throbber
578
`
`Mini-Reviews in Medicinal Chemistry, 2010, 10, 578-595
`
`Recent Progress in Chemically Modified siRNAs
`
`M. Gaglione and A. Messere*
`
`Department of Environmental Sciences, Second University of Naples, via Vivaldi 43, 81100, Caserta, Italy
`
`Abstract: RNA interference technology has become a powerful laboratory tool to study gene function. Small interfering
`RNAs (siRNAs) have provided unprecedented opportunities for the development of new therapeutics in human diseases.
`Unfortunately, siRNA duplexes are not optimal drug-like molecules. The problems for their effective application are fun-
`damentally delivery, stability and off-target effects. Chemical modification provides solutions to many of the challenges
`facing siRNA therapeutics. In this review, we recapitulate and discuss the development of the latest described chemical
`modifications of siRNAs, with a special focus on novel chemical modifications of siRNA structure, architecture and
`siRNA conjugates.
`
`Keywords: RNAi, siRNAs, chemical modification, siRNA conjugates.
`
`INTRODUCTION
`
` The completion of human genome sequencing project
`and the elucidation of many molecular pathways that are
`important in diseases have made the use of nucleic acid-
`based inhibitors of gene expression an immense attractive-
`ness in functional genomics and therapeutic strategies. The
`increase in Oligonucleotide-Mediated Gene Silencing [1]
`(OMGS) is largely a result of improvements in the rational
`design of ONs and in the methods used for synthesizing oli-
`gonucleotides with relatively inexpensive cost. The concept
`underlying nucleic acids based technology is relatively
`straightforward: the use of a sequence that recognizes a spe-
`cific mRNA through Watson-Crick base pair hybridization
`inhibits gene expression at either the transcriptional or post-
`transcriptional level [2]. The major classes of inhibitory
`agents are Antisense Oligonucleotides (AS-ONs or ASOs)
`and more recently Small Interfering RNA (siRNAs).
`
`From Antisense to siRNA
`
` The notion that gene expression could be modified
`through the use of small ODNs derives from studies by
`Paterson et al. [3], who first used singlestranded DNA to
`inhibit translation of a complementary RNA in a cell-free
`system in 1977. The following year, Zamecnik and Stephen-
`son [1, 4] showed that a short (13-mer) DNA that was AN-
`TISENSE to the Rous sarcoma virus could inhibit viral rep-
`lication in culture. In the mid 1980s, the existence of natu-
`rally occurring antisense RNAs and their role in regulating
`gene expression was shown [5, 6]. Since then, the antisense
`strategy has enjoyed exponential gains in interest and has
`been the subject of thousands of published reports. The basic
`concept and the mechanisms of inhibition of AS-ONs have
`been characterized and discerned in two main mechanisms:
`steric-blockage and RNase-H activation
`[7, 8]. The
`
`*Address correspondence to this author at the Department of Environmental
`Sciences, Second University of Naples, via Vivaldi 43, 81100, Caserta,
`Italy; Tel: +39-(0)823-274602; Fax: +39-(0)823-274605;
`E-mail: anna.messere@unina2.it
`
`steric-blocker AS-ONs physically prevent or inhibit the pro-
`gression of splicing or the translation machinery, while the
`RNase H dependent oligonucleotides induce the degradation
`of mRNA. Most of the oligonucleotides capable of inhibiting
`splicing are not RNase-H dependent [9-11]. In spite of the
`theoretical simplicity, the molecular mechanism of action of
`AS-ONs has critical drawbacks such as poor stability versus
`nuclease activity in vitro and in vivo, low intracellular pene-
`tration and low bioavailability [12, 13]. Synthetic nucleic
`acids can be chemically modified to improve their general
`pharmacodynamic properties for their therapeutic use. Hun-
`dreds of different modifications have been studied for utility
`in antisense applications [14-16]. Three generations of
`chemically modified ASOs have been developed to enhance
`nuclease resistance, prolong tissue half-life, reduce off-target
`effects and increase affinity and potency [7, 17-24]. The
`most notable discovery was the introduction of a phos-
`phorothioate backbone in oligonucleotides, leading to a sig-
`nificant increase in their stability without major changes in
`ability to hybridize their target mRNA [7]. Other chemical
`modifications, including the development of DNA/RNA
`mixed-backbone oligonucleotides [25] and other oligonu-
`cleotides barely resembling DNA (such as peptide nucleic
`acid (PNA) and locked nucleic acid (LNA) structures) [7],
`have been made to increase the efficacy and stability of the
`antisense molecules. Although a few first generation ASOs
`have shown promising therapeutic performances, newer gen-
`erations have entered into clinical trials.
`
`RNA interference (RNAi) is the most recent explotion of
`interest in antisense world following the discoveries of Mello
`and colleagues [26] that double-stranded RNAs (dsRNAs)
`elicit potent degradation of targeted mRNA sequences in C.
`elegans and in mammalian cells [27, 28]. The active compo-
`nents of the RNAi are dsRNAs, small interfering RNA (siR-
`NAs), that tipically contain 19-21 bp and 2-nt 3’-overhangs.
`These short species are naturally produced by Dicer- medi-
`ated cleavage of larger dsRNAs and they are functional in
`mammalian cells. Synthetic siRNAs can also be introduced
`into cells exogenously in order to experimentally activate
`RNAi [27]. When an exogenous 19-21 bp siRNA is intro-
`
`1389-5575/10 $55.00+.00
`
`© 2010 Bentham Science Publishers Ltd.
`
`Alnylam Exh. 1032
`
`

`

`Recent Progress in Chemically Modified siRNAs
`
`Mini-Reviews in Medicinal Chemistry, 2010, Vol. 10, No. 7 579
`
`duced into a mammalian cell the 5’-end is phosphorylated.
`The duplex is assembled to form the RNA-Induced Silencing
`Complex (RISC), a multiprotein complex including Argo-
`naute 2 (AGO2), Dicer, TRBP (HIV-1 TAR RNA binding
`protein) and PACT (a dsRNA-binding protein), as well as
`other proteins, some of which are yet unknown [29]. The
`RISC proteins facilitate searching through the genome for
`RNA sequences that are complementary to one of the two
`strands of the siRNA duplex. One strand of the siRNA (the
`sense or passenger strand) is lost from the complex, while
`the other strand (the antisense or guide strand) is matched
`with its complementary RNA. In particular, the targets of
`siRNA-loaded RISC are mRNAs presenting a perfect se-
`quence complementarity. When siRNA-mediated silencing
`occurs, after the cleavage, release and degradation of the
`products, RISC-complex can interact with other molecules
`from the mRNA pool [30]. It was shown that endogenously
`expressed small RNA molecules, named microRNAs (miR-
`NAs), elicit gene-silencing in higher organisms [31, 32].
`MiRNAs are not involved in the pathway that leads to the
`production of a protein, instead they regulate the expression
`of mRNAs. The targets of miRNA-loaded RISC are mRNAs
`presenting a perfect sequence complementarity with nucleo-
`tides 2-7 in the 5’ region of the miRNA (the so-called seed
`region), and additional base pairings with its 3’-region. RISC
`mediates downregulation of gene expression by cleavage or
`translational inhibition of the target mRNA. The choice be-
`tween these two modes of action is dictated by the degree of
`complementarity between miRNA and its target. Near-
`perfect complementarity produces cleavage of the mRNA, as
`well as siRNAs, followed by its complete degradation,
`whereas partial complementarity causes translational inhibi-
`tion [33]. In vertebrates, most of the time, the consequence is
`an inhibition of translation. In addition to repressing transla-
`tion, miRNA interactions can promote the deadenylation or
`decapping, leading to rapid mRNA decay [34]. MicroRNA
`target sequences are usually located in the 3’-UTR of
`mRNAs. A single miRNA is expected to target at least 100
`transcripts from various genes, and one mRNA may be tar-
`geted, at its 3’-end, by different miRNAs [35]. In mammal-
`ian cells, long double strand RNAs (> 30 nt) induce immune
`responses [36] resulting in global gene silencing and in cer-
`tain cases cell death [37, 38]. This fact prevented the applica-
`tion of RNAi to mammalian cells until it was discovered that
`21-mer siRNA duplexes were capable of effecting potent and
`specific gene knockdown without inducing of the interferon
`response [27]. However, the immunogenic activity of short
`RNA duplexes is more significant than it was originally
`thought. Recent findings report off-target effects resulting
`from immunostimulation caused by dsRNAs that are shorter
`than 30 bp [39-46]. Immune response activation by short
`RNAs is a complex process. Briefly, RNAs are recognized
`by three main types of immunoreceptors: Toll-like receptors
`(specifically TLR3, TLR7 and TLR8), protein kinase R
`(PKR) and helicases such as RIG-I and MDA5. Recognition
`by these receptors can lead to a number of cellular responses
`including release of cytokines and changes in gene expres-
`sion [42]. The receptors involved are expressed on cell sur-
`faces (TLR3) [47], in endosomes (TLR3/7/8) [48-50] and in
`the cytoplasm (RIG-I, MDA5, PKR) [49]. Given its reliabil-
`ity and ease of use, RNAi has become the most widely used
`
`technology in functional genomic studies in vitro and in sev-
`eral model organisms. To translate this potential into a broad
`new family of therapeutics, it is necessary to optimize the
`efficacy of the RNA-based drugs. As discussed in this re-
`view, it might be possible to achieve this optimization using
`chemical modifications that improve, just like for ASOs,
`their in vivo stability, cellular delivery, biodistribution,
`pharmacokinetics, potency, and specificity. Previous experi-
`ence with antisense oligonucleotides is directly relevant to
`the clinical progress of siRNAs. Indeed, siRNAs and an-
`tisense oligonucleotides have more similarities than differ-
`ences. ASOs and siRNAs are natural phosphodiester com-
`pounds, they are short (~20 base) nucleic acids and are large,
`negatively charged molecules [51, 52]. Similar protocols
`exist for the large-scale synthesis of nucleic acid drugs in
`amounts needed for clinical trials. Such as ASOs, siRNAs do
`not readly cross the cellular membrane because of their nega-
`tive charge and size [53]. There are biological barriers that
`stand between initial administration of oligonucleotides and
`their final actions within cells. One the first biological barri-
`ers encountered by administered naked ASOs and siRNAs is
`represented by the nuclease activity in plasma and tissues.
`Single-stranded nucleic acids are rapidly degraded in serum
`or inside cells. Double-stranded nucleic acids, including
`siRNAs, are more stable than their single stranded counter-
`parts, but are still degraded and must be protected from nu-
`clease attack [54]. The systemic administration of ASOs and
`siRNAs results in primary localization to liver while their
`local administration is an option for many diseases. They
`have a common target: siRNAs are also intended to bind by
`Watson-Crick base-pairing complementary mRNA, and they
`can induce target RNA destruction. Both ASOs and siRNAs
`can incorporate chemical modifications in order to improve
`their in vivo properties. SiRNAs and ASOs, because of syn-
`thetic nature can be recognized as “foreign” by the innate
`immune system. In addition, for siRNAs both strands have
`the potential to trigger unwanted side effects [55 and cited
`references]. Finally, antisense and siRNA molecules are in-
`volved in ongoing testing in multiple clinical trials. There are
`also significant differences between antisense oligonucleo-
`tides and siRNAs. siRNAs contain two strands, which must
`be synthesized separately and then hybridized. Each strand
`has a molecular weight of approximately 7000 Da and the
`large size, as well as the charged backbone, discourage easy
`passage of RNA duplex through cell membranes. The target
`recognition of siRNA is mediated through the RISC com-
`plex. Moreover, very few molecules of siRNA are needed to
`inhibit gene expression [56]: siRNAs co-opt a natural silenc-
`ing pathway and the RISC proteins facilitate efficient recog-
`nition of target sequences by siRNAs, whereas ASOs must
`find their targets unassisted. Because endogenous duplex
`RNAs control important physiologic processes, introducing
`synthetic RNA may pertub this native machinery. ASOs
`have one strand that can block RNA by a steric mechanism
`or form a DNA-mRNA hybrid that recruits RNase H. Other
`mechanisms described for the ASOs include interference
`with mRNA processing and transport, and formation of a
`triplex directly with DNA into nucleus (Triplex Forming
`Oligonucleotides, TFO) [57, 58]. The mechanism by which
`antisenses exert their effects, largely depends on their struc-
`ture and chemistry: morpholino, PNA and 2’-O-alkyl modi-
`
`

`

`580 Mini-Reviews in Medicinal Chemistry, 2010, Vol. 10, No. 7
`
`Gaglione and Messere
`
`fied ASOs are capable of acting by other mechanisms e.g.
`they can inhibit intron excision, a key step in the processing
`of mRNA. Splicing occurs into nucleus during the matura-
`tion step of pre-mRNA and can be inhibited by hybridization
`of an oligonucleotide to the 5’ and 3’-regions involved in
`this process. ASOs can act like splicing regulator while siR-
`NAs cannot target nuclear RNAs or introns [59]. When
`chemical modifications are introduced into siRNAs it is im-
`portant to underline that modified siRNAs work through an
`only pathway and must interact with a number of different
`cellular proteins. Many or all of these proteins may be sensi-
`tive to changes in siRNA structure caused by the modifying
`group. SiRNA duplexes have been modified in a wide vari-
`ety of ways and the reported results seem to contradict one
`another.
`
` The better and recent understanding of both ASOs and
`siRNA properties have raised an important discussion about
`the future of antisense and siRNA in clinic applications. But
`the theoretical discussion of the relative value of siRNA and
`antisense oligonucleotides can only be resolved by clinical
`development. In fact, the widespread and rapid adoption of
`siRNA in cell culture, due to the potency of the natural
`mechanism of RNAi, has supported the conclusion that
`siRNA strategy has advantages for gene silencing respect to
`ASOs approach. However, further work will be needed to
`improve in vivo potency, biodistribution and toxicity of siR-
`NAs. Certainly the future of siRNA as therapy will involve
`advanced medicinal chemistry to improve both biodistribu-
`tion to tissue and uptake by specific cell types to reduce off-
`target effects and enhance the efficiency of silencing by
`those duplexes that ultimately reach target cells.
`
` A logical starting point for the reaching of this goal has
`lied with the previous generation of research on the proper-
`ties of ASOs. Because of this head start, progress that re-
`quired 10-15 years for antisense oligonucleotides has re-
`quired just 2-4 years for siRNA. The lesson learned from
`antisense drug clinical development has provided insights
`into how to choose target genes, into experimental pitfalls
`and the criteria for designing well-controlled experiments in
`drug based RNAi. In conclusion, the chemical modifications
`that have been developed to optimize the properties of
`ASOs, have had a major impact on the properties and effi-
`cacy of siRNAs.
`
`Functional Anatomy of siRNA: The Rationale for Chemi-
`cal Modification of siRNAs
`
` On the basis of analyses of a small number of silenced
`genes in mammalian cells, a set of empirical guidelines have
`already been proposed for siRNA design. These rules require
`the generation of RNA duplexes containing a 19-nt duplexed
`region, symmetric 2-3-nt 3’ overhangs, 5’-phosphate and 3’-
`hydroxyl groups targeting a region in the gene to be silenced.
`In most cases, only one of the two strands of a siRNA enters
`the RISC with high efficiency. This so-called guide strand is
`selected on the basis of thermodynamic bias and is responsi-
`ble for mediating knockdown of target genes. The targeted
`mRNA is than cleaved by activated RISC at a single site that
`is defined with regard to where the 5’-end of the antisense
`strand is bound to the mRNA target sequence [60, 61]. For
`RNAi-mediated mRNA cleavage and degradation to be suc-
`
`cessful, the double helix of the antisense-target mRNA du-
`plex must be in the A-form [62]. On the basis of these crite-
`ria, an effective siRNA has high stability (high content of G
`or C) at the 5’-terminus on the sense strand, in order to block
`incorporation of sense strand into RISC, lower stability at the
`5’-terminus of antisense (AU-richness) to promote incorpo-
`ration of antisense strand into RISC, and at the cleavage site
`(U at position 10 of sense strand), to help RISC-AS-
`mediated cleavage of mRNA and the RISC-AS-complex
`release. Effective gene silencing by RNAi machinery re-
`quires complete understanding of the elements that influence
`siRNA functioanality and specificity. These include, as well
`as only just described structural and sequence features re-
`quired, sequence space restrictions that define the boundaries
`of siRNA targeting. The sequence space is defined as the
`region of a gene that can be targeted for effective gene
`knockdown. Actually, targets are limited to regions of the
`gene that are transcribed: the 5’ and 3’ UTRs (untraslated
`regions) regions within 75 bases of the start codon and se-
`quences with > 50% C+G content and the ORF (open read-
`ing frame). Many computer programs are available for iden-
`tifying the optimal target sequences for a given gene [63,
`64]. The abundance of 3’UTR seed complements in off-
`target gene [65] suggested that, in the future, bioinformatic
`techniques that minimize off-target effects may be a standard
`component in siRNA design. There are multiple types of
`chemical modifications that are introduced into siRNAs in
`order to enhance thermodynamic and nuclease stability, to
`increase the half-life of the siRNA duplexes in vivo, to im-
`prove the biodistribution and pharmacokinetic properties of
`siRNAs, to target small RNAs to certain cell types and to
`improve the potency of siRNAs in terms of target binding
`affinity, and finally to reduce off-target effects. The synthe-
`sis and the properties of siRNAs bearing phosphodiester,
`carbohydrate and nucleobase modifications have been revi-
`wed extensively [66, 67]. Consequently, this review particu-
`larly covers the recent advances in this area that have been
`published over the past recent years, with particular attention
`on modified structure, architecture and conjugated siRNAs.(cid:1)
`
`CLASSICAL AND NOVEL CHEMICAL MODIFICA-
`TIONS OF siRNAs
`
` The current and most popular approach in the synthesis
`of oligoribonucleotides is based on phosphoramidite chemis-
`try which was originally developed for the synthesis of DNA
`oligonucleotides by Beaucage and Caruters in the early 1980
`[68]. Most siRNAs used in research today are made by
`chemical synthesis using phosphoramidite building blocks as
`single-stranded form. This approach permits incorporation of
`a wide variety of modifications into the siRNAs. A rational
`design of effective chemically modified siRNA must con-
`sider as general principle that the two strands of a siRNA
`function differently and as pratical hint that the nucleotides
`are different according to positions and nature. It has been
`recently demonstrated that novel sense and/or antisense
`strand modifications can greatly enhance siRNA specificity
`targeting, distinct and separate the events in interference
`cycle [69, 70].
`
`In this section, we will briefly review the most significant
`
`siRNA modifications described in literature, drawing atten-
`tion to those that have improved siRNA performance. We
`
`

`

`Recent Progress in Chemically Modified siRNAs
`
`Mini-Reviews in Medicinal Chemistry, 2010, Vol. 10, No. 7 581
`
`will point out the useful and universal modifications as well
`as the most innovative modifications to move siRNA toward
`the clinic.
`
`Base Modifications
`
` Base-modified siRNAs (Fig. 1) have been investigated so
`far to a very limited extent [71, 72] despite the central in-
`volvement in target recognition of nucleobases. The report
`by Parrish et al. describe induction of RNAi in C. elegans by
`dsRNAs containing 4-thiouridine, 5-bromo-, 5-iodo-, 5-(3-
`aminoallyl)-uridine, or inosine [71]. While these experiments
`did not demonstrate any remarkable differences in silencing
`activity regardless of the introduced modifications, the work
`by Chiu and Rana [72] describes reduced silencing activity
`of duplexes containing 5-bromouridine, 5-iodouridine, 2,6-
`diaminopurine, and N-3-methyl-uridine. The patent literature
`also claims several base-modified nucleosides as compo-
`nents of small interfering nucleic acids, albeit no details are
`provided with regard to properties of such modified siRNAs
`[73]. In other studies [74], Sipa et al. evaluated the thermo-
`dynamic stability and gene-silencing activity of base-
`modified siRNAs containing three modified nucleosides: 2-
`thiouridine (s2U), pseudouridine ((cid:1)), and dihydrouridine (D).
`They demonstrated that these three naturally occurring modi-
`fied nucleosides, when introduced into a siRNA duplex,
`modulate its silencing potency. The extent of this effect de-
`pends on the modification position and is most advantageous
`when the s2U, (cid:1), and D nucleosides participate in an en-
`hancement of the siRNA duplex asymmetry. More recently,
`Kool at al. have evaluated steric and stability effects on
`RNAi activity by use of siRNAs modified with propynyl and
`methyl functionalities at the C-5 position of pyrimidine nu-
`
`cleobases [75]. Their results suggest that at the 5’-half of the
`guide RNA, large increases in the size of the functionality at
`the position 5 of pyrimidine nucleobases can be detrimental
`to RNAi activity. Despite the strong stabilizing effects of the
`5-propynyl modification, this bulky substitution caused
`negative effects in RNAi activity, probably due to disruption
`of interactions in the major groove of the active RISC com-
`plex. However, the smaller 5-methyl substitution did not
`adversely affect gene silencing activity. Surprisingly, some
`atypical base structures have been used in siRNA [76].
`SiRNA duplexes containing the 2,4-difluorotoluyl ribonu-
`cleoside (rF) were synthesized to evaluate the effect of non-
`canonical nucleoside mimetics on RNA interference. 5´-
`Modification of the guide strand with rF did not alter silenc-
`ing relative to unmodified control. Internal uridine to rF sub-
`stitutions were well-tolerated and thermal melting analysis
`showed that the base pair between rF and adenosine (A) was
`destabilizing relative to a uridine-adenosine pair, although it
`was slightly less destabilizing than other mismatches. siR-
`NAs with the rF modification effectively silenced gene ex-
`pression and offered improved nuclease resistance in serum.
`Base modifications can also help to reduce immune activa-
`tion: modification with pseudouracil or 2-thiouracil prevents
`the RIG-I-mediated
`immunostimulation due
`to a 5’-
`triphosphate [40], and 5-methyl-C, N6-methyl-A pseu-
`douridine prevents recognition of RNA by TLR3, TLR7 and
`TLR8 [77].
`
`Sugar Modifications
`
` The siRNA modifications on the sugar moiety are the
`most widely described (Fig. 2). The milestone study report-
`ing the pioneering analysis of chemical modified siRNA
`
`F
`
`R
`
`rF
`
`N
`
`N
`
`R
`
`F
`
`NH2
`
`N
`
`Br
`
`N
`
`NH2
`
`O
`
`N
`
`R
`
`I
`
`NH
`
`O
`
`O
`
`N
`
`R
`
`NH
`
`O
`
`2,6-DAP
`
`5-BrU
`
`5-IU
`
`O
`
`O
`
`HN
`
`HN
`
`HN
`
`NH
`
`~) ~) 4
`Y~ Y~ Y~
`
`O
`
`R
`
`(cid:1)
`
`O
`
`NH
`
`O
`
`NH2
`
`N
`
`N
`
`I
`
`R
`
`O
`
`O
`
`N
`
`R
`
`s2U
`
`S
`
`O
`
`N
`
`R
`
`D
`
`O
`
`NH
`
`O
`
`N
`
`I
`
`R
`
`N
`
`I
`
`R
`
`pU
`
`mU
`
`mC
`
`
`
`Fig. (1). Nucleobase modifications in siRNA. rF: 2,4-difluorotoluyl ribonucleoside; 2,6-DAP: 2,6-diaminopurine; 5-BrU: 5-bromouridine; 5-
`IU: 5-Iodouridine; s2U: 2-thiouridine; D: dihydrouridine; (cid:1): pseudouridine; pU: 5-propynyluridine; mU: 5-methyluridine; mC: 5-
`methylcitydine
`
`

`

`582 Mini-Reviews in Medicinal Chemistry, 2010, Vol. 10, No. 7
`
`Gaglione and Messere
`
`function [72] showed that while A-form helix is required for
`the mechanism of RNAi, the 2’-OH function was not an es-
`sential requirement for interfering activity. This study
`strongly indicates that RNAi machinery does not require the
`2’-OH for recognition of siRNAs and catalytic ribonuclease
`activity of RISC does not involve the 2’-hydroxyl group of
`guide antisense of RNA. Therefore, the 2’ position of ribose
`has been heavily modified. Modification of the 2’ position of
`the ribose can increase Tm of duplex and confers varying
`degrees of nuclease resistance. It may also provide protection
`from immune activation. Briefly, we list here the most ex-
`tensively studied 2’ modifications in siRNA effective mole-
`cules. 2’-fluoro (2’-F) was among the first modifications that
`were independently tested by several groups and they result
`well tolerated in siRNA applications [71, 72, 78-80]. 2’-O-
`methyl (2’O-Me) was extensively tested in siRNA by differ-
`ent groups since 2003 [72, 79-87]. Partial modification of
`siRNA strands with 2’O-Me (less than 8 nt) led to improved
`performance of siRNA in cell [86], but siRNAs completely
`made of 2’O-Me lost interference activity [72]. Position-
`specific 2’-O-methylation of siRNAs reduces “off target”
`transcript silencing [69]. The sharp position dependence of
`2’-O-Me modification contrasts with the broader position
`dependence of base-pair within the seed region, suggesting a
`role for position 2 of the guide strand distinct from its effects
`on pairing to target transcripts. Besides aforementioned sta-
`bility, 2’-O-Me siRNAs exhibit increased potency [88] and
`reduced immunostimulatory activity [41, 89], with the ex-
`ception 2’-O-Me cytidine [90]. Introduction of 2’-O-Me
`uridine or guanosine into one strand of the siRNA duplex
`generates noninflammatory siRNAs. 2’-O-Me siRNAs, con-
`taining less than 20% modified nucleosides and targeting
`apolipoprotein B (apoB), can mediate potent silencing of its
`target mRNA and cause significant decreases in serum apoB
`and cholesterol. This is achieved at therapeutical viable
`siRNA doses without cytokine induction, toxicity or off tar-
`get effects [90]. It was been showed that 2’-O-Me RNAs act
`as potent antagonists of immunostimulatory RNA and they
`are able significantly to reduce both interferon-(cid:1), and inter-
`leukin-6 induction by small-molecule TLR7 agonist loxorib-
`ine in human peripheral blood mononuclear cells, murine
`Flt3L dendritic cells and in vivo mice [89]. Further, 2’-O-
`methylation selectively protects the particularly vulnerable
`5’-end of the guide strand against exonucleases in human
`blood serum [91]. Specific chemical modification thus re-
`solves the asymmetric degradation of the guide and sense
`strands, which is inherent to the thermodynamic asymmetry
`of the siRNA termini as required for proper utilization of the
`guide strand in RNAi pathway. The 2’-O-(2-methoxyethyl)
`residues (2’-O-MOE) can be incorporated into siRNAs much
`like 2’-O-Me or 2’-F. The 2’-MOE modification in the an-
`tisense strand resulted in less active siRNA constructs re-
`gardless of placement position in the construct. The incorpo-
`ration of modified residues in the sense strand did not show a
`strong positional preference [85]. This modification is not
`generally available for use but a successful gene target in
`vivo is reported [92]. Likewise, 2’-O-allyl modification is
`well tolerated in the 3’-overhangs but not at most positions
`in siRNA duplexes [87]. Another class of 2’ modification is
`2’-deoxy-2’-fluoro-(cid:2)-D-arabino nucleic acids
`(FANA).
`FANA nucleotides have the stereochemistry at the 2’ posi-
`
`tion inverted relative to that found in 2’-O-Me and 2’-F
`RNA. siRNAs that have completely FANA-substituted sense
`strands are 4-fold more potent than unmodified siRNAs and
`have a longer half-life in serum. The antisense strand is less
`tolerant of the FANA modification [93, 94]. DNA itself rep-
`resents a further modification in which there is not the elec-
`tronegative OH-group in 2’ position. The use of DNA in 3’-
`overhangs of synthetic siRNAs is well known [27]. Substitu-
`tion with double stranded DNA in the 8-bp region at the
`5’end of the guide strand gives active duplexes with reduced
`off-target effects [95]. Interestingly, 2’deoxy bases have re-
`cently been reported to also block immune detection, particu-
`larly T or dU bases [96]. Locked nucleic acids is a family of
`conformationally locked nucleotide analogues containing a
`methylene bridge between the 2’ and 4’ carbons of the ribose
`ring [97]. This linkage constrains the ribose ring, “locking” it
`into the 3’-endo conformation close to that formed by RNA
`after hybridization. The incorporation of LNA in siRNAs
`enhances serum half-life substantially and a few LNA units
`at the 5’-end of the sense strand improves thermodynamic
`bias, reduces immunostimulatory and off-target effects [39,
`98]. Particular LNA based siRNAs were designed and de-
`veloped against the highly structured 5’-UTR of cox-
`sackievirus B3 (CVB-3) [99]. The best siLNA improved
`viability of infected cells by 92% and exerted good antiviral
`activity in plaque reduction assays. 2’-O-(2,4-dinitrophenyl)
`RNAs were among the 2’modification that have been more
`recently tested in the context of RNAi. DPN-ssRNA and
`DPN-siRNA outperform previous generation of antisense
`and unmodified siRNAs in gene silencing potency, stability,
`hybridization affinity and delivery maintaining specific bind-
`ing properties. Poly-2’-O-(2,4-dinitrophenyl)-oligoribonucl-
`eotide (DNP-RNA) represents a promising new gene silenc-
`ing platform. DNA-RNAs with a DNP nucleotide molar ratio
`of about 0.7 can spontaneously cross viral envelopes and
`mammalian cell membranes. DNP-RNAs are resistant to
`degradation by ribonucleases, including RNases A, B, H, S,
`T 1 and 2 and phosphodiesterase I and II [100] and hybridize
`with their complementary RNA. Interestingly, 2’-O-DNP
`modified siRNA and its single stranded counter part have
`shown similar silencing activities [101-103]. The ring oxy-
`gen has also been modified: 4’-thio modified nucleosides
`have a sulfur atom at the 4 position of the ribose ring. Ho-
`shika et al. report that 4’-thio RNAs form a thermally stable
`duplex with the complementary RNA and show high nucle-
`ase resistance, despite the 2’-OH groups. In addition, struc-
`tural analysis by CD spectra
`indicates
`that
`the 4’-
`thioRNA:RNA duplex adopts an A-form conformation as
`does the natural RNA duplex. 4’-S-RNA is very well toler-
`ated near the termini of siRNA duplexes [104-106]. Modifi-
`cations at the sense-strand preserve RNAi activity of modi-
`fied siRNA, except
`for
`full modification with 4’-
`thioribonucleoside and the activity of siRNAs modified at
`the antisense-strand was dependent on the position and the
`number of modifications with 4’-thioribonucleosides [104].
`For example, the 5’-end of the antisense strand could be
`modified with a few 4’S-RNA inserts, without significant
`loss of potency [105]. Furthermore, Dande et al. describe
`that significant improvements in siRNA activity and plasma
`stability were achieved by judicious combination of 4’-
`thioribose with 2’-O-Me and 2’-O-MOE modifications. It is
`
`

`

`Recent Progress in Chemically Modified siRNAs
`
`Mini-Reviews in Medicinal Chemistry, 2010, Vol. 10, No. 7 583
`
`O
`
`/1-
`
`B
`
`B
`
`O
`
`O
`
`O
`
`O
`
`)-,..
`
`B
`
`O
`
`/1-
`
`O
`
`B
`
`O
`
`O
`
`O
`
`2'-MOE
`
`OCH3
`
`O
`
`B
`
`O
`
`O
`
`2'-OMe
`
`~ I
`
`F
`
`B
`
`O
`
`O
`
`OH
`
`O
`
`F
`
`O
`
`B
`
`O
`
`2'-F
`
`O
`
`O
`
`)-,..
`
`B
`
`2'-OH
`
`O
`
`/1-
`
`O
`
`O
`
`O
`
`O
`
`O
`
`O
`
`1, c l,c>-:c
`L 1\)-,.. I, C
`I" j__
`>-
`l,c
`I,{ l,c
`I, C I, C
`
`B
`
`S
`
`O
`
`OH
`
`4'-THIO
`
`;,..,,,
`
`O
`
`B
`
`O
`
`I-((
`
`O
`
`O
`
`LNA
`
`~
`
`O
`
`2'-FANA
`
`O
`
`2'-deoxy
`
`O2N
`2'-DNP
`
`;!'
`
`2'-O-allyl
`
`)-,.. 0
`
`B
`
`O
`
`O
`
`NH2
`
`2'-amino
`
`/1-
`
`O
`
`B
`
`O
`
`NO2
`
`O
`
`O
`
`OH
`
`UNA
`
`B
`
`
`
`A
`
`O
`
`1-ztr
`
`O
`
`O
`
`ENA
`
`Fig. (2). Sugar modifications in siRNA. 2’-F: 2’-fluoroRNA; 2’-OMe: 2’-O-Methyl RNA; 2’-MOE: 2’-O-(2-methoxyethyl)RNA; 2’-O-allyl:
`2’-O-allyl RNA; 2’-DNP: 2’-O-(2,4-dinitrophenyl) RNA; 2’-FANA: 2’-deoxy-2’-fluoro-(cid:2)-D-arabino nucleic acids; 2’-deoxy: 2’-
`deoxyribonucleotide; LNA: locked nucleic acid; 4’-Thio: 4’-thioribonucleoside; UNA: unlocked nucleic acid; 2’-amino: 2’-amino RNA;
`ENA: ethylene-bridge nucleic acid.
`
`also possible to combine 4(cid:1)-thio substitution with FANA
`nucleotides, and 4(cid:1)-S-FANA–modified siRNAs have been
`shown to possess potencies for silencing in cell culture that
`were
`similar
`to unmodified
`siRNAs
`[95]. 2’,3’-
`Seconucleosid

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket