You are on page 1of 16

Adv Exp Med Biol – Cell Biology and Translational Medicine

https://doi.org/10.1007/5584_2018_331
# Springer Nature Switzerland AG 2019

CRISPR/Cas9 for Sickle Cell Disease:


Applications, Future Possibilities,
and Challenges

Selami Demirci, Alexis Leonard, Juan J. Haro-Mora,


Naoya Uchida, and John F. Tisdale

Abstract induced pluripotent stem cells (iPSCs) among


Sickle cell disease (SCD) is an inherited other approaches. Recent discovery of
monogenic disorder resulting in serious CRISPR/Cas9 has not only revolutionized
mortality and morbidity worldwide. Although genome engineering but has also brought the
the disease was characterized more than a cen- possibility of translating these concepts into a
tury ago, there are only two FDA approved clinically meaningful reality. Here we
medications to lessen disease severity, and a summarize genome engineering applications
definitive cure available to all patients with using CRISPR/Cas9, addressing challenges
SCD is lacking. Rapid and substantial progress and future perspectives of CRISPR/Cas9 as a
in genome editing approaches have proven curative option for SCD.
valuable as a curative option given plausibility
to either correct the underlying mutation in Keywords
patient-derived hematopoietic stem/progenitor Gene editing · Gene therapy · Hematopoietic
cells (HSPCs), induce fetal hemoglobin stem cell transplantation ·
expression to circumvent sickling of red Hemoglobinopathies · Programmable
blood cells (RBCs), or create corrected endonucleases

S. Demirci (*)
Cellular and Molecular Therapeutics Branch, NHLBI/
NIDDK, National Institutes of Health, Abbreviations
Bethesda, MD, USA AAV Adeno-associated virus
Cellular and Molecular Therapeutics Branch, National BM Bone marrow
Heart, Lung and Blood Institute, Bethesda, MD, USA Cas9 CRISPR associated protein 9
e-mail: selami.demirci@nih.gov;
CRISPR Clustered regularly interspaced
s.demirci1996@gmail.com
short palindromic repeats
A. Leonard, J. J. Haro-Mora, and N. Uchida
DSB Double strand breaks
Cellular and Molecular Therapeutics Branch, NHLBI/
NIDDK, National Institutes of Health, dCas9 Dead Cas9
Bethesda, MD, USA ddPCR Droplet digital PCR
J. F. Tisdale (*) eSpCas9 Enhanced specificity Streptococcus
Cellular and Molecular Therapeutics Branch, National pyogenes Cas9
Heart, Lung and Blood Institute, Bethesda, MD, USA GVHD Graft-vs-host disease
e-mail: johntis@mail.nih.gov
S. Demirci et al.

HbA Adult hemoglobin gene, SCD encompasses a group of disorders


HbF Fetal hemoglobin with variable clinical phenotypes yet share a com-
HbS Hemoglobin S mon pathophysiologic consequence derived from
HDR Homology directed repair a single monogenic change. The modified β-globin
HLA Human leukocyte antigen gene produces an abnormal hemoglobin S (HbS)
HPFH Hereditary persistence of fetal which rapidly polymerizes in the deoxygenated
globin state altering red blood cell (RBC) rheology and
HPLC High performance liquid lifespan. This single substitution leads to multiple
chromatography downstream effects and devastating clinical
HRI Heme-regulated inhibitor complications including chronic anemia, chronic
HSCT Hematopoietic stem cell inflammation, recurrent vaso-occlusion, acute and
transplantation chronic pain, stroke, organ failure, and early mor-
HSPCs Hematopoietic stem/progenitor tality (Paulukonis et al. 2016).
cells SCD is the most common inherited hemoglo-
HU Hydroxyurea binopathy worldwide, and despite knowledge of
INDELs Insertions/deletions the disorder for over 100 years, it remains a life-
iPSCs Induced pluripotent stem cells limiting disease with few therapeutic options to
LCR Locus control region reduce disease severity. Unlike other more
MUD Matched unrelated donor recently identified molecular disorders that have
NHEJ Non-homologous end-joining benefited from higher federal, foundational, and
OTEs Off-target effects per person funding (Smith et al. 2006; Lobner
PACE Phage-assisted continuous et al. 2013), there are only two FDA approved
evolution medications to lessen disease severity, hydroxy-
PAM Protospacer-adjacent motif urea (HU) (approved for adults in 1998; children
QTL Quantitative trait loci in 2017) and L-glutamine (approved in 2018).
RBCs red blood cells There remains misinformation, poor adherence,
ScCas9 Streptococcus canis Cas9 and a reluctance to prescribe HU despite benefit
SCD Sickle cell disease (Wang et al. 2011; Zimmerman et al. 2007;
shRNAmiR MicroRNA-adapted small hairpin Steinberg et al. 2003; Ware 2010), while insur-
(sh) RNAs ance companies will often not cover the cost of
SpCas9- high fidelity Streptococcus the highly purified form of L-glutamine approved
HF1 pyogenes Cas9 by the FDA. Whereas the two mainstay
TALENs TAL-effector nucleases treatments for SCD, blood transfusions and HU
UCBT Umbilical cord blood do not fully eliminate the consequences of the
transplantation disease, simple public health measures such as
ZFNs Zinc finger nucleases newborn screening, penicillin prophylaxis, and
vaccinations have significantly reduced early
childhood mortality. Between 1979 and 2005,
childhood mortality for children with SCD
decreased by 3% per year; however, a 1% per
1 Introduction
year increase during the same period was
observed for adults (Lanzkron et al. 2013). As
Sickle cell disease (SCD) is an inherited
more than 94% of children with SCD in well-
monogenic disorder characterized by a single sub-
resourced countries now survive until age
stitution on chromosome 11 where glutamic acid is
18, and with an expected rise in birth rate for
replaced by valine in the sixth codon of the
babies with severe hemoglobin disorders to be
β-globin gene. Whether inherited either in a homo-
over 400,000 by 2050 (Piel et al. 2013), disease
zygous state or with another abnormal β-globin
management needs to shift to a two-tiered system
CRISPR Editing in Sickle Cell Disease

addressing acute and chronic disease needs while two major barriers in the cure of SCD: the lack of
simultaneously searching for curative options to suitable donors, and the morbidity and mortality
address the global burden and public health issues associated with GVHD. After decades of scientific
of the disease. Hematopoietic stem cell transplan- progress, gene therapy for the cure of SCD is
tation (HSCT) and gene therapy offer a way to currently in multiple clinical trials with promising
reduce disease burden, improve outcomes and initial results. Potential methods for gene therapy
quality of life for patients with SCD, and poten- in SCD are multiple: (i) addition of therapeutic
tially reduce health care costs over the long term globin such as β-globin or βT87Q-globin to make
(Ballas 2009; Arnold et al. 2017; Saenz and adult hemoglobin (HbA), or γ-globin to enhance
Tisdale 2015; Bhatia et al. 2015). fetal hemoglobin (HbF) levels, (ii) HbF induction
Since the first HSCT in 1984 for a pediatric by editing of globin regulatory elements or knock-
patient with SCD and acute myelogenous leuke- down of HbF repressors, or (iii) direct gene correc-
mia, numerous patients have successfully tion of the SCD mutation with programmable
undergone bone marrow (BM) HSCT with a nucleases. Here we focus on the challenges of
human leukocyte antigen (HLA)-matched sibling CRISPR-Cas9 editing, it’s implications, and future
donor. Whether using a myeloablative or possibilities as a curative option for SCD.
non-myeloablative preparative regimen, greater
than 90% of all patients are cured of SCD with a
BM HSCT (Walters et al. 1996; Hsieh et al. 2014; 2 Genome Editing in SCD
Walters et al. 2001; Gluckman et al. 2017).
Between 1986 and 2013, over 1,000 patients Given the prospect for genotypic and therefore
have received an HLA-matched sibling HSCT phenotypic correction in a monogenic disorder
with a 5-year event free survival and overall sur- like SCD, significant effort has been devoted to
vival of 91.4% and 92.9%, respectively find critical genes/chromosomal areas
(Gluckman et al. 2017). HSCT should be consid- contributing to the pathophysiology of the dis-
ered standard of care when a patient has a clinical ease. Antisickling genes such as wild type
indication and an HLA-matched sibling donor, yet β-globin, modified β-globin (T87Q) which
less than 15% of patients with SCD have an appro- confers additional antisickling properties,
priately matched donor (Walters et al. 2001). Fur- γ-globin or β/γ hybrids have been transferred to
thermore, only 10% of eligible patients have sickle HSPCs using various viral constructs; of
undergone curative HSCT despite patient willing- those, some are currently being tested in clinical
ness to consider HSCT morbidity and mortality at trials for both safety and efficacy (reviewed in
the chance for cure (Chakrabarti and Bareford (Demirci et al. 2018)).
2007). HLA-matched unrelated donor (MUD) Genome editing is desirable as it leads to
transplantation, umbilical cord blood transplanta- permeant removal or correction of a detrimental
tion (UCBT), and haploidentical transplantation mutation, or by the creation of protective
offer more patients the chance for cure, though insertions or deletions. Theoretically, program-
high rates of complications currently limit the mable nucleases create double strand breaks
broad use of these therapies. Such complications, (DSB) at a specific genomic locus followed by
including graft rejection and graft-vs-host disease recruitment of DNA repair mechanism through
(GVHD), are addressed in gene therapy models either non-homologous end-joining (NHEJ) or
where a patient’s autologous hematopoietic stem homology directed repair (HDR) (using homolo-
and progenitor cells (HSPCs) are modified thereby gous sequences found in sister chromatids,
eliminating such complications. homologous chromosomes or extrachromosomal
The premise of gene therapy either by gene donor DNA sequence provided for correction
editing or insertion into autologous HSPCs raises purposes) to the DSB site. Until 5 years ago,
the promise of a safer cure for SCD that is avail- three major nucleases including meganucleases
able to all patients. Such methodology eliminates also known as homing endonucleases (reviewed
S. Demirci et al.

in (Stoddard 2011)), zinc finger nucleases (ZFNs, 6 months after birth. Both HbA and HbF are
reviewed in (Urnov et al. 2010)), and maintained on chromosome 11, with switching
TAL-effector nucleases (TALENs, reviewed in from HbF to adult globin mainly controlled by a
(M Scharenberg et al. 2013)) were introduced powerful upstream enhancer known as the locus
for various genome editing purposes. These tools control region (LCR) that loops to each globin
have been successfully used ex vivo to correct the promoter to activate their expression (Li et al.
SCD mutation and induce fetal globin by editing 2002). After the switch to HbA, HbF is not
regulatory sequences such as promoters or other entirely suppressed, though it is not evenly
regulatory sequences including BCL11A, KLF1 distributed among RBCs. When there is not a
and MYB to circumvent the severity of the muta- genotypic cause for persistence of HbF in all
tion in sickle HSPCs (reviewed in (Tasan et al. RBCs, HbF can be minimal in some cells or
2016)). While these nucleases are highly specific concentrated in specific cells referred to as
thereby diminishing off-target effects (OTEs), F-cells (Demirci et al. 2018).
programming of these enzymes is difficult, time After the initial observation by Janet Watson
consuming, and requires significant expertise. and colleagues that newborn babies do not show
In 2012, Doudna et al. presented a new SCD complications for a certain period due to
genome editing technology (Wiedenheft et al. high levels of HbF in the infant’s blood (Watson
2012; Jinek et al. 2012), referred to as Clustered et al. 1948), more work has been devoted to
regularly interspaced short palindromic repeats increase HbF levels in the adult body. The impor-
(CRISPR)/CRISPR associated protein 9 (Cas9), tant role of elevated HbF for SCD protection was
in which a specific RNA (guide RNA) sequence further confirmed with the reports showing
recognizes the target DNA region of interest and asymptomatic patients with SCD with elevated
directs the effector Cas protein there for editing. HbF as a result of coinheriting hereditary persis-
This strategy not only revolutionized genome tence of fetal globin (HPFH) mutations (Forget
editing strategies but also brought forth the 1998; Stamatoyannopoulos et al. 1975). Such
improved possibility of translation of genome mutations occur either in the form of large
editing approaches to the clinical setting due to deletions in the β-globin gene, or smaller
its advantages: easy to design, highly efficient, deletions/single nucleotide polymorphisms
and inexpensive. Once introduced into target (SNPs) in γ-globin promoter or HbF regulating
cells, CRISPR/Cas9 directed DSBs result in acti- quantitative trait loci (QTL) (Paikari and Sheehan
vation of DNA repair mechanisms. This machin- 2018). In line with these reports, deletion or
ery would lead to either some insertions/deletions inversion of 13.6 kb chromosomal region to
(INDELs), which ideally results in loss-of-func- obtain a HPFH-like phenotype in SCD patient
tion for a given gene, or would repair the DNA derived HSPCs resulted in elevated levels of
break using homology strands if HDR is HbF in erythroblast and ameliorated the ex vivo
activated. In this manner, CRISPR/Cas9 technol- sickling (Antoniani et al. 2018). Similarly, point
ogy can target correction of the SCD mutation or mutations created by CRISPR/Cas9 approach in
induce fetal hemoglobin expression by editing the 115 and  200 clusters of the γ-globin
chromosomal areas controlling its expression promoter, inhibiting the binding of validated
(Fig. 1), yet challenges in the use of this technol- HbF transcriptional repressors BCL11A and LRF
ogy remain surrounding efficiency, safety, and (also known as ZBTB7A), respectively (Wang
delivery. and Thein 2018), de-repressed the expression of
HbF (Martyn et al. 2018; Liu et al. 2018). To
show the applicability of these approaches, ani-
2.1 HbF Induction mal models are required for in vivo evaluations
prior to human studies. Immunodeficient mice are
HbF is the predominant globin type after the first generally used for human cell engraftment studies
trimester of gestation and is replaced by HbA by but are not proper for in vivo erythropoiesis. To
CRISPR Editing in Sickle Cell Disease

Fig. 1 Potential CRISPR/Cas9 applications for sickle cell and iPSCs, and subsequent normal red blood cell deriva-
disease (SCD). The proof-of-principle experiments have tion for transfusion purposes. However, these advances are
proven the possibility of SCD mutation correction and waiting to be addressed by clinical trials to explore the full
fetal hemoglobin (HbF) induction in SCD derived HSCs potential

overcome this problem, Li et al. used a human considered potential candidates, direct targeting
β-globin locus transgenic (β-YAC) mice model to of these factors for HbF induction is challenging
study the in vivo effect of disruption of the repres- as all of them have either broader roles in
sor binding region within the γ-globin promoter non-erythroid linages or have significant roles in
(Li et al. 2018). Along with significant target site normal erythropoiesis. Significant candidates,
distribution which was sustained in the secondary LFR and BCL11A, are validated HbF silencers
transplantation experiments, no hematological (Uda et al. 2008; Menzel et al. 2007), and have
abnormality was seen and pronounced switch been edited in the erythrocyte progenitor cell line
from human β to γ globin expression in RBCs (HUDEP-2) leading to robust HbF expression
of adult mice was noted. (Masuda et al. 2016). BCL11A is important for
Gene edition of transcriptional regulators is an HSPC function (Tsang et al. 2015) and normal
alternative methodology to stimulate rare natu- lymphoid development (Liu et al. 2003), with
rally occurring HPFH mutations to control HbF only one paper demonstrating very low level
expression. Several transcription factors includ- indels and a slight increase in γ-globin expression
ing SCA/TAL1, GATA1 and KLF1 are reported to in a non-human primate model using TALE
be involved in HbF regulation (Sankaran and nuclease mRNA targeting the BCL11A coding
Orkin 2013). While all of them could be sequence with respect to control transplants
S. Demirci et al.

(Humbert et al. 2018). The safety and feasibility to clinically meaningful levels that reverse the
of the BCL11A knockdown is still awaiting to be sickling of RBCs and reverse the disease pheno-
addressed by large animal models with high indel type as seen in patients with HPFH.
ratios and subsequent clinical trials with large
patients cohorts. The first clinical trial launched
in February 2018 uses a lentiviral gene transfer 2.2 SCD Mutation Correction
vector encoding a microRNA-adapted small hair-
pin (sh) RNAs (shRNAmiR) targeting BCL11A in As the pathologic mutation for SCD is already
patients with severe SCD is currently ongoing clearly identified, correction of the SCD mutation
with the first patient demonstrating 23% HbF seems the most difficult but potentially the most
(NCT03282656, Shim et al. 2017; Esrick et al. feasible and promising approach as Cas9 cuts
2018). Recently, Daniel Bauer and colleagues sickle β-globin and this break can be repaired if
have presented a different approach in which a normal β-globin sequence flanked with homol-
they induce comparable levels of HbF in CD34+ ogy arms to the DSB is supplied. Genotypic cor-
cells by targeting the +58 intronic site of the rection appears possible by targeting the specific
BCL11A gene that acts as an erythroid specific locus at the genome and providing the correct
enhancer (Bauer et al. 2013; Canver et al. 2015). sequence for β-globin without the necessity of
They were able to show that while guide RNA exogenous transgene activation.
directed disruption of the enhancer site provided To ensure proper correction, an increasing
substantial reduction in Bcl11a expression in number of researchers are using gene editing
erythrocyte cells leading to elevated HbF expres- technologies for correcting the SCD mutation
sion in mice, it did not affect the expression in in different cell types (Table 1). Most of these
non-erythroid lineages (Smith et al. 2016). The works use the CRISPR/Cas9 system as it has
results were extended to erythroid cells derived shown better correction efficiency and lower
from progenitor cells of patients with OTEs than other gene editing tools such as
β-Thalassemia major (Psatha et al. 2018), TALENs (Bak et al. 2018; Hoban et al.
supporting that this enhancer disruption strategy 2016a). The HSPC source is historically bone
would be favorable for clinical use if it is proven marrow derived CD34+ HSPCs, currently used
safe with preclinical and clinical studies. in the majority of genome editing studies,
With the establishment of guide RNA screen- though recently peripherally mobilized CD34 +
ing models, it has become possible to discover HSPCs using plerixafor has shown promise in
novel genomic sites/genes controlling HbF patients with SCD given safety concerns
expression. In a recent paper, protein kinase regarding granulocyte colony stimulating factor
domain–focused CRISPR/Cas9–based genetic use in these patients. These CD34+ cells can be
screening revealed that heme-regulated inhibitor modified to be infused back into the patient.
HRI (also known as EIF2AK1), an erythroid- However, differences in the cell cycle or the
specific kinase that controls protein translation presence of specific nucleases that might dis-
as an HbF repressor, could be used as a potential rupt the correction pathways used by the cells
candidate for the treatment of hemoglobi- after the DSBs offer overall resistance to suc-
nopathies (Grevet et al. 2018). Using similar cessful gene editing (Lomova et al. 2018). In
methodology, the same group also identified that order to maximize success, the preferred deliv-
SPOP, a substrate adaptor of the CUL3 ubiquitin ery method in these studies is electroporation
ligase complex, as a HbF repressor in both with an Adeno-associated virus (AAV)-6 viral
HUDEP-2 and CD34+ cells (Lan et al. 2018). vector for the delivery of the CRISPR/Cas9
Extending these guide RNA screening strategies system with the donor DNA. For evaluating
to non-coding regions and epigenetics would the correction of the SCD mutation, several
allow identification of stronger candidates or studies analyzed gene editing at the DNA
gene combinations to enhance HbF expression level using either targeted deep sequencing
CRISPR Editing in Sickle Cell Disease

Table 1 Selected sickle cell disease (SCD) mutation correction studies using CRISPR/Cas9
Genome
editing Mouse transplantation
Gene Cell types tool Outcomes/comments experiments References
HBB iPSCs CRISPR/ Correction of the SCD mutation ND Huang
Cas9 detectable at RNA expression and et al.
western blot (2015)
HBB BM CD34+ TALEN Correction of SCD mutation with ND Hoban
CRISPR/ higher performance (7.3% HbA et al.
Cas9 production evaluated by HPLC) (2016a)
using CRISPR/Cas9
HBB BM CD34+ CRISPR/ Correction of SCD mutation Long term engraftment of Dever
Mobilized Cas9 using an anti-sickling β-globin enriched CD34+ edited et al.
CD34+ cDNA donor with a 29% population measured by flow (2016)
efficiency at the RNA expression cytometry in femur BM
level (4–30%)
HBB Mobilized CRISPR/ Correction of SCD mutation up to Long term engraftment ability DeWitt
CD34+ Cas9 33% correction evaluated by of non-enriched CD34+ edited et al.
RNAseq and up to 29.3% HbA cells evaluated in BM (2016)
production evaluated by HPLC (2.3  1.8%) and Spleen
(3.7  1.4%)
HBB iPSCs CRISPR/ Correction of SCD mutation ND Park et al.
Cas9 evaluated by sequencing. No (2017)
functional studies
HBB CD34+ CRISPR/ 9.4–9.6% genome editing ND Wen et al.
selected Cas9 efficiency evaluated by (2017)
from sequencing. Correction also
PBMCs evaluated at protein level by
HPLC
HBB iPSCs CRISPR/ Up to 67.9% correction efficiency ND Li et al.
Cas9 evaluated by sequencing. No (2016)
functional studies
HBB Mobilized CRISPR/ 20% SCD mutation correction Long term engraftment of Magis
CD34+ Cas9 evaluated by RNAseq enriched CD34+ edited cells et al.
evaluated in BM (23.4% (2018)
average)
HBB ESCs CRISPR/ 63% SCD mutation correction ND Martin
iPSCs Cas9 evaluated by nested ddPCR et al.
(2018)
HBB CD34+ CRISPR/ More than 50% HbA evaluated ND Vakulskas
HiFiCas9 by HPLC and nested ddPCR et al.
(2018)
HBB CD34+ CRISPR/ Evaluation of the SCD mutation Levels of long-term engraftment Lomova
Cas9 correction by High-throughput of non-enriched CD34+ edited et al.
sequencing cells up to 2.5% in BM (2018)

(Lomova et al. 2018; Wen et al. 2017) or nested correction of the SCD mutation in the β-globin
droplet digital (dd)PCR (Vakulskas et al. 2018), gene (Hoban et al. 2016a; Vakulskas et al.
while others used more functional studies like 2018; DeWitt et al. 2016).
RNAseq or RNA expression levels (Dever et al. Since the publication in 2008 of a protocol for
2016; DeWitt et al. 2016; Chung et al. 2018; generating human iPSCs from somatic cells, many
Magis et al. 2018) with only three studies using groups have developed protocols for the differenti-
High performance liquid chromatography ation of iPSCs into different cell lineages such as
(HPLC) for measuring protein levels after the hematopoietic cells to become another viable source
S. Demirci et al.

of autologous HSPCs (Fujita et al. 2016; Ferreira editing of long-term engrafting HSPCs are not
et al. 2018; Sugimura et al. 2017). Currently how- yet fully explored. Treating cells with CRISPR/
ever, the hematopoietic cells derived from iPSCs are Cas9 and a β-globin donor might result with cells
primitive rather than definitive hematopoietic cells in their native state (uncorrected), as sickle trait
and are therefore unable to engraft in a xenograft (one allele corrected), as healthy (both alleles
mouse model. The available protocols for the dif- corrected), as β-thalassemia major (both alleles
ferentiation of iPSCs towards HSPCs mainly mimic disrupted), as β-thalassemia trait (one allele
primitive hematopoiesis, which can be noticed corrected and other disrupted), and/or
when the generated HSPCs are differentiated into sickle/β-thalassemia (one allele disrupted) due to
erythroid cells containing mainly ε-globin and NHEJ/HDR machinery of the cells (Esrick and
γ-globin, with very low amounts of β-globin if Bauer 2018). As precise correction in long-term
present at all. In order to realize the available gene HSPCs is not yet efficient and editing results in
editing tools to correct the SCD mutation in iPSCs reduction in engrafting HSPCs (Hoban et al.
for therapeutic purpose, a proper differentiation pro- 2016a; Dever et al. 2016), transplantation of
tocol is needed to produce engraftable HSPCs from mixed culture could be clinically problematic
iPSCs. Such therapy, as with other autologous mod- and possible unintended consequences should be
ification strategies, would ultimately eliminate two addressed before clinical trials.
major hurdles in allogeneic transplantation; rejec-
tion and GVHD in transplantation therapies. In
addition, as low efficiency of correction is a prob-
3 Challenges
lem for HSPC studies, cloning corrected cells from
a bulk iPSC population would allow derivation of a
Genome editing has been the most attractive tool
population with 100% of cells corrected.
for scientists seeking to correct genetic mutations
In addition to the difficulties differentiating
either as gene knockout or knock-in. Conventional
iPSCs towards HSPCs, only one study has
methods for genome engineering, however, are
presented the correction of the SCD mutation in
costly, time-consuming, labor-intensive, and
SCD-derived iPSCs at the RNA and protein levels
require expertise in protein engineering to design
by qPCR and Western blot analyses, respectively
specific nucleases (Roy et al. 2018). On the con-
(Huang et al. 2015), though several groups have
trary, CRISPR/Cas9 genome editing is a system
shown the correction of the SCD mutation at the
that is relatively easier, cheaper and more efficient,
DNA level using nested ddPCR or DNA sequenc-
and is being used in a large variety of model cells
ing (Park et al. 2017; Li et al. 2016; Martin et al.
and species. It has not only led to easier and
2018). Correction of underlying mutation in both
cheaper development of knock-out animal models
CD34+ and iPSCs seems promising, yet while
but has also contributed to the establishment of
significant correction rates are reported in ex vivo
whole-genome screening libraries that identify
conditions, limited corrected human cell engraft-
therapeutic genes/chromosomal regions that may
ment are reported in immunodeficient mouse
directly affect a targeted phenotype. While there is
models (Table 1). While immunodeficient mice
a huge international interest in CRISPR/Cas9-
transplantation models for human cell engraftment
based editing approaches, there is still much to
studies are being widely accepted, it is not clear
improve upon such as the efficiency of cutting
that whether these results completely reflect the
and editing (both NHEJ and HDR), improving
clinical outcome of these approaches. After opti-
specificity, and improving delivery methods.
mization of the correction methodologies, larger
Lastly, there is a world-wide concern about safety,
animal models are necessary to explore the poten-
particularly as it relates to OTEs, that needs to be
tial of the application.
clarified and addressed before transferring this
Though editing of CD34+ cells is possible,
approach into routine clinical care.
multiple genotypic outcomes are possible and
CRISPR Editing in Sickle Cell Disease

3.1 Efficiency of Editing recently generated Cas9 variants with various


PAM compatibilities (including NG, GAA and
The limiting factor for diverse application of a given GAT) using phage-assisted continuous evolution
CRISPR/Cas9 system has been the dependency on (PACE) approach (Hu et al. 2018). But more
a protospacer-adjacent motif (PAM) sequence intriguingly, although extending PAM recognition
flanking the target. For instance, as SCD mutation capacity of Cas9 variants would be assumed to
correction studies need to target a specific chromo- augment OTE (Hu et al. 2018; Tsai et al. 2015),
somal area, there are not many guide RNA options they reported greater DNA specificity for Cas9
for different Cas proteins. Therefore, substantial variants with respect to canonical SpCas9 along
effort has been made to engineer various Cas effec- with lower genome-wide off-target. In a different
tor proteins for the recognition of different PAM approach, Sniper-Cas9 (F539S/M763I/K890 N
sequence (Kleinstiver et al. 2015a; Nishimasu et al. variant) was successfully obtained using directed
2018; Kim et al. 2017). While the introduction of evolution, and characterized with high on-target
19 subtypes of CRISPR systems with various Cas and reduced OTEs (Lee et al. 2018). These studies
effector proteins recognizing different PAM sites illustrate the potential and the need for further
have extended targetable genomic loci (Leenay improvements in targetable loci on the genome
and Beisel 2017), not all of them have been widely for various Cas effector proteins. While improving
studied in terms of efficacy and safety. Therefore, the efficiency, safety should also be parallelly
scientists still tend to use well-established Cas types taken into account to realize the approaches in
(i.e Streptococcus pyogenes Cas9-SpCas9 or Cpf1- routine clinical applications.
Cas12a) in their research. SpCas9 has a PAM rec-
ognition of 5’ NGG 30 , while some other Cas9
orthologs have been reported to require longer 3.2 Potential Immunogenicity
PAM sites (Fonfara et al. 2013; Ran et al. 2015). of Editing Tools or Edited Cells
While these have some advantages over classical
SpCas9, their longer PAM sites restrict their use The ultimate goal of CRISPR technology is to
despite potentially more efficient delivery. For edit mutations related with disorders or control
example, smaller Cas effector proteins such as disease associated gene expressions in patient-
Staphylococcus aureus derived Cas9 (SaCas9) derived specific stem/progenitor cells. However,
with a PAM site of NNGRRT, are more efficient in vivo effects of CRISPR/Cas9 systems have a
for viral delivery systems (Kleinstiver et al. 2015b). lot of unanswered questions. In 2019, there are
To extend the boundaries of targeting range for open clinical trials in the United States and abroad
Cas9 proteins, PAM preference can successfully using CRISPR/Cas9 for a potential treatment of
be altered by targeted mutations to residues near SCD, Thalassemia, HIV-1, and several cancer
the PAM DNA duplex (Anders et al. 2016; Hirano types (https://clinicaltrials.gov/keyword
et al. 2016). CRISPR). Though hope remains for these clinical
Understanding the subunits of Cas effector trials, ex vivo work conducted thus far, demon-
proteins have allowed the modification of PAM strate preliminary data pointing toward possible
specificity. In a recent report, Chatterjee et al. adverse effects of the technology. The first ques-
characterized Streptococcus canis Cas9 (ScCas9) tion is whether guide RNAs or Cas9 itself has any
displaying 50 -NNG-30 PAM, reporting an 89.2% effects on the immune system. To partially
sequence similarity to SpCas9 (Chatterjee et al. address this uncertainty, Kim et al. demonstrated
2018). Structural analysis showed that two distinct that in vitro transcribed guide RNAs with a 5-
0
mutational areas [a positive-charged insertion in -triphosphate group (50 -ppp) leads to cytotoxicity
the REC domain (at 367–376) and a KQ insertion due to the activation of innate immune system in
in the PAM-interacting domain (at 1337 and human and mouse cells (Kim et al. 2018). The
1338)] are responsible for having the specificity authors also reported that removal of triphosphate
for a minimal PAM sequence. Another group has resulted in high mutation rate in primary human
S. Demirci et al.

CD4+ cells thus avoiding the innate immune The second important aspect to reduce OTEs is
system. In a recent pre-print article, Charlesworth to enhance Cas9 specificity. A mutated variant of
et al. showed pre-existing antibodies against Cas9 Cas9, nickase (Cas9n), can only cut a single DNA
derived from Staphylococcus aureus (79%) or strand such that two close recognition sites in the
Streptococcus pyogenes (65%) in a small group DNA are required for a double strand break and
of healthy volunteers (Charlesworth et al. 2018). thus OTEs are drastically reduced (50–1500 fold
In a follow up work performed with 200 blood in human cells) (Ran et al. 2013). However, as
samples, prevalence of antibodies against SaCas9 some single nicks can be converted to double
and SpCas9 were reported to be 10% and 2.5%, strand breaks, this approach was further improved
respectively (Simhadri et al. 2018). While these with introduction of a catalytically inactive Cas9
results are not unexpected, triggering of the (dead (d)Cas9) and Fok1 fusion protein (Tsai et al.
immune system by CRISPR/Cas9 is potentially 2014). In this approach, recognition of guide
problematic and harmful in vivo. While these RNAs by dCas9 brings Fok1 enzyme the close
observations and potential immune response are proximity that is required for active dimerized
awaiting to be addressed by large animal models Fok1 nuclease. While these approaches provided
and clinical studies, Cas9 expression levels, significant reduction in the off-target issues,
delivery methods, vector types in case of trans- requirement for a double recognition site might
duction routes, and target cells populations result in less editing efficiencies, and the necessity
should be optimized in any capacity to diminish for double guide RNA usage might limit viral
a severe immune response. delivery approaches. To therefore keep editing
efficiency high enough for clinical application,
active nucleases are being engineered for higher
3.3 Specificity of Editing specificities. The initial idea for high specificity
nucleases was to decrease the interactions of Cas9
Other than a potential immune response, OTEs with its DNA target to lessen OTEs while keeping
are one of the biggest challenges of CRISPR/ enough energy for on-target recognition. With the
Cas9 system. As Cas9-guide RNA complex can introduction of high fidelity Cas9 (SpCas9-HF1,
recognize sequences with up to 5 mismatched N497A/R661A/Q695A/Q926A) (Kleinstiver et al.
bases (Fu et al. 2013), the possibility of OTE for 2016) and enhanced specificity Cas9 (eSpCas9
a given guide RNA cannot be ignored. A number (1.1), K848A/K1003A/R1060A) (Slaymaker
of advances have been taken to increase the et al. 2016), there are no or significantly reduced
specificity of CRISPR/Cas system, but the OTEs compared to wild type nucleases while
guide RNA design is the first critical process maintaining robust on-target activities. Recently,
for reduction of OTEs. There are vast guide Doudna et al. has published that both SpCas9-HF1
RNA design tools available; of those, newer and eSpCas9(1.1) are trapped in an inactive state
ones include supplementary algorithms when bound to mismatched targets and that the
evaluating on-target cutting efficiency other non-catalytic domain of Cas9, REC3, is responsi-
than selectivity for the target. During the synthe- ble for target recognition and direction of nuclease
sis of guide RNA, additional modifications on activity (Chen et al. 2017). Using these
the guide RNA structure including truncation of observations, they were able to create hyper-
spacer RNA (Fu et al. 2014) and chemical accurate Cas9 variant (HypaCas9) with wide-
modifications (Cromwell et al. 2018) have been range genome specificity without compromising
reported to increase Cas9 endonuclease specific- any detectable OTEs.
ity. In addition, chemical modifications with Recently, several publications have raised
20 -O-methyl 30 phosphorothioate (Hendel et al. appropriate concern about the CRISPR/Cas sys-
2015) and 20 -fluoro-ribose (Rahdar et al. 2015) tem showing unintended consequences such as
improve the editing efficiency via increasing the large deletions, insertions, and rearrangement of
stability of guide RNAs in cells. the chromosome when used in clinical trials
CRISPR Editing in Sickle Cell Disease

(Kosicki et al. 2018; Shin et al. 2017; Adikusuma be critical. Jiang et al. showed that Cas9 protein
et al. 2018). It is not clear that this uncertainty is was at maximum level 6 h after delivering Cas9
going to be elucidated, or is clinically relevant, mRNA and not detectable after 24 h in mice
but it is sensible to urge more pre-clinical studies (Jiang et al. 2017). One way to optimize effi-
addressing these valid safety concerns. ciency would be different delivery times or chem-
ical modifications to provide the stability of
RNAs as was mentioned earlier (Safety section).
3.4 Delivery The RNP complex is another alternative in
which native Cas9 protein and guide RNA form
To apply CRISPR/Cas9 system to a given cell a single complex that is readily active once it is in
type/organism, the structure and vehicle of the the cell. Other than the question of whether native
components should be determined based on foreign protein to the human cells is significantly
requirements for protein amount, exposure time, immunogenic to hinder the potential of RNP
efficiency, and restrictions for OTEs and other usage, the main drawback of this application is
safety issues. For the structure of the system, it that Cas9-guide RNA structure is a relatively
could be (i) integrating/non-integrating viral large complex. Non-viral delivery systems
vectors/plasmids expressing both mRNAs for including electroporation, encapsulation, and
guide RNA and Cas9, (ii) Cas9 mRNA and delivery by modification are trending for not
guide RNA, or (iii) ribonucleoprotein complex only transferring this large cargo but also for
(RNP) constituting Cas9 protein and guide other DNA and RNA systems (reviewed in
RNA. A short time after the discovery that (Glass et al. 2018)). Electroporation, a
CRISPR/Cas9 system could be used in human non-selective delivery method, has been used for
cells for genome editing purposes, viral a long time for various DNA, RNA, and protein
constructs providing continuous expression of transfers through the cell membrane by enlarging
Cas9 and guide RNAs were used to explore this the pores on the cell membrane via a strong
potential. However, while it might be advanta- electric field. While this method is highly efficient
geous for gene editing approaches requiring in transferring Cas9 and guide RNA to HSPCs for
long-term expression, it was also recognized that an aim of correction of SCD mutation (Hoban
sustained expression of guide RNAs and Cas9 et al. 2016a; Dever et al. 2016; Magis et al.
augmented the possibility of mismatch bindings 2018), toxicity and the long-term viability issue
and OTEs (Pattanayak et al. 2013). For precise of electroporation for a clinical setting is still
temporal control of expression, several inducible being questioned. From a clinical point of view,
systems have been presented (Nihongaki et al. huge quantities of Cas9 protein might be required
2015; Zetsche et al. 2015). Using vectoral deliv- for a clinical setting, and purification of
ery in the lab is stable and cheap; however, there endotoxin-free Cas9 protein is not economically
is still ongoing debate about the problems of viral feasible at this time. More industrial work is
systems with the immune system (Yin et al. 2014) warranted to explore feasible ways for GMP
and insertional mutagenesis (Hoban et al. 2016b). grade Cas9 production in order for this technique
An alternative method to plasmids/vectors to be practical in a clinical setting.
carrying Cas9 sequence is the introduction of
mRNA for Cas9 that is translated to active protein
once it is transferred to the cell. While this system 4 Future Perspective
avoids the time needed for transcription for Cas9 and Directions
transferred with plasmids, it is also applicable
only for genome editing approaches doable with While SCD was characterized more than century
transient Cas9 expressions. In addition, as ago, definitive treatment for all patients is not
mRNAs are not as stable as DNAs, delivery currently available given a lack of suitable donors
time of RNAs for Cas9 and guide RNA would for curative HSCT. As monogenic disease, SCD is
S. Demirci et al.

one of the most important candidates for Arnold SD, Brazauskas R, He N, Li Y, Aplenc R, Jin Z,
programmable nucleases, particularly CRISPR/ Hall M, Atsuta Y, Dalal J, Hahn T (2017) Clinical risks
and healthcare utilization of haematopoietic cell trans-
Cas9 due to being cost-effective, easily applicable, plantation for sickle cell disease in the US using
and highly efficient. Proof-of-principle studies merged databases. Haematologica 102
have shown that CRISPR/Cas9 can efficiently be (11):1823–1832. https://doi.org/10.3324/haematol.
used to correct the SCD mutation or induce HbF 2017.169581
Bak RO, Gomez-Ospina N, Porteus MH (2018) Gene
expression in ex vivo cell culture conditions and editing on center stage. Trends Genet 34(8):600–611
mouse models. However, there is still concerns Ballas SK (2009) The cost of health care for patients with
about the safety due to random off-target effect sickle cell disease. Am J Hematol 84(6):320–322
and subtherapeutic efficiency. More work should Bauer DE, Kamran SC, Lessard S, Xu J, Fujiwara Y,
Lin C, Shao Z, Canver MC, Smith EC, Pinello L
be conducted in larger animal models to demon- (2013) An erythroid enhancer of BCL11A subject to
strate the safety of the approach along with optimi- genetic variation determines fetal hemoglobin level.
zation studies in ex vivo conditions. Science 342(6155):253–257
Clinical trials investigating the prospective of Bhatia M, Kolva E, Cimini L, Jin Z, Satwani P, Savone M,
George D, Garvin J, Paz ML, Briamonte C (2015)
CRISPR/Cas9 for SCD are in progress or are Health-related quality of life after allogeneic
starting soon, which will certainly direct the future hematopoietic stem cell transplantation for sickle cell
of this approach. The application itself is disease. Biol Blood Marrow Transplant 21(4):666–672
promising but it is not currently feasible for trans- Canver MC, Smith EC, Sher F, Pinello L, Sanjana NE,
Shalem O, Chen DD, Schupp PG, Vinjamur DS,
lation into routine use especially for less developed Garcia SP (2015) BCL11A enhancer dissection by
counties such as Africa where prevalence of SCD Cas9-mediated in situ saturating mutagenesis. Nature
is high. Additional cost-effective manufacturing 527(7577):192–197
processes for clinical grade guide RNAs and Chakrabarti S, Bareford D (2007) A survey on patient
perception of reduced-intensity transplantation in
Cas9 proteins should be implemented to extend adults with sickle cell disease. Bone Marrow Trans-
the use, and ensure a safer, more efficient product. plant 39(8):447–451
The premise of gene therapy for the cure of SCD is Charlesworth CT, Deshpande PS, Dever DP, Dejene B,
moving closer to reality, though questions and Gomez-Ospina N, Mantri S, Pavel-Dinu M,
Camarena J, Weinberg KI, Porteus MH (2018) Identi-
challenges remain to ensure this as a feasible, fication of pre-existing adaptive immunity to Cas9
safe, and lifelong curative strategy. proteins in humans. BioRxiv:243345. https://doi.org/
10.1101/243345
Conflicts of Interest The authors have no commercial, Chatterjee P, Jakimo N, Jacobson JM (2018) Minimal
proprietary, or financial interest in the products described PAM specificity of a highly similarSpCas9 ortholog.
in this article. Sci Adv 4:eaau0766
Chen JS, Dagdas YS, Kleinstiver BP, Welch MM, Sousa
AA, Harrington LB, Sternberg SH, Joung JK,
Yildiz A, Doudna JA (2017) Enhanced proofreading
governs CRISPR–Cas9 targeting accuracy. Nature 550
References (7676):407–410
Chung JE, Magis W, Vu J, Heo S-J, Wartiovaara K,
Adikusuma F, Piltz S, Corbett MA, Turvey M, McColl Walters MC, Kurita R, Nakamura Y, Boffelli D, Mar-
SR, Helbig KJ, Beard MR, Hughes J, Pomerantz RT, tin DI (2018) CRISPR-Cas9 interrogation of a putative
Thomas PQ (2018) Large deletions induced by Cas9 fetal globin repressor in human erythroid cells.
cleavage. Nature 560(7717):E8–E9 BioRxiv:335729. https://doi.org/10.1101/335729
Anders C, Bargsten K, Jinek M (2016) Structural plasticity Cromwell CR, Sung K, Park J, Krysler AR, Jovel J, Kim
of PAM recognition by engineered variants of the SK, Hubbard BP (2018) Incorporation of bridged
RNA-guided endonuclease Cas9. Mol Cell 61 nucleic acids into CRISPR RNAs improves Cas9
(6):895–902 endonuclease specificity. Nat Commun 9(1):1448
Antoniani C, Meneghini V, Lattanzi A, Felix T, Demirci S, Uchida N, Tisdale JF (2018) Gene therapy for
Romano O, Magrin E, Weber L, Pavani G, El sickle cell disease: an update. Cytotherapy 20
Hoss S, Kurita R (2018) Induction of fetal hemoglobin (7):899–910
synthesis by CRISPR/Cas9-mediated editing of the Dever DP, Bak RO, Reinisch A, Camarena J,
human β-globin locus. Blood 131(17):1960–1973. Washington G, Nicolas CE, Pavel-Dinu M,
https://doi.org/10.1182/blood-2017-10-811505 Saxena N, Wilkens AB, Mantri S (2016) CRISPR/
CRISPR Editing in Sickle Cell Disease

Cas9 β-globin gene targeting in human haematopoietic Nat Biotechnol 33(9):985–989. https://doi.org/10.
stem cells. Nature 539(7629):384–389 1038/nbt.3290
DeWitt MA, Magis W, Bray NL, Wang T, Berman JR, Hirano S, Nishimasu H, Ishitani R, Nureki O (2016)
Urbinati F, Heo S-J, Mitros T, Muñoz DP, Boffelli D Structural basis for the altered PAM specificities of
(2016) Selection-free genome editing of the sickle engineered CRISPR-Cas9. Mol Cell 61(6):886–894
mutation in human adult hematopoietic stem/progeni- Hoban MD, Lumaquin D, Kuo CY, Romero Z, Long J,
tor cells. Science Transl Med 8 Ho M, Young CS, Mojadidi M, Fitz-Gibbon S, Cooper
(360):360ra134–360ra134 AR (2016a) CRISPR/Cas9-mediated correction of the
Esrick EB, Bauer DE (2018) Genetic therapies for sickle sickle mutation in human CD34+ cells. Mol Ther 24
cell disease. Semin Hematol 55(8):76–86 (9):1561–1569
Esrick EB, Brendel C, Manis JP, Armant MA, Negre H, Hoban MD, Orkin SH, Bauer DE (2016b) Genetic treat-
Dansereau C, Ciuculescu MF, Patriarca S, ment of a molecular disorder: gene therapy approaches
Mackinnon B, Daley H (2018) Flipping the switch: to sickle cell disease. Blood 127(7):839–848
initial results of genetic targeting of the fetal to adult Hsieh MM, Fitzhugh CD, Weitzel RP, Link ME, Coles
globin switch in sickle cell patients. Blood 132:1023 WA, Zhao X, Rodgers GP, Powell JD, Tisdale JF
Ferreira AF, Calin GA, Picanço-Castro V, Kashima S, (2014) Nonmyeloablative HLA-matched sibling allo-
Covas DT, de Castro FA (2018) Hematopoietic stem geneic hematopoietic stem cell transplantation for
cells from induced pluripotent stem cells–considering severe sickle cell phenotype. JAMA 312(1):48–56
the role of microRNA as a cell differentiation regula- Hu JH, Miller SM, Geurts MH, Tang W, Chen L, Sun N,
tor. J Cell Sci 131(4):jcs203018 Zeina CM, Gao X, Rees HA, Lin Z (2018) Evolved
Fonfara I, Le Rhun A, Chylinski K, Makarova KS, Cas9 variants with broad PAM compatibility and high
Lecrivain A-L, Bzdrenga J, Koonin EV, Charpentier DNA specificity. Nature 556(7699):57–63
E (2013) Phylogeny of Cas9 determines functional Huang X, Wang Y, Yan W, Smith C, Ye Z, Wang J,
exchangeability of dual-RNA and Cas9 among Gao Y, Mendelsohn L, Cheng L (2015) Production of
orthologous type II CRISPR-Cas systems. Nucleic gene-corrected adult beta globin protein in human
Acids Res 42(4):2577–2590 erythrocytes differentiated from patient iPSCs after
Forget BG (1998) Molecular basis of hereditary persis- genome editing of the sickle point mutation. Stem
tence of fetal hemoglobin. Ann N Y Acad Sci 850 Cells 33(5):1470–1479
(1):38–44 Humbert O, Peterson CW, Norgaard ZK, Radtke S, Kiem
Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung H-P (2018) A nonhuman primate transplantation
JK, Sander JD (2013) High-frequency off-target muta- model to evaluate hematopoietic stem cell gene editing
genesis induced by CRISPR-Cas nucleases in human strategies for β-hemoglobinopathies. Mol Ther
cells. Nat Biotechnol 31(9):822–826 Methods Clin Dev 8:75–86
Fu Y, Sander JD, Reyon D, Cascio VM, Joung JK (2014) Jiang C, Mei M, Li B, Zhu X, Zu W, Tian Y, Wang Q,
Improving CRISPR-Cas nuclease specificity using Guo Y, Dong Y, Tan X (2017) A non-viral CRISPR/
truncated guide RNAs. Nat Biotechnol 32(3):279–284 Cas9 delivery system for therapeutically targeting
Fujita A, Uchida N, Haro-Mora JJ, Winkler T, Tisdale J HBV DNA and pcsk9 in vivo. Cell Res 27(3):440–443
(2016) β-globin-expressing definitive erythroid pro- Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA,
genitor cells generated from embryonic and induced Charpentier E (2012) A programmable dual-RNA–
pluripotent stem cell-derived sacs. Stem Cells 34 guided DNA endonuclease in adaptive bacterial immu-
(6):1541–1552 nity. Science 337(6096):816–821. https://doi.org/10.
Glass Z, Lee M, Li Y, Xu Q (2018) Engineering the 1126/science.1225829
delivery system for CRISPR-based genome editing. Kim YB, Komor AC, Levy JM, Packer MS, Zhao KT, Liu
Trends Biotechnol 36(2):173–185 DR (2017) Increasing the genome-targeting scope and
Gluckman E, Cappelli B, Bernaudin F, Labopin M, Volt F, precision of base editing with engineered Cas9-
Carreras J, Simões BP, Ferster A, Dupont S, De La cytidine deaminase fusions. Nat Biotechnol 35
Fuente J (2017) Sickle cell disease: an international (4):371–376
survey of results of HLA-identical sibling Kim S, Koo T, Jee H-G, Cho H-Y, Lee G, Lim D-G, Shin
hematopoietic stem cell transplantation. Blood 129 HS, Kim J-S (2018) CRISPR RNAs trigger innate
(11):1548–1556 immune responses in human cells. Genome Res
Grevet JD, Lan X, Hamagami N, Edwards CR, 28:367–373. https://doi.org/10.1101/gr.231936.117
Sankaranarayanan L, Ji X, Bhardwaj SK, Face CJ, Kleinstiver BP, Prew MS, Tsai SQ, Topkar VV, Nguyen
Posocco DF, Abdulmalik O (2018) Domain-focused NT, Zheng Z, Gonzales AP, Li Z, Peterson RT, Yeh
CRISPR screen identifies HRI as a fetal hemoglobin J-RJ (2015a) Engineered CRISPR-Cas9 nucleases with
regulator in human erythroid cells. Science 361 altered PAM specificities. Nature 523(7561):481–485
(6399):285–290 Kleinstiver BP, Prew MS, Tsai SQ, Nguyen NT, Topkar
Hendel A, Bak RO, Clark JT, Kennedy AB, Ryan DE, VV, Zheng Z, Joung JK (2015b) Broadening the
Roy S, Steinfeld I, Lunstad BD, Kaiser RJ, Wilkens targeting range of Staphylococcus aureus CRISPR-
AB (2015) Chemically modified guide RNAs enhance Cas9 by modifying PAM recognition. Nat Biotechnol
CRISPR-Cas genome editing in human primary cells. 33(12):1293–1298
S. Demirci et al.

Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen M Scharenberg A, Duchateau P, Smith J (2013) Genome
NT, Zheng Z, Joung JK (2016) High-fidelity CRISPR– engineering with TAL-effector nucleases and alterna-
Cas9 nucleases with no detectable genome-wide tive modular nuclease technologies. Curr Gene Ther 13
off-target effects. Nature 529(7587):490–495 (4):291–303
Kosicki M, Tomberg K, Bradley A (2018) Repair of Magis W, DeWitt MA, Wyman SK, Vu JT, Heo S-J, Shao
double-strand breaks induced by CRISPR–Cas9 leads SJ, Hennig F, Romero ZG, Campo-Fernandez B,
to large deletions and complex rearrangements. Nat McNeill M (2018) In vivo selection for corrected
Biotechnol 36(8):765–771 β-globin alleles after CRISPR/Cas9 editing in human
Lan X, Khandros E, Grevet JD, Peslak SA, Bhardwaj S, sickle hematopoietic stem cells enhances therapeutic
Keller CA, Giardine B, Garcia BA, Hardison RC, Shi J potential. BioRxiv:432716. https://doi.org/10.1101/
(2018) Domain-focused CRISPR-Cas9 screen 432716
identifies the E3 ubiquitin ligase substrate adaptor pro- Martin R, Ikeda K, Uchida N, Cromer MK, Nishimura T,
tein SPOP as a novel repressor of fetal hemoglobin. Dever DP, Camarena J, Bak R, Lausten A, Jakobsen
Blood 132:414 MR (2018) Selection-free, high frequency genome
Lanzkron S, Carroll CP, Haywood C Jr (2013) Mortality editing by homologous recombination of human plu-
rates and age at death from sickle cell disease: US, ripotent stem cells using Cas9 RNP and AAV6.
1979–2005. Public Health Rep 128(2):110–116 BioRxiv:252163. https://doi.org/10.1101/252163
Lee JK, Jeong E, Lee J, Jung M, Shin E, Kim Y-h, Lee K, Martyn GE, Wienert B, Yang L, Shah M, Norton LJ,
Jung I, Kim D, Kim S (2018) Directed evolution of Burdach J, Kurita R, Nakamura Y, Pearson RC,
CRISPR-Cas9 to increase its specificity. Nat Commun Funnell AP (2018) Natural regulatory mutations ele-
9(1):3048 vate the fetal globin gene via disruption of BCL11A or
Leenay RT, Beisel CL (2017) Deciphering, communicat- ZBTB7A binding. Nat Genet 50(4):498–503
ing, and engineering the CRISPR PAM. J Mol Biol Masuda T, Wang X, Maeda M, Canver MC, Sher F,
429(2):177–191 Funnell AP, Fisher C, Suciu M, Martyn GE, Norton
Li Q, Peterson KR, Fang X, Stamatoyannopoulos G LJ (2016) Transcription factors LRF and BCL11A
(2002) Locus control regions. Blood 100 independently repress expression of fetal hemoglobin.
(9):3077–3086 Science 351(6270):285–289
Li C, Ding L, Sun C-W, Wu L-C, Zhou D, Pawlik KM, Menzel S, Garner C, Gut I, Matsuda F, Yamaguchi M,
Khodadadi-Jamayran A, Westin E, Goldman FD, Heath S, Foglio M, Zelenika D, Boland A, Rooks H
Townes TM (2016) Novel HDAd/EBV (2007) A QTL influencing F cell production maps to a
reprogramming vector and highly efficient Ad/ gene encoding a zinc-finger protein on chromosome
CRISPR-Cas sickle cell disease gene correction. Sci 2p15. Nat Genet 39(10):1197–1199
Rep 6:30422 Nihongaki Y, Kawano F, Nakajima T, Sato M (2015)
Li C, Psatha N, Sova P, Gil S, Wang H, Kim J, Kulkarni C, Photoactivatable CRISPR-Cas9 for optogenetic
Valensisi C, Hawkins RD, Stamatoyannopoulos G genome editing. Nat Biotechnol 33(7):755–760
(2018) Reactivation of γ-globin in adult β-YAC mice Nishimasu H, Shi X, Ishiguro S, Gao L, Hirano S,
after ex vivo and in vivo hematopoietic stem cell Okazaki S, Noda T, Abudayyeh OO, Gootenberg JS,
genome editing. Blood 131:2915–2928. https://doi. Mori H (2018) Engineered CRISPR-Cas9 nuclease
org/10.1182/blood-2018-03-838540 with expanded targeting space. Science 361
Liu P, Keller JR, Ortiz M, Tessarollo L, Rachel RA, (6408):1259–1262
Nakamura T, Jenkins NA, Copeland NG (2003) Paikari A, Sheehan VA (2018) Fetal haemoglobin induc-
Bcl11a is essential for normal lymphoid development. tion in sickle cell disease. Br J Haematol 180
Nat Immunol 4(6):525–532 (2):189–200
Liu N, Hargreaves VV, Zhu Q, Kurland JV, Hong J, Park S, Gianotti-Sommer A, Molina-Estevez FJ,
Kim W, Sher F, Macias-Trevino C, Rogers JM, Kurita Vanuytsel K, Skvir N, Leung A, Rozelle SS, Shaikho
R (2018) Direct promoter repression by BCL11A EM, Weir I, Jiang Z (2017) A comprehensive, ethni-
controls the fetal to adult hemoglobin switch. Cell cally diverse library of sickle cell disease-specific
173(2):430–442. e417 induced Pluripotent stem cells. Stem Cell Rep 8
Lobner K, Lanzkron S, Haywood C (2013) NIH and (4):1076–1085
National Foundation Expenditures for sickle cell dis- Pattanayak V, Lin S, Guilinger JP, Ma E, Doudna JA, Liu
ease and cystic fibrosis are associated with Pubmed DR (2013) High-throughput profiling of off-target
publications and FDA approvals. Blood 122:1739 DNA cleavage reveals RNA-programmed Cas9 nucle-
Lomova A, Clark DN, Campo-Fernandez B, Flores- ase specificity. Nat Biotechnol 31(9):839–843
Bjurström C, Kaufman ML, Fitz-Gibbon S, Wang X, Paulukonis ST, Eckman JR, Snyder AB, Hagar W,
Miyahira EY, Brown D, DeWitt MA (2018) Improving Feuchtbaum LB, Zhou M, Grant AM, Hulihan MM
gene editing outcomes in human hematopoietic stem (2016) Defining sickle cell disease mortality using a
and progenitor cells by temporal control of DNA population-based surveillance system, 2004 through
repair. Stem Cells:1–11. https://doi.org/10.1002/stem. 2008. Public Health Rep 131(2):367–375
2935
CRISPR Editing in Sickle Cell Disease

Piel FB, Hay SI, Gupta S, Weatherall DJ, Williams TN of fetal hemoglobin in blacks and its association with
(2013) Global burden of sickle cell anaemia in children sickle cell trait. Blood 46(5):683–692
under five, 2010–2050: modelling based on Steinberg MH, Barton F, Castro O, Pegelow CH, Ballas
demographics, excess mortality, and interventions. SK, Kutlar A, Orringer E, Bellevue R, Olivieri N,
PLoS Med 10(7):e1001484 Eckman J (2003) Effect of hydroxyurea on mortality
Psatha N, Reik A, Phelps S, Zhou Y, Dalas D, Yannaki E, and morbidity in adult sickle cell anemia: risks and
Levasseur DN, Urnov FD, Holmes MC, benefits up to 9 years of treatment. JAMA 289
Papayannopoulou T (2018) Disruption of the (13):1645–1651
BCL11A erythroid enhancer reactivates fetal hemoglo- Stoddard BL (2011) Homing endonucleases: from micro-
bin in erythroid cells of patients with β-thalassemia bial genetic invaders to reagents for targeted DNA
major. Mol Ther Methods Clin Dev 10:313–326 modification. Structure 19(1):7–15
Rahdar M, McMahon MA, Prakash TP, Swayze EE, Sugimura R, Jha DK, Han A, Soria-Valles C, da Rocha
Bennett CF, Cleveland DW (2015) Synthetic CRISPR EL, Lu Y-F, Goettel JA, Serrao E, Rowe RG,
RNA-Cas9–guided genome editing in human cells. Malleshaiah M (2017) Haematopoietic stem and pro-
Proc Natl Acad Sci 112(51):E7110–E7117 genitor cells from human pluripotent stem cells. Nature
Ran FA, Hsu PD, Lin C-Y, Gootenberg JS, Konermann S, 545(7655):432–438
Trevino AE, Scott DA, Inoue A, Matoba S, Zhang Y Tasan I, Jain S, Zhao H (2016) Use of genome-editing
(2013) Double nicking by RNA-guided CRISPR Cas9 tools to treat sickle cell disease. Hum Genet 135
for enhanced genome editing specificity. Cell 154 (9):1011–1028
(6):1380–1389 Tsai SQ, Wyvekens N, Khayter C, Foden JA, Thapar V,
Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz Reyon D, Goodwin MJ, Aryee MJ, Joung JK (2014)
AJ, Zetsche B, Shalem O, Wu X, Makarova KS (2015) Dimeric CRISPR RNA-guided FokI nucleases for
In vivo genome editing using Staphylococcus aureus highly specific genome editing. Nat Biotechnol 32
Cas9. Nature 520(7546):186–191 (6):569–576
Roy B, Zhao J, Yang C, Luo W, Xiong T, Li Y, Fang X, Tsai SQ, Zheng Z, Nguyen NT, Liebers M, Topkar VV,
Gao G, Singh CO, Madsen L (2018) CRISPR/cascade Thapar V, Wyvekens N, Khayter C, Iafrate AJ, Le LP
9-mediated genome editing-challenges and (2015) GUIDE-seq enables genome-wide profiling of
opportunities. Front Genet 9:240–252 off-target cleavage by CRISPR-Cas nucleases. Nat
Saenz C, Tisdale JF (2015) Assessing costs, benefits, and Biotechnol 33(2):187–197
risks in chronic disease: taking the long view. Biol Tsang JC, Yu Y, Burke S, Buettner F, Wang C,
Blood Marrow Transplant 21(7):1149–1150 Kolodziejczyk AA, Teichmann SA, Lu L, Liu P
Sankaran VG, Orkin SH (2013) The switch from fetal to (2015) Single-cell transcriptomic reconstruction
adult hemoglobin. Cold Spring Harb Perspect Med 3 reveals cell cycle and multi-lineage differentiation
(1):a011643 defects in Bcl11a-deficient hematopoietic stem cells.
Shim G, Kim D, Park GT, Jin H, Suh S-K, Oh Y-K (2017) Genome Biol 16(1):178
Therapeutic gene editing: delivery and regulatory Uda M, Galanello R, Sanna S, Lettre G, Sankaran VG,
perspectives. Acta Pharmacol Sin 38(6):738–753 Chen W, Usala G, Busonero F, Maschio A, Albai G
Shin HY, Wang C, Lee HK, Yoo KH, Zeng X, Kuhns T, (2008) Genome-wide association study shows
Yang CM, Mohr T, Liu C, Hennighausen L (2017) BCL11A associated with persistent fetal hemoglobin
CRISPR/Cas9 targeting events cause complex and amelioration of the phenotype of β-thalassemia.
deletions and insertions at 17 sites in the mouse Proc Natl Acad Sci 105(5):1620–1625
genome. Nat Commun 8:15464 Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD
Simhadri VL, McGill J, McMahon S, Wang J, Jiang H, (2010) Genome editing with engineered zinc finger
Sauna ZE (2018) Prevalence of pre-existing antibodies nucleases. Nat Rev Genet 11(9):636–646
to CRISPR-associated nuclease Cas9 in the US popu- Vakulskas CA, Dever DP, Rettig GR, Turk R, Jacobi AM,
lation. Mol Ther Methods Clin Dev 10:105–112 Collingwood MA, Bode NM, McNeill MS, Yan S,
Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, Camarena J (2018) A high-fidelity Cas9 mutant deliv-
Zhang F (2016) Rationally engineered Cas9 nucleases ered as a ribonucleoprotein complex enables efficient
with improved specificity. Science 351(6268):84–88 gene editing in human hematopoietic stem and progen-
Smith LA, Oyeku SO, Homer C, Zuckerman B (2006) itor cells. Nat Med 24(8):1216–1224
Sickle cell disease: a question of equity and quality. Walters MC, Patience M, Leisenring W, Eckman JR, Scott
Pediatrics 117(5):1763–1770 JP, Mentzer WC, Davies SC, Ohene-Frempong K,
Smith EC, Luc S, Croney DM, Woodworth MB, Greig Bernaudin F, Matthews DC (1996) Bone marrow
LC, Fujiwara Y, Nguyen M, Sher F, Macklis JD, Bauer transplantation for sickle cell disease. N Engl J Med
DE (2016) Strict in vivo specificity of the Bcl11a 335(6):369–376
erythroid enhancer. Blood 128(19):2338–2342. Walters M, Patience M, Leisenring W, Rogers Z,
https://doi.org/10.1182/blood-2016-08-736249 Aquino V, Buchanan G, Roberts I, Yeager A, Hsu L,
Stamatoyannopoulos G, Wood W, Papayannopoulou T, Adamkiewicz T (2001) Stable mixed hematopoietic
Nute P (1975) A new form of hereditary persistence chimerism after bone marrow transplantation for sickle
S. Demirci et al.

cell anemia. Biol Blood Marrow Transplant 7 Wen J, Tao W, Hao S, Zu Y (2017) Cellular function
(12):665–673 reinstitution of offspring red blood cells cloned from
Wang X, Thein SL (2018) Switching from fetal to adult the sickle cell disease patient blood post CRISPR
hemoglobin. Nat Genet 50(4):478–480 genome editing. J Hematol Oncol 10(1):119
Wang WC, Ware RE, Miller ST, Iyer RV, Casella JF, Wiedenheft B, Sternberg SH, Doudna JA (2012)
Minniti CP, Rana S, Thornburg CD, Rogers ZR, RNA-guided genetic silencing systems in bacteria
Kalpatthi RV (2011) Hydroxycarbamide in very and archaea. Nature 482(7385):331–338
young children with sickle-cell anaemia: a multicentre, Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin JR,
randomised, controlled trial (BABY HUG). Lancet 377 Anderson DG (2014) Non-viral vectors for gene-based
(9778):1663–1672 therapy. Nat Rev Genet 15(8):541–555
Ware RE (2010) How I use hydroxyurea to treat young Zetsche B, Volz SE, Zhang F (2015) A split-Cas9 archi-
patients with sickle cell anemia. Blood 115 tecture for inducible genome editing and transcription
(26):5300–5311. https://doi.org/10.1182/blood-2009- modulation. Nat Biotechnol 33(2):139–142
Blood Zimmerman SA, Schultz WH, Burgett S, Mortier NA,
Watson J, Stahman AW, Bilello FP (1948) The signifi- Ware RE (2007) Hydroxyurea therapy lowers
cance of the paucity of sickle cells in newborn Negro transcranial Doppler flow velocities in children with
infants. Obstet Gynecol Surv 3(6):819–820 sickle cell anemia. Blood 110(3):1043–1047

You might also like