Vous êtes sur la page 1sur 13

Please cite this article in press as: Esposito et al.

, Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review

Hacking the Cancer Genome: Profiling


Therapeutically Actionable Long Non-coding RNAs
Using CRISPR-Cas9 Screening
Roberta Esposito,1,2 Núria Bosch,1,2,3 Andrés Lanzós,1,2,3 Taisia Polidori,1,2,3 Carlos Pulido-Quetglas,1,2,3
and Rory Johnson1,2,*
1Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
2Department for BioMedical Research, University of Bern, Bern, Switzerland
3Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland

*Correspondence: rory.johnson@dbmr.unibe.ch
https://doi.org/10.1016/j.ccell.2019.01.019

Long non-coding RNAs (lncRNAs) represent a huge reservoir of potential cancer targets. Such ‘‘onco-
lncRNAs’’ have resisted traditional RNAi methods, but CRISPR-Cas9 genome editing now promises func-
tional screens at high throughput and low cost. The unique biology of lncRNAs demands screening strategies
distinct from protein-coding genes. The first such screens have identified hundreds of onco-lncRNAs pro-
moting cell proliferation and drug resistance. Ongoing developments will further improve screen perfor-
mance and translational relevance. This Review aims to highlight the potential of CRISPR screening technol-
ogy for discovering new onco-lncRNAs, and to guide molecular oncologists wishing to apply it to their cancer
of interest.

Introduction ship to target gene (cis or trans). With exceptions, lncRNAs are
The past decade has witnessed a dramatic expansion in both the thought to function as mature RNAs (Guttman and Rinn, 2012).
number and nature of cancer-related genes (Khurana et al., They interact with other RNAs, genomic DNA (gDNA) sites, and
2016; Bhan et al., 2017). Technological advances in tumor proteins, through both structural and sequence-specific ele-
genome sequencing and functional genomics have identified a ments (Guttman and Rinn, 2012; Johnson and Guigó, 2014).
diverse cast of genomic elements whose mutation, epigenetic Their presumed ability to adopt complex tertiary structures un-
modification or altered expression contribute to ‘‘cancer hall- derlies the formation of multi-component complexes, increasing
marks’’ (Hanahan and Weinberg, 2000; Futreal et al., 2004; Li lncRNAs’ potential biological roles, but hugely complicating the
et al., 2014a). Among these, long non-coding RNAs (lncRNAs) study of their mechanisms (Guttman and Rinn, 2012; Johnson
are a numerous yet poorly understood gene class that have and Guigó, 2014). They are found in both the nucleus and cyto-
generated widespread interest as novel cancer targets (Du plasm of the cell, with a slight nuclear tendency (Ulitsky and Bar-
et al., 2013; Yan et al., 2015; Zhu et al., 2016; Lanzós et al., tel, 2013; Cabili et al., 2015). Finally, they can act both in cis or
2017; Liu et al., 2017). trans, influencing the chromatin structure, gene transcription,
LncRNAs are defined for what they are not: RNA transcripts, in and RNA processing of either neighboring or distantly encoded
the range of 200–10,000 nucleotides, where no evidence can be target genes (Quinodoz and Guttman, 2014; Vance and Ponting,
found for protein-coding capacity (Derrien et al., 2012). They 2014; Kopp and Mendell, 2018). So far, they have been impli-
comprise a large and heterogeneous class that, in contrast cated in diverse cellular processes, ranging from the control of
to other ncRNAs, have resisted functional classification and pre- gene expression to protein translation to ion channel activity
diction (Ulitsky and Bartel, 2013; Kopp and Mendell, 2018). With (Ulitsky and Bartel, 2013; Johnson and Guigó, 2014).
exceptions, lncRNAs have gene structures, chromatin, tran- The numerical population of lncRNAs is particularly daunting:
scriptional regulation, and post-transcriptional processing remi- recent estimates have up to 50,000 or more, far exceeding the
niscent of protein-coding genes (Derrien et al., 2012). Although number of protein-coding genes (Uszczynska-Ratajczak et al.,
many are species specific, a core shows evidence for ancient 2018). Ever-increasing volumes of transcriptome sequence
evolutionary conservation (Ulitsky et al., 2011). In general, data are unlikely to have yet saturated the full extent of lncRNAs
lncRNAs are characterized by low expression and high tissue- (Nellore et al., 2016; Lagarde et al., 2017). Thus, even in a pessi-
and cell-specificity (Derrien et al., 2012). While the biological mistic scenario where just a fraction of lncRNAs are functional,
functionality of many lncRNAs remains debated (Engreitz et al., there are likely to remain many thousands of undiscovered genes
2016; Kopp and Mendell, 2018), it is beyond doubt that func- with therapeutic potential.
tional examples exist (Wutz et al., 2002; Gutschner et al., 2013; For a number of years, we have been unable to capitalize on
Lee et al., 2016; Leucci et al., 2016; Hosono et al., 2017), this wealth of novel genes due to lack of effective and econom-
including those with clearly established cancer roles. ical loss-of-function (LOF) tools. The widely used RNAi has
The molecular mechanisms of lncRNAs are highly heteroge- proven generally ineffective for lncRNAs, due to their unique
neous and can be interpreted along three principal axes: molec- localization and expression (Gutschner et al., 2011; Maamar
ular interactions, subcellular localization, and regulatory relation- et al., 2013). A solution has recently appeared thanks to recent

Cancer Cell 35, April 15, 2019 ª 2019 Elsevier Inc. 1


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review
Growth erties in most types of solid tumors (Chakravarty et al., 2014),
ANRIL supression GAS5
through diverse and complex mechanisms (Chakravarty et al.,
Apoptosis 2014). Perhaps the best studied of these is through the organiza-
PANDA MEG3
tion of nuclear architecture. It has been shown that NEAT1 is
necessary for the formation of paraspeckles, poorly understood
- 3’ nuclear structures (Clemson et al., 2009; West et al., 2014).
5’-
Tumor-suppressor Recently, NEAT1 was shown to be a direct target of p53
Onco-lncRNA
lncRNA (Adriaens et al., 2016; Mello et al., 2017). ANRIL is an antisense
- 3’
5’- lncRNA, transcribed from the locus INK4b-ARF-INK4a, encod-
ing for three tumor-suppressor proteins, p15(INK4b), p14(ARF),
HULC NBAT1 and p16(INK4a). ANRIL overexpression in tumors results in the
Proliferation transcriptional repression of the three genes, and subsequent
evading of growth suppression mechanism (Yap et al., 2010).
HOTAIR Invasion LINC-PINT
In addition, MALAT1 has been reclassified as a cancer-lncRNA
instead of onco-lncRNA, since Kim et al. (2018) characterized
Figure 1. LncRNAs in Cancer Phenotypes
Selected examples of onco-lncRNAs (left) and tumor-suppressor lncRNAs it as tumor-suppressor in in vivo models of breast cancer.
(right) shown to contribute to four cancer hallmarks. Green and red arrows The number of known oncogenic lncRNAs is growing rapidly,
represent positive and negative regulation, respectively. Colored ovals of which a small number of illustrative cases are described here.
represent proteins or RNAs that bind to lncRNAs and are necessary for their
The lncRNA PANDA, induced in a p53-dependent manner, re-
function.
presses apoptosis by interacting with the transcription factor
NF-YA and preventing the transcription of pro-apoptotic genes
advances in genome-engineering technology. The CRISPR- (Hung et al., 2011). SAMMSON is a direct target of the metas-
Cas9 technology is a potent and versatile toolkit with which tasis marker, SOX10, and promotes melanoma cell survival by
to manipulate a wide variety of genomic elements (Cong et al., enhancing mitochondrial metabolism. It interacts with a key
2013). Most importantly, CRISPR appears to overcome the regulator of mitochondria metabolism, p32, involved in formation
drawbacks of previous methods and can effectively target of functional 12S ribosomal RNA (Leucci et al., 2016). Another,
lncRNAs (Aparicio-Prat et al., 2015). LINK-A, localizes to the cytosolic membrane, directly interacts
The aim of this Review is to provide a comprehensive introduc- with phosphatidylinositol-3,4,5-trisphosphate and AKT (Lin
tion to CRISPR-based screening for cancer-promoting lncRNAs. et al., 2017), mediating AKT recruitment and activation. Hyperac-
We summarize recent successful screens of lncRNAs involved in tivation of this signal has been associated with tumorigenesis
cell proliferation and drug resistance. We advise the reader on and resistance to AKT inhibitors in breast cells (Lin et al., 2017).
achieving high-sensitivity, high-specificity screens by means of LncRNAs can act also as tumor-suppressors: GAS5, MEG3,
optimal design and analysis strategies, while avoiding common NBAT, and LINC-PINT, often downregulated in cancer, play
pitfalls. Finally, we discuss how future developments in CRISPR important roles in key cellular processes (Zhou et al., 2012;
screens promise to unlock a new generation of treatments for Marin-Bejar et al., 2013; Pandey et al., 2014; Pickard and Wil-
unmet clinical needs in cancer. liams, 2015) (Figure 1). For example, LINC-PINT lies downstream
of p53 and acts as its regulatory effector, inhibiting tumor cell in-
LncRNAs: The Next Frontier of Cancer Targets vasion (Marin-Bejar et al., 2013). LINC-PINT downregulation
In a remarkably short period of time, lncRNAs have been linked leads to increased tumorigenesis in mouse models, and its
to essentially every major cancer type (for a detailed review expression is lost in many tumors (Marin-Bejar et al., 2017).
see Huarte, 2015; DiStefano, 2018; Lin and Yang, 2018), contrib- The rapid growth in known onco-lncRNAs has begun to feed
uting to each of the ten cancer hallmarks (Hanahan and Wein- into therapies. This has been facilitated by progress in devel-
berg, 2000, 2011; Gutschner and Diederichs, 2012; Bartonicek oping reagents capable of downregulating RNAs in vivo. Anti-
et al., 2016; Arun et al., 2018) (Figure 1). We here define lncRNAs sense oligonucleotides (ASOs) are a broad class of nucleic acids
that positively contribute to cancer hallmarks as ‘‘onco- that bind to RNAs and trigger their degradation by the RNaseH
lncRNAs,’’ and those that contribute either negatively or posi- pathway (Dias and Stein, 2002). In contrast to RNAi, this degra-
tively as ‘‘cancer-lncRNAs.’’ dation takes place in the nucleus, meaning that nuclear, chro-
Iconic onco-lncRNAs such as HOTAIR, NEAT1, ANRIL, and matin-associated, and even primary (unspliced) lncRNAs, can
MALAT1 fulfill the definitions of conventional oncogenes: they be efficiently targeted (Fang and Fullwood, 2016). Chemical
are recurrently overexpressed in tumors, their introduction to modifications can improve the stability of ASOs in vivo, most
or removal from cultured cancer cells alters growth and invasive- notably DNA:RNA chimeric ‘‘Gapmers’’ (Watts and Corey,
ness, and they are involved in core biological processes in 2012). Other methods may be used to tune the tissue- or cell-
healthy cells (Gupta et al., 2010; Yap et al., 2010; Gutschner specific targeting of ASOs (Juliano, 2016; Ross et al., 2017;
et al., 2013; Chakravarty et al., 2014; Leucci et al., 2016; Sun Scharner et al., 2018).
et al., 2016). HOTAIR, originally identified in metastatic breast Attempts to discover functional lncRNAs have historically
cancer (Gupta et al., 2010), was subsequently linked to a variety relied on indirect transcriptomic evidence, using microarrays or
of cancers (revised in Tang and Hann, 2018). However, the RNA sequencing (RNA-seq) to find differentially expressed
mechanisms of action remain uncertain and debated (Portoso genes between tumor and healthy samples (Iyer et al., 2015).
et al., 2017). NEAT1 is upregulated and shows oncogenic prop- For example, HULC was first detected through its deregulation

2 Cancer Cell 35, April 15, 2019


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review

Table 1. CRISPR-Based Screens for Cancer-lncRNAs


Pooled Library Library lncRNA Identified PubMed
Perturbation Cas9 Variant In Vitro Model Size (Targets) Addgene IDs Phenotype (Validated) ID
CRISPR-del wild-type Cas9 Huh7.5OC 671 89640 cell growth 51 (9) 27798563
CRISPRi dCas9-KRAB K562, U87, HeLa, 16,401 86538–86550 cell growth 499 (65) 27980086
HEK293T, MCF-7,
MDA-MB-231, iPSC
CRISPRa dCas9-VP64 + MCF-7 241 – AKT activation 5 (1) 28176758
pSM2-p65-HSF1
CRISPRa dCas9-VP64 + A375 10,504 1000000106 vemurafenib 16 (16) 28792927
pSM2-p65-HSF1 resistance
CRISPRa dCas9-VP64 + MOLM14 14,701 – Ara-C 2,874 (13) 29677511
pSM2-p65-HSF1 resistance
CRISPR-del wild-type Cas9 K562 10,996 – cell growth 230 (35) 30395134

in hepatocellular carcinoma (Panzitt et al., 2007), while RMRP been successful across diverse perturbation modalities, lncRNA
has been linked to multiple cancers through expression deregu- targets, cell models, and phenotypes (Table 1).
lation (Meng et al., 2016; Shao et al., 2016; Feng et al., 2017). Three LOF screens have been presented, using CRISPR-dele-
This approach suffers from the underlying assumptions that tion (CRISPR-del) and CRISPR-inhibition (CRISPRi) (explained in
differential expression implies function, and, inversely, that can- detail in the next section). Zhu and colleagues used a paired-
cer functions must be reflected in tumor-specific expression sgRNA (pgRNA) promoter deletion strategy with a library target-
changes. ing 700 previously identified cancer-associated lncRNAs (Du
Cancer-lncRNAs have been also identified through copy- et al., 2013), yielding 51 hits (7%) for proliferation-regulating
number alterations (CNAs) in tumor genomes. For example, lncRNAs in a liver cancer cell line (Zhu et al., 2016). Forty-
PVT1 and SAMMSON were initially detected due to their co- three of these promoted proliferation, corresponding to onco-
amplification with nearby protein-coding oncogenes MYC and lncRNAs. Very recently, the same group developed an alterna-
MITF, respectively. Further experiments revealed key roles for tive LOF screen targeting the splice site of 10,996 multi-exonic
these lncRNAs in tumor progression (Shtivelman and Bishop, lncRNAs and identified 230 (2%) to be essential for cell growth
1989; Leucci et al., 2016). Recently, the advent of whole-genome of K562 cells (Liu et al., 2018).
sequencing has enabled the search for ‘‘driver’’ mutations in A CRISPRi-based study screened a much larger library target-
lncRNAs, which are under positive selection (Rheinbay et al., ing 16,401 lncRNAs in seven cell lines (Liu et al., 2017). Here, 499
2017). For example, SAMMSON (Lanzós et al., 2017), MIAT (Mu- hits (3%) were identified (Liu et al., 2017). Surprisingly, the major-
laroni et al., 2016), and RMRP (Rheinbay et al., 2017) are en- ity yielded phenotypes in just one cell type, despite being ex-
riched for somatic mutations in tumors. Although promising, pressed in multiple cells, suggesting that lncRNAs exert cell-
driver discovery methods are restricted by presently small type-specific functions independent of steady-state expression
numbers of entire tumor genome sequences. Both driver and levels and highlighting the advantage of CRISPR screening
CNA analyses only identify relatively early mutations in tumori- over expression-based methods.
genesis, and likely represent a small subset of clinically action- Other studies have used the gain-of-function (GOF) perturba-
able onco-lncRNAs. tion, CRISPR-activation (CRISPRa). A screen in MCF7 cells iden-
Despite their value, these methods for identifying onco- tified 5 out 241 lncRNAs (2%) to be involved in the AKT activation
lncRNAs have failed to keep pace with the rate at which new pathway (Koirala et al., 2017). Another study in melanoma cells
lncRNAs are annotated, and generally do not yield direct func- identified 16 lncRNA loci involved in resistance to the targeted
tional evidence for cancer roles. What is more, lncRNAs’ unique therapeutic vemurafenib (Joung et al., 2017a, 2017b). Bester
biology present challenges compared with protein-coding et al. (2018) identified 2,874 hits out of 14,701 (19%) lncRNAs
genes, and their functions cannot be predicted based on primary for their response to Ara-C drug, in an acute myeloid leukemia
sequence. The result is that just a tiny fraction of lncRNAs, 500 cell lines.
(1%), have been linked to cancer so far (Chen et al., 2013; Quek Independently from the different modalities used (LOF, GOF),
et al., 2015; Ning et al., 2016; Carlevaro-Fita et al., 2017). This these studies provide several insights. First, they confirm that
has created a pressing need for methods capable of functionally many novel lncRNAs mediate cancer hallmarks, and may be
screening thousands of lncRNAs at low cost. identified using cell-based genome-wide screens. Second, hit
rates appear to lie in the range 2%–20%––surprisingly high,
CRISPR Screening for Onco-LncRNAs considering that neither LOF study specifically targeted lncRNAs
CRISPR-Cas9 technology represents the long-awaited break- that were expressed in their respective cell lines.
through technology for perturbing lncRNAs at high throughput,
using a pooled screening format requiring low capital invest- CRISPR-Cas9 Toolkit for LncRNAs
ment. Recent landmark studies have demonstrated CRISPR- CRISPR-Cas9 genome engineering is an adaptation of the
Cas9’s power to discover onco-lncRNAs. These studies have Streptococcus pyogenes immunity system (Cong et al., 2013;

Cancer Cell 35, April 15, 2019 3


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review

A Cas9 B Wild-type CRISPR-deletion


Cas9

LOF
-3’

GTACATCGTACGCATTGTACNCC 0.5-5 kb
5’- CATGTTACGCATGCTACATG

Protospacer (20 nt)


NGG

PAM

CRISPR-inhibition
KRAB
C
dCas9

Cargo

F
LO
25-75 bp

CRISPR-activation
VP64
G
O
F

100-150 bp

Figure 2. Flavors of CRISPR Perturbations


(A) CRISPR-Cas9 system. The basic CRISPR-Cas9 system consists of two components. The single guide RNA (sgRNA) hybridizes to the 20-nt protospacer in
genomic DNA through its variable region, while recruiting Cas9 protein through a constant, structured domain. The protospacer must precede a protospacer-
associated motif (PAM), which in the case of Cas9 is ‘‘NGG.’’ Recruitment of Cas9 results in a double-strand break (DSB) three nucleotides upstream the PAM.
(B) CRISPR-deletion. LncRNA knockout may be achieved by CRISPR-deletion (CRISPR-del). Here, a pair of wild-type Cas9 endonucleases are recruited to sites
flanking the target transcription start site (TSS) (arrow). Non-homologous end-joining (NHEJ) repairs the lesion, resulting in promoter deletion and gene silencing.
(C) CRISPR-inhibition/activation. Transient perturbations may be achieved through mutated nuclease-deficient Cas9 (dCas9). dCas9 can be fused with a variety
of cargoes, i.e., effector domains, to modulate lncRNA expression. The KRAB domain (red, upper panel) inhibits transcription (CRISPR-inhibition [CRISPRi]),
while activators such as VP64 (green, lower panel) activate transcription (CRISPR-activation [CRISPRa]). Efficiency of all methods depends strongly on relative
locations of sgRNAs to lncRNA TSS, shown below.

Mali et al., 2013). It comprises two components: Cas9 endonu- duce simultaneous DSBs at sites flanking the target region,
clease protein, capable of inducing a double-strand break relying on a non-homologous end-joining (NHEJ) process to
(DSB) in bound DNA, and a single guide RNA (sgRNA) repair the break and remove the intervening fragment (Ho
(Figure 2A). The sgRNA is itself a synthetic fusion of the et al., 2015; Vidigal and Ventura, 2015) (Figure 2B). CRISPR-
CRISPR-RNA (crRNA), containing a variable 20-nt sequence del has been widely used on lncRNAs and other genomic
complementary to the targeted protospacer, and a constant targets, and typically yields target homozygous deletion effi-
trans-activating crRNA (tracrRNA) recognized by Cas9 (Fig- ciencies in the range 10%–40% (Canver et al., 2014; Ho et al.,
ure 2A) (Jinek et al., 2012). The Cas9 will only cut when an 2015). Factors affecting this efficiency are poorly understood,
NGG protospacer adjacent motif is found immediately 30 to the but likely include target length (Canver et al., 2014) and the
protospacer (Jinek et al., 2012) (Figure 2A). In effect, CRISPR- rate of NHEJ. This approach suffers from a number of chal-
Cas9 may be considered a genomic search engine serving to lenges, in terms of co-delivery of two sgRNAs, and efficiency
deliver the Cas9 cargo to a defined genomic site. One of the of the NHEJ process.
key benefits of CRISPR/Cas9 is its versatility: the Cas9 protein An important first decision is what exactly to delete: the entire
can be modified to deliver protein cargoes––and hence, pertur- lncRNA gene, or just part of it? The first lncRNA targeted through
bations––of choice to desired genomic locations. genomic deletion was the mouse Rian lncRNA gene, where a
LncRNAs introduce special constraints to CRISPR-Cas9 as 23-kb fragment was eliminated (Han et al., 2014). This was sub-
originally designed for protein-coding genes. For the latter, it is sequently extended to other lncRNAs (Ho et al., 2015; Holdt
sufficient to recruit a single Cas9 to the open reading frame et al., 2016; Koirala et al., 2017; Xing et al., 2017). Full-gene dele-
(ORF), where indel mutations lead to frameshifts and complete tion has two main drawbacks. First, when deleting large regions,
LOF of the encoded peptide (Shalem et al., 2014). As lncRNAs there is the risk of false-positive phenotypes by inadvertently
have no ORF, it is unlikely their functional domains can be inac- removing other overlapping genomic elements (Bassett et al.,
tivated in this way (Guttman and Rinn, 2012). Consequently, 2014; Groff et al., 2016). An example is provided by Linc-p21,
elaborations of the basic CRISPR-Cas9 approach have been thought to be a cis-acting lncRNA, which actually contains mul-
developed to target lncRNAs. These can be divided into perma- tiple enhancer elements within the DNA sequence. The deletion
nent genome editing (deletions) or transitory transcriptional of these elements is responsible for the transcriptional regulation
modulation (inhibition and activation) (Figures 2B and 2C). Dis- of surrounding genes (Groff et al., 2016). Second, long gene-
cussion is restricted to methods that can presently be scaled sized deletions have low efficiency (Canver et al., 2014).
to screening throughputs. As a result, subsequent studies have silenced lncRNAs by
Genomic Deletion removing smaller regions of 0.5–5 kb encompassing promoter
Perhaps the most obvious LOF method is by genomic deletion. and transcriptional start site (TSS) (Zhu et al., 2016) (Figure 2B).
CRISPR-del uses a pair of CRISPR-Cas9 complexes to intro- This has been successfully applied to lncRNAs such as Meteor

4 Cancer Cell 35, April 15, 2019


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review

(Alexanian et al., 2017) and MALAT1 (Aparicio-Prat et al., 2015). tions. Because it targets exonic sequences, it should specifically
Cells carrying promoter deletions display strongly reduced target lncRNAs without affecting any overlapping protein-coding
lncRNA expression levels, with allelic deletion efficiencies up to genes. Moreover, it could resolve the present controversy over
60% in unsorted cells (Pulido-Quetglas et al., 2017). Anecdotally, lncRNAs’ (non-mutually exclusive) modes-of-action: whether
one problem arising is that mutated genes may switch expres- they function (1) as mature RNA molecules or (2) simply through
sion to nearby, cryptic promoters, although this has not been the ‘‘act of transcription’’ (Latos et al., 2012; Kopp and Men-
tested systematically. Certainly, residual expression of the dell, 2018).
lncRNA is often observed (Bassett et al., 2014); therefore, in in-
stances where complete silencing is required, a full-gene dele- Screening for LncRNAs: A User’s Guide
tion might be preferable. When designing a CRISPR/Cas9 screen, one must address
Transcriptional Repression by CRISPRi three key considerations: Which perturbation––activation,
Cas9 can also be engineered for transient LOF. Here, a catalyt- repression or deletion? Which sgRNA library to choose (i.e.,
ically inactive version of Cas9 (dCas) is used as an RNA-guided targeting which lncRNAs)? Which cell model and pheno-
targeting platform to which effector domain cargoes are fused typic assay?
(Qi et al., 2013) (Figure 2C). Strong repression is achieved by Here we guide new users in addressing these questions. We
addition of repressor domains, such as the Kru €ppel-associated introduce the pooled CRISPR screening workflow, and guide
box or concatenated mSin3 interaction domains (SID4X) (Gilbert the reader through challenges and benefits of CRISPR ap-
et al., 2013; Dominguez et al., 2016). Such configurations typi- proaches, with key design considerations for achieving high-
cally achieve knockdown efficiencies up to 15-fold, and are sensitivity, high-specificity screens.
applicable to both protein-coding and lncRNA genes (Gilbert Pooled Screening
et al., 2013; Stojic et al., 2018). CRISPRi produces fewer false- Pooled CRISPR screening allows one to assay hundreds to thou-
positive results than RNAi (Buehler et al., 2012; Smith et al., sands of lncRNAs in a rapid and cost-effective way, without the
2017a, 2017b; Peretz et al., 2018). In contrast to CRISPR-del, requirement of robotic equipment. The approach is based on a
LOF is reversible because no genetic mutation takes place (Man- decade of development of analogous short hairpin RNA (shRNA)
degar et al., 2016). screens utilizing the RNAi pathway, but avoiding its pitfalls
Transcriptional Activation by CRISPRa of low knockdown efficacy and frequent off-targeting (Stojic
Analogous to CRISPRi, transient GOF can be achieved by et al., 2018).
CRISPRa (Figure 2C). Although early setups gave relatively Pooled screens entail a ‘‘phenotype-to-genotype’’ evaluation
weak effects (Gilbert et al., 2013; Perez-Pinera et al., 2013; Cha- of gene function, and, in common with all screening modalities,
vez et al., 2015), more complex systems achieve potent activa- follow a common three-step structure: (1) perturbation, (2) selec-
tion, such as the fusion of dCas9 with a SunTag array that re- tion, and (3) hit identification (Figure 3). What sets pooled screens
cruits multiple copies of VP64 (Tanenbaum et al., 2014). The apart from more common arrayed screening, is that instead of
combination of p65 and HSF1 fused to the sgRNA (by means introducing perturbations one-by-one in separate culture wells,
of an aptamer) showed among the best activation profile by re- they are performed in a single mixed population of cells.
cruiting numerous downstream activation proteins (Konermann The perturbation step of pooled library screening is achieved
et al., 2015). CRISPRa overcomes numerous drawbacks associ- by means of a library of sgRNAs in a lentiviral backbone
ated with cDNA overexpression. CRISPRa achieves endoge- (Figure 3A), where virions carry one of many thousands of
nous gene upregulation, which is expected to more accurately distinct targeting constructs (either shRNAs or sgRNAs). This is
recreate natural patterns of transcript isoform expression (Ko- used to infect the cell model (Figure 3B), at low multiplicity of
nermann et al., 2015), and also to faithfully model cis transcrip- infection (MOI, 0.3), thereby ensuring that each individual cell
tional effects that appear to be lost with plasmid overexpression receives a single virion and hence perturbation (Figure 3C). Every
(Seiler et al., 2017). Finally, CRISPRa oversteps the difficulty of infected cell carries a unique, retrievable record of this perturba-
cloning large cDNAs into vectors with limited capacity. tion, in the form of a genomically inserted sgRNA sequence.
The major concern of CRISPRi/a is the risk of off-target effects Cells are cultured under antibiotic selection for a determined
arising through perturbations of nearby genes (Goyal et al., period post-infection, to allow perturbation to take place and re-
2017). Therefore, these techniques should ideally only be applied move uninfected cells.
for the manipulation of lncRNA promoters distal from protein- In the selection step, sgRNA-expressing cells are subjected to
coding genes (Goyal et al., 2017). an assay that is capable of physically selecting cells according
Direct RNA Targeting: The Future of Screening? to the phenotype of interest (Pheno+). As a reference, non-
Although not yet implemented for screening, a new CRISPR mo- phenotype cells (Pheno) are also harvested as appropriate
dality holds great promise: ‘‘CRISPR-R,’’ or direct degradation of (Figure 3C). Commonly used assays are cell proliferation (where
RNA. Wild-type Cas9 can recognize, bind, and cut RNA mole- faster proliferating cells dominate the population over time), sort-
cules, even in the absence of DNA (O’Connell et al., 2014; Nelles ing by a fluorescent marker, or resistance to a drug. sgRNAs
et al., 2016). Degradation has also been achieved by fusing from selected cells can be retrieved, sequenced, and used to
dCas9 to the PIN RNA endonuclease domain from SMG6 (Batra infer the identity of genes whose perturbation led to the pheno-
et al., 2017), or using another Cas homolog, Cas13 (Abudayyeh type of interest. The requirement for a selectable phenotypic
et al., 2017). assay is a key constraint of such screens. Fortunately, a
Knocking down RNA without altering transcriptional rates range of widely used assays are available that model cancer
potentially gives CRISPR-R advantages over other perturba- hallmarks.

Cancer Cell 35, April 15, 2019 5


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review

A B Figure 3. Pooled Screening Workflow of the


CRISPR-Del Approach for lncRNAs
Library design Oligo synthesis
(A) Library construction. A pooled oligonucleotide
library of pgRNAs is designed, targeting pro-
dCas9 dCas9 moters of 1,000 lncRNAs with n = 10 indepen-
Cas9 or KRAB or VPR
LcnRNA1 LcnRNA2 dent constructs. Here the perturbation is CRISPR-
10 pgRNA designs 10K Oligos del, but the same workflow also applies for
x 1000 LncRNAs Transduction CRISPRi/a, which only differ in employing one
sgRNA rather than a pair (represented by scis-
sors). The resulting library of 103 unique se-
Virus production Plasmid library cloning
Cell model quences are synthesized as on oligonucleotide
Selection pool, cloned into a vector backbone, and used to
produce infectious lentivirus.
(B) Stable cell line production. Cells stably ex-
>500K colonies for 50x coverage Cas9-expressing population pressing Cas9/dCas9 are established by trans-
fection or infection, and selected by antibiotics to
remove non-expressing cells.
(C) Screening. Stable cells are infected with the
C lentivirus pool, at a low multiplicity of infction (MOI,
0.3), so that the majority of cells receive one
Pooled infection Expansion single genomically inserted library sequence. After
MOI 0.3
antibiotic selection to remove uninfected cells,
30M cells Proliferation cells are subjected to the phenotypic assay, which
Drug response separates those displaying phenotype of interest
Phenotypic selection Invasion (Pheno+) from the rest (Pheno). Finally, cell pop-
Migration ulations are harvested and stored. It is crucial to
Apoptosis maintain sufficient coverage of the library at every
point in the protocol, so that each library sequence
is represented by a large number of individual
cells, and no bottlenecks occur that could
10M cells randomly eliminate certain library sequences.
Coverage 1000x (D) PCR amplification. Genomic DNA is purified,
and PCR used to amplify lentivirally inserted
sgRNAs. PCR primers contain adaptamers and
barcodes, in yellow and blue, respectively, needed
Pheno- Pheno+
for the subsequent NGS sequencing. The PCR
product is sequenced and mapped to the library
design, in order to count the instances of each li-
D brary sequence.
(E) Sequencing, read alignment and statistical
Barcode analysis. Three different lncRNAs are shown, each
Adaptamer gDNA – vector – sgRNA – vector – sgRNA – vector – gDNA represented by three unique sgRNAs. In the
example, NGS reads are represented by colored
bars. Examples of neutral (in gray), depleted
(in red) and enriched (in green) lncRNAs, are
E provided.

Neutral Depleted Enriched


lncRNA lncRNA lncRNA
is compared between Pheno+ and
sgRNAs 1 2 3 1 2 3 1 2 3 Pheno cell populations, and statistically
significantly enriched or depleted
Pheno- sgRNAs implicate their target genes as
modulators of the phenotype of interest
Pheno+
(Figure 3E). Correctly performed, pooled
screens can be rapid, economical, and
yield hits with strong statistical confi-
Fold
~1 <1 >1 dence and direct relevance to cancer.
change
Choosing an Optimal Perturbation
FDR >0.05 ≤0.05 ≤0.05 Selecting your CRISPR perturbation
should begin with several considerations.
LncRNA LncRNA
Function None promotes represses Are the lncRNAs of interest expressed in
phenotype phenotype the selected cell model? If the answer is
affirmative, then LOF by CRISPR-del or
CRISPRi are obvious choices. Inciden-
In the final hit identification step, gDNA is extracted, the lenti- tally, for lncRNA screens, no study has yet systematically inves-
virally inserted sgRNA sequences are amplified by PCR and the tigated which is most effective (for a systemic comparison
frequency of all sgRNAs in the library quantified by next-genera- of platforms see Rosenbluh et al., 2017; Sanson et al.,
tion sequencing (NGS) (Figure 3D). The frequency of each sgRNA 2018). CRISPRi involves cloning single sgRNAs, whereas the

6 Cancer Cell 35, April 15, 2019


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review

pgRNA-expressing vectors for CRISPR-del require a two-step better represents the baseline impact caused by DSBs (Aguirre
cloning process (Aparicio-Prat et al., 2015). On the other hand, et al., 2016; Haapaniemi et al., 2018).
GOF by CRISPRa is useful when lncRNAs are not expressed in TSS Annotation. TSSs of lncRNAs are often mis-annotated
the cell model (Joung et al., 2017a, 2017b). From a therapeutic by hundreds of kilobases (Lahens et al., 2014; Lagarde
point of view, LOF methods are preferable over GOF, because et al., 2017). Correct TSS identification is crucial because
they more closely model available ASO drugs, which are gener- dCas9 and CRISPR-del constructs are only effective within a
ally inhibitory. Ideally, studies should integrate multiple parallel small window around the TSS. Consequently, public lncRNA
CRISPR perturbations to maximize sensitivity and specificity gene annotations like GENCODE (Derrien et al., 2012) should
of results, for example CRISPR-del and CRISPRi in the same be combined with other experimental evidence to validate
cell model. However, for laboratories embarking on CRISPR TSS predictions. Most prominently Cap Analysis of Gene
screening for the first time, we would suggest that CRISPRi Expression (CAGE) is regarded as the single most valuable ev-
probably represents the best starting point for LOF screens at idence, and datasets are available for thousands of human and
this point, thanks to the availability of tools, pre-constructed li- mouse organs and cell types (FANTOM Consortium and the
braries, and previous literature. RIKEN PMI and CLST (DGT) et al., 2014). Additional evidence
Library Selection and Design sources are DNAseI hypersensitive regions and chromatin
The sgRNA library should ideally target the entire set of lncRNAs states (ChromHMM) (Ernst and Kellis, 2017). Ideally such
expressed in the cell model of interest. In practice, this is impos- data should have been collected from cells or organs of rele-
sible to achieve, due to our relatively poor annotations of vance to the screening cells. More detailed discussion of this
lncRNAs, even for deeply studied cell lines (Uszczynska-Ratajc- topic may be found in (Radzisheuskaya et al., 2016; Sanson
zak et al., 2018). In practice, researchers face the choice of (1) et al., 2018).
using an available pre-made library targeting lncRNAs that sgRNA Design. Design tools aim to select optimal 20-mer pro-
may not be an ideal match to the user’s cell type of interest, tospacer sequences to create sgRNAs with maximal on-target
or (2) create a new custom library for the cell type of interest, efficacy and minimal off-target effects. Typical libraries are de-
but with the considerable expertise and investment that this signed with three to ten unique constructs per target lncRNA,
entails. in order to remove false-positive sgRNAs and boost statistical
Pre-made libraries are available from AddGene’s collection power at the analysis stage (Figure 3A). As a consequence,
(https://www.addgene.org/crispr/libraries/) (see Table 1). They off-target filters may be set less stringent than for other applica-
target generic sets of cancer-associated lncRNAs, or sets of tions. Recent sgRNA design tools incorporate nucleotide/dinu-
lncRNAs that are expressed in a specific cell line (the latter tar- cleotide composition, favored and disfavored nucleotides at
gets far more genes, and hence entails larger-scale screens). each position, length, and RNA thermodynamics, to successfully
Using such libraries in a first project is clearly a practical option, distinguish high-efficacy sgRNAs (Doench et al., 2014, 2016;
provided they cover reasonable numbers of lncRNAs expressed Moreno-Mateos et al., 2015; Xu et al., 2015).
in the cells of interest. Perhaps more important is the relative position of the proto-
For the majority of cancers, pre-made libraries are not yet spacer to TSS of targeted lncRNA (Horlbeck et al., 2016a; San-
available, necessitating creation of a customized library. This op- son et al., 2018). CRISPRi and CRISPRa have strong require-
tion is more challenging and time-consuming, but allows one to ments for proximity to the TSS: 25–250 bp downstream and
create a reagent tailored closely to the experimental objective. approximately 150–75 bp upstream of the TSS, respectively
Library design is composed of three main steps: (1) selection (Figures 2B and 2C) (Horlbeck et al., 2016a, 2016b; Radzisheus-
of candidate target genes, (2) identification of their transcription kaya et al., 2016; Rosenbluh et al., 2017; Sanson et al., 2018). On
start site (TSS), and (3) design of optimal sgRNAs (Horlbeck the other hand, CRISPR-del constraints are less well under-
et al., 2016a; Sanson et al., 2018). stood, but certainly deletion efficiency is inversely correlated
Candidate Selection. LncRNAs may be selected on the basis of with pgRNA distance (Canver et al., 2014).
two main criteria: their suspected involvement in the cancer of in- An important hurdle is that most sgRNA design tools either
terest, and their favorable gene properties. Suspected lncRNAs require some bioinformatics knowledge to create large-scale li-
could be identified by expression in a cancer of interest (Du et al., brary designs (Hodgkins et al., 2015; Xu et al., 2015; Doench
2013; Zhu et al., 2016). Our ability to specifically target lncRNAs et al., 2016), or else only provide low-throughput webservers,
depends on organization of their gene locus. More specifically, like GPP Web Portal for CRISPRi/a/del (Sanson et al., 2018),
for the 50% of lncRNA overlapping protein-coding genes, there MiT for CRISPR-del (Hsu et al., 2013), and CRISPOR (Haeussler
is a risk of off-target effects through inadvertently affecting the et al., 2016). An exception is CRISPETa, which performs large-
latter (Derrien et al., 2012; Goyal et al., 2017; Bester et al., scale designs via webserver but presently only for CRISPR-del
2018). For this reason, one should consider selecting only inter- (Pulido-Quetglas et al., 2017).
genic (non-overlapping) lncRNAs (Goyal et al., 2017). In addition, Cell Model
a number of control elements should be included: positive Screens performed in vitro in cancer cell lines will be the focus
controls (targeting mRNAs and lncRNA known to be involved here, although shRNA studies have demonstrated the feasibility
in the phenotype under study), negative controls (sgRNAs target- of in vivo screens (Delas et al., 2017). Cell lines have several ad-
ing non-genic genomic regions), and non-targeting controls vantages in terms of handling and culture, rapid and indefinite
(sgRNAs that do not match any genomic region). The distinction growth, and ease in deriving stable expressing clones. Selected
between the latter two is that although non-targeting controls cell lines should model the cancer of interest as closely as
may better represent neutral controls, using negative controls possible. Furthermore, cells should ideally have available

Cancer Cell 35, April 15, 2019 7


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review

genomic data, such as RNA-seq, CAGE, and histone modifica- One approach to overcome this impasse is to convert negative
tions. Particularly valuable in this regard are ENCODE and selection screens to positive selection screens. Take as an
the Cancer Cell Atlas (https://www.genome.gov/26524238/ example an LOF screen for genes that protect cells from death
encode-project-common-cell-types/) (ENCODE Project Con- in response to a drug. In a conventional negative selection
sortium et al., 2012). (dropout) setup, one would collect surviving cells as Pheno+,
An important drawback of CRISPR-del screens in cancer cell and search for dropout sgRNAs. This can be converted to a pos-
models is the somatic CNAs of genes. Two studies have shown itive selection screen by collecting dying cells as Pheno+. Now,
that double-stranded breaks caused by targeting amplified sgRNAs targeting protective lncRNAs will be enriched in Pheno+
genomic regions in human cancer cell lines reduces proliferation cells (Arroyo et al., 2016).
or survival, and may cause false-positive results (Aguirre et al., The screening phenotypes attempted to date (Table 1) repre-
2016; Munoz et al., 2016). Although these false-positive hits sent a small fraction of the wide variety of available assays (Zhu
affect only a small fraction of essential genes identified by et al., 2016; Joung et al., 2017a, 2017b; Liu et al., 2017; Bester
CRISPR (Munoz et al., 2016), it is fundamental to control these et al., 2018). Genes affecting migration or apoptosis have been
potential artifacts. Delivery of Cas9 is another key consideration. dissected in the past for mRNA shRNA screening (Li et al.,
The Cas9 sequence is long (4 kb) (Graham and Root, 2015), 2014a; Seo et al., 2014). Adapting such phenotypic assays to
sharply reducing the infection efficiency of lentivirus. Two main CRISPR screening will allow the exploration of lncRNAs
alternatives are to deliver the Cas9 gene in the same lentiviral involved in other cancer hallmarks with high biomedical rele-
vectors as the sgRNA, or separately. The former option suffers vance. For instance, it is possible to sort apoptotic cells by
from low library infection efficiency, due to length. Improved li- fluorescence-activated cell sorting (FACS) after annexin V
brary infection efficiency can be achieved by separate delivery: staining (Arroyo et al., 2016). Similarly, Transwell inserts can
stable cell lines expressing Cas9 are first created (Figure 3B), be adapted to physically sort cells on the basis of migration
and the sgRNA library is subsequently delivered from a separate rate (Smolen et al., 2010; Seo et al., 2014). Nevertheless, the
vector at high efficiency (Sanjana et al., 2014). It is preferable to technical challenges of such assays should not be underesti-
work with heterogeneous populations of Cas9-expressing cells, mated. FACS can be time-consuming: genome-wide screens
rather than isolated clonal lines, due to artifactual epigenetic require tens of millions of cells, equating to several hours of
characteristics of the latter (Stojic et al., 2018). sorting. Similarly, scaling presently available Transwell sizes
Phenotypic Assay to the number of cells will involve hundreds of parallel experi-
In principle, CRISPR screens can be combined with a wide variety ments, and may also require multiple rounds of selection to
of established cell-based phenotypic assays, ranging from the achieve sufficient population separation (Smolen et al., 2010;
simplest, such as proliferation, to more complex phenotypes Seo et al., 2014).
including drug resistance, migration, and marker gene expression. Optimal Screening Considerations
Pooled screening suffers from an important trade-off between During library creation and screening there are certain critical pa-
practicality and biomedical relevance. Under a phenotypic rameters that must be controlled to ensure sensitivity and spec-
screen, cells can experience one of two outcomes when a can- ificity of final results.
cer-lncRNA is perturbed: negative and positive selection. Nega- Firstly, it is fundamentally important to maintain adequate li-
tively selected perturbations are particularly relevant in cancer brary representation throughout the experiment. Representation
LOF screens, where the overarching goal is to identify drug tar- refers to the number of unique molecules (i.e., plasmids, infected
gets, which, when inhibited, lead to a loss in cancer phenotype. cells, NGS reads) that carry each library sequence, and should
In LOF screens, negatively selected sgRNAs will be depleted or far exceed the number of sequences in the library (ideally,
lost in the selected (Pheno+) population, and are commonly known 500-fold or more). This ensures that individual library sequences
as ‘‘dropouts.’’ A classic dropout screen is represented by simple do not randomly disappear from the population due to stochastic
LOF proliferation screens, where sgRNAs that disappear from the effects, and yield sufficient statistical power for hit identification.
population after a defined period identify genes that positively Suggestions for maintaining library coverage are shown in
regulate growth (Shalem et al., 2014). The drawback of relying Figure 3.
on dropouts is limited sensitivity: the dynamic range of detection A second critical issue relates to various artifacts that can arise
of dropouts is between 1-fold (of the Pheno population) and during two main PCR steps, being amplification of (1) the oligo-
zero, and will be proportionally reduced where perturbation effi- nucleotide pool during library cloning, and (2) genomic inserts for
ciency is <100%. False-positive dropouts can also appear if bot- NGS. For both, it is important to rigorously minimize amplifica-
tlenecks occur in the cell population due to low library coverage. tion cycles. A promising solution is to track individual library mol-
Conversely, positively selected cells are collected because ecules using unique molecular identifiers (UMI), which can be
they display an aberrant phenotype, and will be enriched in incorporated into the library design (Roy et al., 2018), or before
Pheno+ populations. If the perturbation is LOF, then such cells PCR amplification of the library preparation for sequencing
will have developed a cancer phenotype due to loss of a targeted (Hong and Gresham, 2017; Smith et al., 2017a, 2017b) or into
lncRNA––for example, a growth-inhibitory gene in a proliferation the lentiviral backbone (Michlits et al., 2017). By enabling one
assay. Such lncRNAs have lower biomedical relevance (due to to count individual cells and molecules, instead of PCR ampli-
the difficulty of therapeutically activating a target gene), but are cons, UMIs should improve the accuracy of final hit identifica-
typically more readily identified, because their dynamic range tion. However, even without them, the use of multiple unique tar-
of detection varies from 1 to infinity, and, in contrast to dropouts, geting constructs per lncRNA should go a long way to eliminating
should be largely unaffected by perturbation efficiency. PCR artifacts.

8 Cancer Cell 35, April 15, 2019


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review

Finally, it is recommended to perform at least biological dupli- The Path Ahead


cates for each screen for reasons of statistical sensitivity in hit CRISPR screening promises to unlock the potential of lncRNAs
identification (Li et al., 2014b). as therapeutic targets in cancer. Its simplicity, low cost, and flex-
ibility bring transcriptome-wide screens within reach of the
Identification of Screen Hits average molecular oncology laboratory. We anticipate that
The final step of a CRISPR-screen is the identification of lncRNA CRISPR screening for lncRNAs will be widely implemented
‘‘hits’’ from raw NGS data. Hits are defined on the basis of enrich- across all the major cancer types in the coming years. Neverthe-
ment or depletion of their sgRNAs in Pheno+ cells, compared less, it has clear conceptual differences to classical arrayed
with Pheno (Figure 3). screening, and labs embarking on such a project for the first
From each selected cell sample, gDNA is extracted and lenti- time must have a clear understanding of the optimal screen set
viral inserts amplified by PCR using generic, barcoded primers up for their system of interest. We have sought here to address
flanking single or paired gRNAs (Figure 3D). gPCR products this, by introducing the rationale for CRISPR screening and prac-
carry unique sequence identifying their target, in a fragment tical steps for its implementation.
compatible with Illumina HiSeq, while barcodes allow one We foresee a number of future developments that will improve
to track individual samples and multiplex samples during NGS. the sensitivity and selectivity of screens, while improving the
When working with pgRNAs for CRISPR-del, paired-end biomedical relevance of hits. Firstly, screening phenotypes will
sequencing should be used to capture both sgRNA sequences. become more sophisticated and focused. So far, published
After NGS, raw sequencing reads are mapped to the original screens have been limited to the two most straightforward
library design and used to count instances of each sequence phenotypes: proliferation and drug resistance. However, the
in the library (Figure 3E). The output will be a file containing the oncology field has developed a wide array of in vitro cell-based
read counts for each sgRNA across all the Pheno+/– and repli- phenotypic assays that model classical cancer ‘‘hallmarks,’’
cated samples. A table of these counts represents the input for and can often be readily adapted to pooled screening format.
bioinformatic hit identification software. We see the exciting prospect of such assays being used to
Several statistical algorithms have been used to identify hits screen for lncRNAs mediating more focused hallmarks such as
in pooled screens using read counts: negative binomial distri- migration, invasion, or sensitivity to apoptosis.
butions (MAGeCK and ENCoRE), Bayesian analysis (BAGEL), With the broad adoption of CRISPR screening technology for
or probability mass function of a binomial distribution (STARS) studying the function of lncRNAs, a systematic evaluation of
(Li et al., 2014b; Doench et al., 2016; Hart and Moffat, 2016). CRISPR is essential. An important goal for the field is to create
Requirements are different for each tool; while STARS and confident gold standard sets of lncRNAs known to influence
BAGEL require as input file the read count table, MAGeCK phenotypes such as proliferation. Such sets can be incorporated
and ENCoRE accept raw reads and perform median normaliza- into targeting libraries, in order to benchmark screen perfor-
tion on read counts of the different samples to adjust for the mance across cell lines and labs. Such efforts will benefit from
effect of library sizes and read count distributions. Finally, previous efforts such as ProjectDRIVE, which created lists of
ENCoRE provides a graphical interface, enabling non-bio- essential protein-coding genes across hundreds of cell lines us-
informaticians to perform a complete analysis starting from ing RNAi (McDonald et al., 2017).
raw sequencing data (Trumbach et al., 2017). Of these Although cell lines are the most widespread model used for
methods, MAGeCK would appear to be the most widely screening up-to-date, they often do not faithfully recapitulate
used. Importantly, all these methods leverage the presence of the complex biology of their original tumor. In theory, any cell
multiple sgRNAs per target and experimental replicates to type can be screened, including primary cells (Parnas et al.,
improve statistical confidence in hits. 2015), organoids (Matano et al., 2015), and in vivo xenografts
The final outcome is a ranked series of lncRNA hit predictions (Parnas et al., 2015). Although they carry important limitations,
with false discovery rate (FDR) estimates and effect sizes to these models better represent the tumor environment and
serve as the starting point for experimental validation and should be adapted to screens. The most obvious challenge for
follow-up. Before embarking on time-consuming experimental all these models is generating sufficient cell numbers, and this
validation, it is prudent to first examine the performance of nega- may be overcome by screening smaller libraries of candidates
tive and positive control sgRNAs and targets. Furthermore, given obtained from prior, larger screens performed in cell lines (Wil-
that the FDR correction of some methods appears to be exces- liams et al., 2017).
sively stringent (specifically, MAGeCK; Zhu et al., 2016), a helpful Another area for improvement is lncRNA annotation: present
parallel method for hit identification is through the generation gene collections are poor, both in terms of missing many
of quantile-quantile plots, to examine the behavior of uncorrec- lncRNAs, and in the TSS definition of included genes (Uszczyn-
ted p values. ska-Ratajczak et al., 2018). The effectiveness of CRISPRi/a and
Validations can be carried out using different perturbations to CRISPR-del depend crucially on identifying the correct TSS, so
deeply characterize lncRNA functions. For example, the PVT1 inaccurate annotations are likely to result in numerous false-
promoter was predicted to have tumor-suppressor activity by negative predictions. Advances in lncRNA annotations will result
a CRISPRi screen (Liu et al., 2017), in contrast to the PVT1 in larger and more potent CRISPR libraries.
mature RNA, known to act as an oncogene (Tseng et al., CRISPR technologies themselves are in a phase of rapid
2014). In a follow-up study, Cho et al. (2018) integrated development. CRISPRi and CRISPRa enzymes are being engi-
CRISPR-del, CRISPRa, CRISPRi, and other additional methods, neered for increased potency. Similarly, CRISPR-del efficiency
to validate the tumor-suppressor hypothesis. falls far short of 100% and may be improved in the future

Cancer Cell 35, April 15, 2019 9


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review

(Song et al., 2017). Indeed, future technologies might implement Canver, M.C., Bauer, D.E., Dass, A., Yien, Y.Y., Chung, J., Masuda, T., Maeda,
T., Paw, B.H., and Orkin, S.H. (2014). Characterization of genomic deletion
some kind of blunt end insertion to silence and tag edited cells efficiency mediated by CRISPR/Cas9 in mammalian cells. J. Biol. Chem.
simultaneously (Roy et al., 2018). 289, 21312–21324.
In summary, we anticipate that CRISPR screening will
Carlevaro-Fita, J., Lanzós, A., Feuerbach, L., Chen, H., Mas-Ponte, D.,
continue to evolve and improve in the coming years to become PCAWG Driver Identification Working Group, Guigó, R., Pedersen, J.S., and
an indispensable part of the functional cancer genomics toolkit Johnson, R. (2017). Unique genomic features and deeply-conserved functions
and uncover a wealth of new lncRNA drug targets. of long non-coding RNAs in the Cancer LncRNA Census (CLC). bioRxiv,
152769.

ACKNOWLEDGMENTS Chakravarty, D., Sboner, A., Nair, S.S., Giannopoulou, E., Li, R., Hennig, S.,
Mosquera, J.M., Pauwels, J., Park, K., Kossai, M., et al. (2014). The oestrogen
receptor alpha-regulated lncRNA NEAT1 is a critical modulator of prostate
We acknowledge administrative support from Deborah Re and Silvia Roesse- cancer. Nat. Commun. 5, 5383.
let (DBMR). Work in the Johnson laboratory is funded by the Swiss National
Science Foundation through the National Center of Competence in Research Chavez, A., Scheiman, J., Vora, S., Pruitt, B.W., Tuttle, M., P R Iyer, E., Lin, S.,
(NCCR) ‘‘RNA & Disease,’’ the Medical Faculty of the University and University Kiani, S., Guzman, C.D., Wiegand, D.J., et al. (2015). Highly efficient Cas9-
Hospital of Bern, the Helmut Horten Stiftung, and the Swiss Cancer League. mediated transcriptional programming. Nat. Methods 12, 326–328.

Chen, G., Wang, Z., Wang, D., Qiu, C., Liu, M., Chen, X., Zhang, Q., Yan, G.,
REFERENCES
and Cui, Q. (2013). LncRNADisease: a database for long-non-coding RNA-
associated diseases. Nucleic Acids Res. 41 (Database issue), D983–D986.
Abudayyeh, O.O., Gootenberg, J.S., Essletzbichler, P., Han, S., Joung, J., Be-
lanto, J.J., Verdine, V., Cox, D.B.T., Kellner, M.J., Regev, A., et al. (2017). RNA Cho, S.W., Xu, J., Sun, R., Mumbach, M.R., Carter, A.C., Chen, Y.G., Yost,
targeting with CRISPR-Cas13. Nature 550, 280–284. K.E., Kim, J., He, J., Nevins, S.A., et al. (2018). Promoter of lncRNA gene
PVT1 is a tumor-suppressor DNA boundary element. Cell 173, 1398–1412.e22.
Adriaens, C., Standaert, L., Barra, J., Latil, M., Verfaillie, A., Kalev, P., Boeckx,
B., Wijnhoven, P.W., Radaelli, E., Vermi, W., et al. (2016). p53 induces forma- Clemson, C.M., Hutchinson, J.N., Sara, S.A., Ensminger, A.W., Fox, A.H.,
tion of NEAT1 lncRNA-containing paraspeckles that modulate replication Chess, A., and Lawrence, J.B. (2009). An architectural role for a nuclear non-
stress response and chemosensitivity. Nat. Med. 22, 861–868. coding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol.
Cell 33, 717–726.
Aguirre, A.J., Meyers, R.M., Weir, B.A., Vazquez, F., Zhang, C.Z., Ben-David,
U., Cook, A., Ha, G., Harrington, W.F., Doshi, M.B., et al. (2016). Genomic copy Cong, L., Ran, F.A., Cox, D., Lin, S., Barretto, R., Habib, N., Hsu, P.D., Wu, X.,
number dictates a gene-independent cell response to CRISPR/Cas9 targeting. Jiang, W., Marraffini, L.A., et al. (2013). Multiplex genome engineering using
Cancer Discov. 6, 914–929. CRISPR/Cas systems. Science 339, 819–823.

Alexanian, M., Maric, D., Jenkinson, S.P., Mina, M., Friedman, C.E., Ting, C.C., Delas, M.J., Sabin, L.R., Dolzhenko, E., Knott, S.R., Munera Maravilla, E.,
Micheletti, R., Plaisance, I., Nemir, M., Maison, D., et al. (2017). A transcribed Jackson, B.T., Wild, S.A., Kovacevic, T., Stork, E.M., Zhou, M., et al. (2017).
enhancer dictates mesendoderm specification in pluripotency. Nat. Commun. lncRNA requirements for mouse acute myeloid leukemia and normal differen-
8, 1806. tiation. Elife 6, https://doi.org/10.7554/eLife.25607.
Aparicio-Prat, E., Arnan, C., Sala, I., Bosch, N., Guigó, R., and Johnson, R. Derrien, T., Johnson, R., Bussotti, G., Tanzer, A., Djebali, S., Tilgner, H., Guer-
(2015). DECKO: single-oligo, dual-CRISPR deletion of genomic elements nec, G., Martin, D., Merkel, A., Knowles, D.G., et al. (2012). The GENCODE v7
including long non-coding RNAs. BMC Genomics 16, 846. catalog of human long noncoding RNAs: analysis of their gene structure, evo-
lution, and expression. Genome Res. 22, 1775–1789.
Arroyo, J.D., Jourdain, A.A., Calvo, S.E., Ballarano, C.A., Doench, J.G., Root,
D.E., and Mootha, V.K. (2016). A genome-wide CRISPR death screen identifies Dias, N., and Stein, C.A. (2002). Antisense oligonucleotides: basic concepts
genes essential for oxidative phosphorylation. Cell Metab. 24, 875–885. and mechanisms. Mol. Cancer Ther. 1, 347–355.
Arun, G., Diermeier, S.D., and Spector, D.L. (2018). Therapeutic targeting of
DiStefano, J.K. (2018). The emerging role of long noncoding RNAs in human
long non-coding RNAs in cancer. Trends Mol. Med. 24, 257–277.
disease. Methods Mol. Biol. 1706, 91–110.
Bartonicek, N., Maag, J.L.V., and Dinger, M.E. (2016). Long noncoding RNAs
Doench, J.G., Hartenian, E., Graham, D.B., Tothova, Z., Hegde, M., Smith, I.,
in cancer: mechanisms of action and technological advancements. Mol.
Sullender, M., Ebert, B.L., Xavier, R.J., Root, D.E., et al. (2014). Rational design
Cancer 15, 43.
of highly active sgRNAs for CRISPR-Cas9-mediated gene inactivation. Nat.
Bassett, A.R., Akhtar, A., Barlow, D.P., Bird, A.P., Brockdorff, N., Duboule, D., Biotechnol. 32, 1262–1267.
Ephrussi, A., Ferguson-Smith, A.C., Gingeras, T.R., Haerty, W., et al. (2014).
Considerations when investigating lncRNA function in vivo. Elife 3, e03058. Doench, J.G., Fusi, N., Sullender, M., Hegde, M., Vaimberg, E.W., Donovan,
K.F., Smith, I., Tothova, Z., Wilen, C., Orchard, R., et al. (2016). Optimized
Batra, R., Nelles, D.A., Pirie, E., Blue, S.M., Marina, R.J., Wang, H., Chaim, I.A., sgRNA design to maximize activity and minimize off-target effects of
Thomas, J.D., Zhang, N., Nguyen, V., et al. (2017). Elimination of toxic micro- CRISPR-Cas9. Nat. Biotechnol. 34, 184–191.
satellite repeat expansion RNA by RNA-targeting Cas9. Cell 170, 899–
912.e10. Dominguez, A.A., Lim, W.A., and Qi, L.S. (2016). Beyond editing: repurposing
CRISPR-Cas9 for precision genome regulation and interrogation. Nat. Rev.
Bester, A.C., Lee, J.D., Chavez, A., Lee, Y.R., Nachmani, D., Vora, S., Victor, J., Mol. Cell Biol. 17, 5–15.
Sauvageau, M., Monteleone, E., Rinn, J.L., et al. (2018). An integrated
genome-wide CRISPRa approach to functionalize lncRNAs in drug resistance. Du, Z., Fei, T., Verhaak, R.G., Su, Z., Zhang, Y., Brown, M., Chen, Y., and Liu,
Cell 173, 649–664.e20. X.S. (2013). Integrative genomic analyses reveal clinically relevant long non-
coding RNAs in human cancer. Nat. Struct. Mol. Biol. 20, 908–913.
Bhan, A., Soleimani, M., and Mandal, S.S. (2017). Long noncoding RNA and
cancer: a new paradigm. Cancer Res. 77, 3965–3981. ENCODE Project Consortium (2012). An integrated encyclopedia of DNA ele-
ments in the human genome. Nature 489, 57–74.
Buehler, E., Khan, A.A., Marine, S., Rajaram, M., Bahl, A., Burchard, J., and
Ferrer, M. (2012). siRNA off-target effects in genome-wide screens identify Engreitz, J.M., Haines, J.E., Perez, E.M., Munson, G., Chen, J., Kane, M.,
signaling pathway members. Sci. Rep. 2, 428. McDonel, P.E., Guttman, M., and Lander, E.S. (2016). Local regulation of
gene expression by lncRNA promoters, transcription and splicing. Nature
Cabili, M.N., Dunagin, M.C., McClanahan, P.D., Biaesch, A., Padovan-Merhar, 539, 452–455.
O., Regev, A., Rinn, J.L., and Raj, A. (2015). Localization and abundance anal-
ysis of human lncRNAs at single-cell and single-molecule resolution. Genome Ernst, J., and Kellis, M. (2017). Chromatin-state discovery and genome anno-
Biol. 16, 20. tation with ChromHMM. Nat. Protoc. 12, 2478–2492.

10 Cancer Cell 35, April 15, 2019


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review
Fang, Y., and Fullwood, M.J. (2016). Roles, functions, and mechanisms of long Holdt, L.M., Stahringer, A., Sass, K., Pichler, G., Kulak, N.A., Wilfert, W., Kohl-
non-coding RNAs in cancer. Genomics Proteomics Bioinformatics 14, 42–54. maier, A., Herbst, A., Northoff, B.H., Nicolaou, A., et al. (2016). Circular non-
coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis
Feng, W., Li, L., Xu, X., Jiao, Y., and Du, W. (2017). Up-regulation of the long in humans. Nat. Commun. 7, 12429.
non-coding RNA RMRP contributes to glioma progression and promotes gli-
oma cell proliferation and invasion. Arch. Med. Sci. 13, 1315–1321. Hong, J., and Gresham, D. (2017). Incorporation of unique molecular identifiers
in TruSeq adapters improves the accuracy of quantitative sequencing.
FANTOM Consortium and the RIKEN PMI and CLST (DGT), Forrest, A.R., Ka- BioTechniques 63, 221–226.
waji, H., Rehli, M., Baillie, J.K., de Hoon, M.J., Haberle, V., Lassmann, T., Ku-
lakovskiy, I.V., Lizio, M., et al. (2014). A promoter-level mammalian expression Horlbeck, M.A., Gilbert, L.A., Villalta, J.E., Adamson, B., Pak, R.A., Chen, Y.,
atlas. Nature 507, 462–470. Fields, A.P., Park, C.Y., Corn, J.E., Kampmann, M., et al. (2016a). Compact
and highly active next-generation libraries for CRISPR-mediated gene repres-
Futreal, P.A., Coin, L., Marshall, M., Down, T., Hubbard, T., Wooster, R., Rah- sion and activation. Elife 5, https://doi.org/10.7554/eLife.19760.
man, N., and Stratton, M.R. (2004). A census of human cancer genes. Nat. Rev.
Cancer 4, 177–183. Horlbeck, M.A., Witkowsky, L.B., Guglielmi, B., Replogle, J.M., Gilbert, L.A.,
Villalta, J.E., Torigoe, S.E., Tjian, R., and Weissman, J.S. (2016b). Nucleo-
Gilbert, L.A., Larson, M.H., Morsut, L., Liu, Z., Brar, G.A., Torres, S.E., Stern- somes impede Cas9 access to DNA in vivo and in vitro. Elife 5, https://doi.
Ginossar, N., Brandman, O., Whitehead, E.H., Doudna, J.A., et al. (2013). org/10.7554/eLife.12677.
CRISPR-mediated modular RNA-guided regulation of transcription in eukary-
otes. Cell 154, 442–451. Hosono, Y., Niknafs, Y.S., Prensner, J.R., Iyer, M.K., Dhanasekaran, S.M.,
Mehra, R., Pitchiaya, S., Tien, J., Escara-Wilke, J., Poliakov, A., et al. (2017).
Goyal, A., Myacheva, K., Groß, M., Klingenberg, M., Duran Arqué, B., and Oncogenic role of THOR, a conserved cancer/testis long non-coding RNA.
Diederichs, S. (2017). Challenges of CRISPR/Cas9 applications for long non- Cell 171, 1559–1572.e20.
coding RNA genes. Nucleic Acids Res. 45, e12.
Hsu, P.D., Scott, D.A., Weinstein, J.A., Ran, F.A., Konermann, S., Agarwala, V.,
Graham, D.B., and Root, D.E. (2015). Resources for the design of CRISPR Li, Y., Fine, E.J., Wu, X., Shalem, O., et al. (2013). DNA targeting specificity of
gene editing experiments. Genome Biol. 16, 260. RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832.

Groff, A.F., Larson, M.H., Morsut, L., Liu, Z., Brar, G.A., Torres, S.E., Stern-Gi- Huarte, M. (2015). The emerging role of lncRNAs in cancer. Nat. Med. 21,
nossar, N., Brandman, O., Whitehead, E.H., Doudna, J.A., et al. (2016). In vivo 1253–1261.
characterization of linc-p21 reveals functional cis-regulatory DNA elements.
Cell Rep. 16, 2178–2186. Hung, T., Wang, Y., Lin, M.F., Koegel, A.K., Kotake, Y., Grant, G.D., Horlings,
H.M., Shah, N., Umbricht, C., Wang, P., et al. (2011). Extensive and coordi-
Gupta, R.A., Shah, N., Wang, K.C., Kim, J., Horlings, H.M., Wong, D.J., Tsai, nated transcription of noncoding RNAs within cell-cycle promoters. Nat.
M.C., Hung, T., Argani, P., Rinn, J.L., et al. (2010). Long non-coding RNA Genet. 43, 621–629.
HOTAIR reprograms chromatin state to promote cancer metastasis. Nature
464, 1071–1076. Iyer, M.K., Niknafs, Y.S., Malik, R., Singhal, U., Sahu, A., Hosono, Y., Barrette,
T.R., Prensner, J.R., Evans, J.R., Zhao, S., et al. (2015). The landscape of long
€mmerle, M., Eissmann, M., Hsu, J., Kim, Y., Hung, G., Re-
Gutschner, T., Ha noncoding RNAs in the human transcriptome. Nat. Genet. 47, 199–208.
venko, A., Arun, G., Stentrup, M., Gross, M., et al. (2013). The noncoding
RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J.A., and Charpentier,
cells. Cancer Res. 73, 1180–1189. E. (2012). A programmable dual-RNA-guided DNA endonuclease in adaptive
bacterial immunity. Science 337, 816–821.
Gutschner, T., Baas, M., and Diederichs, S. (2011). Noncoding RNA gene
silencing through genomic integration of RNA destabilizing elements using Johnson, R., and Guigó, R. (2014). The RIDL hypothesis: transposable ele-
zinc finger nucleases. Genome Res. 21, 1944–1954. ments as functional domains of long noncoding RNAs. RNA 20, 959–976.

Gutschner, T., and Diederichs, S. (2012). The hallmarks of cancer: a long non- Joung, J., Engreitz, J.M., Konermann, S., Abudayyeh, O.O., Verdine, V.K.,
coding RNA point of view. RNA Biol. 9, 703–719. Aguet, F., Gootenberg, J.S., Sanjana, N.E., Wright, J.B., Fulco, C.P., et al.
(2017a). Genome-scale activation screen identifies a lncRNA locus regulating
Guttman, M., and Rinn, J.L. (2012). Modular regulatory principles of large non- a gene neighbourhood. Nature 548, 343–346.
coding RNAs. Nature 482, 339–346.
Joung, J., Konermann, S., Gootenberg, J.S., Abudayyehv, O.O., Platt, R.J.,
Haapaniemi, E., Botla, S., Persson, J., Schmierer, B., and Taipale, J. (2018). Brigham, M.D., Sanjana, N.E., and Zhang, F. (2017b). Genome-scale
CRISPR-Cas9 genome editing induces a p53-mediated DNA damage CRISPR-Cas9 knockout and transcriptional activation screening. Nat. Protoc.
response. Nat. Med. 24, 927–930. 12, 828–863.

Haeussler, M., Schönig, K., Eckert, H., Eschstruth, A., Mianné, J., Renaud, Juliano, R.L. (2016). The delivery of therapeutic oligonucleotides. Nucleic
J.B., Schneider-Maunoury, S., Shkumatava, A., Teboul, L., Kent, J., et al. Acids Res. 44, 6518–6548.
(2016). Evaluation of off-target and on-target scoring algorithms and integra-
tion into the guide RNA selection tool CRISPOR. Genome Biol. 17, 148. Khurana, E., Fu, Y., Chakravarty, D., Demichelis, F., Rubin, M.A., and Gerstein,
M. (2016). Role of non-coding sequence variants in cancer. Nat. Rev. Genet.
Han, J., Zhang, J., Chen, L., Shen, B., Zhou, J., Hu, B., Du, Y., Tate, P.H., 17, 93–108.
Huang, X., Zhang, W., et al. (2014). Efficient in vivo deletion of a large imprinted
lncRNA by CRISPR/Cas9. RNA Biol. 11, 829–835. Kim, J., Piao, H.L., Kim, B.J., Yao, F., Han, Z., Wang, Y., Xiao, Z., Siverly, A.N.,
Lawhon, S.E., Ton, B.N., et al. (2018). Long noncoding RNA MALAT1 sup-
Hanahan, D., and Weinberg, R.A. (2000). The hallmarks of cancer. Cell presses breast cancer metastasis. Nat. Genet. 50, 1705–1715.
100, 57–70.
Koirala, P., Huang, J., Ho, T.T., Wu, F., Ding, X., and Mo, Y.Y. (2017). LncRNA
Hanahan, D., and Weinberg, R.A. (2011). Hallmarks of cancer: the next gener- AK023948 is a positive regulator of AKT. Nat. Commun. 8, 14422.
ation. Cell 144, 646–674.
Konermann, S., Brigham, M.D., Trevino, A.E., Joung, J., Abudayyeh, O.O.,
Hart, T., and Moffat, J. (2016). BAGEL: a computational framework for identi- Barcena, C., Hsu, P.D., Habib, N., Gootenberg, J.S., Nishimasu, H., et al.
fying essential genes from pooled library screens. BMC Bioinformatics (2015). Genome-scale transcriptional activation by an engineered CRISPR-
17, 164. Cas9 complex. Nature 517, 583–588.

Ho, T.-T., Zhou, N., Huang, J., Koirala, P., Xu, M., Fung, R., Wu, F., and Mo, Kopp, F., and Mendell, J.T. (2018). Functional classification and experimental
Y.Y. (2015). Targeting non-coding RNAs with the CRISPR/Cas9 system in hu- dissection of long noncoding RNAs. Cell 172, 393–407.
man cell lines. Nucleic Acids Res. 43, e17.
Lagarde, J., Uszczynska-Ratajczak, B., Carbonell, S., Pérez-Lluch, S., Abad,
Hodgkins, A., Farne, A., Perera, S., Grego, T., Parry-Smith, D.J., Skarnes, A., Davis, C., Gingeras, T.R., Frankish, A., Harrow, J., Guigo, R., et al.
W.C., and Iyer, V. (2015). WGE: a CRISPR database for genome engineering. (2017). High-throughput annotation of full-length long noncoding RNAs with
Bioinformatics 31, 3078–3080. capture long-read sequencing. Nat. Genet. 49, 1731–1740.

Cancer Cell 35, April 15, 2019 11


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review
Lahens, N.F., Kavakli, I.H., Zhang, R., Hayer, K., Black, M.B., Dueck, H., Pi- is a p53-inducible lincRNA essential for transformation suppression. Genes
zarro, A., Kim, J., Irizarry, R., Thomas, R.S., et al. (2014). IVT-seq reveals Dev. 31, 1095–1108.
extreme bias in RNA sequencing. Genome Biol. 15, R86.
Meng, Q., Ren, M., Li, Y., and Song, X. (2016). LncRNA-RMRP acts as an
Lanzós, A., Carlevaro-Fita, J., Mularoni, L., Reverter, F., Palumbo, E., Guigó, oncogene in lung cancer. PLoS One 11, e0164845.
R., and Johnson, R. (2017). Discovery of cancer driver long noncoding RNAs
across 1112 tumour genomes: new candidates and distinguishing features. Michlits, G., Hubmann, M., Wu, S.H., Vainorius, G., Budusan, E., Zhuk, S.,
Sci. Rep. 7, 41544. Burkard, T.R., Novatchkova, M., Aichinger, M., Lu, Y., et al. (2017). CRISPR-
UMI: single-cell lineage tracing of pooled CRISPR-Cas9 screens. Nat.
Latos, P.A., Pauler, F.M., Koerner, M.V., Sxenergin, H.B., Hudson, Q.J., Stoc- Methods 14, 1191–1197.
sits, R.R., Allhoff, W., Stricker, S.H., Klement, R.M., Warczok, K.E., et al.
(2012). Airn transcriptional overlap, but not its lncRNA products, induces im- Moreno-Mateos, M.A., Vejnar, C.E., Beaudoin, J.D., Fernandez, J.P., Mis, E.K.,
printed Igf2r silencing. Science 338, 1469–1472. Khokha, M.K., and Giraldez, A.J. (2015). CRISPRscan: designing highly effi-
cient sgRNAs for CRISPR-Cas9 targeting in vivo. Nat. Methods 12, 982–988.
Lee, S., Kopp, F., Chang, T.C., Sataluri, A., Chen, B., Sivakumar, S., Yu, H., Xie,
Y., and Mendell, J.T. (2016). Noncoding RNA NORAD regulates genomic sta- Mularoni, L., Sabarinathan, R., Deu-Pons, J., Gonzalez-Perez, A., and López-
bility by sequestering PUMILIO proteins. Cell 164, 69–80. Bigas, N. (2016). OncodriveFML: a general framework to identify coding and
non-coding regions with cancer driver mutations. Genome Biol. 17, 128.
Leucci, E., Vendramin, R., Spinazzi, M., Laurette, P., Fiers, M., Wouters, J., Ra-
daelli, E., Eyckerman, S., Leonelli, C., Vanderheyden, K., et al. (2016). Mela- Munoz, D.M., Cassiani, P.J., Li, L., Billy, E., Korn, J.M., Jones, M.D., Golji, J.,
noma addiction to the long non-coding RNA SAMMSON. Nature 531, Ruddy, D.A., Yu, K., McAllister, G., et al. (2016). CRISPR screens provide a
518–522. comprehensive assessment of cancer vulnerabilities but generate false-posi-
tive hits for highly amplified genomic regions. Cancer Discov. 6, 900–913.
Li, H., Zhang, Y., Ströse, A., Tedesco, D., Gurova, K., and Selivanova, G.
(2014a). Integrated high-throughput analysis identifies Sp1 as a crucial deter- Nelles, D.A., Fang, M.Y., O’Connell, M.R., Xu, J.L., Markmiller, S.J., Doudna,
minant of p53-mediated apoptosis. Cell Death Differ. 21, 1493–1502. J.A., and Yeo, G.W. (2016). Programmable RNA tracking in live cells with
CRISPR/Cas9. Cell 165, 488–496.
Li, W., Xu, H., Xiao, T., Cong, L., Love, M.I., Zhang, F., Irizarry, R.A., Liu, J.S.,
Brown, M., Liu, X.S., et al. (2014b). MAGeCK enables robust identification Nellore, A., Jaffe, A.E., Fortin, J.P., Alquicira-Hernández, J., Collado-Torres, L.,
of essential genes from genome-scale CRISPR/Cas9 knockout screens. Wang, S., Phillips, R.A., III, Karbhari, N., Hansen, K.D., Langmead, B., et al.
Genome Biol. 15, 554. (2016). Human splicing diversity and the extent of unannotated splice junctions
across human RNA-seq samples on the Sequence Read Archive. Genome
Lin, A., Hu, Q., Li, C., Xing, Z., Ma, G., Wang, C., Li, J., Ye, Y., Yao, J., Liang, K., Biol. 17, 266.
et al. (2017). The LINK-A lncRNA interacts with PtdIns(3,4,5)P3 to hyperacti-
vate AKT and confer resistance to AKT inhibitors. Nat. Cell Biol. 19, 238–251. Ning, S., Zhang, J., Wang, P., Zhi, H., Wang, J., Liu, Y., Gao, Y., Guo, M., Yue,
M., Wang, L., et al. (2016). Lnc2Cancer: a manually curated database of exper-
Lin, C., and Yang, L. (2018). Long noncoding RNA in cancer: wiring signaling imentally supported lncRNAs associated with various human cancers. Nucleic
circuitry. Trends Cell Biol. 28, 287–301. Acids Res. 44, D980–D985.

Liu, S.J., Horlbeck, M.A., Cho, S.W., Birk, H.S., Malatesta, M., He, D., Atte- O’Connell, M.R., Oakes, B.L., Sternberg, S.H., East-Seletsky, A., Kaplan, M.,
nello, F.J., Villalta, J.E., Cho, M., Chen, Y., et al. (2017). CRISPRi-based and Doudna, J.A. (2014). Programmable RNA recognition and cleavage by
genome-scale identification of functional long noncoding RNA loci in human CRISPR/Cas9. Nature 516, 263–266.
cells. Science 355, https://doi.org/10.1126/science.aah7111.
Pandey, G.K., Mitra, S., Subhash, S., Hertwig, F., Kanduri, M., Mishra, K., Frans-
Liu, Y., Cao, Z., Wang, Y., Guo, Y., Xu, P., Yuan, P., Liu, Z., He, Y., and Wei, W. son, S., Ganeshram, A., Mondal, T., Bandaru, S., et al. (2014). The risk-associ-
(2018). Genome-wide screening for functional long noncoding RNAs in human ated long noncoding RNA NBAT-1 controls neuroblastoma progression by regu-
cells by Cas9 targeting of splice sites. Nat. Biotechnol. 36, 1203–1210. lating cell proliferation and neuronal differentiation. Cancer Cell 26, 722–737.

Maamar, H., Cabili, M.N., Rinn, J., and Raj, A. (2013). linc-HOXA1 is a noncod- Panzitt, K., Tschernatsch, M.M., Guelly, C., Moustafa, T., Stradner, M., Stroh-
ing RNA that represses HoXa1 transcription in cis. Genes Dev. 27, 1260–1271. maier, H.M., Buck, C.R., Denk, H., Schroeder, R., Trauner, M., et al. (2007).
Characterization of HULC, a novel gene with striking up-regulation in hepato-
Mali, P., Yang, L., Esvelt, K.M., Aach, J., Guell, M., DiCarlo, J.E., Norville, J.E., cellular carcinoma, as noncoding RNA. Gastroenterology 132, 330–342.
and Church, G.M. (2013). RNA-guided human genome engineering via Cas9.
Science 339, 823–826. Parnas, O., Jovanovic, M., Eisenhaure, T.M., Herbst, R.H., Dixit, A., Ye, C.J.,
Przybylski, D., Platt, R.J., Tirosh, I., Sanjana, N.E., Shalem, O., et al. (2015).
Mandegar, M.A., Huebsch, N., Frolov, E.B., Shin, E., Truong, A., Olvera, M.P., A genome-wide CRISPR screen in primary immune cells to dissect regulatory
Chan, A.H., Miyaoka, Y., Holmes, K., Spencer, C.I., Judge, L.M., et al. (2016). networks. Cell 162, 675–686.
CRISPR interference efficiently induces specific and reversible gene silencing
in human iPSCs. Cell Stem Cell 18, 541–553. Peretz, L., Besser, E., Hajbi, R., Casden, N., Ziv, D., Kronenberg, N., Gigi, L.B.,
Sweetat, S., Khawaled, S., Aqeilan, R., et al. (2018). Combined shRNA over
Marin-Bejar, O., Marchese, F.P., Athie, A., Sánchez, Y., González, J., Segura, CRISPR/cas9 as a methodology to detect off-target effects and a potential
V., Huang, L., Moreno, I., Navarro, A., Monzó, M., Garcı́a-Foncillas, J., et al. compensatory mechanism. Sci. Rep. 8, 93.
(2013). Pint lincRNA connects the p53 pathway with epigenetic silencing by
the Polycomb repressive complex 2. Genome Biol. 14, R104. Perez-Pinera, P., Kocak, D.D., Vockley, C.M., Adler, A.F., Kabadi, A.M., Pol-
stein, L.R., Thakore, P.I., Glass, K.A., Ousterout, D.G., Leong, K.W., et al.
Marin-Bejar, O., Mas, A.M., González, J., Martinez, D., Athie, A., Morales, X., (2013). RNA-guided gene activation by CRISPR-Cas9-based transcription
Galduroz, M., Raimondi, I., Grossi, E., Guo, S., Rouzaut, A., et al. (2017). The factors. Nat. Methods 10, 973–976.
human lncRNA LINC-PINT inhibits tumor cell invasion through a highly
conserved sequence element. Genome Biol. 18, 202. Pickard, M.R., and Williams, G.T. (2015). Molecular and cellular mechanisms of
action of tumour suppressor GAS5 LncRNA. Genes 6, 484–499.
Matano, M., Date, S., Shimokawa, M., Takano, A., Fujii, M., Ohta, Y., Wata-
nabe, T., Kanai, T., and Sato, T. (2015). Modeling colorectal cancer using Portoso, M., Ragazzini, R., Brencic  Moiani, A., Michaud, A., Vassilev, I., Was-
, Z.,
CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat. sef, M., Servant, N., Sargueil, B., Margueron, R., et al. (2017). PRC2 is dispens-
Med. 21, 256–262. able for HOTAIR-mediated transcriptional repression. EMBO J. 36, 981–994.

McDonald, E.R., 3rd, de Weck, A., Schlabach, M.R., Billy, E., Mavrakis, K.J., Pulido-Quetglas, C., Aparicio-Prat, E., Arnan, C., Polidori, T., Hermoso, T., Pal-
Hoffman, G.R., Belur, D., Castelletti, D., Frias, E., Gampa, K., et al. (2017). Proj- umbo, E., Ponomarenko, J., Guigo, R., and Johnson, R. (2017). Scalable
ect DRIVE: a compendium of cancer dependencies and synthetic lethal rela- design of paired crispr guide RNAs for genomic deletion. PLoS Comput.
tionships uncovered by large-scale, deep RNAi screening. Cell 170, 577– Biol. 13, e1005341.
592.e10.
Qi, L.S., Larson, M.H., Gilbert, L.A., Doudna, J.A., Weissman, J.S., Arkin, A.P.,
Mello, S.S., Sinow, C., Raj, N., Mazur, P.K., Bieging-Rolett, K., Broz, D.K., and Lim, W.A. (2013). Repurposing CRISPR as an RNA-guided platform for
Imam, J.F.C., Vogel, H., Wood, L.D., Sage, J., Hirose, T., et al. (2017). Neat1 sequence-specific control of gene expression. Cell 152, 1173–1183.

12 Cancer Cell 35, April 15, 2019


Please cite this article in press as: Esposito et al., Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using
CRISPR-Cas9 Screening, Cancer Cell (2019), https://doi.org/10.1016/j.ccell.2019.01.019

Cancer Cell

Review
Quek, X.C., Thomson, D.W., Maag, J.L., Bartonicek, N., Signal, B., Clark, M.B., Stojic, L., Lun, A.T.L., Mangei, J., Mascalchi, P., Quarantotti, V., Barr, A.R., Ba-
Gloss, B.S., and Dinger, M.E. (2015). lncRNAdb v2.0: expanding the reference kal, C., Marioni, J.C., Gergely, F., Odom, D.T., et al. (2018). Specificity of RNAi,
database for functional long noncoding RNAs. Nucleic Acids Res. 43, D168–D173. LNA and CRISPRi as loss-of-function methods in transcriptional analysis. Nu-
cleic Acids Res. 46, 5950–5966.
Quinodoz, S., and Guttman, M. (2014). Long noncoding RNAs: an emerging
link between gene regulation and nuclear organization. Trends Cell Biol. 24, Sun, R., Qin, C., Jiang, B., Fang, S., Pan, X., Peng, L., Liu, Z., Li, W., Li, Y., Li,
651–663. G., et al. (2016). Down-regulation of MALAT1 inhibits cervical cancer cell inva-
sion and metastasis by inhibition of epithelial-mesenchymal transition. Mol.
Radzisheuskaya, A., Shlyueva, D., Mu €ller, I., and Helin, K. (2016). Optimizing Biosyst. 12, 952–962.
sgRNA position markedly improves the efficiency of CRISPR/dCas9-mediated
transcriptional repression. Nucleic Acids Res. 44, e141. Tanenbaum, M.E., Gilbert, L.A., Qi, L.S., Weissman, J.S., and Vale, R.D.
(2014). A protein-tagging system for signal amplification in gene expression
Rheinbay, E., Nielsen, M.M., Abascal, F., Tiao, G., Hornshøj, H., Hess, J.M., and fluorescence imaging. Cell 159, 635–646.
Pedersen, R.I.I., Feuerbach, L., Sabarinathan, R., Madsen, H.T., et al. (2017).
Discovery and characterization of coding and non-coding driver mutations in Tang, Q., and Hann, S.S. (2018). HOTAIR: an oncogenic long non-coding RNA
more than 2,500 whole cancer genomes. bioRxiv, 237313. in human cancer. Cell Physiol. Biochem. 47, 893–913.
Rosenbluh, J., Xu, H., Harrington, W., Gill, S., Wang, X., Vazquez, F., Root, Trumbach, D., Pfeiffer, S., Poppe, M., Scherb, H., Doll, S., Wurst, W., and
D.E., Tsherniak, A., and Hahn, W.C. (2017). Complementary information Schick, J.A. (2017). ENCoRE: an efficient software for CRISPR screens iden-
derived from CRISPR Cas9 mediated gene deletion and suppression. Nat. tifies new players in extrinsic apoptosis. BMC Genomics 18, 905.
Commun. 8, 15403.
Tseng, Y.-Y., Moriarity, B.S., Gong, W., Akiyama, R., Tiwari, A., Kawakami, H.,
Ross, S.J., Revenko, A.S., Hanson, L.L., Ellston, R., Staniszewska, A., Ronning, P., Reuland, B., Guenther, K., Beadnell, T.C., et al. (2014). PVT1
Whalley, N., Pandey, S.K., Revill, M., Rooney, C., Buckett, L.K., et al. (2017). dependence in cancer with MYC copy-number increase. Nature 512, 82–86.
Targeting KRAS-dependent tumors with AZD4785, a high-affinity therapeutic
antisense oligonucleotide inhibitor of KRAS. Sci. Transl Med. 9, https://doi. Ulitsky, I., Shkumatava, A., Jan, C.H., Sive, H., and Bartel, D.P. (2011).
org/10.1126/scitranslmed.aal5253. Conserved function of lincRNAs in vertebrate embryonic development despite
rapid sequence evolution. Cell 147, 1537–1550.
Roy, K.R., Smith, J.D., Vonesch, S.C., Lin, G., Tu, C.S., Lederer, A.R., Chu, A.,
Suresh, S., Nguyen, M., Horecka, J., et al. (2018). Multiplexed precision Ulitsky, I., and Bartel, D.P. (2013). XLincRNAs: genomics, evolution, and
genome editing with trackable genomic barcodes in yeast. Nat. Biotechnol. mechanisms. Cell 154, 26–46.
36, 512–520.
Uszczynska-Ratajczak, B., Lagarde, J., Frankish, A., Guigó, R., and Johnson,
Sanjana, N.E., Shalem, O., and Zhang, F. (2014). Improved vectors and R. (2018). Towards a complete map of the human long non-coding RNA tran-
genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784. scriptome. Nat. Rev. Genet. 19, 535–548.
Sanson, K.R., Hanna, R.E., Hegde, M., Donovan, K.F., Strand, C., Sullender,
Vance, K.W., and Ponting, C.P. (2014). Transcriptional regulatory functions of
M.E., Vaimberg, E.W., Goodale, A., Root, D.E., Piccioni, F., et al. (2018). Up,
nuclear long noncoding RNAs. Trends Genet. 30, 348–355.
down, and out: optimized libraries for CRISPRa, CRISPRi, and CRISPR-
knockout genetic screens. bioRxiv, 356626. Vidigal, J.A., and Ventura, A. (2015). Rapid and efficient one-step generation of
paired gRNA CRISPR-Cas9 libraries. Nat. Commun. 6, 8083.
Scharner, J., Rigo, F., Bennett, C.F., and Krainer, A.R. (2018). Delivery of Gal-
NAc-conjugated splice-switching ASOs to non-hepatic cells through ectopic
Watts, J.K., and Corey, D.R. (2012). Silencing disease genes in the laboratory
expression of asialoglycoprotein receptor. bioRxiv, 382606. and the clinic. J. Pathol. 226, 365–379.
Seiler, J., Breinig, M., Caudron-Herger, M., Polycarpou-Schwarz, M., Boutros,
West, J.A., Davis, C.P., Sunwoo, H., Simon, M.D., Sadreyev, R.I., Wang, P.I.,
M., and Diederichs, S. (2017). The lncRNA VELUCT strongly regulates viability
Tolstorukov, M.Y., and Kingston, R.E. (2014). The long noncoding RNAs
of lung cancer cells despite its extremely low abundance. Nucleic Acids Res.
NEAT1 and MALAT1 bind active chromatin sites. Mol. Cell 55, 791–802.
45, 5458–5469.
Williams, S.P., Barthorpe, A.S., Lightfoot, H., Garnett, M.J., and McDermott, U.
Seo, M., Lee, S., Kim, J.H., Lee, W.H., Hu, G., Elledge, S.J., and Suk, K. (2014).
(2017). High-throughput RNAi screen for essential genes and drug synergistic
RNAi-based functional selection identifies novel cell migration determinants
dependent on PI3K and AKT pathways. Nat. Commun. 5, 5217. combinations in colorectal cancer. Sci. Data 4, 170139.

Shalem, O., Sanjana, N.E., Hartenian, E., Shi, X., Scott, D.A., Mikkelson, T., Wutz, A., Rasmussen, T.P., and Jaenisch, R. (2002). Chromosomal silencing
Heckl, D., Ebert, B.L., Root, D.E., Doench, J.G., et al. (2014). Genome-scale and localization are mediated by different domains of Xist RNA. Nat. Genet.
CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87. 30, 167–174.

Shao, Y., Shao, Y., Ye, M., Li, Q., Sun, W., Ye, G., Zhang, X., Yang, Y., Xiao, B., Xing, Y.-H., Yao, R.W., Zhang, Y., Guo, C.J., Jiang, S., Xu, G., Dong, R., Yang,
Guo, J., et al. (2016). LncRNA-RMRP promotes carcinogenesis by acting as a L., and Chen, L.L. (2017). SLERT regulates DDX21 rings associated with Pol I
miR-206 sponge and is used as a novel biomarker for gastric cancer. Oncotar- transcription. Cell 169, 664–678.e16.
get 7, 37812–37824.
Xu, H., Xiao, T., Chen, C.H., Li, W., Meyer, C.A., Wu, Q., Wu, D., Cong, L.,
Shtivelman, E., and Bishop, J.M. (1989). The PVT gene frequently amplifies Zhang, F., Liu, J.S., et al. (2015). Sequence determinants of improved CRISPR
with MYC in tumor cells. Mol. Cell. Biol. 9, 1148–1154. sgRNA design. Genome Res. 25, 1147–1157.

Smith, I., Greenside, P.G., Natoli, T., Lahr, D.L., Wadden, D., Tirosh, I., Nar- Yan, X., Hu, Z., Feng, Y., Hu, X., Yuan, J., Zhao, S.D., Zhang, Y., Yang, L.,
ayan, R., Root, D.E., Golub, T.R., Subramanian, A., et al. (2017a). Evaluation Shan, W., He, Q., et al. (2015). Comprehensive genomic characterization of
of RNAi and CRISPR technologies by large-scale gene expression profiling long non-coding RNAs across human cancers. Cancer Cell 28, 529–540.
in the connectivity map. PLoS Biol. 15, e2003213.
Yap, K.L., Li, S., Muñoz-Cabello, A.M., Raguz, S., Zeng, L., Mujtaba, S., Gil, J.,
Smith, T., Heger, A., and Sudbery, I. (2017b). UMI-tools: modeling sequencing Walsh, M.J., and Zhou, M.M. (2010). Molecular interplay of the noncoding RNA
errors in Unique Molecular Identifiers to improve quantification accuracy. ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcrip-
Genome Res. 27, 491–499. tional silencing of INK4a. Mol. Cell 38, 662–674.

Smolen, G.A., Zhang, J., Zubrowski, M.J., Edelman, E.J., Luo, B., Yu, M., Ng, Zhou, Y., Zhang, X., and Klibanski, A. (2012). MEG3 noncoding RNA: a tumor
L.W., Scherber, C.M., Schott, B.J., Ramaswamy, S., et al. (2010). A genome- suppressor. J. Mol. Endocrinol. 48, R45–R53.
wide RNAi screen identifies multiple RSK-dependent regulators of cell migra-
tion. Genes Dev. 24, 2654–2665. Zhu, S., Li, W., Liu, J., Chen, C.H., Liao, Q., Xu, P., Xu, H., Xiao, T., Cao, Z.,
Peng, J., et al. (2016). Genome-scale deletion screening of human long non-
Song, Y., Lai, L., and Li, Z. (2017). Large-scale genomic deletions mediated by coding RNAs using a paired-guide RNA CRISPR-Cas9 library. Nat. Biotechnol.
CRISPR/Cas9 system. Oncotarget 8, 5647. 34, 1279–1286.

Cancer Cell 35, April 15, 2019 13

Vous aimerez peut-être aussi