throbber
J. Med. Chem. 2005, 48, 901-904
`
`901
`
`Fully 2¢-Modified Oligonucleotide
`Duplexes with Improved in Vitro
`Potency and Stability Compared to
`Unmodified Small Interfering RNA
`
`Charles R. Allerson,*,† Namir Sioufi,‡ Russell Jarres,‡
`Thazha P. Prakash,† Nishant Naik,† Andres Berdeja,§
`Lisa Wanders,§ Richard H. Griffey,†
`Eric E. Swayze,† and Balkrishen Bhat†
`Departments of Medicinal Chemistry, Antisense Lead
`Identification, and Research Chemistry,
`Isis Pharmaceuticals, 2292 Faraday Avenue,
`Carlsbad, California 92008
`Received October 18, 2004
`
`Abstract: We have identified a small
`interfering RNA
`(siRNA) motif, consisting entirely of 2¢ -O-methyl and 2¢ -fluoro
`nucleotides, that displays enhanced plasma stability and
`increased in vitro potency. At one site, this motif showed
`remarkable >500-fold improvement in potency over the un-
`modified siRNA. This marks the first report of such a potent
`fully modified motif, which may represent a useful design for
`therapeutic oligonucleotides.
`The specific and reversible modulation of gene ex-
`pression through the use of short synthetic oligonucle-
`otides has proven useful in the study of gene function
`and as a therapeutic mechanism in man.1 Recently,
`RNA interference (RNAi) has emerged as a novel
`mechanism that is activated in mammalian cells by
`small interfering RNAs (siRNAs), short RNA duplexes
`with strands of 21-23 nucleotides.2,3 Once inside the
`cell, siRNAs associate with proteins to form an RNA-
`induced silencing complex (RISC).4 A helicase activity
`associated with RISC separates the two strands of the
`duplex,5 releasing the sense strand and permitting the
`binding of the antisense strand to its messenger RNA
`(mRNA) target. The resulting duplex is a substrate for
`a RISC-associated nuclease, recently identified as Ago2
`(also known as eIF2C2 in man),6 which cleaves the
`target transcript at a single site.5,7
`Despite attempts to use siRNA in vivo,8-14 the reduc-
`tion of endogenous target mRNAs through the systemic
`delivery of siRNA has proven difficult. Although there
`have been conflicting reports on the nuclease stability
`of unmodified siRNA duplexes, there is evidence that
`many are degraded within minutes in mammalian
`serum.9,15,16 It seems likely that siRNAs with increased
`nuclease stability will have a better chance at eliciting
`an in vivo response. Efforts to determine the optimal
`use of stabilizing chemistries have been the focus of
`several recent reports.15,17-21 From these studies, the
`2¢ -O-methyl (2¢ -OMe) and 2¢ -deoxy-2¢ -fluoro (2¢ -F) modi-
`fications have shown promise in stabilizing siRNA
`without disrupting the efficiency of mRNA target reduc-
`tion. To further optimize the use of these and other
`chemistries in siRNA, we are conducting an extensive
`
`* To whom correspondence should be addressed. Phone: 760-603-
`4697. Fax: 760-603-4654. E-mail: callerson@isisph.com.
`† Department of Medicinal Chemistry.
`‡ Department of Antisense Lead Identification.
`§ Department of Research Chemistry.
`
`SAR analysis of chemically modified siRNA. One of the
`most promising designs from these studies is a fully
`modified duplex that consists of alternating 2¢ -OMe and
`2¢-F nucleotides. Surprisingly, duplexes with this sub-
`stitution display increased in vitro potency and in-
`creased stability. Here, we report on this remarkably
`active motif and present our preliminary findings.
`The duplexes used in our SAR study were designed
`to target one of two sites within the coding region of
`the human PTEN mRNA, both previously reported as
`valid target sites for siRNA.22 Modified and unmodified
`duplexes were introduced into HeLa cells using a
`cationic lipid transfection reagent (lipofectin). In vitro
`activity was measured by performing RT-PCR on the
`PTEN mRNA and comparing to mRNA levels of un-
`treated cells. All PTEN signals were normalized to total
`RNA, as measured with RiboGreen.23
`Natural Dicer-generated siRNAs have 2-nucleotide 3¢
`overhangs on both ends of the short duplex.2 In an effort
`to mimick this design, most synthetic siRNAs are
`designed with 3¢ overhangs, typically with the sequence
`dTdT. Several recent reports have provided evidence
`that these overhangs bind to the PAZ domains found
`in numerous proteins, including the Ago2 component
`of RISC, perhaps functioning as a specificity determi-
`nant.24-28 However, there have been other reports that
`blunt-ended RNA duplexes can function equally well
`and may be more stable to exonucleolytic degradation.21
`Given our goal of developing duplexes with optimal
`stability and activity, we explored the use of blunt-ended
`duplexes at both PTEN target sites (sites A and B). The
`activities of 19-base-pair duplexes having 3¢ -dTdT over-
`hangs (1 and 4) were compared to those of otherwise
`identical blunt-ended duplexes (2 and 5) in a 10-point
`dose-response experiment (Figure 1). We found only a
`minimal impact on activity with the removal of the
`overhangs. To confirm the specificity of PTEN mRNA
`reduction, duplexes containing six mismatches (3 and
`6) to the target site were included as negative controls
`in each experiment. Neither control affected PTEN
`mRNA or total RNA levels.
`After validation of the use of blunt-ended constructs,
`the remaining SAR was performed without the use of
`3¢ overhangs. Among the duplex designs examined were
`several that belonged to a class of “alternating” motifs
`in which one type of modified nucleotide was placed in
`alternating positions with a ribonucleotide or a different
`modified nucleotide. In a recent publication, motifs of
`this type, using 2¢-OMe and unmodified (2¢-OH) nucle-
`otides, were shown to have enhanced serum stability
`and single dose in vitro activities similar to those of the
`corresponding unmodified blunt-ended duplexes.21 Dur-
`ing the course of our studies, we examined siRNAs with
`the same 2¢-OMe/2¢-OH substitution. We found that
`duplexes with this motif had in vitro potencies similar
`to those of unmodified siRNAs (see Supporting Informa-
`tion). As part of our broader SAR studies, however, we
`also examined the effect of combining the 2¢-OMe
`substitution with other chemistries, such as 2¢ -F, which
`has already been shown to be well-tolerated in siRNA.15
`This led to the identification of our most potent con-
`
`10.1021/jm049167j CCC: $30.25 © 2005 American Chemical Society
`Published on Web 01/20/2005
`
`Alnylam Exh. 1016
`
`

`

`902 Journal of Medicinal Chemistry, 2005, Vol. 48, No. 4
`
`Letters
`
`Site A
`AAGUAAGGACCAGAGACAAdTdT-3'
`5•.
`3'-dTdTUUCAUUCCUGGUCUCUGUU
`-5'
`AAGUAAGGACCAGAGACAA
`5•.
`UUCAUUCCUGGUCUCUGUU
`3'·
`s·-
`3'-
`
`-3'
`-5'
`
`-3'
`-5'
`
`AAGiAAiGAWuG~GA~AA
`UUC UU CU UC CU UU
`
`2
`
`3
`
`A
`
`140
`
`120
`
`! 100
`e
`~ 80
`
`80 J 40
`
`20
`
`0 ...J....L...1.-'-~-'-'-'--'-'--'-....... -'-.L...l.-'-~-'-'-'--'-'--'-....... -'-.L...1....J...L..l.-'-L...J....L.J..J
`~~~~,§§~~~:~:~i! §§~~~~~:~~! §§S~~~q~~~~
`0000
`0000
`OOOOOON~
`-
`3
`
`2
`
`SiteB
`
`4
`
`5
`
`6
`
`s·-
`GGGUAAAUACAUUCUUCAUdTdT-3'
`3'-dTdTCCCAUUUAUGUAAGAAGUA
`-5'
`s·-
`GGGUAAAUACAUUCUUCAU
`3'-
`CCCAUUUAUGUAAGAAGUA
`GGGAAA~UAW.UU~UUUAU
`S'-
`3'-
`CCCUUU£AUA!,.AA£AAAUA
`
`-3'
`-s·
`-3'
`-5'
`
`B
`
`140
`
`120
`
`" ~ 100
`e
`z 80
`t:
`c 60
`~ 40
`if
`
`20
`
`4
`5
`6
`Figure 1. Reduction of endogenous PTEN mRNA in HeLa
`cells by siRNAs having 3¢ -dTdT overhangs or blunt ends. HeLa
`cells were transfected with siRNAs at the indicated concentra-
`tions in the presence of lipofectin and treated for 20 h followed
`by lysis and RT-PCR. Message levels are reported as percent
`of PTEN mRNA from untreated cells. The bottom strand of
`each duplex is complementary to the target mRNA (mis-
`matches to the target site are indicated with an underline).
`(A) PTEN mRNA reduction by siRNAs targeted to site A. Cells
`were also treated with a duplex containing six mismatches to
`the target site as a negative control (3). (B) PTEN mRNA
`reduction by siRNAs targeted to site B. Cells were also treated
`with a duplex containing six mismatches to the target site as
`a negative control (6).
`
`struct design, in which both strands were substituted
`with alternating 2¢-OMe and 2 ¢-F nucleotides (Figure
`2). From an eight-point dose-response analysis in HeLa
`cells, we were able to estimate IC50 values of each duplex
`(Table 1; see also Supporting Information).
`At site A, the duplex with alternating 2¢-F/2¢-OMe
`chemistry (7, Figure 2) had biological activity that was
`roughly equivalent to that of the parent siRNA (2). At
`site B, however, the 2¢-F/2¢-OMe duplex ( 10) displayed
`dramatically improved potency. Even at the lowest
`concentration (2 pM), target reduction was greater than
`75% relative to untreated control, minimally reflecting
`500-fold improvement in potency over the unmodified
`siRNA (5). Throughout these studies, we were cognizant
`of the importance of the 5¢ -phosphates normally present
`on Dicer-generated siRNA duplexes. The 5¢-phosphate
`on the antisense strand has been shown to be critical
`for efficient assembly and activation of RISC in mam-
`malian systems.29 However, it has been shown that
`
`A
`
`120
`
`100
`
`80
`
`"' z
`0: e
`z
`t: 80
`! 40
`i 20
`
`B
`
`120
`
`2
`
`7
`
`8
`
`9
`
`-3'
`.5•
`
`SlteA
`5'- AAGUAAGGACCAGAGACAA
`3'- UUCAUUCCUGGUCUCUGUU
`S'- AAGUAAGGACCAGAGACAA -3'
`3'- UUCAUUCCUGGUCUCUGUU -s· N: 2'-0Me
`N=2'-F
`S'-P-AAGUAAGGACCAGAGACAA -3'
`P • 5'aphosphate
`3'- UUCAUUCCUGGUCUCUGUU.P-5'
`s·- AAGCAACGAGAAGCGAUAA
`-3'
`3'- UUC~ UU~CUJ.ll.UC~CUalJU -5'
`
`2
`
`7
`
`8
`
`9
`
`5
`
`10
`
`11
`
`12
`
`Site B
`-3'
`S'- GGGUAAAUACAUUCUUCAU
`3•. CCCAUUUAUGUAAGAAGUA
`-5'
`s·- GGGUAAAUACAUUCUUCAU -3'
`3•. CCCAUUUAUGUAAGAAGUA -5'
`S'-P-GGGUAAAUACAUUCUUCAU -3'
`3'- CCCAUUUAUGUAAGAAGUA.P-5'
`s·- ggg~~~~~~~~~l -3'
`
`3'-
`
`-5'
`
`N = 2'-0Mc
`N = 2'-F
`P - S'.phosphate
`
`100
`
`80
`
`80
`
`" z
`0: e
`z
`"'
`Ii: I 40
`
`0.
`
`20
`
`ONCO .... NOOOO
`
`!!!~ONIDIO~
`
`10
`11
`12
`5
`Figure 2. Reduction of endogenous PTEN mRNA in HeLa
`cells by unmodified or fully 2¢ -F/2¢ -OMe modified 19-base-pair
`oligonucleotide duplexes. HeLa cells were transfected with
`siRNAs at the indicated concentrations in the presence of
`lipofectin and treated for 20 h followed by lysis and RT-PCR.
`Message levels are reported as percent of PTEN mRNA from
`untreated cells. The bottom strand of each duplex is comple-
`mentary to the target mRNA (mismatches to the target site
`are indicated with an underline). The 2¢-F modification is
`indicated in green, the 2¢-OMe modification is indicated in
`purple, while the presence of a 5¢-phosphate is indicated by
`the letter “P”. (A) PTEN mRNA reduction by duplexes targeted
`to site A. (B) PTEN mRNA reduction by duplexes targeted to
`site B.
`
`Tm (°C)
`72.8
`93.9
`nd
`62.0
`nd
`82.0
`
`plasma half-life (min)
`(cid:24)30
`nd
`>420
`nd
`nd
`>420
`
`Table 1. Summary of in Vitro Activity, Plasma Stability, and
`Duplex Thermal Stabilitya
`duplex
`site
`IC50 (nM)
`2
`A
`0.26
`7
`A
`0.31
`8
`A
`0.068
`5
`B
`0.81
`<0.002
`10
`B
`<0.002
`11
`B
`a nd ) not determined.
`prephosphorylation is generally unnecessary because
`the siRNAs are phosphorylated by endogenous kinases.
`Anticipating that chemically modified duplexes might
`be less efficiently processed by these kinases,30 we
`compared the activities of 2¢ -F/2¢ -OMe duplexes with
`preestablished 5¢-phosphates (Figure 2, 8 and 11). At
`site A, addition of a synthetic 5¢ -phosphate produced
`
`

`

`Letters
`
`Journal of Medicinal Chemistry, 2005, Vol. 48, No. 4 903
`
`modest improvement (5-fold) in potency (Table 1). Given
`the already potent activity of 10, we were unable to
`resolve any beneficial effect of 5¢ -phosphorylation at site
`B. Similar effects from phosphorylation have been
`observed with this motif on other targets, where mod-
`estly active duplexes show improvement of in vitro
`potency upon addition of the 5¢ -phosphate, while the
`effect on extremely potent duplexes is difficult to resolve
`(data not shown). Furthermore, the 5¢ -phosphate on the
`antisense strand appears to be largely responsible for
`this improvement in potency, although the magnitude
`of the effect may depend on cell type (data not shown).
`We also prepared and tested modified duplexes that
`contained six mismatches to either of the target sites
`(9 and 12). Neither mismatch-containing duplex pro-
`duced a significant reduction in PTEN mRNA or total
`RNA.
`To examine the relative serum stabilities of these
`duplexes, we tested unmodified siRNA 2 and 2¢ -F/2¢ -
`OMe modified duplexes 8 and 11 for their ability to
`resist degradation in mouse plasma. Each of the du-
`plexes was treated with 25% mouse plasma at 37 °C
`for up to 7 h. At various time points, aliquots were
`removed and examined for intact duplex by capillary
`gel electrophoresis.31,32 From these measurements, we
`plotted percent intact duplex against time and assessed
`the relative stabilities of the three duplexes (Figure 3A,
`Table 1). From this straightforward analysis, the stabil-
`ity of the 2¢ -F/2¢ -OMe duplexes was striking. Even after
`7 h, the duplexes were greater than 60% (8) or 70% (11)
`intact. While there was an initial loss of duplex in each
`case, this may correspond to rapid loss of imperfectly
`annealed strands. Regardless, the remaining duplex
`degrades at a rate that suggests a half-life of much
`greater than 7 h.
`It has previously been shown that the use of 2¢-F
`modifications can increase the thermal stability of
`oligonucleotide duplexes.33 Because duplexes 7, 8, 10,
`and 11 each contain a total of 19 2¢-F nucleotides, we
`anticipated that these duplexes might have much
`greater thermal stabilities than their unmodified coun-
`terparts. To examine this possibility, we measured the
`thermal stabilities of two control duplexes (2 and 5) and
`two modified duplexes (7 and 11) (Figure 3B). In each
`case we see a roughly 20 °C increase in Tm with the
`fully modified duplexes. This corresponds to slightly
`more than 1 °C increase in Tm per 2¢ -F substitution. This
`increase in thermal stability is likely to explain in part
`the enhanced plasma stability of the modified duplexes
`and may improve the interaction between the antisense
`strand and the target mRNA.
`It is surprising that duplexes with such high thermal
`stabilities can function so efficiently and potently rela-
`tive to the unmodified siRNAs. Recent studies have
`suggested that RISC chooses which strand to retain on
`the basis of differences in thermodynamic stability at
`the 5¢ ends of the strands, with the strand having the
`5¢ end of lower stability being more likely to load into
`RISC.34,35 It is worth noting that one end of the site B
`duplex contains three consecutive G-C base pairs.
`Although the introduction of 2¢-F nucleotides should
`raise the thermodynamic stability of both ends of the
`duplex, it may increase the already more-stable end
`above a critical threshold that the RISC-associated
`
`A
`
`100
`
`~
`
`80
`
`60
`
`><
`GI
`Q.
`:I
`C
`tl
`J!
`.5
`c 40
`GI
`I:?
`GI
`D.
`
`20
`
`0
`
`0
`
`60
`
`120
`
`180
`
`240
`
`300
`
`360
`
`420
`
`Time (minutes)
`
`B 1.4
`
`(.)
`
`GI 1.3
`C .,,
`-e
`0
`ti) 1.2
`.a cc
`"C -~ ii 1.1
`E ...
`0 z
`
`1.0
`
`0.9
`
`•
`(cid:127)
`...
`,.
`
`2
`7
`5
`11
`
`20
`
`30
`
`40
`
`50
`
`60
`
`70
`
`80
`
`90
`
`100
`
`Temperature (degrees C)
`Figure 3. Comparison of the biophysical properties of un-
`modified blunt-ended siRNAs and 2¢ -F/2¢ -OMe modified
`duplexes. (A) Stability of duplexes in 25% (v/v) mouse plasma.
`Duplexes were treated with 25% mouse plasma at 37 °C for
`the indicated times, then examined by capillary gel electro-
`phoresis to determine the amount of intact duplex (see
`Supporting Information for details). The plots shown are the
`result of two independent experiments. (B) Thermal denatur-
`ation profiles of both unmodified and modified duplexes in 100
`mM NaCl, 10 mM sodium phosphate, pH 7.5, 0.1 mM EDTA,
`and 4 (cid:237)M of each strand. Absorbance at 260 nm was measured
`as the temperature was raised from 15 to 85 °C or from 30 to
`95 °C. Absorbances were normalized to facilitate graphical
`depiction. Melting temperatures (Tm) were calculated from
`first-derivative curves of at least two separate experiments and
`are summarized in Table 1.
`helicase cannot overcome, shifting the bias for strand-
`loading more in favor of the antisense strand. We also
`observed that among the unmodified duplexes, the best
`activity comes from the duplex having the highest Tm,
`whereas the opposite is true for the 2¢-F/2¢-OMe motif.
`Although not definitive, these relationships may hint
`at the existence of an optimal thermal stability.
`From these observations, the alternating 2¢ -F/2¢ -OMe
`motif appears to be an attractive design for creating
`functionally active and stable RNA duplexes. Clearly,
`these duplexes potently reduce levels of endogenous
`target mRNA, with the addition of a 5¢ -phosphate to the
`antisense strand further enhancing in vitro potency. The
`biophysical properties of this duplex motif, reflected in
`its enhanced serum and thermal stability, also favor its
`chances at surviving in serum, which will hopefully
`
`

`

`904 Journal of Medicinal Chemistry, 2005, Vol. 48, No. 4
`
`translate to improved in vivo potency. The identification
`of this motif from our SAR analysis highlights the value
`of screening oligonucleotides containing multiple chem-
`istries and may prove a useful strategy in related areas
`such as micro-RNA. Although promising, the utility and
`optimal design of this motif for in vivo applications and
`a biochemical explanation for the remarkable increase
`in potency remain to be determined and are the focus
`of ongoing studies. We anticipate that this and other
`chemically modified duplex motifs will ultimately prove
`useful in the design of clinically active nucleic acids.
`
`Acknowledgment. The authors thank Bruce Ross,
`Mystie Nguyen, and Mingming Han for providing the
`2¢ -O-methyl and 2¢ -deoxy-2¢ -fluoro nucleoside phos-
`phoramidites, Hans Gaus, Sam Lee, and Jodee Stein-
`berg for their assistance in oligonucleotide analysis, and
`Prasad Dande, Sue Freier, Bridget Lollo, and Brenda
`Baker for their thoughtful discussions about this project.
`
`Supporting Information Available: Methods for the
`synthesis and purification of the oligonucleotides, mass
`spectral and capillary gel electrophoresis data of each com-
`pound, and details of the biological and biochemical assays.
`This material is available free of charge via the Internet at
`http://pubs.acs.org.
`
`References
`(1) Crooke, S. T. Molecular mechanisms of action of antisense drugs.
`Biochim. Biophys. Acta 1999, 1489, 31-44.
`(2) Elbashir, S. M.; Harborth, J.; Lendeckel, W.; Yalcin, A.; Weber,
`K.; et al. Duplexes of 21-nucleotide RNAs mediate RNA interfer-
`ence in cultured mammalian cells. Nature 2001, 411, 494-498.
`(3) Caplen, N. J.; Parrish, S.; Imani, F.; Fire, A.; Morgan, R. A.
`Specific inhibition of gene expression by small double-stranded
`RNAs in invertebrate and vertebrate systems. Proc. Natl. Acad.
`Sci. U.S.A. 2001, 98, 9742-9747.
`(4) Hammond, S. M.; Bernstein, E.; Beach, D.; Hannon, G. J. An
`RNA-directed nuclease mediates post-transcriptional gene
`silencing in Drosophila cells. Nature 2000, 404, 293-296.
`(5) Martinez, J.; Patkaniowska, A.; Urlaub, H.; Luhrmann, R.;
`Tuschl, T. Single-Stranded Antisense siRNAs Guide Target RNA
`Cleavage in RNAi. Cell 2002, 110, 563-574.
`(6) Liu, J.; Carmell, M. A.; Rivas, F. V.; Marsden, C. G.; Thomson,
`J. M.; et al. Argonaute2 Is the Catalytic Engine of Mammalian
`RNAi. Science (Washington, D.C.) 2004, 305, 1437-1441.
`(7) Haley, B.; Zamore, P. D. Kinetic analysis of the RNAi enzyme
`complex. Nat. Struct. Mol. Biol. 2004, 11, 599-606.
`(8) Zhang, X.; Shan, P.; Jiang, D.; Noble, P. W.; Abraham, N. G.; et
`al. Small Interfering RNA Targeting Heme Oxygenase-1
`Enhances Ischemia-Reperfusion-Induced Lung Apoptosis. J.
`Biol. Chem. 2004, 279, 10677-10684.
`(9) Braasch, D. A.; Paroo, Z.; Constantinescu, A.; Ren, G.; Oz, O.
`K.; et al. Biodistribution of phosphodiester and phosphorothioate
`siRNA. Bioorg. Med. Chem. Lett. 2004, 14, 1139-1143.
`(10) Song, E.; Lee, S. K.; Wang, J.; Ince, N.; Ouyang, N.; et al. RNA
`interference targeting Fas protects mice from fulminant hepa-
`titis. Nat. Med. 2003, 9, 347-351.
`(11) Zender, L.; Huetker, S.; Liedtke, C.; Tillmann, H. L.; Zender,
`S.; et al. Caspase 8 small interfering RNA prevents acute liver
`failure in mice. Proc. Natl. Acad. Sci. U.S.A. 2003, 100, 7797-
`7802.
`(12) Sorensen, D. R.; Leirdal, M.; Sioud, M. Gene Silencing by
`Systemic Delivery of Synthetic siRNAs in Adult Mice. J. Mol.
`Biol. 2003, 327, 761-766.
`(13) Layzer, J. M.; McCaffrey, A. P.; Tanner, A. K.; Huang, Z.; Kay,
`M. A.; et al. In vivo activity of nuclease-resistant siRNAs. RNA
`2004, 10, 766-771.
`(14) Soutschek, J.; Akinc, A.; Bramlage, B.; Charisse, K.; Constien,
`R.; et al. Therapeutic silencing of an endogenous gene by
`systemic administration of modified siRNAs. Nature (London)
`2004, 432, 173-178.
`
`Letters
`
`(15) Chiu, Y. L.; Rana, T. M. siRNA function in RNAi: A chemical
`modification analysis. RNA 2003, 9, 1034-1048.
`(16) Braasch, D. A.; Jensen, S.; Liu, Y.; Kaur, K.; Arar, K.; et al. RNA
`Interference in Mammalian Cells by Chemically-Modified RNA.
`Biochemistry 2003, 42, 7967-7975.
`(17) Capodici, J.; Kariko, K.; Weissman, D. Inhibition of HIV-1
`Infection by Small Interfering RNA-Mediated RNA Interference.
`J. Immunol. 2002, 169, 5196-5201.
`(18) Harborth, J.; Elbashir, S. M.; Vandenburgh, K.; Manninga, H.;
`Scaringe, S. A.; et al. Sequence, Chemical, and Structural
`Variation of Small Interfering RNAs and Short Hairpin RNAs
`and the Effect on Mammalian Gene Silencing. Antisense Nucleic
`Acid Drug Dev. 2003, 13, 83-105.
`(19) Amarzguioui, M.; Holen, T.; Babaie, E.; Prydz, H. Tolerance for
`mutations and chemical modifications in a siRNA. Nucleic Acids
`Res. 2003, 31, 589-595.
`(20) Holen, T.; Amarzguioui, M.; Babaie, E.; Prydz, H. Similar
`behaviour of single-strand and double-strand siRNAs suggests
`they act through a common RNAi pathway. Nucleic Acids Res.
`2003, 31, 2401-2407.
`(21) Czauderna, F.; Fechtner, M.; Dames, S.; Aygun, H.; Klippel, A.;
`et al. Structural variations and stabilising modifications of
`synthetic siRNAs in mammalian cells. Nucleic Acids Res. 2003,
`31, 2705-2716.
`(22) Vickers, T. A.; Koo, S.; Bennett, C. F.; Crooke, S. T.; Dean, N.
`M.; et al. Efficient Reduction of Target RNAs by Small Interfer-
`ing RNA and RNase H-Dependent Antisense Agents. J. Biol.
`Chem. 2003, 278, 7108-7118.
`(23) Hashimoto, J. G.; Beadles-bohling, A. S.; Wiren, K. M. Com-
`parison of RiboGreen and 18S rRNA quantitation for normal-
`izing real-time RT-PCR expression analysis. BioTechniques
`2004, 36, 54, 56, 58-60.
`(24) Lingel, A.; Simon, B.; Izaurralde, E.; Sattler, M. Structure and
`nucleic-acid binding of the Drosophila Argonaute 2 PAZ domain.
`Nature (London) 2003, 426, 465-469.
`(25) Song, J.-J.; Liu, J.; Tolia, N. H.; Schneiderman, J.; Smith, S. K.;
`et al. The crystal structure of the Argonaute2 PAZ domain
`reveals an RNA binding motif in RNAi effector complexes. Nat.
`Struct. Biol. 2003, 10, 1026-1032.
`(26) Yan, K. S.; Yan, S.; Farooq, A.; Han, A.; Zeng, L.; et al. Structure
`and conserved RNA binding of the PAZ domain. Nature (London)
`2003, 426, 469-474.
`(27) Ma, J.-B.; Ye, K.; Patel, D. J. Structural basis for overhang-
`specific small interfering RNA recognition by the PAZ domain.
`Nature (London) 2004, 429, 318-322.
`(28) Lingel, A.; Simon, B.; Izaurralde, E.; Sattler, M. Nucleic acid
`3¢ -end recognition by the Argonaute2 PAZ domain. Nat. Struct.
`Mol. Biol. 2004, 11, 576-577.
`(29) Schwarz, D. S.; Hutva´gner, G.; Haley, B.; Zamore, P. D. Evidence
`That siRNAs Function as Guides, Not Primers, in the Drosophila
`and Human RNAi Pathways. Mol. Cell 2002, 10, 537-548.
`(30) Nykanen, A.; Haley, B.; Zamore, P. D. ATP requirements and
`small
`interfering RNA structure in the RNA interference
`pathway. Cell (Cambridge, Mass.) 2001, 107, 309-321.
`(31) Leeds, J. M.; Graham, M. J.; Truong, L.; Cummins, L. L.
`Quantitation of phosphorothioate oligonucleotides in human
`plasma. Anal. Biochem. 1996, 35, 36-43.
`(32) Geary, R. S.; Matson, J.; Levin, A. A. A nonradioisotope biomedi-
`cal assay for intact oligonucleotide and its chain-shortened
`metabolites used for determination of exposure and elimination
`half-life of antisense drugs in tissue. Anal. Biochem. 1999, 274,
`241-248.
`(33) Kawasaki, A. M.; Casper, M. D.; Freier, S. M.; Lesnik, E. A.;
`Zounes, M. C.; et al. Uniformly modified 2¢ -deoxy-2¢ -fluoro
`phosphorothioate oligonucleotides as nuclease-resistant anti-
`sense compounds with high affinity and specificity for RNA
`targets. J. Med. Chem. 1993, 36, 831-841.
`(34) Schwarz, D. S.; Hutvagner, G.; Du, T.; Xu, Z.; Aronin, N.; et al.
`Asymmetry in the assembly of the RNAi enzyme complex. Cell
`2003, 115, 199-208.
`(35) Khvorova, A.; Reynolds, A.; Jayasena, S. D. Functional siRNAs
`and miRNAs exhibit strand bias. Cell 2003, 115, 209-216.
`JM049167J
`
`

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