throbber
Published online 23 March 2015
`
`Nucleic Acids Research, 2015, Vol. 43, No. 7 3407–3419
`doi: 10.1093/nar/gkv226
`
`SURVEY AND SUMMARY
`Advances in CRISPR-Cas9 genome engineering:
`lessons learned from RNA interference
`Rodolphe Barrangou1,†, Amanda Birmingham2,†, Stefan Wiemann3, Roderick
`L. Beijersbergen4, Veit Hornung5 and Anja van Brabant Smith2,*
`
`1Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695,
`USA, 2Dharmacon, part of GE Healthcare, Lafayette, CO 80026, USA, 3Division of Molecular Genome Analysis, and
`Genomic & Proteomics Core Facility, German Cancer Research Center, 69120 Heidelberg, Germany, 4The
`Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands and 5Institute of Molecular Medicine, University
`Hospital, University of Bonn, 53128 Bonn, Germany
`
`Received January 15, 2015; Revised March 4, 2015; Accepted March 5, 2015
`
`ABSTRACT
`The discovery that the machinery of the Clustered
`Regularly Interspaced Short Palindromic Repeats
`(CRISPR)-Cas9 bacterial immune system can be re-
`purposed to easily create deletions, insertions and
`replacements in the mammalian genome has revo-
`lutionized the field of genome engineering and re-
`invigorated the field of gene therapy. Many paral-
`lels have been drawn between the newly discov-
`ered CRISPR-Cas9 system and the RNA interfer-
`ence (RNAi) pathway in terms of their utility for
`understanding and interrogating gene function in
`mammalian cells. Given this similarity, the CRISPR-
`Cas9 field stands to benefit immensely from lessons
`learned during the development of RNAi technology.
`We examine how the history of RNAi can inform to-
`day’s challenges in CRISPR-Cas9 genome engineer-
`ing such as efficiency, specificity, high-throughput
`screening and delivery for in vivo and therapeutic
`applications.
`
`INTRODUCTION
`From early classical genetic studies to present-day molec-
`ular ones, the ability to modulate gene content and ex-
`pression has been essential to understanding the function
`of genes within biological pathways and their correlation
`with disease phenotypes. The discovery of RNAi and its
`reduction to practice in mammalian cells in the early to
`mid 2000’s made reverse genetics approaches feasible on a
`
`genome scale in higher eukaryotes (1). In the last 24 months,
`another gene modulation technique, Clustered Regularly
`Interspaced Short Palindromic Repeats (CRISPR)-Cas9
`genome engineering (referred to as CRISPR-Cas9), has
`emerged; in that remarkably brief window, this approach
`has proven to be a powerful tool for studying individual
`gene function, performing genome-wide screens, creating
`disease models and perhaps developing therapeutic agents
`(2). These lightning advances have largely followed the path
`blazed by RNAi studies and we argue that further leverage
`is to be gained by examining relevant successes and failures
`in the last 14 years of RNAi.
`RNAi and CRISPR-Cas9 have many clear similarities.
`Indeed, the mechanisms of both use small RNAs with an
`on-target specificity of ∼18–20 nt. Both methods have been
`extensively reviewed recently (3–5) so we only highlight their
`main features here. RNAi operates by piggybacking on
`the endogenous eukaryotic pathway for microRNA-based
`gene regulation (Figure 1A). microRNAs (miRNAs) are
`small, ∼22-nt-long molecules that cause cleavage, degra-
`dation and/or translational repression of RNAs with ad-
`equate complementarity to them (6). RNAi reagents for re-
`search aim to exploit the cleavage pathway using perfect
`complementarity to their targets to produce robust down-
`regulation of only the intended target gene. The CRISPR-
`Cas9 system, on the other hand, originates from the bac-
`terial CRISPR-Cas system, which provides adaptive im-
`munity against invading genetic elements (7). Generally,
`CRISPR-Cas systems provide DNA-encoded (7), RNA-
`mediated (8), DNA- (9) or RNA-targeting(10) sequence-
`specific targeting. Cas9 is the signature protein for Type
`II CRISPR-Cas systems (11), in which gene editing is me-
`
`*To whom correspondence should be addressed. Tel: +1 720 890 5188; Fax: +1 303 604 3286; Email: anja.smith@ge.com
`†
`These authors contributed equally to this work.
`C(cid:3) The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research.
`This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which
`permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.
`
`

`
`3408 Nucleic Acids Research, 2015, Vol. 43, No. 7
`
`Figure 1. The RNAi and CRISPR-Cas9 pathways in mammalian cells. (A) miRNA genes code for primary miRNAs that are processed by the
`Drosha/DGCR8 complex to generate pre-miRNAs with a hairpin structure. These molecules are exported from the nucleus to the cytoplasm, where
`they are further processed by Dicer to generate ∼22-nt-long double-stranded mature miRNAs. The RNA duplex associates with an Argonaute (Ago)
`protein and is then unwound; the strand with a more unstable 5(cid:4) end (known as the guide strand) is loaded into Ago to create the RNA-induced silencing
`complex (RISC) while the unloaded strand is discarded. Depending on the degree of complementarity to their targets, miRNAs cause either transcript
`cleavage and/or translational repression and mRNA degradation. siRNAs directly mimic mature miRNA duplexes, while shRNAs enter the miRNA path-
`way at the pre-miRNA hairpin stage and are processed into such duplexes. (B) CRISPR-Cas9-mediated genome engineering in mammalian cells requires
`crRNA, tracrRNA and Cas9. crRNA and tracrRNA can be provided exogenously through a plasmid for expression of a sgRNA, or chemically synthesized
`crRNA and tracrRNA molecules can be transfected along with a Cas9 expression plasmid. The crRNA and tracrRNA are loaded into Cas9 to form an
`RNP complex which targets complementary DNA adjacent to the PAM. Using the RuvC and HNH nickases, Cas9 generates a double-stranded break
`(DSB) that can be either repaired precisely (resulting in no genetic change) or imperfectly repaired to create a mutation (indel) in the targeted gene. There
`are a myriad of mutations that can be generated; some mutations will have no effect on protein function while others will result in truncations or loss of
`protein function. Shown are mutations that will induce a frameshift in the coding region of the mRNA (indicated by red X’s), resulting in either a truncated,
`non-functional protein or loss of protein expression due to nonsense-mediated decay of the mRNA.
`
`

`
`diated by a ribonucleoprotein (RNP) complex consisting
`of a CRISPR RNA (crRNA) (8) in combination with
`a trans-activating CRISPR RNA (tracrRNA) (12) and a
`Cas9 nuclease (13–16) that targets complementary DNA
`flanked by a protospacer-adjacent motif (PAM) (17–19).
`The molecular machinery from the CRISPR-Cas9 bacte-
`rial immune system can be repurposed for genome edit-
`ing in mammalian cells by introduction of exogenous cr-
`RNAs and tracrRNAs or a single guide RNA chimeric
`molecule (sgRNA) which combines crRNA and tracrRNA
`sequences, together with the Cas9 endonuclease to create a
`double-strand break (DSB) in the targeted DNA (16,20–22)
`(Figure 1B). The DSB is repaired either by non-homologous
`end joining (NHEJ) or homology-directed repair (HDR)
`(23). The error-prone NHEJ pathway typically generates
`small insertions or deletions (indels) that are unpredictable
`in nature, but frequently cause impactful and inactivating
`mutations in the targeted sequence; conversely, the HDR
`pathway is useful for precise insertion of donor DNA into
`the targeted site.
`Both RNAi and CRISPR-Cas9 have experienced signifi-
`cant milestones in their technological development, as high-
`lighted in Figure 2 (7–14,16–22,24–51) (highlighted top-
`ics have been detailed in recent reviews (2,4,52–58)). The
`CRISPR-Cas9 milestones to date have mimicked a com-
`pressed version of those for RNAi, underlining the prac-
`tical benefit of leveraging similarities to this well-trodden
`research path. While RNAi has already influenced many
`advances in the CRISPR-Cas9 field, other applications of
`CRISPR-Cas9 have not yet been attained but will likely
`continue to be inspired by the corresponding advances in
`the RNAi field (Table 1). Of particular interest are the po-
`tential parallels in efficiency, specificity, screening and in
`vivo/therapeutic applications, which we discuss further be-
`low.
`
`EFFICIENCY
`Work performed during the first few years of intensive
`RNAi investigations demonstrated that, when taking 70–
`75% reduction in RNA levels as a heuristic threshold for ef-
`ficiency (59), only a small majority of siRNAs and shRNAs
`function efficiently (24,60) when guide strand sequences are
`chosen randomly. This observation led to the development
`in 2004 of rational design algorithms for siRNA molecules
`(Figure 2), followed later by similar algorithms for shRNAs.
`These methods have been able to achieve ∼75% correlation
`and >80% positive predictive power in identifying func-
`tional siRNAs (61) but have been somewhat less effective
`for shRNAs (62) (perhaps because in most cases, shRNAs
`produce less knockdown than do siRNAs, likely due to a
`smaller number of active molecules in each cell). crRNAs
`also vary widely in efficiency: reports have demonstrated
`indel (insertion and deletion) creation rates between 5 and
`65% (20,25), though the average appears to be between 10
`and 40% in un-enriched cell populations. Indeed, a growing
`amount of evidence suggests a wide range of crRNA effi-
`ciency between genes and even between exons of the same
`gene, yielding some ‘super’ crRNAs that are more func-
`tional (26,27). However, such high-functioning crRNAs are
`
`Nucleic Acids Research, 2015, Vol. 43, No. 7 3409
`
`likely to make up only a small percentage of those randomly
`selected for any given gene (28).
`Following the RNAi playbook, efforts to develop rational
`design algorithms for crRNA have already begun: Doench
`et al. (28) assayed thousands of sgRNAs in a functional
`assay and identified sequence features that predict sgRNA
`activity. In addition, the CRISPR-Cas9 field has moved
`quickly to determine relevant structures and details on the
`separate binding and cleavage processes (29–30,63), thus
`avoiding a deficit of mechanistic information that impeded
`early efforts to predict RNAi efficiency. From these early
`CRISPR-Cas9 studies, it is already understood that the ef-
`ficiency of the crRNA:tracrRNA:Cas9 RNP complex is de-
`termined by PAM-dependent, crRNA-driven Cas9 binding
`to target DNA as well as crRNA sequence complementarity
`to the target DNA (particularly in the first PAM-proximal
`8–10 nt, the ‘seed’ region of the crRNA). Nonetheless, many
`details of CRISPR-Cas9 activity remain to be learned. Be-
`cause an efficient sgRNA or crRNA must not only create a
`DSB in the sequence of interest, but also create a mutation
`that results in functional disruption of the resulting protein
`or non-coding structure, the type of indel as well as its posi-
`tion along the length of the coding sequence are likely to be
`important; thus, effective rational design efforts for func-
`tional knockouts may need to incorporate attributes gov-
`erning these characteristics as well. Furthermore, more than
`a decade of efforts in the RNAi field demonstrates that even
`a well-characterized system is not necessarily easy to pre-
`dict: crRNA design algorithms, like those for siRNAs and
`shRNAs, may plateau in their predictive power at a level
`that still necessitates testing of multiple reagents per target
`in order to guarantee selection of a functional one.
`It is worth noting that efficient gene silencing by an RNAi
`reagent requires a process of ongoing, active repression; this
`mechanism may be impaired by gene-specific factors that
`increase mRNA turnover and/or decrease RNA-induced
`silencing complex (RISC) turnover. As such, there are some
`genes for which no RNAi reagents can be found that qual-
`ify as effective. In contrast, efficiency of a crRNA depends
`upon the probability of a one-time event––the editing of a
`DNA site. Because of this difference, the success of rational
`design efforts may be less critical to the CRISPR field on an
`individual gene basis, as it will be widely possible to find an
`effective crRNA if one is willing to evaluate enough treated
`cells; highly effective crRNA designs, however, will still be
`necessary for genome-scale studies, as discussed further be-
`low.
`
`SPECIFICITY AND OFF-TARGET EFFECTS
`Perhaps in no other area are the lessons of RNAi as obvious
`as in that of specificity. While RNAi was originally hailed
`as exquisitely specific (64), subsequent research has shown
`that in some circumstances it can trigger non-specific ef-
`fects and/or sequence-specific off-target effects (65). Many
`non-specific effects seen with this approach are mediated by
`the inadvertent activation of pattern recognition receptors
`(PRRs) of the innate immune system that have evolved to
`sense the presence of nucleic acids in certain sub-cellular
`compartments. siRNA length, certain sequence motifs, the
`absence of 2-nt 3(cid:4) overhangs and cell type are important fac-
`
`

`
`3410 Nucleic Acids Research, 2015, Vol. 43, No. 7
`
`Figure 2. Timeline of milestones for RNAi and CRISPR-Cas9. Milestones in the RNAi field are noted above the line and milestones in the CRISPR-Cas9
`field are noted below the line. These milestones have been covered in depth in recent reviews (2,4,52–29).
`
`Table 1. Summary of improvements in the CRISPR-Cas9 field that can be anticipated by corresponding RNAi advances
`
`Milestone
`
`IND application
`
`Off-target driver
`identification
`
`Off-target-reducing
`modifications
`
`Large-scale arrayed
`screening
`
`in vivo use (human)
`
`Phase III entry
`
`RNAi
`
`2004
`
`2006
`
`2006
`
`2007
`
`2010
`
`2014
`
`CRISPR
`
`This step is undoubtedly imminent. The drug that was the subject of the first RNAi IND
`failed clinical trials when its effect was shown to be due to non-RNAi-related
`mechanisms; especially since CRISPR therapeutics require the delivery not only of a
`targeting RNA but of exogenous Cas9 (delivered as DNA, mRNA or protein),
`pharmaceutical developers must avoid allowing history to repeat itself.
`Current work is characterizing the nature and extent of the PAM-proximal crRNA
`‘seed’. Until it is complete, novel outcomes must be demonstrated using multiple reagents
`to the same target, as is routinely done for RNAi. Once the crRNA seed is understood,
`researchers should determine whether it could be leveraged to develop sequence-specific
`off-target controls such as RNAi’s C911 controls.
`While effective specificity-enhancing chemical modifications for CRISPR may have to
`wait until off-target drivers are more fully understood, synthetic crRNAs should be
`modifiable by precisely the same methods as synthetic siRNAs.
`Genome-wide arrayed screens using CRISPR are likely to be more challenging because
`the percentage of edited cells is typically lower than for RNAi. Nonetheless, CRISPR
`screening and analysis practices will build on and extend those designed for RNAi
`screening, just as the latter did with those for small-molecule screening.
`As CRISPR-driven editing in adult human cells has already been achieved, in vivo
`human use seems inevitable. Efficacious delivery, including that of the exogenous Cas9
`protein (or Cas9 mRNA) necessary to make integration-less DNA modifications, is likely
`to present a significant hurdle. Novel delivery formulations developed in pursuit of
`RNAi therapeutics will undoubtedly be among those tried first.
`CRISPRa and other dCas9-based approaches raise the hope of addressing conditions
`untreatable purely via RNAi-like down-regulation while retaining the reversible nature of
`RNAi. The two modalities might profitably be used in parallel.
`
`tors for induction of the mammalian interferon response
`(66–68). Additionally, the general perturbation of cellular
`or tissue homeostasis by the delivery process itself can also
`trigger unwanted responses (most likely secondary to innate
`immune damage-sensing pathways) such as the wide-spread
`alteration of gene expression caused by cationic lipids, es-
`pecially when used at high concentrations (69). Such non-
`
`specific effects associated with delivery will still exist for
`CRISPR-Cas9 but can likely be overcome by minimizing
`lipid concentration as is now routinely done in RNAi stud-
`ies. Similarly, the introduction of chemical modifications
`into the backbone of an siRNA duplex (e.g. 2(cid:4)-O-methyl
`ribosyl) can block the recognition of RNA molecules by
`PRRs (66,70–71), so such modifications may also address
`
`

`
`innate immune system recognition caused by synthetic cr-
`RNAs. Researchers would do well to investigate whether
`additional effects may result, potentially in a cell-line or
`cell-type dependent manner, as a response to creation of
`DSBs or the abundant expression of Cas9 or an sgRNA
`molecule (such as that seen when strong shRNA expression
`outcompetes that of endogenous miRNAs, leading to the
`breakdown of cellular regulation (72)). These types of non-
`specific off-target effects have already been reported with
`other genome engineering techniques (e.g. zinc finger nucle-
`ases (73)) but the ease-of-use and simplicity of the CRISPR-
`Cas9 system should allow researchers to address these types
`of questions fully in the near future.
`RNAi can also produce sequence-specific off-target ef-
`fects, which were initially described in early 2003 (31),
`but whose potential impact was not fully appreciated un-
`til well after the method had become a widely used re-
`search and screening technique (e.g. (74)). Cleavage-based
`off-targeting, which occurs when RISC encounters an
`unintended transcript target with perfect or near-perfect
`complementarity to its guide strand, can induce knock-
`down equivalent to that of intended target down-regulation
`and was originally hypothesized to be the main cause of
`sequence-specific off-target effects. It took several years to
`determine that these effects were in fact primarily caused
`by RNAi reagents acting in a ‘miRNA-like’ fashion, down-
`regulating unintended targets by small (usually <2-fold)
`amounts primarily through seed-based interactions with
`the 3(cid:4) UTR of those unintended targets. Because miRNA-
`like off-targeting is generally seed-based and all transcripts
`contain matches to a variety of 6–8-base motifs, such off-
`targeting can affect tens to hundreds of transcripts. Further-
`more, if the RNAi reagent contains a seed mimicking that
`of an endogenous miRNA, the off-targeting may affect the
`pathway or family of targets evolutionarily selected for reg-
`ulation by that miRNA. It is not possible to design RNAi
`reagents that do not contain seed regions found in the tran-
`scriptome’s 3(cid:4) UTRs and the non-seed factors that conclu-
`sively determine whether or not a seed-matched transcript is
`in fact off-targeted have not yet been identified. Both ratio-
`nal design and chemical modifications such as 2(cid:4) O-methyl
`ribosyl substitutions can mitigate seed-based off-target ef-
`fects (32), but without a full solution, specificity remains a
`well-known pain point for RNAi users.
`Inspired by these concerns, an initial evaluation of the
`off-target potential of CRISPR-Cas9 was published within
`months of the technique’s debut and work to refine these
`early findings has continued apace. Studies have revealed
`some sequence flexibility, and tolerance for mismatches
`and bulges, that have generated concerns about specificity
`and sequence-directed off-target cleavage (25,75–79). Sev-
`eral variations of CRISPR-Cas9 have been developed to ad-
`dress specificity including paired nickases (77), short sgR-
`NAs (76) and Cas9 fused to FokI (80), and the rapid ad-
`vances in understanding CRISPR-Cas9 mechanism and
`structure are likely to further fuel such developments. Re-
`cent papers, however, have uncovered very few to no off-
`target mutations that can be attributed to CRISPR-Cas9
`(33,34) and conclude that clonal artifacts that derive from
`isolating CRISPR-Cas9-edited cells may be a larger con-
`cern. This apparent discrepancy between prediction and re-
`
`Nucleic Acids Research, 2015, Vol. 43, No. 7 3411
`
`ality, while encouraging, highlights a treacherous pitfall in
`studying off-target gene editing: to date the primary ap-
`proach has been to predict putative off-target sites and then
`search for editing at those sites, but this approach risks
`falling prey to the ‘streetlight effect’, in which one searches
`only where it is easy to look. The RNAi field learned this
`lesson painfully: early off-target prediction efforts focused
`on strong overall complementarity as a determinant and
`thus largely failed to identify genes that were actually off-
`targeted due to short, seed-based complementary (35). Un-
`less the CRISPR-Cas9 field learns from RNAi’s mistakes,
`it is in danger of repeating the same very one, especially as
`CRISPR-Cas9 specificity has recently also been shown to
`depend on an as-yet-not-fully-defined seed region (20,81).
`At this stage, computational predictions of putative off-
`target gene-editing sites are at best questionable guesses and
`thus cannot be depended upon in assessing the full effect of
`off-targeting. Unfortunately, while RNAi specificity stud-
`ies are limited to the transcriptome, analysis of CRISPR-
`Cas9 specificity requires identifying effects that may occur
`anywhere in the genome, generally through next-generation
`sequencing. This process is costly and depends upon non-
`trivial data analysis and processing, so it remains unclear
`whether the field has the will to commit to this work. Until
`such time as a less biased understanding of CRISPR-Cas9
`off-targeting emerges, researchers are advised to emulate the
`best-practice of RNAi by using multiple crRNAs or sgR-
`NAs in order to show redundancy of phenotype by multi-
`ple reagents targeting the same gene, thus ensuring that the
`phenotype is due to on-target effects rather than off-target
`effects.
`
`GENOME-SCALE SCREENING TOOLS
`Interest in genome-scale CRISPR-Cas9-based screening
`has blossomed, with some pooled screening resources al-
`ready available (26,27) and arrayed ones likely to emerge in
`the near future. Because genome editing screens will also be
`affected by a large number of the factors that make RNAi
`screens more challenging than small-molecule ones (82),
`practitioners would do well to study the hard-won victories
`in this field since the first published whole-genome synthetic
`lethal screen for sensitization to paclitaxel (36) (Figure 2)
`before diving into these costly experiments.
`Of particular importance is evaluating whether the lower
`efficiencies seen using CRISPR-Cas9 are sufficient to gen-
`erate a desired phenotype in the screening assay––that is,
`determining whether the phenotype is detectable in the tar-
`geted cell population. In this regard, two factors are of spe-
`cial concern: the ploidy of the gene locus of interest (as
`tumor cell lines are often aneuploid) and the likelihood
`of disrupting the reading frame by the induced mutation
`(since +3 or −3 indels would not serve this purpose). Tak-
`ing these factors into account, the chance of obtaining a
`high percentage of cells that have a functional knockout
`in a bulk cell culture is relatively low under typical screen-
`ing conditions. Consequently, it is unlikely that traditional
`arrayed loss-of-signal screens such as those common in
`RNAi will be widely feasible in bulk-transfected cells using
`CRISPR-Cas9. Nevertheless, the CRISPR-Cas9 technol-
`ogy may have an advantage in screens for which a complete
`
`

`
`3412 Nucleic Acids Research, 2015, Vol. 43, No. 7
`
`knockout is required to uncover a phenotype (for exam-
`ple, when targeting kinases where a residual expression of
`10% is sufficient for activity). The use of analysis techniques
`that examine effects at the single-cell level (using, for exam-
`ple, high-content microscopy or fluorescence-activated cell
`sorting-based read-outs) could also be informative, as it has
`been for gene silencing screens. A more novel possibility
`is leveraging HDR to develop systems in which CRISPR-
`Cas9-mediated DSBs are repaired using an exogenous tem-
`plate harboring a marker or resistance gene that can later be
`used to select cells with a functional, inactivating, recombi-
`nation event (83).
`Homologous recombination could similarly be used to
`validate RNAi screening hits by introducing variants in
`the transcript that do not alter the protein but create mis-
`matches with the siRNA or shRNA reagents such that tar-
`get knockdown no longer occurs, providing a fast method
`to distinguish on-target and off-target effects. More simply,
`gene editing can be used to knock out a putative hit to deter-
`mine if the knockout results in a similar phenotype to that
`obtained with knockdown during the RNAi screen (84). Al-
`though (as discussed further below) knockout phenotypes
`may differ from knockdown phenotypes (so that a negative
`result may not necessarily indicate a false positive), confir-
`mation of the knockdown phenotype with a CRISPR-Cas9
`knockout would be a strong indication of a true positive.
`Conversely, RNAi reagents may be an effective validation
`strategy for CRISPR-Cas9-based screens and the numer-
`ous other effective approaches optimized for RNAi screens,
`such as confirmation of assay phenotype by multiple inde-
`pendent reagents and the use of additional related assays
`(58), will prove directly applicable to genome editing vali-
`dation.
`The development and application of RNAi-based pooled
`screening approaches have greatly enhanced the field of
`functional genomics screening in mammalian cells: the
`ability to quantify in large populations the relative abun-
`dance of individual cells, each carrying a gene-specific gene-
`modifying reagent, allows for different screening models
`such as identification of genotype-specific essential genes,
`synthetic lethal genes or genes involved in resistance to spe-
`cific drugs (85). Crucial requirements for this approach are
`the availability of reagents with high efficiency and speci-
`ficity, the presence of tractable markers for integrated con-
`structs and the availability of large collections of gene-
`perturbing reagents in retroviral or lentiviral vectors. In
`contrast to the current lack of arrayed screening resources
`for CRISPR-Cas9, large, genome-scale collections of sgR-
`NAs that fulfill these criteria are available alongside anal-
`ogous shRNA collections. Numerous examples of both
`pooled RNAi screens and pooled CRISPR-Cas9 screens
`have been published (26–27,86–90,37).
`CRISPR-Cas9 pooled screens share with their RNAi-
`based cousins the necessity of inactivating gene activity at
`single-copy integration of the shRNA- or sgRNA-encoding
`expression cassette. This proves a difficulty for both tech-
`nologies, as efficiency of shRNA-mediated knockdown
`varies for different platforms but never reaches complete
`knockdown for all vectors targeting a specific gene (91,92)
`and the observed frequency of CRISPR-Cas9-based gene
`inactivation using single-copy sgRNAs shows great vari-
`
`ability among the different studies, on average not reaching
`frequencies higher than 50% (20,25). RNAi screeners have
`demonstrated that such limited efficiency is more challeng-
`ing for screening models aimed at the identification of lethal
`genes, synthetic lethal genes or response enhancers than for
`enrichment experiments like resistance screens or positive
`selection of a phenotype such as expression of a cell surface
`marker. To combat this limitation, libraries for CRISPR-
`Cas9 pooled screening are already including multiple inde-
`pendent reagents for each gene. As shown by shRNA-based
`pooled screens, this approach insures against false nega-
`tives due to individual inefficient reagents and provides a
`means of corroborating each reagent’s result, thereby reduc-
`ing false positives.
`False-positive off-target effects in both RNAi and
`CRISPR-Cas9 pooled libraries will also be mitigated by ra-
`tional design, as discussed above. Further, recently devel-
`oped C911 seed match controls (93) can be implemented
`in large-scale shRNA screening collections as internal off-
`target matched controls; such a technique would be highly
`desirable for CRISPR-Cas9 screening but its feasibility will
`depend on further understanding of the relevant specificity
`mechanisms. So far, the levels of off-target effects in pooled
`screens using CRISPR-Cas9-based gene editing remains
`unclear, although results from the comparison of shRNA
`and CRISPR-Cas9 screens in the same screening model (for
`genes whose loss confer resistance to vemurafenib, a BRAF
`protein kinase inhibitor) in A375 melanoma cells support
`a low frequency of off-target effects (26) in the CRISPR-
`Cas9 system. With regards to false negatives, one would
`expect that a considerable advantage of the CRISPR-Cas9
`technology over shRNA-mediated knockdown would be in-
`creased strength of phenotype due to the ability to com-
`pletely abolish expression of the targeted gene. As a re-
`sult, one would predict improved recovery of genes involved
`in the phenotype of interest, and indeed, it has been re-
`ported that the recovery rate for essential genes is higher for
`CRISPR-Cas9 than for shRNA (94). However, the afore-
`mentioned genome-wide CRISPR-Cas9 screen for cellu-
`lar resistance to vemurafenib identified a limited number
`of hits compared to a similar screen with a genome-wide
`shRNA collection (26). It may be that CRISPR-Cas9-based
`screens are unable to identify genes that are lethal upon
`complete loss, but are associated with the desired pheno-
`type when knocked down by 70–90%. An example of such
`a gene is SOX10, which causes a slow-growth phenotype
`upon knockdown; this phenotype is associated with resis-
`tance to vemurafenib (95). CRISPR-Cas9 screeners should
`take this potential bias into account when analyzing their
`screening results.
`
`INVIVOSTUDIES
`Following the footsteps of RNAi, CRISPR-Cas9 has
`quickly advanced beyond studies in cell lines and primary
`cell cultures to in vivo studies aimed at everything from
`examination of the biology of particular genes and dis-
`ease phenotypes to development of potential therapeutic
`agents. Notably, however, this technology provides signifi-
`cant advances in the creation of animal models for mecha-
`nistic studies that RNAi, given its transient and partial na-
`
`

`
`ture, cannot offer. Focusing on in vivo studies in the mouse,
`Wang et al. (96) demonstrated that CRISPR-Cas9 can be
`introduced into embryonic stem cells in a multiplex fash-
`ion to create animals carrying multiple specific mutations
`in several genes in a manner requiring only one genera-
`tion, thereby dramatically decreasing the time required to
`generate transgenic animal models. Additional studies have
`used CRISPR-Cas9 to create mouse models of various can-
`cers by mutating a combination of tumor suppressor genes
`and oncogenes in the livers of wild-type mice (97) or in
`mouse hematopoietic stem cells (98), eliminating the need
`for time-consuming creation and crossbreeding of genet-
`ically engineered mouse strains. The modification of the
`mouse genome using CRISPR-Cas9 technology is not lim-
`ited to gene mutations: large chromosomal deletions, inver-
`sions and translocations can be produced by using multiple
`sgRNAs (99–101).
`The final goal of much in vivo work is the development
`of therapeutic tools. In spite of challenges regarding deliv-
`ery and non-specific effects (including those that caused the
`first RNAi-based therapeutic candidate by OPKO Health
`to fail phase III clinical trials in 2009), considerable efforts
`and investments continue in the pursuit of RNA-targeting
`therapeutics. More than 30 clinical trials are currently in
`progress or completed on indications from pachyonychia
`congenita to high cholesterol (102,103). Recently, advances
`in non-viral delivery systems have been made with the de-
`velopment of lipopeptide nanoparticles that offer the op-
`portunity to treat disease via in vivo delivery to endothe-
`lial cells or hepatocytes (104,105). Given this enduring in-
`terest in gene-modulation-based drugs, it seems certain that
`CRISPR-Cas9-based treatments will shortly enter the ther-
`apeutics pipeline; recent proof-of-principle studies (Table 2)
`point to likely indications (106–115). Gene-editing thera-
`peutics may enjoy a smoother road than gene-silencing-
`based ones since they have no requirement for continuous
`delivery of siRNAs or continuous expression of integrated
`shRNAs. As a consequence, gene editing can be done with-
`out leaving a footprint in the genome other than the cor-
`rected DNA sequence. While gene-editing therapeutics may
`have the advantage of not requiring continuous delivery
`or expression of RNAs, RNAi has the advantage of

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