throbber
Published online 16 January 2012
`
`4125–4136
`Nucleic Acids Research, 2012, Vol. 40, No. 9
`doi:10.1093/nar/gkr1301
`
`mRNA knockdown by single strand RNA is
`improved by chemical modifications
`
`Henry J. Haringsma, Jenny J. Li, Ferdie Soriano, Denise M. Kenski,
`W. Michael Flanagan and Aarron T. Willingham*
`
`Sirna Therapeutics, 1700 Owens Street, Fourth Floor, San Francisco, CA 94158, USA
`
`Received November 8, 2011; Revised December 16, 2011; Accepted December 19, 2011
`
`ABSTRACT
`
`While RNAi has traditionally relied on RNA duplexes,
`early evaluation of siRNAs demonstrated activity of
`the guide strand in the absence of the passenger
`strand. However, these single strands lacked the
`activity of duplex RNAs. Here, we report the system-
`atic use of chemical modifications to optimize
`single-strand
`RNA
`(ssRNA)-mediated mRNA
`0
`F ribose modifica-
`knockdown. We identify that 2
`0
`-end phosphorylation vastly
`tions coupled with 5
`improves ssRNA activity both in vitro and in vivo.
`The impact of specific chemical modifications on
`ssRNA activity implies an Ago-mediated mechanism
`but the hallmark mRNA cleavage sites were not
`observed which suggests ssRNA may operate
`through a mechanism beyond conventional Ago2
`slicer activity. While currently less potent
`than
`duplex siRNAs, with additional chemical optimiza-
`tion and alternative routes of delivery, chemically
`modified ssRNAs could represent a powerful RNAi
`platform.
`
`INTRODUCTION
`
`RNA interference is a process inherently driven by RNA
`duplexes. The double-strand endonuclease Dicer processes
`long double-stranded RNAs (dsRNA) into small 21–23 nt
`0
`phosphate (1–4). The re-
`RNA duplexes containing a 5
`sulting siRNAs and miRNAs load into the RNA-induced
`silencing complex (RISC) where the passenger strand
`(sense) is dissociated, leaving the active guide strand (anti-
`sense) loaded in Ago2 and available to base pair with
`target mRNAs and initiate degradation (5,6). Exogenous
`delivery of small interfering dsRNAs (siRNAs) to mam-
`malian cells was shown to be an effective strategy to
`engage this endogenous mechanism of RNA-mediated
`regulation (7,8). This
`seminal discovery resulted in
`
`for studying
`siRNA becoming an indispensible tool
`gene function and now represents a new therapeutic
`modality (9,10).
`Despite the central role of duplex RNAs in triggering
`RNAi, in the early days of the development of siRNAs
`there were reports of single-strand RNA (ssRNA) acting
`to initiate mRNA knockdown. Using extracts from
`lines, Martinez et al.
`human cell
`(11) reported that
`ssRNA could reconstitute RISC; however, compared to
`duplex siRNAs, 10- to 100-fold higher concentrations of
`ssRNA were required. They also transfected single strands
`and duplexes into cells, thus demonstrating that ssRNA
`knockdown was optimal with a size >17 nt and improved
`0
`by the addition of a 5
`phosphate. Again, duplex siRNAs
`remained more potent. Schwarz et al. (12) evaluated both
`miRNA and siRNA, demonstrating that ssRNAs corres-
`ponding to the let-7 miRNA or luciferase could trigger
`0
`target cleavage but required a 5
`phosphate. The observa-
`tion that phosphorylation improves ssRNA activity indir-
`ectly argues for Ago-mediated activity. Phosphorylation
`0
`of the 5
`-end of the guide strand plays an important role
`in guide strand binding within the MID domain of Ago2
`and the subsequent positioning of the guide strand for
`accurate cleavage (13,14).
`Single-strand-mediated knockdown was shown to be
`dose-dependent but was 8-fold less effective than the
`corresponding duplex. Another study found that ssRNA
`targeting the human blood clotting initiator tissue factor
`(TF) was as effective in human cells as the duplex siRNA
`at high concentrations of 100–200 nM (15). In contrast to
`other reports, exogenous phosphorylation of the ssRNA
`was not found to improve knockdown activity. In a
`follow-up study, a dose–response comparison of
`the
`same single strand and duplex siRNAs
`found the
`dsRNA was 5- to 6-fold more potent than the correspond-
`ing ssRNA and analysis by northern blot demonstrated
`that ssRNA produced mRNA cleavage fragments (16).
`Core components of RISC have been shown to bind to
`ssRNAs. In vitro binding studies with purified human
`Ago2 demonstrate significantly higher binding affinity
`
`*To whom correspondence should be addressed. Tel: +1 650 496 1284; Fax: +1 650 496 6556; Email: aarron.willingham@merck.com
`
`ß The Author(s) 2012. Published by Oxford University Press.
`This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/
`by-nc/3.0), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
`
`Alnylam Exh. (cid:20)(cid:19)(cid:22)(cid:26)
`
`

`

`4126 Nucleic Acids Research, 2012, Vol. 40, No. 9
`
`for ssRNA which bound 70- to 100-fold tighter than
`dsRNA (17). Recombinant Dicer has also been shown
`to bind 21 nt ssRNAs in vitro with preferential affinity
`0
`for ssRNAs containing a 5
`phosphate (18). This led the
`authors to hypothesize that Dicer may play a role in
`ssRNA knockdown by either facilitating loading of
`ssRNA into Ago2 or by binding to ssRNAs and thus pro-
`tecting them from nucleases (18).
`Chemical modifications have been employed to improve
`the pharmacokinetic properties of siRNAs including
`increasing
`nuclease
`stability,
`improving
`potency,
`reducing off target effects and abrogating immune stimu-
`0
`0
`ribose modifications (2
`-
`lation (19–21). Generally, 2
`0
`methoxy and 2
`-fluoro in particular) are broadly tolerated
`within the guide strand of duplex siRNAs. Holen et al.
`reported the first evaluation of the impact of chemical
`modifications on ssRNA activity and compared them to
`corresponding duplex siRNAs
`(16). The positional
`chemical sensitivity seen for duplex siRNAs (15) was
`mirrored by the ssRNAs which the authors interpret as
`indicating ssRNA and dsRNA act through a common
`RNAi pathway (16). This hypothesis was supported by
`their observation that an excess of dsRNA can competi-
`tively block the activity of ssRNAs in vitro (16).
`Hall et al. (22) reported that ssRNA potency can be
`significantly improved by the replacement of some of
`the phosphodiester backbone with boranophosphate
`modfications, where a non-bridging oxygen is replaced
`with an isoelectronic borane. Inclusion of 5–8 borano-
`phosphate modifications allowed ssRNAs to achieve
`knockdown comparable to duplex siRNAs at 10–
`25 nM concentrations.
`Importantly, HeLa cells pre-
`treated with siRNAs targeting Ago2 had significantly
`reduced target knockdown when they were subsequently
`transfected with ssRNAs, suggesting that ssRNAs act
`through Ago2 (22). Despite their beneficial properties,
`boranophosphate modifications present a number of syn-
`thesis and chemistry challenges and have thus far not been
`widely adopted (21).
`Recently,
`in unpublished work presented at the 7th
`Annual Oligonucleotide Therapeutics Society meeting
`et al.
`(September 2011), Swayze
`(23) described a
`modified ssRNA that included phosphorothioate back-
`0
`-end vinyl phosphonate modifications that
`bone and 5
`were intended to improve the stability of the ssRNA
`molecule. This modification pattern was a refinement of
`0
`F and
`an earlier published pattern of evenly spaced 2
`0
`2
`OMe modifications reported to improve the activity of
`duplex siRNAs (24). Swayze et al. (23) also reported that
`ssRNAs were inactive in Ago2 knockout cells indicating
`Ago2 mediates ssRNA activity. Importantly, subcutane-
`ously administered unformulated ssRNAs were active in
`rodents and comparable to the potency of second gener-
`ation RNase H antisense oligonucleotides.
`As discussed above, the limited exploration of chemical
`modifications has shown promise for improving the
`activity of ssRNA. Therefore, we conducted a systematic
`0
`ribose modifications on the ac-
`analysis of the impact of 2
`tivity of both duplex and ssRNA-mediated knockdown—
`screening six different RNAs to ensure the generality of
`0
`our observations. Modifications of the 2
`ribose ring
`
`0
`0
`F), and deoxy
`OMe), fluoro (2
`included methoxy (2
`0
`H), which applied to either purines or pyrimidines
`(2
`present on the guide strand. Additionally, guide strands
`were synthesized with or without phosphorylation of the
`0
`-end. In vitro screening in tissue culture cells identified
`5
`0
`0
`that 2
`F content and 5
`phosphorylation are key for
`optimal ssRNA-mediated knockdown of target mRNAs.
`Optimized ssRNAs were tested in mice using a lipid
`nanoparticle delivery vehicle. Knockdown of
`target
`mRNA by ssRNA exceeded 90% at the highest dose
`tested; however, the duplex siRNAs in the same study
`had greater overall knockdown and much longer duration.
`0
`Modification of the guide strand 5
`-end with abasic nu-
`cleotides negatively impacted the activity of both duplex
`and ssRNAs, consistent with an earlier report that an
`abasic nucleotide reduced Ago2 binding affinity and
`0
`cleavage activity (17). Taken together with the role of 5
`phosphorylation,
`these
`observations
`offer
`indirect
`evidence that ssRNAs act
`through an Ago-mediated
`mechanism. However, analysis of cleavage products by
`0
`5
`RACE and sequencing failed to identify distinct
`cleavage sites for ssRNAs despite the identification of
`hallmark Ago2 cleavage for duplex RNAs. This suggests
`ssRNA may act through a mechanism beyond canonical
`Ago2 slicer activity.
`0
`0
`While 2
`phosphorylation were
`-fluoro content and 5
`identified as greatly improving ssRNA activity, our side
`by side comparison with duplex siRNAs demonstrates
`that duplex RNAs are consistently more potent than
`their corresponding single strands both in vitro and
`in vivo.
`Importantly, as highlighted by preliminary
`results from Swayze et al. (23), with further improvements
`in chemical optimization and evaluation of alternative
`dosing strategies, such as sub-cutaneous delivery, ssRNA
`could represent a powerful alternative to duplex siRNAs.
`
`MATERIALS AND METHODS
`
`RNA oligo sequence and synthesis
`0
`
`A systematic screen of 2
`ribose modifications on six dif-
`ferent target sites (Table 1) resulted in a total of 180
`siRNAs which were then compared as duplex and single
`strands (360 in total). Individual sequences and modifica-
`tion patterns are listed in Supplementary Tables S3
`0
`and S4. Inverted abasic modifications present at the 5
`0
`and 3
`of the passenger strand serve to block loading
`into Ago2 (25,26). Sequences for siRNAs were selected
`using a previously published algorithm developed to
`
`Table 1. Target gene, site, accession and sequence for siRNAs
`
`Target
`gene
`
`ApoB
`ApoB
`ApoB
`ApoB
`SSB
`SSB
`
`Target
`site
`
`6981
`8786
`9470
`10127
`386
`963
`
`Target
`accession
`
`NM_009693
`NM_009693
`NM_009693
`NM_009693
`NM_009278
`NM_009278
`
`Target sequence
`
`CACAATGCATTTAGATCAA
`CCAGTAAGGCTTCTCTTAA
`CTTTAACAATTCCTGAAAT
`TCATCACACTGAATACCAA
`TAACAACAGACTTTAATGT
`AAATCATGGTGAAATAAAA
`
`

`

`Nucleic Acids Research, 2012, Vol. 40, No. 9 4127
`
`predict maximal knockdown while minimizing off-target
`hybridizations (27). siRNAs were synthesized using previ-
`ously described methods (28,29). Individual oligonucleo-
`tide strands were synthesized separately using solid-phase
`methods, purified by ion-exchange
`chromatography
`and duplexed when appropriate. Knockdown studies
`were normalized using a non-targeting control siRNA
`0
`0
`0
`OH (r), 2
`F (f), 2
`OMe
`sequence which contains ribose 2
`0
`H (d) residues at the indicated positions as well
`(m) and 2
`as inverted abasic caps (iB) on the passenger strand: guide
`0
`0
`–3
`)
`(fC;fC;fU;mG;mA;mA;mG;mA;mG;mA;mG;fU;
`(5
`0
`0
`fU;mA;mA;mA;rA;rG;rA;mU;mU) and passenger (5
`–3
`)
`( iB; fU; fC;fU;fU;fU;fU;dA;dA;fC;fU;fC;fU;fC;fU;fU;fC;
`dA;dG;dG;dT;dT;iB).
`
`diafiltration as previously described (30,32). The cat-
`ionic lipid CLinDMA (2-{4-[(3b)-cholest-5-en-3-yloxy]-
`butoxy}-N,N-dimethyl-3-[(9Z,12Z)-octadeca-9,12-dien-1-
`yloxy]propan-1-amine),
`cholesterol
`and PEG-DMG
`(monomethoxy(polyethyleneglycol)-1,2-dimyristoylglycer-
`ol) were mixed together at a molar ratio of 50:44:6.
`PEG-DMG was purchased from NOF Corporation, chol-
`esterol
`from Northern Lipids, and CLinDMA was
`synthesized by Merck & Co., Inc. Particle size was
`measured by dynamic light scattering using a Wyatt
`DynaPro plate reader and percent encapsulation was
`determined using a SYBR Gold fluorescence assay
`(Invitrogen) and were within pre-established quality
`metrics (32).
`
`In vitro mRNA knockdown
`
`In vivo mRNA knockdown
`
`Mouse Hepa1–6 cells were cultured in Dulbecco’s-
`modified Eagle Medium supplemented with 10% fetal
`bovine
`serum, 1% penicillin–steptomycin and 1%
`sodium bicarbonate. These cells were plated in a 96-well
`culture plates at a density of 3000 cells/well 24 h prior to
`transfection. Transfections were
`performed
`using
`Opti-MEM I Reduced Serum Media and Lipofectamine
`RNAiMAX as previously described (30). Final siRNA
`concentrations range from 100 to 1 nM for in vitro
`cell-based screens with concentrations varying for
`ssRNA (100 nM, 10 nM) and dsRNA (100 nM, 10 nM,
`and 1 nM) (see Supplementary Table S3). Final siRNA
`concentrations for the dose–response curves range from
`40 to 0.002 nM along an eight-point, 4-fold titration
`curve. Twenty-four hours post-transfection cells were
`washed with phosphate-buffered saline and processed
`using
`the TaqMan Gene Expression Cells-to-CT
`(Invitrogen), per manufacturer’s instructions, to extract
`RNA, synthesize cDNA and perform RT–qPCR using
`an Ssb (Mm00447374_m1) or ApoB (Mm01545154_g1)
`specific Taqman primer/probe set on an ABI Prism
`7900HT Sequence Detector. Reverse transcription condi-
`
`tions were as follows: 60 min at 37
`C followed by 5 min
`
`at 95
`C. RT–qPCR conditions were as follows: 2 min at
`
`
`50
`C, 10 min at 95
`C, followed by 40 cycles of 15 s at
`
`
`95
`C, and 1 min at 60
`C. Gapdh mRNA levels were
`used for data normalization (Taqman part number
`4308313). Knockdown of Ssb/ApoB was calculated as
`the percent knockdown in Ssb/ApoB cDNA measured in
`experimentally treated cells relative to the Ssb/ApoB
`cDNA levels measured in non-targeting, control-treated
`cells. The
`comparative Ct
`calculation method for
`knockdown has previously been described (31). Briefly,
`mRNA CtGapdh
` Ct = CtTarget
` Ct =
`and
` Ct(siRNA) Ct(non-targeting control) and relative expres-
` Ct
`sion
`level = 2
`and % knockdown =
`100 (1 2
` Ct). Potency (EC50) was calculated using
`a four-parameter curve fit
`tool and Prism graphing
`software (GraphPad Software).
`
`Lipid nanoparticle formulation
`
`siRNA lipid nanoparticles (LNPs) were assembled by
`simultaneous mixing of a lipid mixture in ethanol
`with an aqueous
`solution of
`siRNA followed by
`
`All in vivo work was approved by an Institutional Animal
`Care and Use Committee and adhered to standards rec-
`ommended by the Association for Assessment and
`Accreditation of Laboratory Animal Care, International.
`C57/BL6 mice (Jackson Labs) were dosed by intravenous
`injection with siRNA formulated in lipid nanoparticle at 3
`or 6 mg/kg as previously described (30,33). Animals were
`euthanized by CO2 inhalation and immediately after eu-
`thanasia liver sections were excised, placed in RNA Later
`
`C until ready for analysis. Liver
`(Qiagen) and stored at 4
`tissue was homogenized in Qiazol using stainless steel
`beads and a TissueLyser (Qiagen). Following homogen-
`ization,
`chloroform was added and samples were
`centrifuged. The aqueous layer was combined with an
`equal volume of 70% ethanol and samples were purified
`using an RNeasy purification kit per manufacturer’s
`directions
`(Qiagen). The
`resulting RNA was
`then
`normalized, cDNA was synthesized, and RT-qPCR was
`performed using ApoB specific Taqman primer/probe sets
`on an ABI Prism 7900HT Sequence Detector. Reverse
`transcription conditions were as follows: 60 minutes at
`
`
`37
`C followed by 5 min at 95
`C. RT–qPCR conditions
`
`
`were as follows: 2 min at 50
`C, 10 min at 95
`C, followed
`
`
`by 40 cycles of 15 s at 95
`C and 1 min at 60
`C. Gapdh
`mRNA levels were used for data normalization.
`Knockdown of ApoB was calculated using the same
`method described for the in vitro experiments.
`0
`0
`
`-RLM-RACE was performed using GeneRacer kit
`5
`(Invitrogen) following the manufacturer’s instructions.
`Briefly, 100 ng untreated RNA was directly ligated to the
`RNA linker. After phenol/chloroform extraction and pre-
`cipitation, the first-strand cDNA was synthesized using
`0
`the Oligo-dT primer. For
`the first-round 5
`RACE
`reaction, 1 ml of the first-strand cDNA was amplified
`0
`using the GeneRacer 5
`primer and a target gene specific
`primer (Supplementary Table S5) with cycling as follow-
`
`
`ing: 1 cycle of 94
`C for 2 min, then 5 cycles of 94
`C for 30 s
`
`
`and 72
`C for 1 min, and then 5 cycles of 94
`C for 30 s, and
`
`
`70
`C for 1 min, followed by 20 cycles of 94
`C for 30 s,
`
`
`65
`C for 30 s and 68
`C for 1 min. After additional
`
`
`C, ml of
`C and cool down to 4
`10-min incubation at 68
`the first PCR reaction was used for the second round
`
`5
`
`-RACE and sequencing of cleavage products
`
`

`

`4128 Nucleic Acids Research, 2012, Vol. 40, No. 9
`
`0
`
`0
`Nested
`RACE (Nested PCR) using the GeneRacer 5
`5
`primer and gene-specific nested primer with the 1 cycle
`
`
`
`C for 2 min, and 25 cycles of 94
`C for 30 s, 65
`C
`at 94
`
`C for 1 min. From this reaction, 10 ml was
`for 30 s, and 68
`analyzed on a 2% agarose gel. See Supplementary Table
`S5 for sequence and other information for the primers
`used. To confirm the cleavage site of target mRNA, the
`gel bands corresponding to the size of the expected
`0
`RACE amplicon was
`excised and purified using
`5
`QIAquick Gel Extraction kit (QIAGEN). The purified
`PCR products were sequenced by GENEWIZ (South
`Plainfield, NJ, USA).
`
`RESULTS
`
`Selection of siRNAs and modification patterns
`
`ApoB has been a frequent target of siRNA knockdown
`because of its potential therapeutic value in lowering
`serum cholesterol. Furthermore, given its predominant
`expression in liver hepatocytes, ApoB transcripts can be
`readily targeted in vivo using currently available siRNA
`delivery technologies (34–36). The Ssb gene encodes the
`La antigen which is involved in transfer RNA maturation
`(37) and has been implicated in the autoimmune disease
`Sjogren’s syndrome (38). Ssb was selected primarily for its
`ubiquitous cellular expression pattern. Four siRNAs tar-
`geting ApoB and two siRNAs targeting SSB were chosen
`to evaluate the generality of our findings across multiple
`siRNAs (Table 1).
`0
`0
`0
`H),
`-deoxy (2
`OH), 2
`Various combinations of ribose (2
`0
`0
`0
`0
`-fluoro (2
`F), and 2
`-methoxy (2
`OMe) modifications
`2
`were assigned based on the pyrimidine (Y) or purine (R)
`identity of the siRNA sequence (Table 2). Methoxy modi-
`fication of position 14 of
`the guide strand was not
`permitted due to reported sensitivity of this position to
`modifications
`(17). These
`combinations were
`also
`0
`phosphorylation
`evaluated in the presence or absence of 5
`0
`PO4). Placement of abasic residues
`of the guide strand (5
`0
`at the 5
`of the guide strand have been reported to reduce
`Ago2 binding and cleavage activity (17). Therefore, as a
`negative control, a separate set of oligos were synthesized
`
`with three abasic residues (i.e. ribose sugar without nu-
`cleotide base) at positions 1–3. The chemical structures
`0
`modifications are shown with adenosine as the
`of the 2
`representative nucleotide. All together, 30 different siRNA
`modification patterns were evaluated for each of the ApoB
`or SSB target sites (a total of 180 unique guide strands).
`Each of the different guide strands was evaluated as single
`strands or as duplexes with a passenger strand to create a
`conventional siRNA duplex. Passenger strands are largely
`0
`OMe uridine overhangs and
`ribonucleotides with 2
`inverted abasic residues on each end. Inverted abasics
`block the loading of these strands into Ago2, thus prevent-
`ing passenger strand competition with the guide strand
`(25,26). Sequence information for each guide strand and
`passenger strand are provided in Supplementary Tables S3
`and S4. For in vitro studies, all knockdown data were
`normalized to a chemically modified non-specific siRNA
`duplex which was designed to lack homology to nearly all
`mammalian transcripts (see ‘Materials and Methods’
`section).
`
`0
`F modifications are critical for the activity of single
`2
`strand RNA in vitro
`
`The mouse hepatocyte derived cell line Hepa1-6 was trans-
`fected with single strand and duplex siRNAs at 100 and
`10 nM concentrations. For dsRNAs, 10 nM is likely a
`saturating concentration as 100 nM did not result in sig-
`nificant improvements in observed target knockdown.
`Therefore,
`two siRNAs were also screened at 1 nM.
`Target knockdown was measured 24 h later
`and
`compared to unmodified controls. Figure 1 shows the
`results for ApoB (8786), though a similar trend was
`observed
`for
`the
`other
`five
`siRNAs
`tested
`(Supplementary Figure S1). Generally, dsRNAs were
`broadly tolerant of modifications
`though there was
`0
`-deoxy which is consistent with
`notable sensitivity to 2
`Ago2 preference for RNA (rather than DNA) substrates
`(13,14). High percentage methoxy modification of the
`guide strand was tolerated within dsRNAs though a loss
`in knockdown was seen at the submaximal 1 nM concen-
`tration. Such tolerance of methoxy modification is likely
`
`Table 2. Summary of modification strategy used to evaluate structure–activity relationship for single strand and duplex siRNAs
`0
`0
`p
`5
`aba
`5
`+/ +/
`+/ +/
`+/ +/
`+/ +/
`+/ +/
`+/ +/
`+/ +/
`+/ +/
`+/ +/
`+/ +/
`
`Key
`
`Modifications
`
`Y=r, R=r
`Y=d, R=d
`Y=f, R=f
`Y=m, R=m
`Y=f, R=r
`Y=f, R=d
`Y=f, R=m
`Y=r, R=f
`Y=d, R=f
`Y=m, R=f
`0
`
`Pyr (Y)
`0
`2
`0
`2
`0
`2
`0
`2
`0
`2
`0
`2
`0
`2
`0
`2
`0
`2
`0
`2
`
`OH
`H
`F
`OMe
`F
`F
`F
`OH
`H
`OMe
`
`2
`2
`2
`2
`2
`2
`2
`2
`2
`2
`
`Pur (R)
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`
`OH
`H
`F
`OMe
`OH
`H
`OMe
`F
`F
`F
`
`0
`0
`PO4 were tested and with or without three abasic
`p). Additionally, oligos containing a 5
`phosphate (5
`Oligos were prepared with or without 5
`0
`0
`0
`0
`0
`0
`0
`0
`0
`end of the guide strand (5
`aba). Unmodified ribose (2
`OH) is compared to 2
`-fluoro (2
`F), 2
`-deoxy (2
`H), and 2
`-methoxy (2
`OMe)
`residues at the 5
`modifications assigned to pyrimidines (Pyr) or purines (Pur). Key column lists nomenclature used in subsequent figures. Structures are shown with
`adenosine as the representative nucleotide. A total of 30 different siRNAs were designed for each target site. Sequences and modification patterns are
`listed in Supplementary Table S3.
`
`

`

`Nucleic Acids Research, 2012, Vol. 40, No. 9 4129
`
`dsRNAi (100nM)
`dsRNAi (10nM)
`dsRNAi (1nM)
`ssRNAi (100nM)
`ssRNAi (10nM)
`
`120
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`-20
`
`-40
`
`ApoB (8786) target site
`
`ApoB mRNA knockdown (%)
`
`Y=f, R=r
`
`Y=f, R=d
`
`Y=f, R= m
`
`Y=r, R=f
`
`Y=d, R=f
`Y= m, R=f
`
`-60
`
`Y=r, R=r
`Y=d, R=d
`
`Y=f, R=f
`Y= m, R= m
`
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`
`2'OH
`
`2'F
`
`2'H
`
`2'OMe
`
`Figure 1. Comparison of ssRNA and dsRNA oligos containing pyrimidine (Y) and purine (R) 2’ modifications. ApoB (8786) sequence shown.
`In vitro cell-based evaluation of ApoB mRNA knockdown (error bars represent standard deviation of four replicates). Unmodified ribose (‘r’) is
`compared to 2’-fluoro (‘f’), 2’-methoxy (‘m’), and 2’-deoxy (‘d’) in all combinations on purines (R) and pyrimidines (Y). Oligos do not contain a 5’
`phosphate. Guide strand oligo sequence and color-coded modification patterns are depicted. Five other siRNAs were tested and a similar require-
`ment of 2’F for ssRNA knockdown was observed (Supplementary Figure S1).
`
`0
`
`OH at position 14 of the
`due to our inclusion of 2
`guide strand which has previously been reported as
`highly sensitive to methoxy modifications (17).
`0
`F content exhibit
`Strikingly, single strands containing 2
`the most pronounced mRNA knockdown of the various
`single-strand modification patterns
`tested with 67%
`knockdown compared to the negligible activity seen
`for unmodified (Figure 1, Supplementary Table S3). Five
`other siRNA sequences were also evaluated and a similar
`0
`F dependence for ssRNA activity was observed, though
`2
`the magnitude of response was variable (Supplementary
`Figure S1). This variance is likely attributable to the fact
`that these RNA oligos were not phosphorylated, and there
`0
`phosphorylation of the
`is precedent in the literature that 5
`guide strand is key for ssRNA knockdown (11,12).
`0
`
`5
`
`phosphorylation improves the potency of ssRNA
`
`The same series of RNA modifications evaluated in
`0
`phosphor-
`Figure 1 were then tested in the presence of 5
`ylation of the guide strand. Figure 2 shows that addition
`0
`phosphate significantly improves the potency of
`of a 5
`ssRNA, resulting in significant knockdown even at a
`lower concentration of 10 nM. The overall effectiveness
`0
`of 2
`F-modified ssRNA activity for
`the other five
`siRNAs is also more evident when the ssRNAs are
`phosphorylated (Supplementary Figure S2).
`In the
`0
`presence of 5
`phosphorylation, the beneficial impact of
`0
`F content is significantly more obvious (compare to
`2
`Supplementary Figure S1). While the ssRNA knockdown
`at
`this
`lower
`concentration still
`lags
`that of
`the
`
`0
`
`F
`the combination of 2
`comparable duplex siRNAs,
`0
`modifications and 5
`phosphorylation are key to
`improving
`the
`potency
`of
`ssRNA knockdown:
`improving knockdown from negligible levels to 65–95%
`(Supplementary Table S3).
`0
`Pyrimidines modified with 2
`-fluoro appear to confer
`improved
`knockdown
`to
`ssRNAs
`than
`similarly
`0
`F
`modified purines. It does not seem that overall 2
`content of the RNA oligo can explain this difference as
`the ApoB siRNAs are either approximately evenly split
`between purine and pyrimidine content or are purine
`rich (58% purine). The Ssb sequences are predominantly
`pyrimidine rich (58–74%). The underlying mechanism for
`0
`F pyrimidine preference for ssRNA silencing activity
`2
`remains unclear. Overall, phosphorylated ssRNAs con-
`0
`F modifications of both purines and pyrimidines
`taining 2
`exhibited the best knockdown activity in vitro.
`
`ssRNA potency significantly underperforms
`duplex siRNAs
`
`At the two concentrations tested, ssRNAs appear less
`potent than their corresponding duplex siRNAs. Two
`ApoB siRNAs were selected for dose response measure-
`ments and calculation of EC50 values
`(Figure 3).
`0
`F-modified RNAs were compared as
`Unmodified and 2
`single strands and duplexes over an eight-point dose–
`response ranging from 40 nM down to 0.002 nM (all
`0
`F modifica-
`oligos were phosphorylated). The value of 2
`tions for ssRNA activity was immediately evident for both
`ApoB (8786) and (6981) sequences: unmodified ssRNAs
`
`

`

`dsRNAi (100nM)
`dsRNAi (10nM)
`dsRNAi (1nM)
`ssRNAi (100nM)
`ssRNAi (10nM)
`
`4130 Nucleic Acids Research, 2012, Vol. 40, No. 9
`
`120
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`-20
`
`-40
`
`ApoB (8786) target site
`
`ApoB mRNA knockdown (%)
`
`-60
`
`Y=f, R=f, 5'p
`Y=r, R=r, 5'p
`Y=d, R=f, 5'p
`Y=f, R=d, 5'p
`Y=r, R=f, 5'p
`Y=f, R=r, 5'p
`Y= m, R=f, 5'p
`Y=d, R=d, 5'p
`Y=f, R= m, 5'p
`Y= m, R= m, 5'p
`
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`U U A A G A G A A G C C U U A C U G G U U
`
`p
`
`p
`
`p
`
`p
`
`p
`
`p
`
`p
`
`p
`
`p
`
`p
`
`2'OH
`
`2'F
`
`2'H
`0
`0
`phosphorylation of the guide strand.
`modifications for ApoB (8786) in the presence of 5
`Figure 2. Comparison of purine versus pyrimidine 2
`In vitro cell-based evaluation of ApoB mRNA knockdown (error bars represent standard deviation of four replicates). Unmodified ribose (‘r’) is
`0
`0
`0
`0
`-fluoro (‘f’), 2
`-methoxy (‘m’), and 2
`-deoxy (‘d’) in all combinations on purines (R) and pyrimidines (Y). All RNA oligos contain 5
`compared to 2
`0
`0
`phosphate (5
`p). Guide strand oligo sequence and color-coded modification patterns are depicted. Five other siRNAs with were also tested with 5
`0
`phosphorylation for optimal in vitro ssRNA knockdown was observed
`phosphorylation (Supplementary Figure S2) and a similar requirement for 5
`(compare to Supplementary Figure S1).
`
`2'OMe
`
`0
`0
`Figure 3. In vitro potency of single strands compared to duplex siRNAs. (A) ApoB (8786) and (B) ApoB (6981) compared as 2
`OH and 2
`F guide
`phosphorylation. Single strands are 20- to 80-fold less potent than the corresponding duplex siRNAs containing identical guide
`0
`strands with 5
`strands. Potency and mRNA knockdown values listed in Supplementary Table S1.
`
`essentially had no knockdown over the dose range while
`0
`2
`F ssRNA exhibited a dose response. However, the
`duplex siRNAs were found to be 20- to 80-fold more
`potent
`than their corresponding single strands with
`picomolar potencies
`for dsRNAs compared to the
`nanomolar
`potencies
`measured
`for
`ssRNAs
`(Supplementary Table S1). ApoB (8786) duplexes
`0
`possess 15 pM potencies (for both unmodified and 2
`F)
`
`F ssRNA EC50 is 1.2 nM. The ApoB (6981)
`0
`while the 2
`F duplex is 2-fold more potent than the unmodified
`0
`2
`0
`duplex (2
`OH); however, both are significantly more
`F ssRNA (0.9 nM). Despite the
`0
`potent
`than the 2
`lower overall potencies relative to duplexes, it is important
`to note that ssRNAs are still capable of maximal mRNA
`knockdown >90% and that this level of knockdown was
`enabled by chemical modifications.
`
`

`

`Nucleic Acids Research, 2012, Vol. 40, No. 9 4131
`
`ApoB (6981) target site
`
`B
`
`120
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`(%) (mean +/- SD)
`
`ApoB mRNA Knockdown
`
`ApoB (8786) target site
`
`120
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`A
`
`(%) (mean +/- SD)
`
`ApoB mRNA Knockdown
`
`-20
`
`P B S
`
`single (2'F, 5'p)
`duplex (2'F, 5'p)duplex (2'O H, 5'p)single (2'O H, 5'p)
`
`
`
`
`
`-20
`
`P B S
`
`single (2'F, 5'p)
`duplex (2'F, 5'p)
`duplex (2'O H, 5'p)single (2'O H, 5'p)
`
`
`
`ApoB (6981) target site
`Day7
`
`Day14
`
`Day2
`
`D
`
`120
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`-20
`
`(%) (mean +/- SD)
`
`ApoB mRNA Knockdown
`
`ApoB (8786) target site
`Day2
`Day7
`Day14
`
`120
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`-20
`
`C
`
`(%) (mean +/- SD)
`
`ApoB mRNA Knockdown
`
`P B S
`
`P B S
`
`P B S
`
`duplex (2'F, 5'p)
`single (2'F, 5'p)
`duplex (2'F, 5'p)single (2'F, 5'p)
`
`duplex (2'F, 5'p)
`single (2'F, 5'p)
`
`P B S
`
`
`
`duplex (2'F, 5'p)single (2'F, 5'p)duplex (2'F, 5'p)
`
`
`
`P B S
`
`P B S
`
`
`
`single (2'F, 5'p)single (2'F, 5'p)duplex (2'F, 5'p)
`
`Figure 4. In vivo knockdown of single strand versus duplex siRNA demonstrates ssRNAi activity in vivo. RNAs formulated in a lipid nanoparticle
`(LNP) delivery vehicle. Modified guide strands (2’F) are compared to unmodified (2’OH). All oligos contain 5’ phosphorylation. Comparison of Day
`3 ApoB mRNA knockdown after an LNP delivered 3 mg/kg dose of dsRNA or ssRNA for target sites (A) ApoB (8786) and (B) ApoB (6981).
`Knockdown of 60–75% was observed for phosphorylated single strands containing 2’F modifications. Duration of mRNA knockdown after an LNP
`delivered 6 mg/kg dose was measured over 2 weeks (Day 2, Day 7, Day 14 time points) for (C) ApoB (8786) and (D) ApoB (6981). Knockdown
`values shown in Supplementary Table S2.
`
`ssRNA knockdown in vivo
`
`We extended our comparison of ssRNA and dsRNA into
`mice to identify whether ssRNAs were competent for
`knockdown in vivo. Single strand and duplex RNAs
`were formulated in lipid nanoparticles (LNP) which
`permitted side by side comparison of these RNAs while
`using a well-established delivery vehicle with proven
`capabilities (30,32). Since the nanoparticle encapsulates
`the siRNAs, it is likely shielded from serum nucleases
`which eliminated an additional
`round of
`siRNA
`chemical optimization for serum stability. The same
`ApoB siRNAs assayed for cell-based potency (Figure 3)
`were formulated in LNPs and intravenously dosed at
`3 mg/kg into mice. Three days later the mice were
`sacrificed and ApoB mRNA knockdown was measured
`from harvested livers. Compared to buffer control, the
`duplex siRNAs reduced ApoB expression by 85–98%
`(Figure 4A and B and Supplementary Table S2).
`Unmodified ssRNAs had no significant knockdown
`consistent with in
`vitro observations.
`which was
`0
`However 2
`F-modified single strands were quite effective,
`reducing ApoB expression levels by 61–74% and
`demonstrating that ssRNA functions in vivo.
`
`To evaluate duration of knockdown, a separate in vivo
`0
`F-modified ssRNA
`study was then conducted with the 2
`and dsRNA by using a higher dose (6 mg/kg) and con-
`ducting a 2-week time-course. Groups of mice were
`sacrificed at Days 2, 7, and 14, and ApoB mRNA
`knockdown was measured (Figure 4C and D). At the
`higher dose, ssRNA knockdown increased to nearly
`90% at Day 2, although the effect was transient—
`knockdown reduced to 71–77% at 7 days and returned
`to baseline after 14 days. In marked contrast, duplex
`siRNAs still had 95% knockdown two weeks after
`dosing. In summary, chemical modifications permit effec

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