ArticlePDF Available

CRISPR/Cas9 applications in gene therapy for primary immunodeficiency diseases

Authors:
  • Cimeio Therapeutics

Abstract and Figures

Primary immunodeficiency diseases (PIDs) encompass a range of diseases due to mutations in genes that are critical for immunity. Haploinsufficiency and gain-of-function mutations are more complex than simple loss-of-function mutations; in addition to increased susceptibility to infections, immune dysregulations like autoimmunity and hyperinflammation are common presentations. Hematopoietic stem cell (HSC) gene therapy, using integrating vectors, provides potential cure of disease, but genome-wide transgene insertions and the lack of physiological endogenous gene regulation may yet present problems, and not applicable in PIDs where immune regulation is paramount. Targeted genome editing addresses these concerns; we discuss some approaches of CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas system applicable for gene therapy in PIDs. Preclinical repair of gene mutations and insertion of complementary DNA restore endogenous gene regulation and they have shown very promising data for clinical application. However, ongoing studies to characterize off-target genotoxicity, careful donor designs to ensure physiological expression, and maneuvers to optimize engraftment potential are critical to ensure successful application of this next-gen targeted HSC gene therapy.
Content may be subject to copyright.
Review Article
CRISPR/Cas9 applications in gene therapy for
primary immunodeficiency diseases
Suk See De Ravin and Julie Brault
Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, U.S.A.
Correspondence: Suk See De Ravin (sderavin@niaid.nih.gov)
Primary immunodeciency diseases (PIDs) encompass a range of diseases due to mutations
in genes that are critical for immunity. Haploinsufciency and gain-of-function mutations are
more complex than simple loss-of-function mutations; in addition to increased susceptibility
to infections, immune dysregulations like autoimmunity and hyperinammation are common
presentations. Hematopoietic stem cell (HSC) gene therapy, using integrating vectors, pro-
vides potential cure of disease, but genome-wide transgene insertions and the lack of
physiological endogenous gene regulation may yet present problems, and not applicable in
PIDs where immune regulation is paramount. Targeted genome editing addresses these
concerns; we discuss some approaches of CRISPR (Clustered Regularly Interspaced Short
Palindromic Repeats)/Cas system applicable for gene therapy in PIDs. Preclinical repair of
gene mutations and insertion of complementary DNA restore endogenous gene regulation
and they have shown very promising data for clinical application. However, ongoing studies
to characterize off-target genotoxicity, careful donor designs to ensure physiological expres-
sion, and maneuvers to optimize engraftment potential are critical to ensure successful
application of this next-gen targeted HSC gene therapy.
Introduction
Primary immunodeciency diseases (PIDs) encompass a range of disorders due to mutations in genes
that are critical for immune function and result in absent, non-functioning or abnormally active
immune cells. Over 300 genes have been associated with PIDs [1] and, with the availability of high-
throughput next-generation sequencing, the list is rapidly growing longer. Better characterization
of genetic defects provides the opportunity to design specic gene correction strategies for the treatment
of PIDs, thus paving the way to precision medicine. In addition to delineating mutation types in PIDs,
such as point mutations, chromosomal alterations (deletions, insertions, translocations) or copy number
variation, mechanistic understanding of the genetic end products is also crucial as loss-of-function
(LOF), gain-of-function (GOF), haploinsufciency, or dominant negative phenotypes probably require
different approaches tailored to each scenario when contemplating treatment strategies [2].
PIDs most commonly present with increased susceptibility to infections and evidence of immune dys-
regulation that may present as autoimmunity or hyperinammation.Manyrecentlydened PIDs
include GOF mutations in genes that affect cellular signaling pathways or immune regulation. We will
discuss the versatile applications of targeted genome editing using the CRISPR (Clustered Regularly
Interspaced Short Palindromic Repeats)/Cas system to both LOF PIDs and more challenging GOF PIDs.
The clinical manifestations of the PID may inuence the approaches for treatment of the disease.
Previously well-described LOF mutations, such as those leading to severe combined immunodeciency
(SCID) due to IL2RG defects (X-linked SCID or SCID-X1) or adenosine deaminase deciency (ADA
SCID), or chronic granulomatous disease (CGD), present early with severe infections. In contrast, some
recently described PIDs with abnormalities in cellular immune signaling pathways may be diagnosed
later in life with signicant immune dysregulation presenting as autoimmunity [3,4]. Clinical manage-
ment of PIDs with increased susceptibility to infections has included lifelong anti-microbial prophylaxis,
Version of Record published:
23 May 2019
Received: 19 March 2019
Revised: 9 May 2019
Accepted: 13 May 2019
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology 1
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
immunoglobulin supplementation and, generally, consideration of hematopoietic stem cell (HSC) transplant
(HSCT) from a human leukocyte antigen-matched allogeneic donor. For patients with defects in immune cell
signaling, medical intervention can be tailored to enhance regulatory pathways or to inhibit inammatory signals
[58]. The design of genome-editing approaches to treat PID, therefore, requires consideration of the specic
causative genetic mutation as well as the clinical presentation and condition of the patient.
The rst successful allogeneic HSCT of a SCID-X1 patient over 50 years ago demonstrated that replacement with
normal HSCs can cure this PID [9]. Although potentially curative, allogeneic HSCT still suffers from the following
problems: potential for graft versus host disease, graft rejection, toxicities related to chemotherapeutic or immunosup-
pressive agents and most critically, the availability of matched allogeneic donor grafts [10,11]. HSC gene therapy,
however, can circumvent some of these problems by correction of autologous HSCs by insertion of a therapeutic
gene into the chromosomal DNA of the HSC that engrafts and differentiates ideally with normal function. The inser-
tion of the therapeutic gene relies on integrating vectors that deliver and insert one or more copies into the HSC.
Using γ-retroviral vectors, initial HSC gene therapy trials demonstrated signicant clinical benets, especially where
the insertion conferred a survival advantage to gene-corrected cells [1214]. Unfortunately, lymphoid and/or
myeloid insertional mutagenesis was observed in some PIDs including SCID-X1, CGD, and WiskottAldrich syn-
drome (WAS) [1517]. More recently, human lentivirus vectors have been used, with improved safety features and
an integration pattern in the chromosomal DNA that does not favor promoters and enhancers, to reduce the risk of
oncogenic activating integrations [18]. Replication-decient lentivectors have been examined in clinical trials for the
treatment of PIDs, including SCID-X1, CGD, WAS, ADASCID with some positive results [1922]. Lentivector
gene therapy products for ADASCID, SCID-X1, and CGD are soon likely to become approved drugs [23,24]. The
unequivocal clinical benets following HSC gene therapy provide further incentive to design better strategies for spe-
cicgenemodication of autologous HSCs for transplant to treat PIDs. Lentivector HSC gene therapy may yet suffer
from two disadvantages (the potential long-term genotoxicity of the quasi-random genome-wide vector integrations
and the lack of physiological regulation of gene expression) that may be eliminated by specically targeted
gene-editing techniques.
Targeted genetic modication may offer the greatest potential yet for denitively curing the widest spectrum
of PIDs. Recent advances in nucleases have made possible precise genome editing by the use of programmable
endonucleases including zinc nger nucleases, transcription activator-like effector nucleases, or megaTALs and
more recently, the CRISPR/Cas9 system [2531]. These genome engineering systems share an ability to recog-
nize and target specic genomic DNA sequences and create a site-specic double-strand break (DSB) or a
Figure 1. Overview of the gene-editing therapy for primary PIDs.
Autologous hematopoietic stem and immune cells apheresed from PID patients processed in vitro with CRISPR/Cas9
gene-editing tools may be administered fresh or cryopreserved following validation of product for correction efciency, safety
and hematopoietic function before infusion.
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology2
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
single-strand nickin the DNA, thus stimulating the endogenous cellular repair machinery to bring about the
genomic alteration. The main repair pathways are the error-prone non-homologous end joining (NHEJ) that
results in insertions and/or deletions (indels) at the site of the break, and the homology-directed repair (HDR)
that uses the sister chromatid or an exogenous DNA donor template to repair the break [32]. In the context of
PIDs, correction of genetic defects most frequently relies on HDR, although the NHEJ repair pathway can be
harnessed to functionally inactivate a targeted locus (knock-out) by introducing small indels.
We focus our discussion on the use of the CRISPR/Cas9 system as a representative of this class of
gene-editing tools and on the genome-editing strategies that can be applied to potential cures of specic PIDs.
Although gene-edited HSC may be the best denitive therapy, for some PIDs, for example in CD40L deciency
causing Hyper IgM syndrome, correction of primary immune cells, e.g. T-cells, may be sufcient for control
of disease, either in isolation or to improve control of disease to reduce risks of denitive transplant, whether
allogeneic or autologous gene-modied HSCs (Figure 1)[33,34].
CRISPR/Cas9 tools for gene editing
In addition to its versatility, the wide availability of CRISPR/Cas9 reagents has resulted in the rapid develop-
ment of new tools, as summarized in Figure 2, and growing knowledge leading to creative solutions to solve
the range of genetic mutations in PIDs. We present here some tools available to date and illustrate their use in
specic PIDs.
The CRISPR/Cas9 system relies on the recognition of a specic genomic DNA target complementary to a short
guide RNA sequence (gRNA, generally 20 nucleotides), a protospacer, adjacent to an appropriate protospacer adja-
centmotif(PAM).TheformationofanRNADNA pairing induces a conformational change in the Cas9 nuclease
that promotes its nuclease activity and the generation of a double-strand cleavage 3 bp upstream of the PAM
sequence [35]. The rst described Cas9 nuclease, a class 2 type II CRISPR/Cas system derived from Streptococcus
pyogenes (SpCas9), recognizes a 50-NGG-30PAM sequence theoretically found every 812 bp in the genome
[26,31,36]. Excellent works in the eld have created and uncovered Cas9 variants requiring different PAM sequences
[35,37] and nucleases from alternate bacterial species [Streptococcus thermophilus (PAM = 50-NNAGAAW-30and
Figure 2. CRISPR/Cas9 system for targeted genome editing.
The steps for CRISPR/Cas9 gene-editing tool selection from an array of CRISPR/Cas9 components are summarized (a), delivery
methods (b), and approaches harnessing HDR for gene correction (c). LHA: left homology arm; RHA: right homology arm.
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology 3
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
50-NGGNG-30for CRISPR1 and 3, respectively, every 64 bp) or Neisseria meningitidis (PAM = 50-NNNNGATT-30
every 128 bp)] [25,38,39] have greatly extended the scope of targetable genomic sites.
The safety prole of the CRISPR/Cas system can be improved in many ways, most simply with the use of
engineered SpCas9 High Fidelity variants [40,41]. Alternatively, studies using Cas9 nickases (Cas9n; D10A or
H840A mutations) [26,42] or catalytically inactive Cas9 (fCas9) or dead Cas9 (dCas9) fused to a FokI nuclease
domain [4345] reported improved specicity by creating a DSB between the two binding sites. Cas9n can
improve specicity by up to 1,500-fold compared with the wild-type (WT) Cas9 with fewer off-target cleavage
events [46,47]. Also reported to possess greater specicities are Cas orthologs of the class 2 type V CRISPR/Cas
system like Cpf1/Cas12a and Cas12b nucleases from other bacterial species [4852] compared with S. pyogenes
Cas9. Ingenious base editing approaches that convert mutants to WT sequence without DNA cleavage are
thought to limit indels at target sites. Current strategy fuses Cas9 with a cytidine or adenosine deaminase to
cause an irreversible conversion of C:G to T:A or A:T to G:C, respectively, without DNA cleavage [5356]. The
current limitations of this approach are the correction efciency and a relatively limited scope of targetable
mutations. Furthermore, more sensitive approaches revealed substantial off-target effects, with a 20-fold higher
off-target frequency with cytosine base editors compared with adenosine base editors [57].
The next critical component of the CRISPR genome-editing system is the guide. Initially, the length of guide
RNAs was around 100 nucleotides (spacer of 20 nucleotides complementary to the target genomic sequence) and
they were generally produced by in vitro transcription. The use of single-guide RNA (sgRNA) with a shorter
spacer of 17, 18, or 19 nucleotides improves specicity but may reduce the efciency of genome-editing rates [58],
and requires careful evaluation for each PID. Production of gRNA by in vitro transcription may be challenging for
some groups and prohibitory for large-scale clinical grade manufacturing. Synthetic sgRNAs have potential advan-
tages including chemical modications that increase the stability of the sgRNA and genome-editing frequencies
[59,60]. Recently, many have found Cas9 nuclease complexed with sgRNA to form a ribonucleoprotein (RNP) that
supports earlier editing activity with less associated toxicity an attractive option (Figure 2a)[40,61,62].
Since the donor for the gene-editing system determines the end product, this component of the
genome-editing system requires careful attention and will be discussed next in greater detail in the context of
specic mutations and PIDs. Donors that have been used include single-stranded oligodeoxynucleotide
(ssODN) [63,64], long ssDNA and linear or circular dsDNA [6568], or complementary DNA (cDNA) within
an engineered virus cassette such as integrase-defective lentivirus (IDLV; transient expression and weak integra-
tion capability) [69] or recombinant adeno-associated virus (rAAV), depending on the size of the donor
(Figure 2). The rAAV as a donor delivery tool has low immunogenic potential and has high infectious capabil-
ity even in non-dividing cells and low integration rates in the genome of the host cell [7072]. They are gener-
ally capable of delivering up to 4.55 kb but by using two co-transduced AAV vectors, DNA cargos of up to
6.5 kb have been reported [73]. For targeting human HSCs, rAAV serotype 6 was shown to have the greatest
afnity, and a mutant AAV6 capsid provided further improvement [7477]. In general, donors should have
homology arms of 400 bp, codon optimization of the cDNA and modication of the PAM sequence to prevent
re-cutting of the donor or gene-modied cells (Figure 2c)[75].
CRISPR/Cas9 approaches for HSC genome editing in PIDs
Two main considerations in determining a genome-editing strategy for PIDs are rstly, the mutation size
(single or few nucleotides versus large deletions) and frequency (mutation hotspots versus disease-causing
mutations distributed throughout the gene) that affect the choice of donor template, and secondly, the func-
tional effect of the mutation. While LOF mutation can be boosted by correction of 1 or 2 alleles, diseases that
result from heterozygote mutations are challenging due to the risks of unintended cuts on the normal allele
that results in disease, or the residual effects of uncorrected mutant allele. Ideally, in haploinsufciency muta-
tions, one needs correction of the mutant allele without disrupting the other allele. However, in GOF muta-
tions, the goal is to knock-out or repair the mutant allele while avoiding unintended cuts of the other allele.
Great work in the design of broader choices for nucleases and with improved specicity may provide the neces-
sary tools necessary for correction of these more challenging mutations [37,78,79].
LOF mutations that involve a single or few nucleotides are amenable to the use of a short oligonucleotide donor
to repair the mutation resulting in a normal genetic sequence, i.e. gene repair(Figure 2c). About two-thirds of
human genetic diseases arise from single-base mutations. To correct such mutations in situ,thesimplestapproach
is to use a short donor template, such as a ssODN [8085] although linear or plasmid dsDNA donors have also
been used [86,87]. Various studies have evaluated the effects of the length, sense, and symmetry of the donor
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology4
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
ssODN as well as chemical modications (such as phosphorothioate linkages both 50and 30ends) to induce higher
HDR efciency [60,81,86,88]. This strategy was employed to repair a relatively frequent mutation in the CYBB
gene that encodes for gp91
phox
, a critical catalytic subunit of the phagocyte NADPH oxidase complex, which
results in X-linked chronic granulomatous disease (X-CGD). Using a chemically modied 100 bp ssODN with
symmetrical homology arms, delivered by electroporation with Cas9 mRNA and in vitro transcribed sgRNA, in
vitro correction rates of 3040% of HSC were achieved that, upon transplantation into NSG immunodecient
mice, resulted in 1520% stable corrected cells at 5 months [80]. Subsequent improvements to this system have
included the use of chemically modied synthetic guides and non-targeting donor oligos that improved in vitro
correction rates to 5060% (unpublished). This level of correction is sufcient to reverse the disease phenotype of
X-CGD as it relates to susceptibility of infections only. In contrast with some gene therapy approaches using
exogenous promoters, the gene-edited corrected cells restored normal physiological regulation with normal
amounts of protein expression in the appropriate mature differentiated phagocytes and functionally normal
amounts of NADPH oxidase activity. Although consideration of individual mutations seems daunting at present, it
is possible that with the rapid progress in producing synthetic guides and donor oligonucleotides, an individual
mutation-specic repair system can be optimized for treatment within a few months for patients not needing emer-
gency intervention. Optimization of this approach may provide proof-of-concept that gene-repaired HSCs can
restore HSC compartment for gene therapy in other diseases, where a single LOF mutation may account for most
or all patients, such as sickle cell disease [89]orp47
phox
-decient CGD [90]. Of note, the level of correction for
reversal of disease phenotype for different LOF mutations likely varies and is important to dene in the preclinical
studies to provide the rationale for treatment. For example, diseases, such as SCID-X1 in which functional cells
have survival advantage, require lower levels of (10%) correction to reverse disease phenotype [91].
While short donor oligos may be ideal for small mutations, repair of large deletions or insertions is not pos-
sible with this approach. Instead, an alternative strategy to address large mutations or simply address mutations
located widely in the gene is to insert a functional cDNA into a targeted site, generally downstream of the
endogenous promoter to restore physiological regulation (Figure 2c). Targeted integration (TI) near the start
site could potentially address > 95% of the mutations. This universalor one-size-ts-allapproach has been
shown to achieve excellent rates of TI of the cDNA, or portions of genes at a targeted site (a knock-in) where
the nal genomic sequence is altered to provide a functional gene product under the control of the endogenous
promoter. Although the donors can be delivered many ways, including IDLV [83,92], the best outcomes are
observed following delivery by rAAV. Very encouraging preclinical data pave the way for imminent clinical
application for treatment of SCID-X1 due to IL2RG gene defects [9193] and CD40L for XHIM [34,94]. Other
PIDs reporting encouraging results following targeted knock-ins include BTK for X-linked agammaglobuline-
mia (XLA) [95] and CYBB for X-CGD [76,96,97].
Recent advances in optimizing the CRISPR/Cas system have generally resulted in excellent TI rates over 50% in
vitro. Critical factors to be taken into consideration to ensure successful clinical outcomes for gene-edited HSC gene
therapy include an evaluation of the functional end product ( protein expressed) as well as the engraftment capabilities
following CRISPR/Cas9 gene editing. In addition to endogenous gene regulation, factors such as the Kozak consensus
sequence, intronic regulatory elements, and polyadenylation length ( poly(A)) should be taken into consideration
when designing donors (Figure 2). To illustrate, we evaluated the knock-inapproach targeting the insertion of a
partial cDNA (CYBB Exon 713) using our previously reported guide targeting CYBB E7 c.676 with the ssODN gene
repair correction described above [98]. Expression of the donor encoding CYBB Exons 7 to 13 is regulated by the
endogenous CYBB promoter. We observed signicantly reduced amounts of gp91
phox
protein per cell produced that
was independent of TI rates (>5070%). Substituting the poly(A) with a longer poly(A) signicantly improved the
amount of protein produced. Similarly, the addition of Woodchuck Hepatitis Virus Post Transcriptional Regulatory
Element (WPRE) signicantly increases expression of genes delivered by viral vectors [94,99,100]. Furthermore, there
may be critical regulatory elements in introns lost with cDNAs that are devoid of all introns. This was illustrated by
Sweeney et al. who showed that TI in CYBB E1 with a CYBB E113 cDNA failed to restore the gp91
phox
expression,
while the expression was restored when the donor was inserted in Exon 2, suggesting the presence of critical regula-
tory elements in Intron 1 [98]. This problem may be circumvented by the retention of the critical intronic sequences
that may also allow the expression of isoforms under the physiological regulation of the promoter.
The engraftment capabilities of gene-edited HSCs need careful evaluation. We previously observed good engraft-
ment rates of 3040% following transplants of 1 million CRISPR/Cas9/ssODN gene-repaired HSCs per mouse
(NOD SCIDyc-decient strain) conditioned with 20 mg/kg busulfan. Our current experiences with AAV-mediated
gene knock-inapproaches have observed lower engraftment rates in general (unpublished). Several groups have
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology 5
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
reported immune responses to viruses such as interferon-inducible antiviral factors or activation of p53-mediated
DNA damage response induced by DSBs which may alter stemness of the HSCs and impair the ability to engraft
[101104]. Better understanding of the mechanisms involved may lead to discovery of agents that may counteract
these responses, such as Cyclosporin H [105,106], or agents that enhance HSC self-renewal such as
pyrimido-indole derivative UM171 and aryl hydrocarbon receptor antagonist StemRegenin 1 [77,107,108].
Since HDR occurs most efciently during the S/G2 phases of the cell cycle when DNA is relaxed and more access-
ible [109], there are molecules targeting factors critical for DNA repair mechanisms that may also promote HDR.
Synchronizing Cas9 expression with activation of the HDR machinery may also enhance templated repair [110112]
or by the use of inhibitors of key enzymes of the NHEJ pathway like SCR7, an inhibitor of the DNA ligase IV
DNA-binding domain, or an inhibitor of 53BP1 (i53) [113,114]. A 5070% increase in gene correction rates in vitro
(both gene repair and targeted gene knock-in) was achieved with the use of the inhibitor of 53BP1 (unpublished).
Long-term safety and efcacy studies of NSG transplants for gene-modied HSCs with the aid of i53 are ongoing.
GOF PIDs include recently characterized PIDs, such as phosphoinositide-3-kinase δ-syndrome (PIK3CD), or
cytotoxic lymphocyte antigen-4 (CTLA-4) haploinsufciency with pathological increased cellular signaling and
hyperactivation, resulting in clinical disease [115119]. In PIDs due to GOF mutations, transplants from allo-
geneic donors have shown that strategies that result in chimeric populations of gene-mutant cells and genetic-
ally normal cells will likely not reverse patients clinical phenotype. Strategies to cause new mutant alleles may
result in mutant phenotype, while TI into the normal allele will not alleviate the end product due to the
mutant allele. There are challenging strategies that at least in theory may revert phenotype, such as targeted
knock-out of the mutant allele and replacing the mutant allele with a TI of normal cDNA. Single nucleotide
GOF mutations may also benet from specic base editing that corrects the specic mutation without risks of
DSB that potentially creates new mutants.
Conclusion
In conclusion, the CRISPR/Cas9 technology has transformed biomedical research and preclinical data strongly
support the translation of specic genome editing for the treatment of a variety of primary immunodeciencies.
There are critical ongoing research efforts to develop more sensitive techniques to detect chromosomal altera-
tions due to unintended effects of endonucleases. Thorough evaluation of engraftment capabilities of
gene-edited HSCs particularly with AAV-delivered donors and detailed analysis of functional outcomes for
each corrected gene defect is essential for effective correction of PIDs. Despite some concerns for off-target
indels, the potential for circumventing genome-wide vector integration concerns, and to restore physiological
control under endogenous promoters should greatly improve the safety of HSC gene therapy. Continued pro-
gress in gene-editing tools will hopefully provide broader range of choices for correcting currently challenging
PIDs, especially those due to GOF mutations.
Summary
HSC gene therapy is clinically benecial for some PIDs.
Targeted genome editing can avoid genome-wide integration and constitutive expression of
transgene.
CRISPR/Cas approach to repair gene mutations or insertion of cDNA at endogenous gene
locus can restore physiological regulation.
Preclinical CRISPR/Cas9 studies for some LOF mutation PID are ready for translation to the
clinic.
Future direction CRISPR/Cas9 is versatile and may provide innovative tools and strategies
to address the range of genetic mutations in PIDs, such as haploinsufciency, dominant
negative or GOF mutations.
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology6
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
Abbreviations
ADA, adenosine deaminase deciency; cDNA, complementary DNA; CGD, chronic granulomatous disease;
CRISPR, Clustered Regularly Interspaced Short Palindromic Repeats; DSB, double-strand break;
GOF, gain-of-function; HDR, homology-directed repair; HSC, Hematopoietic stem cell; LOF, loss-of-function;
NHEJ, non-homologous end joining; PAM, protospacer adjacent motif; PIDs, Primary immunodeciency
diseases; rAAV, recombinant adeno-associated virus; RNP, ribonucleoprotein; SCID, severe combined
immunodeciency; sgRNA, single-guide RNA; ssODN, single-stranded oligodeoxynucleotide; TI, targeted
integration; WAS, WiskottAldrich syndrome; WT, wild-type; X-CGD, X-linked chronic granulomatous disease.
Funding
This work is supported by the Intramural Research Program of the National Institutes of Allergy and Infectious
Diseases and the National Institutes of Health.
Acknowledgements
We owe our gratitude to our patients who have volunteered and supported us in our research, our colleagues
at the Dowling Apheresis Center, and the Center for Cell Engineering at the Clinical Center for support with
research stem cell collections. We also thank Dr Kol Zarember for the helpful comments for the review.
Competing Interests
The Authors declare that there are no competing interests associated with the manuscript.
References
1 Picard, C., Bobby Gaspar, H., Al-Herz, W., Bousha, A., Casanova, J.L., Chatila, T. et al. (2018) International Union of Immunological Societies: 2017
Primary Immunodeciency Diseases Committee Report on inborn errors of immunity. J. Clin. Immunol. 38,96128 https://doi.org/10.1007/
s10875-017-0464-9
2 Lodish, H., Berk, A., Zipursky, S.L., Matsudaira, P., Baltimore, D. and Darnell, J. (2000) Genetic Analysis in Cell Biology, 4th edn, W. H. Freeman,
New York
3 Allenspach, E. and Torgerson, T.R. (2016) Autoimmunity and primary immunodeciency disorders. J. Clin. Immunol. 36, S57S67
https://doi.org/10.1007/s10875-016-0294-1
4 Schmidt, R.E., Grimbacher, B. and Witte, T. (2018) Autoimmunity and primary immunodeciency: two sides of the same coin? Nat. Rev. Rheumatol. 14,
718 https://doi.org/10.1038/nrrheum.2017.198
5 Lee, S., Moon, J.S., Lee, C.R., Kim, H.E., Baek, S.M., Hwang, S. et al. (2016) Abatacept alleviates severe autoimmune symptoms in a patient carrying
a de novo variant in CTLA-4. J. Allergy Clin. Immun. 137, 327330 https://doi.org/10.1016/j.jaci.2015.08.036
6OShea, J.J. and Plenge, R. (2012) JAK and STAT signaling molecules in immunoregulation and immune-mediated disease. Immunity 36, 542550
https://doi.org/10.1016/j.immuni.2012.03.014
7 Schwartz, D.M., Bonelli, M., Gadina, M. and OShea, J.J. (2016) Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases.
Nat. Rev. Rheumatol. 12,2536 https://doi.org/10.1038/nrrheum.2015.167
8 Weinacht, K.G., Charbonnier, L.M., Alroqi, F., Plant, A., Qiao, Q., Wu, H. et al. (2017) Ruxolitinib reverses dysregulated T helper cell responses and
controls autoimmunity caused by a novel signal transducer and activator of transcription 1 (STAT1) gain-of-function mutation. J. Allergy Clin. Immun.
139, 16291640 https://doi.org/10.1016/j.jaci.2016.11.022
9 Gatti, R.A., Meuwissen, H.J., Allen, H.D., Hong, R. and Good, R.A. (1968) Immunological reconstitution of sex-linked lymphopenic immunological
deciency. Lancet 2, 13661369 https://doi.org/10.1016/S0140-6736(68)92673-1
10 Shamriz, O. and Chandrakasan, S. (2019) Update on advances in hematopoietic cell transplantation for primary immunodeciency disorders.
Immunol. Allergy Clin. North Am. 39, 113128 https://doi.org/10.1016/j.iac.2018.08.003
11 Kang, E. and Gennery, A. (2014) Hematopoietic stem cell transplantation for primary immunodeciencies. Hematol. Oncol. Clin. North Am. 28,
11571170 https://doi.org/10.1016/j.hoc.2014.08.006
12 Malech, H.L., Maples, P.B., Whiting-Theobald, N., Linton, G.F., Sekhsaria, S., Vowells, S.J. et al. (1997) Prolonged production of NADPH
oxidase-corrected granulocytes after gene therapy of chronic granulomatous disease. Proc. Natl Acad. Sci. U.S.A. 94, 1213312138
https://doi.org/10.1073/pnas.94.22.12133
13 Cavazzana-Calvo, M., Hacein-Bey, S., de Saint Basile, G., Gross, F., Yvon, E., Nusbaum, P. et al. (2000) Gene therapy of human severe combined
immunodeciency (SCID)-X1 disease. Science 288, 669672 https://doi.org/10.1126/science.288.5466.669
14 Gaspar, H.B., Parsley, K.L., Howe, S., King, D., Gilmour, K.C., Sinclair, J. et al. (2004) Gene therapy of X-linked severe combined immunodeciency by
use of a pseudotyped gammaretroviral vector. Lancet 364, 21812187 https://doi.org/10.1016/S0140-6736(04)17590-9
15 Hacein-Bey-Abina, S., Von Kalle, C., Schmidt, M., McCormack, M.P., Wulffraat, N., Leboulch, P. et al. (2003) LMO2-associated clonal T cell proliferation
in two patients after gene therapy for SCID-X1. Science 302, 415419 https://doi.org/10.1126/science.1088547
16 Stein, S., Ott, M.G., Schultze-Strasser, S., Jauch, A., Burwinkel, B., Kinner, A. et al. (2010) Genomic instability and myelodysplasia with monosomy 7
consequent to EVI1 activation after gene therapy for chronic granulomatous disease. Nat. Med. 16, 198204 https://doi.org/10.1038/nm.2088
17 Braun, C.J., Boztug, K., Paruzynski, A., Witzel, M., Schwarzer, A., Rothe, M. et al. (2014) Gene therapy for Wiskott-Aldrich syndromelong-term efcacy
and genotoxicity. Sci. Transl. Med. 6, 227ra33 https://doi.org/10.1126/scitranslmed.3007280
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology 7
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
18 Zufferey, R., Dull, T., Mandel, R.J., Bukovsky, A., Quiroz, D., Naldini, L. et al. (1998) Self-inactivating lentivirus vector for safe and efcient in vivo gene
delivery. J. Virol. 72, 98739880 PMID:9811723
19 Aiuti, A., Biasco, L., Scaramuzza, S., Ferrua, F., Cicalese, M.P., Baricordi, C. et al. (2013) Lentiviral hematopoietic stem cell gene therapy in patients
with Wiskott-Aldrich syndrome. Science 341, 1233151 https://doi.org/10.1126/science.1233151
20 Abina S, H.-B., Gaspar, H.B., Blondeau, J., Caccavelli, L., Charrier, S., Buckland, K. et al. (2015) Outcomes following gene therapy in patients with
severe Wiskott-Aldrich syndrome. J. Am. Med. Assoc. 313, 15501563 https://doi.org/10.1001/jama.2015.3253
21 De Ravin, S.S., Wu, X., Moir, S., Anaya-OBrien, S., Kwatemaa, N., Littel, P. et al. (2016) Lentiviral hematopoietic stem cell gene therapy for X-linked
severe combined immunodeciency. Sci. Transl. Med. 8, 335ra57 https://doi.org/10.1126/scitranslmed.aad8856
22 Carbonaro, D.A., Zhang, L., Jin, X., Montiel-Equihua, C., Geiger, S., Carmo, M. et al. (2014) Preclinical demonstration of lentiviral vector-mediated
correction of immunological and metabolic abnormalities in models of adenosine deaminase deciency. Mol. Ther. 22, 607622
https://doi.org/10.1038/mt.2013.265
23 Aiuti, A., Roncarolo, M.G. and Naldini, L. (2017) Gene therapy for ADA-SCID, the rst marketing approval of an ex vivo gene therapy in Europe: paving
the road for the next generation of advanced therapy medicinal products. EMBO Mol. Med. 9, 737740 https://doi.org/10.15252/emmm.201707573
24 Ferrua, F. and Aiuti, A. (2017) Twenty-ve years of gene therapy for ADA-SCID: from bubble babies to an approved drug. Hum. Gene Ther. 28,
972981 https://doi.org/10.1089/hum.2017.175
25 Gasiunas, G., Barrangou, R., Horvath, P. and Siksnys, V. (2012) Cas9-crRNA ribonucleoprotein complex mediates specic DNA cleavage for adaptive
immunity in bacteria. Proc. Natl Acad. Sci. U.S.A. 109, E2579E2586 https://doi.org/10.1073/pnas.1208507109
26 Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J.A. and Charpentier, E. (2012) A programmable dual-RNA-guided DNA endonuclease in
adaptive bacterial immunity. Science 337, 816821 https://doi.org/10.1126/science.1225829
27 Sander, J.D. and Joung, J.K. (2014) CRISPR-Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 32, 347355
https://doi.org/10.1038/nbt.2842
28 Jinek, M., East, A., Cheng, A., Lin, S., Ma, E. and Doudna, J. (2013) RNA-programmed genome editing in human cells. eLife 2, e00471
https://doi.org/10.7554/eLife.00471
29 Hsu, P.D., Lander, E.S. and Zhang, F. (2014) Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 12621278
https://doi.org/10.1016/j.cell.2014.05.010
30 Doudna, J.A. and Charpentier, E. (2014) Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096
https://doi.org/10.1126/science.1258096
31 Cong, L., Ran, F.A., Cox, D., Lin, S., Barretto, R., Habib, N. et al. (2013) Multiplex genome engineering using CRISPR/Cas systems. Science 339,
819823 https://doi.org/10.1126/science.1231143
32 Chapman, J.R., Taylor, M.R.G. and Boulton, S.J. (2012) Playing the end game: DNA double-strand break repair pathway choice. Mol. Cell 47, 497510
https://doi.org/10.1016/j.molcel.2012.07.029
33 Hubbard, N., Hagin, D., Sommer, K., Song, Y., Khan, I., Clough, C. et al. (2016) Targeted gene editing restores regulated CD40L function in X-linked
hyper-IgM syndrome. Blood 127, 25132522 https://doi.org/10.1182/blood-2015-11-683235
34 Kuo, C.Y., Long, J.D., Campo-Fernandez, B., de Oliveira, S., Cooper, A.R., Romero, Z. et al. (2018) Site-specic gene editing of human hematopoietic
stem cells for X-linked hyper-IgM syndrome. Cell Rep. 23, 26062616 https://doi.org/10.1016/j.celrep.2018.04.103
35 Wilkinson, R.A., Martin, C., Nemudryi, A.A. and Wiedenheft, B. (2019) CRISPR RNA-guided autonomous delivery of Cas9. Nat. Struct. Mol. Biol. 26,
1424 https://doi.org/10.1038/s41594-018-0173-y
36 Hsu, P.D., Scott, D.A., Weinstein, J.A., Ran, F.A., Konermann, S., Agarwala, V. et al. (2013) DNA targeting specicity of RNA-guided Cas9 nucleases.
Nat. Biotechnol. 31, 827832 https://doi.org/10.1038/nbt.2647
37 Kleinstiver, B.P., Prew, M.S., Tsai, S.Q., Topkar, V.V., Nguyen, N.T., Zheng, Z. et al. (2015) Engineered CRISPR-Cas9 nucleases with altered PAM
specicities. Nature 523, 481485 https://doi.org/10.1038/nature14592
38 Zhang, Y., Heidrich, N., Ampattu, B.J., Gunderson, C.W., Seifert, H.S., Schoen, C. et al. (2013) Processing-independent CRISPR RNAs limit natural
transformation in Neisseria meningitidis.Mol Cell. 50, 488503 https://doi.org/10.1016/j.molcel.2013.05.001
39 Esvelt, K.M., Mali, P., Braff, J.L., Moosburner, M., Yaung, S.J. and Church, G.M. (2013) Orthogonal Cas9 proteins for RNA-guided gene regulation and
editing. Nat. Methods 10, 11161121 https://doi.org/10.1038/nmeth.2681
40 Vakulskas, C.A., Dever, D.P., Rettig, G.R., Turk, R., Jacobi, A.M., Collingwood, M.A. et al. (2018) A high-delity Cas9 mutant delivered as a
ribonucleoprotein complex enables efcient gene editing in human hematopoietic stem and progenitor cells. Nat. Med. 24, 12161224
https://doi.org/10.1038/s41591-018-0137-0
41 Kleinstiver, B.P., Pattanayak, V., Prew, M.S., Tsai, S.Q., Nguyen, N.T. and Joung, J.K. (2016) High-delity CRISPR-Cas9 nucleases with no detectable
genome-wide off-target effects. Nature 529, 490495 https://doi.org/10.1038/nature16526
42 Mali, P., Yang, L., Esvelt, K.M., Aach, J., Guell, M., DiCarlo, J.E. et al. (2013) RNA-guided human genome engineering via Cas9. Science 339,
823826 https://doi.org/10.1126/science.1232033
43 Tsai, S.Q., Wyvekens, N., Khayter, C., Foden, J.A., Thapar, V., Reyon, D. et al. (2014) Dimeric CRISPR RNA-guided FokI nucleases for highly specic
genome editing. Nat. Biotechnol. 32, 569576 https://doi.org/10.1038/nbt.2908
44 Guilinger, J.P., Thompson, D.B. and Liu, D.R. (2014) Fusion of catalytically inactive Cas9 to FokI nuclease improves the specicity of genome
modication. Nat. Biotechnol. 32, 577582 https://doi.org/10.1038/nbt.2909
45 Wyvekens, N., Topkar, V.V., Khayter, C., Joung, J.K. and Tsai, S.Q. (2015) Dimeric CRISPR RNA-guided FokI-dCas9 nucleases directed by truncated
gRNAs for highly specic genome editing. Hum. Gene Ther. 26, 425431 https://doi.org/10.1089/hum.2015.084
46 Ran, F.A., Hsu, P.D., Lin, C.Y., Gootenberg, J.S., Konermann, S., Trevino, A.E. et al. (2013) Double nicking by RNA-guided CRISPR Cas9 for enhanced
genome editing specicity. Cell 154, 13801389 https://doi.org/10.1016/j.cell.2013.08.021
47 Cho, S.W., Kim, S., Kim, Y., Kweon, J., Kim, H.S., Bae, S. et al. (2014) Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases
and nickases. Genome Res. 24, 132141 https://doi.org/10.1101/gr.162339.113
48 Zetsche, B., Gootenberg, J.S., Abudayyeh, O.O., Slaymaker, I.M., Makarova, K.S., Essletzbichler, P. et al. (2015) Cpf1 is a single-RNA-guided
endonuclease of a Class 2 CRISPR-Cas system. Cell 163, 759771 https://doi.org/10.1016/j.cell.2015.09.038
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology8
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
49 Kleinstiver, B.P., Tsai, S.Q., Prew, M.S., Nguyen, N.T., Welch, M.M., Lopez, J.M. et al. (2016) Genome-wide specicities of CRISPR-Cas Cpf1 nucleases
in human cells. Nat. Biotechnol. 34, 869874 https://doi.org/10.1038/nbt.3620
50 Fagerlund, R.D., Staals, R.H.J. and Fineran, P.C. (2015) The Cpf1 CRISPR-Cas protein expands genome-editing tools. Genome Biol. 16, 251
https://doi.org/10.1186/s13059-015-0824-9
51 Teng, F., Cui, T.T., Feng, G.H., Guo, L., Xu, K., Gao, Q.Q. et al. (2018) Repurposing CRISPR-Cas12b for mammalian genome engineering. Cell Discov.
4,63https://doi.org/10.1038/s41421-018-0069-3
52 Strecker, J., Jones, S., Koopal, B., Schmid-Burgk, J., Zetsche, B., Gao, L.Y. et al. (2019) Engineering of CRISPR-Cas12b for human genome editing.
Nat. Commun. 10, 212 https://doi.org/10.1038/s41467-018-08224-4
53 Nishida, K., Arazoe, T., Yachie, N., Banno, S., Kakimoto, M., Tabata, M. et al. (2016) Targeted nucleotide editing using hybrid prokaryotic and vertebrate
adaptive immune systems. Science 353, 919921 https://doi.org/10.1126/science.aaf7573
54 Komor, A.C., Kim, Y.B., Packer, M.S., Zuris, J.A. and Liu, D.R. (2016) Programmable editing of a target base in genomic DNA without double-stranded
DNA cleavage. Nature 533, 420424 https://doi.org/10.1038/nature17946
55 Gaudelli, N.M., Komor, A.C., Rees, H.A., Packer, M.S., Badran, A.H., Bryson, D.I. et al. (2017) Programmable base editing of ATtoGC in genomic
DNA without DNA cleavage. Nature 551, 464471 https://doi.org/10.1038/nature24644
56 Eid, A., Alshareef, S. and Mahfouz, M.M. (2018) CRISPR base editors: genome editing without double-stranded breaks. Biochem. J. 475, 19551964
https://doi.org/10.1042/BCJ20170793
57 Zuo, E.W., Sun, Y.D., Wei, W., Yuan, T.L., Ying, W.Q., Sun, H. et al. (2019) Cytosine base editor generates substantial off-target single-nucleotide
variants in mouse embryos. Science 364, 289292 https://doi.org/10.1126/science.aav9973
58 Fu, Y.F., Sander, J.D., Reyon, D., Cascio, V.M. and Joung, J.K. (2014) Improving CRISPR-Cas nuclease specicity using truncated guide RNAs. Nat.
Biotechnol. 32, 279284 https://doi.org/10.1038/nbt.2808
59 Moon, S.B., Kim, D.Y., Ko, J.H., Kim, J.S. and Kim, Y.S. (2019) Improving CRISPR genome editing by engineering guide RNAs. Trends Biotechnol.
https://doi.org/10.1016/j.tibtech.2019.01.009
60 Hendel, A., Bak, R.O., Clark, J.T., Kennedy, A.B., Ryan, D.E., Roy, S. et al. (2015) Chemically modied guide RNAs enhance CRISPR-Cas genome
editing in human primary cells. Nat. Biotechnol. 33, 985989 https://doi.org/10.1038/nbt.3290
61 Dever, D.P., Bak, R.O., Reinisch, A., Camarena, J., Washington, G., Nicolas, C.E. et al. (2016) CRISPR/Cas9 β-globin gene targeting in human
haematopoietic stem cells. Nature 539, 384389 https://doi.org/10.1038/nature20134
62 Gundry, M.C., Brunetti, L., Lin, A., Mayle, A.E., Kitano, A., Wagner, D. et al. (2016) Highly efcient genome editing of murine and human hematopoietic
progenitor cells by CRISPR/Cas9. Cell Rep. 17, 14531461 https://doi.org/10.1016/j.celrep.2016.09.092
63 Miura, H., Gurumurthy, C.B., Sato, T., Sato, M. and Ohtsuka, M. (2015) CRISPR/Cas9-based generation of knockdown mice by intronic insertion of
articial microRNA using longer single-stranded DNA. Sci. Rep. 5, 12799 https://doi.org/10.1038/srep12799
64 Yoshimi, K., Kunihiro, Y., Kaneko, T., Nagahora, H., Voigt, B. and Mashimo, T. (2016) ssODN-mediated knock-in with CRISPR-Cas for large genomic
regions in zygotes. Nat. Commun. 7, 10431 https://doi.org/10.1038/ncomms10431
65 Jun, S., Lim, H., Jang, H., Lee, W., Ahn, J., Lee, J.H. et al. (2018) Straightforward delivery of linearized double-stranded DNA encoding sgRNA and
donor DNA for the generation of single nucleotide variants based on the CRISPR/Cas9 system. ACS Synth. Biol. 7, 16511659
https://doi.org/10.1021/acssynbio.7b00345
66 Quadros, R.M., Miura, H., Harms, D.W., Akatsuka, H., Sato, T., Aida, T. et al. (2017) Easi-CRISPR: a robust method for one-step generation of mice
carrying conditional and insertion alleles using long ssDNA donors and CRISPR ribonucleoproteins. Genome Biol. 18,92
https://doi.org/10.1186/s13059-017-1220-4
67 Ma, Y.W., Zhang, X., Shen, B., Lu, Y.D., Chen, W., Ma, J. et al. (2014) Generating rats with conditional alleles using CRISPR/Cas9. Cell Res. 24,
122125 https://doi.org/10.1038/cr.2013.157
68 Yang, H., Wang, H.Y., Shivalila, C.S., Cheng, A.W., Shi, L.Y. and Jaenisch, R. (2013) One-step generation of mice carrying reporter and conditional
alleles by CRISPR/Cas-mediated genome engineering. Cell 154, 13701379 https://doi.org/10.1016/j.cell.2013.08.022
69 Ortinski, P.I., ODonovan, B., Dong, X.Y. and Kantor, B. (2017) Integrase-decient lentiviral vector as an all-in-one platform for highly efcient CRISPR/
Cas9-mediated gene editing. Mol. Ther. Methods Clin. D. 5, 153164 https://doi.org/10.1016/j.omtm.2017.04.002
70 Miller, D.G., Petek, L.M. and Russell, D.W. (2003) Human gene targeting by adeno-associated virus vectors is enhanced by DNA double-strand breaks.
Mol. Cell. Biol. 23, 35503557 https://doi.org/10.1128/MCB.23.10.3550-3557.2003
71 Russell, D.W. and Hirata, R.K. (1998) Human gene targeting by viral vectors. Nat. Genet. 18, 325330 https://doi.org/10.1038/ng0498-325
72 Khan, I.F., Hirata, R.K. and Russell, D.W. (2011) AAV-mediated gene targeting methods for human cells. Nat. Protoc. 6, 482501
https://doi.org/10.1038/nprot.2011.301
73 Bak, R.O. and Porteus, M.H. (2017) CRISPR-mediated integration of large gene cassettes using AAV donor vectors. Cell Rep. 20, 750756
https://doi.org/10.1016/j.celrep.2017.06.064
74 Song, L.J., Kauss, M.A., Kopin, E., Chandra, M., Ul-Hasan, T., Miller, E. et al. (2013) Optimizing the transduction efciency of capsid-modied AAV6
serotype vectors in primary human hematopoietic stem cells in vitro and in a xenograft mouse model in vivo. Cytotherapy 15, 986998
https://doi.org/10.1016/j.jcyt.2013.04.003
75 Bak, R.O., Dever, D.P. and Porteus, M.H. (2018) CRISPR/cas9 genome editing in human hematopoietic stem cells. Nat. Protoc. 13, 358376
https://doi.org/10.1038/nprot.2017.143
76 De Ravin, S.S., Reik, A., Liu, P.Q., Li, L.H., Wu, X.L., Su, L. et al. (2016) Targeted gene addition in human CD34
+
hematopoietic cells for correction of
X-linked chronic granulomatous disease. Nat. Biotechnol. 34, 424429 https://doi.org/10.1038/nbt.3513
77 Charlesworth, C.T., Camarena, J., Cromer, M.K., Vaidyanathan, S., Bak, R.O., Carte, J.M. et al. (2018) Priming human repopulating hematopoietic stem
and progenitor cells for Cas9/sgRNA gene targeting. Mol. Ther. Nucl. Acids 12,89104 https://doi.org/10.1016/j.omtn.2018.04.017
78 Gao, L.Y., Cox, D.B.T., Yan, W.X., Manteiga, J.C., Schneider, M.W., Yamano, T. et al. (2017) Engineered Cpf1 variants with altered PAM specicities.
Nat. Biotechnol. 35, 789792 https://doi.org/10.1038/nbt.3900
79 Slaymaker, I.M., Gao, L.Y., Zetsche, B., Scott, D.A., Yan, W.X. and Zhang, F. (2016) Rationally engineered Cas9 nucleases with improved specicity.
Science 351,8488 https://doi.org/10.1126/science.aad5227
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology 9
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
80 De Ravin, S.S., Li, L.H., Wu, X.L., Choi, U., Allen, C., Koontz, S. et al. (2017) CRISPR-Cas9 gene repair of hematopoietic stem cells from patients with
X-linked chronic granulomatous disease. Sci. Transl. Med. 9, eaah3480 https://doi.org/10.1126/scitranslmed.aah3480
81 Richardson, C.D., Ray, G.J., DeWitt, M.A., Curie, G.L. and Corn, J.E. (2016) Enhancing homology-directed genome editing by catalytically active and
inactive CRISPR-Cas9 using asymmetric donor DNA. Nat. Biotechnol. 34, 339344 https://doi.org/10.1038/nbt.3481
82 Hoban, M.D., Lumaquin, D., Kuo, C.Y., Romero, Z., Long, J., Ho, M. et al. (2016) CRISPR/Cas9-mediated correction of the sickle mutation in human
CD34
+
cells. Mol. Ther. 24, 15611569 https://doi.org/10.1038/mt.2016.148
83 Hoban, M.D., Cost, G.J., Mendel, M.C., Romero, Z., Kaufman, M.L., Joglekar, A.V. et al. (2015) Correction of the sickle cell disease mutation in human
hematopoietic stem/progenitor cells. Blood 125, 25972604 https://doi.org/10.1182/blood-2014-12-615948
84 DeWitt, M.A., Magis, W., Bray, N.L., Wang, T., Berman, J.R., Urbinati, F. et al. (2016) Selection-free genome editing of the sickle mutation in human
adult hematopoietic stem/progenitor cells. Sci. Transl. Med. 8, 360ra134 https://doi.org/10.1126/scitranslmed.aaf9336
85 Agudelo, D., Duringer, A., Bozoyan, L., Huard, C.C., Carter, S., Loehr, J. et al. (2017) Marker-free coselection for CRISPR-driven genome editing in
human cells. Nat. Methods 14, 615620 https://doi.org/10.1038/nmeth.4265
86 Renaud, J.B., Boix, C., Charpentier, M., De Cian, A., Cochennec, J., Duvernois-Berthet, E. et al. (2016) Improved genome editing efciency and exibility
using modied oligonucleotides with TALEN and CRISPR-Cas9 nucleases. Cell Rep. 14,22632272 https://doi.org/10.1016/j.celrep.2016.02.018
87 Song, F. and Stieger, K. (2017) Optimizing the DNA donor template for homology-directed repair of double-strand breaks. Mol. Ther. Nucl. Acids 7,
5360 https://doi.org/10.1016/j.omtn.2017.02.006
88 Yang, L., Guell, M., Byrne, S., Yang, J.L., De Los Angeles, A., Mali, P. et al. (2013) Optimization of scarless human stem cell genome editing. Nucl.
Acids Res. 41, 90499061 https://doi.org/10.1093/nar/gkt555
89 Uchida, N., Li, L., Haro-Mora, J.J., Demirci, S., Nassehi, T., Gamer, J. et al. (2018) Development of a clinically applicable method for high-efciency
gene correction of plerixafor-mobilized CD34+ cells from patients with sickle cell disease.ASH Annual Meeting, San Diego, CA
90 Merling, R.K., Kuhns, D.B., Sweeney, C.L., Wu, X., Burkett, S., Chu, J. et al. (2017) Gene-edited pseudogene resurrection corrects p47
phox
-decient
chronic granulomatous disease. Blood Adv. 1, 270278 https://doi.org/10.1182/bloodadvances.2016001214
91 Schiroli, G., Ferrari, S., Conway, A., Jacob, A., Capo, V., Albano, L. et al. (2017) Preclinical modeling highlights the therapeutic potential of
hematopoietic stem cell gene editing for correction of SCID-X1. Sci. Transl. Med. 9, eaan0820 https://doi.org/10.1126/scitranslmed.aan0820
92 Genovese, P., Schiroli, G., Escobar, G., Di Tomaso, T., Firrito, C., Calabria, A. et al. (2014) Targeted genome editing in human repopulating
haematopoietic stem cells. Nature 510, 235240 https://doi.org/10.1038/nature13420
93 Pavel-Dinu, M., Wiebking, V., Dejene, B.T, Srifa, W., Mantri, S., Nicolas, C.E et al. (2019) A safe and efcient universal genome targeting therapeutic
approach for SCID-X1 in human long-term hematopoietic stem cells. Nat. Commun. 10, 2021 https://doi.org/10.1038/s41467-019-09614-y
94 Hubbard, N.W., Hagin, D., Sommer, K., Song, Y.M., Clough, C., Khan, I.F. et al. (2016) Targeted gene editing restores regulated CD40L expression and
function in X-Higm T cells. Blood 127, 25132522 https://doi.org/10.1182/blood-2015-11-683235
95 Maerken, M., MacEwan, D., Harper, N., Slupsky, J.R. and Linley, A. (2018) Gene editing of BTK in acute myeloid leukaemia using CRISPR-Cas9.ASH
2018, San Diego, CA
96 Flynn, R., Grundmann, A., Renz, P., Hanseler, W., James, W.S., Cowley, S.A. et al. (2015) CRISPR-mediated genotypic and phenotypic correction of a
chronic granulomatous disease mutation in human iPS cells. Exp. Hematol. 43, 838848.e3 https://doi.org/10.1016/j.exphem.2015.06.002
97 Sweeney, C.L., Choi, U., Pavel-Dinu, M., Koontz, S., Li, L., Theobald, N. et al. (2018) CRISPR-mediated targeted insertion of CYBB cDNAs into the
CYBB locus for correction of X-linked chronic granulomatous disease patient CD34+ cells.ASGCT Annual Meeting, Chicago, IL
98 Sweeney, C.L., Zou, J.Z., Choi, U., Merling, R.K., Liu, A., Bodansky, A. et al. (2017) Targeted repair of CYBB in X-CGD iPSCs requires retention of
intronic sequences for expression and functional correction. Mol. Ther. 25, 321330 https://doi.org/10.1016/j.ymthe.2016.11.012
99 Paterna, J.C., Moccetti, T., Mura, A., Feldon, J. and Bueler, H. (2000) Inuence of promoter and WHV post-transcriptional regulatory element on
AAV-mediated transgene expression in the rat brain. Gene Ther. 7, 13041311 https://doi.org/10.1038/sj.gt.3301221
100 Zufferey, R., Donello, J.E., Trono, D. and Hope, T.J. (1999) Woodchuck hepatitis virus posttranscriptional regulatory element enhances expression of
transgenes delivered by retroviral vectors. J. Virol. 73, 28862892 PMID:10074136
101 Cromer, M.K., Vaidyanathan, S., Ryan, D.E., Curry, B., Lucas, A.B., Camarena, J. et al. (2018) Global transcriptional response to CRISPR/Cas9-AAV 6- b a s e d
genome editing in CD34
+
hematopoietic stem and progenitor cells. Mol. Ther. 26,24312442 https://doi.org/10.1016/j.ymthe.2018.06.002
102 Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. and Taipale, J. (2018) CRISPR-Cas9 genome editing induces a p53-mediated DNA damage
response. Nat. Med. 24, 927930 https://doi.org/10.1038/s41591-018-0049-z
103 Schoggins, J.W. and Rice, C.M. (2011) Interferon-stimulated genes and their antiviral effector functions. Curr. Opin. Virol. 1, 519525
https://doi.org/10.1016/j.coviro.2011.10.008
104 Schiroli, G., Conti, A., Ferrari, S., Della Volpe, L., Jacob, A., Albano, L. et al. (2019) Precise gene editing preserves hematopoietic stem cell function
following transient p53-mediated DNA damage response. Cell Stem Cell 24, 551565 https://doi.org/10.1016/j.stem.2019.02.019
105 Petrillo, C., Thorne, L.G., Unali, G., Schiroli, G., Giordano, A.M.S., Piras, F. et al. (2018) Cyclosporine H overcomes innate immune restrictions to
improve lentiviral transduction and gene editing in human hematopoietic stem cells. Cell Stem Cell 23, 820832.e9 https://doi.org/10.1016/j.stem.
2018.10.008
106 Zonari, E., Desantis, G., Petrillo, C., Boccalatte, F.E., Lidonnici, M.R., Kajaste-Rudnitski, A. et al. (2017) Efcient ex vivo engineering and expansion of
highly puried human hematopoietic stem and progenitor cell populations for gene therapy. Stem Cell Rep. 8, 977990 https://doi.org/10.1016/j.
stemcr.2017.02.010
107 Boitano, A.E., Wang, J.A., Romeo, R., Bouchez, L.C., Parker, A.E., Sutton, S.E. et al. (2010) Aryl hydrocarbon receptor antagonists promote the
expansion of human hematopoietic stem cells. Science 329, 13451348 https://doi.org/10.1126/science.1191536
108 Fares, I., Chagraoui, J., Gareau, Y., Gingras, S., Ruel, R., Mayotte, N. et al. (2014) Pyrimidoindole derivatives are agonists of human hematopoietic stem
cell self-renewal. Science 345, 15091512 https://doi.org/10.1126/science.1256337
109 Branzei, D. and Foiani, M. (2008) Regulation of DNA repair throughout the cell cycle. Nat. Rev. Mol. Cell Biol. 9, 297308 https://doi.org/10.1038/
nrm2351
110 Gutschner, T., Haemmerle, M., Genovese, G., Draetta, G.F. and Chin, L. (2016) Post-translational regulation of Cas9 during G1 enhances
homology-directed repair. Cell Rep. 14, 15551566 https://doi.org/10.1016/j.celrep.2016.01.019
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology10
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
111 Liu, M.J., Rehman, S., Tang, X.D., Gu, K., Fan, Q.L., Chen, D.K. et al. (2019) Methodologies for improving HDR efciency. Front. Genet. 9, 691
https://doi.org/10.3389/fgene.2018.00691
112 Savic, N., Ringnalda, F.C.A.S., Lindsay, H., Berk, C., Bargsten, K., Li, Y.Z. et al. (2018) Covalent linkage of the DNA repair template to the CRISPR-Cas9
nuclease enhances homology-directed repair. eLife 7, e33761 https://doi.org/10.7554/eLife.33761
113 Maruyama, T., Dougan, S.K., Truttmann, M.C., Bilate, A.M., Ingram, J.R. and Ploegh, H.L. (2016) Increasing the efciency of precise genome editing
with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat. Biotechnol. 34, 210 https://doi.org/10.1038/nbt0216-210c
114 Canny, M.D., Moatti, N., Wan, L.C.K., Fradet-Turcotte, A., Krasner, D., Mateos-Gomez, P.A. et al. (2018) Inhibition of 53BP1 favors
homology-dependent DNA repair and increases CRISPR-Cas9 genome-editing efciency. Nat. Biotechnol. 36,95102 https://doi.org/10.1038/nbt.4021
115 Deau, M.C., Heurtier, L., Frange, P., Suarez, F., Bole-Feysot, C., Nitschke, P. et al. (2015) A human immunodeciency caused by mutations in the
PIK3R1 gene. J. Clin. Invest. 125, 17641765 https://doi.org/10.1172/JCI81746
116 Elgizouli, M., Lowe, D.M., Speckmann, C., Schubert, D., Hulsdunker, J., Eskandarian, Z. et al. (2016) Activating PI3Kδmutations in a cohort of 669
patients with primary immunodeciency. Clin. Exp. Immunol. 183, 221229 https://doi.org/10.1111/cei.12706
117 Kuehn, H.S., Ouyang, W., Lo, B., Deenick, E.K., Niemela, J.E., Avery, D.T. et al. (2014) Immune dysregulation in human subjects with heterozygous
germline mutations in CTLA4. Science 345, 16231627 https://doi.org/10.1126/science.1255904
118 Lucas, C.L., Kuehn, H.S., Zhao, F., Niemela, J.E., Deenick, E.K., Palendira, U. et al. (2014) Dominant-activating germline mutations in the gene
encoding the PI(3)K catalytic subunit p110δresult in T cell senescence and human immunodeciency. Nat. Immunol. 15,8897
https://doi.org/10.1038/ni.2771
119 Schubert, D., Bode, C., Kenefeck, R., Hou, T.Z., Wing, J.B., Kennedy, A. et al. (2014) Autosomal dominant immune dysregulation syndrome in humans
with CTLA4 mutations. Nat. Med. 20, 14101416 https://doi.org/10.1038/nm.3746
© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society and the Royal Society of Biology 11
Emerging Topics in Life Sciences (2019)
https://doi.org/10.1042/ETLS20180157
... A critical limitation of the gene therapy approach for IEIs is the broad spectrum of disease categories as well as the varying mutations, which require the development of personalized medicine approaches depending on the specific mutation a patient carries (250). To ensure successful clinical outcomes for gene therapy of IEIs, a sufficient number of gene-corrected HSCs with engraftment capabilities following CRISPR/Cas9 gene editing is of paramount importance (251). HDR occurs most efficiently in dividing cells, and the low efficiency of HDR in HSCs can be improved by manipulating critical factors of DNA repair mechanisms to inhibit the NHEJ pathway (72,73). ...
... Optimizing CRISPR/Cas genome editing tools to maximize editing efficiency while minimizing off-target effects presents directions for advancing genome editing agents (247). The engraftment capability and selfrenewal ability of gene-corrected HSCs are critical factors that should be taken into account because manipulating and expanding gene-corrected cells in vitro increases the risk of losing the multilineage potential of HSCs (251). Exposure to nucleases and the toxic effects caused by condition agents largely affect the selfrenew ability of gene corrected HSCs, implementation of "hit-andrun" strategy to reduce nucleases exposure time and development of advanced conditioning regimens could ameliorate these harmfulness to enhance HSC self-renewal ability (14, 251). ...
Article
Full-text available
Inborn errors of immunity (IEIs) are a group of inherited disorders caused by mutations in the protein-coding genes involved in innate and/or adaptive immunity. Hematopoietic stem cell transplantation (HSCT) is a mainstay definitive therapy for many severe IEIs. However, the lack of HLA-matched donors increases the risk of developing severe immunological complications. Gene therapy provides long-term clinical benefits and could be an attractive therapeutic strategy for IEIs. In this review, we describe the development and evolution of clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated proteins (Cas) gene-editing systems, including double-strand break (DSB)-based gene editing and DSB-free base editing or prime editing systems. Here, we discuss the advances in and issues associated with CRISPR/Cas gene editing tools and their potential as therapeutic alternatives for IEIs. We also highlight the progress of preclinical studies for the treatment of human genetic diseases, including IEIs, using CRISR/Cas and ongoing clinical trials based on this versatile technology.
... The primary immune deficiency diseases most studied for gene therapy have been X-linked SCID, ADA-SCID, X-linked CGD, and WAS. [30][31][32][33][34][35][36][37][38][39][40] Over the past 25 years, work in the field of gene therapy for primary immunodeficiencies has made significant progress and also expanded the possibilities to include treatments for other diseases. Early gene therapy using gammaretroviral vectors was associated with insertional mutagenesis near proto-oncogenes, resulting in malignancy in some patients. ...
Article
Full-text available
Over the past 20 years, the rapid evolution in the diagnosis and treatment of primary immunodeficiencies (PI) and the recognition of immune dysregulation as a feature in some have prompted the use of “inborn errors of immunity” (IEI) as a more encompassing term used to describe these disorders [1, 2] . This article aims to review the future of therapy of PI/IEI (referred to IEI throughout this paper). Historically, immune deficiencies have been characterized as monogenic disorders resulting in immune deficiencies affecting T cells, B cells, combination of T and B cells, or innate immune disorders. More recently, immunologists are also recognizing a variety of phenotypes associated with one genotype or similar phenotypes across genotypes and a role for incomplete penetrance or variable expressivity of some genes causing inborn errors of immunity [3]. The IUIS classification of immune deficiencies (IEIs) has evolved over time to include 10 categories, with disorders of immune dysregulation accounting for a new subset, some treatable with small molecule inhibitors or biologics. [1] Until recently, management options were limited to prompt treatment of infections, gammaglobulin replacement, and possibly bone marrow transplant depending on the defect. Available therapies have expanded to include small molecule inhibitors, biologics, gene therapy, and the use of adoptive transfer of virus-specific T cells to fight viral infections in immunocompromised patients. Several significant contributions to the field of clinical immunology have fueled the rapid advancement of therapies over the past two decades. Among these are educational efforts to recruit young immunologists to the field resulting in the growth of a world-wide community of clinicians and investigators interested in rare diseases, efforts to increase awareness of IEI globally contributing to international collaborations, along with advancements in diagnostic genetic testing, newborn screening, molecular biology techniques, gene correction, use of immune modulators, and ex vivo expansion of engineered T cells for therapeutic use. The development and widespread use of newborn screening have helped to identify severe combined immune deficiency (SCID) earlier resulting in better outcomes [4]. Continual improvements and accessibility of genetic sequencing have helped to identify new IEI diseases at an accelerated pace [5]. Advances in gene therapy and bone marrow transplant have made treatments possible in otherwise fatal diseases. Furthermore, the increased awareness of IEI across the world has driven networks of immunologists working together to improve the diagnosis and treatment of these rare diseases. These improvements in the diagnosis and treatment of IEI noted over the past 20 years bring hope for a better future for the IEI community. This paper will review future directions in a few of the newer therapies emerging for IEI. For easy reference, most of the diseases discussed in this paper are briefly described in a summary table, in the order mentioned within the paper (Appendix).
... We sought to develop a gene therapy approach that would address most of the mutations and maintain endogenous regulation to ensure physiological expression using the CRISPR/Cas9 approach to target insertion (TI) of a corrective MAGT1 complementary DNA (cDNA) near the transcription start site. [16][17][18][19] Exciting developments in CRISPR gene correction for several PIDs 20-25 are unfortunately not matched by the pace of clinical translation. 26 A major barrier to clinical translation of promising CRISPR gene correction procedures is the low engraftment rates of edited hematopoietic stem and progenitor cells (HSPCs) with substantially reduced correction rates after transplantation in mouse xenograft models, which indicates poor targeting of engrafting human HSPCs. ...
Article
'X-linked MAGT1 deficiency with increased susceptibility to Epstein-Barr virus-infection and N-linked glycosylation defect' (XMEN) disease is a recently described primary immunodeficiency marked by defective T and Natural Killer (NK) cells. Potentially curative hematopoietic stem cell transplant is associated with high mortality rates. We sought to develop an ex vivo targeted gene therapy approach for XMEN patients using CRISPR/Cas9/adeno-associated vector (AAV) to insert a therapeutic MAGT1 gene at the constitutive locus under the regulation of the endogenous promoter. Clinical translation of CRISPR/Cas9/AAV-targeted gene editing (GE) is hampered by low engraftable GE hematopoietic stem/progenitor cells (HSPCs). Here, we optimized GE conditions by transient enhancement of homology-directed repair while suppressing AAV-associated DNA damage response to achieve highly efficient (>60%) genetic correction in engrafting XMEN HSPCs in transplanted mice. Restored MAGT1-glycosylation function in human NK and CD8+ T cells restored NKG2D expression and function in XMEN lymphocytes for potential treatment of infections, and corrected HSPCs for long-term gene therapy, thus offering two efficient therapeutic options for XMEN poised for clinical translation.
... Gene and cell-based therapies currently show immense promise. Targeted genome editing using the clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) system could be used for correcting gene errors in patients' cells ex vivo, followed by return of genetically repaired cells (De Ravin and Brault, 2019), thus avoiding immune system responses against foreign cells. This approach can be termed "precision medicine", which involves narrowly targeting specific genes or proteins. ...
... In the case of CRISPR-Cas9 approaches for HSC genome editing in PIDs, the design of the donor template is challenging and both the nature (single/multiple mutations or deletions in one or more hotspots distributed along the gene) and the functional effect of the mutation (gain of function versus loss of function) have to be taken into consideration [77]. ...
Article
Full-text available
Primary immunodeficiency diseases (PIDs) are rare diseases that are characterized by genetic mutations that damage immunological function, defense, or both. Some of these rare diseases are caused by aberrations in the normal development of natural killer cells (NKs) or affect their lytic synapse. The pathogenesis of these types of diseases as well as the processes underlying target recognition by human NK cells is not well understood. Utilizing induced pluripotent stem cells (iPSCs) will aid in the study of human disorders, especially in the PIDs with defects in NK cells for PID disease modeling. This, together with genome editing technology, makes it possible for us to facilitate the discovery of future therapeutics and/or cell therapy treatments for these patients, because, to date, the only curative treatment available in the most severe cases is hematopoietic stem cell transplantation (HSCT). Recent progress in gene editing technology using CRISPR/Cas9 has significantly increased our capability to precisely modify target sites in the human genome. Among the many tools available for us to study human PIDs, disease- and patient-specific iPSCs together with gene editing offer unique and exceptional methodologies to gain deeper and more thorough understanding of these diseases as well as develop possible alternative treatment strategies. In this review, we will discuss some immunodeficiency disorders affecting NK cell function, such as classical NK deficiencies (CNKD), functional NK deficiencies (FNKD), and PIDs with involving NK cells as well as strategies to model and correct these diseases for further study and possible avenues for future therapies.
... Strategies to manipulate these pathways may be required for efficient, high yield editing of stem cell populations. Romito et al. [11] provide an update on the potential of genome editing of blood stem cells, and DeRavin and Brault [12] consider specific applications for inherited immunodeficiencies, some of which are close to clinical phase testing. The attraction of such conditions for genome editing derives from the possibility of providing therapeutic 'cure' through the correction of a relatively modest number of stem cells if upon correction, cells acquire a powerful survival advantage compared with defective populations. ...
Article
The rapid evolution of tools for genome editing has created a dizzying array of possibilities for novel therapeutic strategies, even though to date only a handful of clinical applications have been realised. Proof-of-concept demonstrations of targeted genome modification in vitro and in small animal models of inherited single gene disorders have to be translated into effective therapies. Interest has naturally gravitated towards opportunities for collection, ex vivo modification and return of blood, immune and stem cells. Initial applications designed to modify T cells to protect against HIV or to confer potent anti-leukaemic effects have reached clinical phase, and further applications to modify blood stem cells are close to being applied. There are generic considerations of safety, on- and off-target effects and possible genotoxicity as well as issues relating to more sophisticated systemic approaches where niche occupation and host immunity become relevant. Such issues will be likely addressed over time, with carefully designed clinical trials required to determine therapeutic risks and benefits.
Article
Full-text available
Aim: This research examines the safety and effectiveness of CRISPR-Cas technologies in correcting genetic defects and restoring immunological function. Method: This study reviewed CRISPR-Cas research on treating paediatric immunodeficiencies using PRISMA criteria. Databases: The searches conducted in PubMed, Web of Science, and Embase primarily focused on scholarly papers published in English Findings: Five of seventeen papers matched the requirements for more complete research after a thorough examination. Numerous studies have demonstrated that CRISPR-Cas may correct genetic abnormalities in SCID and Wiskott-Aldrich Syndrome. Thus, animal models have showed considerable immune function improvements and disease amelioration. Due to a lack of clinical application data, laboratory results differ from paediatric patient outcomes. Conclusion: In conclusion, CRISPR-Cas technology allows precise genetic changes, promising therapy for paediatric immunodeficiencies. Despite promising preclinical results, longer-term clinical trials are needed to determine safety and effectiveness. To maximise the utility of CRISPR-Cas for paediatric immunodeficiency therapies, laboratory discoveries must be translated into clinical practice.
Thesis
Full-text available
Heterozygote Keimbahn-Mutationen in Cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) beeinträchtigen die suppressive Funktion von regulatorischen T-Zellen im menschlichen Immunsystem. Dadurch kommt es zu einer überschießenden Immunantwort und Fehlregulationen, vor allem bei Interaktionen des zellulären Immunsystems. Klinisch präsentieren sich betroffene Mutationsträger mit häufigen und wiederkehrenden Infektionen. Darüber hinaus entwickeln manche Patienten lebensbedrohliche Antikörper-vermittelte Autoimmunerkrankungen sowie massive Lymphoproliferationen. Doch es existieren bislang keine einheitlichen therapeutischen Empfehlungen. Ein Teil der Mutationsträger entwickelt bösartige Tumore; doch hierzu gibt es bislang noch keine ausreichenden Kenntnisse. Die vorliegende Arbeit setzt an diesen Punkten an und präsentiert erstmals Behandlungsempfehlungen für CTLA4-Mutationsträger und neue klinisch-relevante Erkenntnisse. Dies wurde durch eine Zusammentragung weltweiter Fälle und eine detaillierte Analyse der bisherigen Therapien und deren Wirkungen möglich. Es wurde herausgefunden, wodurch die Effektivität der immunsuppressiven Therapien verbessert werden kann und wie groß das Potential neuer therapeutischer Ansätze ist. Eine bereits existierende Kohorte konnte im Rahmen dieser Arbeit um 42 CTLA4- Mutationsträger auf insgesamt 175 Mutationsträger erweitert werden. 123 benötigten immunsuppressive Therapien, weshalb diese Patienten als klinisch betroffen klassifiziert worden sind (entspricht einer Penetranz von 70%). Unter den 42 neuen Mutationsträgern wurden 39 erstmals veröffentlicht. Zehn Mutationen wurden erstmals publiziert. 19 Varianten im Gen CTLA4 wurden im Labor des Centrums für chronische Immundefizienz in Freiburg funktionell getestet. Darunter waren vier, von denen noch keine Testung publiziert war. Insgesamt 17 Mutationsträger entwickelten im Laufe ihres Lebens eine Neoplasie, was im Vergleich zum Wildtyp einen Hinweis auf ein erhöhtes Krebsrisiko darstellt. Da mehr als die Hälfte der Neoplasien mit onkogenen Viren assoziiert waren, scheint deren unzureichende Kontrolle Teil des Phänotyps zu sein. Es wurde eine Webbrowser-basierte Onlinedatenbank aufgebaut. Diese sog. CTLA-4- Registry ist in Zukunft multizentrisch nutzbar. Weitere Forschung sollte einerseits die unvollständige Penetranz und andererseits die Effektivität und Sicherheit von Abatacept (CTLA-4-Ig) untersuchen. Dieses zeigte zum Teil sehr gute Ergebnisse. Unsere Behandlungsempfehlungen legten den Grundstein für eine Therapie der CTLA-4- Insuffizienz. Durch einen internationalen Austausch können aus ihrer Anwendung neue Erkenntnisse gewonnen werden, die schlussendlich jedem einzelnen CTLA4-Mutationsträger dienen.
Article
As the list of regulatory agency-approved gene therapies grows, these products are now in the therapeutic spotlight with the potential to cure or dramatically alleviate several benign and malignant hematologic diseases. The mechanisms for gene manipulation are diverse, and include the use of a variety of cell sources and both viral vector- and nuclease-based targeted approaches. Gene editing has also reached the realm of blood component therapy and testing, where cultured products are being developed to improve transfusion support for individuals with rare blood types. In this review, we summarize the milestones in the development of gene therapies for hematologic diseases, mechanisms for gene manipulation, and implications for transfusion medicine and blood centers as these therapies continue to advance and grow.
Article
Full-text available
Sickle cell disease (SCD) is caused by a 20A>T mutation in the β-globin gene, and can be cured by therapeutic β-globin gene addition into hematopoietic stem cells (HSCs) with lentiviral transduction. However, this method relies upon random integration, leaving the SCD mutation intact and potentially inducing insertional mutagenesis. Genome editing technologies have the potential to correct the SCD mutation without integration, producing adult hemoglobin (Hb) while simultaneously eliminating sickle Hb. In this study, we investigated CRISPR/Cas9-based gene correction for SCD CD34+ cells. Plerixafor-mobilized SCD CD34+ cells were transfected by electroporation using the GMP-compliant, FDA Master File-supported, and scalable MaxCyte GT System to deliver SCD mutation-specific guide RNA at 200mg/ml, SpCas9 mRNA at 200mg/ml or protein at 120mg/ml, and single strand donor DNA with a normal β-globin sequence at 80, 120, or 200mg/ml. We chose Cas9 mRNA and single strand donor DNA due to the ease of clinical grade large-scale production and to avoid the need for viral vector manufacturing. Following erythroid differentiation, gene correction efficiency was evaluated at DNA levels by deep sequencing and at protein levels by reverse-phase HPLC. Cell viability was reduced to 76-87% after electroporation, compared to 90% in the control. We observed high-efficiency genome editing (29-34% gene correction and 49-58% indels) with Cas9 mRNA, showing donor DNA concentration dependence, and editing levels were comparable to Cas9 protein (39% correction and 43% indels). 15-23% Biallelic and 17-26% monoallelic gene correction were detected at the clonal level by colony assay. After erythroid differentiation, up to 54% normal β-globin production was observed with Cas9 mRNA (Figure), comparable to Cas9 protein (67%), while βs-globin amounts were markedly reduced under both conditions (6-10%). Similar correction efficiencies were obtained from two additional SCD patients' CD34+ cells at DNA levels (28-35%) and protein levels (33-56%). These data demonstrate that Cas9 mRNA and single strand donor DNA allow for efficient gene correction in SCD CD34+ cells, exceeding the therapeutic threshold of 20% in SCD. We then evaluated off-target effects on the δ-globin gene, which was reported as a major off-target site in β-globin gene editing due to high homology; however, almost no off-target effects (0.6-1.3% indels) were detected. Interestingly, gene conversion in the 9T>C polymorphism (11bp upstream of SCD mutation) on the β-globin gene was observed, and this conversion always occurred with SCD gene correction (26-33% of SCD gene correction), suggesting that gene conversion is strongly affected by distance from the target site. In addition, we evaluated genome editing among subpopulations of CD34+ cells from 3 healthy donors under the same conditions (normal β-globin to SCD mutation). We observed similar editing efficiencies (conversion and indels) among more immature (CD34+CD133+CD90+) and relatively differentiated populations (CD34+CD133+CD90-, CD34+CD133-, and CD34-) as well as among cells at different phases of the cell cycle (G0/G1, S, and G2/M), suggesting that similar gene correction efficiencies are obtained in all CD34+ cell populations, including the HSC population. We have begun efforts to evaluate gene-corrected SCD CD34+ cell engraftment in the mouse xenograft model, as similarly corrected X-CGD CD34+ cells were engrafted in immunodeficient mice. To examine the effects of indels in the β-globin gene, we next evaluated Hb production from genome-edited SCD CD34+ cells (2 patients) without donor DNA. Editing without donor DNA resulted in 63-70% indels (compared to 26-29% correction and 46-53% indels with donor DNA) and increased non-adult Hb production (small amounts of fetal Hb and significant amounts of a Hb variant), which will require further investigation to characterize. In summary, we observed efficient gene correction in SCD CD34+ cells with a simple Cas9 mRNA, single strand donor DNA, and guide RNA method, resulting in ~30% gene correction and ~50% indels. After erythroid differentiation, the majority of Hb detected was adult Hb; we detected up to 54% normal β-globin production with a marked reduction of βs-globin to ~10%. Evaluation of engraftment potential is required for gene-corrected CD34+ cells, but these methods would be clinically applicable for gene correction in SCD. Figure. Figure. Disclosures Li: MaxCyte, Inc.: Employment. Allen:MaxCyte, Inc.: Employment. Peshwa:MaxCyte, Inc.: Employment.
Article
Full-text available
Gene correction in human long-term hematopoietic stem cells (LT-HSCs) could be an effective therapy for monogenic diseases of the blood and immune system. Here we describe an approach for X-linked sSevere cCombined iImmunodeficiency (SCID-X1) using targeted integration of a cDNA into the endogenous start codon to functionally correct disease-causing mutations throughout the gene. Using a CRISPR-Cas9/AAV6 based strategy, we achieve up to 20% targeted integration frequencies in LT-HSCs. As measures of the lack of toxicity we observe no evidence of abnormal hematopoiesis following transplantation and no evidence of off-target mutations using a high-fidelity Cas9 as a ribonucleoprotein complex. We achieve high levels of targeting frequencies (median 45%) in CD34⁺ HSPCs from six SCID-X1 patients and demonstrate rescue of lymphopoietic defect in a patient derived HSPC population in vitro and in vivo. In sum, our study provides specificity, toxicity and efficacy data supportive of clinical development of genome editing to treat SCID-Xl.
Article
Full-text available
Precise gene editing in hematopoietic stem and progenitor cells (HSPCs) holds promise for treating genetic diseases. However, responses triggered by programmable nucleases in HSPCs are poorly characterized and may negatively impact HSPC engraftment and long-term repopulation capacity. Here, we induced either one or several DNA double-stranded breaks (DSBs) with optimized zinc-finger and CRISPR/Cas9 nucleases and monitored DNA damage response (DDR) foci induction, cell-cycle progression, and transcriptional responses in HSPC subpopulations, with up to single-cell resolution. p53-mediated DDR pathway activation was the predominant response to even single-nuclease-induced DSBs across all HSPC subtypes analyzed. Excess DSB load and/or adeno-associated virus (AAV)-mediated delivery of DNA repair templates induced cumulative p53 pathway activation, constraining proliferation, yield, and engraftment of edited HSPCs. However, functional impairment was reversible when DDR burden was low and could be overcome by transient p53 inhibition. These findings provide molecular and functional evidence for feasible and seamless gene editing in HSPCs. Precise gene editing has the potential to treat immune and hematological diseases. Genovese, Naldini, Di Micco, and colleagues now show that gene-editing procedures are well tolerated by hematopoietic stem cells and provide molecular evidence of the feasibility of seamless gene editing, strengthening translation of such approaches to humans.
Article
Full-text available
Spotting off-targets from gene editing Unintended genomic modifications limit the potential therapeutic use of gene-editing tools. Available methods to find off-targets generally do not work in vivo or detect single-nucleotide changes. Three papers in this issue report new methods for monitoring gene-editing tools in vivo (see the Perspective by Kempton and Qi). Wienert et al. followed the recruitment of a DNA repair protein to DNA breaks induced by CRISPR-Cas9, enabling unbiased detection of off-target editing in cellular and animal models. Zuo et al. identified off-targets without the interference of natural genetic heterogeneity by injecting base editors into one blastomere of a two-cell mouse embryo and leaving the other genetically identical blastomere unedited. Jin et al. performed whole-genome sequencing on individual, genome-edited rice plants to identify unintended mutations. Cytosine, but not adenine, base editors induced numerous single-nucleotide variants in both mouse and rice. Science , this issue p. 286 , p. 289 , p. 292 ; see also p. 234
Article
Full-text available
The type-V CRISPR effector Cas12b (formerly known as C2c1) has been challenging to develop for genome editing in human cells, at least in part due to the high temperature requirement of the characterized family members. Here we explore the diversity of the Cas12b family and identify a promising candidate for human gene editing from Bacillus hisashii, BhCas12b. However, at 37 °C, wild-type BhCas12b preferentially nicks the non-target DNA strand instead of forming a double strand break, leading to lower editing efficiency. Using a combination of approaches, we identify gain-of-function mutations for BhCas12b that overcome this limitation. Mutant BhCas12b facilitates robust genome editing in human cell lines and ex vivo in primary human T cells, and exhibits greater specificity compared to S. pyogenes Cas9. This work establishes a third RNA-guided nuclease platform, in addition to Cas9 and Cpf1/Cas12a, for genome editing in human cells. The Cas12b family of CRISPR nucleases has been underutilized in mammalian cells due to the high temperature requirement of known members. Here the authors engineer BhCas12b to overcome this limitation for robust and specific genome editing applications in human cells.
Article
Full-text available
Clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) is a precise genome manipulating technology that can be programmed to induce double-strand break (DSB) in the genome wherever needed. After nuclease cleavage, DSBs can be repaired by non-homologous end joining (NHEJ) or homology-directed repair (HDR) pathway. For producing targeted gene knock-in or other specific mutations, DSBs should be repaired by the HDR pathway. While NHEJ can cause various length insertions/deletion mutations (indels), which can lead the targeted gene to lose its function by shifting the open reading frame (ORF). Furthermore, HDR has low efficiency compared with the NHEJ pathway. In order to modify the gene precisely, numerous methods arose by inhibiting NHEJ or enhancing HDR, such as chemical modulation, synchronized expression, and overlapping homology arm. Here we focus on the efficiency and other considerations of these methodologies.
Article
Full-text available
Cas9 is an endonuclease that can be programed to autonomously deliver diverse effectors to specified genetic addresses. High-resolution structures of this protein and its associated CRISPR RNA guide explain the molecular mechanisms of CRISPR-RNA-guided DNA recognition and provide a molecular blueprint that has facilitated structure-guided functional remodeling. Here we retrace events that led from early efforts to understand the central role of Cas9 in CRISPR-mediated adaptive immunity to contemporary efforts aimed at developing and deploying this enzyme for programmable genetic editing.
Article
Full-text available
The prokaryotic CRISPR-Cas adaptive immune systems provide valuable resources to develop genome editing tools, such as CRISPR-Cas9 and CRISPR-Cas12a/Cpf1. Recently, CRISPR-Cas12b/C2c1, a distinct type V-B system, has been characterized as a dual-RNA-guided DNA endonuclease system. Though being active in vitro, its cleavage activity at endogenous genome remains to be explored. Furthermore, the optimal cleavage temperature of the reported Cas12b orthologs is higher than 40 °C, which is unsuitable for mammalian applications. Here, we report the identification of a Cas12b system from the Alicyclobacillus acidiphilus (AaCas12b), which maintains optimal nuclease activity over a wide temperature range (31 °C–59 °C). AaCas12b can be repurposed to engineer mammalian genomes for versatile applications, including single and multiplex genome editing, gene activation, and generation of gene mutant mouse models. Moreover, whole-genome sequencing reveals high specificity and minimal off-target effects of AaCas12b-meditated genome editing. Our findings establish CRISPR-Cas12b as a versatile tool for mammalian genome engineering.
Article
Acute myeloid leukemia (AML) blood cancer is found primarily in adults and the elderly, being the third biggest blood cancer killer. In the last decades there has been some improved rates in younger patients, but little improvement in older patients. AML survival rates are the worst of all blood cancers, therefore finding new therapeutic targets to treat AML is crucial. Bruton's tyrosine kinase (BTK) is involved in the signalling of multiple receptors including growth factor receptors, cytokine receptors, G-protein coupled receptors, antigen receptors and integrins. BTK in turn activates many of the major downstream signalling pathways that control cell migration, adhesion, survival and proliferation. BTK is best known for its role in B-cell receptor signalling, but BTK is also expressed and constitutively active in AML cell lines and AML patient blasts. Targeting BTK in B-cell malignancies with the BTK inhibitor ibrutinib has shown clinical effectiveness and tolerability in recent patient trials. Ibrutinib is orally active and covalently binds at the Cys-481 in the ATP-binding domain of BTK, to irreversibly block its enzymatic activity. A proportion of CLL and MCL patients treated with ibrutinib relapse during the therapy, with secondary resistance developing against ibrutinib. This resistance is mainly due to a BTK cysteine to serine mutation at position 481 (C481S) which disrupts ibrutinib's covalent binding to BTK. As little is known about BTK's role and therapeutic potential in AML, we aimed to study the effects and functional consequences of pharmacological inhibition of BTK using ibrutinib. We furthermore generated the BTK-C481S mutation in AML cell lines using the CRISPR-Cas9 gene-editing system. Shown here for the first time is a gene-edited mutation introduced into a human leukemia cell clone that has been purified to homogeneity. We explored the functional impact of BTK-C481S mutation on ibrutinib sensitivity, and mutation-induced adaptation of the phosphoproteome and kinome in AML cells. HEK-293T cells were used for validation purposes to find the most efficient knock-in strategy. We tested the most commonly used CRISPR endonuclease spCas9 as well as two other Cas9 orthologs called SaCas9 and AsCas12a. The designed spacer sequences within the sgRNA that target the gene of interest were delivered either via plasmid or in vitro transcripts. A repair template for homology directed repair (HDR) was designed containing the necessary base changes to change the amino acid at position 481 in BTK from cysteine to serine. The HDR template was delivered via transfection as single-stranded oligodeoxynucleotide (ssODN) or incorporated into plasmid. All strategies tested were successful but due to the poor transfectability of AML cell lines, we used lentiviral delivery to introduce BTK-C481S mutation into AML cell lines. The BTK-C481S mutation was introduced in THP-1 and OCI-AML3 cells. Mutant-positive cells were found via clonal selection and Sanger sequencing. Wild-type (WT) as well as mutant cells were treated with various ibrutinib concentrations ranging from 1 nM to 1 µM and analysed for BTK expression/phosphorylation via Western blot, with effects on cell proliferation, viability and cell cycle measured via flow cytometry. We also performed phosphoproteomics and kinome assays comparing untreated as well as treated (30 nM ibrutinib, 2h) THP-1 WT and THP-1 BTK-C481S cells. Western blot results showed that the effective concentration required for BTK inhibition is 30-fold higher in BTK-C481S mutant compared to WT cells (300 nM vs 10 nM). Wash-out experiments revealed that ibrutinib binding can be reversed in BTK-C481S mutant cells but not WT cells. Little effect of ibrutinib on proliferation and viability were seen comparing BTK-C481S mutant and WT AML cells. These signalling adaptations in BTK-C481S cells are consistent with changes observed in ibrutinib-resistant leukemia patient samples. Disclosures Slupsky: Verastem: Research Funding.
Article
CRISPR technology is a two-component gene editing system in which the effector protein induces genetic alterations with the aid of a gene targeting guide RNA. Guide RNA can be produced through chemical synthesis, in vitro transcription, or intracellular transcription. Guide RNAs can be engineered to have chemical modifications, alterations in the spacer length, sequence modifications, fusion of RNA or DNA components, and incorporation of deoxynucleotides. Engineered guide RNA can improve genome editing efficiency and target specificity, regulation of biological toxicity, sensitive and specific molecular imaging, multiplexing, and editing flexibility. Therefore, engineered guide RNA will enable more specific, efficient, and safe gene editing, ultimately improving the clinical benefits of gene therapy.