You are on page 1of 31

HHS Public Access

Author manuscript
Br J Haematol. Author manuscript; available in PMC 2018 April 06.
Author Manuscript

Published in final edited form as:


Br J Haematol. 2016 October ; 175(2): 208–225. doi:10.1111/bjh.14297.

CRISPR-Cas9 technology and its application in haematological


disorders
Han Zhang and Nami McCarty
The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases
(IMM), University of Texas-Health Science Centre at Houston, Houston, TX, USA
Author Manuscript

Summary
The recent advent of the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-
CRISPR associated protein 9 (Cas9) system for precise genome editing has revolutionized
methodologies in haematology and oncology studies. CRISPR-Cas9 technology can be used to
remove and correct genes or mutations, and to introduce site-specific therapeutic genes in human
cells. Inherited haematological disorders represent ideal targets for CRISPR-Cas9-mediated gene
therapy. Correcting disease-causing mutations could alleviate disease-related symptoms in the near
future. The CRISPR-Cas9 system is also a useful tool for delineating molecular mechanisms
involving haematological malignancies. Prior to the use of CRISPR-Cas9-mediated gene
correction in humans, appropriate delivery systems with higher efficiency and specificity must be
identified, and ethical guidelines for applying the technology with controllable safety must be
established. Here, the latest applications of CRISPR-Cas9 technology in haematological disorders,
Author Manuscript

current challenges and future directions are reviewed and discussed.

Keywords
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-associated protein 9 (Cas9)
system; haematological disorders; gene editing; haematological malignancies

Introduction
Over the past two decades, advances in molecular biology and genetics have greatly
extended our knowledge of haemaological diseases. However, the characterization of genes
of interest remains a great challenge. The optimal way to unravel the roles of particular
Author Manuscript

genes is to manipulate their functions via gene deletion, insertion or modification. Earlier
genome editing technologies, such as homologous recombination (HR), have facilitated the
manipulation of genes by generating knockout and knock-in cells or mouse models (Thomas
et al, 1986; Mansour et al, 1988; Capecchi, 1989a, b). However HR-mediated targeting is a
less efficient, time-consuming and labour-intensive process, rendering its widespread

Correspondence: Nami McCarty, PhD, University of Texas-Health Science Centre at Houston, 1825 Pressler St., IMM-630H,
Houston, TX 77030, USA. nami.mccarty@uth.tmc.edu.
Author contributions
Han Zhang reviewed the literature, wrote the paper and made corrections. Nami McCarty designed the review, wrote the paper and
made corrections.
Zhang and McCarty Page 2

application difficult. In subsequent decades, targeting of DNA double-stranded breaks


Author Manuscript

(DSBs) using engineered nucleases, such as zinc-finger nucleases (ZFNs) and transcription
activator-like effector nucleases (TALENs), was introduced to stimulate cellular DNA repair
pathways: non-homologous end-joining (NHEJ) and homology-directed repair (HDR)
(Rudin et al, 1989; Rouet et al, 1994; Choulika et al, 1995; Bibikova et al, 2002; Moscou &
Bogdanove, 2009). In most cases, NHEJ-mediated error-prone DNA repair generates
random insertions or deletions (indels), leading to frameshift mutations. If an exogenous
donor DNA template is available surrounding the DSB, then the DNA damage can be
repaired via an HDR pathway, which leads to precise gene modifications or gene insertions.
Although ZFNs and TALENs have resulted in more efficient and applicable site-specific
genome engineering (Joung & Sander, 2013; Urnov, 2014), the expansion of their
applications has also been hampered by complex procedures, low specificity and high cost
(Peng et al, 2015; Sanchez-Rivera & Jacks, 2015).
Author Manuscript

The most recent development, the Clustered Regularly Interspaced Short Palindromic
Repeats (CRISPR)-CRISPR associated protein 9 (Cas9) technique, has initiated a new era of
genome editing by overcoming the limitations of these earlier methods (Cong et al, 2013;
Hsu et al, 2014; Niu et al, 2014; Konermann et al, 2015). During the past 4 years, the
CRISPR-Cas9 system has been overwhelmingly adopted as a powerful tool in biological
research, ranging from the modification of plants to the creation of animal models and the
alleviation of human diseases including haematological diseases.

Development of the CRISPR-Cas9 system


CRISPR was first described in Escherichia coli as an odd 29-nucleotide repeat sequence
with 32-nucleotide spacing by a Japanese research group in 1987, although no further
Author Manuscript

detailed observation was reported (Ishino et al, 1987). Several years later, Francisco Mojica
discovered similar repeats in several microbial species and reported their potential use in
prokaryotes (Mojica et al, 1993, 1995). By 2002, the novel repeats were formally named as
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR); meanwhile,
scientists discovered specific CRISPR-associated (Cas) genes that were closely related to the
function of CRISPR (Jansen et al, 2002). In 2003, Mojica discovered the function of the
CRISPR system, proposing that CRISPR is an adaptive immune system that protects
microbes against specific infections (Mojica et al, 2005). Two independent groups (Bolotin
et al, 2005; Pourcel et al, 2005) reported similar conclusions, and their finding was
confirmed by experiments showing that CRISPR provides acquired immunity against viruses
in prokaryotes (Barrangou et al, 2007).

Following these discoveries, more researchers adopted CRISPR technologies into their
Author Manuscript

research. In 2008, scientists completed the first attempt to program CRISPR in bacteria, and
for the first time they hypothesized that the target of CRISPR was not RNA, but DNA
(Brouns et al, 2008). This hypothesis was confirmed within the same year by Marraffini and
Sontheimer (2008), clarifying the mechanism by which CRISPR cleaves DNA: Cas9
nuclease guided by CRISPR RNAs (crRNAs) creates DSBs in DNA at precise positions
(Garneau et al, 2010). These findings as well as the later discovery of trans-activating

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 3

CRISPR RNA (tracrRNA) (Deltcheva et al, 2011; Jinek et al, 2012) positioned CRISPR-
Author Manuscript

Cas9 systems on a novel stage for genome engineering.

Since the release of several milestone studies of bacteria utilizing the CRISPR-Cas9 system
(Gasiunas et al, 2012; Jinek et al, 2012), additional groups have adopted the system as an
editing tool for mammalian cells. Researchers from the Massachusetts Institute of
Technology have developed and optimized the Streptococcus pyogenes CRISPR-Cas9
system to facilitate efficient genome editing in mammalian cells (Cong et al, 2013).
Simultaneously, another group established a CRISPR-Cas9 system as an RNA-guided
editing tool for human genome engineering using a chimeric single-guide RNA (sgRNA)
instead of a tracrRNA:crRNA duplex (Mali et al, 2013a). Inspired by successful outcomes in
vitro, scientists have started to apply a CRISPR-Cas9 system in vivo. Several studies have
reported efficient genome editing in multiple organisms (e.g., zebrafish, yeast, mouse,
drosophila, pig and monkey, among others) (Jackson et al, 2005; Soda et al, 2007; Hwang et
Author Manuscript

al, 2013; Maddalo et al, 2014; Niu et al, 2014; Xue et al, 2014; Akiyama & Gibson, 2015;
DiCarlo et al, 2015; Gantz & Bier, 2015; Redel & Prather, 2015). More interestingly, a
CRISPR-Cas9 system was applied to screen for loss-of-function using a high-throughput
screening platform in human cells (Koike-Yusa et al, 2014; Shalem et al, 2014; Wang et al,
2014; Zhou et al, 2014). These genome-scale knockout screens provide an alternative
screening system to RNA interference (RNAi), which is limited by partial depletion of target
gene levels, toxicity and transient silencing (Echeverri et al, 2006; Kaelin, 2012; Sanchez-
Rivera & Jacks, 2015).

Further modifications of the CRISPR system have been reported. Qi et al (2013)


demonstrated a catalytically inactive Cas9 protein (dCas9) lacking endonuclease activity that
can be used as a platform for RNA-guided transcription regulation; this modified system is
Author Manuscript

called CRISPR interference (CRISPRi). Unlike RNAi-based silencing, which destructs


transcribed mRNAs, the CRISPR-dCas9 system directly blocks transcription elongation
within protein-coding regions and leads to dramatic suppression of transcription, with no
detectable off-target effects (Qi et al, 2013). The authors subsequently reported that both
repressive and activating effector domains, such as KRAB and VP64, can be fused to dCas9
to either repress (CRISPRi) or activate (CRISPRa) the transcription of target genes (Gilbert
et al, 2013). This CRISPRi/a system was further modified to control the transcription levels
of endogenous genes up to ~1000-fold, allowing researchers to determine how gene dosage
affects cellular functions of interest (Gilbert et al, 2014). Other than targeting dsDNAs,
CRISPR-Cas9 was modified to bind or cut RNA targets in vitro by providing the protospacer
adjacent motif (PAM) as part of an oligonucleotide (PAMmer). This strategy enables Cas9 to
cleave RNA targets in a programmable manner and facilitates direct transcript detection
Author Manuscript

(O’Connell et al, 2014). The latest study further established nuclear-localized RNA-targeting
Cas9 (RCas9) as a tool to track RNAs in living cells, in which RCas9 is capable of tracking
the localization and movement of endogenous RNAs (Nelles et al, 2016). This work enables
scientists to further measure other RNA features and manipulate these processes to correct
disease-causing RNA behaviours (Fig 1).

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 4

Applications of CRISPR-Cas9 technology in haematological disorders


Author Manuscript

Of the three types of CRISPR systems, the type II system is the simplest for genome editing
technology, requiring only Cas9 proteins and two RNAs: tracrRNA and crRNA (Brouns et
al, 2008; Wiedenheft et al, 2011; Jinek et al, 2012). The tracrRNA:crRNA duplex was
further simplified with a programmed sgRNA, which directs Cas9 endonuclease toward any
double-stranded DNA sequence of interest to trigger NHEJ or HDR-mediated genome
editing (Jinek et al, 2012; Mali et al, 2013a). The functions and mechanisms of the type II
CRISPR-Cas9 system have been extensively reviewed in detail (Hsu et al, 2014; Sanchez-
Rivera & Jacks, 2015; Lander, 2016).

Non-cancerous haematological disorders


β-thalassaemia—β-thalassaemia is one of the most common inherited blood disorders
and is characterized by mutations in the human haemoglobin beta (HBB, also known as β-
Author Manuscript

globin) gene. Decreased HBB expression results in the accumulation of α-globin chains,
leading to ineffective erythropoiesis and thus severe anaemia (Ribeil et al, 2013; Finotti et al,
2015). One curative treatment for β-thalassaemia is allogeneic haematopoietic stem cell
transplantation (HSCT); however, this treatment is only employed for those patients with
suitable matching donors. Although extensive progress has been achieved in allogeneic
HSCT, there is always an elevated risk of suffering graft-versus-host disease (GVHD), graft
rejection or even transplant-related mortality (King & Shenoy, 2014).

Compared with allogeneic HSCT, gene therapy has been considered an alternative
therapeutic option, as several clinical trials have been successfully conducted in patients
with β-thalassaemia (Cavazzana-Calvo et al, 2010; Finotti et al, 2015). Virus-based vectors
have been widely used in gene therapy due to their ability to integrate permanently into
Author Manuscript

genomic DNA. Nevertheless, insertional activation of protooncogenes remains a major


problem associated with vector-mediated gene therapy and can lead to secondary cancers,
such as leukaemia (Wu & Dunbar, 2011).

Similar to virus-mediated gene therapy, CRISPR-Cas9-mediated genome editing is used to


correct HBB gene mutations in patients via HDR, leading to normal erythropoiesis. In the
past two years, several research groups have successively applied CRISPR-Cas9 technology
to correct β-thalassaemia mutations in patient-derived induced pluripotent stem cells (iPSCs)
(Xie et al, 2014a; Song et al, 2015; Xu et al, 2015). Gene-corrected iPSCs can restore HBB
expression with a minimal off-target effect. In a recent study, the fibroblasts of a β-
thalassaemia patient were reprogrammed to become transgene-free naïve-state iPSCs, which
showed significantly higher targeting efficiencies in the CRISPR-Cas9 genome editing
Author Manuscript

system compared with primed iPSCs (Yang et al, 2016).

In addition to correcting β-thalassaemia-causing mutations, other interesting attempts have


been reported in the treatment of β-thalassaemia via reactivation of fetal haemoglobin
(HbF); elevated HbF is beneficial in β-haemoglobinopathies [e.g., β-thalassaemia and
sickle-cell disease (SCD)] (Bauer et al, 2012) Unfortunately, therapy via HbF reactivation
has lagged for many years due to the unclear regulatory relationship between HbF and adult
haemoglobin (HbA) during the development of erythroid cells. This gap was not filled until

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 5

the BCL11A locus, a critical regulator of the fetal-to-adult switch and HbF silencing, was
Author Manuscript

identified in genome-wide association studies (GWAS) of HbF (Lettre et al, 2008; Uda et al,
2008). Recently, Canver et al (2015) identified an erythroid-enhancer region within the
BCL11A gene via CRISPR-Cas9-mediated in situ saturating mutagenesis. As an ideal
therapeutic target for β-haemoglobin disorders, this region can be disrupted by individual
sgRNAs, leading to efficient HbF reactivation.

Sickle-cell disease—Along with β-thalassaemia, SCD remains a global health burden,


particularly in underdeveloped countries (Weatherall, 2010). SCD is another human genetic
disease that is caused by the single-nucleotide mutation of the HBB gene. Under
deoxygenated conditions, sickle haemoglobin polymerization within erythrocytes deforms
red blood cells (RBCs) to assume a sickle shape, clogs blood vessels and results in painful
symptoms, such as haemolysis, ischaemia, anaemia and multi-organ injury (Gewin, 2015;
Hoban et al, 2016).
Author Manuscript

Several groups have attempted to correct the SCD mutation in patient-derived iPSCs through
ZFN-based (Zou et al, 2011) or TALENs-induced HDR (Sun & Zhao, 2014). One recent
study utilized a CRISPR-Cas9 system to target the endogenous HBB locus in human iPSCs
generated from SCD patients, resulting in higher efficiencies compared with other nucleases
(Huang et al, 2015). In addition, HbF reactivation by targeting the BCL11A gene using a
CRISPR-Cas9, as detailed above, is also a promising treatment for SCD. Alternatively,
epigenetic modifiers are beginning to emerge as potential therapeutic targets for SCD. In
murine erythroleukaemia cells, CRISPR-Cas9-mediated Ehmt2 knockout increased the
expression of embryonic Hbb genes Hbb-y (also termed Hbb-εy) and Hbb-bh1 (Hbb-βh1),
providing genetic evidence for EHMT1/2 inhibition, which induced HbF synthesis. This
approach requires further investigation as a novel therapeutic approach for the treatment of
Author Manuscript

SCD (Renneville et al, 2015).

Fanconi anaemia—Fanconi anaemia (FA) is a genetic DNA repair-deficient human


disorder that results from mutations in FA genes. Patients with FA develop bone marrow
failure and congenital defects, and exhibit a high predisposition to malignancies such as
myelodysplastic syndrome (MDS), acute myeloid leukaemia (AML) and solid tumours
(Taniguchi & D’Andrea, 2006; Auerbach, 2009; Soulier, 2011; Kottemann &
Smogorzewska, 2013).

To overcome the limitations of HSCT-based treatment in FA patients, gene therapies have


been adopted (Bogliolo & Surralles, 2015). Somatic cells from FA patients, for example,
have been reprogrammed to generate patient-specific iPSCs; notably, the corrected FA-
Author Manuscript

specific iPSCs generated the first disease-free haematopoietic progenitor cells (Raya et al,
2009). Subsequently, more efficient gene correction was performed by targeting FANCA
insertion into the AAVS1 safe harbour locus using ZFN (Rio et al, 2014). Recently, Osborn
et al (2015) generated a CRISPR-Cas9 system for the FANCC locus to analyse repair of the
FANCC c.456 + 4A>T point mutation in FA fibroblasts. This result indicates that CRISPR-
Cas9-mediated HDR offers a novel gene correction strategy for FA patients and can provide
a proof of principle for precise genome editing.

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 6

Diamond-Blackfan anaemia—Diamond-Blackfan anaemia (DBA) is a rare inherited


Author Manuscript

pure erythroid aplasia that only affects RBCs with decreased erythroid progenitors in the
bone marrow. The main cause of DBA is the mutation of ribosomal protein genes, which
lead to ribosomal stress and TP53 (p53) activation (Dutt et al, 2011; Jaako et al, 2011).

In addition to bone marrow transplantation, glucocorticoid hormones as well as other


supplementary therapies have been used to improve RBC production in DBA; however,
many side effects and unresponsive cases have been reported (Vlachos et al, 2001, 2008;
Ball, 2011). Recently, one research group successfully rescued haematological defects in a
zebra-fish DBA model by inactivating tp53 (p53) in erythrocytes via a CRISPR-Cas9-based
vector system (Ablain et al, 2015). This finding exemplifies the effective use of a CRISPR-
Cas9 in vivo genetic model and its further development in model organisms other than
zebrafish.
Author Manuscript

Thrombocytopenia—Thrombocytopenia refers to a blood disorder that involves a


decrease in thrombocytes caused by either inherited or acquired alterations (Drachman,
2004). To date, only a few cases have documented the use of CRISPR-Cas9 for the treatment
of thrombocytopenia. In one study, Zhang et al (2016a) used a CRISPR-Cas9 gene editing to
convert megakaryocyte-like cells and iPSCs from HPA-1a (human platelet alloantigen-1a) to
the HPA-1b alloantigenic epitope, which is relatively rare and difficult to obtain. This work
highlights the potential of the CRISPR-Cas9 system to produce designer platelets for
diagnostic and therapeutic purposes.

Another study focused on the thrombocytopenia caused by solid-organ xenotransplantation


(Butler et al, 2016). The authors genetically modified pigs using CRISPR-Cas9 and found
that porcine kidneys failed to consume human platelets; in contrast, porcine livers exhibited
Author Manuscript

a significant ability to consume human platelets. This finding reveals different responses of
porcine organs to human platelets. Further genetic modification of donor pigs is needed to
prevent the development of thrombocytopenia in pig-to-primate xenotransplantation.

Haemophilia—Haemophilia refers to a group of hereditary disorders of blood coagulation


(Ratnoff & Bennett, 1973). The most common type of this disorder is haemophilia A (HA),
which is caused by a deficiency in coagulation factor VIII (FVIII). Another type,
haemophilia B (HB), is caused by coagulation factor IX (FIX) deficiency.

Several approaches have been reported to treat haemophilia using gene therapy (High,
2012). Adenovirus-associated virus (AAV) vectors expressing either human FVIII
(McIntosh et al, 2013) or human FIX (Nathwani et al, 2011) have been used in the past
decade as the first HA or HB gene therapy. Because AAV-mediated gene transfer is
Author Manuscript

applicable only to a limited group of patients, alternative strategies have been proposed
(High, 2012). Last year, one research group developed a CRISPR-Cas9 system to revert two
inverted chromosomal regions back to the normal orientation in HA patient-derived iPSCs;
the endothelial cells differentiated from the inversion-corrected iPSCs efficiently rescued the
FVIII deficiency in an HA mouse with no off-target mutations (Park et al, 2015). More
recently, a group from China identified a novel mutation, Y371D, in the human F9 (FIX)
gene. Introduction of this mutation into mice via CRISPR-Cas9 led to a severe HB

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 7

phenotype, whereas correction of this mutation by CRISPR-Cas9-mediated in situ genome


Author Manuscript

editing restored haemostasis in HB mice (Guan et al, 2016). These preliminary data
demonstrate the feasibility of CRISPR-Cas9 as a versatile tool in precision medicine.

Others—Similar to human blood disorders, genetic correction using a CRISPR-Cas9


system has been reported in the context of other haematological disorders. Using
myeloproliferative neoplasm patient-derived iPSCs carrying a JAK2-V617F point mutation,
Smith et al (2015) demonstrated that Cas9 could specifically target the mutant allele with
little disruption to the other allele. This result highlights the advantages of CRISPR-Cas9
technology in allele-specific genome targeting, offering a unique targeting specificity that
has not been previously achieved. Another group utilized a CRISPR-Cas9 system to create
Kdelr1 mutant mice, which had reduced expression of the T-cell receptor and increased
expression of CD44 (Siggs et al, 2015). This study indicated that KDELR1 is responsible for
T-cell lymphopenia, although the precise mechanism of T lymphopenia in Kdelr1 mutants
Author Manuscript

has not yet been elucidated. Applications of a CRISPR-Cas9 system to non-cancerous


haematological disorders are summarized in Table I and Fig 2.

Malignant haematological disorders


Lymphoma—Despite improved responses with frontline therapies in lymphoma
(Shankland et al, 2012), relapsed and refractory cases remain a significant challenge (Brice,
2008; Gangatharan & Kuruvilla, 2013). Genome-editing technology has become a potential
therapeutic option in lymphoid malignancies, although most applications of CRISPR-Cas9
to date have been limited to bench studies.

Malina et al (2013) first established the lentiviral/retroviral-based ‘all-in-one’ CRISPR-


Cas9-mediated ex vivo editing of Trp53 in chemoresistant lymphomas in the Eµ-myc mouse
Author Manuscript

model. Notably, the authors extended the CRISPR-Cas9 platform to positive selection
screenings (identifying those cells that ‘pass’ the selection mechanism) both in vitro and in
vivo using TP53 as a model genetic target. This work demonstrated the efficiency and
simplicity of editing unique genomic loci using a CRISPR-Cas9 system. Recently, an
exciting work unveiled the target gene for the therapeutic activity of NUTLIN3A (Valente et
al, 2016), a novel small-molecule antagonist of MDM2 that promotes TP53 activation.
Using mouse models lacking TP53 target genes, the authors demonstrated that BBC3
(PUMA) is responsible for the resistance of NUTLIN3A in lymphomas. Further CRISPR-
Cas9-mediated silencing of the BBC3 gene confirmed that BBC3 expression might predict
NUTLIN3A treatment outcomes in patients.

More recently, a study reported the application of a CRISPR-Cas9 system to disrupt CXCR4
Author Manuscript

expression in mantle cell lymphomas (MCL), a highly aggressive subset of B-cell non-
Hodgkin lymphomas (NHLs) (Chen et al, 2016). Using lentiviral-based CRISPR-Cas9-
mediated silencing of CXCR4, the researchers found that reactive oxygen-mediated CXCR4
expression is a key signal inducing autophagy, which contributes to the survival of
bortezomib-resistant MCL cells (Chen et al, 2016). The same group also reported that
Hedgehog inhibitor LDE225 induces autophagy to promote MCL cell survival via the
upregulation of CXCR4 because LDE225 treatment failed to induce autophagy in CXCR4

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 8

knockout cells generated by CRISPR-Cas9 (Zhang et al, 2016b). To summarize, the


Author Manuscript

CRISPR-Cas9 system generates numerous exciting opportunities to unravel the mechanisms


of drug resistance and to identify potential therapeutic targets in lymphomas.

Myeloma—Multiple myeloma (MM) is the second most frequent haematological


malignancy in the US after NHLs and is characterized by the presence of excess numbers of
malignant plasma cells in the bone marrow. Over the past several years, novel treatment
options have markedly improved the outcomes of MM patients (Richardson et al, 2007;
Raab et al, 2009; Moreau et al, 2015). However, important challenges remain that necessitate
new strategies (Moreau et al, 2015).

Wu et al (2014) first reported genome-editing in MM cells. The authors discovered that


TALENs are a very robust and efficient genome-editing tool in MM cells. Recently, Tagde et
al (2016) utilized CRISPR-Cas9 editing to silence MUC1-C, an oncogenic transmembrane
Author Manuscript

protein, and found that MUC1-C occupies the MYC promoter and activates the MYC gene
via a β-catenin/TCF4-mediated mechanism. Further analysis using CRISPR-Cas9-mediated
microarray datasets demonstrated that MUC1-C drives MYC expression and contributes to
MM progression. Therefore, TALENs and CRISPR-Cas9 technologies may be efficient tools
for targeting specific genes, facilitating the construction of knockout reagents. In the near
future, the use of these tools will be extended to clinical treatment in MM patients.

Leukaemia—In comparison to lymphoma and MM, the CRISPR-Cas9 genome editing has
been overwhelmingly applied in leukaemia research, especially in myeloid leukaemia. Heckl
et al (2014) used the CRISPR-Cas9 system to generate mouse models of AML using
lentivirus-mediated editing of multiple genes (e.g., Runx1 and Tp53, among others) in
murine haematopoietic stem and progenitor cells (HSPC). This work highlights the
Author Manuscript

application of CRISPR-Cas9 to generate ex vivo genome editing to delineate the complexity


of human blood cancers. Later that year, Chen et al (2014) discovered a novel tumour
suppressor gene, KMT2C (lysine methyltransferase 2C, also termed MLL3), in chromosome
7q using an ex vivo CRISPR-Cas9-mediated approach. Further experiments in a mouse
model confirmed that KMT2C is a haploinsufficient tumour suppressor in AML and its
inhibition impairs differentiation of the myeloid lineage and produces an MDS-like
syndrome in mice. Interestingly, murine AMLs with KMT2C suppression are resistant to
conventional chemotherapy. This study provides a valuable clue for the treatment of AML
patients harbouring −7/del(7q) lesions.

In the past 2 years, there have been an increasing number of publications related to CRISPR-
Cas9 technology in leukaemia research; however most studies have focused primarily on the
Author Manuscript

delivery of gene function studies in vitro. For example, one group reported that CRISPR-
Cas9-mediated loss of EPAS1 (HIF-2α) alone had no impact on the growth of human AML
cells under hypoxic conditions (Vukovic et al, 2015). In another study, the authors combined
an RNA sequencing method and CRISPR/TALEN-based knockout techniques to identify
genes associated with drug resistance in AML (Rathe et al, 2014). In addition to gene
silencing, CRISPR-Cas9 has also been reported in the context of gene insertions and
corrections. Cooper et al (2015), using CRISPR-Cas9-induced site-mutation, found an
enhancer that is critical for normal haematopoiesis and, potentially, during myeloid

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 9

transformation. In human myeloid leukaemia K562 cells, mutation of CRISPR-Cas9-


Author Manuscript

induced to SRSF2 revealed the mechanism of the disease-associated SRSF2 mutation in the
regulation of splicing, thereby providing a group of mis-spliced mRNA isoforms for
potential therapeutic targeting (Zhang et al, 2015a). In another recent study, CRISPR-Cas9-
mediated ASXL1 homozygous correction not only restored ASXL1 expression in chronic
myeloid leukaemia (CML) cells, but it also increased survival in mouse xenografts (Valletta
et al, 2015).

Along with many studies investigating AML, the application of a CRISPR-Cas9 system to
other types of leukaemia has been reported. In human T-cell acute lymphoblastic leukaemia
(T-ALL) cells, a CRISPR-Cas9 editing tool was used to disrupt TAL1 (Navarro et al, 2015)
or TRIB1 (TRB1) (Miyajima et al, 2015) genes to delineate their biological functions. More
recently, Dronabinol (Tetrahydrocannabinol, THC), a US Food and Drug Administration-
approved cannabinoid receptor (CNB) agonist for the treatment of chemotherapy-induced
Author Manuscript

nausea and vomiting, was found to induce apoptosis in acute leukaemia cells, as evidenced
by the abrogation of pro-apoptotic effects of CRISPR-mediated knockout of CNB1 (CB1) or
CNB2 (CB2) following THC treatment. (Kampa-Schittenhelm et al, 2016).

Because CRISPR-Cas9 can achieve complete silencing of target genes, many research
groups have employed a CRISPR-Cas9 system in large-scale, loss-of-function screens in
blood cancer cells. One earlier study established a lentivirus-mediated library containing 73
000 sgRNAs to generate knockout collections for genome-wide screening in myeloid
leukaemia cells (Wang et al, 2014). This system provides several advancements: (i)
CRISPR-Cas9-mediated gene depletion leads to a complete loss of gene function, which
enables the targeting of non-coding regions that are conventionally considered non-
targetable by RNAi; (ii) most sgRNAs can efficiently create mutations at target sites with
Author Manuscript

perfect discrimination between true and false positives at the significance threshold; (iii) the
screens showed minimal off-target effects. This study pioneered whole-genome analysis
using sgRNA-mediated screens in blood cancers. Recently, Vakoc and his team designed an
sgRNA library to target 192 chromatin regulatory domains (Shi et al, 2015). From this
screen, they discovered 6 known drug targets and 19 novel binding domains in murine AML
cells. This sgRNA-targeted screening required 2 weeks to unveil the 6 previously described
drug targets, an achievement that had taken researchers over half a century to identify using
conventional methods (Ricks, 2015). Applications of CRISPR-Cas9 in haematological
malignancies are summarized in Table II and Fig 2.

Secondary haematological disorders


Chronic granulomatous disease—Chronic granulomatous disease (CGD) is a rare
Author Manuscript

genetic immunodeficiency disorder of phagocytic cells (Seger, 2008). The abnormalities of


nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits result in the lack
of an anti-pathogen oxidative burst, thereby leading to recurrent life-threatening bacterial
and fungal infections (Nienhuis, 2013). So far, only one reported study has utilized CRISPR-
Cas9 in CGD. Flynn et al (2015) corrected a single point mutation (T>G) at the end of the
first intron of the CYBB gene using CRISPR-Cas9. Mutation of this gene is responsible for
most cases of CGD. Surprisingly, the gene correction resulted in the restoration of oxidative

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 10

burst function in iPSCs derived from a CGD patient, offering a potential new gene
Author Manuscript

therapeutic approach to CGD treatment using CRISPR-Cas9.

Human immunodeficiency virus (HIV) infection—HIV infection has been associated


with a broad range of haematological disorders (Scadden et al, 1989). Before the advent of
CRISPR-Cas9 technology, ZFN-induced disruption of the CCR5 gene, an essential HIV-1
co-receptor required for virus entry, had become a promising gene therapy and was
evaluated in clinical trials (Tebas et al, 2014). Recent advances in CRISPR-Cas9 have
further revolutionized studies of HIV-1. Since Cho et al (2013) first tested the DNA cleavage
activity of a CRISPR-Cas9 system for silencing the CCR5 gene in vitro, several studies have
reported CRISPR-Cas9-mediated disruption of CCR5 (Cradick et al, 2013; Yang et al, 2013;
Ye et al, 2014) and other co-factors, including CXCR4 (Hou et al, 2015) and XPO1
(exportin 1) (Boons et al, 2015).
Author Manuscript

Despite much progress in combined antiviral therapy, it remains a significant challenge to


completely eradicate HIV latent infection. One strategy to achieve this goal is to remove the
proviral HIV-1 DNA from the host cell genome, and several research groups have reported
the potential of the CRISPR-Cas9 system to disrupt latent HIV-1 provirus by targeting the
long terminal repeat (LTR) region (Ebina et al, 2013; Hu et al, 2014; Zhu et al, 2015;
Kaminski et al, 2016). However, the feasibility of this approach in HIV therapy has been
questioned by the latest finding that showed viral escape from CRISPR gene-editing attack
due to the Cas9/sgRNA-derived mutations (Wang et al, 2016). Last year, investigators fused
a synergistic activation mediator (SAM) to dCas9 to target the enhancer of the LTR
promoter in HIV-1 latent cells. This target-specific compulsory reactivation led to cellular
suicide of viral proteins in HIV-1-infected cells and provided a novel therapeutic tool for the
permanent eradication of the HIV virus (Zhang et al, 2015b). More recently, another study
Author Manuscript

identified a targeted hotspot for CRISPR activation. Among five effective sgRNA candidates
targeting different regions of the LTR, sg362F was the most effective for stimulating potent
transcriptional activation of latent HIV-1 infection in human T cells. Unlike chemical-
activating agents, CRISPR-dCas9-mediated reactivation of HIV-1 is specific in all latency
models and does not induce off-target effects (Saayman et al, 2016).

Epstein-Barr virus (EBV) infection—Wang and Quake (2014) first used CRISPR-Cas9
as a novel nuclease system for targeting key EBV-DNA in a Burkitt lymphoma (BL) cell
line. Remarkably, nearly 25% of the treated cells were completely cleared of EBV with no
detectable viral DNA, and another 50% of cells showed a substantial decrease in EBV load
relative to the untreated samples. To apply this strategy to clinical therapy, a safer and more
efficient delivery system for genome editing in patients will be required.
Author Manuscript

Malaria—Since the CRISPR-Cas9 was successfully applied in Drosophila in vivo (Gantz &
Bier, 2015), scientists have attempted to apply this system to target either malaria parasites
(Ghorbal et al, 2014; Zhang et al, 2014) or malaria mosquitoes (Gantz & Bier, 2015;
Hammond et al, 2016). In one representative study, researchers identified a gene that is
required for female fertility in Anopheles mosquitoes and inserted it into a CRISPR-Cas9
gene-drive construct targeting female fertility loci (Hammond et al, 2016). This technique
impaired the fertility of female mosquitoes, leading to extinction of the population over

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 11

several years. In another study, parasite transmission was hindered by insertion of an anti-
Author Manuscript

malarial gene into malaria mosquitoes using CRISPR-Cas9 (Gantz et al, 2015). This strategy
decreased the spread of malaria while maintaining the integrity of the mosquito species.
Although the same genome-editing tool was applied in these two studies, distinct endings
(extinct vs reserve species) might be achieved (Pennisi, 2015; Alphey, 2016).

Leishmaniasis—Last year, one research group from France reported the first efficient
CRISPR-Cas9-mediated genome editing in Leishmania parasites (Sollelis et al, 2015). They
incorporated the CRISPR-Cas9 system into Leishmania major by disrupting the
paraflagellar rod (PFR)-2 loci, the major constituent of the trypanosomatid-specific
filamentous network. Targeting PFR-2 genes, the authors obtained parasites in a single
round of transfection within only 1 month; importantly, no off-targets effects were detected.
Another group subsequently reported the successful implantation of the CRISPR-Cas9
technology in Leishmania donovani (L. donovani). Using this efficient system, the authors
Author Manuscript

introduced drug selection markers and the GFP tag precisely into the L. donovani genome,
leading to improved isolation of gene disruption mutants. (Zhang & Matlashewski, 2015).
Collectively, these landmark reports have facilitated the use of CRISPR-(d)Cas9 for the
treatment of secondary haematological disorders (Table III, Fig 2).

Challenges of CRISPR-Cas9 technology


Technical limitations
CRISPR-Cas9-mediated gene modification can be achieved via transient or stable delivery
of the CRISPR components. Transient transfection in cells has been reported in several
studies (Cho et al, 2013; Cong et al, 2013; Mali et al, 2013a), although viral-based
transfection of CRISPR components is a more efficient alternative to generate stably-
Author Manuscript

modified cells (Malina et al, 2013; Heckl et al, 2014; Chen et al, 2016; Zhang et al, 2016b).
Nevertheless, the delivery efficiency remains a significant challenge in hard-to-transfect
blood cancer cells, especially when electroporation is used as a delivery method. To
overcome these limitations, Han et al (2015) developed rapid cell mechanical deformation.
This technique generates transient holes in the membrane that enable the delivery of
biomaterials via the medium. This approach achieves high delivery efficiency in different
cell types, including lymphoma cells and embryonic stem cells, broadening the cell type
applicability of transient transfection of CRISPR plasmids. In addition to transient
transfection, viral transfection is frequently used to deliver CRISPR-Cas9 system into
haematological malignant cancer cells, although some technical issues persist, such as
relatively low infection rates.
Author Manuscript

The critical role of target genes for cell survival is an additional challenge. For example,
MCL1, an anti-apoptotic BCL2 family member, is required for the sustained survival of
human BL cell lines because silencing of MCL1 leads to rapid cell death. In this case, the
drug-inducible CRISPR-Cas9 lentiviral system provides an option to identify the functions
of tumour-essential genes (Aubrey et al, 2015).

Due to the increased utilization of the NHEJ pathway over HDR in mammalian cells, the
low efficiency of CRISPR-Cas9-induced HDR is an additional limitation (Mali et al, 2013a;

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 12

Wang et al, 2013). Therefore, increasing the efficiency of HDR facilitates the rapid
Author Manuscript

generation of cell lines with precisely edited genes. To resolve this issue, a recent study
identified a RAD51-stimulatory compound, RS-1, which enhances the efficiency of Cas9-
stimulated HDR by 3- to 6-fold (Pinder et al, 2015).To some extent, this discovery improves
efficiency, although greater improvement will be needed in the future.

Off-target effects
With more successful applications of CRISPR-Cas9 in mammalian cells, the issue of
specificity remains a major concern. In 2013, Nature Biotechnology simultaneously
published a series of letters regarding NHEJ-mediated off-target mismatches in human cells
(Fu et al, 2013; Hsu et al, 2013; Mali et al, 2013b; Pattanayak et al, 2013). To overcome
these hurdles, several methods have been developed to detect off-target effects. Deep
sequencing (Fu et al, 2013; Hsu et al, 2013) and whole genome sequencing (Smith et al,
Author Manuscript

2014; Veres et al, 2014) have been considered options to identify mismatches; however, the
possibility of overlooking off-target sites and the higher cost make it difficult to perform this
step in every experiment. Alternatively, a series of online bioinformatics tools were
developed to predict off-target mutation sites (Ran et al, 2013; Fine et al, 2014; Heigwer et
al, 2014; Xie et al, 2014b). However, these computational methods failed to predict bona
fide off-target integration sites (Tsai et al, 2015). Instead, a novel method called Genome-
wide Unbiased Identification of DSBs Enabled by Sequencing (GUIDE-seq) provides an
unbiased and genome-wide method for detecting CRISPR RNA-guided DSBs in cells (Tsai
et al, 2015). In addition, other approaches, such as modified high-throughput genome-wide
translocation sequencing (HTGTS), and the integrase-defective lentiviral vector (IDLV)
assay, were developed to identify off-target effects produced by CRISPR-Cas9 (Frock et al,
2015; Wang et al, 2015). These pioneering studies have increased the technical feasibility of
Author Manuscript

off-target detection (Koo et al, 2015; Bolukbasi et al, 2016), although ‘it is impossible to
draw broad conclusions about the specificity of engineered nucleases’, as recently reported
(Gabriel et al, 2015).

Knockdown or knockout?
Although CRISPR-Cas9 provides a tool for complete silencing, gene knockdown can still be
a valuable tool depending on the experimental objectives. Technically, shRNA-mediated
gene knockdown does not require single clone isolation, whereas CRISPR-Cas9-based gene
silencing usually involves a long duration to generate stable cell lines. Despite many
concerns about which method is more appropriate for delineating gene functions, genome-
editing technology is the apparent option for creating a true genetic null allele, introducing a
point mutation, and correcting a specific mutation (Ul Ain et al, 2015). Currently, more
Author Manuscript

CRISPR-Cas9-based platforms have evolved to meet different demands. For example, DNA-
free CRISPR-Cas9 gene editing can be used to avoid potential integration events; the
specificity of the CRIPSR system can be enhanced using paired Cas9 nickases by targeting
two sites on opposite DNA strands; a CRISPR vector with a fluorescent marker has become
available for positive cell selection; and an inducible Cas9 expression vector has increased
experimental flexibility. These novel CRISPR-Cas9 platforms facilitate its application for
more investigative purposes and, ultimately, for therapeutic use.

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 13

Collectively, the challenges of CRISPR-Cas9 technology discussed herein only reflect those
Author Manuscript

limitations of the studies in vitro, ex vivo or in vivo. The greatest challenge is yet to come.
Many questions remain unanswered regarding the use of this technology in humans. For
example, which delivery system is suitable for humans? How will the human immune
system respond to the administration of genome editing tools in vivo? How can off-target
effects be minimized rather than predicted or detected upon application in humans? With
these uncertainties, further applications in gene and cell therapies will require a long
duration to be fully realized (Maeder & Gersbach, 2016).

Future directions and prospects


Hereditary blood disorders, such as haemoglobin abnormalities, anaemias or coagulation
disorders, represent ideal targets for gene therapy with genome editing technology because
the partial restoration of gene functions are sufficient to reverse the symptoms. Although
Author Manuscript

scientists are striving to unlock new possibilities of CRISPR-Cas9 in different areas, for
example, to eliminate pathogen-carrying species (e.g., malaria and Zika), to resurrect a
woolly mammoth, or to make micro-pig pets even designer babies (Reardon, 2016),
legitimate concerns are arising due to its simple and highly versatile potential to edit the
entire genome of the creatures. Last year, a team of researchers, for the first time, applied
CRISPR-Cas9 to edit the endogenous HBB genes in human embryos; however, the edited
embryos were all mosaic due to the low efficiency of HDR (Liang et al, 2015). Therefore,
exploration in human cells can also raise bioethical issues.

Any intentional or unintentional misuse of this technology may result in unexpected effects
on future generations and lead to substantial or even permanent changes in the human gene
pool. Thus, the international summit on human gene editing was held in Washington, D.C.
Author Manuscript

last winter, where the closing statement recommended the rigorous continuation of basic and
preclinical research and emphasized that any clinical use of germline editing is irresponsible
(Olson, 2016).

In conclusion, CRISPR-Cas9 technology has sparked advancements in haematological


disorders. The scientific breakthroughs with respect to programmable nucleases will allow
us to delineate the mechanisms underlying the development of blood diseases and eventually
contribute to the alleviation of disease symptoms or the cure of certain diseases via safe and
controllable gene therapy.

Acknowledgments
This work is supported by R01 (R01CA181319, NCI) and American Society Scholar Award (RSG-11-157) given to
Author Manuscript

NM.

References
Ablain J, Durand EM, Yang S, Zhou Y, Zon LI. A CRISPR/Cas9 vector system for tissue-specific gene
disruption in zebrafish. Developmental Cell. 2015; 32:756–764. [PubMed: 25752963]
Akiyama T, Gibson MC. Decapentaplegic and growth control in the developing Drosophila wing.
Nature. 2015; 527:375–378. [PubMed: 26550824]
Alphey L. Can CRISPR-Cas9 gene drives curb malaria? Nature Biotechnology. 2016; 34:149–150.

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 14

Aubrey BJ, Kelly GL, Kueh AJ, Brennan MS, O’Connor L, Milla L, Wilcox S, Tai L, Strasser A,
Herold MJ. An inducible lentiviral guide RNA platform enables the identification of tumor-essential
Author Manuscript

genes and tumor-promoting mutations in vivo. Cell Reports. 2015; 10:1422–1432. [PubMed:
25732831]
Auerbach AD. Fanconi anemia and its diagnosis. Mutation Research. 2009; 668:4–10. [PubMed:
19622403]
Ball S. Diamond Blackfan anemia. Hematology: The Education Program of the American Society of
Hematology. 2011; 2011:487–491.
Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S, Romero DA, Horvath P.
CRISPR provides acquired resistance against viruses in prokaryotes. Science. 2007; 315:1709–
1712. [PubMed: 17379808]
Bauer DE, Kamran SC, Orkin SH. Reawakening fetal hemoglobin: prospects for new therapies for the
beta-globin disorders. Blood. 2012; 120:2945–2953. [PubMed: 22904296]
Bibikova M, Golic M, Golic KG, Carroll D. Targeted chromosomal cleavage and mutagenesis in
Drosophila using zinc-finger nucleases. Genetics. 2002; 161:1169–1175. [PubMed: 12136019]
Bogliolo M, Surralles J. Fanconi anemia: a model disease for studies on human genetics and advanced
Author Manuscript

therapeutics. Current Opinion in Genetics & Development. 2015; 33:32–40. [PubMed: 26254775]
Bolotin A, Quinquis B, Sorokin A, Ehrlich SD. Clustered regularly interspaced short palindrome
repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology. 2005; 151:2551–
2561. [PubMed: 16079334]
Bolukbasi MF, Gupta A, Wolfe SA. Creating and evaluating accurate CRISPR-Cas9 scalpels for
genomic surgery. Nature Methods. 2016; 13:41–50. [PubMed: 26716561]
Boons E, Vanstreels E, Jacquemyn M, Nogueira TC, Neggers JE, Vercruysse T, van den Oord J, Tamir
S, Shacham S, Landesman Y, Snoeck R, Pannecouque C, Andrei G, Daelemans D. Human
Exportin-1 is a target for combined therapy of HIV and AIDS related lymphoma. EBioMedicine.
2015; 2:1102–1113. [PubMed: 26501108]
Brice P. Managing relapsed and refractory Hodgkin lymphoma. British Journal of Haematology. 2008;
141:3–13. [PubMed: 18279457]
Brouns SJ, Jore MM, Lundgren M, Westra ER, Slijkhuis RJ, Snijders AP, Dickman MJ, Makarova KS,
Koonin EV, van der Oost J. Small CRISPR RNAs guide antiviral defense in prokaryotes. Science.
Author Manuscript

2008; 321:960–964. [PubMed: 18703739]


Butler JR, Martens GR, Li P, Wang ZY, Estrada JL, Ladowski JM, Tector M, Tector AJ. The fate of
human platelets exposed to porcine renal endothelium: a singlepass model of platelet uptake in
domestic and genetically modified porcine organs. Journal of Surgical Research. 2016; 200:698–
706. [PubMed: 26375504]
Canver MC, Smith EC, Sher F, Pinello L, Sanjana NE, Shalem O, Chen DD, Schupp PG, Vinjamur
DS, Garcia SP, Luc S, Kurita R, Nakamura Y, Fujiwara Y, Maeda T, Yuan GC, Zhang F, Orkin SH,
Bauer DE. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature.
2015; 527:192–197. [PubMed: 26375006]
Capecchi MR. Altering the genome by homologous recombination. Science. 1989a; 244:1288–1292.
[PubMed: 2660260]
Capecchi MR. The new mouse genetics: altering the genome by gene targeting. Trends in Genetics.
1989b; 5:70–76. [PubMed: 2660363]
Cavazzana-Calvo M, Payen E, Negre O, Wang G, Hehir K, Fusil F, Down J, Denaro M, Brady T,
Westerman K, Cavallesco R, Gillet-Legrand B, Caccavelli L, Sgarra R, Maouche-Chretien L,
Author Manuscript

Bernaudin F, Girot R, Dorazio R, Mulder GJ, Polack A, Bank A, Soulier J, Larghero J, Kabbara N,
Dalle B, Gourmel B, Socie G, Chretien S, Cartier N, Aubourg P, Fischer A, Cornetta K, Galacteros
F, Beuzard Y, Gluckman E, Bushman F, Hacein-Bey-Abina S, Leboulch P. Transfusion
independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature.
2010; 467:318–322. [PubMed: 20844535]
Chen C, Liu Y, Rappaport AR, Kitzing T, Schultz N, Zhao Z, Shroff AS, Dickins RA, Vakoc CR,
Bradner JE, Stock W, LeBeau MM, Shannon KM, Kogan S, Zuber J, Lowe SW. MLL3 is a
haploinsufficient 7q tumor suppressor in acute myeloid leukemia. Cancer Cell. 2014; 25:652–665.
[PubMed: 24794707]

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 15

Chen Z, Teo AE, McCarty N. ROS-Induced CXCR4 Signaling Regulates Mantle Cell Lymphoma
(MCL) cell survival and drug resistance in the bone marrow microenvironment via autophagy.
Author Manuscript

Clinical Cancer Research. 2016; 22:187–199. [PubMed: 26350264]


Cho SW, Kim S, Kim JM, Kim JS. Targeted genome engineering in human cells with the Cas9 RNA-
guided endonuclease. Nature Biotechnology. 2013; 31:230–232.
Choulika A, Perrin A, Dujon B, Nicolas JF. Induction of homologous recombination in mammalian
chromosomes by using the I-SceI system of Saccharomyces cerevisiae. Molecular and Cellular
Biology. 1995; 15:1968–1973. [PubMed: 7891691]
Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, Zhang
F. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013; 339:819–823.
[PubMed: 23287718]
Cooper S, Guo H, Friedman AD. The +37 kb Cebpa enhancer is critical for cebpa myeloid gene
expression and contains functional sites that bind SCL, GATA2, C/EBPalpha, PU.1, and additional
Ets factors. PLoS ONE. 2015; 10:e0126385. [PubMed: 25938608]
Cradick TJ, Fine EJ, Antico CJ, Bao G. CRISPR/Cas9 systems targeting beta-globin and CCR5 genes
have substantial off-target activity. Nucleic Acids Research. 2013; 41:9584–9592. [PubMed:
Author Manuscript

23939622]
Deltcheva E, Chylinski K, Sharma CM, Gonzales K, Chao Y, Pirzada ZA, Eckert MR, Vogel J,
Charpentier E. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III.
Nature. 2011; 471:602–607. [PubMed: 21455174]
DiCarlo JE, Chavez A, Dietz SL, Esvelt KM, Church GM. Safeguarding CRISPR-Cas9 gene drives in
yeast. Nature Biotechnology. 2015; 33:1250–1255.
Drachman JG. Inherited thrombocytopenia: when a low platelet count does not mean ITP. Blood. 2004;
103:390–398. [PubMed: 14504084]
Dutt S, Narla A, Lin K, Mullally A, Abayasekara N, Megerdichian C, Wilson FH, Currie T, Khanna-
Gupta A, Berliner N, Kutok JL, Ebert BL. Haploinsufficiency for ribosomal protein genes causes
selective activation of p53 in human erythroid progenitor cells. Blood. 2011; 117:2567–2576.
[PubMed: 21068437]
Ebina H, Misawa N, Kanemura Y, Koyanagi Y. Harnessing the CRISPR/Cas9 system to disrupt latent
HIV-1 provirus. Scientific Reports. 2013; 3:2510. [PubMed: 23974631]
Author Manuscript

Echeverri CJ, Beachy PA, Baum B, Boutros M, Buchholz F, Chanda SK, Downward J, Ellenberg J,
Fraser AG, Hacohen N, Hahn WC, Jackson AL, Kiger A, Linsley PS, Lum L, Ma Y, Mathey-
Prevot B, Root DE, Sabatini DM, Taipale J, Perrimon N, Bernards R. Minimizing the risk of
reporting false positives in large-scale RNAi screens. Nature Methods. 2006; 3:777–779.
[PubMed: 16990807]
Fine EJ, Cradick TJ, Zhao CL, Lin Y, Bao G. An online bioinformatics tool predicts zinc finger and
TALE nuclease off-target cleavage. Nucleic Acids Research. 2014; 42:e42. [PubMed: 24381193]
Finotti A, Breda L, Lederer CW, Bianchi N, Zuccato C, Kleanthous M, Rivella S, Gambari R. Recent
trends in the gene therapy of beta-thalassemia. Journal of Blood Medicine. 2015; 6:69–85.
[PubMed: 25737641]
Flynn R, Grundmann A, Renz P, Hanseler W, James WS, Cowley SA, Moore MD. CRISPR-mediated
genotypic and phenotypic correction of a chronic granulomatous disease mutation in human iPS
cells. Experimental Hematology. 2015; 43:e833.
Frock RL, Hu J, Meyers RM, Ho YJ, Kii E, Alt FW. Genome-wide detection of DNA double-stranded
breaks induced by engineered nucleases. Nature Biotechnology. 2015; 33:179–186.
Author Manuscript

Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung JK, Sander JD. High-frequency off-target
mutagenesis induced by CRISPR-Cas nucleases in human cells. Nature Biotechnology. 2013;
31:822–826.
Gabriel R, von Kalle C, Schmidt M. Mapping the precision of genome editing. Nature Biotechnology.
2015; 33:150–152.
Gangatharan S, Kuruvilla J. Relapsed and refractory aggressive NHL: time for a change. Transfusion
and Apheresis Science. 2013; 49:72–79. [PubMed: 23835116]
Gantz VM, Bier E. Genome editing. The mutagenic chain reaction: a method for converting
heterozygous to homozygous mutations. Science. 2015; 348:442–444. [PubMed: 25908821]

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 16

Gantz VM, Jasinskiene N, Tatarenkova O, Fazekas A, Macias VM, Bier E, James AA. Highly efficient
Cas9-mediated gene drive for population modification of the malaria vector mosquito Anopheles
Author Manuscript

stephensi. Proceedings of the National Academy of Sciences of the United States of America.
2015; 112:E6736–E6743. [PubMed: 26598698]
Garneau JE, Dupuis ME, Villion M, Romero DA, Barrangou R, Boyaval P, Fremaux C, Horvath P,
Magadan AH, Moineau S. The CRISPR/Cas bacterial immune system cleaves bacteriophage and
plasmid DNA. Nature. 2010; 468:67–71. [PubMed: 21048762]
Gasiunas G, Barrangou R, Horvath P, Siksnys V. Cas9-crRNA ribonucleoprotein complex mediates
specific DNA cleavage for adaptive immunity in bacteria. Proceedings of the National Academy of
Sciences of the United States of America. 2012; 109:E2579–E2586. [PubMed: 22949671]
Gewin V. Medicine: expanding possibilities. Nature. 2015; 528:S10–S11. [PubMed: 26630591]
Ghorbal M, Gorman M, Macpherson CR, Martins RM, Scherf A, Lopez-Rubio JJ. Genome editing in
the human malaria parasite Plasmodium falciparum using the CRISPR-Cas9 system. Nature
Biotechnology. 2014; 32:819–821.
Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA, Torres SE, Stern-Ginossar N, Brandman O,
Whitehead EH, Doudna JA, Lim WA, Weissman JS, Qi LS. CRISPR-mediated modular RNA-
Author Manuscript

guided regulation of transcription in eukaryotes. Cell. 2013; 154:442–451. [PubMed: 23849981]


Gilbert LA, Horlbeck MA, Adamson B, Villalta JE, Chen Y, Whitehead EH, Guimaraes C, Panning B,
Ploegh HL, Bassik MC, Qi LS, Kampmann M, Weissman JS. Genome-Scale CRISPR-mediated
control of gene repression and activation. Cell. 2014; 159:647–661. [PubMed: 25307932]
Guan Y, Ma Y, Li Q, Sun Z, Ma L, Wu L, Wang L, Zeng L, Shao Y, Chen Y, Ma N, Lu W, Hu K, Han
H, Yu Y, Huang Y, Liu M, Li D. CRISPR/Cas9-mediated somatic correction of a novel coagulator
factor IX gene mutation ameliorates hemophilia in mouse. EMBO Molecular Medicine. 2016;
8:477–488. [PubMed: 26964564]
Hammond A, Galizi R, Kyrou K, Simoni A, Siniscalchi C, Katsanos D, Gribble M, Baker D, Marois
E, Russell S, Burt A, Windbichler N, Crisanti A, Nolan T. A CRISPR-Cas9 gene drive system
targeting female reproduction in the malaria mosquito vector Anopheles gambiae. Nature
Biotechnology. 2016; 34:78–83.
Han X, Liu Z, Jo MC, Zhang K, Li Y, Zeng Z, Li N, Zu Y, Qin L. CRISPR-Cas9 delivery to hard-to-
transfect cells via membrane deformation. Science Advances. 2015; 1:e1500454. [PubMed:
26601238]
Author Manuscript

Heckl D, Kowalczyk MS, Yudovich D, Belizaire R, Puram RV, McConkey ME, Thielke A, Aster JC,
Regev A, Ebert BL. Generation of mouse models of myeloid malignancy with combinatorial
genetic lesions using CRISPR-Cas9 genome editing. Nature Biotechnology. 2014; 32:941–946.
Heigwer F, Kerr G, Boutros M. E-CRISP: fast CRISPR target site identification. Nature Methods.
2014; 11:122–123. [PubMed: 24481216]
High KA. The gene therapy journey for hemophilia: are we there yet? Blood. 2012; 120:4482–4487.
[PubMed: 22829631]
Hoban MD, Orkin SH, Bauer DE. Genetic treatment of a molecular disorder: gene therapy approaches
to sickle cell disease. Blood. 2016; 127:839–848. [PubMed: 26758916]
Hou P, Chen S, Wang S, Yu X, Chen Y, Jiang M, Zhuang K, Ho W, Hou W, Huang J, Guo D. Genome
editing of CXCR4 by CRISPR/cas9 confers cells resistant to HIV-1 infection. Scientific Reports.
2015; 5:15577. [PubMed: 26481100]
Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S, Agarwala V, Li Y, Fine EJ, Wu X, Shalem O,
Cradick TJ, Marraffini LA, Bao G, Zhang F. DNA targeting specificity of RNA-guided Cas9
Author Manuscript

nucleases. Nature Biotechnology. 2013; 31:827–832.


Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for genome
engineering. Cell. 2014; 157:1262–1278. [PubMed: 24906146]
Hu W, Kaminski R, Yang F, Zhang Y, Cosentino L, Li F, Luo B, Alvarez-Carbonell D, Garcia-Mesa Y,
Karn J, Mo X, Khalili K. RNA-directed gene editing specifically eradicates latent and prevents
new HIV-1 infection. Proceedings of the National Academy of Sciences of the United States of
America. 2014; 111:11461–11466. [PubMed: 25049410]
Huang X, Wang Y, Yan W, Smith C, Ye Z, Wang J, Gao Y, Mendelsohn L, Cheng L. Production of
gene-corrected adult beta globin protein in human erythrocytes differentiated from patient iPSCs

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 17

after genome editing of the sickle point mutation. Stem Cells. 2015; 33:1470–1479. [PubMed:
25702619]
Author Manuscript

Hwang WY, Fu Y, Reyon D, Maeder ML, Tsai SQ, Sander JD, Peterson RT, Yeh JR, Joung JK.
Efficient genome editing in zebrafish using a CRISPR-Cas system. Nature Biotechnology. 2013;
31:227–229.
Ishino Y, Shinagawa H, Makino K, Amemura M, Nakata A. Nucleotide sequence of the iap gene,
responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of
the gene product. Journal of Bacteriology. 1987; 169:5429–5433. [PubMed: 3316184]
Jaako P, Flygare J, Olsson K, Quere R, Ehinger M, Henson A, Ellis S, Schambach A, Baum C, Richter
J, Larsson J, Bryder D, Karlsson S. Mice with ribosomal protein S19 deficiency develop bone
marrow failure and symptoms like patients with Diamond-Blackfan anemia. Blood. 2011;
118:6087–6096. [PubMed: 21989989]
Jackson EL, Olive KP, Tuveson DA, Bronson R, Crowley D, Brown M, Jacks T. The differential
effects of mutant p53 alleles on advanced murine lung cancer. Cancer Research. 2005; 65:10280–
10288. [PubMed: 16288016]
Jansen R, Embden JD, Gaastra W, Schouls LM. Identification of genes that are associated with DNA
Author Manuscript

repeats in prokaryotes. Molecular Microbiology. 2002; 43:1565–1575. [PubMed: 11952905]


Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-
guided DNA endonuclease in adaptive bacterial immunity. Science. 2012; 337:816–821. [PubMed:
22745249]
Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nature
Reviews: Molecular Cell Biology. 2013; 14:49–55. [PubMed: 23169466]
Kaelin WG Jr. Molecular biology. Use and abuse of RNAi to study mammalian gene function. Science.
2012; 337:421–422. [PubMed: 22837515]
Kaminski R, Chen Y, Fischer T, Tedaldi E, Napoli A, Zhang Y, Karn J, Hu W, Khalili K. Elimination
of HIV-1 Genomes from Human T-lymphoid Cells by CRISPR/Cas9 Gene Editing. Scientific
Reports. 2016; 6:22555. [PubMed: 26939770]
Kampa-Schittenhelm KM, Salitzky O, Akmut F, Illing B, Kanz L, Salih HR, Schittenhelm MM.
Dronabinol has preferential antileukemic activity in acute lymphoblastic and myeloid leukemia
with lymphoid differentiation patterns. BMC Cancer. 2016; 16:25. [PubMed: 26775260]
Author Manuscript

King A, Shenoy S. Evidence-based focused review of the status of hematopoietic stem cell
transplantation as treatment of sickle cell disease and thalassemia. Blood. 2014; 123:3089–3094.
quiz 3210. [PubMed: 24511087]
Koike-Yusa H, Li Y, Tan EP, Velasco-Herrera Mdel C, Yusa K. Genome-wide recessive genetic
screening in mammalian cells with a lentiviral CRISPR-guide RNA library. Nature Biotechnology.
2014; 32:267–273.
Konermann S, Brigham MD, Trevino AE, Joung J, Abudayyeh OO, Barcena C, Hsu PD, Habib N,
Gootenberg JS, Nishimasu H, Nureki O, Zhang F. Genomescale transcriptional activation by an
engineered CRISPR-Cas9 complex. Nature. 2015; 517:583–588. [PubMed: 25494202]
Koo T, Lee J, Kim JS. Measuring and reducing off-target activities of programmable nucleases
including CRISPR-Cas9. Molecules and Cells. 2015; 38:475–481. [PubMed: 25985872]
Kottemann MC, Smogorzewska A. Fanconi anaemia and the repair of Watson and Crick DNA
crosslinks. Nature. 2013; 493:356–363. [PubMed: 23325218]
Lander ES. The heroes of CRISPR. Cell. 2016; 164:18–28. [PubMed: 26771483]
Lettre G, Sankaran VG, Bezerra MA, Araujo AS, Uda M, Sanna S, Cao A, Schlessinger D, Costa FF,
Author Manuscript

Hirschhorn JN, Orkin SH. DNA polymorphisms at the BCL11A, HBS1L-MYB, and beta-globin
loci associate with fetal hemoglobin levels and pain crises in sickle cell disease. Proceedings of the
National Academy of Sciences of the United States of America. 2008; 105:11869–11874.
[PubMed: 18667698]
Liang P, Xu Y, Zhang X, Ding C, Huang R, Zhang Z, Lv J, Xie X, Chen Y, Li Y, Sun Y, Bai Y,
Songyang Z, Ma W, Zhou C, Huang J. CRISPR/Cas9-mediated gene editing in human
tripronuclear zygotes. Protein Cell. 2015; 6:363–372. [PubMed: 25894090]
Maddalo D, Manchado E, Concepcion CP, Bonetti C, Vidigal JA, Han YC, Ogrodowski P, Crippa A,
Rekhtman N, de Stanchina E, Lowe SW, Ventura A. In vivo engineering of oncogenic

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 18

chromosomal rearrangements with the CRISPR/Cas9 system. Nature. 2014; 516:423–427.


[PubMed: 25337876]
Author Manuscript

Maeder ML, Gersbach CA. Genome-editing technologies for gene and cell therapy. Molecular
Therapy. 2016; 24:430–446. [PubMed: 26755333]
Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, Norville JE, Church GM. RNA-guided
human genome engineering via Cas9. Science. 2013a; 339:823–826. [PubMed: 23287722]
Mali P, Aach J, Stranges PB, Esvelt KM, Moosburner M, Kosuri S, Yang L, Church GM. CAS9
transcriptional activators for target specificity screening and paired nickases for cooperative
genome engineering. Nature Biotechnology. 2013b; 31:833–838.
Malina A, Mills JR, Cencic R, Yan Y, Fraser J, Schippers LM, Paquet M, Dostie J, Pelletier J.
Repurposing CRISPR/Cas9 for in situ functional assays. Genes & Development. 2013; 27:2602–
2614. [PubMed: 24298059]
Mansour SL, Thomas KR, Capecchi MR. Disruption of the proto-oncogene int-2 in mouse embryo-
derived stem cells: a general strategy for targeting mutations to non-selectable genes. Nature.
1988; 336:348–352. [PubMed: 3194019]
Marraffini LA, Sontheimer EJ. CRISPR interference limits horizontal gene transfer in staphylococci by
Author Manuscript

targeting DNA. Science. 2008; 322:1843–1845. [PubMed: 19095942]


McIntosh J, Lenting PJ, Rosales C, Lee D, Rabbanian S, Raj D, Patel N, Tuddenham EG, Christophe
OD, McVey JH, Waddington S, Nienhuis AW, Gray JT, Fagone P, Mingozzi F, Zhou SZ, High KA,
Cancio M, Ng CY, Zhou J, Morton CL, Davidoff AM, Nathwani AC. Therapeutic levels of FVIII
following a single peripheral vein administration of rAAV vector encoding a novel human factor
VIII variant. Blood. 2013; 121:3335–3344. [PubMed: 23426947]
Miyajima C, Itoh Y, Inoue Y, Hayashi H. Positive regulation of interleukin-2 expression by a
pseudokinase, tribbles 1, in activated T cells. Biological & Pharmaceutical Bulletin. 2015;
38:1126–1133. [PubMed: 26235576]
Mojica FJ, Juez G, Rodriguez-Valera F. Transcription at different salinities of Haloferax mediterranei
sequences adjacent to partially modified PstI sites. Molecular Microbiology. 1993; 9:613–621.
[PubMed: 8412707]
Mojica FJ, Ferrer C, Juez G, Rodriguez-Valera F. Long stretches of short tandem repeats are present in
the largest replicons of the Archaea Haloferax mediterranei and Haloferax volcanii and could be
involved in replicon partitioning. Molecular Microbiology. 1995; 17:85–93. [PubMed: 7476211]
Author Manuscript

Mojica FJ, Diez-Villasenor C, Garcia-Martinez J, Soria E. Intervening sequences of regularly spaced


prokaryotic repeats derive from foreign genetic elements. Journal of Molecular Evolution. 2005;
60:174–182. [PubMed: 15791728]
Moreau P, Attal M, Facon T. Frontline therapy of multiple myeloma. Blood. 2015; 125:3076–3084.
[PubMed: 25838345]
Moscou MJ, Bogdanove AJ. A simple cipher governs DNA recognition by TAL effectors. Science.
2009; 326:1501. [PubMed: 19933106]
Nathwani AC, Tuddenham EG, Rangarajan S, Rosales C, McIntosh J, Linch DC, Chowdary P, Riddell
A, Pie AJ, Harrington C, O’Beirne J, Smith K, Pasi J, Glader B, Rustagi P, Ng CY, Kay MA, Zhou
J, Spence Y, Morton CL, Allay J, Coleman J, Sleep S, Cunningham JM, Srivastava D, Basner-
Tschakarjan E, Mingozzi F, High KA, Gray JT, Reiss UM, Nienhuis AW, Davidoff AM.
Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. New England Journal
of Medicine. 2011; 365:2357–2365. [PubMed: 22149959]
Navarro JM, Touzart A, Pradel LC, Loosveld M, Koubi M, Fenouil R, Le Noir S, Maqbool MA,
Author Manuscript

Morgado E, Gregoire C, Jaeger S, Mamessier E, Pignon C, Hacein-Bey-Abina S, Malissen B, Gut


M, Gut IG, Dombret H, Macintyre EA, Howe SJ, Gaspar HB, Thrasher AJ, Ifrah N, Payet-Bornet
D, Duprez E, Andrau JC, Asnafi V, Nadel B. Site- and allele-specific polycomb dysregulation in T-
cell leukaemia. Nature Communications. 2015; 6:6094.
Nelles DA, Fang MY, O’Connell MR, Xu JL, Markmiller SJ, Doudna JA, Yeo GW. Programmable
RNA tracking in live cells with CRISPR/Cas9. Cell. 2016; 165:488–496. [PubMed: 26997482]
Nienhuis AW. Development of gene therapy for blood disorders: an update. Blood. 2013; 122:1556–
1564. [PubMed: 23843498]

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 19

Niu Y, Shen B, Cui Y, Chen Y, Wang J, Wang L, Kang Y, Zhao X, Si W, Li W, Xiang AP, Zhou J, Guo
X, Bi Y, Si C, Hu B, Dong G, Wang H, Zhou Z, Li T, Tan T, Pu X, Wang F, Ji S, Zhou Q, Huang
Author Manuscript

X, Ji W, Sha J. Generation of gene-modified cynomolgus monkey via Cas9/RNA-mediated gene


targeting in one-cell embryos. Cell. 2014; 156:836–843. [PubMed: 24486104]
O’Connell MR, Oakes BL, Sternberg SH, East-Seletsky A, Kaplan M, Doudna JA. Programmable
RNA recognition and cleavage by CRISPR/Cas9. Nature. 2014; 516:263–266. [PubMed:
25274302]
Olson, S. International Summit on Human Gene Editing: A Global Discussion: MEETING IN BRIEF.
In: Olson, S., editor. International Summit on Human Gene Editing: A Global Discussion. National
Academies Press (US); Washington (DC): 2016. p. 3-8.
Osborn MJ, Gabriel R, Webber BR, DeFeo AP, McElroy AN, Jarjour J, Starker CG, Wagner JE, Joung
JK, Voytas DF, von Kalle C, Schmidt M, Blazar BR, Tolar J. Fanconi anemia gene editing by the
CRISPR/Cas9 system. Human Gene Therapy. 2015; 26:114–126. [PubMed: 25545896]
Park CY, Kim DH, Son JS, Sung JJ, Lee J, Bae S, Kim JH, Kim DW, Kim JS. Functional correction of
large factor VIII gene chromosomal inversions in Hemophilia A patient-derived iPSCs using
CRISPR-Cas9. Cell Stem Cell. 2015; 17:213–220. [PubMed: 26212079]
Author Manuscript

Pattanayak V, Lin S, Guilinger JP, Ma E, Doudna JA, Liu DR. High-throughput profiling of off-target
DNA cleavage reveals RNA-programmed Cas9 nuclease specificity. Nature Biotechnology. 2013;
31:839–843.
Peng R, Lin G, Li J. Potential Pitfalls of CRISPR/Cas9-mediated genome editing. FEBS Journal. 2015;
283:1218–1231. [PubMed: 26535798]
Pennisi E. SCIENCE AND SOCIETY. Gene drive turns mosquitoes into malaria fighters. Science.
2015; 350:1014. [PubMed: 26612928]
Pinder J, Salsman J, Dellaire G. Nuclear domain ‘knock-in’ screen for the evaluation and identification
of small molecule enhancers of CRISPR-based genome editing. Nucleic Acids Research. 2015;
43:9379–9392. [PubMed: 26429972]
Pourcel C, Salvignol G, Vergnaud G. CRISPR elements in Yersinia pestis acquire new repeats by
preferential uptake of bacteriophage DNA, and provide additional tools for evolutionary studies.
Microbiology. 2005; 151:653–663. [PubMed: 15758212]
Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, Lim WA. Repurposing CRISPR
as an RNA-guided platform for sequence-specific control of gene expression. Cell. 2013;
Author Manuscript

152:1173–1183. [PubMed: 23452860]


Raab MS, Podar K, Breitkreutz I, Richardson PG, Anderson KC. Multiple myeloma. Lancet. 2009;
374:324–339. [PubMed: 19541364]
Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-
Cas9 system. Nature Protocols. 2013; 8:2281–2308. [PubMed: 24157548]
Rathe SK, Moriarity BS, Stoltenberg CB, Kurata M, Aumann NK, Rahrmann EP, Bailey NJ, Melrose
EG, Beckmann DA, Liska CR, Largaespada DA. Using RNA-seq and targeted nucleases to
identify mechanisms of drug resistance in acute myeloid leukemia. Scientific Reports. 2014;
4:6048. [PubMed: 25116387]
Ratnoff OD, Bennett B. The genetics of hereditary disorders of blood coagulation. Science. 1973;
179:1291–1298. [PubMed: 4568863]
Raya A, Rodriguez-Piza I, Guenechea G, Vassena R, Navarro S, Barrero MJ, Consiglio A, Castella M,
Rio P, Sleep E, Gonzalez F, Tiscornia G, Garreta E, Aasen T, Veiga A, Verma IM, Surralles J,
Bueren J, Izpisua Belmonte JC. Disease-corrected haematopoietic progenitors from Fanconi
Author Manuscript

anaemia induced pluripotent stem cells. Nature. 2009; 460:53–59. [PubMed: 19483674]
Reardon S. Welcome to the CRISPR zoo. Nature. 2016; 531:160–163. [PubMed: 26961640]
Redel BK, Prather RS. Meganucleases revolutionize the production of genetically engineered pigs for
the study of human diseases. Toxicologic Pathology. 2015; 44:428–433. [PubMed: 26516165]
Renneville A, Van Galen P, Canver MC, McConkey M, Krill-Burger JM, Dorfman DM, Holson EB,
Bernstein BE, Orkin SH, Bauer DE, Ebert BL. EHMT1 and EHMT2 inhibition induces fetal
hemoglobin expression. Blood. 2015; 126:1930–1939. [PubMed: 26320100]
Ribeil JA, Arlet JB, Dussiot M, Moura IC, Courtois G, Hermine O. Ineffective erythropoiesis in beta -
thalassemia. Scientific World Journal. 2013; 2013:394295. [PubMed: 23606813]

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 20

Richardson PG, Hideshima T, Mitsiades C, Anderson KC. The emerging role of novel therapies for the
treatment of relapsed myeloma. Journal of the National Comprehensive Cancer Network. 2007;
Author Manuscript

5:149–162. [PubMed: 17335684]


Ricks D. Scientists journey into genomes via CRISPR-Cas9. Journal of the National Cancer Institute.
2015; 10:7. djv352.
Rio P, Banos R, Lombardo A, Quintana-Bustamante O, Alvarez L, Garate Z, Genovese P, Almarza E,
Valeri A, Diez B, Navarro S, Torres Y, Trujillo JP, Murillas R, Segovia JC, Samper E, Surralles J,
Gregory PD, Holmes MC, Naldini L, Bueren JA. Targeted gene therapy and cell reprogramming
in Fanconi anemia. EMBO Molecular Medicine. 2014; 6:835–848. [PubMed: 24859981]
Rouet P, Smih F, Jasin M. Expression of a site-specific endonuclease stimulates homologous
recombination in mammalian cells. Proceedings of the National Academy of Sciences of the
United States of America. 1994; 91:6064–6068. [PubMed: 8016116]
Rudin N, Sugarman E, Haber JE. Genetic and physical analysis of double-strand break repair and
recombination in Saccharomyces cerevisiae. Genetics. 1989; 122:519–534. [PubMed: 2668114]
Saayman SM, Lazar DC, Scott TA, Hart JR, Takahashi M, Burnett JC, Planelles V, Morris KV,
Weinberg MS. Potent and targeted activation of Latent HIV-1 using the CRISPR/dCas9 activator
Author Manuscript

complex. Molecular Therapy. 2016; 24:488–498. [PubMed: 26581162]


Sanchez-Rivera FJ, Jacks T. Applications of the CRISPR-Cas9 system in cancer biology. Nature
Reviews: Cancer. 2015; 15:387–395. [PubMed: 26040603]
Scadden DT, Zon LI, Groopman JE. Pathophysiology and management of HIV-associated hematologic
disorders. Blood. 1989; 74:1455–1463. [PubMed: 2676010]
Seger RA. Modern management of chronic granulomatous disease. British Journal of Haematology.
2008; 140:255–266. [PubMed: 18217895]
Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mikkelsen TS, Heckl D, Ebert BL, Root DE,
Doench JG, Zhang F. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science.
2014; 343:84–87. [PubMed: 24336571]
Shankland KR, Armitage JO, Hancock BW. Non-Hodgkin lymphoma. Lancet. 2012; 380:848–857.
[PubMed: 22835603]
Shi J, Wang E, Milazzo JP, Wang Z, Kinney JB, Vakoc CR. Discovery of cancer drug targets by
CRISPR-Cas9 screening of protein domains. Nature Biotechnology. 2015; 33:661–667.
Author Manuscript

Siggs OM, Popkin DL, Krebs P, Li X, Tang M, Zhan X, Zeng M, Lin P, Xia Y, Oldstone MB, Cornall
RJ, Beutler B. Mutation of the ER retention receptor KDELR1 leads to cell-intrinsic
lymphopenia and a failure to control chronic viral infection. Proceedings of the National
Academy of Sciences of the United States of America. 2015; 112:E5706–E5714. [PubMed:
26438836]
Smith C, Gore A, Yan W, Abalde-Atristain L, Li Z, He C, Wang Y, Brodsky RA, Zhang K, Cheng L,
Ye Z. Whole-genome sequencing analysis reveals high specificity of CRISPR/Cas9 and TALEN-
based genome editing in human iPSCs. Cell Stem Cell. 2014; 15:12–13. [PubMed: 24996165]
Smith C, Abalde-Atristain L, He C, Brodsky BR, Braunstein EM, Chaudhari P, Jang YY, Cheng L, Ye
Z. Efficient and allele-specific genome editing of disease loci in human iPSCs. Molecular
Therapy. 2015; 23:570–577. [PubMed: 25418680]
Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H,
Kurashina K, Hatanaka H, Bando M, Ohno S, Ishikawa Y, Aburatani H, Niki T, Sohara Y,
Sugiyama Y, Mano H. Identification of the transforming EML4-ALK fusion gene in non-small-
cell lung cancer. Nature. 2007; 448:561–566. [PubMed: 17625570]
Author Manuscript

Sollelis L, Ghorbal M, MacPherson CR, Martins RM, Kuk N, Crobu L, Bastien P, Scherf A, Lopez-
Rubio JJ, Sterkers Y. First efficient CRISPR-Cas9-mediated genome editing in Leishmania
parasites. Cellular Microbiology. 2015; 17:1405–1412. [PubMed: 25939677]
Song B, Fan Y, He W, Zhu D, Niu X, Wang D, Ou Z, Luo M, Sun X. Improved hematopoietic
differentiation efficiency of gene-corrected beta-thalassemia induced pluripotent stem cells by
CRISPR/Cas9 system. Stem Cells and Development. 2015; 24:1053–1065. [PubMed: 25517294]
Soulier J. Fanconi anemia. Hematology: The Education Program of the American Society of
Hematology. 2011; 2011:492–497.

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 21

Sun N, Zhao H. Seamless correction of the sickle cell disease mutation of the HBB gene in human
induced pluripotent stem cells using TALENs. Biotechnology and Bioengineering. 2014;
Author Manuscript

111:1048–1053. [PubMed: 23928856]


Tagde A, Rajabi H, Bouillez A, Alam M, Gali R, Bailey S, Tai YT, Hideshima T, Anderson K, Avigan
D, Kufe D. MUC1-C drives MYC in multiple myeloma. Blood. 2016; 127:2587–2597. [PubMed:
26907633]
Taniguchi T, D’Andrea AD. Molecular pathogenesis of Fanconi anemia: recent progress. Blood. 2006;
107:4223–4233. [PubMed: 16493006]
Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt SK, Surosky RT, Giedlin MA, Nichol G,
Holmes MC, Gregory PD, Ando DG, Kalos M, Collman RG, Binder-Scholl G, Plesa G, Hwang
WT, Levine BL, June CH. Gene editing of CCR5 in autologous CD4 T cells of persons infected
with HIV. New England Journal of Medicine. 2014; 370:901–910. [PubMed: 24597865]
Thomas KR, Folger KR, Capecchi MR. High frequency targeting of genes to specific sites in the
mammalian genome. Cell. 1986; 44:419–428. [PubMed: 3002636]
Tsai SQ, Zheng Z, Nguyen NT, Liebers M, Topkar VV, Thapar V, Wyvekens N, Khayter C, Iafrate AJ,
Le LP, Aryee MJ, Joung JK. GUIDE-seq enables genome-wide profiling of off-target cleavage by
Author Manuscript

CRISPR-Cas nucleases. Nature Biotechnology. 2015; 33:187–197.


Uda M, Galanello R, Sanna S, Lettre G, Sankaran VG, Chen W, Usala G, Busonero F, Maschio A,
Albai G, Piras MG, Sestu N, Lai S, Dei M, Mulas A, Crisponi L, Naitza S, Asunis I, Deiana M,
Nagaraja R, Perseu L, Satta S, Cipollina MD, Sollaino C, Moi P, Hirschhorn JN, Orkin SH,
Abecasis GR, Schlessinger D, Cao A. Genome-wide association study shows BCL11A
associated with persistent fetal hemoglobin and amelioration of the phenotype of beta-
thalassemia. Proceedings of the National Academy of Sciences of the United States of America.
2008; 105:1620–1625. [PubMed: 18245381]
Ul Ain Q, Chung JY, Kim YH. Current and future delivery systems for engineered nucleases: ZFN,
TALEN and RGEN. Journal of Controlled Release. 2015; 205:120–127. [PubMed: 25553825]
Urnov FD. Biological techniques: edit the genome to understand it. Nature. 2014; 513:40–41.
[PubMed: 25141180]
Valente LJ, Aubrey BJ, Herold MJ, Kelly GL, Happo L, Scott CL, Newbold A, Johnstone RW, Huang
DC, Vassilev LT, Strasser A. Therapeutic response to nongenotoxic activation of p53 by Nutlin3a
is driven by PUMA-mediated apoptosis in lymphoma cells. Cell Reports. 2016; 14:1858–1866.
Author Manuscript

[PubMed: 26904937]
Valletta S, Dolatshad H, Bartenstein M, Yip BH, Bello E, Gordon S, Yu Y, Shaw J, Roy S, Scifo L,
Schuh A, Pellagatti A, Fulga TA, Verma A, Boultwood J. ASXL1 mutation correction by
CRISPR/Cas9 restores gene function in leukemia cells and increases survival in mouse
xenografts. Oncotarget. 2015; 6:44061–44071. [PubMed: 26623729]
Veres A, Gosis BS, Ding Q, Collins R, Ragavendran A, Brand H, Erdin S, Cowan CA, Talkowski ME,
Musunuru K. Low incidence of off-target mutations in individual CRISPR-Cas9 and TALEN
targeted human stem cell clones detected by whole-genome sequencing. Cell Stem Cell. 2014;
15:27–30. [PubMed: 24996167]
Vlachos A, Klein GW, Lipton JM. The Diamond Blackfan Anemia Registry: tool for investigating the
epidemiology and biology of Diamond-Blackfan anemia. Journal of Pediatric Hematology/
Oncology. 2001; 23:377–382. [PubMed: 11563775]
Vlachos A, Ball S, Dahl N, Alter BP, Sheth S, Ramenghi U, Meerpohl J, Karlsson S, Liu JM, Leblanc
T, Paley C, Kang EM, Leder EJ, Atsidaftos E, Shimamura A, Bessler M, Glader B, Lipton JM,
Author Manuscript

Participants of Sixth Annual Daniella Maria Arturi International Consensus, Conference.


Diagnosing and treating Diamond Blackfan anaemia: results of an international clinical
consensus conference. British Journal of Haematology. 2008; 142:859–876. [PubMed:
18671700]
Vukovic M, Guitart AV, Sepulveda C, Villacreces A, O’Duibhir E, Panagopoulou TI, Ivens A,
Menendez-Gonzalez J, Iglesias JM, Allen L, Glykofrydis F, Subramani C, Armesilla-Diaz A,
Post AE, Schaak K, Gezer D, So CW, Holyoake TL, Wood A, O’Carroll D, Ratcliffe PJ, Kranc
KR. Hif-1alpha and Hif-2alpha synergize to suppress AML development but are dispensable for
disease maintenance. Journal of Experimental Medicine. 2015; 212:2223–2234. [PubMed:
26642852]

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 22

Wang J, Quake SR. RNA-guided endonuclease provides a therapeutic strategy to cure latent
herpesviridae infection. Proceedings of the National Academy of Sciences of the United States of
Author Manuscript

America. 2014; 111:13157–13162. [PubMed: 25157128]


Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW, Zhang F, Jaenisch R. One-step generation of
mice carrying mutations in multiple genes by CRISPR/Casmediated genome engineering. Cell.
2013; 153:910–918. [PubMed: 23643243]
Wang T, Wei JJ, Sabatini DM, Lander ES. Genetic screens in human cells using the CRISPR-Cas9
system. Science. 2014; 343:80–84. [PubMed: 24336569]
Wang X, Wang Y, Wu X, Wang J, Wang Y, Qiu Z, Chang T, Huang H, Lin RJ, Yee JK. Unbiased
detection of off-target cleavage by CRISPR-Cas9 and TALENs using integrase-defective
lentiviral vectors. Nature Biotechnology. 2015; 33:175–178.
Wang Z, Pan Q, Gendron P, Zhu W, Guo F, Cen S, Wainberg MA, Liang C. CRISPR/Cas9-derived
mutations both inhibit HIV-1 replication and accelerate viral escape. Cell Reports. 2016; 15:481–
489. [PubMed: 27068471]
Weatherall DJ. The inherited diseases of hemoglobin are an emerging global health burden. Blood.
2010; 115:4331–4336. [PubMed: 20233970]
Author Manuscript

Wiedenheft B, Lander GC, Zhou K, Jore MM, Brouns SJ, van der Oost J, Doudna JA, Nogales E.
Structures of the RNA-guided surveillance complex from a bacterial immune system. Nature.
2011; 477:486–489. [PubMed: 21938068]
Wu C, Dunbar CE. Stem cell gene therapy: the risks of insertional mutagenesis and approaches to
minimize genotoxicity. Frontiers of Medicine. 2011; 5:356–371. [PubMed: 22198747]
Wu X, Blackburn PR, Tschumper RC, Ekker SC, Jelinek DF. TALEN-mediated genetic tailoring as a
tool to analyze the function of acquired mutations in multiple myeloma cells. Blood Cancer
Journal. 2014; 4:e210. [PubMed: 24813078]
Xie F, Ye L, Chang JC, Beyer AI, Wang J, Muench MO, Kan YW. Seamless gene correction of beta-
thalassemia mutations in patient-specific iPSCs using CRISPR/Cas9 and piggyBac. Genome
Research. 2014a; 24:1526–1533. [PubMed: 25096406]
Xie S, Shen B, Zhang C, Huang X, Zhang Y. sgRNAcas9: a software package for designing CRISPR
sgRNA and evaluating potential off-target cleavage sites. PLoS ONE. 2014b; 9:e100448.
[PubMed: 24956386]
Author Manuscript

Xu P, Tong Y, Liu XZ, Wang TT, Cheng L, Wang BY, Lv X, Huang Y, Liu DP. Both TALENs and
CRISPR/Cas9 directly target the HBB IVS2-654 (C > T) mutation in beta-thalassemia-derived
iPSCs. Scientific Reports. 2015; 5:12065. [PubMed: 26156589]
Xue W, Chen S, Yin H, Tammela T, Papagiannakopoulos T, Joshi NS, Cai W, Yang G, Bronson R,
Crowley DG, Zhang F, Anderson DG, Sharp PA, Jacks T. CRISPR-mediated direct mutation of
cancer genes in the mouse liver. Nature. 2014; 514:380–384. [PubMed: 25119044]
Yang L, Guell M, Byrne S, Yang JL, De Los Angeles A, Mali P, Aach J, Kim-Kiselak C, Briggs AW,
Rios X, Huang PY, Daley G, Church G. Optimization of scarless human stem cell genome
editing. Nucleic Acids Research. 2013; 41:9049–9061. [PubMed: 23907390]
Yang Y, Zhang X, Yi L, Hou Z, Chen J, Kou X, Zhao Y, Wang H, Sun XF, Jiang C, Wang Y, Gao S.
Naive induced pluripotent stem cells generated from beta-thalassemia fibroblasts allow efficient
gene correction with CRISPR/Cas9. Stem Cells Translational Medicine. 2016; 5:8–19. [PubMed:
26676643]
Ye L, Wang J, Beyer AI, Teque F, Cradick TJ, Qi Z, Chang JC, Bao G, Muench MO, Yu J, Levy JA,
Kan YW. Seamless modification of wild-type induced pluripotent stem cells to the natural
Author Manuscript

CCR5Delta32 mutation confers resistance to HIV infection. Proceedings of the National


Academy of Sciences of the United States of America. 2014; 111:9591–9596. [PubMed:
24927590]
Zhang WW, Matlashewski G. CRISPR-Cas9-mediated genome editing in Leishmania donovani. MBio.
2015; 6:e00861. [PubMed: 26199327]
Zhang C, Xiao B, Jiang Y, Zhao Y, Li Z, Gao H, Ling Y, Wei J, Li S, Lu M, Su XZ, Cui H, Yuan J.
Efficient editing of malaria parasite genome using the CRISPR/Cas9 system. MBio. 2014;
5:e01414–01414. [PubMed: 24987097]

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 23

Zhang J, Lieu YK, Ali AM, Penson A, Reggio KS, Rabadan R, Raza A, Mukherjee S, Manley JL.
Disease-associated mutation in SRSF2 misregulates splicing by altering RNA-binding affinities.
Author Manuscript

Proceedings of the National Academy of Sciences of the United States of America. 2015a;
112:E4726–E4734. [PubMed: 26261309]
Zhang Y, Yin C, Zhang T, Li F, Yang W, Kaminski R, Fagan PR, Putatunda R, Young WB, Khalili K,
Hu W. CRISPR/gRNA-directed synergistic activation mediator (SAM) induces specific,
persistent and robust reactivation of the HIV-1 latent reservoirs. Scientific Reports. 2015b;
5:16277. [PubMed: 26538064]
Zhang N, Zhi H, Curtis BR, Rao S, Jobaliya C, Poncz M, French DL, Newman PJ. CRISPR/Cas9-
mediated conversion of human platelet alloantigen allotypes. Blood. 2016a; 127:675–680.
[PubMed: 26634302]
Zhang H, Chen Z, Neelapu SS, Romaguera J, McCarty N. Hedgehog inhibitors selectively target cell
migration and adhesion of mantle cell lymphoma in bone marrow microenvironment. Oncotarget.
2016b; 7:14350–14365. [PubMed: 26885608]
Zhou Y, Zhu S, Cai C, Yuan P, Li C, Huang Y, Wei W. High-throughput screening of a CRISPR/Cas9
library for functional geno-mics in human cells. Nature. 2014; 509:487–491. [PubMed:
Author Manuscript

24717434]
Zhu W, Lei R, Le Duff Y, Li J, Guo F, Wainberg MA, Liang C. The CRISPR/Cas9 system inactivates
latent HIV-1 proviral DNA. Retrovirology. 2015; 12:22. [PubMed: 25808449]
Zou J, Mali P, Huang X, Dowey SN, Cheng L. Site-specific gene correction of a point mutation in
human iPS cells derived from an adult patient with sickle cell disease. Blood. 2011; 118:4599–
4608. [PubMed: 21881051]
Author Manuscript
Author Manuscript

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 24
Author Manuscript
Author Manuscript

Fig 1.
Key discoveries and advances in the development of the CRISPR-Cas9 system. CRISPR,
clustered regularly interspaced short palindromic repeats; Cas, CRISPR-associated; DSBs,
DNA double-stranded breaks; crRNAs: CRISPR RNAs; tracrRNA, trans-activating CRISPR
RNA; sgRNA, single-guide RNA; dCas9, inactive Cas9; RCas9, RNA-targeting Cas9.
Author Manuscript
Author Manuscript

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Zhang and McCarty Page 25
Author Manuscript
Author Manuscript

Fig 2.
Applications of CRISPR-(d)Cas9 in haematological disorders. Hereditary blood disorders,
such as haemoglobin abnormalities, anaemias or coagulation disorders represent ideal
targets for CRISPR-Cas9-mediated gene therapy. Genetic controls of cells with CRISPR-
Cas9 also allow us to delineate the molecular mechanisms underlying the development of
Author Manuscript

blood diseases, and further contribute to the alleviation or cure of human diseases. CRISPR,
clustered regularly interspaced short palindromic repeats; dCas9, inactive Cas9; EBV,
Epstein–Barr virus; HIV, human immunodeficiency virus; HSPCs, haematopoietic stem and
progenitor cells; iPSCs, induced pluripotent stem cells.
Author Manuscript

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Table I

Applications of CRISPR-Cas9 technology in non-cancerous haematological disorders.

Off-target
Targeted genes Cell types Genome editing effects Outcomes/comments References
β-thalassaemia HBB Patient-derived iPSCs HDR-induced correction No Restored HBB expression and (Xie et al, 2014a)
exhibited normal karyotypes
Zhang and McCarty

HBB Patient-derived iPSCs HDR-induced correction No Restored HBB expression and (Song et al,
induced haematopoietic 2015)
differentiation
HBB Patient-derived iPSCs HDR-induced correction More Higher efficiency of CRISPR-Cas9- (Xu et al, 2015)
induced DSBs, but lower efficiency
of homologous gene targeting
compared to TALENs
HBB Patient-derived naïve iPSCs HDR-induced correction No Naïve iPSCs exhibited improved (Yang et al,
gene-correction efficiencies 2016)
BCL11A (enhancer) CD34+ HSPC-derived erythroid NHEJ-induced knockout N/A Resulted in near-complete loss of (Canver et al,
precursor cell line BCL11A expression and induction 2015)
of γ-globin and HbF protein
HBB Human embryos HDR-induced correction Apparent Mosaicism and mutations at non- (Liang et al,
target sites were apparent in the 2015)
edited embryos
Sickle-cell disease HBB Patient-derived iPSCs HDR-induced correction Less Corrected a sickle point mutation (Huang et al,
2015)
Ehmt2 Murine erythroleukaemia cells NHEJ-induced knockout N/A Increased the expression of Hbb-εy (Renneville et al,
and Hbb-βh1 embryonic globin 2015)
genes
Fanconi anaemia FANCC Patient-derived fibroblast cells HDR-induced correction No Corrected a Fanconi anaemia point (Osborn et al,
mutation 2015)
Diamond-Blackfan anaemia tp53 Zebrafish embryos NHEJ-induced knockout Maybe Rescued haematological defects in a (Ablain et al,
zebrafish DBA model 2015)

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Thrombocytopenia ITGB3 Polyploid megakaryocytic cells HDR-induced knock-in No Converted from HPA-1a to HPA-1b (Zhang et al,
and iPSCs 2016a)
CMAH, GGTA1 Pigs NHEJ-induced knockout N/A Revealed different responses of (Butler et al,
porcine organs to human platelets 2016)
Haemophilia F8 Patient-derived iPSCs HDR-induced correction No Rescued the FVIII deficiency in HA (Park et al, 2015)
mice
F9 Mouse HDR-induced knock-in/correction No Identified a novel Y371D mutation (Guan et al,
in F9 gene; targeting Y371D 2016)
mutation could restore haemostasis
in HB mice
Myeloproliferative neoplasm JAK2 Patient-derived iPSCs HDR-induced correction No Specifically targeted the mutant (Smith et al,
allele 2015)
Page 26
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Off-target
Targeted genes Cell types Genome editing effects Outcomes/comments References
B-/T-lymphopenia Kdelr1 Mouse NHEJ-induced knockout N/A Reduced expression of TCR and (Siggs et al,
increased expression of CD44 in 2015)
Kdelr1 mutant mice

DBA, Diamond-Blackfan anaemia; DSBs, double strand breaks; HA, haemophilia A; HB, haemophilia B; HDR, homology-directed repair; HPA, human platelet antigen; iPSCs, induced pluripotent stem
cells; N/A, not applicable; NHEJ, non-homologous end joining; TALENs, transcription activator- like effector nucleases; TCR, T cell receptor.
Zhang and McCarty

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Page 27
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Table II

Applications of CRISPR-Cas9 technology in haematological malignancies.

Off–
target
Targeted genes Cell types Genome editing effects Outcomes/comments References
Lymphoma & Myeloma Trp53 Mouse lymphoma cells NHEJ-induced knockout Limited Established the virus-based CRISPR-Cas9 (Malina et al, 2013)
Zhang and McCarty

mediated ex vivo genome editing in the


lymphoma mouse model
BBC3 (PUMA) Human cancer cell lines NHEJ-induced knockout N/A Impaired treatment responses to NUTLIN3A (Valente et al, 2016)
CXCR4 Human MCL cell lines NHEJ-induced knockout N/A Failed to induce autophagy formation upon (Chen et al, 2016;
treatment Zhang et al, 2016b)
MUC1-C Human MM cell lines NHEJ-induced knockout N/A Downregulated MYC mRNA and protein (Tagde et al, 2016)
expression
MCL1 Human BL cell lines NHEJ-induced knockout N/A Identified tumour-essential genes using inducible (Aubrey et al, 2015)
CRISPR-Cas9
Leukaemia Runx1, etc. Mouse HSPCs NHEJ-induced knockout No Generated the virus-based CRISPR-Cas9 (Heckl et al, 2014)
mediated ex vivo genome editing for modelling of
myeloid malignancies
KMT2C (MLL3) Mouse HSPCs NHEJ-induced knockout N/A KMT2C (MLL3) is a haploinsufficient tumour (Chen et al, 2014)
suppressor in AML
EPAS1 (HIF-2α) Human AML cell lines NHEJ-induced knockout N/A Loss of EPAS1 alone had no impact on human (Vukovic et al, 2015)
AML cells under hypoxic conditions
Ly6c2, etc. Murine AML cell lines NHEJ-induced knockout Little Identified genes that are associated with (Rathe et al, 2014)
cytarabine resistance in AML
Cebpa(enhancer) Murine myeloid cell line HDR-induced knock-in N/A Mutated enhancer reduced C/EBPα expression, (Cooper et al, 2015)
indicating the importance of the enhancer for
myeloid Cebpa gene regulation
SRSF2 Human CML cell line HDR-induced knock-in N/A Mutant SRSF2 misregulated hundreds of splicing (Zhang et al, 2015a)
events, thereby providing a group of mis-spliced

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


isoforms for potential therapeutic targeting
ASXL1 Human CML cell line HDR-induced correction No Restored ASXL1 expression in CML cells and (Valletta et al, 2015)
increased survival in mice xenografts
TAL1 Human T-ALL cell line HDR-induced knock-in N/A Induced a selective epigenetic switch (Navarro et al, 2015)
TRIB1 Human T-ALL cell line NHEJ-induced knockout N/A Loss of TRIB1 reduced interleukin-2 expression (Miyajima et al, 2015)
CNB1, CNB2 Human T-ALL, AML, CML cell NHEJ-induced knockout N/A Resulted in abrogation of pro-apoptotic effects (Kampa-Schittenhelm
lines upon THC treatment et al, 2016)
sgRNAs pool Human myeloid leukaemia cell NHEJ-induced knockout Minimal Initiated sgRNA-mediated screens for whole- (Wang et al, 2014)
lines genome analysis in human cells
sgRNAs pool Murine AML cell line NHEJ-induced knockout Low Discovered 6 known drug targets and 19 novel (Shi et al, 2015)
binding domains in murine AML cells
Page 28
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
AML, acute myeloid leukaemia; BL, Burkitt lymphoma; CML, chronic myeloid leukaemia; HDR, homology-directed repair; HSPCs, haematopoietic stem and progenitor cells; MCL, mantle cell
lymphoma; MM, multiple myeloma; N/A, not applicable; NHEJ, non-homologous end joining; sgRNA, single-guide RNA; T-ALL, T-cell acute lymphoblastic leukaemia; THC, Tetrahydrocannabinol
(Dronabinol).
Zhang and McCarty

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Page 29
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Table III

Applications of CRISPR-(d)Cas9 technology in secondary haematological disorders.

Off-target
Targeted genes Cell types Genome editing effects Outcomes/comments References
CGD CYBB Patient-derived iPSCs HDR-induced correction 35% (21/60) Resulted in the recovery of ROS activity (Flynn et al, 2015)
Zhang and McCarty

HIV infection CCR5 HEK 293T cell line NHEJ-induced knockout No First tested the CRISPR-Cas9 system to knockout (Cho et al, 2013)
CCR5 gene in vitro
CCR5, HBB HEK 293T cell line NHEJ-induced knockout More Resulted in a wide variety of indels and point (Cradick et al, 2013)
mutations
CCR5 Human iPSCs NHEJ-induced knockout and N/A Optimized the process for genome editing in (Yang et al, 2013)
HDR-induced correction human stem cell
CCR5 Human iPSCs HDR-induced knock-in No PiggyBac technology plus CRISPR-Cas9 (Ye et al, 2014)
mediated the natural CCR5 mutation in human
iPSCs
CXCR4 Primary human CD4+ T cells NHEJ-induced knockout No Conferred cell resistance to HIV-1 infection (Hou et al, 2015)
and T cell line
XPO1 HEK 293T cell line HDR-induced knock-in N/A XPO1 mutants failed to affect viral replication (Boons et al, 2015)
upon treatment
HIV-1 (LTR region) Human 293T, Hela and T cell NHEJ-induced indels N/A Restricted transcriptionally active provirus and (Ebina et al, 2013)
lines blocked the expression of latently integrated
provirus
HIV-1 (LTR region) Human microglial, NHEJ-induced indels No Completely excised integrated proviral DNA; (Hu et al, 2014)
promonocytic and T cell lines inactivated viral gene expression and replication
in HIV latent cells
HIV-1 (LTR, pol, rev) HIV-GFP Jurkat cell line NHEJ-induced indels N/A Efficiently mutated and deactivated HIV-1 (Zhu et al, 2015)
proviral DNA in latently infected Jurkat cells
HIV-1 (LTR region) Human T cell line and NHEJ-induced indels No Excised integrated proviral DNA and eliminated (Kaminski et al, 2016)
primary T cells HIV-1 production in infected cells; reduced viral
replication in primary T cells

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


HIV-1 (LTR, pol, rev) SupT1 CD4+ T cell line NHEJ-induced indels Many indels HIV-1 escaped from CRISPR-Cas9-mediated (Wang et al, 2016)
suppression due to the Cas9/sgRNA-derived
mutations in the viral DNA sequence
HIV-1 (LTR region) Human microglial and T cell dCas9-induced activation No Reactivated HIV-1 latent viruses and led to (Zhang et al, 2015b)
lines suicide of the HIV-1-infectd cells
HIV-1 (LTR region) HEK 293T and human T cell dCas9-induced activation No Identified a targeted hotspot for CRISPR-dCas9 (Saayman et al, 2016)
lines activation, leading to potent transcriptional
activation of latent HIV infection
EBV infection EBV BL cell lines NHEJ-induced indels No Led to proliferation arrest and apoptosis in EBV- (Wang & Quake,
infected cells 2014)
Malaria Pysera1, etc. Parasite P. yoelii NHEJ-induced knockout No CRISPR-Cas9 can be applied to modify the (Zhang et al, 2014)
rodent malaria P. yoelii genome
Page 30
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Off-target
Targeted genes Cell types Genome editing effects Outcomes/comments References
Egfp, etc. Parasite P. falciparum NHEJ-induced knockout and No Specific gene knockouts and single-nucleotide (Ghorbal et al, 2014)
HDR-induced knock-in substitutions can be achieved in P. falciparum via
CRISPR-Cas9
AGAP007280, etc. Anopheles gambiae mosquito NHEJ-induced knockout and N/A Gene disruption conferred a recessive female- (Hammond et al,
embryos HDR-induced knock-in sterility phenotype; gene insertion at female- 2016)
fertility loci showed highly efficient gene drive
Zhang and McCarty

activity and can spread in a caged population


m2A10-m1C3 Anopheles stephensi HDR-induced knock-in No Blocked parasite development and modified (Gantz et al, 2015)
mosquito embryos mosquito population
Leishmaniasis PFR2 Parasite L. major NHEJ-induced knockout No Got null parasites in a single round of (Sollelis et al, 2015)
transfection
LdMT, etc. Parasite L. donovani NHEJ-induced knockout and N/A Identified a novel single point mutation caused by (Zhang &
HDR-induced knock-in CRISPR-Cas9 in LdMT; precisely insert a drug Matlashewski, 2015)
selection marker into genome

BL, Burkitt lymphoma; CGD, chronic granulomatous disease; EBV, Epstein–Barr virus; HDR, homology-directed repair; HEK, human embryonic kidney; HIV, human immunodeficiency virus; indels,
insertions or deletions; iPSCs, induced pluripotent stem cells; LTR, long terminal repeat; N/A, not applicable; NHEJ, non-homologous end joining; ROS, reactive oxygen species.

Br J Haematol. Author manuscript; available in PMC 2018 April 06.


Page 31

You might also like