-2 or hAgo-3 dose required for siRNA can further contribute to the off-target effects
[110]. Further work is needed to resolve the specicity of miRNA to be discussed later.
loading onto different Ago containing RISCs. Although most miRNA Understanding the mechanism and the dynamics of siRNA and
target sites have been identied to be located at the 3-UTR region, shRNA at the cellular and molecular level greatly enhanced the effort
recent systemic identication of mRNAs recruited to hAgo-2 have in developing therapeutic siRNAs for various diseases. As a result,
identied many mRNAs with target sites located at the coding region modications can be made to improve the efcacy and stability of
and some at the 5-UTR [111]. hAgo-2 could initiate the target mRNA RNAi agents. Chemically synthesized siRNA is easier to modify
degradation with its slicing activity in the coding region. Tay et al. through chemistry; however, bulk manufacturing of complex struc-
recently found many naturally occurring miRNA targets are located in tures such as modied siRNA is more expensive. Vector based shRNA
the coding region of embryonic regulated genes to modulate relies on the host machinery for expression, but, on the other hand, is
embryonic stem cell differentiation [112], further support that more difcult to modify. Modication can only be achieved through
miRNA can act through mRNA regions other than 3-UTR. manipulating expression strategy (e.g. bi-functional shRNA), rede-
signing shRNA structure, or by varying promoter regulation. The
3.4. Summary of si/sh/bi shRNA expression units can be incorporated into varieties of plasmids
and viral vectors for delivery and integration. In addition, vector based
In summary, exogenously introduced siRNA with appropriate shRNA expression can also be regulated or induced [118120].
length (1921 nt) and 2 nt 3 overhang, can be loaded onto RISC for Numerous siRNAs have been demonstrated to be effective for in-
RNAi function without interacting with either Dicer, TRBP or PACT; vivo tumor growth modulations via intratumoral, ex-vivo, or systemic
however, the loading process is10 less efcient than shRNA. routes of application (For review see [121123]. Vector based shRNA
Increasing the length of the siRNA duplex to 2930 nt with a 2 nt 3 has, likewise, demonstrated in-vivo effectiveness (For review see
overhang only at one end of the duplex (specically antisense [113]) [121,123,124]. In-vivo studies employ a variety of delivery methods
appears to improve efcacy [114]. If so, it is possibly because and may not ensure equivalency of strand biasing; therefore, it is hard
increasing the length of an siRNA duplex with an unprocessed end to perform direct comparison between siRNA and shRNA. McAnuff et
forces directionality as a result of imposed thermodynamic instability al., using a luciferase expression system, compared the potency of
determining the guide strand motif and thereby enhancing its siRNA versus shRNA mediated knockdown in vivo; they found that
association with Dicer/TRBP/PACT complex for more efcient loading siRNA and shRNA are equivalent in potency at 10 mg dose; however,
onto RLC [114]. shRNA, on the other hand, assimilates into the on a molar basis, the shRNA was 250 fold more effective than the
endogenous miRNA pathway and in so doing is signicantly more siRNA [115]. In an effort to assess the potential of RNAi as a therapeutic
efcient [115117]. Additionally, uorescent tagged siRNA tracing for hepatitis C (HCV), siRNANS5B was targeted against the non-
indicated high degradation and turnover of exogenously introduced structural protein 5B viral polymerase coding region fused with
siRNA. Less than 1% of the introduced duplex remains in the cell 48 h luciferase gene [125]. Luciferase expression in vivo was reduced by
after administration. shRNA can be continuously synthesized by the 75%. Using a cognate shRNANS5B produced a 92.8% average inhibition
host cell, therefore, its effect should be much more durable. over 3 experiments. McCleary et al. incubated shRNA, directed against
Concentrations necessary for effective knockdown are usually in the rey luciferase and containing 29 mer stems and 2-nt 3 overhangs,
D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759 753

with recombinant human Dicer [116]. The resulting 22-nt shRNA counterparts though their transfection efciency is generally lower
products, with predictable guide strands, were compared to identi- [139141]. Non-viral vehicles for delivery of siRNA and shRNA are
cally targeted 21 mer siRNA in HeLa cells. More effective inhibition typically cationic preparations. Their positive charge facilitates
was seen with the shRNA. In another study of the feasibility of RNAi for complexation with negatively charged nucleic acids and also binding
treatment of HCV, 19 and 25 bp shRNAs were compared with 19 and to the negatively charged glycocalyx on external cell membranes
25 bp siRNA directed against the HCV IRES (internal ribosomal entry promoting endocytosis. Both tumor targeting and cell entry can be
site) using a luciferase reporter in the AVA5 cell line with stable enhanced by decoration or complexation of the vehicle with targeting
expression of the HCV subgenomic genotype 1b replicon [117]. The moieties, such as monoclonal antibodies, peptides, small molecule
19 bp shRNAs were more potent than either the 19- or 25-bp siRNAs. ligands, and aptamers to recognize cell surface markers [124,142].
A recent paper evaluated the levels of Dicer and Drosha in cell lines Once endocytosed, the vehicle's positive charge facilitates early escape
and in tumor samples from patients with ovarian cancer [126]. The from the endosome [143,144]. Though the positive charge of these
distribution of Dicer mRNA levels was bimodal and 60% of specimens vehicles improves their transfection efciency, it is also associated
had decreased Dicer mRNA. Furthermore, low Dicer levels were found with increased toxicity [140,145].
to be a predictor of reduced disease-specic survival in multivariate A wide variety of potential vehicles are being developed to address
analysis. Of particular interest was the nding that, compared to siRNA the different issues associated with the delivery of shRNA and siRNA.
mediated silencing of the galectin-3 gene, poor silencing was achieved There are three major classes of non-viral delivery vehicle systems:
with shRNA in ovarian cancer cell lines with low versus high Dicer synthetic polymers, natural/biodegradable polymers, and lipids;
expression. These data will need to be conrmed and evaluated many of the vehicles that are showing promise are actually hybrids
further. Although there is a global downregulation of miRNA of these classes. For instance, there is a cyclodextrin-based cationic
expression in cancer [127], whether this is, in large part, due to low polymer which has been used successfully to deliver siRNA targeted to
Dicer expression or to shortened 3 UTRs with fewer miRNA-binding RRM2 in various in vivo cancer models [134,146]. This preparation is
sites [128] in highly proliferating tumors with modulated feedback currently in Phase I clinical trials. Lipid based nanoparticles are
mechanisms is not known. miRNA functionality has been conrmed in showing potential for the delivery of shRNA and siRNA [147]. Protiva
the three tumor types (ovary, breast, and lung) evaluated in this study Biotherapeutics and Alnaylam have developed nanoparticles com-
raising questions regarding both qualitative and quantitative issues. posed of a lipidPEG conjugate that is capable of encapsulating and
As the authors note, there are data correlating high Dicer expression protecting nucleic acids for the purpose of systemic delivery. These
with poor prognostic features in other tumor types [129,130]. In their stable nucleic acid lipid particles (SNALPs) were used in the rst
retrospective evaluation of lung cancer, the role of let-7 as both a successful administration of siRNAs to a non-human primate
regulator of Dicer and ras and their feedback networks could not be [148,149]. Silence Therapeutics has developed a lipid-based delivery
assessed [131,132]. The Dicer levels in the tumors used for functional vehicle specically designed for siRNA delivery to endothelial cells.
assay of gene silencing are not given nor is there conrmation of the This vehicle, called AtuPLEX, contains a mix of cationic and fusogenic
equivalency of strand biasing between the siRNA and shRNA lipids [150,151]. This vehicle has been used effectively to knockdown
constructs. protein kinase N3 in murine prostate and pancreatic cancer models,
RNAi therapeutics have been shown to be well tolerated in inhibiting cancer progression [152,153]. More detailed discussions of
numerous animal models allowing for transition into the clinic. At delivery vehicles for shRNA [124,154] and siRNA [141,155157] as well
least 10 RNAi-based drugs are currently in early phase clinical trials as general discussions of organ and tissue specic RNAi delivery
[133], two of which are cancer related; one targeted against the M2 [138,158,159] may be found elsewhere.
subunit of ribonucleotide reductase (RRM2) [134] and the other The magnitude of cytokine induction associated with in vivo
targeted against tenascin-C [135]. Animal studies with siRNA delivery of siRNA has been noted to vary widely based on the delivery
inhibitors for RRM2 show efcacy [134] and safety in non-human vehicle used [160]. The well recognized conundrum in cationic non-
primates [136]. shRNA for the treatment of hepatitis B was also viral nucleic delivery is that transfection efciency usually correlates
approved for clinical trial by FDA. with toxicity [161,162]. Effective strategies being pursued to break this
Both siRNA and shRNA have their respective advantages and correlation include molecular modications to shield positive charge
disadvantages from the mechanistic point of view. However, safety of and the use of biodegradable polymers [154,163].
this new therapeutic paradigm is of the utmost importance. Although
no signicant adverse event involving initial RNAi-based clinical trials 5. Off-target effects
has been reported, there are concerns over the potential off-target
effect of RNAi-based agents which will be discussed separately below. Despite initial results showing excellent specicity in RNAi
mediated gene silencing, over the past 5 years many studies have
4. Delivery shown that there are multiple specic and nonspecic mechanisms
through which siRNA and shRNA can cause effects other than the
Efcacy of an RNAi cancer therapeutic is limited by the quantity of intended mRNA suppression. Unintended effects on gene expression
the oligomer that effectively enters the tumor cells. In the clinical mediated by RNAi are termed off-target effects. Specic off-target
setting this is primarily dependent on the method of delivery. An ideal effects are mediated by partial sequence complementarity of the RNAi
delivery vehicle must be able to selectively and differentially target construct to mRNAs other than the intended target. Nonspecic off-
tumors versus normal tissue, homogeneously distribute through the target effects include a wide variety of immune and toxicity related
tumor mass and penetrate the tumor cells following systemic effects that are intrinsic to the RNAi construct itself or its delivery
administration. If cell entry is mediated by endocytosis the delivery vehicle. The following provides a brief review to the mechanisms
vehicle must negotiate endosomal/lysosomal escape and, in the case surrounding off-target effects and addresses strategies that are being
of shRNA, the payload must penetrate the nuclear membrane as well. developed to minimize those effects.
Viral vectors are popular for laboratory delivery of shRNA because of
their high transfection efciency and effective integration of exogen- 5.1. Specic off-target effects
ous DNA, but they have been losing support in recent years because of
concerns over safety and immunogenicity [137,138]. Non-viral poly- Expression proling experiments have shown that partial sequence
meric delivery systems, in particular those with biodegradable complementarity in the passenger or guide strands of the RNAi
components, have much better safety proles than their viral construct can produce off-target gene suppression. The rst group to
754 D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759

denitively demonstrate this used an unbiased genome-wide micro- reductions in off-target effects are sometimes accompanied by
array prole to search for downregulated mRNA immediately after deceases in the overall potency of suppression. [178180].
transfecting a variety of different siRNA constructs directed to different Chemical modications can also be asymmetrically applied to the
genes into HeLa cells. This study demonstrated that few off-target genes passenger strand of the siRNA construct in order to specically inhibit its
were regulated in common. Furthermore, the off-target expression participation in silencing. Annealing of the guide strand to a passenger
patterns observed for each individual siRNA were consistent, with strand composed of two segments has been shown to reduce off-target
repeated runs revealing the same off-target expression prole. It was effects mediated by passenger strand complementarity [181]. The
also noted that the number and identity of the off-target transcripts was addition of other chemical moieties to one or both strands can also
unrelated to the ability of the siRNA to silence the target gene [164]. It limit specic off-target effects. The 5-phosphorylation status of the
has subsequently been shown that both siRNA and shRNA constructs siRNA strands has been shown to be a determinant of which strand is
with complementarity in the seed region can produce the same off- involved in silencing, and thus replacement of the 5 phosphate by of an
target expression proles, even across cell lines, independent of delivery siRNA strand with a methyl group has been shown to reduce its
method [165]. participation in silencing [182].
Even limited sequence siRNA:mRNA complementarity with the Chemical modications to nucleotides within the seed region of
intended target, which is usually less than optimal, can produce off- the passenger or guide strand can reduce unintended entry of siRNA
target suppression. Off-target silencing effects have been demon- constructs into the endogenous miRNA gene silencing pathway by
strated in transcripts with complementarity as low as 7 nucleotides inhibiting the interaction of the RISC complex with mRNA. It has been
with the guide siRNA strand [166]. Due to a variety of known and shown that 2-O-methyl ribosyl substitution at position 2 in the siRNA
unknown mechanisms, not all transcripts with this level of similarity guide strand can reduce off-target silencing of transcripts with
are silenced. The location of the region of complementarity within the complementarity to the seed region of the siRNA guide strand [165].
RNAi construct and the mRNA transcript are important predictors of Asymmetric replacement of seed region nucleotides with DNA bases
potential for suppression. Complementarity within nucleotides 27 at has also been shown to reduce off-targeting as a result of seed region
the 5 end of either the siRNA passenger or guide strands has been complementarity within the passenger strand [183].
shown to be a key determinant in directing off-target effects [167]. Recent in vitro studies have shown that shRNA produces fewer off-
This region of the construct is reminiscent of the seed region within target effects than siRNA. In one study shRNA and siRNA of the same
miRNA, i.e., a heptameric sequence beginning at the rst or second core sequence directed towards TP53 were applied to HCT-116 colon
position from the 5 end of the miRNA that is complementary to sites carcinoma cells in concentrations necessary to achieve comparable
in the mRNA 3-UTRs, which guide silencing in endogenous RNAi. levels of target knockdown. Microarray proling demonstrated a
Indeed, RNAi off-targeting seems to be mechanistically related to an much higher degree of up- and downregulation of off-target
miRNA-like effect in that complementarity between the miRNA seed transcripts in the siRNA transfected cells (M. Mehaffey, T. Ward, and
hexamer and the 3UTR of the off-targeted mRNA produce effective M. Cleary, in prep.). It has been suggested that these differences arise
suppression of gene expression [103,168,169]. from the fact that shRNA is transcribed in the nucleus and is therefore
Various in vitro and in silico methods are either available or under subject to endogenous processing and regulatory mechanisms.
development for analysis of the off-target effects of a given RNAi Additionally, siRNA is more susceptible to degradation in the
construct. Screening can be accomplished effectively using mRNA cytoplasm, which may also lead to off-target silencing [184].
expression data from transfected cells. Expression proling for off-
target screening must be temporally controlled to ensure observation 5.2. Nonspecic off-target effects
of primary changes in mRNA levels and not secondary changes as a
result of downregulation of target protein expression. Results can then Nonspecic off-target effects are those unintended perturbations
be correlated to qRT-PCR data and to protein expression data through in gene expression not resulting from the direct interaction of an RNAi
Western blots. Methods for microarray-based off-target screening are construct with an mRNA transcript. Included in this category are
reviewed in detail elsewhere [169,170]. interferon (IFN) and other immune mediated responses to exogenous
Computational methods are also being developed so that RNAi RNAi, cellular toxicities due to the nucleotide construct, and effects
constructs may be more effectively screened prior to in vitro testing. related to the delivery vehicle. There is strong reason to believe that
Global complementarity search algorithms such as BLASTn and shRNA and siRNA would have very different proles of nonspecic off-
SmithWaterman have been shown to be poor measures of off-target target effects because of the mechanistic differences between the two
potential in an RNAi construct [168]. Methods for in silico screening of approaches.
RNAi constructs must be optimized to consider seed complementarity Introduction of dsRNA longer than 2930 bp into mammalian cells
in the constructs as well as 3 UTR complementarity in the results in a potent induction of the innate immune system via PKR,
transcriptome. Several groups are working to develop algorithms similar to the mammalian cell defense mechanism against viral
and some are freely available online [171174]. infection [185]. Activation of the innate immune response by receptors
The specic off-target effects of a given construct can be mitigated by sensitive to exogenous nucleic acids leads to global degradation of
several methods. siRNAs with an asymmetric unilateral 2-nt-overhang mRNA and thus broad inhibition of translation as well as global
on the antisense strand have greater potency than conventional siRNAs upregulation of IFN-stimulated gene expression. Although shorter
as well as reduced off-target effects due to preferential strand selection siRNA constructs have been shown to avoid receptor activation and
[113]. Sequence based modications designed to reduce specic off- were initially considered to be non-immunogenic [18], subsequent in
target effects are likely to benet both siRNA and shRNA approaches. vitro data have shown that the introduction of both synthetic siRNA
Single and double base mismatches between an RNAi construct and its oligomers and shRNA can induce a partial interferon response [186
target transcript are often tolerated without reducing the potency of 188] some of which are sequence dependent (e.g. GU-dependent, 5-
suppression [168,175177]. This allows for the optimization of the UGUGU-3 and GU-independent, 5-GUCCUUCAA-3) and, therefore,
construct sequence to minimize complementarity with 3UTRs of avoidable. Activation of the innate immune system in the case of
unintended targets. exogenous RNAi is likely mediated through several cytoplasmic and
Although the vector-driven shRNA approach to RNAi does not endosomal mechanisms attuned to recognize exogenous nucleic acids
permit specic chemical modication of the silencing construct, siRNA from infectious agents. The relevant mediators of nucleic acid-
oligomers can be chemically modied in order to reduce direct off- stimulated immunoactivation at the level of the endosome are Toll-
target effects. These modications must be carefully applied as like receptors (TLRs) 7 and 8 (typically activated by ssRNA), TLR 9 (via
D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759 755

unmethylated CpG activation) and TLR 3 (via dsRNA) [189193]. toxicity of siRNAs in mice and hamsters showed that systemic
Immune activation by nucleic acids at the cytoplasmic level is introduction of synthetic siRNAs does not result in the suppression
mediated through RNA sensing receptors such as RIG-I and MDA-5 of endogenous miRNA levels. In this study siRNAs targeting two
[114,194]. Mechanisms surrounding immune activation by RNAi are hepatocyte-specic genes (apolipoprotein B and factor VII) and a
reviewed more thoroughly elsewhere [160,195]. scramble control were administered to mice and one siRNA targeting
Recently, naked siRNA has been shown to activate TLR-3 on the the hepatocyte-expressed gene Scap was administered to hamsters.
surface of vascular endothelial cells and trigger the release of IFN- Robust suppression of target genes was achieved in all cases. No
and IL-12 that mediate nonspecic anti-angiogenic effects in-vivo changes in levels of the hepatocyte endogenously expressed miR-122
[196]. Immune stimulation often is largely responsible for the were observed in the mice or the hamsters and no changes in the
observed therapeutic effects of siRNA rather than the direct targeted broadly expressed miRNAs, miR-16 and let-7a, were observed in the
effect [197]. Misinterpreting the therapeutic effect of siRNA needs to mice [211].
be carefully monitored. Though siRNA likely does not compete with endogenous miRNA
Immune activation at the endosomal level is more readily avoidable for processing proteins, care must be taken when using shRNA as an
by shRNA constructs because the construct is presented on a DNA effector of RNAi in order to minimize the potential for damage
plasmid obviating dsRNA activation of TLR 3. However, TLR 9, as noted, is mediated by over-saturation of exportin-5. In one in vitro study, over-
activated by unmethylated CpG motifs, which are typically found in expression of the exportin-5 protein has been shown to eliminate the
bacterial DNA [198,199]. Careful shRNA-encoding plasmid design, nuclear export bottleneck and allow cells to tolerate higher dosages of
avoiding unmethylated CpG motifs, can effectively attenuate if not shRNA without toxicity [212]. Another study showed that an adeno-
eliminate TLR 9 mediated endosomal immunoactivation [200]. associated virus construct using a Pol-II promoter was able to achieve
There is also reason to believe that shRNA is less likely to induce an stable target gene suppression at high shRNA doses for over 1 year
inammatory response through cytoplasmic dsRNA receptors in vivo after the initial dosing [213]. In order to minimize the risk of toxicity
because shRNA is spliced by endogenous mechanisms. It has been from over-saturation of miRNA professing systems, data to date
suggested that the 5 ends of the endogenous-dicer spliced RNA suggest both selective promoter integration (e.g. pol II versus pol III)
oligomers are less immunogenic than the 5 ends of exogenous siRNA and limiting the dosage of shRNA so as to stay below the threshold of
oligomers [114,194]. This has been supported in vitro in an experiment competitive inhibition of the endogenous miRNA biogenesis
that compared liposome delivered siRNAs versus Pol III promoter machinery.
expressed shRNAs of the same sequence in primary CD34+ progenitor
derived hematopoietic cells. In this study it was found that siRNA 6. The future outlook
induced IFN-alpha and type I IFN genes, while the shRNA of the same
sequence did not induce an immune response [201]. The ability to precisely and differentially target functionally bio-
Sequence modications can be made to shRNA or siRNA in order to relevant molecular signals in patient's cancers will establish a new
reduce immunogenicity. It has been shown that endosomal immu- paradigm in cancer management; one which focuses on dening the
noactivation by siRNA through TLR7 and TLR8 can be sequence uniqueness of each patient's tumor and tumor-host processes and
dependant [189,190,202]. Some simple sequence modications, such interactions following rational target prioritization using computa-
as the introduction of G:U mismatches into the sequence also seem to tional systems biology algorithms. This, then, would allow for
lower the IFN response in vitro [203,204]. One recent study showed exploitation of the attack vulnerability of the rewired cancer
that a marked reduction in the expression of the interferon-stimulated network by deconstructing essential hubs and linkages, multiply
gene oligoadenylate synthetase 1 (Oas1) could be achieved by targeting and eliminating them. Both siRNA and shRNA effectors are
modifying the shRNA to contain features of the naturally occurring attractive opportunities. The capability of potentiating activity using a
microRNA-30 (miR-30) precursor [205]. bi-functional design may further enhance safety and efcacy. The
As in the case of specic off-target effects, chemical modication to simplicity of siRNA manufacturing and the transient nature of the
siRNA oligomers can make them less immunostimulatory, however effect per dose may be optimal for certain medical disorders in which
these modications must be ne-tuned so as not to negatively affect high vector doses are required, e.g. some of the viral infections,
the potency of intended target silencing. Suppression of the TLR however, by using the endogenous processing machinery, optimized
mediated immune response has been achieved by substituting the 2- shRNA constructs allow for high potency sustainable effects using
hydroxyl uridines in the construct with 2-uoro, 2-deoxy, or 2-O- low-copy numbers resulting in less off-target effects (particularly if
methyl uridines [206208], the products of which do retain target- embedded in an miRNA scaffold) thereby ensuring greater safety.
silencing potency while reducing immunogenicity. Though shRNA seems ideal for cancer-related therapeutic develop-
Usage of endogenous processing systems gives shRNA an advan- ment, new technology such as bi-functional RNA interference may
tage over siRNA in terms of its propensity for induction of IFN but its provide an even greater opportunity for enhancement in potency as
over-saturation of these systems has been shown to have other well as heightening safety thereby increasing the opportunities for
consequences that are more easily avoided by siRNA. In a key in vivo multiple target therapy. This, of course, is contingent on optimization
study of the safety effects of long term expression of shRNA in the of delivery and minimization of off-target effect which will need to be
livers of adult mice, a type 8 adeno-associated virus with a Pol III established through early clinical testing.
promoter was used to drive expression of 49 different shRNAs of
different lengths and sequences directed against six targets. 36 of the 7. Conclusions
constructs tested resulted in a dose dependant liver injury that was
determined to be associated with the downregulation of critical Our understanding and application of RNAi has dramatically
endogenous miRNAs. The degree of miRNA downregulation was advanced over the last 5 years. Despite limitations in developing
related neither to the shRNA sequence nor to the degree of effective delivery vehicles and concerns regarding potential off-target
downregulation of the target mRNA [209]. Subsequent transfection activity, clinical development has been initiated. As the science of this
studies suggested the degree of miRNA downregulation to result from edgling technology advances, it is evident that issues such as target
a competitive bottleneck in shared miRNA/shRNA processing, most selection, effector potency, delivery vehicle design, and off-target
likely at the level of exportin-5 (the nuclear membrane export protein effects will continue to be addressed and resolved. Bi-functional RNAi
used to transfer pre-miRNAs into the cytoplasm) and the RISC products are evolving components in this transition to clinically
component, Argonaute-2 [210]. A similar in vivo study of hepatic effective and safer therapeutics.
756 D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759

Acknowledgements [28] M. Yoshinouchi, T. Yamada, M. Kizaki, J. Fen, T. Koseki, Y. Ikeda, T. Nishihara, K.


Yamato, In vitro and in vivo growth suppression of human papillomavirus 16-
positive cervical cancer cells by E6 siRNA, Mol. Ther. 8 (2003) 762768.
The authors would like to acknowledge Brenda Marr and Susan [29] A. Choudhury, J. Charo, S.K. Parapuram, R.C. Hunt, D.M. Hunt, B. Seliger, R.
Mill for their competent and knowledgeable assistance in the Kiessling, Small interfering RNA (siRNA) inhibits the expression of the Her2/neu
gene, upregulates HLA class I and induces apoptosis of Her2/neu positive tumor
preparation of this manuscript, and M. Cleary for providing the data cell lines, Int. J. Cancer 108 (2004) 7177.
before publication. [30] G. Yang, K.Q. Cai, J.A. Thompson-Lanza, R.C. Bast Jr., J. Liu, Inhibition of breast and
ovarian tumor growth through multiple signaling pathways by using retrovirus-
mediated small interfering RNA against Her-2/neu gene expression, J. Biol. Chem.
References 279 (2004) 43394345.
[31] B. Farrow, P. Rychahou, C. Murillo, K.L. O'Connor, T. Iwamura, B.M. Evers,
[1] V.G. Kaklamani, W.J. Gradishar, Gene expression in breast cancer, Curr. Treatm. Inhibition of pancreatic cancer cell growth and induction of apoptosis with novel
Opt. Oncol. 7 (2006) 123128. therapies directed against protein kinase A, Surgery 134 (2003) 197205.
[2] L. Marchionni, R.F. Wilson, S.S. Marinopoulos, A.C. Wolff, G. Parmigiani, E.B. Bass, [32] E. Yague, C.F. Higgins, S. Raguz, Complete reversal of multidrug resistance by stable
S.N. Goodman, Impact of gene expression proling tests on breast cancer expression of small interfering RNAs targeting MDR1, Gene Ther.11 (2004) 11701174.
outcomes, Evid. Rep./Technol. Assess. (Full Rep.) (2007) 1105. [33] B.A. Kosciolek, K. Kalantidis, M. Tabler, P.T. Rowley, Inhibition of telomerase activity
[3] J.K. Habermann, J. Doering, S. Hautaniemi, U.J. Roblick, N.K. Bundgen, D. Nicorici, U. in human cancer cells by RNA interference, Mol. Cancer Ther. 2 (2003) 209216.
Kronenwett, S. Rathnagiriswaran, R.K. Mettu, Y. Ma, S. Kruger, H.P. Bruch, G. Auer, N.L. [34] D.P. Cioca, Y. Aoki, K. Kiyosawa, RNA interference is a functional pathway with
Guo, T. Ried, The gene expression signature of genomic instability in breast cancer is therapeutic potential in human myeloid leukemia cell lines, Cancer Gene Ther.
an independent predictor of clinical outcome, Int. J. Cancer. 124 (7) (2009 Apr 1) 10 (2003) 125133.
15521564. [35] H. Kawasaki, K. Taira, Short hairpin type of dsRNAs that are controlled by tRNA(Val)
[4] P. Wirapati, C. Sotiriou, S. Kunkel, P. Farmer, S. Pradervand, B. Haibe-Kains, C. promoter signicantly induce RNAi-mediated gene silencing in the cytoplasm of
Desmedt, M. Ignatiadis, T. Sengstag, F. Schutz, D.R. Goldstein, M. Piccart, M. human cells, Nucleic Acids Res. 31 (2003) 700707.
Delorenzi, Meta-analysis of gene expression proles in breast cancer: toward a [36] K. Li, S.Y. Lin, F.C. Brunicardi, P. Seu, Use of RNA interference to target cyclin
unied understanding of breast cancer subtyping and prognosis signatures, E-overexpressing hepatocellular carcinoma, Cancer Res. 63 (2003) 35933597.
Breast Cancer Res. 10 (2008) R65. [37] U.N. Verma, R.M. Surabhi, A. Schmaltieg, C. Becerra, R.B. Gaynor, Small interfering
[5] T. Sorlie, R. Tibshirani, J. Parker, T. Hastie, J.S. Marron, A. Nobel, S. Deng, H. Johnsen, RNAs directed against beta-catenin inhibit the in vitro and in vivo growth of colon
R. Pesich, S. Geisler, J. Demeter, C.M. Perou, P.E. Lonning, P.O. Brown, A.L. Borresen- cancer cells, Clin. Cancer Res. 9 (2003) 12911300.
Dale, D. Botstein, Repeated observation of breast tumor subtypes in independent [38] S. Aharinejad, P. Paulus, M. Sioud, M. Hofmann, K. Zins, R. Schafer, E.R. Stanley, D.
gene expression data sets, Proc. Natl. Acad. Sci. U. S. A. 100 (2003) 84188423. Abraham, Colony-stimulating factor-1 blockade by antisense oligonucleotides and
[6] M. Davies, B. Hennessy, G.B. Mills, Point mutations of protein kinases and small interfering RNAs suppresses growth of human mammary tumor xenografts in
individualised cancer therapy, Expert Opin. Pharmacother. 7 (2006) 22432261. mice, Cancer Res. 64 (2004) 53785384.
[7] A.G. Letai, Diagnosing and exploiting cancer's addiction to blocks in apoptosis, [39] H. Uchida, T. Tanaka, K. Sasaki, K. Kato, H. Dehari, Y. Ito, M. Kobune, M. Miyagishi,
Nat. Rev., Cancer 8 (2008) 121132. K. Taira, H. Tahara, H. Hamada, Adenovirus-mediated transfer of siRNA against
[8] O.I. Olopade, T.A. Grushko, R. Nanda, D. Huo, Advances in breast cancer: pathways survivin induced apoptosis and attenuated tumor cell growth in vitro and in vivo,
to personalized medicine, Clin. Cancer Res. 14 (2008) 79887999. Mol. Ther. 10 (2004) 162171.
[9] J. Nemunaitis, N. Senzer, I. Khalil, Y. Shen, P. Kumar, A. Tong, J. Kuhn, J. Lamont, M. [40] A.J. Salisbury, V.M. Macaulay, Development of molecular agents for IGF receptor
Nemunaitis, D. Rao, Y.A. Zhang, Y. Zhou, J. Vorhies, P. Maples, C. Hill, D. Shanahan, targeting, Horm. Metab. Res. 35 (2003) 843849.
Proof concept for clinical justication of network mapping for personalized [41] M.S. Duxbury, H. Ito, M.J. Zinner, S.W. Ashley, E.E. Whang, Focal adhesion kinase
cancer therapeutics, Cancer Gene Ther. 14 (2007) 686695. gene silencing promotes anoikis and suppresses metastasis of human pancreatic
[10] C.A. Stein, J.S. Cohen, Oligodeoxynucleotides as inhibitors of gene expression: a adenocarcinoma cells, Surgery 135 (2004) 555562.
review, Cancer Res. 48 (1988) 26592668. [42] M.S. Duxbury, E. Matros, H. Ito, M.J. Zinner, S.W. Ashley, E.E. Whang, Systemic
[11] E. Zamaratski, P.I. Pradeepkumar, J. Chattopadhyaya, A critical survey of the siRNA-mediated gene silencing: a new approach to targeted therapy of cancer,
structurefunction of the antisense oligo/RNA heteroduplex as substrate for Ann. Surg. 240 (2004) 667674 [discussion 675666].
RNase H, J. Biochem. Biophys. Methods 48 (2001) 189208. [43] S. Filleur, A. Courtin, S. Ait-Si-Ali, J. Guglielmi, C. Merle, A. Harel-Bellan, P.
[12] S.T. Crooke, Molecular mechanisms of action of antisense drugs, Biochim. Clezardin, F. Cabon, SiRNA-mediated inhibition of vascular endothelial growth
Biophys. Acta 1489 (1999) 3144. factor severely limits tumor resistance to antiangiogenic thrombospondin-1 and
[13] D. Castanotto, M. Scherr, J.J. Rossi, Intracellular expression and function of slows tumor vascularization and growth, Cancer Res. 63 (2003) 39193922.
antisense catalytic RNAs, Methods Enzymol. 313 (2000) 401420. [44] Y. Takei, K. Kadomatsu, Y. Yuzawa, S. Matsuo, T. Muramatsu, A small interfering
[14] J.J. Rossi, Ribozymes in the nucleolus, Science 285 (1999) 1685. RNA targeting vascular endothelial growth factor as cancer therapeutics, Cancer
[15] S.W. Santoro, G.F. Joyce, A general purpose RNA-cleaving DNA enzyme, Proc. Natl. Res. 64 (2004) 33653370.
Acad. Sci. U. S. A. 94 (1997) 42624266. [45] S.S. Lakka, C.S. Gondi, N. Yanamandra, W.C. Olivero, D.H. Dinh, M. Gujrati, J.S. Rao,
[16] Y. Wu, L. Yu, R. McMahon, J.J. Rossi, S.J. Forman, D.S. Snyder, Inhibition of bcr-abl Inhibition of cathepsin B and MMP-9 gene expression in glioblastoma cell line via
oncogene expression by novel deoxyribozymes (DNAzymes), Hum. Gene Ther. 10 RNA interference reduces tumor cell invasion, tumor growth and angiogenesis,
(1999) 28472857. Oncogene 23 (2004) 46814689.
[17] T.E. Ichim, M. Li, H. Qian, I.A. Popov, K. Rycerz, X. Zheng, D. White, R. Zhong, W.P. Min, [46] A. Singh, S. Boldin-Adamsky, R.K. Thimmulappa, S.K. Rath, H. Ashush, J. Coulter, A.
RNA interference: a potent tool for gene-specic therapeutics, Am. J. Transplant. Blackford, S.N. Goodman, F. Bunz, W.H. Watson, E. Gabrielson, E. Feinstein, S.
4 (2004) 12271236. Biswal, RNAi-mediated silencing of nuclear factor erythroid-2-related factor 2
[18] S.M. Elbashir, J. Harborth, W. Lendeckel, A. Yalcin, K. Weber, T. Tuschl, Duplexes of gene expression in non-small cell lung cancer inhibits tumor growth and
21-nucleotide RNAs mediate RNA interference in cultured mammalian cells, increases efcacy of chemotherapy, Cancer Res. 68 (2008) 79757984.
Nature 411 (2001) 494498. [47] S. Nakahira, S. Nakamori, M. Tsujie, Y. Takahashi, J. Okami, S. Yoshioka, M.
[19] J. Liu, M.A. Carmell, F.V. Rivas, C.G. Marsden, J.M. Thomson, J.J. Song, S.M. Yamasaki, S. Marubashi, I. Takemasa, A. Miyamoto, Y. Takeda, H. Nagano, K. Dono,
Hammond, L. Joshua-Tor, G.J. Hannon, Argonaute2 is the catalytic engine of K. Umeshita, M. Sakon, M. Monden, Involvement of ribonucleotide reductase M1
mammalian RNAi, Science 305 (2004) 14371441. subunit overexpression in gemcitabine resistance of human pancreatic cancer,
[20] Z. Paroo, D.R. Corey, Challenges for RNAi in vivo, Trends Biotechnol. 22 (2004) Int. J. Cancer 120 (2007) 13551363.
390394. [48] A. Fire, S. Xu, M.K. Montgomery, S.A. Kostas, S.E. Driver, C.C. Mello, Potent and
[21] Y. Aoki, D.P. Cioca, H. Oidaira, J. Kamiya, K. Kiyosawa, RNA interference may be specic genetic interference by double-stranded RNA in Caenorhabditis elegans,
more potent than antisense RNA in human cancer cell lines, Clin. Exp. Pharmacol. Nature 391 (1998) 806811.
Physiol. 30 (2003) 96102. [49] J.Y. Yu, S.L. DeRuiter, D.L. Turner, RNA interference by expression of short-interfering
[22] B.L. Bass, RNA interference. The short answer, Nature 411 (2001) 428429. RNAs and hairpin RNAs in mammalian cells, Proc. Natl. Acad. Sci. U. S. A. 99 (2002)
[23] S. Coma, V. Noe, C. Lavarino, J. Adan, M. Rivas, M. Lopez-Matas, R. Pagan, F. 60476052.
Mitjans, S. Vilaro, J. Piulats, C.J. Ciudad, Use of siRNAs and antisense oligonucleo- [50] M. Miyagishi, K. Taira, U6 promoter-driven siRNAs with four uridine 3 overhangs
tides against survivin RNA to inhibit steps leading to tumor angiogenesis, efciently suppress targeted gene expression in mammalian cells, Nat.
Oligonucleotides 14 (2004) 100113. Biotechnol. 20 (2002) 497500.
[24] M. Miyagishi, M. Hayashi, K. Taira, Comparison of the suppressive effects of [51] A. Jarve, J. Muller, I.H. Kim, K. Rohr, C. MacLean, G. Fricker, U. Massing, F. Eberle, A.
antisense oligonucleotides and siRNAs directed against the same targets in Dalpke, R. Fischer, M.F. Trendelenburg, M. Helm, Surveillance of siRNA integrity
mammalian cells, Antisense Nucleic Acid Drug Dev. 13 (2003) 17. by FRET imaging, Nucleic Acids Res. 35 (2007) e124.
[25] M. Scherr, K. Battmer, T. Winkler, O. Heidenreich, A. Ganser, M. Eder, Specic inhibition [52] J.P. Leonetti, N. Mechti, G. Degols, C. Gagnor, B. Lebleu, Intracellular distribution of
of bcr-abl gene expression by small interfering RNA, Blood 101 (2003) 15661569. microinjected antisense oligonucleotides, Proc. Natl. Acad. Sci. U. S. A. 88 (1991)
[26] T.R. Brummelkamp, R. Bernards, R. Agami, Stable suppression of tumorigenicity 27022706.
by virus-mediated RNA interference, Cancer Cells 2 (2002) 243247. [53] G.B. Robb, K.M. Brown, J. Khurana, T.M. Rana, Specic and potent RNAi in the
[27] L.A. Martinez, I. Naguibneva, H. Lehrmann, A. Vervisch, T. Tchenio, G. Lozano, A. nucleus of human cells, Nat. Struct. Mol. Biol. 12 (2005) 133137.
Harel-Bellan, Synthetic small inhibiting RNAs: efcient tools to inactivate oncogenic [54] S.Y. Berezhna, L. Supekova, F. Supek, P.G. Schultz, A.A. Deniz, siRNA in human cells
mutations and restore p53 pathways, Proc. Natl. Acad. Sci. U. S. A. 99 (2002) selectively localizes to target RNA sites, Proc. Natl. Acad. Sci. U. S. A. 103 (2006)
14,84914,854. 76827687.
D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759 757

[55] B.A. Janowski, K.E. Huffman, J.C. Schwartz, R. Ram, R. Nordsell, D.S. Shames, J.D. [88] Y. Lee, C. Ahn, J. Han, H. Choi, J. Kim, J. Yim, J. Lee, P. Provost, O. Radmark, S. Kim, V.N.
Minna, D.R. Corey, Involvement of AGO1 and AGO2 in mammalian transcriptional Kim, The nuclear RNase III Drosha initiates microRNA processing, Nature 425 (2003)
silencing, Nat. Struct. Mol. Biol. 13 (2006) 787792. 415419.
[56] D.H. Kim, L.M. Villeneuve, K.V. Morris, J.J. Rossi, Argonaute-1 directs siRNA-mediated [89] H. Zhang, F.A. Kolb, V. Brondani, E. Billy, W. Filipowicz, Human Dicer
transcriptional gene silencing in human cells, Nat. Struct. Mol. Biol. 13 (2006) preferentially cleaves dsRNAs at their termini without a requirement for ATP,
793797. EMBO J. 21 (2002) 58755885.
[57] S. Rudel, A. Flatley, L. Weinmann, E. Kremmer, G. Meister, A multifunctional [90] Y. Lee, K. Jeon, J.T. Lee, S. Kim, V.N. Kim, MicroRNA maturation: stepwise
human Argonaute2-specic monoclonal antibody, RNA 14 (2008) 12441253. processing and subcellular localization, EMBO J. 21 (2002) 46634670.
[58] T. Ohrt, J. Mutze, W. Staroske, L. Weinmann, J. Hock, K. Crell, G. Meister, P. Schwille, [91] B.R. Cullen, Transcription and processing of human microRNA precursors, Mol.
Fluorescence correlation spectroscopy and uorescence cross-correlation spectro- Cell 16 (2004) 861865.
scopy reveal the cytoplasmic origination of loaded nuclear RISC in vivo in human [92] R. Yi, Y. Qin, I.G. Macara, B.R. Cullen, Exportin-5 mediates the nuclear export of
cells, Nucleic Acids Res. 36 (2008) 64396449. pre-microRNAs and short hairpin RNAs, Genes Dev. 17 (2003) 30113016.
[59] L. Weinmann, J. Hock, T. Ivacevic, T. Ohrt, J. Mutze, P. Schwille, E. Kremmer, V. [93] E. Lund, S. Guttinger, A. Calado, J.E. Dahlberg, U. Kutay, Nuclear export of
Benes, H. Urlaub, G. Meister, Importin 8 is a gene silencing factor that targets microRNA precursors, Science 303 (2004) 9598.
argonaute proteins to distinct mRNAs, Cell 136 (2009) 496507. [94] Y.S. Lee, K. Nakahara, J.W. Pham, K. Kim, Z. He, E.J. Sontheimer, R.W. Carthew,
[60] S. Guang, A.F. Bochner, D.M. Pavelec, K.B. Burkhart, S. Harding, J. Lachowiec, S. Distinct roles for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA silencing
Kennedy, An Argonaute transports siRNAs from the cytoplasm to the nucleus, pathways, Cell 117 (2004) 6981.
Science 321 (2008) 537541. [95] M. Landthaler, D. Gaidatzis, A. Rothballer, P.Y. Chen, S.J. Soll, L. Dinic, T. Ojo, M. Hafner,
[61] A. Grunweller, C. Gillen, V.A. Erdmann, J. Kurreck, Cellular uptake and localization M. Zavolan, T. Tuschl, Molecular characterization of human Argonaute-containing
of a Cy3-labeled siRNA specic for the serine/threonine kinase Pim-1, ribonucleoprotein complexes and their bound target mRNAs, RNA 14 (2008)
Oligonucleotides 13 (2003) 345352. 25802596.
[62] T. Ohrt, D. Merkle, K. Birkenfeld, C.J. Echeverri, P. Schwille, In situ uorescence [96] S.M. Hammond, S. Boettcher, A.A. Caudy, R. Kobayashi, G.J. Hannon, Argonaute2, a
analysis demonstrates active siRNA exclusion from the nucleus by exportin 5, link between genetic and biochemical analyses of RNAi, Science 293 (2001)
Nucleic Acids Res. 34 (2006) 13691380. 11461150.
[63] Y.L. Chiu, A. Ali, C.Y. Chu, H. Cao, T.M. Rana, Visualizing a correlation between [97] E.J. Sontheimer, R.W. Carthew, Molecular biology. Argonaute journeys into the
siRNA localization, cellular uptake, and RNAi in living cells, Chem. Biol. 11 (2004) heart of RISC, Science 305 (2004) 14091410.
11651175. [98] G. Hutvagner, P.D. Zamore, A microRNA in a multiple-turnover RNAi enzyme
[64] K. Forstemann, Y. Tomari, T. Du, V.V. Vagin, A.M. Denli, D.P. Bratu, C. Klattenhoff, complex, Science 297 (2002) 20562060.
W.E. Theurkauf, P.D. Zamore, Normal microRNA maturation and germ-line stem [99] S. Yekta, I.H. Shih, D.P. Bartel, MicroRNA-directed cleavage of HOXB8 mRNA,
cell maintenance requires Loquacious, a double-stranded RNA-binding domain Science 304 (2004) 594596.
protein, PLoS. Biol. 3 (2005) e236. [100] D.T. Humphreys, B.J. Westman, D.I. Martin, T. Preiss, MicroRNAs control translation
[65] F. Jiang, X. Ye, X. Liu, L. Fincher, D. McKearin, Q. Liu, Dicer-1 and R3D1-L catalyze initiation by inhibiting eukaryotic initiation factor 4E/cap and poly(A) tail function,
microRNA maturation in Drosophila, Genes Dev. 19 (2005) 16741679. Proc. Natl. Acad. Sci. U. S. A. 102 (2005) 16,96116,966.
[66] K. Saito, A. Ishizuka, H. Siomi, M.C. Siomi, Processing of pre-microRNAs by the [101] R.S. Pillai, S.N. Bhattacharyya, C.G. Artus, T. Zoller, N. Cougot, E. Basyuk, E.
Dicer-1-Loquacious complex in Drosophila cells, PLoS. Biol. 3 (2005) e235. Bertrand, W. Filipowicz, Inhibition of translational initiation by Let-7 MicroRNA
[67] X. Liu, J.K. Park, F. Jiang, Y. Liu, D. McKearin, Q. Liu, Dicer-1, but not Loquacious, is in human cells, Science 309 (2005) 15731576.
critical for assembly of miRNA-induced silencing complexes, RNA 13 (2007) [102] R. Thermann, M.W. Hentze, Drosophila miR2 induces pseudo-polysomes and
23242329. inhibits translation initiation, Nature 447 (2007) 875878.
[68] E.P. Murchison, J.F. Partridge, O.H. Tam, S. Chelou, G.J. Hannon, Characterization of [103] M.A. Valencia-Sanchez, J. Liu, G.J. Hannon, R. Parker, Control of translation and
Dicer-decient murine embryonic stem cells, Proc. Natl. Acad. Sci. U. S. A. 102 (2005) mRNA degradation by miRNAs and siRNAs, Genes Dev. 20 (2006) 515524.
12,13512,140. [104] R. Parker, U. Sheth, P bodies and the control of mRNA translation and degradation,
[69] E. Bernstein, A.A. Caudy, S.M. Hammond, G.J. Hannon, Role for a bidentate Mol. Cell 25 (2007) 635646.
ribonuclease in the initiation step of RNA interference, Nature 409 (2001) 363366. [105] K. Okamura, A. Ishizuka, H. Siomi, M.C. Siomi, Distinct roles for Argonaute
[70] P. Provost, D. Dishart, J. Doucet, D. Frendewey, B. Samuelsson, O. Radmark, proteins in small RNA-directed RNA cleavage pathways, Genes Dev. 18 (2004)
Ribonuclease activity and RNA binding of recombinant human Dicer, EMBO J. 16551666.
21 (2002) 58645874. [106] K. Miyoshi, H. Tsukumo, T. Nagami, H. Siomi, M.C. Siomi, Slicer function of
[71] S.M. Hammond, A.A. Caudy, G.J. Hannon, Post-transcriptional gene silencing by Drosophila Argonautes and its involvement in RISC formation, Genes Dev. 19 (2005)
double-stranded RNA, Nat. Rev., Genet. 2 (2001) 110119. 28372848.
[72] M.A. Carmell, G.J. Hannon, RNase III enzymes and the initiation of gene silencing, [107] Y. Tomari, T. Du, P.D. Zamore, Sorting of Drosophila small silencing RNAs, Cell
Nat. Struct. Mol. Biol. 11 (2004) 214218. 130 (2007) 299308.
[73] I.J. Macrae, K. Zhou, F. Li, A. Repic, A.N. Brooks, W.Z. Cande, P.D. Adams, J.A. [108] K. Forstemann, M.D. Horwich, L. Wee, Y. Tomari, P.D. Zamore, Drosophila
Doudna, Structural basis for double-stranded RNA processing by Dicer, Science microRNAs are sorted into functionally distinct argonaute complexes after
311 (2006) 195198. production by dicer-1, Cell 130 (2007) 287297.
[74] K.H. Kok, M.H. Ng, Y.P. Ching, D.Y. Jin, Human TRBP and PACT directly interact with [109] F.A. Steiner, S.W. Hoogstrate, K.L. Okihara, K.L. Thijssen, R.F. Ketting, R.H. Plasterk,
each other and associate with dicer to facilitate the production of small T. Sijen, Structural features of small RNA precursors determine Argonaute loading
interfering RNA, J. Biol. Chem. 282 (2007) 17,64917,657. in Caenorhabditis elegans, Nat. Struct. Mol. Biol. 14 (2007) 927933.
[75] G.B. Robb, T.M. Rana, RNA helicase A interacts with RISC in human cells and [110] A. Azuma-Mukai, H. Oguri, T. Mituyama, Z.R. Qian, K. Asai, H. Siomi, M.C. Siomi,
functions in RISC loading, Mol. Cell 26 (2007) 523537. Characterization of endogenous human Argonautes and their miRNA partners in
[76] C. Matranga, Y. Tomari, C. Shin, D.P. Bartel, P.D. Zamore, Passengerstrand RNA silencing, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 79647969.
cleavage facilitates assembly of siRNA into Ago2-containing RNAi enzyme [111] D.G. Hendrickson, D.J. Hogan, D. Herschlag, J.E. Ferrell, P.O. Brown, Systematic
complexes, Cell 123 (2005) 607620. identication of mRNAs recruited to argonaute 2 by specic microRNAs and
[77] T.A. Rand, S. Petersen, F. Du, X. Wang, Argonaute2 cleaves the anti-guide strand of corresponding changes in transcript abundance, PLoS ONE 3 (2008) e2126.
siRNA during RISC activation, Cell 123 (2005) 621629. [112] Y. Tay, J. Zhang, A.M. Thomson, B. Lim, I. Rigoutsos, MicroRNAs to Nanog, Oct4 and Sox2
[78] P.J. Leuschner, S.L. Ameres, S. Kueng, J. Martinez, Cleavage of the siRNA passenger coding regions modulate embryonic stem cell differentiation, Nature 455 (2008)
strand during RISC assembly in human cells, EMBO Rep. 7 (2006) 314320. 11241128.
[79] J.B. Preall, E.J. Sontheimer, RNAi: RISC gets loaded, Cell 123 (2005) 543545. [113] M. Sano, M. Sierant, M. Miyagishi, M. Nakanishi, Y. Takagi, S. Sutou, Effect of
[80] B. Haley, P.D. Zamore, Kinetic analysis of the RNAi enzyme complex, Nat. Struct. asymmetric terminal structures of short RNA duplexes on the RNA interference
Mol. Biol. 11 (2004) 599606. activity and strand selection, Nucleic Acids Res. 36 (2008) 58125821.
[81] R.I. Gregory, T.P. Chendrimada, N. Cooch, R. Shiekhattar, Human RISC couples [114] D.H. Kim, M.A. Behlke, S.D. Rose, M.S. Chang, S. Choi, J.J. Rossi, Synthetic dsRNA Dicer
microRNA biogenesis and posttranscriptional gene silencing, Cell 123 (2005) substrates enhance RNAi potency and efcacy, Nat. Biotechnol. 23 (2005) 222226.
631640. [115] M.A. McAnuff, G.R. Rettig, K.G. Rice, Potency of siRNA versus shRNA mediated
[82] E. Maniataki, Z. Mourelatos, A human, ATP-independent, RISC assembly machine knockdown in vivo, J. Pharm. Sci. 96 (2007) 29222930.
fueled by pre-miRNA, Genes Dev. 19 (2005) 29792990. [116] D. Siolas, C. Lerner, J. Burchard, W. Ge, P.S. Linsley, P.J. Paddison, G.J. Hannon, M.A.
[83] R.E. Collins, X. Cheng, Structural and biochemical advances in mammalian RNAi, Cleary, Synthetic shRNAs as potent RNAi triggers, Nat. Biotechnol. 23 (2005) 227231.
J. Cell. Biochem. 99 (2006) 12511266. [117] A.V. Vlassov, B. Korba, K. Farrar, S. Mukerjee, A.A. Seyhan, H. Ilves, R.L. Kaspar, D.
[84] R.L. Boudreau, A.M. Monteys, B.L. Davidson, Minimizing variables among hairpin- Leake, S.A. Kazakov, B.H. Johnston, shRNAs targeting hepatitis C: effects of
based RNAi vectors reveals the potency of shRNAs, RNA 14 (2008) 18341844. sequence and structural features, and comparision with siRNA, Oligonucleotides
[85] B.R. Cullen, RNAi the natural way, Nat. Genet. 37 (2005) 11631165. 17 (2007) 223236.
[86] Y. Zeng, R. Yi, B.R. Cullen, Recognition and cleavage of primary microRNA [118] M. Gossen, H. Bujard, Tight control of gene expression in mammalian cells by
precursors by the nuclear processing enzyme Drosha, EMBO J. 24 (2005) tetracycline-responsive promoters, Proc. Natl. Acad. Sci. U. S. A. 89 (1992) 55475551.
138148. [119] S. Gupta, R.A. Schoer, J.E. Egan, G.J. Hannon, V. Mittal, Inducible, reversible, and stable
[87] J.M. Silva, M.Z. Li, K. Chang, W. Ge, M.C. Golding, R.J. Rickles, D. Siolas, G. Hu, P.J. RNA interference in mammalian cells, Proc. Natl. Acad. Sci. U. S. A. 101 (2004)
Paddison, M.R. Schlabach, N. Sheth, J. Bradshaw, J. Burchard, A. Kulkarni, G. Cavet, 19271932.
R. Sachidanandam, W.R. McCombie, M.A. Cleary, S.J. Elledge, G.J. Hannon, Second- [120] R.A. Dickins, M.T. Hemann, J.T. Zilfou, D.R. Simpson, I. Ibarra, G.J. Hannon, S.W.
generation shRNA libraries covering the mouse and human genomes, Nat. Genet. Lowe, Probing tumor phenotypes using stable and regulated synthetic microRNA
37 (2005) 12811288. precursors, Nat. Genet. 37 (2005) 12891295.
758 D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759

[121] A.W. Tong, Y.A. Zhang, J. Nemunaitis, Small interfering RNA for experimental [153] M. Aleku, P. Schulz, O. Keil, A. Santel, U. Schaeper, B. Dieckhoff, O. Janke, J. Endruschat,
cancer therapy, Curr. Opin. Mol. Ther. 7 (2005) 114124. B. Durieux, N. Roder, K. Lofer, C. Lange, M. Fechtner, K. Mopert, G. Fisch, S. Dames, W.
[122] A. Aigner, Cellular delivery in vivo of siRNA-based therapeutics, Curr. Pharm. Des. Arnold, K. Jochims, K. Giese, B. Wiedenmann, A. Scholz, J. Kaufmann, Atu027, a
14 (2008) 36033619. liposomal small interfering RNA formulation targeting protein kinase N3, inhibits
[123] A.W. Tong, C.M. Jay, N. Senzer, P.B. Maples, J. Nemunaitis, Systemic therapeutic gene cancer progression, Cancer Res. 68 (2008) 97889798.
delivery for cancer: crafting Paris' arrow, Curr. Gene Ther. 9 (1) (2009 Feb) 4560. [154] J.S. Vorhies, J.J. Nemunaitis, Synthetic vs. natural/biodegradable polymers for
[124] J.S. Vorhies, J. Nemunaitis, Nonviral delivery vehicles for use in short hairpin RNA- delivery of shRNA-based cancer therapies, Methods Mol. Biol. 480 (2009) 1129.
based cancer therapies, Expert Rev. Anticancer Ther. 7 (2007) 373382. [155] P. Kumar, H.S. Ban, S.S. Kim, H. Wu, T. Pearson, D.L. Greiner, A. Laouar, J. Yao, V.
[125] A.P. McCaffrey, L. Meuse, T.T. Pham, D.S. Conklin, G.J. Hannon, M.A. Kay, RNA Haridas, K. Habiro, Y.G. Yang, J.H. Jeong, K.Y. Lee, Y.H. Kim, S.W. Kim, M. Peipp, G.H.
interference in adult mice, Nature 418 (2002) 3839. Fey, N. Manjunath, L.D. Shultz, S.K. Lee, P. Shankar, T cell-specic siRNA delivery
[126] W.M. Merritt, Y.G. Lin, L.Y. Han, A.A. Kamat, W.A. Spannuth, R. Schmandt, D. Urbauer, suppresses HIV-1 infection in humanized mice, Cell 134 (2008 Dec 30) 577586.
L.A. Pennacchio, J.F. Cheng, A.M. Nick, M.T. Deavers, A. Mourad-Zeidan, H. Wang, P. [156] K. Gao, L. Huang, Nonviral methods for siRNA Delivery, Mol. Pharmacol. 2008 Dec 30.
Mueller, M.E. Lenburg, J.W. Gray, S. Mok, M.J. Birrer, G. Lopez-Berestein, R.L. Coleman, [157] A. Akinc, A. Zumbuehl, M. Goldberg, E.S. Leshchiner, V. Busini, N. Hossain, S.A.
M. Bar-Eli, A.K. Sood, Dicer, Drosha, and outcomes in patients with ovarian cancer, N. Bacallado, D.N. Nguyen, J. Fuller, R. Alvarez, A. Borodovsky, T. Borland, R. Constien,
Engl. J. Med. 359 (2008) 26412650. A. de Fougerolles, J.R. Dorkin, K. Narayanannair Jayaprakash, M. Jayaraman, M.
[127] J. Lu, G. Getz, E.A. Miska, E. Alvarez-Saavedra, J. Lamb, D. Peck, A. Sweet-Cordero, B.L. John, V. Koteliansky, M. Manoharan, L. Nechev, J. Qin, T. Racie, D. Raitcheva, K.G.
Ebert, R.H. Mak, A.A. Ferrando, J.R. Downing, T. Jacks, H.R. Horvitz, T.R. Golub, Rajeev, D.W. Sah, J. Soutschek, I. Toudjarska, H.P. Vornlocher, T.S. Zimmermann, R.
MicroRNA expression proles classify human cancers, Nature 435 (2005) 834838. Langer, D.G. Anderson, A combinatorial library of lipid-like materials for delivery
[128] R. Sandberg, J.R. Neilson, A. Sarma, P.A. Sharp, C.B. Burge, Proliferating cells of RNAi therapeutics, Nat. Biotechnol. 26 (2008) 561569.
express mRNAs with shortened 3 untranslated regions and fewer microRNA [158] S. Ghatak, V.C. Hascall, F.G. Berger, M.M. Penas, C. Davis, E. Jabari, X. He, J.S. Norris,
target sites, Science 320 (2008) 16431647. Y. Dang, R.R. Markwald, S. Misra, Tissue-specic shRNA delivery: a novel
[129] S. Chiosea, E. Jelezcova, U. Chandran, M. Acquafondata, T. McHale, R.W. Sobol, R. approach for gene therapy in cancer, Connect. Tissue Res. 49 (2008) 265269.
Dhir, Up-regulation of dicer, a component of the MicroRNA machinery, in prostate [159] P. Kumar, H. Wu, J.L. McBride, K.E. Jung, M.H. Kim, B.L. Davidson, S.K. Lee, P.
adenocarcinoma, Am. J. Pathol. 169 (2006) 18121820. Shankar, N. Manjunath, Transvascular delivery of small interfering RNA to the
[130] N. Sugito, H. Ishiguro, Y. Kuwabara, M. Kimura, A. Mitsui, H. Kurehara, T. Ando, R. central nervous system, Nature 448 (2007) 3943.
Mori, N. Takashima, R. Ogawa, Y. Fujii, RNASEN regulates cell proliferation and affects [160] A. Judge, I. MacLachlan, Overcoming the innate immune response to small
survival in esophageal cancer patients, Clin. Cancer Res. 12 (2006) 73227328. interfering RNA, Hum. Gene Ther. 19 (2008) 111124.
[131] S. Tokumaru, M. Suzuki, H. Yamada, M. Nagino, T. Takahashi, Let-7 regulates Dicer [161] H. Lv, S. Zhang, B. Wang, S. Cui, J. Yan, Toxicity of cationic lipids and cationic
expression and constitutes a negative feedback loop, Carcinogenesis 29 (2008) polymers in gene delivery, J. Control. Release 114 (2006) 100109.
20732077. [162] T.G. Park, J.H. Jeong, S.W. Kim, Current status of polymeric gene delivery systems,
[132] J.J. Forman, A. Legesse-Miller, H.A. Coller, A search for conserved sequences in Adv. Drug Deliv. Rev. 58 (2006) 467486.
coding regions reveals that the let-7 microRNA targets Dicer within its coding [163] M. Breunig, U. Lungwitz, R. Liebl, A. Goepferich, Breaking up the correlation between
sequence, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 14,87914,884. efcacy and toxicity for nonviral gene delivery, Proc. Natl. Acad. Sci. U. S. A.104 (2007)
[133] Breakdown of RNAi-Based Drugs in the Clinic, RNAi News, vol. 6, 2008. 14,45414,459.
[134] J.D. Heidel, J.Y. Liu, Y. Yen, B. Zhou, B.S. Heale, J.J. Rossi, D.W. Bartlett, M.E. Davis, [164] A.L. Jackson, S.R. Bartz, J. Schelter, S.V. Kobayashi, J. Burchard, M. Mao, B. Li, G.
Potent siRNA inhibitors of ribonucleotide reductase subunit RRM2 reduce cell Cavet, P.S. Linsley, Expression proling reveals off-target gene regulation by RNAi,
proliferation in vitro and in vivo, Clin. Cancer Res. 13 (2007) 22072215. Nat. Biotechnol. 21 (2003) 635637.
[135] R. Zukiel, S. Nowak, E. Wyszko, K. Rolle, I. Gawronska, M.Z. Barciszewska, J. [165] A.L. Jackson, J. Burchard, J. Schelter, B.N. Chau, M. Cleary, L. Lim, P.S. Linsley,
Barciszewski, Suppression of human brain tumor with interference RNA specic Widespread siRNA off-target transcript silencing mediated by seed region
for tenascin-C, Cancer Biol. Ther. 5 (2006) 10021007. sequence complementarity, RNA 12 (2006) 11791187.
[136] J.D. Heidel, Z. Yu, J.Y. Liu, S.M. Rele, Y. Liang, R.K. Zeidan, D.J. Kornbrust, M.E. Davis, [166] X. Lin, X. Ruan, M.G. Anderson, J.A. McDowell, P.E. Kroeger, S.W. Fesik, Y. Shen,
Administration in non-human primates of escalating intravenous doses of siRNA-mediated off-target gene silencing triggered by a 7 nt complementation,
targeted nanoparticles containing ribonucleotide reductase subunit M2 siRNA, Nucleic Acids Res. 33 (2005) 45274535.
Proc. Natl. Acad. Sci. U. S. A. 104 (2007) 57155721. [167] A.L. Jackson, J. Burchard, D. Leake, A. Reynolds, J. Schelter, J. Guo, J.M. Johnson, L.
[137] E. Check, A tragic setback, Nature 420 (2002) 116118. Lim, J. Karpilow, K. Nichols, W. Marshall, A. Khvorova, P.S. Linsley, Position-
[138] T. Nguyen, E.M. Menocal, J. Harborth, J.H. Fruehauf, RNAi therapeutics: an update specic chemical modication of siRNAs reduces off-target transcript silencing,
on delivery, Curr. Opin. Mol. Ther. 10 (2008) 158167. RNA 12 (2006) 11971205.
[139] J. Luten, C.F. van Nostrum, S.C. De Smedt, W.E. Hennink, Biodegradable polymers as [168] A. Birmingham, E.M. Anderson, A. Reynolds, D. Ilsley-Tyree, D. Leake, Y. Fedorov, S.
non-viral carriers for plasmid DNA delivery, J. Control. Release 126 (2008) 97110. Baskerville, E. Maksimova, K. Robinson, J. Karpilow, W.S. Marshall, A. Khvorova, 3
[140] S. Zhang, B. Zhao, H. Jiang, B. Wang, B. Ma, Cationic lipids and polymers mediated UTR seed matches, but not overall identity, are associated with RNAi off-targets, Nat.
vectors for delivery of siRNA, J. Control. Release 123 (2007) 110. Methods 3 (2006) 199204.
[141] A. Aigner, Nonviral in vivo delivery of therapeutic small interfering RNAs, Curr. [169] E. Anderson, Q. Boese, A. Khvorova, J. Karpilow, Identifying siRNA-induced off-
Opin. Mol. Ther. 9 (2007) 345352. targets by microarray analysis, Methods Mol. Biol. 442 (2008) 4563.
[142] J.A. Hughes, G.A. Rao, Targeted polymers for gene delivery, Expert Opin. Drug [170] S. van Dongen, C. Abreu-Goodger, A.J. Enright, Detecting microRNA binding and
Deliv. 2 (2005) 145157. siRNA off-target effects from expression data, Nat. Methods 5 (2008) 10231025.
[143] W.T. Godbey, M.A. Barry, P. Saggau, K.K. Wu, A.G. Mikos, Poly(ethylenimine)- [171] S. Qiu, C.M. Adema, T. Lane, A computational study of off-target effects of RNA
mediated transfection: a new paradigm for gene delivery, J. Biomed. Mater. Res. interference, Nucleic Acids Res. 33 (2005) 18341847.
51 (2000) 321328. [172] Y.K. Park, S.M. Park, Y.C. Choi, D. Lee, M. Won, Y.J. Kim, AsiDesigner: exon-based
[144] M. Thomas, A.M. Klibanov, Enhancing polyethylenimine's delivery of plasmid siRNA design server considering alternative splicing, Nucleic Acids Res. 36 (2008)
DNA into mammalian cells, Proc. Natl. Acad. Sci. U. S. A. 99 (2002) 14,64014,645. W97103.
[145] Y. Kim, M. Tewari, J.D. Pajerowski, S. Cai, S. Sen, J. Williams, S. Sirsi, G. Lutz, D.E. [173] T. Yamada, S. Morishita, Accelerated off-target search algorithm for siRNA,
Discher, Polymersome delivery of siRNA and antisense oligonucleotides, J. Control. Bioinformatics 21 (2005) 13161324.
Release 134 (2) (2009 Mar 4) 132140. [174] Y. Naito, T. Yamada, T. Matsumiya, K. Ui-Tei, K. Saigo, S. Morishita, dsCheck: highly
[146] J.D. Heidel, Linear cyclodextrin-containing polymers and their use as delivery sensitive off-target search software for double-stranded RNA-mediated RNA
agents, Expert Opin. Drug Deliv. 3 (2006) 641646. interference, Nucleic Acids Res. 33 (2005) W589591.
[147] J.N. Moreira, A. Santos, V. Moura, M.C. Pedroso de Lima, S. Simoes, Non-viral lipid- [175] Q. Du, H. Thonberg, J. Wang, C. Wahlestedt, Z. Liang, A systematic analysis of the
based nanoparticles for targeted cancer systemic gene silencing, J. Nanosci. silencing effects of an active siRNA at all single-nucleotide mismatched target
Nanotechnol. 8 (2008) 21872204. sites, Nucleic Acids Res. 33 (2005) 16711677.
[148] N. Blow, Small RNAs: delivering the future, Nature 450 (2007) 11171120. [176] D.S. Schwarz, H. Ding, L. Kennington, J.T. Moore, J. Schelter, J. Burchard, P.S. Linsley,
[149] T.S. Zimmermann, A.C. Lee, A. Akinc, B. Bramlage, D. Bumcrot, M.N. Fedoruk, J. N. Aronin, Z. Xu, P.D. Zamore, Designing siRNA that distinguish between genes
Harborth, J.A. Heyes, L.B. Jeffs, M. John, A.D. Judge, K. Lam, K. McClintock, L.V. that differ by a single nucleotide, PLoS Genet. 2 (2006) e140.
Nechev, L.R. Palmer, T. Racie, I. Rohl, S. Seiffert, S. Shanmugam, V. Sood, J. [177] C. Dahlgren, H.Y. Zhang, Q. Du, M. Grahn, G. Norstedt, C. Wahlestedt, Z. Liang,
Soutschek, I. Toudjarska, A.J. Wheat, E. Yaworski, W. Zedalis, V. Koteliansky, M. Analysis of siRNA specicity on targets with double-nucleotide mismatches,
Manoharan, H.P. Vornlocher, I. MacLachlan, RNAi-mediated gene silencing in Nucleic Acids Res. 36 (2008) e53.
non-human primates, Nature 441 (2006) 111114. [178] J.K. Watts, G.F. Deleavey, M.J. Damha, Chemically modied siRNA: tools and
[150] A. Santel, M. Aleku, O. Keil, J. Endruschat, V. Esche, B. Durieux, K. Lofer, M. applications, Drug Discov. Today 13 (2008) 842855.
Fechtner, T. Rohl, G. Fisch, S. Dames, W. Arnold, K. Giese, A. Klippel, J. Kaufmann, [179] O. Snove Jr., J.J. Rossi, Chemical modications rescue off-target effects of RNAi,
RNA interference in the mouse vascular endothelium by systemic administration ACS Chem. Biol. 1 (2006) 274276.
of siRNA-lipoplexes for cancer therapy, Gene Ther. 13 (2006) 13601370. [180] M.A. Behlke, Chemical modication of siRNAs for in vivo use, Oligonucleotides
[151] A. Santel, M. Aleku, O. Keil, J. Endruschat, V. Esche, G. Fisch, S. Dames, K. Lofer, M. 18 (2008) 305319.
Fechtner, W. Arnold, K. Giese, A. Klippel, J. Kaufmann, A novel siRNA-lipoplex [181] J.B. Bramsen, M.B. Laursen, C.K. Damgaard, S.W. Lena, B.R. Babu, J. Wengel, J. Kjems,
technology for RNA interference in the mouse vascular endothelium, Gene Ther. Improved silencing properties using small internally segmented interfering RNAs,
13 (2006) 12221234. Nucleic Acids Res. 35 (2007) 58865897.
[152] M. Aleku, G. Fisch, K. Mopert, O. Keil, W. Arnold, J. Kaufmann, A. Santel, [182] P.Y. Chen, L. Weinmann, D. Gaidatzis, Y. Pei, M. Zavolan, T. Tuschl, G. Meister,
Intracellular localization of lipoplexed siRNA in vascular endothelial cells of Strand-specic 5-O-methylation of siRNA duplexes controls guide strand
different mouse tissues, Microvasc. Res. 76 (2008) 3141. selection and targeting specicity, RNA 14 (2008) 263274.
D.D. Rao et al. / Advanced Drug Delivery Reviews 61 (2009) 746759 759

[183] K. Ui-Tei, Y. Naito, S. Zenno, K. Nishi, K. Yamato, F. Takahashi, A. Juni, K. Saigo, [199] H. Hemmi, O. Takeuchi, T. Kawai, T. Kaisho, S. Sato, H. Sanjo, M. Matsumoto, K.
Functional dissection of siRNA sequence by systematic DNA substitution: modied Hoshino, H. Wagner, K. Takeda, S. Akira, A toll-like receptor recognizes bacterial
siRNA with a DNA seed arm is a powerful tool for mammalian gene silencing with DNA, Nature 408 (2000) 740745.
signicantly reduced off-target effect, Nucleic Acids Res. 36 (2008) 21362151. [200] H.K. de Wolf, N. Johansson, A.T. Thong, C.J. Snel, E. Mastrobattista, W.E. Hennink,
[184] D. Rao, N. Senzer, M.A. Cleary, J. Nemunaitis, Comparative assessment of siRNA G. Storm, Plasmid CpG depletion improves degree and duration of tumor gene
and shRNA off target effects: what is slowing clinical development. Cancer Gene expression after intravenous administration of polyplexes, Pharm. Res. 25 (2008)
Therapy (2009). 16541662.
[185] D. Bumcrot, M. Manoharan, V. Koteliansky, D.W. Sah, RNAi therapeutics: a [201] M.A. Robbins, M. Li, I. Leung, H. Li, D.V. Boyer, Y. Song, M.A. Behlke, J.J. Rossi, Stable
potential new class of pharmaceutical drugs, Nat. Chem. Biol. 2 (2006) 711719. expression of shRNAs in human CD34+ progenitor cells can avoid induction of
[186] A.J. Bridge, S. Pebernard, A. Ducraux, A.L. Nicoulaz, R. Iggo, Induction of an interferon responses to siRNAs in vitro, Nat. Biotechnol. 24 (2006) 566571.
interferon response by RNAi vectors in mammalian cells, Nat. Genet. 34 (2003) [202] M. Sioud, Induction of inammatory cytokines and interferon responses by
263264. double-stranded and single-stranded siRNAs is sequence-dependent and
[187] C.A. Sledz, M. Holko, M.J. de Veer, R.H. Silverman, B.R. Williams, Activation of the requires endosomal localization, J. Mol. Biol. 348 (2005) 10791090.
interferon system by short-interfering RNAs, Nat. Cell Biol. 5 (2003) 834839. [203] H. Akashi, M. Miyagishi, T. Yokota, T. Watanabe, T. Hino, K. Nishina, M. Kohara, K.
[188] K. Kariko, P. Bhuyan, J. Capodici, D. Weissman, Small interfering RNAs mediate Taira, Escape from the interferon response associated with RNA interference using
sequence-independent gene suppression and induce immune activation by vectors that encode long modied hairpin-RNA, Mol. Biosyst. 1 (2005) 382390.
signaling through toll-like receptor 3, J. Immunol. 172 (2004) 65456549. [204] T. Watanabe, M. Sudoh, M. Miyagishi, H. Akashi, M. Arai, K. Inoue, K. Taira, M.
[189] V. Hornung, M. Guenthner-Biller, C. Bourquin, A. Ablasser, M. Schlee, S. Uematsu, Yoshiba, M. Kohara, Intracellular-diced dsRNA has enhanced efcacy for silencing
A. Noronha, M. Manoharan, S. Akira, A. de Fougerolles, S. Endres, G. Hartmann, HCV RNA and overcomes variation in the viral genotype, Gene Ther. 13 (2006)
Sequence-specic potent induction of IFN-alpha by short interfering RNA in 883892.
plasmacytoid dendritic cells through TLR7, Nat. Med. 11 (2005) 263270. [205] M. Bauer, N. Kinkl, A. Meixner, E. Kremmer, M. Riemenschneider, H. Forstl, T.
[190] A.D. Judge, V. Sood, J.R. Shaw, D. Fang, K. McClintock, I. MacLachlan, Sequence- Gasser, M. Uefng, Prevention of interferon-stimulated gene expression using
dependent stimulation of the mammalian innate immune response by synthetic microRNA-designed hairpins, Gene Ther. 16 (2009) 142147.
siRNA, Nat. Biotechnol. 23 (2005) 457462. [206] M. Sioud, Single-stranded small interfering RNA are more immunostimulatory
[191] S.S. Diebold, T. Kaisho, H. Hemmi, S. Akira, C. Reis e Sousa, Innate antiviral than their double-stranded counterparts: a central role for 2-hydroxyl uridines
responses by means of TLR7-mediated recognition of single-stranded RNA, in immune responses, Eur. J. Immunol. 36 (2006) 12221230.
Science 303 (2004) 15291531. [207] J.M. Layzer, A.P. McCaffrey, A.K. Tanner, Z. Huang, M.A. Kay, B.A. Sullenger, In vivo
[192] F. Heil, H. Hemmi, H. Hochrein, F. Ampenberger, C. Kirschning, S. Akira, G. Lipford, activity of nuclease-resistant siRNAs, RNA 10 (2004) 766771.
H. Wagner, S. Bauer, Species-specic recognition of single-stranded RNA via toll- [208] K. Kariko, M. Buckstein, H. Ni, D. Weissman, Suppression of RNA recognition by
like receptor 7 and 8, Science 303 (2004) 15261529. Toll-like receptors: the impact of nucleoside modication and the evolutionary
[193] M. Karin, Y. Yamamoto, Q.M. Wang, The IKK NF-kappa B system: a treasure trove origin of RNA, Immunity 23 (2005) 165175.
for drug development, Nat. Rev., Drug Discov. 3 (2004) 1726. [209] D. Grimm, K.L. Streetz, C.L. Jopling, T.A. Storm, K. Pandey, C.R. Davis, P. Marion, F.
[194] J.T. Marques, T. Devosse, D. Wang, M. Zamanian-Daryoush, P. Serbinowski, R. Salazar, M.A. Kay, Fatality in mice due to oversaturation of cellular microRNA/short
Hartmann, T. Fujita, M.A. Behlke, B.R. Williams, A structural basis for hairpin RNA pathways, Nature 441 (2006) 537541.
discriminating between self and nonself double-stranded RNAs in mammalian [210] D. Grimm, M.A. Kay, Therapeutic application of RNAi: is mRNA targeting nally
cells, Nat. Biotechnol. 24 (2006) 559565. ready for prime time? J. Clin. Invest. 117 (2007) 36333641.
[195] M. Sioud, RNA interference and innate immunity, Adv. Drug Deliv. Rev. 59 (2007) [211] M. John, R. Constien, A. Akinc, M. Goldberg, Y.A. Moon, M. Spranger, P. Hadwiger, J.
153163. Soutschek, H.P. Vornlocher, M. Manoharan, M. Stoffel, R. Langer, D.G. Anderson, J.D.
[196] M.E. Kleinman, K. Yamada, A. Takeda, V. Chandrasekaran, M. Nozaki, J.Z. Baf, R.J. Horton, V. Koteliansky, D. Bumcrot, Effective RNAi-mediated gene silencing without
Albuquerque, S. Yamasaki, M. Itaya, Y. Pan, B. Appukuttan, D. Gibbs, Z. Yang, K. interruption of the endogenous microRNA pathway, Nature 449 (2007) 745747.
Kariko, B.K. Ambati, T.A. Wilgus, L.A. DiPietro, E. Sakurai, K. Zhang, J.R. Smith, E.W. [212] R. Yi, B.P. Doehle, Y. Qin, I.G. Macara, B.R. Cullen, Overexpression of exportin 5
Taylor, J. Ambati, Sequence- and target-independent angiogenesis suppression by enhances RNA interference mediated by short hairpin RNAs and microRNAs, RNA
siRNA via TLR3, Nature 452 (2008) 591597. 11 (2005) 220226.
[197] M. Robbins, A. Judge, E. Ambegia, C. Choi, E. Yaworski, L. Palmer, K. McClintock, I. [213] J.C. Giering, D. Grimm, T.A. Storm, M.A. Kay, Expression of shRNA from a tissue-
Maclachlan, Misinterpreting the therapeutic effects of siRNA caused by immune specic pol II promoter is an effective and safe RNAi therapeutic, Mol. Ther.16 (2008)
stimulation, Hum. Gene Ther. 19 (10) (2008 Oct) 991999. 16301636.
[198] S. Bauer, C.J. Kirschning, H. Hacker, V. Redecke, S. Hausmann, S. Akira, H. Wagner,
G.B. Lipford, Human TLR9 confers responsiveness to bacterial DNA via species-
specic CpG motif recognition, Proc. Natl. Acad. Sci. U. S. A. 98 (2001) 92379242.

Você também pode gostar