Abstract
Sickle cell disease (SCD) is one of the most common life threatening monogenic diseases affecting millions of people worldwide. Allogenic hematopietic stem cell transplantation is the only known cure for the disease with high success rates but the limited availability of matched sibling donors and the high risk of transplantation-related side effects force the scientific community to envision additional therapies. Ex vivo gene therapy through globin gene addition has been investigated extensively, and is currently being tested in clinical trials which have begun reporting encouraging data. Recent improvements in our understanding of the molecular pathways controlling mammalian erythropoiesis and hemoglobin offer new and exciting therapeutic options. Rapid and substantial advances in genome engineering tools, particularly CRISPR/Cas9, have raised the possibility of genetic correction in induced pluripotent stem cells (iPSCs) as well as patient-derived hematopoietic stem and progenitor cells (HSPCs). However, these techniques are still in their infancy and safety/efficacy issues remain that must be addressed before translating these promising techniques into clinical practice.
Keywords: Sickle cell anemia, genome engineering, BCL11A, gene addition, fetal globin
Introduction
Sickle cell disease (SCD) is a severe hereditary form of anemia that results from a single mutation at the sixth codon of the β-globin chain (from glutamic acid to valine) of the adult hemoglobin (Hb) tetramer (α2β2) [1], that is prone to polymerization at low oxygen levels. It is one of the most prevalent and severe monogenetic disorders, and more than 100,000 individuals in the United States and several million around the world are affected by both acute and chronic manifestations of SCD such as frequent pain crises, silent cerebral infarct, stroke, end organ damage and early death [2]. Polymerized sickle hemoglobin (HbS, α2βS2) interferes with red blood cell biconcave architecture and flexibility, resulting in crescent shaped cells with enhanced adherence to the vascular endothelium, and hemolysis, that obstructs the blood flow [3]. More detailed reviews on pathophysiology [4] and genetics [5] of SCD are available in the literature for further reading.
Since its original description more than a century ago [6], treatments that only reduce the symptoms and complications of SCD such as blood transfusions, preventive therapies including penicillin prophylaxis and pneumococcal vaccination, and hydroxyurea therapy have been leveraged in the clinics. Blood transfusion, however, does not correct the phenotype and results in iron overload when not accompanied by aggressive chelation therapy. Hydroxyurea treatment provides clinical benefit through the induction of fetal globin (HbF, α2γ2) which competes with sickle globin; thus, reducing SCD symptoms, but response to hydroxyurea is not uniform among patients and concerns for long term use remain despite abundant evidence for safety [4]. On the other hand, substantial advances in cellular and molecular biology have led to some powerful tools that we have begun to employ. Of those, allogenic hematopoietic stem cell (HSC) transplantation is as of yet the only available curative option for patients with severe disease [7]. Despite the considerably high success rate of HSC transplantation, a significant proportion of the candidates (>80%) do not have a suitable matched sibling donor, and there remains a risk for graft rejection, graft-versus host disease (GVHD) and transplant related mortality [8–11]. Improvements have been made with reduced intensity condition; however, this approach also remains limited by donor availability [9].
For those lacking a suitable allogeneic HSC donor, genetic strategies targeting autologous HSCs remains an alternative (Figure 1). In theory, as genetically modified therapeutic cells are of patient origin, the risk for GVHD and transplant rejection can be virtually abrogated, abrogating the need for immunosuppression as part of the conditioning regimen. Primary results obtained from clinical trials with genetically modified autologous HSCs expressing potential therapeutic genes for immunodeficiency disorders [12–21] have encouraged a focus in blood related diseases. The globin disorders, though long held as a therapeutic target, have proven much more difficult because of the necessity of regulated, lineage specific, high-level globin expression. In this review, general gene therapy approaches for SCD including gene addition and genome editing technologies for decreasing SCD symptoms by enhancing HbF or correcting the mutation in the β-globin sequence will be outlined.
Stable gene addition with lentiviral vectors
HSCs are limited in the human body and prolonged cultivation of HCSs in in vitro conditions changes the potential of stem cells. Therefore, determining efficient gene transfer systems providing stable expression of a target gene at therapeutic levels without leading to any safety concerns after modification, such as an immunogenic response or oncogenesis due to random insertional mutagenesis, is one of the main hurdles to clinical application. After the first gene therapy trial using a γ-retroviral vector to transduce mobilized CD34+ cells for the treatment of severe combined immunodeficiency (SCID) was reported [22], the potential of these viral vectors has been widely investigated for various hematological disorders. However, the use of the γ-retroviral vectors in clinical trials were marred because they are not capable of transducing non-dividing cells including HCSs in quiescent state, cannot carry large gene sets such as β-globin and its regulatory elements required for high level of expression, and present relatively instable RNA to be reverse transcribed and delivered to the nucleus [23, 24]. Most importantly, treatment with γ-retroviral vector-transduced HSCs in various disorders led to leukemia or myelodysplasia, which was attributed to vector insertion near protooncogenes, activated due to enhancer sequences in the retroviral long terminal repeats (LTRs) [25]. The development of human immunodeficiency virus type 1 (HIV-1), belonging to lentivirus family, derived vectors circumvent these safety and efficacy issues. Apart from transducing non-dividing cells and transferring large sequences, they have displayed a safe profile without any sign of insertional oncogenesis or mutagenesis in SCD and β-thalassemia patients for 4 to 30 months [26, 27]. The self-inactivating (SIN) design in the U3 region of the 3’ LTR region, removing accessory genes, and separating packaging components, have made lentiviral vectors a relatively safer option, reducing the possibility of insertional oncogenesis, genotoxicity and the development of replication competent counterparts [28]. While lentiviral vectors have the confidence of the scientific community, recent works suggests that foamy viral vectors might also provide a safe option [29, 30].
β-globin gene addition
The initial idea in gene therapy applications to inhibit HbS polymerization emerged with the introduction of a functional β-globin transgene into HSCs. However, efficient and erythroid-specific expression of transgene in reconstituting HSCs remained insufficient in in vivo studies [31, 32]. Incorporation of the human β-globin locus control region (LCR) became feasible with the development of the lentiviral vector systems based upon HIV-1, and the model was first demonstrated in a murine model of β-thalassemia [33]. These results were repeated and extended in both thalassemia and SCD mouse models [34–40], we subsequently established a preclinical, rhesus macaque model for lentiviral globin gene transfer. Mobilized peripheral blood progenitor cells, transduced with a vesicular stomatitis virus-G (VSV-G) pseudotyped, modified HIV-1 based vector [41], expressed significant levels of human β-globin (>50%) among erythroid progeny generated in vitro. In vivo studies showed more modest levels of human β-globin expression at around 5% early post-transplantation and stabilized at lower levels at two years [42], due to species-specific transduction restriction of rhesus progenitor cells by HIV-1 vector. Our newly developed chimeric vector containing the SIV capsid to circumvent the species-specific block to HIV in old world monkeys has allowed us to obtain high-level engraftment of genetically modified cells carrying erythroid specific cassettes at levels now exceeding 20% [43], and predicting ultimate clinical success [44].
Anti-sickling β-globin gene addition
Sickle hemoglobin polymerization is the main reason for structural deformation in red blood cells. Initial studies proved that fetal globin or its mixed hybrid tetramer (α2βSγ) do not contribute (or to a much lesser extent) to the deoxyHbS polymerization [45]. The amino acid, threonine (T87), at position 87, is replaced by a glutamine (Q87) in beta globin providing less tendency to contact with sickle beta subunit valine at position 6 [46].
In a series of studies, the β-globin sequence has been modified to increase the anti-sickling activity based on the observation of less polymerization for α2βSγ with respect to α2βSβ. Kinetic studies have proved that conversion from T87 to Q87 is the main parameter responsible for most of the sickle inhibiting activity among other 10 amino acid differences between β and γ-globin sequences [47]. Mutation in the sequence of β-globin (βA(T87Q)) provided strong anti-sickling properties as efficient as γ-globin in reversing the phenotype in two relevant SCD mouse models, BERK and SAD [48]. In addition to its anti-sickling activity, the modified β-globin can be separated from other globins in reverse-phase high-performance liquid chromatography analysis which makes it more convenient in clinical trials [49]. Results for a multi-center phase ½ clinical study for adults with severe SCD have been reported and updated recently [26, 50]. A pretreatment transfusion regimen along with improved CD34+ cell collection and manufacturing conditions, along with improvement in myeloablation have resulted in higher peripheral blood vector copy numbers (VCNs) in SCD patients (HGB-206, ClinicalTrials.gov Identifier: NCT02140554) with levels reaching the therapeutic range. In addition, the use of plerixafor, a CXCR4 receptor antagonist, to drive CD34+ mobilization has now been adopted in the same clinical trial [51], providing less invasive cell collection with respect to bone marrow harvest and safer mobilization as compared G-CSF treatment which historically has led to life-threatening complications when used in individuals with SCD [52]. Research scale levels of transduction with the drug products remained similar for peripheral blood or bone marrow derived cells from the same patient, indicating plerixafor-based HSC mobilization would be a safer and feasible method for clinical gene therapy applications.
An updated report for the HGB-205 trial recently published demonstrated that one patient with severe SCD showed no SCD complications for up to 30 months after the LentiGlobin treatment, had stable Hb (12.4 g/dL), βA(T87Q) (6.1 g/dL) and peripheral blood VCN (2.3) levels [53]. Consistently, expression of βA(T87Q) in the patient that was transplanted with autologous CD34+ cells transduced with LentiGlobin BB305 was sufficient to reverse markers of hemolysis and provide stable hemoglobin levels [54], similar to that seen in sickle cell trait (SCT) for 15 months of follow up [55]. While the initial results are quite encouraging, clinical trials testing these approaches are still ongoing (Table 1, as of February 2018), and larger clinical trials along with extended follow-up will be required to establish the safety and efficacy of these vector-based approaches for broad application in SCD patients.
Table 1.
ClinicalTrials.gov Identifier | Official Title | Therapeutic Gene | Age Eligibility/Phase | Status | Sponsor |
---|---|---|---|---|---|
NCT02186418 | Gene Transfer for Patients With Sickle Cell Disease Using a Gamma Globin Lentivirus Vector: An Open Label Phase I/II Pilot Study | γ-globin | 18-35/Phase I/II | Recruiting | Children’s Hospital Medical Center, Cincinnati |
NCT02247843 | Clinical Research Study of Autologous Bone Marrow Transplantation for Sickle Cell Disease (SCD) Using Bone Marrow CD34+ Cells Modified With the Lenti/βAS3-FB Lentiviral Vector | βAS3-FB (Antisickling β-globin) | 18 and older/Phase I | Recruiting | Donald B. Kohn, M.D. (University of California, Los Angeles) |
NCT02193191 | Safety and Efficacy Trial of Escalation of Plerixafor for Mobilization of CD34+ Hematopoietic Progenitor Cells and Evaluation of Globin Gene Transfer in Patients With Sickle Cell Disease | β-globin | 18-65/Phase I | Recruiting | Memorial Sloan Kettering Cancer Center |
NCT02140554 | A Phase 1 Study Evaluating Gene Therapy by Transplantation of Autologous CD34+ Stem Cells Transduced Ex Vivo With the LentiGlobin BB305 Lentiviral Vector in Subjects With Severe Sickle Cell Disease | T87Q (Antisickling β-globin) | 18 and older/Phase I | Recruiting | Bluebird Bio |
NCT02633943 | Longterm Follow-up of Subjects With Hemoglobinopathies Treated With Ex Vivo Gene Therapy Using Autologous Hematopoietic Stem Cells Transduced With a Lentiviral Vector | T87Q (Antisickling β-globin) | 5-50/NA | Enrolling by invitation | Bluebird Bio |
NCT02151526 | Phase 1/2 Open Label Study Evaluating the Safety and Efficacy of Gene Therapy of the Beta-Hemoglobinopathies (Sickle Cell Disease and Beta- Thalassemia Major) by Transplantation of Autologous CD34+ Stem Cells Transduced Ex Vivo With a Lentiviral Beta- A-T87Q Globin Vector (LentiGlobin BB305 Drug Product) | T87Q (Antisickling β-globin) | 5-35/Phase I/II | Active, not recruiting | Bluebird Bio |
As modification at β16, glycine to aspartic acid, serves a competitive advantage over sickle globin (βS, HbS) for binding to α chain, and modification at β22, glutamic acid to alanine, partially enhances axial interaction with α20 histidine, double mutant (βAS2; T87Q and E22A) [56] and triple-mutant β-globin variants (βAS3; T87Q, E22A and G16D) [57] have also been developed. These modifications provide anti-sickling properties greater than the T87Q alone modified variant, and comparable to fetal globin. In a SCD murine model, transplantation of bone marrow stem cells transduced with SIN lentivirus carrying βAS3 reversed red blood cell physiology and SCD clinical symptoms [58]. Based on these encouraging in vitro and in vivo data, this variant is now currently being tested in a clinical trial (Identifier no: NCT02247843).
γ-globin gene addition
Globin synthesis in humans is control by developmentally regulated gene expression, known as globin switching. HbF is the dominant hemoglobin type after the first trimester of gestation, which is gradually replaced by adult hemoglobin after birth. The contribution of HbF in adults is generally lower than 1% and is not evenly distributed in red cells but rather, concentrated in particular cells, referred to as F-cells [59]. The appreciation of the ameliorative role of HbF in the blood of patients with SCD began 70 years ago, when Janet Watson and colleagues demonstrated deoxygenated erythrocytes derived from infants with SCD showed delayed sickling compared to their sickle mother’s erythrocytes in in vitro conditions [60]. This low rate of sickling was attributed to high levels of HbF in the infants’ blood, with increased sickling noted at later time points after a decline of HbF. This initial observation was further confirmed by others, with the demonstration that SCD and β-thalassemia patients with hereditary persistence of fetal hemoglobin (HPFH) syndrome, having red blood cells with pancellular distribution of HbF with a typical level of 30%, have a marked reduction of their clinical and hematologic severity [61–63].
After discovering the ameliorative effect of HbF for SCD patients, efforts to increase γ-globin expression by direct or indirect approaches were vigorously followed. As observed for β-globin encoding vectors, γ-globin encoding vectors were similarly insufficient [64]. The issue of tissue specificity have been partially overcome using different vector designs such as linking the ankyrin promoter to the γ-globin cassette to provide erythroid specific expression [65] or using β-globin regulatory sequences and promoter elements to provide cell specificity and high level expression [66]. As a different approach, Samakoglu and her colleagues used a γ-globin transgene expression and concomitant βS RNA interference under the control of LCR and γ-globin promoters in sickle cell patient-derived CD34+ cells [67]. With both a reduction in βS and an increase in γ-globin, an enhancement of anti-sickling activity would be expected [68]. To provide stable and therapeutic level of gene expression for clinical studies, γ/β hybrids have been constructed to benefit from LCR regulatory elements of β-globin and anti-sickling properties of γ-globin. One of those γ/β hybrids gene carrying construct, driven by ankyrin-1 promoter and controlled by two erythroid specific enhancers (HS-40 plus GATA-1 or HS-40 plus 5-aminolevulinate synthase intron 8 [I8] enhancers) provided 43-113% human γ-globin/copy of murine α-globin [69].
Targeted γ-globin induction
Having the knowledge of chromosomal orientation, various transcription factors regulating expression of the globins; controlling these parameters to induce HbF has always been an attractive research area for SCD. γ-to-β globin switching in neonatal period is driven by β-globin LCR, modulating β-like globin expressions by direct contact with their promoters. The shift of interaction between promoters and the LCR region is driven by transcription factors including Gata1, Tal1, E2A, Lmo2, and Ldb1 [70]. Recently, an innovative strategy based on artificially changing the binding site of LCR has emerged to mediate globin switching. Deng and colleagues reported that fusion protein of artificial zinc finger (ZF) and Ldb1, or its self-association domain, can rescue the interaction between β-globin promoter and LCR in Gata-null erythroblasts [71]. The ZF-Ldb1 can be effectively targeted to the γ-globin promoter, leading to reactivation of γ-globin expression (85% of total β-like globins) and a concomitant reduction in β-globin expression in primary adult human erythroblasts [72]. The approach was tested in hematopoietic progenitor cells derived from sickle patients as well [73]. Transducing SCD CD34+ cells with a lentiviral vector encoding ZF-Ldb1 upregulated HbF production and reduced erythrocyte sickling in vitro with greater efficiency than chemical HbF inducers including butyrate, hydroxyurea and debicatine. From a SCD perspective, decreasing sickle globin along with augmenting anti-sickling γ-globin expressions would increase the clinical benefits.
ZF designs were also fused with transcription factors regulating γ-globin expressions. ZF directed to −117-position of γ-globin promoter, that is open to DNA-binding proteins and close enough to known transcriptional regulators of the gene, were fused with the potential transcriptional factors [74]. Transducing K562 cells with retrovirus expressing gg-VP64, a transcriptional activator domain, induced HbF expression 7-16-fold. Wilber and colleagues brought this technology to human CD34+ cells [75]. Significant induction of HbF (up to 20%) was achieved by overexpressing gg-VP64 domain driven by an erythrocyte specific promoter, ankyrin-1.
γ-globin repressor silencing
Early works and genome-wide association studies (GWAS) in blood donors revealed DNA polymorphisms in BCL11A and HBS1L-MYB genes that were associated with elevated HbF expression [63, 76, 77]. The newly discovered fetal globin regulating candidate, bcl11a, was confirmed by its γ-globin repressing roles in primary erythroid cells by re-configuration of the locus region by binding to transcription factors such as GATA-1 and SOX-6 [78]. Moreover, conditional deletion of bcl11a in erythroid cells using a SCD mouse model suggested that targeting this repressor was sufficient enough to reverse the SCD manifestations in vivo [79], showing the importance of this gene as potential target for further clinical studies. The question of “to what extent does BCL11A control HbF?” has partly been answered by two recent clinical studies reporting autism spectrum disorder patients with rare microdeletions of 2p15-p16.1, who had haploinsufficient bcl11a, expressed around 20% HbF levels, indicating the central and strong role of bcl11a on γ-globin silencing [80, 81]. In addition to BCL11A, other key trans-acting factors such as klf1 and myb have been studied in genetic and functional models [82, 83]. Continued investigation of these and other factors involved in fetal hemoglobin repression will help to inform clinical approaches leveraging genome editing technologies.
Genome editing technology
Along with the development of human induced pluripotent stem (iPS) cells by Yamanaka and colleagues [84] through the introduction of specific reprogramming factors, circumventing the ethical issues surrounding embryonic stem cells, solid advances in genome editing with methods including zinc finger nucleases (ZFNs), transcription-activator like effector nucleases (TALENs), and clustered regularly interspaced palindromic repeats (CRISPR)-associated protein-9 nuclease (Cas9) have brought the potential to transform promising ideas to the clinics. These nucleases are proven to efficiently edit genomes of various animal models and human cells, and great efforts to bring these tools to the clinic have been pursued by academia and industry. These editing tools efficiently disrupt genes at defined loci, and can also be used to promote homology directed repair (HDR) as a means of gene correction, although HDR is less efficient. As β-globin disorders such as SCD are monogenic diseases, genome editing aiming to correct the mutated β-globin gene would be applicable to all affected patients, making this disorder an ideal target. Additionally, the higher efficiency of cutting (over correcting) genes at specific loci along with the ameliorative effect of HPFH on the SCD phenotype makes genome editing at HPFH loci an attractive initial approach. The induction of HbF synthesis by editing regulatory sequences such as promoters or other regulatory sequences such as BCL11A are straightforward approaches for a radical cure of the beta-hemoglobinopathies. Genome editing tools are mainly based on the creation of double strand-break using an endonuclease at a specific site in the genome leading to activation of auto-repair machinery system using either non-homologous end joining (NHEJ) or the error-free homology directed repair (HDR) pathways [85]. The CRISPR/Cas9 system for genome editing contains an endonuclease that cuts DNA, a guide RNA to direct where the DNA cut occurs, and template DNA which serves as the correct copy of the gene which is targeted. There are a number of delivery systems for transporting these tools to cells that depend on the application, but efficiency is increasing such that double strand breaks at specific loci can be achieved at 90% or higher rates, with lower rates of HDR achieved thus far. As HCSs are mostly quiescent (G0/G1), gene disruption will prove easier than gene correction, which is more likely among committed progenitor cells (S/G2) [86]. Regardless of such drawbacks, understanding the mechanism of action for each editing system, introduction of new chimeric technologies and continuous interest of scientists would help to meet these kinds of technical challenges.
Gene correction through genome editing
While gene addition strategies have enjoyed outstanding progress recently, with translation into clinical research now ongoing, genome editing methods with programmable nucleases targeting precise sites of disease associated mutations have been introduced to tackle fundamental concerns of gene addition such as insertional mutagenesis and low transduction efficiency of the viral systems transferring large cargos. The holy grail of genome editing for the β-hemoglobinopathies is the correction of the β-globin mutation either in patient-derived HSCs or patient derived iPSCs which while easier, would then require conversion to engraftable HSCs. Indeed, iPSCs are infinitely more amenable to genome editing than primary cells [87], they remain encumbered as HSC derivation from pluripotent stem cells with engraftment potential remains insufficient at the moment, and safety concerns remain to be addressed before the introduction of iPSCs to treat the β-globin disorders. Patient-derived HSCs, however, remain a suitable option for corrective applications for now as iPSCs are not currently applicable in the clinical practice due to safety concerns. However, experimental data obtained from animal models conducted with HSC-like cells derived from iPSCs are quite encouraging. In a humanized SCD mouse model, transplantation of hematopoietic progenitors obtained from autologous skin fibroblast-derived iPSCs, that were corrected with normal β-globin, rescued sickle cell manifestations [88]. As a starting point to transfer the method to human, earlier attempts to correct β-globin mutation in human SCD patient-derived blood progenitor cells or iPSCs by ZFNs [87, 89, 90] and TALENs [91–93] have shown the potential of genome engineering to cure the disease. Even though these nucleases can be designed quite specific and off-target problem is not generally an issue, they are expensive, labor intensive, time consuming and requiring expertise. The CRISPR/Cas9 system has emerged as another option that has proven easy to design and can deliver high efficiency genome editing. As with all such systems, minimization of off-target effects remains crucial. Improving DNA specificity by modified nucleases such as Cas9 nickases (Cas9n), dimerization dependent dead Cas9-FokI chimeric enzyme, or high-fidelity versions of Cas9 mutated in DNA binding grove to increase the nuclease specificity, have reduced off-target activity (reviewed in [86]). Also, the correction rates in sickle patient derived progenitor cells are encouraging. Hoban et al. presented 20% sickle correction rate in bone marrow CD34+ cells electroporated with Cas9 mRNA and transduced with integrase-free lentiviral vector carrying gRNA and β-globin gene donor template [94]. This level of correction might be clinically relevant as 20% donor chimerism provides significant improvements and reverse the disease manifestations in allogenic HSC transplanted SCD patients [44]. Almost 30% correction of red blood cells derived from edited sickle CD34+ cells was recently reported [95]. Transplantation of these corrected cells to NSG mouse revealed long-term maintenance and repopulation of HDR-edited HSCs in both bone marrow (2.3%) and spleen (3.7%). While the results are quite encouraging, correction of cells with long-term repopulating activity requires further optimization. To overcome this limitation, the truncated nerve growth factor receptor (tNGFR) was recently used to enrich HBB-targeted HSCs using magnetic bead separation [96]. These strategies are beginning to show some promise for SCD patients, and a focus on limiting off-target activity is currently being vigorously pursued.
Targeted γ-globin induction through genome editing
Elevated HbF has a long-known beneficial role in SCD and experiments of nature have suggested increasing HbF as a treatment for SCD such as in HPFH. Increasing the HbF levels in sickle erythrocytes before severe organ damage might be beneficial in the preventing the relentless complications of this disease. Elevating the HbF levels through specific genome editing tools has emerged as a promising strategy for SCD treatment. BCL11A was one of the first targets for gene editing after the introduction of these robust and relatively simple genome engineering approaches. However, complete knockout of bcl11a is lethal in mice due to the deficiencies in neural and lymphoid development [97]. Therefore, rather than creating full knockout of BCL11A activity in HSCs, a search for the erythroid specific enhancer could provide a target free of these complications. In a recent study, CRISPR/Cas9 mediated saturating mutations have been introduced to erythroid cells to evaluate effects on HbF levels in red blood cells. This tour de force led to the identification of a particular site in the BCL11A erythroid specific enhancer region, enabling the production of significant HbF without effecting HSC behavior or lymphoid development [98, 99]. In another similar approach, Tan et al. used specific ZFNs for targeting two different regions of bcl11a to increase HbF production in bone marrow derived CD34+ cells. Ablation of bcl11a resulted in reactivation of HbF in erythroid cells and maintained the engraftment capacity indicating the clinical relevance of current gene editing strategy [100]. More recently, disruption of exon-2 and GATAA motif in the intronic erythroid-specific enhancer of bcl11a in bone marrow-derived CD34+ using targeted zinc finger nucleases was reported to elevate HbF expression while exon-2 deletion negatively impacted ex vivo enucleation and engraftment in immunodeficient mice [101].
Other than BCL11A, known HPFH mutations in the β-globin locus were also created in CD34+ cells using CRISPR/Cas9 that increased γ-globin expression in RBC generated in ex vivo culture [102, 103]. HbF upregulation in modified progenitor cells is promising, and efforts to achieve reliable engraftment should be pursued as the technologies require electroporation of HSCs, a process that initially led to the development of viral vectors more than a quarter of a century ago due to the deleterious effects of electroporation on viability.
Although there is a great focus on genome editing researches especially for monogenic disorders such as SCD, there are still serious limitations and considerations. Several delivery methods for genome editing tools in the forms of DNA, RNA or protein have been presented in the literature including electroporation, cationic lipids, cell penetrating peptides, nanoparticles and viral vectors, offering variable pros and cons (reviewed in [104, 105]). The use of integrating viral vectors with persistent expression of Cas9 and guides RNAs allows one to study the activity of the system, but persistent Cas9 expression is not desirable for most clinical application [106]; hence, transient nuclease activity would be preferable and convenient from a clinical perspective. Nucleases can be applied both ex vivo and in vivo to target the HSCs but ex vivo editing would be advantageous as HSCs can be cultured and manipulated ex vivo allowing controlled delivery of enzymes especially where off-target modifications are problematic, and higher levels of editing rates are needed. However, HSCs can lose their stem cell properties and engraftment ability in culture conditions due to epigenetic instability in laboratory conditions [107]. A reduction in engraftment ability can be augmented by delivering ablative conditioning, but ablative conditioning carries significant risks [106].
Improved conditioning for better tolerability
Better conditioning regimens offering less overall toxicity and higher specificity would circumvent the concerns over current chemoradiotherapy based regimens and increase the success rate of HSC-based genetic therapies. Targeting HSCs in bone marrow while sparing non-hematopoietic cells using specific antibodies has emerged as an attractive alternative to genotoxic chemoradiotherapy. These nontoxic approaches are especially appropriate for blood disorders like SCD where end accumulated organ damage compounds toxicity of existing conditioning regimens. A monoclonal antibody, ACK2, targeting c-kit was recently demonstrated with or without low-dose irradiation to eliminate host blood progenitor cells before the donor cell transplantation [108, 109]. Considerable level of endogenous HSC clearance and 90% donor chimerism after subsequent transplantation were achieved. As a different strategy, others have targeted CD45 with antibody conjugated to ribosome-inactivating protein, Saporin (CD45-SAP) to target HSCs in an immunocompetent mouse model [110]. Efficient donor cell engraftment (> 90%) was obtained with a single dose of CD45-SAP administration without neutropenia and anemia, and most interestingly, rapid recovery of T and B cells, and sustained anti-fungal immunity were reported along with correction of sickle manifestations in vivo. These promising strategies could transform transplantation approaches for genetic diseases affecting the blood if these results can be replicated in large animals and humans.
Conclusion
Recent advances in gene transfer and editing technologies, better clarification of erythropoiesis regulation, and improved transplantation methods have driven optimism in developing a onetime definitive cure for SCD. Gene addition trials has already shown that high-level expression of anti-sickling β-globin can reverse the complications of the disease (NEJM article). With the improvements in cell processing, viral vectors, and transduction methods, we anticipate validation of these encouraging results over the short term, allowing broader application of potentially curative strategies to patients with SCD. In addition, nature has already shown us that persistent, high HbF expression through coinheritance of HBFH mutations can ameliorate the clinical severity of SCD, opening another potentially curative strategy. If genetic approaches can be adapted and standardized at high efficiency, increasing HbF protein levels in the blood of patients with SCD by gene addition or controlling gene repressors and activators of γ-globin through gene editing would be an additional option, and preclinical results are already promising for clinical trials to open in the coming year. Finally, a dream therapy for SCD has recently become imaginable through correction of the disease-causing mutation in patient-derived iPSCs and blood progenitor/stem cells with the introduction of relatively easy and feasible genome editing tools, particularly CRISPR/Cas9. There remain several issues to be addressed including delivery of these tools to HSCs without compromising their engrafting ability, achieving high efficiency correction and minimizing/eliminating undesirable off-target editing. Overcoming the current technical challenges of the newer candidate approaches appear surmountable, and continued rapid progress should finally lead to widely available curative approaches for the hemoglobinopathies.
Acknowledgments
The authors thank to Bjorg Gudmundsdottir for her valuable opinions on the paper.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosure of interest
The authors have no commercial, proprietary, or financial interest in the products or companies described in this article.
References
- 1.Ingram V. A specific chemical difference between the globins of normal human and sickle cell anemia hemoglobin. Nature. 1956 Oct 13;178:792–794. doi: 10.1038/178792a0. [DOI] [PubMed] [Google Scholar]
- 2.Piel FB, Patil AP, Howes RE, Nyangiri OA, Gething PW, Dewi M, Temperley WH, Williams TN, Weatherall DJ, Hay SI. Global epidemiology of sickle haemoglobin in neonates: a contemporary geostatistical model-based map and population estimates. The Lancet. 2013;381(9861):142–151. doi: 10.1016/S0140-6736(12)61229-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Atkins RC, Walters MC. Haematopoietic cell transplantation in the treatment of sickle cell disease. Expert opinion on biological therapy. 2003;3(8):1215–1224. doi: 10.1517/14712598.3.8.1215. [DOI] [PubMed] [Google Scholar]
- 4.Ware RE, de Montalembert M, Tshilolo L, Abboud MR. Sickle cell disease. The Lancet. 2017 doi: 10.1016/S0140-6736(17)30193-9. [DOI] [PubMed] [Google Scholar]
- 5.Steinberg MH, Sebastiani P. Genetic modifiers of sickle cell disease. American journal of hematology. 2012;87(8):795–803. doi: 10.1002/ajh.23232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Savitt TL, Goldberg MF. Herrick’s 1910 case report of sickle cell anemia: the rest of the story. Jama. 1989;261(2):266–271. [PubMed] [Google Scholar]
- 7.Bhatia M, Walters M. Hematopoietic cell transplantation for thalassemia and sickle cell disease: past, present and future. Bone marrow transplantation. 2008;41(2):109–117. doi: 10.1038/sj.bmt.1705943. [DOI] [PubMed] [Google Scholar]
- 8.Hsieh MM, Fitzhugh CD, Weitzel RP, Link ME, Coles WA, Zhao X, Rodgers GP, Powell JD, Tisdale JF. Nonmyeloablative HLA-matched sibling allogeneic hematopoietic stem cell transplantation for severe sickle cell phenotype. Jama. 2014;312(1):48–56. doi: 10.1001/jama.2014.7192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hsieh MM, Kang EM, Fitzhugh CD, Link MB, Bolan CD, Kurlander R, Childs RW, Rodgers GP, Powell JD, Tisdale JF. Allogeneic hematopoietic stem-cell transplantation for sickle cell disease. New England Journal of Medicine. 2009;361(24):2309–2317. doi: 10.1056/NEJMoa0904971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Robinson TM, Fuchs EJ. Allogeneic stem cell transplantation for sickle cell disease. Current Opinion in Hematology. 2016;23(6):524–529. doi: 10.1097/MOH.0000000000000282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Soni S, Gross TG, Rangarajan H, Baker KS, Sturm M, Rhodes M. Outcomes of matched sibling donor hematopoietic stem cell transplantation for severe sickle cell disease with myeloablative conditioning and intermediate-dose of rabbit anti-thymocyte globulin. Pediatric blood & cancer. 2014;61(9):1685–1689. doi: 10.1002/pbc.25059. [DOI] [PubMed] [Google Scholar]
- 12.Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G, Gross F, Yvon E, Nusbaum P, Selz F, Hue C, Certain S, Casanova JL, Bousso P, Deist FL, Fischer A. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease [see comments] Science. 2000;288(5466):669–72. doi: 10.1126/science.288.5466.669. [DOI] [PubMed] [Google Scholar]
- 13.Aiuti A, Vai S, Mortellaro A, Casorati G, Ficara F, Andolfi G, Ferrari G, Tabucchi A, Carlucci F, Ochs HD, Notarangelo LD, Roncarolo MG, Bordignon C. Immune reconstitution in ADA-SCID after PBL gene therapy and discontinuation of enzyme replacement. Nat Med. 2002;8(5):423–5. doi: 10.1038/nm0502-423. [DOI] [PubMed] [Google Scholar]
- 14.Aiuti A, Slavin S, Aker M, Ficara F, Deola S, Mortellaro A, Morecki S, Andolfi G, Tabucchi A, Carlucci F, Marinello E, Cattaneo F, Vai S, Servida P, Miniero R, Roncarolo MG, Bordignon C. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science. 2002;296(5577):2410–3. doi: 10.1126/science.1070104. [DOI] [PubMed] [Google Scholar]
- 15.Ott MG, Schmidt M, Schwarzwaelder K, Stein S, Siler U, Koehl U, Glimm H, Kuhlcke K, Schilz A, Kunkel H, Naundorf S, Brinkmann A, Deichmann A, Fischer M, Ball C, Pilz I, Dunbar C, Du Y, Jenkins NA, Copeland NG, Luthi U, Hassan M, Thrasher AJ, Hoelzer D, von Kalle C, Seger R, Grez M. Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nat Med. 2006;12(4):401–9. doi: 10.1038/nm1393. [DOI] [PubMed] [Google Scholar]
- 16.Aiuti A, Cassani B, Andolfi G, Mirolo M, Biasco L, Recchia A, Urbinati F, Valacca C, Scaramuzza S, Aker M, Slavin S, Cazzola M, Sartori D, Ambrosi A, Di Serio C, Roncarolo MG, Mavilio F, Bordignon C. Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy. J Clin Invest. 2007;117(8):2233–40. doi: 10.1172/JCI31666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Aiuti A, Cattaneo F, Galimberti S, Benninghoff U, Cassani B, Callegaro L, Scaramuzza S, Andolfi G, Mirolo M, Brigida I, Tabucchi A, Carlucci F, Eibl M, Aker M, Slavin S, Al-Mousa H, Al Ghonaium A, Ferster A, Duppenthaler A, Notarangelo L, Wintergerst U, Buckley RH, Bregni M, Marktel S, Valsecchi MG, Rossi P, Ciceri F, Miniero R, Bordignon C, Roncarolo MG. Gene therapy for immunodeficiency due to adenosine deaminase deficiency. N Engl J Med. 2009;360(5):447–58. doi: 10.1056/NEJMoa0805817. [DOI] [PubMed] [Google Scholar]
- 18.Gaspar HB, Cooray S, Gilmour KC, Parsley KL, Zhang F, Adams S, Bjorkegren E, Bayford J, Brown L, Davies EG, Veys P, Fairbanks L, Bordon V, Petropoulou T, Kinnon C, Thrasher AJ. Hematopoietic stem cell gene therapy for adenosine deaminase-deficient severe combined immunodeficiency leads to long-term immunological recovery and metabolic correction. Sci Transl Med. 2011;3(97):97ra80. doi: 10.1126/scitranslmed.3002716. [DOI] [PubMed] [Google Scholar]
- 19.Hacein-Bey-Abina S, Hauer J, Lim A, Picard C, Wang GP, Berry CC, Martinache C, Rieux-Laucat F, Latour S, Belohradsky BH, Leiva L, Sorensen R, Debre M, Casanova JL, Blanche S, Durandy A, Bushman FD, Fischer A, Cavazzana-Calvo M. Efficacy of gene therapy for X-linked severe combined immunodeficiency. The New England journal of medicine. 2010;363(4):355–64. doi: 10.1056/NEJMoa1000164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hacein-Bey-Abina S, Pai SY, Gaspar HB, Armant M, Berry CC, Blanche S, Bleesing J, Blondeau J, de Boer H, Buckland KF, Caccavelli L, Cros G, De Oliveira S, Fernandez KS, Guo D, Harris CE, Hopkins G, Lehmann LE, Lim A, London WB, van der Loo JC, Malani N, Male F, Malik P, Marinovic MA, McNicol AM, Moshous D, Neven B, Oleastro M, Picard C, Ritz J, Rivat C, Schambach A, Shaw KL, Sherman EA, Silberstein LE, Six E, Touzot F, Tsytsykova A, Xu-Bayford J, Baum C, Bushman FD, Fischer A, Kohn DB, Filipovich AH, Notarangelo LD, Cavazzana M, Williams DA, Thrasher AJ. A modified gamma-retrovirus vector for X-linked severe combined immunodeficiency. The New England journal of medicine. 2014;371(15):1407–17. doi: 10.1056/NEJMoa1404588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Candotti F, Shaw KL, Muul L, Carbonaro D, Sokolic R, Choi C, Schurman SH, Garabedian E, Kesserwan C, Jagadeesh GJ, Fu PY, Gschweng E, Cooper A, Tisdale JF, Weinberg KI, Crooks GM, Kapoor N, Shah A, Abdel-Azim H, Yu XJ, Smogorzewska M, Wayne AS, Rosenblatt HM, Davis CM, Hanson C, Rishi RG, Wang X, Gjertson D, Yang OO, Balamurugan A, Bauer G, Ireland JA, Engel BC, Podsakoff GM, Hershfield MS, Blaese RM, Parkman R, Kohn DB. Gene therapy for adenosine deaminase-deficient severe combined immune deficiency: clinical comparison of retroviral vectors and treatment plans. Blood. 2012;120(18):3635–46. doi: 10.1182/blood-2012-02-400937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Blaese RM, Culver KW, Anderson WF, Nienhuis A, Dunbar C, Chang L, Mullen C, Carter C, Leitman S, Berger M. Treatment of Severe Combined Immunodeficiency Disease (SCID) due to Adenosine Deaminase Deficiency with CD34+ Selected Autologous Peripheral Blood Cells Transduced with a Human ADA Gene (Amendment) National Institutes of Health, Human gene therapy. 1993;4(4):521–527. doi: 10.1089/hum.1993.4.4-521. [DOI] [PubMed] [Google Scholar]
- 23.Bank A, Dorazio R, Leboulch P. A Phase I/II Clinical Trial of β-Globin Gene Therapy for β-Thalassemia. Annals of the New York Academy of Sciences. 2005;1054(1):308–316. doi: 10.1196/annals.1345.007. [DOI] [PubMed] [Google Scholar]
- 24.Chandrakasan S, Malik P. Gene therapy for hemoglobinopathies: the state of the field and the future. Hematology/oncology clinics of North America. 2014;28(2):199–216. doi: 10.1016/j.hoc.2013.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Aiuti A, Biasco L, Scaramuzza S, Ferrua F, Cicalese MP, Baricordi C, Dionisio F, Calabria A, Giannelli S, Castiello MC. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science. 2013;341(6148):1233151. doi: 10.1126/science.1233151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kanter J, Walters MC, Hsieh MM, Krishnamurti L, Kwiatkowski J, Kamble RT, von Kalle C, Kuypers FA, Cavazzana M, Leboulch P. Interim results from a phase 1/2 clinical study of lentiglobin gene therapy for severe sickle cell disease. Am Soc Hematology. 2016 [Google Scholar]
- 27.Thompson AA, Kwiatkowski J, Rasko J, Hongeng S, Schiller GJ, Anurathapan U, Cavazzana M, Ho PJ, von Kalle C, Kletzel M. Lentiglobin Gene Therapy for Transfusion-Dependent β-Thalassemia: Update from the Northstar Hgb-204 Phase 1/2 Clinical Study. Am Soc Hematology. 2016 [Google Scholar]
- 28.Pauwels K, Gijsbers R, Toelen J, Schambach A, Willard-Gallo K, Verheust C, Debyser Z, Herman P. State-of-the-art lentiviral vectors for research use: risk assessment and biosafety recommendations. Current gene therapy. 2009;9(6):459–474. doi: 10.2174/156652309790031120. [DOI] [PubMed] [Google Scholar]
- 29.Browning DL, Collins CP, Hocum JD, Leap DJ, Rae DT, Trobridge GD. Insulated foamy viral vectors. Human gene therapy. 2015;27(3):255–266. doi: 10.1089/hum.2015.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Browning DL, Everson EM, Leap DJ, Hocum JD, Wang H, Stamatoyannopoulos G, Trobridge GD. Evidence for the in vivo safety of insulated foamy viral vectors. Gene therapy. 2017;24(3):187–198. doi: 10.1038/gt.2016.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Bender M, Gelinas R, Miller AD. A majority of mice show long-term expression of a human beta-globin gene after retrovirus transfer into hematopoietic stem cells. Molecular and cellular biology. 1989;9(4):1426–1434. doi: 10.1128/mcb.9.4.1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Karlsson S, Bodine DM, Perry L, Papayannopoulou T, Nienhuis AW. Expression of the human beta-globin gene following retroviral-mediated transfer into multipotential hematopoietic progenitors of mice. Proceedings of the National Academy of Sciences. 1988;85(16):6062–6066. doi: 10.1073/pnas.85.16.6062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.May C, Rivella S, Callegari J, Heller G, Gaensler KM, Luzzatto L, Sadelain M. Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin. Nature. 2000;406(6791):82–6. doi: 10.1038/35017565. [DOI] [PubMed] [Google Scholar]
- 34.Persons DA, Allay ER, Sawai N, Hargrove PW, Brent TP, Hanawa H, Nienhuis AW, Sorrentino BP. Successful treatment of murine beta-thalassemia using in vivo selection of genetically modified, drug-resistant hematopoietic stem cells. Blood. 2003;102(2):506–13. doi: 10.1182/blood-2003-03-0677. [DOI] [PubMed] [Google Scholar]
- 35.Persons DA, Hargrove PW, Allay ER, Hanawa H, Nienhuis AW. The degree of phenotypic correction of murine beta -thalassemia intermedia following lentiviral-mediated transfer of a human gamma-globin gene is influenced by chromosomal position effects and vector copy number. Blood. 2003;101(6):2175–83. doi: 10.1182/blood-2002-07-2211. [DOI] [PubMed] [Google Scholar]
- 36.Pawliuk R, Westerman KA, Fabry ME, Payen E, Tighe R, Bouhassira EE, Acharya SA, Ellis J, London IM, Eaves CJ, Humphries RK, Beuzard Y, Nagel RL, Leboulch P. Correction of sickle cell disease in transgenic mouse models by gene therapy. Science. 2001;294(5550):2368–71. doi: 10.1126/science.1065806. [DOI] [PubMed] [Google Scholar]
- 37.Imren S, Payen E, Westerman KA, Pawliuk R, Fabry ME, Eaves CJ, Cavilla B, Wadsworth LD, Beuzard Y, Bouhassira EE, Russell R, London IM, Nagel RL, Leboulch P, Humphries RK. Permanent and panerythroid correction of murine beta thalassemia by multiple lentiviral integration in hematopoietic stem cells. Proc Natl Acad Sci U S A. 2002;99(22):14380–5. doi: 10.1073/pnas.212507099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Moreau-Gaudry F, Xia P, Jiang G, Perelman NP, Bauer G, Ellis J, Surinya KH, Mavilio F, Shen CK, Malik P. High-level erythroid-specific gene expression in primary human and murine hematopoietic cells with self-inactivating lentiviral vectors. Blood. 2001;98(9):2664–72. doi: 10.1182/blood.v98.9.2664. [DOI] [PubMed] [Google Scholar]
- 39.Perumbeti A, Higashimoto T, Urbinati F, Franco R, Meiselman HJ, Witte D, Malik P. A novel human gamma-globin gene vector for genetic correction of sickle cell anemia in a humanized sickle mouse model: critical determinants for successful correction. Blood. 2009;114(6):1174–85. doi: 10.1182/blood-2009-01-201863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Arumugam PI, Scholes J, Perelman N, Xia P, Yee JK, Malik P. Improved human beta-globin expression from self-inactivating lentiviral vectors carrying the chicken hypersensitive site-4 (cHS4) insulator element. Mol Ther. 2007;15(10):1863–71. doi: 10.1038/sj.mt.6300259. [DOI] [PubMed] [Google Scholar]
- 41.Kootstra NA, Munk C, Tonnu N, Landau NR, Verma IM. Abrogation of postentry restriction of HIV-1-based lentiviral vector transduction in simian cells. Proc Natl Acad Sci U S A. 2003;100(3):1298–303. doi: 10.1073/pnas.0337541100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hayakawa J, Ueda T, Lisowski L, Hsieh MM, Washington K, Phang O, Metzger M, Krouse A, Donahue RE, Sadelain M, Tisdale JF. Transient in vivo beta-globin production after lentiviral gene transfer to hematopoietic stem cells in the nonhuman primate. Hum Gene Ther. 2009;20(6):563–72. doi: 10.1089/hum.2008.186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Uchida N, Washington KN, Hayakawa J, Hsieh MM, Bonifacino AC, Krouse AE, Metzger ME, Donahue RE, Tisdale JF. Development of a human immunodeficiency virus type 1-based lentiviral vector that allows efficient transduction of both human and rhesus blood cells. Journal of virology. 2009;83(19):9854–9862. doi: 10.1128/JVI.00357-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Fitzhugh CD, Cordes S, Taylor T, Coles W, Roskom K, Link M, Hsieh MM, Tisdale JF. At least 20% donor myeloid chimerism is necessary to reverse the sickle phenotype after allogeneic HSCT. Blood. 2017 doi: 10.1182/blood-2017-03-772392. blood-2017-03-772392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Eaton WA, Hofrichter J. Hemoglobin S gelation and sickle cell disease. Blood. 1987;70(5):1245–66. [PubMed] [Google Scholar]
- 46.Eaton WA, Bunn HF. Treating sickle cell disease by targeting HbS polymerization. Blood. 2017;129(20):2719–2726. doi: 10.1182/blood-2017-02-765891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Adachi K, Konitzer P, Surrey S. Role of gamma 87 Gln in the inhibition of hemoglobin S polymerization by hemoglobin F. Journal of Biological Chemistry. 1994;269(13):9562–9567. [PubMed] [Google Scholar]
- 48.Pawliuk R, Westerman KA, Fabry ME, Payen E, Tighe R, Bouhassira EE, Acharya SA, Ellis J, London IM, Eaves CJ. Correction of sickle cell disease in transgenic mouse models by gene therapy. Science. 2001;294(5550):2368–2371. doi: 10.1126/science.1065806. [DOI] [PubMed] [Google Scholar]
- 49.Negre O, Bartholomae C, Beuzard Y, Cavazzana M, Christiansen L, Courne C, Deichmann A, Denaro M, de Dreuzy E, Finer M. Preclinical evaluation of efficacy and safety of an improved lentiviral vector for the treatment of β-thalassemia and sickle cell disease. Current gene therapy. 2015;15(1):64–81. doi: 10.2174/1566523214666141127095336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kanter J, Walters MC, Hsieh M, Krishnamurti L, Kwiatkowski JL, Kamble R, von Kalle C, Joseney-Antoine M, Pierciey FJ, Shi W. Interim Results from a Phase 1/2 Clinical Study of Lentiglobin Gene Therapy for Severe Sickle Cell Disease; 59th ASH Annual Meeting & Exposition, Am Soc Hematology; Atlanta, GA. 2017. p. 527. [Google Scholar]
- 51.Tisdale JF, Pierciey FJ, Kamble R, Kanter J, Krishnamurti L, Kwiatkowski JL, Thompson AA, Shestopalov I, Bonner M, Joseney-Antoine M. Successful Plerixafor-Mediated Mobilization, Apheresis, and Lentiviral Vector Transduction of Hematopoietic Stem Cells in Patients with Severe Sickle Cell Disease; 59th ASH anuual meeting & exposition, Am Soc Hematology; Atlanta, GA. 2017. p. 990. [Google Scholar]
- 52.Fitzhugh CD, Hsieh MM, Bolan CD, Saenz C, Tisdale JF. Granulocyte colony-stimulating factor (G-CSF) administration in individuals with sickle cell disease: time for a moratorium? Cytotherapy. 2009;11(4):464–471. doi: 10.1080/14653240902849788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cavazzana M, Hacein-Bey-Abina S, Payen E, Magrin E, Magnani A, Semeraro M, Caccavelli L, Touzot F, Lefrere F, Suarez F. Longer Term Follow-up on the First Patients with Severe Hemoglobinopathies Treated with Lentiglobin Gene Therapy; 59th ASH Annual Meeting & Exposition, Am Soc Hematology; Atlanta, GA. 2017. p. 4609. [Google Scholar]
- 54.Ribeil J-A, Hacein-Bey-Abina S, Payen E, Magnani A, Semeraro M, Magrin E, Caccavelli L, Neven B, Bourget P, El Nemer W. Gene therapy in a patient with sickle cell disease. New England Journal of Medicine. 2017;376(9):848–855. doi: 10.1056/NEJMoa1609677. [DOI] [PubMed] [Google Scholar]
- 55.Walters MC, Rasko J, Hongeng S, Kwiatkowski J, Schiller GJ, Kletzel M, Ho PJ, Vichinsky E, von Kalle C, Cavazzana M. Update of results from the Northstar Study (HGB-204): a phase 1/2 study of gene therapy for beta-thalassemia major via transplantation of autologous hematopoietic stem cells transduced ex-vivo with a lentiviral beta AT87Q-globin vector (LentiGlobin BB305 Drug Product) Blood. 2015;126(23):201–201. [Google Scholar]
- 56.McCune SL, Reilly MP, Chomo MJ, Asakura T, Townes TM. Recombinant human hemoglobins designed for gene therapy of sickle cell disease. Proceedings of the National Academy of Sciences. 1994;91(21):9852–9856. doi: 10.1073/pnas.91.21.9852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Levasseur DN, Ryan TM, Reilly MP, McCune SL, Asakura T, Townes TM. A recombinant human hemoglobin with anti-sickling properties greater than fetal hemoglobin. Journal of Biological Chemistry. 2004;279(26):27518–27524. doi: 10.1074/jbc.M402578200. [DOI] [PubMed] [Google Scholar]
- 58.Levasseur DN, Ryan TM, Pawlik KM, Townes TM. Correction of a mouse model of sickle cell disease: lentiviral/antisickling β-globin gene transduction of unmobilized, purified hematopoietic stem cells. Blood. 2003;102(13):4312–4319. doi: 10.1182/blood-2003-04-1251. [DOI] [PubMed] [Google Scholar]
- 59.Lettre G, Bauer DE. Fetal haemoglobin in sickle-cell disease: from genetic epidemiology to new therapeutic strategies. The Lancet. 2016;387(10037):2554–2564. doi: 10.1016/S0140-6736(15)01341-0. [DOI] [PubMed] [Google Scholar]
- 60.Watson J, Stahman AW, Bilello FP. The significance of the paucity of sickle cells in newborn Negro infants. Am J Med Sci. 1948;215(4):419–23. doi: 10.1097/00000441-194804000-00008. [DOI] [PubMed] [Google Scholar]
- 61.Conley CL, Weatherall DJ, Richardson SN, Shepard MK, Charache S. Hereditary persistence of fetal hemoglobin: a study of 79 affected persons in 15 Negro families in Baltimore. Blood. 1963;21(3):261–281. [PubMed] [Google Scholar]
- 62.Stamatoyannopoulos G, Wood W, Papayannopoulou T, Nute P. A new form of hereditary persistence of fetal hemoglobin in blacks and its association with sickle cell trait. Blood. 1975;46(5):683–692. [PubMed] [Google Scholar]
- 63.Uda M, Galanello R, Sanna S, Lettre G, Sankaran VG, Chen W, Usala G, Busonero F, Maschio A, Albai G. Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of β-thalassemia. Proceedings of the National Academy of Sciences. 2008;105(5):1620–1625. doi: 10.1073/pnas.0711566105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Li Q, Emery DW, Fernandez M, Han H, Stamatoyannopoulos G. Development of viral vectors for gene therapy of β-chain hemoglobinopathies: Optimization of a γ-globin gene expression cassette. Blood. 1999;93(7):2208–2216. [PubMed] [Google Scholar]
- 65.Sabatino DE, Wong C, Cline AP, Pyle L, Garrett LJ, Gallagher PG, Bodine DM. A minimal ankyrin promoter linked to a human γ-globin gene demonstrates erythroid specific copy number dependent expression with minimal position or enhancer dependence in transgenic mice. Journal of Biological Chemistry. 2000;275(37):28549–28554. doi: 10.1074/jbc.M004043200. [DOI] [PubMed] [Google Scholar]
- 66.Pestina TI, Hargrove PW, Jay D, Gray JT, Boyd KM, Persons DA. Correction of murine sickle cell disease using γ-globin lentiviral vectors to mediate high-level expression of fetal hemoglobin. Molecular Therapy. 2009;17(2):245–252. doi: 10.1038/mt.2008.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Samakoglu S, Lisowski L, Budak-Alpdogan T, Usachenko Y, Acuto S, Di Marzo R, Maggio A, Zhu P, Tisdale JF, Riviere I. A genetic strategy to treat sickle cell anemia by coregulating globin transgene expression and RNA interference. Nature biotechnology. 2006;24(1):89–94. doi: 10.1038/nbt1176. [DOI] [PubMed] [Google Scholar]
- 68.Hebbel RP, Boogaerts MA, Eaton JW, Steinberg MH. Erythrocyte adherence to endothelium in sickle-cell anemia: a possible determinant of disease severity, New England. Journal of Medicine. 1980;302(18):992–995. doi: 10.1056/NEJM198005013021803. [DOI] [PubMed] [Google Scholar]
- 69.Moreau-Gaudry F, Xia P, Jiang G, Perelman NP, Bauer G, Ellis J, Surinya KH, Mavilio F, Shen CK, Malik P. High-level erythroid-specific gene expression in primary human and murine hematopoietic cells with self-inactivating lentiviral vectors. Blood. 2001;98(9):2664–2672. doi: 10.1182/blood.v98.9.2664. [DOI] [PubMed] [Google Scholar]
- 70.Cavazzana M, Antoniani C, Miccio A. Gene therapy for β-hemoglobinopathies. Molecular Therapy. 2017 doi: 10.1016/j.ymthe.2017.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Deng W, Lee J, Wang H, Miller J, Reik A, Gregory PD, Dean A, Blobel GA. Controlling long-range genomic interactions at a native locus by targeted tethering of a looping factor. Cell. 2012;149(6):1233–1244. doi: 10.1016/j.cell.2012.03.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Deng W, Rupon JW, Krivega I, Breda L, Motta I, Jahn KS, Reik A, Gregory PD, Rivella S, Dean A. Reactivation of developmentally silenced globin genes by forced chromatin looping. Cell. 2014;158(4):849–860. doi: 10.1016/j.cell.2014.05.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Breda L, Motta I, Lourenco S, Gemmo C, Deng W, Rupon JW, Abdulmalik OY, Manwani D, Blobel GA, Rivella S. Forced chromatin looping raises fetal hemoglobin in adult sickle cells to higher levels than pharmacologic inducers. Blood. 2016;128(8):1139–1143. doi: 10.1182/blood-2016-01-691089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Gräslund T, Li X, Magnenat L, Popkov M, Barbas CF. Exploring strategies for the design of artificial transcription factors targeting sites proximal to known regulatory regions for the induction of γ-globin expression and the treatment of sickle cell disease. Journal of Biological Chemistry. 2005;280(5):3707–3714. doi: 10.1074/jbc.M406809200. [DOI] [PubMed] [Google Scholar]
- 75.Wilber A, Tschulena U, Hargrove PW, Kim YS, Persons DA, Barbas CF, Nienhuis AW. A zinc-finger transcriptional activator designed to interact with the γ-globin gene promoters enhances fetal hemoglobin production in primary human adult erythroblasts. Blood. 2010;115(15):3033–3041. doi: 10.1182/blood-2009-08-240556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Menzel S, Garner C, Gut I, Matsuda F, Yamaguchi M, Heath S, Foglio M, Zelenika D, Boland A, Rooks H. A QTL influencing F cell production maps to a gene encoding a zinc-finger protein on chromosome 2p15. Nature genetics. 2007;39(10):1197. doi: 10.1038/ng2108. [DOI] [PubMed] [Google Scholar]
- 77.Thein SL, Menzel S, Peng X, Best S, Jiang J, Close J, Silver N, Gerovasilli A, Ping C, Yamaguchi M. Intergenic variants of HBS1L-MYB are responsible for a major quantitative trait locus on chromosome 6q23 influencing fetal hemoglobin levels in adults. Proceedings of the National Academy of Sciences. 2007;104(27):11346–11351. doi: 10.1073/pnas.0611393104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sankaran VG, Menne TF, Xu J, Akie TE, Lettre G, Van Handel B, Mikkola HK, Hirschhorn JN, Cantor AB, Orkin SH. Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science. 2008;322(5909):1839–1842. doi: 10.1126/science.1165409. [DOI] [PubMed] [Google Scholar]
- 79.Xu J, Peng C, Sankaran VG, Shao Z, Esrick EB, Chong BG, Ippolito GC, Fujiwara Y, Ebert BL, Tucker PW. Correction of sickle cell disease in adult mice by interference with fetal hemoglobin silencing. Science. 2011;334(6058):993–996. doi: 10.1126/science.1211053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Basak A, Hancarova M, Ulirsch JC, Balci TB, Trkova M, Pelisek M, Vlckova M, Muzikova K, Cermak J, Trka J. BCL11A deletions result in fetal hemoglobin persistence and neurodevelopmental alterations. The Journal of clinical investigation. 2015;125(6):2363. doi: 10.1172/JCI81163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Funnell AP, Prontera P, Ottaviani V, Piccione M, Giambona A, Maggio A, Ciaffoni F, Stehling-Sun S, Marra M, Masiello F. 2p15-p16. 1 microdeletions encompassing and proximal to BCL11A are associated with elevated HbF in addition to neurologic impairment. Blood. 2015;126(1):89–93. doi: 10.1182/blood-2015-04-638528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bianchi E, Zini R, Salati S, Tenedini E, Norfo R, Tagliafico E, Manfredini R, Ferrari S. c-myb supports erythropoiesis through the transactivation of KLF1 and LMO2 expression. Blood. 2010;116(22):e99–e110. doi: 10.1182/blood-2009-08-238311. [DOI] [PubMed] [Google Scholar]
- 83.Borg J, Papadopoulos P, Georgitsi M, Gutiérrez L, Grech G, Fanis P, Phylactides M, Verkerk AJ, van der Spek PJ, Scerri CA. Haploinsufficiency for the erythroid transcription factor KLF1 causes hereditary persistence of fetal hemoglobin. Nature genetics. 2010;42(9):801–805. doi: 10.1038/ng.630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
- 85.Chakraborty A, Tapryal N, Venkova T, Horikoshi N, Pandita RK, Sarker AH, Sarkar PS, Pandita TK, Hazra TK. Classical non-homologous end-joining pathway utilizes nascent RNA for error-free double-strand break repair of transcribed genes. Nature communications. 2016;7 doi: 10.1038/ncomms13049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Genovese P, Schiroli G, Escobar G, Di Tomaso T, Firrito C, Calabria A, Moi D, Mazzieri R, Bonini C, Holmes MC. Targeted genome editing in human repopulating hematopoietic stem cells. Nature. 2014;510(7504):235. doi: 10.1038/nature13420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zou J, Mali P, Huang X, Dowey SN, Cheng L. Site-specific gene correction of a point mutation in human iPS cells derived from an adult patient with sickle cell disease. Blood. 2011;118(17):4599–4608. doi: 10.1182/blood-2011-02-335554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hanna J, Wernig M, Markoulaki S, Sun CW, Meissner A, Cassady JP, Beard C, Brambrink T, Wu LC, Townes TM. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science. 2007;318(5858):1920–1923. doi: 10.1126/science.1152092. [DOI] [PubMed] [Google Scholar]
- 89.Sebastiano V, Maeder ML, Angstman JF, Haddad B, Khayter C, Yeo DT, Goodwin MJ, Hawkins JS, Ramirez CL, Batista LF. In situ genetic correction of the sickle cell anemia mutation in human induced pluripotent stem cells using engineered zinc finger nucleases. Stem cells. 2011;29(11):1717–1726. doi: 10.1002/stem.718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Hoban MD, Cost GJ, Mendel MC, Romero Z, Kaufman ML, Joglekar AV, Ho M, Lumaquin D, Gray D, Lill GR. Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells. Blood. 2015;125(17):2597–2604. doi: 10.1182/blood-2014-12-615948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sun N, Liang J, Abil Z, Zhao H. Optimized TAL effector nucleases (TALENs) for use in treatment of sickle cell disease. Molecular BioSystems. 2012;8(4):1255–1263. doi: 10.1039/c2mb05461b. [DOI] [PubMed] [Google Scholar]
- 92.Sun N, Zhao H. Seamless correction of the sickle cell disease mutation of the HBB gene in human induced pluripotent stem cells using TALENs. Biotechnology and bioengineering. 2014;111(5):1048–1053. doi: 10.1002/bit.25018. [DOI] [PubMed] [Google Scholar]
- 93.Ramalingam S, Annaluru N, Kandavelou K, Chandrasegaran S. TALEN-mediated generation and genetic correction of disease-specific human induced pluripotent stem cells. Current gene therapy. 2014;14(6):461–472. doi: 10.2174/1566523214666140918101725. [DOI] [PubMed] [Google Scholar]
- 94.Hoban MD, Lumaquin D, Kuo CY, Romero Z, Long J, Ho M, Young CS, Mojadidi M, Fitz-Gibbon S, Cooper AR. CRISPR/Cas9-mediated correction of the sickle mutation in human CD34+ cells. Molecular Therapy. 2016;24(9):1561–1569. doi: 10.1038/mt.2016.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.DeWitt MA, Magis W, Bray NL, Wang T, Berman JR, Urbinati F, Heo SJ, Mitros T, Muñoz DP, Boffelli D. Selection-free genome editing of the sickle mutation in human adult hematopoietic stem/progenitor cells. Science translational medicine. 2016;8(360):360ra134–360ra134. doi: 10.1126/scitranslmed.aaf9336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Dever DP, Bak RO, Reinisch A, Camarena J, Washington G, Nicolas CE, Pavel-Dinu M, Saxena N, Wilkens AB, Mantri S. CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells. Nature. 2016;539(7629):384–389. doi: 10.1038/nature20134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.John A, Brylka H, Wiegreffe C, Simon R, Liu P, Jüttner R, Crenshaw EB, Luyten FP, Jenkins NA, Copeland NG. Bcl11a is required for neuronal morphogenesis and sensory circuit formation in dorsal spinal cord development. Development. 2012;139(10):1831–1841. doi: 10.1242/dev.072850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Canver MC, Smith EC, Sher F, Pinello L, Sanjana NE, Shalem O, Chen DD, Schupp PG, Vinjamur DS, Garcia SP. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature. 2015;527(7577):192. doi: 10.1038/nature15521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Bauer DE, Kamran SC, Lessard S, Xu J, Fujiwara Y, Lin C, Shao Z, Canver MC, Smith EC, Pinello L. An erythroid enhancer of BCL11A subject to genetic variation determines fetal hemoglobin level. Science. 2013;342(6155):253–257. doi: 10.1126/science.1242088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Tan S, Chang KH, Smith S, Chen K, Sullivan T, Zhou Q, Reik A, Urnov FD, Rebar EJ, Danos O. Genome editing of the Bcl11A erythroid specific enhancer in bone marrow derived hematopoietic stem and progenitor cells for the treatment of sickle cell disease. Am Soc Hematology. 2015 [Google Scholar]
- 101.Chang KH, Smith SE, Sullivan T, Chen K, Zhou Q, West JA, Liu M, Liu Y, Vieira BF, Sun C. Long-term engraftment and fetal globin induction upon BCL11A gene editing in bone-marrow-derived CD34+ hematopoietic stem and progenitor cells. Molecular Therapy-Methods & Clinical Development. 2017;4:137–148. doi: 10.1016/j.omtm.2016.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Traxler EA, Yao Y, Wang YD, Woodard KJ, Kurita R, Nakamura Y, Hughes JR, Hardison RC, Blobel GA, Li C. A genome-editing strategy to treat [beta]-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition. Nature medicine. 2016;22(9):987–990. doi: 10.1038/nm.4170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ye L, Wang J, Tan Y, Beyer AI, Xie F, Muench MO, Kan YW. Genome editing using CRISPR-Cas9 to create the HPFH genotype in HSPCs: An approach for treating sickle cell disease and β-thalassemia. Proceedings of the National Academy of Sciences. 2016:201612075. doi: 10.1073/pnas.1612075113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.LaFountaine JS, Fathe K, Smyth HD. Delivery and therapeutic applications of gene editing technologies ZFNs, TALENs, and CRISPR/Cas9. International journal of pharmaceutics. 2015;494(1):180–194. doi: 10.1016/j.ijpharm.2015.08.029. [DOI] [PubMed] [Google Scholar]
- 105.Shim G, Kim D, Park GT, Jin H, Suh SK, Oh YK. Therapeutic gene editing: delivery and regulatory perspectives. Acta Pharmacologica Sinica. 2017;38(6):738. doi: 10.1038/aps.2017.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Cox DBT, Platt RJ, Zhang F. Therapeutic genome editing: prospects and challenges. Nature medicine. 2015;21(2):121–131. doi: 10.1038/nm.3793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Varagnolo L, Lin Q, Obier N, Plass C, Dietl J, Zenke M, Claus R, Müller AM. PRC2 inhibition counteracts the culture-associated loss of engraftment potential of human cord blood-derived hematopoietic stem and progenitor cells, Scientific reports. 2015;5 doi: 10.1038/srep12319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Czechowicz A, Kraft D, Weissman IL, Bhattacharya D. Efficient transplantation via antibody-based clearance of hematopoietic stem cell niches. Science. 2007;318(5854):1296–1299. doi: 10.1126/science.1149726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Xue X, Pech NK, Shelley WC, Srour EF, Yoder MC, Dinauer MC. Antibody targeting KIT as pretransplantation conditioning in immunocompetent mice. Blood. 2010;116(24):5419–5422. doi: 10.1182/blood-2010-07-295949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Palchaudhuri R, Saez B, Hoggatt J, Schajnovitz A, Sykes DB, Tate TA, Czechowicz A, Kfoury Y, Ruchika F, Rossi DJ. Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin. Nature biotechnology. 2016;34(7):738. doi: 10.1038/nbt.3584. [DOI] [PMC free article] [PubMed] [Google Scholar]