hydrophobization and bioconjugation for enhanced sirna ......rnai (elbashir et al. 2001b; meister...

27
10.1261/rna.459807 Access the most recent version at doi: 2007 13: 431-456 originally published online February 28, 2007 RNA Daniel De Paula, M. Vitória L.B. Bentley and Ram I. Mahato and targeting Hydrophobization and bioconjugation for enhanced siRNA delivery References http://rnajournal.cshlp.org/content/13/4/431.full.html#related-urls Article cited in: http://rnajournal.cshlp.org/content/13/4/431.full.html#ref-list-1 This article cites 151 articles, 46 of which can be accessed free at: service Email alerting click here top right corner of the article or Receive free email alerts when new articles cite this article - sign up in the box at the http://rnajournal.cshlp.org/subscriptions go to: RNA To subscribe to Copyright © 2007 RNA Society Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.org Downloaded from

Upload: others

Post on 04-Feb-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

  • 10.1261/rna.459807Access the most recent version at doi: 2007 13: 431-456 originally published online February 28, 2007RNA

    Daniel De Paula, M. Vitória L.B. Bentley and Ram I. Mahato and targetingHydrophobization and bioconjugation for enhanced siRNA delivery

    References

    http://rnajournal.cshlp.org/content/13/4/431.full.html#related-urlsArticle cited in:

    http://rnajournal.cshlp.org/content/13/4/431.full.html#ref-list-1This article cites 151 articles, 46 of which can be accessed free at:

    serviceEmail alerting

    click heretop right corner of the article orReceive free email alerts when new articles cite this article - sign up in the box at the

    http://rnajournal.cshlp.org/subscriptions go to: RNATo subscribe to

    Copyright © 2007 RNA Society

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/lookup/doi/10.1261/rna.459807http://rnajournal.cshlp.org/content/13/4/431.full.html#ref-list-1http://rnajournal.cshlp.org/content/13/4/431.full.html#related-urlshttp://rnajournal.cshlp.org/cgi/alerts/ctalert?alertType=citedby&addAlert=cited_by&saveAlert=no&cited_by_criteria_resid=rna;13/4/431&return_type=article&return_url=http://rnajournal.cshlp.org/content/13/4/431.full.pdfhttp://rnajournal.cshlp.org/subscriptionshttp://rnajournal.cshlp.org/http://www.cshlpress.com

  • REVIEW

    Hydrophobization and bioconjugation for enhanced

    siRNA delivery and targeting

    DANIEL DE PAULA,1,2 M. VITÓRIA L.B. BENTLEY,1 and RAM I. MAHATO2

    1Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil2Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA

    ABSTRACT

    RNA interference (RNAi) is an evolutionarily conserved process by which double-stranded small interfering RNA (siRNA)induces sequence-specific, post-transcriptional gene silencing. Unlike other mRNA targeting strategies, RNAi takes advantage ofthe physiological gene silencing machinery. The potential use of siRNA as therapeutic agents has attracted great attention as anovel approach for treating severe and chronic diseases. RNAi can be achieved by either delivery of chemically synthesizedsiRNAs or endogenous expression of small hairpin RNA, siRNA, and microRNA (miRNA). However, the relatively high dose ofsiRNA required for gene silencing limits its therapeutic applications. This review discusses several strategies to improvetherapeutic efficacy as well as to abrogate off-target effects and immunostimulation caused by siRNAs. There is an in-depthdiscussion on various issues related to the (1) mechanisms of RNAi, (2) methods of siRNA production, (3) barriers to RNAi-basedtherapies, (4) biodistribution, (5) design of siRNA molecules, (6) chemical modification and bioconjugation, (7) complexformation with lipids and polymers, (8) encapsulation into lipid particles, and (9) target specificity for enhanced therapeuticeffectiveness.

    Keywords: RNA interference; small interfering RNA; bioconjugation; chemical modification; complex formation

    INTRODUCTION

    RNA interference (RNAi) is an evolutionarily conservedprocess by which double-stranded small interfering RNA(siRNA) induces sequence-specific, post-transcriptionalgene silencing (Hannon 2002). The process of RNAiconsists of an initiator step, in which long double-strandedRNA (dsRNA) is cleaved into siRNA fragments, and aneffector step, in which these fragments are incorporatedinto a protein complex, dissociated, and used as a guidingsequence to recognize homologous mRNA that is sub-sequently cleaved. RNAi is considered a self-defense mech-anism of eukaryotic cells to combat infection by RNAviruses and transposons. It is also assumed to tightlyregulate protein levels in response to various environmentalstimuli (Meister and Tuschl 2004).

    The revolutionary finding of RNAi resulted from thework of Andrew Fire and coworkers at the CarnegieInstitute in Washington, D.C., who demonstrated in 1998that injection of dsRNA into Caenorhabditis elegans leads toefficient sequence-specific gene silencing (Fire et al. 1998).At that time, the state of the art in gene silencing was theuse of antisense oligodeoxynucleotide (ODNs), whichcomprise single strands of short DNA or RNA comple-mentary sequences that hybridize with the targeted mRNA(Mahato et al. 2005). However, the dsRNA seemed toinduce silencing through a pathway distinct from classicalantisense therapies due to the catalytic nature of RNAi, inwhich one siRNA can be used over and over to guide thecleavage of many mRNA molecules (Dykxhoorn et al.2003). Bertrand et al. (2002) have compared the effects ofantisense ODNs and siRNAs targeting green fluorescentprotein (GFP) in vitro and in vivo. siRNA was quantita-tively more efficient, and its effect lasted for a longer timein cell culture. In mice, siRNAs were able to silence geneexpression, whereas no effect was observed in the presenceof antisense ODNs.

    The first evidence that siRNAs can mediate sequence-specific gene silencing in mammalian cells was provided in

    rna4598 De Paula et al. REVIEW RA

    Abbreviations: See list at end of paper.Reprint requests to: Ram I. Mahato, Department of Pharmaceutical

    Sciences, Health Science Center, University of Tennessee, 26 South DunlapStreet, Feurt Building, Room 413, Memphis, TN 38163, USA; e-mail:[email protected]; fax: (901) 448-6092.

    Article published online ahead of print. Article and publication date areat http://www.rnajournal.org/cgi/doi/10.1261/rna.459807.

    RNA (2007), 13:431–456. Published by Cold Spring Harbor Laboratory Press. Copyright � 2007 RNA Society. 431

    JOBNAME: RNA 13#4 2007 PAGE: 1 OUTPUT: Tuesday March 6 18:39:02 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • 2001 when the conversion of dsRNA into short RNAfragments was shown to be bypassed by the transfectionof siRNA molecules into cells (Elbashir et al. 2001a). Afterthat, various in vivo effects of siRNA and short hairpinRNA (shRNA) have been reported (Lewis et al. 2002;McCaffrey et al. 2002; Xia et al. 2002; Song et al. 2003b).For example, McCaffrey et al. (2002) showed that siRNAand shRNA reduce luciferase (Luc) expression in the liverin a sequence-specific manner. Song et al. (2003b) foundthat siRNA targeted to the Fas receptor protects mice fromliver fibrosis.

    Human clinical trials of RNAi-based drugs are currentlyunder way by Acuity Pharmaceuticals and Sirna Therapeu-tics. Both companies are working on intravitreal adminis-tration of siRNA targeting vascular endothelial growthfactor (VEGF), whose overexpression is the primary causeof age-related macular degeneration (AMD). The firstresults are encouraging in terms of tolerability of siRNAcompounds. Other clinical trials involving siRNA fortreating chronic myeloid leukemia and respiratory syncytialvirus infection are also being carried out by the HadassahMedical Organization and Alnylam Pharmaceutics, Inc.,respectively.

    Despite early excitement, siRNAs have shown to activateimmune response in a sequence- and concentration-depen-dent manner, leading to nonspecific gene silencing (Jacksonet al. 2003; Sioud 2005). In this respect, the introduction ofchemical modifications and generation of designed siRNAshave become essential for achieving high gene silencingwith a low degree of undesired effects. Chemical modifica-tions of siRNAs also appear to stabilize these molecules inserum and show enhanced gene silencing (Braasch et al.2003; Chiu and Rana 2003; Layzer et al. 2004). A break-through in this area has recently been achieved by linkingcholesterol (Chol) to siRNAs (Soutschek et al. 2004), andby encapsulating them into stable nucleic acid lipidparticles (Zimmermann et al. 2006). Undoubtedly, effectivein vivo delivery of siRNAs will be a key factor in turningsiRNA into a new class of therapeutics. In this review, wediscuss several strategies to abrogate undesired effects andimprove the efficiency of siRNA, including chemical modifi-cations, siRNA sequence design, bioconjugation, complexformation, and encapsulation into lipid particles.

    MECHANISM OF RNA INTERFERENCE

    The initial studies that unraveled the RNAi machinery wereobtained from experiments using Drosophila extracts.Figure 1 illustrates the mechanism of RNAi. It involvesprocessing of long dsRNA of 500–1000 nucleotides (nt),which triggers into fragments of siRNA. Generally, siRNAis 21–23-nt dsRNA segments with 39-overhangs of 2 nt onboth strands, and unphosphorylated hydroxyl groups at the29 and 39 sites. The siRNA structure is characteristic of anRNase III-like cleavage pattern and plays a crucial role in its

    recognition by other RNAi components. Dicer is theenzyme known to process dsRNA into siRNAs, and itcontains dual catalytic domains and additional helicase andPAZ domains (Elbashir et al. 2001b; Zamore and Haley2005). Processed siRNA is incorporated into a proteincomplex, termed RNA-induced silencing complex (RISC).Dicer is also involved in the early steps of RISC formationand may be required for siRNA entry into RISC (Lee et al.2004). In mammalian cells, the protein Argonaute 2 (Ago2)is the catalytic component of RISC that cleaves targetmRNAs (Liu et al. 2004b; Meister et al. 2004). In the RISCassembly process, siRNAs are initially loaded into Ago2 asduplexes, and then Ago2 cleaves the passenger strand,thereby liberating the guide strand from the siRNA duplexand producing active RISC capable of cleaving targetmRNAs (Matranga et al. 2005; Leuschner et al. 2006).The guide strand serves as a template for the recognitionof homologous mRNA, which upon binding to RISC iscleaved between bases 10 and 11 relative to the 59 base ofthe guide siRNA strand by the catalytic activity of Ago2(Elbashir et al. 2001c). The template siRNA is not affectedby this reaction, so the RISC can undergo numerous cyclesof mRNA cleavage that comprise the high efficiency ofRNAi (Elbashir et al. 2001b; Meister and Tuschl 2004). Themechanism by which passenger strands are cleaved by Ago2follows the same rules established for the siRNA-guidedcleavage of a target mRNA (Leuschner et al. 2006).

    RNAi can also be induced by endogenous expression ofsmall regulatory RNAs known as microRNAs (miRNAs)(Fig. 1). The generation of miRNAs occurs via sequentialprocessing and maturation of long primary transcripts (pri-miRNA). Pre-microRNA (pre-miRNA) exits in the nucleusupon cleavage by Drosha and is recognized by the endo-nuclease Dicer, which processes the pre-miRNA into a22-nt mature miRNA (Bartel 2004). The mature miRNA issubsequently incorporated into the silencing complex(RISC), where it mediates translational repression. DuringRISC assembly, the Ago2-mediated cleavage of the passen-ger strand is blocked by mismatches between the guideand passenger strands of miRNAs. In this case, a slower‘‘backup’’ pathway dissociates and destroys the passengerstrand, liberating active RISC (Matranga et al. 2005;Leuschner et al. 2006). In contrast to siRNA, miRNAs medi-ate RNAi through a translational inhibition mechanisminvolving imperfect complementarity to target mRNAs(Jackson et al. 2003; Bartel 2004).

    One major difference between mammalian RNAi andRNAi in other eukaryotes is the lack of an amplificationsystem for long-term persistence of RNAi in mammaliancells. For example, in Drosophila, z35 molecules of dsRNAcan silence z1000 copies of a target mRNA per cell and canpersist over the course of many generations (Nykanen et al.2001). In contrast, in mammalian cells RNAi persistseffectively only for an average of 66 h due to its dilutionduring cell divisions (Chiu and Rana 2002).

    De Paula et al.

    432 RNA, Vol. 13, No. 4

    JOBNAME: RNA 13#4 2007 PAGE: 2 OUTPUT: Tuesday March 6 18:39:02 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • METHODS OF siRNA PRODUCTION

    The methods of siRNA production can be classified intothree different categories: (1) chemical synthesis, (2) invitro transcription, and (3) endogenous expression. Thechoice of production method depends on the target gene,cell type, in vitro or in vivo use, and desired extent of genesilencing. Table 1 describes the advantages and disadvan-tages of each method of siRNA production.

    Chemical synthesis

    Chemical synthesis is the most direct means of generatingsiRNAs and has several advantages, including precisecontrol of the amount and purity of siRNA, ease incharacterization and scale-up, and ease in chemical mod-ifications for enhanced stability and target specificity(Elbashir et al. 2001a). The chemical synthesis of siRNArequires several steps including the generation of twohomologous strands, annealing of the strands, addition ofchemical entities to increase stability, and ensuring that

    2-nt overhangs are present. The siRNA duplex requires a39-hydroxyl group and a 59-phosphate group for functionalactivity (Dykxhoorn et al. 2003). Unlike DNA, RNA pos-sesses an additional hydroxyl group at the 29 position ofeach ribose building block, which destabilizes RNA underthe basic conditions generally present in DNA synthesisreactions. Hence, the most difficult step in RNA synthesisis the simultaneous protection of the 59- and 29-hydroxylgroups during solid-phase chemistry.

    One drawback of using chemically synthesized siRNA isthat the most effective target sequence is unpredictablesince gene silencing efficiency may vary depending on thesegments of the transcripts that are targeted. For example,Holen et al. (2002) observed that only a few siRNAsresulted in a significant reduction of human tissue factor(HTF) expression after targeting its mRNA with severalsiRNAs synthesized against different sites of the samemRNA. The results suggested that accessible siRNA targetsites may be rare in some human mRNAs. However, moreeffective gene silencing can be achieved by targetingdifferent segments of the same transcript simultaneously.

    FIGURE 1. Mechanisms of RNA interference (RNAi). RNAi is triggered by long double-stranded RNA (dsRNA), small interfering RNA (siRNA),plasmid or virus-based short hairpin RNA (shRNA), or microRNA (miRNA), and siRNA. Long dsRNA, shRNA, and pre-miRNA are processedby Dicer into 21–23-nt siRNA duplexes with symmetric 2-nt 39 overhangs and 59-phosphate groups. Processed siRNAs assemble with cellularproteins to form an RNA-induced silencing complex (RISC). During RISC assembly, one strand (passenger) is eliminated, while the other strand(guide) produces an active RISC, which eventually triggers the sequence-specific mRNA degradation (in case of total complementarity) ortranslational repression (in case of incorporation of partially complementary miRNA).

    siRNA delivery and targeting

    www.rnajournal.org 433

    JOBNAME: RNA 13#4 2007 PAGE: 3 OUTPUT: Tuesday March 6 18:39:02 2007

    csh/RNA/131733/rna4598

    Fig. 1 live 4/C

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • Significant enhancement in gene silencing was achieved bytargeting the HIV-1 coreceptor CXCR4 and the apoptosis-inducing Fas ligand with two or more siRNAs againstdifferent sites of the same mRNA (Ji et al. 2003).

    In vitro transcription and enzymatic digestion

    Since chemically synthesized siRNAs are expensive,attempts are being made to produce siRNAs by in vitrotranscription using T7 RNA polymerase. Synthetic DNAtemplates containing the T7 RNA polymerase promoterregion followed by desired RNA sequence can be producedusing an automated DNA synthesizer and then amplifiedusing PCR. The T7 polymerase binds to the promotersequence, initiates transcription, and then moves along thetemplate strand toward its 59 end, elongating the RNAtranscript as it goes. Although a termination region on theDNA may be used to halt transcription, the runofftranscription, which stops when the 59 end of the DNAtemplate strand is reached, is also commonly used. Tran-scription of the PCR fragment by the T7 RNA polymeraseproduces both sense and antisense RNAs, which spontane-ously anneal, forming full-length dsRNA (Milligan andUhlenbeck 1989; Donze and Picard 2002).

    The in vitro transcription approach is limited by specificsequence requirements related to T7 polymerase. The lastguanosine of the T7 promoter is invariably the firstribonucleotide that is incorporated into the RNA by theT7 polymerase during transcription. Therefore, all siRNAsproduced by this method start with a 59-G residue andrequire a C-39 residue at position 19 (i.e., 59-G-N17-C-39)to allow annealing with the complementary RNA, whichalso has to start with a 59-G residue. Furthermore, since theefficacy of siRNAs targeted to different regions of a genevaries dramatically, the number of sequences that can betargeted using siRNAs generated by this method is limited

    (Donze and Picard 2002). Kim et al. (2004) demonstratedthat siRNAs synthesized from the T7 polymerase systemcan trigger a potent induction of interferon a and b in anumber of cell lines. The induction of these interferons wasalso seen by short single-stranded RNAs transcribed withT3, T7, and Sp6 RNA polymerases. These investigatorsfurther demonstrated that the presence of triphosphateson the 59 end of T7 transcripts is required for interferoninduction, since the treatment of T7 transcripts withphosphatase could abrogate this effect.

    Sohail et al. (2003) reported an alternative approach forproducing a desired siRNA sequence using T7 polymerasein vitro transcription followed by transcript digestion bydeoxyribozymes, which are known as ‘‘DNAzymes’’ orcatalytic DNA. The cleavage of RNA by DNAzymes occursbetween two specific nucleotides, and the requirement forthis dinucleotide sequence is different for different DNA-zyme groups, making them flexible tools for digesting avariety of sequences (Breaker and Joyce 1994; Feldman andSen 2001). The siRNAs produced by this method causeddose-dependent inhibition of type 1 insulin-like growthfactor receptor in human breast cancer cells comparable tothat induced by chemically synthesized siRNAs of the samesequence (Sohail et al. 2003).

    Both recombinant human Dicer (re-hDicer) and Escher-ichia coli RNAse III are used to cleave long dsRNAproduced by in vitro transcription. Treatment with E. coliRNase III or re-hDicer randomly cleaves the RNA into apopulation of siRNA molecules that are effective mediatorsof gene silencing in a manner similar to that observed whenusing synthetic siRNA. Usually, the cleavage products ofE. coli enzyme RNase III range from 12 to 15 base pairs(bp) in length with termini identical to those produced byDicer (Amarasinghe et al. 2001). Although these shortdsRNAs are not long enough to trigger an RNAi inmammalian cells, the average product length generated by

    TABLE 1. Methods of producing siRNA

    Methods of production Characteristics Advantages Disadvantages

    Chemical synthesis d 21–23-bp siRNA d Ease in sequence design d High costd Long dicer substrate

    RNA (disRNA) ;27 bpd Chemical modifications d Transient gene silencing

    d Quick method to silencea specific gene

    In vitro transcription+enzymaticdigestion

    d Pool of siRNAs d Potent silencing d Higher probability of inducingoff-target effects andnonspecific silencing

    d Low cost d Triphosphate induction of interferonby T7 polymerase transcripts

    Endogenous expression(plasmid or viral vectors)

    d siRNA d Allows both long-term andtransient gene silencing

    d Difficult to transfect in caseof plasmid

    d shRNA d Inducible gene silencing d Vector construction is oftentime-consuming

    d pre-miRNA

    De Paula et al.

    434 RNA, Vol. 13, No. 4

    JOBNAME: RNA 13#4 2007 PAGE: 4 OUTPUT: Tuesday March 6 18:39:21 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • RNase III digestion can be increased by altering digestionconditions. Yang et al. (2002) have shown that 20–25-bpsiRNAs produced by RNase III digestion efficientlyinhibited various endogenous genes in different mamma-lian cell lines without nonspecific effects. In addition,RNase III can digest dsRNA faster than re-hDicer andcan be easily overexpressed and purified (Amarasinghe et al.2001; Bernstein et al. 2001). The siRNAs produced byre-hDicer are 21–23 bp in length and contain 2-nt 39 over-hangs with 59-phosphate and 39-hydroxyl termini, whichare essential for RNAi activity. Dicer-generated siRNAs havebeen shown to be effective in silencing exogenous andendogenous gene expression in mammalian cells (Kawasakiet al. 2003; Myers et al. 2003).

    The enzymatic synthesis of siRNA may provide moreeffective gene silencing than chemically synthesized siRNA,as the enzymatically generated siRNA can correspond tosequences overlapping the entire gene. However, complexsiRNA populations may have a higher probability oftargeting other genes, and therefore promoting nonspecificeffects, than do discrete siRNAs. Additionally, enzymaticsynthesis of siRNA requires separation of siRNA fromuncleaved RNA duplexes and residual nucleic acids (Yanget al. 2002; Myers et al. 2003).

    Endogenous expression

    The application of synthetic siRNAs is restricted by bothlow-to-moderate transfection efficiencyand the short-term persistence of tran-sient gene expression. A single trans-fection of siRNA may not provide asufficient window of functional deple-tion for proteins with long half-lives.Another potential problem inherent inchemically synthesized siRNA is vari-ability in transfection efficiency, espe-cially in difficult-to-transfect cells. Tocircumvent these limitations, expressionvectors currently in use employ siRNAor shRNA expression cassettes thatresemble pre-miRNAs and undergoprocessing by Dicer. Like syntheticsiRNAs, they are designed to pair per-fectly with the target mRNA to induceRNAi. These shRNAs are designed foreither transient or long-term genesilencing and can be produced fromplasmid or viral expression vectors(Amarzguioui et al. 2005).

    Plasmid vectors

    shRNA, siRNA, and miRNA can beproduced from plasmid vectors con-taining promoters that are dependent

    on either RNA polymerase (Pol) II or Pol III. Among them,Pol III promoters are used most frequently because it ispossible to express small RNAs that carry the structuralfeature of siRNA. Figure 2 is a schematic representation ofdifferent strategies used to create expression cassettes usingRNA polymerase promoters for generation of siRNA,shRNA, and miRNA. In the first strategy, the sense andantisense strands are expressed as two independent tran-scripts that hybridize within the cells to form functionalsiRNA duplexes (Fig. 2A). In the second strategy, the senseand antisense strands are expressed as a single transcriptseparated by a short loop of 4–10 nt of sequence. Thetranscript forms a hairpin structure that can be processedby Dicer into functional siRNA (Fig. 2B). In the thirdstrategy, miRNAs that are complementary to the targetgene are expressed using the Pol II promoter (Fig. 2C;Dykxhoorn et al. 2003; Sioud 2004).

    Two different Pol III promoters predominantly beingused for shRNA expression are the U6 promoter and theH1 promoter (Brummelkamp et al. 2002; Paddison et al.2002). In addition to these two promoters, other Pol IIIpromoters such as 5S, 7SK, and tRNA promoters have alsobeen used for expressing siRNAs endogenously (Paul et al.2002; Czauderna et al. 2003b; Kawasaki and Taira 2003).The activities of these promoters vary from cell type to celltype. To optimize shRNA expression, it is beneficialto create expression vectors with at least two differentpromoters and transfect them into the cells being targeted

    FIGURE 2. Schematic representation of expression cassettes using RNA polymerase pro-moters for generation of small-interfering RNAs (siRNA). (A) Tandem-type promoters expresssense and antisense strands individually. After transcription, both strands hybridize forming aduplex siRNA. (B) Short hairpin RNAs (shRNA) are expressed as a single transcript separatedby a short loop of 4–10 nt of sequence. The transcripts form a hairpin structure that can beprocessed by Dicer into functional siRNAs. (C) Expression of imperfect duplex hairpinstructures that is based on pre-microRNA (pre-miRNA) structures. pre-miRNAs are processedby Dicer into a mature miRNA, which can direct gene silencing.

    siRNA delivery and targeting

    www.rnajournal.org 435

    JOBNAME: RNA 13#4 2007 PAGE: 5 OUTPUT: Tuesday March 6 18:39:21 2007

    csh/RNA/131733/rna4598

    Fig. 2 live 4/C

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • for gene knockdown. Construction of shRNA expressionvectors poses two serious technical challenges. First, it isdifficult to sequence constructs that contain a hairpinregion, probably because of the tight palindromic structure.Second, z20%–40% of constructs get mutated within thehairpin region (Brummelkamp et al. 2002; Paddison et al.2002; Paul et al. 2002).

    Viral vectors

    The introduction of siRNA expression plasmids into cellsoften requires electroporation, microinjection, or complexformation with synthetic carriers (lipids, polymers, orpeptides). While most rapidly dividing cell lines are easilytransfected using shRNA expression plasmids, these plas-mid vectors are not easily transfected into primary cells,stem cells, and nondividing cells. In the absence of celldivision, the siRNA expression plasmids cannot be intro-duced into the nucleus, where the DNA is transcribed. Toovercome this limitation, different viral vectors encodingshRNA including retroviral, adenoviral, and adeno-associ-ated viral (AAV) are being developed. Typically, thesevectors use a Pol III promoter, such as U6, H1, or transferRNA promoters (Dykxhoorn et al. 2003).

    Retroviral vectors have been reported to mediate anefficient and stable siRNA expression (Rubinson et al. 2003;Liu et al. 2004a). Unlike Moloney murine leukemia virus(MoMLV), lentiviral vectors efficiently integrate into thegenome of nondividing cells, such as pancreatic islets,hematopoietic stem cells, or terminally differentiated cells.A lentiviral vector encoding shRNA has been shown toeffectively silence GFP, BCL-2, and Interleukin (IL) 12receptor (CD25) genes (Tiscornia et al. 2003; Schomberet al. 2004; Wong et al. 2005). Lentiviral vectors encodingshRNA have also been shown to inhibit HIV-1 infection inhematopoietic stem cells and human CD4+ T-cells (Li et al.2003; Nishitsuji et al. 2006). However, lentiviral vectors areassociated with infrequent insertional mutagenesis. Conse-quently, the use of adenoviral vectors is being exploredsince these vectors do not integrate into the host genomeand efficiently transduce both dividing and nondividingcells (Wu et al. 2003). Adenoviruses contain a lineardouble-stranded DNA genome that remains episomal afterinfection. Recombinant adenoviral vectors containingexpression cassettes of interest are readily generated andcan be purified to very high titers (up to 1013 infectionunits/mL) (Huang and Kochanek 2005).

    A variety of properties make AAV vectors an interestingtool for organ-directed shRNA expression, including thelack of pathogenicity, and ease of vector production. TheAAV-2 type is highly prevalent in the human populationand frequently neutralized by antibodies. Therefore, it isimportant to evaluate different AAV serotypes for organ-directed shRNA expression. AAV-8 vectors expressingshRNA are reported to transduce almost 100% of hepato-cytes after intravenous injection into mice (Grimm and Kay

    2006). However, overexpression of shRNA caused length-and dose-dependent liver injury and ultimate death.Morbidity was associated with the down-regulation ofliver-derived miRNA, indicating possible competitionbetween miRNA and shRNA for nuclear Krayopherinexportin 5 (Grimm et al. 2006). Therefore, it is importantto optimize siRNA dose, length, and sequence to avoid over-saturation of endogenous small RNA pathways.

    Inducible expression vectors

    Plasmid- and viral-vector-based constitutive expressions ofshRNAs by RNA Pol III U6 and H1 promoters often fail toadjust the levels of gene silencing necessary for cell survivaland growth. Besides, gene silencing for longer periods mayresult in nonphysiological responses. This problem can becircumvented by generating inducible regulation of RNAi(Czauderna et al. 2003b; Wiznerowicz and Trono 2003;Gupta et al. 2004). Ohkawa and Taira (2000) described thesuccessful regulation of gene silencing by the integration ofthe bacterial tetracycline operon sequence (tetO) into theU6 promoter. Matsukura et al. (2003) have applied thistetracycline-inducible U6 promoter for in vivo transcrip-tion of shRNA, which enables stable transfection followedby conditional expression of shRNA. The main drawback ofthe tetracycline-inducible system is a relatively high back-ground expression in the uninduced state in certain celllines (No et al. 1996; Van Craenenbroeck et al. 2001). Incontrast, the ecdysone-inducible system is tightly regulated,with no expression in the uninduced state and a rapidinductive response (Gupta et al. 2004).

    Owing to the time-consuming process of cloning siRNAinto plasmid constructs and the need for verification of thecloned sequence, an easier approach of screening sequencesinvolving the production of siRNA expression cassettes(SECs) was developed (Castanotto et al. 2002). SECs arePCR-derived siRNA expression templates introduceddirectly into the cells. SECs consist of a Pol III promoter,a sequence encoding an shRNA, and an RNA polymeraseIII termination site. The final result is a PCR product thatcontains a Pol III promoter, a DNA sequence that, oncetranscribed, forms shRNA, and a terminator sequence.After transcription, the shRNA is intracellularly cleavedinto siRNA by the RNAi machinery. By incorporatingrestriction sites at their ends, SECs effectively elicit genesilencing and can be cloned into a plasmid to create ansiRNA expression vector. The expediency and low cost ofthis procedure lends itself to mass screening of siRNAlibraries as well as identification of siRNA target sites(Chang 2004; Wooddell et al. 2005).

    BARRIERS TO RNAi-BASED THERAPIES

    RNAi intersects a number of pathways and siRNA trans-fection often activates immune response leading to unin-tended and off-target effects. Therefore, the potential

    De Paula et al.

    436 RNA, Vol. 13, No. 4

    JOBNAME: RNA 13#4 2007 PAGE: 6 OUTPUT: Tuesday March 6 18:39:31 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • induction of inflammatory cytokines and interferon (IFN)responses by siRNAs represent a major obstacle for theiruse as inhibitors of gene expression. Therapeutic applica-tions of siRNAs require a better understanding of themechanisms that trigger such unwanted effects.

    Immune response activation

    Figure 3 is a schematic representation of the immuneresponse activation by siRNAs. Non-immune and immunecells get activated in the presence of long dsRNA, leading tothe activation of cytoplasmic receptors such as the dsRNA-dependent protein kinase R (PKR) and the retinoic acid-inducible gene-I (RIG-I) (Saunders and Barber 2003;Yoneyama et al. 2004). PKR is activated by autophosphor-ylation following binding to dsRNA. Once activated, itphosphorylates the eukaryotic translation initiation factor(EIF-2)-a, leading to the global suppression of proteinbiosynthesis and subsequent programmed cell death. PKRcan also activate nuclear factor kB (NF-kB) with conse-

    quent induction of type-I IFN production. A family of 29-59-oligoadenylate synthetases (2959-OAS) is also activatedby dsRNA. This leads to the activation of RNase L, whicheventually triggers the nonspecific degradation of mRNA(Marques and Williams 2005).

    RIG-I is an intracellular dsRNA sensor capable oftriggering IFN production. Diverse cell types derived fromRIG-I knockout mice have impaired responses to viraldsRNA, establishing the essential role of RIG-I in themammalian antiviral response (Yoneyama et al. 2004;Marques and Williams 2005). The helicase RIG-I isnecessary for recognizing blunt-ended siRNAs, leading tothe activation of dsRNA signaling. RIG-I unwinds siRNAscontaining blunt ends more efficiently than siRNAs con-taining 39 overhangs. The efficiency in duplex unwinding isthen translated into signaling downstream to interferonregulatory factor 3 (IRF-3) and NF-kB activation (Marqueset al. 2006).

    Mammalian immune cells express a family of toll-likereceptors (TLRs), which recognize pathogen-associated

    molecular patterns including unmethy-lated cytosine-guanine motifs (com-monly known as CpG motifs) and viraldsRNA. TLR3, the receptor for dsRNA,was a logical candidate for recognizingsiRNA in the context of immunostimu-lation. Indeed, TLR3 overexpressedin cultured human embryonic kidney(HEK) 293 cells was capable ofrecognizing siRNAs (Marques andWilliams 2005). However, the activationof immune cells by siRNAs is sequencedependent, and either sense or anti-sense strands individually can induceinflammatory cytokine production asefficiently as duplex siRNAs (Judge et al.2005; Sioud 2005, 2006). Thus, as TLR3neither recognizes single-strandedRNA (ssRNA) nor requires sequencespecificity, it is unlikely involved inactivation of the immune system bysiRNAs (Marques and Williams 2005;Sioud 2005).

    TLR7 and TLR8 were initially shownto mediate the recognition of RNAviruses and small synthetic antiviralcompounds referred to as imidazoqui-nolines. TLR7, TLR8, and TLR9 areexpressed in endosomes and requireendosomal maturation for efficient sig-naling (Marques and Williams 2005).siRNA recognition by TLR7, TLR8, andTLR9 results in activation of NF-kBand IRFs, which induce inflammatorycytokines and IFNs, respectively (Fig. 3;

    FIGURE 3. Activation of immune response by siRNAs. siRNA/shRNA can be delivered bytwo different ways: (1) Delivery of viral vector followed by endogenous expression of shRNA inthe nucleus, which is transported to the cytoplasm and processed by Dicer into siRNAs.The siRNA causes sequence-specific mRNA degradation and does not induce an interferon(IFN) response. (2) Synthetic delivery systems forming an siRNA complex particle, forexample, liposome. In this way, the siRNA is taken up by the cell via endocytosis, causingsequence-specific mRNA cleavage through RNAi and activation of the immune response. Theimmune response can be activated by different pathways: (a) recognition of siRNAs by Toll-like receptors (TLRs); (b) activation of retinoic acid-inducible gene I (RIG-I) by blunt-endsiRNAs; (c) activation of dsRNA-dependent protein kinase R (PKR); (d) activation of 29-59-oligoadenylate synthetases (2959-OAS). Generally, activation of these pathways leads to theinduction of several cellular factors, including nuclear factor kB (NF-kB), interferon regulatoryfactors (IRFs), eukaryotic translation initiation factor 2a (EIF-2a), and RNase L. Induction ofinterferons (IFNs), inflammatory cytokines, nonspecific mRNA degradation, and generalinhibition of protein synthesis are some undesired effects of the immunostimulation caused bysiRNAs.

    siRNA delivery and targeting

    www.rnajournal.org 437

    JOBNAME: RNA 13#4 2007 PAGE: 7 OUTPUT: Tuesday March 6 18:39:32 2007

    csh/RNA/131733/rna4598

    Fig. 3 live 4/C

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • Marques and Williams 2005). Interestingly, not all ssRNAmolecules are capable of activating TLR7 and TLR8, but,rather, U- and G-rich ssRNAs seem to be preferentiallyrecognized (Sioud 2006). It has now become clear thatTLR7 and TLR8 mediate the recognition of siRNAs in asequence-dependent manner (Heil et al. 2004; Hornunget al. 2005; Judge et al. 2005).

    Recognition of siRNAs by TLRs takes place in theendosome, before the siRNAs enter the cytoplasm. There-fore, it is expected that if siRNAs could enter the cytoplasmavoiding the endosome, they should bypass the activationof immune systems but still mediate gene silencing. Robbinset al. (2006) tested the immunostimulatory effects of lipid-delivered siRNAs versus Pol III promoter-expressed shRNAsin primary CD34+ progenitor-derived hematopoietic cells.They observed that in this system, cationic lipid/siRNA com-plexes are potent inducers of IFN-a and type-I IFN geneexpression, whereas the same sequences when expressedendogenously are nonimmunostimulatory.

    The method of siRNA delivery also has a great influenceon the immunostimulatory activity of siRNAs (Fig. 3).Injection of siRNAs after complex formation with cationicliposomes into mice induces the release of inflammatorycytokines, including IL-6, tumor necrosis factor-a (TNF-a),and IFN-a (Judge et al. 2005; Sioud 2005). In contrast,injection of naked, unmodified siRNAs or siRNAs conju-gated to cholesterol has no significant effect on immuno-stimulation (Heidel et al. 2004; Marques and Williams2005). There are two possible explanations for the absenceof immunostimulation by naked siRNAs. First, unmodifiedsiRNAs have a short half-life in serum and may be degradedbefore being recognized by specific receptors. Second,cationic lipid/siRNA complexes are more readily recog-nized by immune cells than are naked siRNAs. In fact,Sioud (2005) reported that naked siRNAs containing aphosphorothioate (PS) backbone were not immunostimu-latory. However, these results should be interpreted care-fully considering that naked CpG ODNs are known to beimmunostimulatory independent of backbone modifica-tions (Marques and Williams 2005).

    Off-target effects and nonspecific gene silencing

    For its therapeutic applications, siRNA must not cause anyunintended effect other than sequence-specific gene silenc-ing. However, recent studies indicate that there are off-target effects associated with the use of siRNA (Jacksonet al. 2003). Off-target effects consist of any gene silencingeffect caused by siRNAs in nontarget mRNAs through theRNAi mechanism. Generally, it occurs due to lack ofcomplementarity between siRNAs and target mRNAs. Anexplanation for the fact that siRNAs can induce silencing ofnontarget genes can be found in the RNAi machinery.Although the actual substrate specificity of individualsiRNAs appears to be very high, RNAi machinery can

    tolerate single mutations located in the center of the siRNAmolecule without losing the ability to induce gene silenc-ing. It makes some siRNAs able to silencing other thanthose genes related to their homologous mRNA. However,complete inactivation of the RNAi mechanism can occur iffour or more mutations are introduced in the guide strand,making the active RISC unable to recognize the targetmRNA (Jackson et al. 2003; Persengiev et al. 2004; Sioud2004).

    siRNAs and miRNAs were found to be functionallyinterchangeable. siRNAs can act as miRNAs dependingon the degree of complementarity with the target mRNA. Ifsynthetic siRNAs bear a sufficiently low degree of comple-mentary bases, target mRNA translation occurs instead ofmRNA degradation, whereas miRNAs will lead to mRNAdegradation if perfect complementarity with target mRNAexists. This process is one of the reasons for the off-targeteffects of siRNAs (Jackson et al. 2003; Bartel 2004). Anotherreason for the occurrence of off-target effects is that notonly the antisense but also the sense strand of an siRNA candirect gene silencing of nontarget genes, and it has beendocumented to occur when as few as 15 bp of comple-mentarity exists between the siRNA and mRNA (Jacksonet al. 2003). Birmingham et al. (2006) also demonstrated thatthe presence of one or more perfect matches between thehexamer or heptamer seed (positions 2–7 or 2–8 of theantisense strand) of an siRNA and the 39-untranslatedregion (UTR), but not the 59-UTR or open reading frame,is associated with off-targeting. A high proportion of ‘‘off-target’’ transcripts silenced by siRNAs have been shown tohave 39-UTR sequence complementarity to the seed regionof the siRNA, since the base mismatches within the siRNAseed region reduced the set of original off-target transcripts.Since there is no algorithm that can eliminate significantnumbers of 7–8-base matches of siRNAs to the tran-scriptome, it will be difficult to achieve perfect specificity(Jackson et al. 2006). These findings suggest a strongmechanistic parallel between siRNA off-targeting andmiRNA-mediated gene regulation.

    Nonspecific gene silencing is those effects caused bysiRNAs through any pathway other than RNAi. Semizarovet al. (2003) investigated the overall cellular effects of siRNAson transcription levels. In a concentration-dependentmanner, siRNAs nonspecifically interfered in the expressionof a significant number of genes, many of which are knownto be involved in apoptosis and stress response. Persengievet al. (2004) reported that treatment with siRNA may resultin diverse cellular activities, such as cell signaling, cytoskel-etal organization, gene expression, metabolism, and celladhesion. Jackson et al. (2003) attributed the nonspecificeffects to cross-hybridization of transcripts containingregions of partial homology with the siRNA sequence.However, the nonspecific effects observed by Persengievet al. (2004) cannot be explained by off-target regulationbecause the siRNAs tested lacked significant sequence

    De Paula et al.

    438 RNA, Vol. 13, No. 4

    JOBNAME: RNA 13#4 2007 PAGE: 8 OUTPUT: Tuesday March 6 18:39:43 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • similarity to any human gene. Moreover, for all of the genestested, expression was affected by two siRNAs withsequences that are completely unrelated. Bridge et al.(2003) also reported that shRNAs can affect the expressionof many genes, including several IFN targets. The activationof sequence-independent inhibition of gene expression bysiRNA or shRNA-expression vectors seems to be, at least inpart, due to the induction of type-I IFN, PKR, andsignaling through TLR3 (Kariko et al. 2004; Marques andWilliams 2005).

    BIODISTRIBUTION OF siRNA

    To develop effective delivery systems of chemically synthe-sized siRNA, it is important to gain in-depth understandingof its biodistribution and pharmacokinetic properties atwhole-body, organ, and cellular levels.

    siRNA duplexes are much more stable than ssRNAs.However, since they are similar to single-stranded antisenseoligonucleotides, they still tend to degrade on incubationwith serum, contributing to their short half-lives in vivo.125I-labeled siRNAs are widely distributed in the body afterintravenous injection into mice, with preferential accumu-lation in the liver and kidney (Braasch et al. 2004).Radiolabeled siRNA is also distributed to the heart, spleen,and lung. However, very little siRNA is detected in thebrain (Braasch et al. 2004; Soutschek et al. 2004; Santelet al. 2006). Intravenous administration of naked siRNA inrats showed a short half-life of 6 min and a clearance of17.6 mL/min. The poor pharmacokinetic properties ofsiRNAs are related to their low in vivo stability, since theycan be degraded by endogenous RNases. Another reason istheir fast elimination by kidney filtration because of theirsmall molecular mass (z7 kDa) (Soutschek et al. 2004).Consequently, improving the pharmacokinetic propertiesof siRNA duplexes is an important goal for developingsiRNA-mediated gene silencing.

    STRATEGIES FOR IMPROVING RNAi-BASEDTHERAPIES

    To develop strategies for improving RNAi, we need toaddress several problems, including the enzymatic stability,cellular uptake, pharmacokinetic properties, and potentialfor off-target effects and immunoactivation of siRNAs. Thesequence design of siRNA molecules is extremely importantto improve their efficacy as well as to reduce the potentialfor off-target effects and activation of the immune response(Reynolds et al. 2004). Backbone modifications and bio-conjugation with lipids and peptides are known to improvethe stability and cellular uptake of siRNAs (Chiu and Rana2003; Lorenz et al. 2004). Several reagents have also beenemployed for systemic administration of siRNAs as analternative to the covalent modifications of the siRNAmolecule (Simeoni et al. 2003; Morrissey et al. 2005b;

    Urban-Klein et al. 2005; Santel et al. 2006). Cationicliposomes are routinely used for transfection of nucleicacids into mammalian cells. Positively charged carriers,such as protamine-antibody fusion proteins, polyethylenei-mine (PEI), and cyclodextrin-containing polycation, havebeen tested for complex formation with siRNAs. To achievecell-specific delivery of siRNAs, several targeting ligands,including cell membrane receptors and antibodies, havebeen explored (Song et al. 2005).

    Design of siRNAs

    A thorough understanding of the sequence, size, andstructural requirements of siRNAs is essential to effectivelymediate RNAi. Table 2 summarizes the most importantsiRNA design rules to improve gene silencing and avoidundesired effects.

    siRNA sequence

    Some research groups have elaborated several guidelines fordesigning siRNAs that can potentially silence gene expres-sion (Elbashir et al. 2002; Paddison et al. 2002; Reynoldset al. 2004). Several sequence motifs are consistent witheffective siRNAs, including AAN19TT, NAN19NN, NAR-N17YNN, and NANN17YNN (where N is any nucleotide, Ris a purine, and Y is a pyrimidine). In principle, any regionof the mRNA can be targeted. However, it should bepointed out that complementarity is often found in the 39-UTR of the off-target sequences. Furthermore, sequencecomplementarity between the 59 end of the guide strandand the mRNA is known to be the key to off-targetsilencing. Additionally, intronic sequences must be avoidedsince RNAi is a cytoplasmic process (Dykxhoorn et al. 2003).

    Ui-Tei et al. (2004) proposed that highly effective RNAican be achieved if siRNA satisfying the following foursequence conditions at the same time is used: (1) AUrichness in the 59-terminal, 7-bp-long region of theantisense strand; (2) G/C at the 59 end of the sense strand;and (3) the absence of any long GC stretch of >9 bp inlength. Even though Elbashir et al. (2002) reported that thetarget region should be at least 50–100 nt downstream fromthe start codon, Dykxhoorn et al. (2003) suggest that thereis a predisposition for effective siRNA-directed silencingtoward the 39 portion of the gene. Moreover, sequenceswith even representation of all nucleotides on the antisensestrand are favored, and those regions with stretches ofa single nucleotide, especially G, should be avoided asG-rich oligonucleotides have a tendency to form quartets.Reynolds et al. (2004) analyzed 180 siRNAs targeting everyother position of two 197-base regions of firefly luciferaseand human cyclophilin B mRNA in a total of 90 siRNAsper gene. A two-base shift in the target position wassufficient to significantly alter siRNA activity, suggestingthat its functionality is determined by the siRNA-specificproperties. Most potent siRNA has a G/C content ranging

    siRNA delivery and targeting

    www.rnajournal.org 439

    JOBNAME: RNA 13#4 2007 PAGE: 9 OUTPUT: Tuesday March 6 18:39:44 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • from 36% to 52%. siRNA duplexes with a GC content>52% may have difficulty in dissociating into passengerand guide strands, while siRNAs with a GC content 19 nt of sequence

    25–30-nt disRNAs are more potent than conventional 21–23-ntsiRNAs

    Long dsRNA (;30 nt) can activate dsRNA-dependent PKRresponse

    Structure The A-form helix of siRNA-mRNA is required for themechanism of RNAi

    29-OH groups of uridines are associated with immunerecognition of siRNAs

    Modifications that do not completely hinder the unwinding ofthe siRNA duplex are allowed since it is not a requirement forhybridization of siRNA with mRNA and induction of RNAi

    Single-strand siRNAs are more immunostimulatory than theirrespective double-strand siRNAs

    29-OH groups are related to enzymatic stability but are notrequired to mediate RNAi

    Blunt structure at the 39 end is the strongest terminal structurefor promoting activation of dsRNA-dependent PKR

    Asymmetrical 25–30-nt siRNAs with 59 blunt ends and 2-nt39 ends are more potent than symmetrical 2-nt overhangscontaining siRNAs

    (disRNA) Dicer substrate siRNA.

    De Paula et al.

    440 RNA, Vol. 13, No. 4

    JOBNAME: RNA 13#4 2007 PAGE: 10 OUTPUT: Tuesday March 6 18:39:44 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • effectively activate innate immunity in the context ofdouble-strand siRNA to a level comparable to that obtainedwith free single-strand RNA. siRNA duplexes seem to beless effective than siRNA in activating innate immunity.The intracellular receptors for single-strand siRNA, inparticular TLR8 and TLR7, do not effectively recognizemost immunostimulatory RNA motifs in the context ofsiRNA duplexes (Sioud 2006).

    A number of academic and commercially affiliated Web-based softwares have been developed to assist researchers inthe identification of efficient siRNA sequences (Naito et al.2004; Yuan et al. 2004). Since the rules that govern efficientsiRNA-directed silencing are still unknown, researchersseeking to silence gene expression should synthesize severalsiRNAs to a gene and validate empirically the efficiency ofeach one. To ensure that the chosen siRNA sequence targetsa single gene, a search of the selected sequence should becarried out against sequence databases. This can be doneusing the Smith–Waterman algorithm or the Basic LocalAlignment Search Tool (BLAST) located at the NationalCenter of Biotechnology Information Web site (http://www.ncbi.nlm.gov). Sequences in these databases thatshare partial homology with siRNAs might be targetedfor silencing by the siRNAs. Potential off-target effects ofthe siRNAs might be minimized by choosing an siRNAwith maximal sequence divergence from the list of geneswith partial sequence identity to the intended mRNA target.

    siRNA size

    Synthetic RNA duplexes of 25–30 nt in length (morespecifically, 27 nt), which are Dicer substrates, have beenshown to be up to 100-fold more potent than correspond-ing conventional 21-nt siRNAs (Kim et al. 2005a). The useof 27-nt dsRNAs, also called Dicer substrate dsRNA(disRNA), allows targeting of some sites within a givensequence that are refractory to suppression with traditional21-nt siRNAs. The use of disRNAs to trigger RNAi canresult in enhanced and prolonged gene silencing at lowerconcentrations than those required for conventional 21-ntsiRNAs applications (Kim et al. 2005a). It is important tonote that in vitro Dicer cleavage of 27-nt disRNAs beforetransfection does not significantly enhance gene silencingefficiency. Additionally, it has been shown that chemicallysynthesized shRNAs that are Dicer substrates are morepotent inducers of RNAi than conventional siRNAs. More-over, a two-base 39 overhang directs Dicer cleavage (Siolaset al. 2005). The enhanced potency of the longer duplexes isattributed to the fact that they are substrates of the Dicerendonuclease, directly linking the production of siRNAs forincorporation into the RISC (Kim et al. 2005a).

    The size of siRNA also plays an important role in theactivation of immune response in a sequence-independentmanner. Although it was initially postulated that aminimum of 30 nt was necessary to activate dsRNAsignaling through PKR activation, shorter siRNAs, as few

    as 21–23 nt, are capable of binding and activating PKR invitro (Marques et al. 2006). In addition, Hornung et al.(2005) observed that 12-nt ssRNAs containing the immu-nostimulatory motif (GUCCUUCAA) were poor inducersof IFN-a in PDCs. However, the investigators observedthat the motif identified needs to be part of a longer ODNsequence, at least 19 bases, to induce IFN-a activation. Theapproach reported by Kim et al. (2005a), which consists ofDicer substrate 27-nt dsRNA with increased RNAi potencyrelative to conventional 21-nt siRNAs, may facilitate theuse of lower concentrations of duplex RNA. Therefore,since the undesired effects can occur in an siRNA dose-dependent manner, the use of Dicer substrate 27-nt dsRNAmay be an alternative to overcome this problem withoutlosing efficacy.

    siRNA structure

    According to Chiu and Rana (2002), the A-form helix of theguide strand-mRNA duplex is required for the mechanism ofRNAi. Chemical modifications disrupting the functionalgroups of the major groove of the A-form helix formed bythe guide strand and its mRNA specifically at the cleavagesite inhibit RNAi. The major groove is also required forpromoting RNA–RISC interactions that subsequently lead tomRNA cleavage (Chiu and Rana 2002, 2003).

    The 29-OH of the ribonucleotide of RNAs is required forthe nucleophilic attack during the hydrolysis of the RNAbackbone, the reaction catalyzed by degradative RNases(Chiu and Rana 2003). However, 29-OH is not required forRNAi-mediated degradation, and, even more specifically, itis not required for nucleotide base-pairing with nucleotideslining the mRNA cleavage site (Chiu and Rana 2003).Additionally, Sioud (2006) has shown that the 29-OH ofuridines is required for immune recognition and signalingof siRNAs, being responsible, at least in part, for activationof the immune response.

    New design approaches that improve the performance oflong dsRNAs as Dicer substrates have been reported (Kimet al. 2005a). These new approaches include 25–30-ntasymmetric dsRNAs with a 59 blunt end and a 2-nt 39overhang on the other end. The improved efficacy ofdisRNAs is postulated to result from their recognitionand cleavage by Dicer, followed by their subsequentincorporation into the RISC complex. This interpretationis supported by observations that Drosophila Dicer is notonly instrumental in handing over siRNA to nascent RISC,but it is itself a component of the latter (Lee et al. 2004).Providing the RNAi machinery with a Dicer substratepresumably results in more efficient incorporation of theactive 21-nt siRNA strand into RISC. These modificationsalso direct the way Dicer processes the dsRNA substrate,resulting in a more homogeneous pool of siRNA products.

    The extremities of the siRNA duplex are critical deter-minants of its capacity to trigger immune response activa-tion. A blunt structure at the 39end is the strongest terminal

    siRNA delivery and targeting

    www.rnajournal.org 441

    JOBNAME: RNA 13#4 2007 PAGE: 11 OUTPUT: Tuesday March 6 18:39:45 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • structure for promoting activation of dsRNA signalingthrough the PKR pathway, followed by a 59overhang(Marques et al. 2006). In contrast, 39overhangs, normallypresent in endogenous Dicer products such as miRNAs,allow RNAi to proceed without activation of dsRNA-dependent PKR (Marques et al. 2006). The presence ofthe 39 overhangs also precludes activation of dsRNAsignaling by siRNAs and reveals an important basis fordiscrimination between self and nonself dsRNAs. Interest-ingly, this type of discrimination appears to take place afterRIG-I binds to dsRNAs (Marques et al. 2006).

    Chemical modifications

    Chemical modifications to sugars, backbones, or bases ofoligonucleotides can be used to control their pharmacoki-netic profiles and reduce nonspecific effects without affect-ing their biological activity. The valuable know-how ofbackbone modifications of antisense ODNs can be readilyadapted to develop new siRNA technologies (Marshall andKaiser 2004). Although siRNA duplexes used for silencingare inherently more stable than ssRNAs, there are reasonsto chemically modify one or both strands. Apart fromimproving stability and reducing off-target effects, chemicalmodifications can aid in broadly targeting siRNAs into cellsand tissues, and certain conjugates can enhance uptake inspecific cell types. Multiple chemical modifications can beintroduced at various positions with the siRNA duplexes.Figure 4 shows the most common chemical modificationsintroduced in siRNAs.

    Phosphodiester modifications

    One of the simplest modifications is the introduction ofphosphorothioate (PS) linkages, which can be prepared byreplacing one of the two nonbridging oxygen atoms in theinternucleotide linkage of RNA by a sulfur atom. PSlinkages are known to reduce cleavage by nucleases andincrease the half-life of ODNs in vivo. However, oligonu-cleotides with extensive PS linkages are also known toincrease binding to serum proteins and can be toxic in vivo(Manoharan 2004; Mahato et al. 2005).

    Braasch et al. (2003) reported that PS siRNAs containing12–21 PS substitutions per strand were stable duringextended incubation in fetal calf serum media, but theirstability was not higher than unmodified siRNA duplexes.The investigators also monitored the inhibition of humancaveolin-1 (hCav) expression by PS siRNAs. The modifiedsiRNAs were able to silence the hCav gene, but lessinhibition was observed by fully modified siRNAs. Thenuclear uptake of PS siRNAs was greater than that ofunmodified siRNAs, which can explain their reducedactivity. Although backbone modifications did not reducethe silencing activity of siRNAs targeting the HTF gene inhuman keratinocytes, most extensively PS siRNAs provedto be cytotoxic, resulting in z70% cell death compared

    with mock-transfected cells (Amarzguioui et al. 2003). Theintroduction of PS linkages to siRNA yields mixed resultsfor siRNA distribution to various organs. In another study,Braasch et al. (2004) observed an increase in the distribu-tion of siRNAs to spleen, heart, and lung, while thedistribution to kidney and liver decreased. However, theeffects were modest, suggesting that the introduction of PSlinkages may not play a major role in determining thedistribution of siRNA.

    An alternative backbone modification that confersincreased biological stability to nucleic acids is the bor-anophosphonate linkage. In boranophosphonate ODNs,the nonbridging phosphodiester oxygen is replaced withan isoelectronic borane (–BH3) moiety. The charge distri-bution of boranophosphonate ODNs also differs from thatof normal phosphate and phosphorothioate ODNs. Thus, itchanges the polarity and increases the hydrophobicity ofthe molecule. While boranophosphonate modification hasbeen less widely studied, it has shown more advantagesthan PSs. Targeting GFP in HeLa cells, Hall et al. (2004)observed that the activity of boranophosphonate siRNAsexceeds that of PS siRNAs, and it was often greater thanthat of unmodified siRNAs, irrespective of the base orstrand modified. However, boranophosphonate modifica-tions placed at the center of the antisense strand reducedRNAi activity. Boranophosphonate siRNAs modified atminimal levels also showed improved stability over unmod-ified siRNAs against nuclease degradation. Additionally,

    FIGURE 4. Most common chemical modifications introduced insiRNAs. (A) Phosphodiester modifications: unmodified siRNA (phos-phodiester RNA); phosphorothioate RNA; boranophosphonate. (B)29-Sugar modifications: 29-O-Methyl RNA; 29-deoxy-29-fluoro RNA;locked nucleic acid (LNA).

    De Paula et al.

    442 RNA, Vol. 13, No. 4

    JOBNAME: RNA 13#4 2007 PAGE: 12 OUTPUT: Tuesday March 6 18:39:45 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • increasing the number of modifications yielded more stablesiRNAs.

    29-Sugar modifications

    Modifications of RNA at the 29-position of the ribose ringhave been shown to increase siRNA stability againstendonucleases and reduce immune response activation.These modifications include 29-O-methyl (29-OMe), 29-deoxy-29-fluoro modifications, and locked nucleic acid.

    Fluoro and methyl linkages. The siRNA motif consistingof 29-OMe and 29-fluoro nucleotides has enhanced plasmastability and increased in vivo potency. The 29-OMe sugarmodification retains the canonical right-handed A-formhelical geometry, which is required for siRNA activity. Thismodification has also been shown to increase the nucleaseresistance of ODNs and siRNA duplexes (Chiu and Rana2003). The effect of 29-OMe modification has been foundto be dependent on both position and extent of incorpo-ration. siRNAs with fully 29-OMe-substituted sense strandswere functional when the duplexes were of 20-bp bluntconstruction, but not for canonical duplexes of 19-bpconstructs with 39 overhangs (Kraynack and Baker 2006).In contrast, siRNAs with alternating 29-OMe and unmod-ified nucleotides (Czauderna et al. 2003a), or alternating29-OMe or 29-O-fluoro nucleotides (Allerson et al. 2005),had activity similar to that of the unmodified duplexes.This suggests that minimal chemical modifications arecompatible with siRNA function. Jackson et al. (2006)reported that 29-OMe modifications to specific positionswithin the siRNA seed region reduce both the number ofoff-target transcripts and the magnitude of their regulation,without significantly affecting silencing of the intendedtargets. Current evidence suggests that 29-OMe sugarmodifications decrease the free energy of hybridization,which would tend to compensate for, rather than impair,weaker base-pairing (Inoue et al. 1987; Lesnik et al. 1993).Additionally, the effects on off-target silencing of the 29-OMe modification show a sharp position dependence, incontrast to the broader position dependence of basesubstitutions (Jackson et al. 2006). It is important to notethat 29-OMe modifications impairing the cleavage of thepassenger strand also impair target RNA cleavage. How-ever, 29-O-methyl ribose groups positioned 1 or 2 ntdownstream from the cleavage site did not impair targetRNA cleavage (Matranga et al. 2005; Leuschner et al. 2006).

    Chiu and Rana (2003) targeted the EGFP gene in HeLacells using different siRNAs modified at the 29-OH site. Theeffects of 29-OH modifications on RNAi were studied byreplacing uridine and cytidine in the antisense strand ofsiRNA by 29-fluoro-uridine (29-FU) and 29-fluoro-cytidine(29-FC), respectively. These modifications increased thesiRNA stability upon exposure to HeLa cell extracts,without losing gene silencing activity. On the other hand,modifying the 29-OH to a bulky methyl group to create

    29-OMe nucleotides in the sense or antisense strand greatlydiminished EGFP gene silencing, whereas double-stranded29-OMe-modified siRNAs completely abolished RNAi. Onereasonable explanation is that the methyl group, as a bulkygroup, may severely limit the interactions among siRNA,target mRNA, and the RNAi machinery that are required tosuccessfully mediate gene silencing.

    Layzer et al. (2004) observed that modification of siRNAwith 29-fluoro pyrimidines does not have an adverse effecton gene silencing and target specificity, as seen in its abilityto distinguish between the GL2 and GL3 luciferase targetmRNAs. In addition, 29-F-modified siRNA showed adramatic increase in the stability over 29-OH siRNAs inhuman plasma. More than 50% of the 29-OH siRNA wasdegraded within a minute of exposure to plasma, andpractically all molecules were degraded after 4 h. Incontrast, >50% of the 29-F siRNA molecules remained fulllength after 24 h of exposure to plasma. However, aftertransfecting 29-F GL2 siRNA into HeLa R19-Luc cells,which stably express the GL2 luciferase gene, little differ-ence was observed in both the duration and magnitude ofsuppression between 29-F and 29-OH siRNAs. These resultsare consistent with those of Song et al. (2003a), whodemonstrated that 29-OH siRNA activity is limited bydilution in cell culture, but not by intracellular stability.Additionally, both 29-OH and 29-F siRNAs reduced lucif-erase expression in vivo by >85% when injected in rodents,but 29-F modification of the siRNA did not grant anyadvantage to either the magnitude or the interval ofresponse. Although 29-F-modified siRNAs have muchgreater stability in plasma than 29-OH siRNAs, these datasuggest that once siRNAs are localized within the cell,modifications that enhance nuclease stability do not influ-ence activity or persistence.

    Locked nucleic acid. Locked nucleic acid (LNA), alsoreferred to as inaccessible RNA, is a family of conforma-tionally locked nucleotide analogs that displays unprece-dented hybridization affinity toward complementary DNAand RNA. Commonly used LNA contains a methylenebridge connecting the 29 oxygen with the 49 carbon of theribose ring. This bridge locks the ribose ring in the 39-endoconformation characteristic of RNA (Bondensgaard et al.2000; Braasch and Corey 2001). LNA has been shown to becompatible with siRNA intracellular machinery, preservingmolecule integrity while offering several improvements thatare relevant to the development of siRNA technology(Braasch et al. 2003; Elmen et al. 2005).

    LNA may be used to increase the functional half-life ofsiRNA in vivo by two different mechanisms: (1) enhancingthe resistance of the constituent RNA strands againstdegradation by ssRNases, and (2) stabilizing the siRNAduplex structure that is crucial for silencing activity. Elmenet al. (2005) reported that the stability of siRNA can beenhanced by conjugating LNA at the 39 ends of the sense

    siRNA delivery and targeting

    www.rnajournal.org 443

    JOBNAME: RNA 13#4 2007 PAGE: 13 OUTPUT: Tuesday March 6 18:39:51 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • strand of siRNA. Introduction of LNA modifications at the39 overhangs in either one or both strands of siRNA againstfirefly luciferase revealed no loss of silencing effect of siRNAin cultured cells. Additionally, one LNA at the 59 end of thesense strand was shown to be fully compatible withsilencing activity, while an LNA at the 59 end of theantisense strand dramatically impaired the silencing effect.Since the strand that displays the weakest binding energy atits closing 59 base pair is incorporated preferentially intothe RISC (Khvorova et al. 2003; Schwarz et al. 2003), theconjugation of LNA at the 59 sense strand might be used toalter strand bias in favor of incorporation of the antisensestrand. It may help in reducing off-target effects andincreasing potency. Additionally, the potency of siRNAsmay be improved by simply lowering the number of RISCsloaded with the sense strand. LNA modifications of the 39or 59 termini of the sense strand of siRNAs can abrogatetheir immunostimulatory activity. However, depending onthe extent and location of LNA modifications, they canblock not only the induction of IFNs but also the genesilencing activity of the siRNA (Hornung et al. 2005).

    Bioconjugation

    Bioconjugation of one or both strands of siRNAs withlipids and polymers is often desirable to (1) further increasetheir thermodynamic and nuclease stability, (2) improve thebiodistribution and pharmacokinetic profiles of siRNAs,and (3) target them to specific cell types.

    Lipid conjugation

    Conjugation with lipids may enhance siRNA uptake viareceptor-mediated endocytosis or by an increased mem-brane permeability of the otherwise negatively chargedRNA. Conjugation of nucleic acids with cholesterol hasbeen demonstrated to enhance cellular uptake in cellculture and hepatic deposition after systemic administra-tion (Cheng et al. 2006). This is because cholesterolconjugation increases the hydrophobicity and cellularassociation of nucleic acids. Lorenz et al. (2004) haveconjugated siRNAs with cholesterol derivatives like lith-ocholic and lauric acids at the 59 end of the sense strand.Cholesterol conjugation was shown to increase the cellularuptake of siRNAs in human liver cells without use ofany transfection reagent. Incubation of cells during 4 h with50 nM of siRNAs with a modified sense strand down-regulated b-galactosidase expression to a higher extent thansiRNAs with a modified antisense strand or two modifiedstrands.

    Apolipoprotein B (ApoB) is expressed predominantly inthe liver and jejunum, and is an essential protein for theassembly and secretion of very-low-density lipoprotein(VLDL) and low-density lipoprotein (LDL), which arerequired for the transport and metabolism of cholesterol.Elevated ApoB and LDL levels are correlated with increased

    risk of coronary artery disease, and inadequate control ofLDL-cholesterol after acute coronary syndromes resultsin increased risk of recurrent cardiac events or death.Soutschek et al. (2004) reported the conjugation of cho-lesterol to the 39 end of the sense strand of an siRNAmolecule targeting ApoB. The concentration of this proteinin human blood samples correlates with those of choles-terol, and higher levels of both compounds are associatedwith an increased risk of coronary heart disease. Intrave-nous administration of radiolabeled siRNA-chol conjugatesinto rats at 50 mg/kg has shown improved pharmacokineticproperties compared to unconjugated siRNAs. siRNA-cholconjugates showed an elimination half-life (t1/2) of 95 minand a plasma clearance (CL) of 0.5 mL/min, whereasunconjugated siRNAs have a t1/2 of 6 min and a CL of17.6 mL/min. Significant levels of this conjugate werefound in the liver, heart, kidney, lung, and adipose tissue.Most importantly, siRNAs efficiently reduced the levels ofApoB mRNA by >50% in the liver and by 70% in thejejunum, which resulted in a lowering of the bloodcholesterol levels comparable to that observed in mice inwhich the ApoB gene had been deleted.

    Peptide conjugation

    Protein transduction domains (PTDs) offer an alternativeto the traditional methods of siRNA delivery. PTDs areshort amino acid sequences that are able to interact withthe plasma membrane in a way that leads to a highly effi-cient uptake into the cytoplasm. Only short stretches,mainly consisting of positively charged amino acids, suchas arginine and lysine, are responsible for translocating thePTDs through the plasma membrane—hence the name cell-penetrating peptides (CPPs) or membrane permeant peptides(MPPs). Stretches of these amino acids help to maintainthe helical structure of the PTD, which is the structuralprerequisite for membrane penetration. Many natural andsynthetic MPPs are known to contain this structural pro-perty of PTDs (Mahat et al. 1999; Schepers 2005).

    Cellular uptake of MPPs takes place in a receptor-independent fashion. MPPs are mostly amphipathic mol-ecules that interact with the negatively charged head groupsof the plasma membrane via their positive amino acidresidues. MPPs can transfect different cell types with veryhigh efficiency and rapid uptake kinetics. Their greatversatility with respect to cargo and cell type, as well astheir high efficiency in delivering cargo molecules into cells,makes the MPPs a valuable tool for transfecting siRNAsinto mammalian cells and even fully grown organisms.Some cargo molecules may only be coupled at one definedsite, where the chemical modification does not alterfunctionality. This can be achieved by making use of singlefunctional groups that naturally occur in the cargo mole-cule or may be introduced by the utilization of chemicallymodified building block in the synthesis of the MPPs.This system can be exploited for efficient delivery of

    De Paula et al.

    444 RNA, Vol. 13, No. 4

    JOBNAME: RNA 13#4 2007 PAGE: 14 OUTPUT: Tuesday March 6 18:39:51 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • siRNAs into cells and into animals. For coupling of thesiRNA, both the MPP and the siRNA must be modified byfunctional groups (Hallbrink et al. 2001; Schepers 2005).

    Muratovska and Eccles (2004) have shown that siRNAscan be delivered directly into the cytoplasm when conju-gated with MPPs, such as penetratin or transportan (Fig.5A). Constitutively expressed luciferase and GFP geneswere successfully silenced in a high proportion of cells ofdifferent types with penetratin– or transportan–siRNAconjugates. For example, in CHO-AA8-Luc Tet Off cells,the percentage of luciferase activity silenced on each of the3 d following incubation of the cells with MPP–siRNA con-jugates was markedly superior to the silencing following

    transfection with Lipofectamine 2000 (Fig. 5B). The salientfeatures of this system are as follows: (1) the peptidefacilitates transport across the plasma membrane, and theMPP–siRNA conjugates are freely translocated into thecytoplasm; (2) the disulfide bond is reduced in the cyto-plasm, releasing the bioactive siRNA to cause sequence-specific mRNA degradation; and (3) the uptake of MPP–siRNA conjugates is rapid and occurs directly through themembrane with no need for classical receptor-mediateduptake or endo- or pinocytosis.

    Simeoni et al. (2003) described a new peptide-based genedelivery system, MPG, which forms stable noncovalentcomplexes with nucleic acids. MPG is a bipartite amphi-

    pathic peptide derived from both thefusion peptide domain of HIV-1 gp41protein and the nuclear localizationsignal (NLS) of Simian virus 40 largeT antigen. The investigators showedthat cell entry is independent of theendosomal pathway and that the NLS ofMPG is involved in both electrostaticinteractions with nucleic acid and nu-clear targeting. MPG/nucleic acid par-ticles interact with the nuclear importmachinery; however, a mutation thataffects the NLS of MPG disrupts theseinteractions and prevents nuclear deliv-ery of nucleic acids. Therefore, thismutated-MPG system was used for siRNAdelivery into the cytoplasm. Theinvestigators demonstrated that thismutation yields a variant of MPG, whichis a powerful tool for siRNA deliveryinto mammalian cells, enabling rapidrelease of the siRNA into the cyto-plasm and promoting robust down-regulation of target mRNA.

    Complex formation

    One of the major challenges of trans-forming siRNAs from laboratoryreagents to therapeutics is to developan effective drug delivery system. Pre-vious efforts to deliver siRNAs to theliver involved high-pressure tail-veininjections, resulting in silencing of areporter gene in the liver (Morrisseyet al. 2005a). However, this harsh treat-ment requires rapid injection of solu-tions two-and-a-half times the totalblood volume of the animal. Althoughtransfection efficiency of siRNAs intocultured cells is satisfactory for mostin vitro applications, its therapeutic

    FIGURE 5. Gene silencing effect of siRNA conjugated with membrane permeant peptides(MPPs). (A) Conjugation of siRNA with two different MPPs: penetratin and transportan.MPPs and siRNAs were synthesized with a thiol group attached. The reaction between the thiolgroups on the siRNAs and the MPPs was catalyzed by the oxidant diamide. (B) Silencing effectof siRNA on luciferase activity. CHO-AA8-Luc Tet-Off cells stably expressing luciferase weretransfected with 25 nM siRNA conjugated with MPPs (penetratin and transportan) orcomplexed with Lipofectamine 2000. Luciferase activity was measured at days 1, 2, and 3 post-transfection. Reproduced with permission from Muratovska and Eccles (2004).

    siRNA delivery and targeting

    www.rnajournal.org 445

    JOBNAME: RNA 13#4 2007 PAGE: 15 OUTPUT: Tuesday March 6 18:39:52 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • applications in vivo present an altogether more dauntingchallenge. Ideally, a delivery mechanism would (1) becapable of binding siRNAs in a reversible manner to ensuresubsequent release of the siRNAs in target cells; (2) protectsiRNAs from nuclease degradation during transit throughthe circulation; (3) escape from endosomal compartment;(4) be biocompatible; and (5) avoid rapid clearance by theliver and kidney.

    Cationic lipids

    Since the introduction in 1987 of the transfection reagentLipofectin, an equimolar mixture of the cationic lipidN-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammoniumchloride (DOTMA) and the colipid dioleoylphosphatidy-lethanolamine (DOPE), many cationic liposomes have beentested for in vitro and in vivo transfection of nucleic acids.The flexibility in the design of cationic lipid structure andliposome composition combined with the diversity ofmethods for their preparation and in vivo efficiency havepromoted the notion that cationic lipids can be efficientlyused for human gene transfer (Mahato et al. 1997). Thephysicochemical properties of lipid/nucleic acid complexesare strongly influenced by the (1) relative proportions ofeach component; (2) structure of the headgroup of thecationic lipids; (3) cationic lipid: colipid molar and chargeratio; (4) particle size of lipid/nucleic complexes; and (5)liposome size (Mahato et al. 1998; Spagnou et al. 2004).Many effective cationic lipids contain protonable poly-amines linked to dialkyl or cholesterol anchors. Thecationic lipids in the liposomal formulation form com-plexes with DNA or RNA, leading to their enhancedcellular association. In general, cationic lipids with multi-valent headgroups have been shown to be more effective ingene transfer compared to their monovalent counterparts(Mahato et al. 1998, 1999; Keller 2005).

    siRNAs interact electrostatically with cationic liposomesto form particles that are able to transfer them into cells.Since the nature of cationic lipid interactions with DNAand RNA is identical, it seems obvious to adapt pDNAformulations for siRNA delivery. However, pDNA andsiRNA are otherwise very different from each other inmolecular weight and topography with potentially impor-tant consequences. In contrast to pDNA, siRNA cannotcondense into particles of nanometric dimensions, beingalready a small subnanometric nucleic acid (Spagnou et al.2004). Additionally, electrostatic interactions betweensiRNA and cationic liposomes pose two potential problems:(1) a relatively uncontrolled interaction process leading tolipid/siRNA complexes of excessive size and poor stabilityand (2) incomplete encapsulation of siRNA molecules,which thereby exposes siRNA to potential enzymatic orphysical degradation prior to delivery to cells. Such con-siderations should make clear the fact that pDNA andsiRNA are completely different kinds of nucleic acids andthat a lipid/siRNA complex should be regarded as different

    from lipid/pDNA complex formulations (Spagnou et al.2004; Keller 2005).

    Torchilin and colleagues (Zhang et al. 2006) successfullydelivered siRNA into lung tumor cells by loading siRNAinto liposomes bearing arginine octamer (R8), a type ofMPP, attached to the liposome surface. siRNA-loadedR8 liposomes were formulated using 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)/siRNA with theaddition of the PEG-phosphatidylethanolamine. The R8liposomes loaded with HDM2–siRNA demonstrated ahigh stability and protected the incorporated siRNA fromdegradation by blood serum even after 24 h of incubation.This siRNA formulation showed very high transfectionefficiency in lung tumor cells. The R8-liposome-mediatedtransfection takes place even in the presence of plasmaproteins.

    Cationic liposomes are known to interact with serumproteins, lipoproteins, heparin, and glycosaminoglycansin the extracellular matrix, leading to aggregation or releaseof nucleic acids from the complexes even before reachingthe target cells. Cationic lipids can also activate the com-plement system, leading to their rapid clearance by macro-phages of the reticuloendothelial systems (Mahato et al.1997). To circumvent these problems, many liposomalcarriers have been coated with PEG to minimize theirinteraction with serum proteins or complement system andto improve the circulation time. In addition, PEGylationmay help to stabilize the lipid/nucleic acid complexes,leading to the reduction in macrophage clearance. Santelet al. (2006) developed a new liposomal system for siRNAdelivery. This system consists of 50 mol% of b-L-arginyl-2,3-L-diaminopropionic acid-N-palmityl-N-oleyl-amidetrihydrochloride (AtuFECT01), 49 mol% of neutral/helperlipid 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine(DPhyPE), and 1 mol% PEGylated lipid N-(carbonyl-methoxypolyethyleneglycol-2000)-1,2-distearoyl-sn-glycero-3-phosphoethanol-amine sodium salt (DSPE-PEG) (Fig.6A). The new liposomal system was able to deliver Cy3–siRNA into different cells lines (HeLa, HUVEC) in a verylow concentration (10 nM). siRNA delivery by this lipo-some was able to silence protein kinase N3 (PKN3)expression in a concentration-dependent manner as shownin Figure 6B. The liposomal system also showed improvedsiRNA cellular uptake and, more importantly, escape fromthe endocytotic/endosomal pathway into the cytoplasm(Fig. 6C,D). Data from IL-12 ELISA demonstrated that1 mol% of DSPE-PEG in the liposome/siRNA complexwas sufficient to reduce unspecific toxic side effects in vivo.The researchers also injected 1.88 mg/kg of complexedCy3–siRNA intravenously in mice to evaluate the effect ofthe liposome on siRNA biodistribution. Complexed siR-NAs were taken up by the vasculature endotheliumin different organs, mainly liver, heart, lung, spleen,and kidney, showing a profound delay in the clearancerate.

    De Paula et al.

    446 RNA, Vol. 13, No. 4

    JOBNAME: RNA 13#4 2007 PAGE: 16 OUTPUT: Tuesday March 6 18:40:03 2007

    csh/RNA/131733/rna4598

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • Cationic polymers

    Due to high cytotoxicity on repeated use and potent anti-inflammatory activity in vivo (Mahato et al. 1998), interestin polymeric gene carriers is growing. Like cationic lipids,cationic polymers spontaneously form complexes withnucleic acids because of electrostatic interactions betweenpositively charged amine groups of the polycations and

    negatively charged phosphate groupsof the nucleic acids. The interactionbetween cationic polymers/nucleic acidcomplex and negatively charged cellmembranes can enhance its uptake bycells and thus increase transfection effi-ciency (Han et al. 2000). Various cat-ionic polymers have been studied aspotential vectors for gene delivery.These include proteins such as histonesand cationized human serum albumin,polypeptides such as poly-L-lysine(PLL) and poly-L-ornithine, and poly-amines such as PEI and starburst poly-amidoamine (pAMAM) dendrimers.Copolymers containing hydrophilic seg-ments such as PEG and dextran havealso been synthesized.

    pAMAM dendrimers are a class ofhighly branched spherical polymerswhose surface charge and diameter aredetermined by the number of syntheticsteps. The three-dimensional sphericalstructure of dendrimers offers synthesiscontrol in terms of degree and genera-tion of branching. This control allowsthe production of polymer particleswith a very low degree of polydispersity,which is a significant advantage overother polymers such as PLL. The pri-mary amino groups present on thesurface of pAMAM dendrimers arepositively charged at physiological pH.These surface amino groups provideuseful moieties for the functionalmodification of dendrimers since theyallow covalent coupling reactions undermild conditions and in a controlledfashion (Mahato et al. 1999). AlthoughpAMAM dendrimers have been largelyapplied for delivery of plasmids andODNs (Santhakumaran 2004; Hollins2004; Choi 2005), this strategy remainsto be explored for siRNA delivery.Generally, it is expected that the advan-tages observed for ODNs may be adap-ted for siRNA. However, Kang et al.

    (2005) reported that pAMAM dendrimers were not effec-tive for delivery of siRNA against multidrug resistance 1protein (MDR-1) in NIH 3T3 MDR cells.

    Among the cationic polymers employed for gene deliv-ery, PEI has been the most widely used polymer for siRNAdelivery. PEIs with various molecular weights, degrees ofbranching, and other modifications have been largely usedfor transfection of siRNAs in different cell lines and live

    FIGURE 6. In vitro evaluation of lipid/siRNA complexes. (A) Chemical structures of lipidsused for preparation of cationic liposomes and complex formation with siRNAs. (AtuFECT01)b-L-Arginyl-2,3-L-diaminopropionic acid-N-palmityl-N-oleyl-amide trihydrochoride; (helperlipid) 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine; (PEG-lipid) distearoyl-sn-glycero-3-phospho-ethanolamine sodium salt. (B) Concentration-dependent inhibition of PKN3 proteinexpression with lipid/siRNA complex, but not with naked siRNA in HeLa cells as assessed byimmunoblot. PTEN served as the loading control. (C) Intracellular distribution of lipid/Cy3–siRNA complexes in HeLa cells analyzed by confocal microscopy. (D) Merged high-magnifi-cation pictures of a cytoplasmic area from HUVECs showing endosomal/lysosomal localizationof siRNAs. Naked siRNAs remain trapped in the endosomal pathway (upper panel), whereasformulated siRNAs escape the late endosomal/lysosomal vesicles (lower panel). Reprinted bypermission from Macmillan Publishers Ltd.: [Gene Therapy] (Santel et al.) � (2006).

    siRNA delivery and targeting

    www.rnajournal.org 447

    JOBNAME: RNA 13#4 2007 PAGE: 17 OUTPUT: Tuesday March 6 18:40:03 2007

    csh/RNA/131733/rna4598

    Fig. 6 live 4/C

    Cold Spring Harbor Laboratory Press on February 12, 2009 - Published by rnajournal.cshlp.orgDownloaded from

    http://rnajournal.cshlp.org/http://www.cshlpress.com

  • animals (Urban-Klein et al. 2005; Grzelinski et al. 2006).The high transfection efficiency of PEI can be attributed toits buffering effect or the ‘‘proton sponge effect’’ due to itssecondary and tertiary amines. The cytotoxicity and trans-fection efficiency of PEI are directly proportional to itsmolecular weight. PEI is often grafted with PEG orcholesterol to reduce its cytotoxicity (Wang et al. 2002;Patil et al. 2005). Using atomic force microscopy, Grzelinskiet al. (2006) showed that complexation of unmodifiedsiRNAs with PEI leads to the formation of complexes thatcondense and completely cover siRNAs. The investigatorsshowed that the delivery of siRNAs against growth factorpleiotrophin complexed with PEI was able to generateantitumoral effects in an orthotropic mouse glioblastomamodel with U87 cells growing intracranially. Urban-Kleinet al. (2005) showed that noncovalent complexation ofsynthetic siRNAs with low-molecular-weight PEI efficientlystabilizes siRNAs and delivers siRNAs into cells wherethey display full bioactivity at nontoxic concentrations.In a subcutaneous mouse tumor model, intraperitonealadministration of PEI/siRNA complexes, but not ofnaked siRNAs, was delivered into the tumors, resulting ina marked reduction of tumor growth through siRNA-mediated HER-2 receptor down-regulation.

    The presence of readily accessible amine groups on thecationic polymers provides a convenient method forintroducing ligands for receptor-mediated gene delivery.A common approach is to modify the amine using theN-hydroxyl succinimide (NHS) moiety, which forms amideand imide bonds with primary and secondary amines,respectively. The carboxyl groups of PEG can be coupledto the amine in the presence of NHS and carbodiimidereagents. In many cases, a hetero-bifunctional linker is usedfor conjugating the ligand to