TO THE EDITOR:

The hemoglobin tetramer is composed of 2 α-like globin chains and 2 β-like globin chains. Sickle cell disease (SCD) and β-thalassemia are β-hemoglobinopathies characterized by abnormal and inadequate production of β-globin, respectively. Patients with SCD and transfusion-dependent β-thalassemia (TDT) are usually born asymptomatic because of high levels of γ-globin, which forms fetal hemoglobin (HbF; α2γ2). Symptoms develop postnatally as γ-globin is replaced by β-globin (Figure 1A).1 However, coinheritance of genetic variants of hereditary persistence of HbF attenuates the severity of both diseases. Therefore, reactivating γ-globin and increasing HbF levels is a promising treatment strategy for SCD and TDT.1,2 

Figure 1.

Editing the HBG1/2 promoters or the BCL11A erythroid enhancer in CD34+ cells with Cas9 RNP robustly increases HbF expression but leads to different levels of transcriptomic perturbation in erythroid progeny in vitro. (A) Developmentally regulated hemoglobin switch during human ontogenesis. (B) BCL11A is a transcription factor that mediates the HbF to adult hemoglobin (HbA) switch shortly after birth by repressing the expression of γ-globin, a component of HbF, via the distal BCL11A binding sites at the γ-globin gene (HBG1/2) promoters. (C) Single-guide RNAs (sgRNAs) were designed to target the +58 GATA1 binding site at the BCL11A erythroid enhancer on chromosome 2 or the −110 distal BCL11A binding sites at the HBG1/2 promoters on chromosome 11. (D) Peripheral blood–mobilized CD34+ cells from healthy donors were prestimulated for 2 days and then electroporated with 1-8 μM of CRISPR-associated protein 9 (Cas9) complexed with guide RNA (gRNA) targeting either the HBG1/2 promoters (HBG1/2) or the BCL11A erythroid enhancer (BCL11A) at a gRNA-to-protein ratio of 4:1 using a Maxcyte GT electroporator, program HBM34-4. The editing rate, expressed as the frequency of sequencing reads with indels at the HBG1 promoter, HBG2 promoter, or hybrid HBG1-2 promoter (the result of simultaneous cleavage of HBG1 and HBG2 promoters and the deletion of the intervening sequence) was determined by next-generation sequencing, 1 day after electroporation. (E) CD34+ cells were placed in erythroid culture conditions for 18 days and the level of HbF expression was evaluated via reverse phase ultrahigh performance liquid chromatography in which individual globin chains (α, β, Aγ, and Gγ) were separated. HbF level was calculated as (Aγ + Gγ)/(β + Aγ + Gγ) × 100%. Data from CD34+ cells electroporated with 4 μM or 8 μM RNP are shown, with maximum levels of editing (typically >95%) achieved in erythroid cells. (F) Day-11 erythroid cells in triplicates from CD34+ cells transfected with 8 μM RNP were collected and the transcriptome profiled via RNA sequencing. High levels of editing were achieved in erythroid cells (mean ± standard deviation: HBG1/2, 98.68% ± 0.09%; BCL11A, 99.22% ± 0.08%). Transcripts expressed at levels of more than or equal to twofold difference from control samples, and FDR of <0.05 were identified (red, blue [BCL11A], and orange [HBG1/2] dots). (G) Differentially expressed genes (DEGs) involved in the cell processes of apoptosis, differentiation, proliferation, or erythropoiesis in BCL11A-edited samples are listed. For panels D-E, each circle represents an independent sample, and the horizontal bar represents the mean value of the respective treatment group. n = 4 to 7 for panel D and n = 6 to 11 for panel E. Mobilized peripheral blood CD34+ cells from multiple donors were used; n = 9 for HBG1/2 and n = 6 for BCL11A. Ordinary 1-way analysis of variance (ANOVA) with Tukey multiple comparisons test was performed to evaluate the statistical significance of the mean values for panel D. ∗P < .05; ∗∗P < .001; ∗∗∗∗P < .0001. BGLT3, β-globin locus transcript 3; BMPR1A, bone morphogenetic protein receptor type 1A gene; CARD9, caspase recruitment domain family member 9 gene; chr, chromosome; ex, exon; FC, fold change; FDR, false discovery rate; FOXC1, forkhead box C1 gene; FZD4, frizzled class receptor 4 gene; GATA5, GATA-binding protein 5 gene; G0S2, G0/G1 switch 2 gene; HBB, β-globin gene; HBD, δ-globin gene; HBE, ε-globin gene; HES4, hes family BHLH transcription factor 4; HS, hypersensitive site; IGF2, insulin-like growth factor 2 gene; IGFL2, IGF-like family member 2 gene; IRS4, insulin receptor substrate 4 gene; JAG1, jagged canonical notch ligand 1 gene; KLF15, KLF transcription factor 15 gene; LCR, locus control region; PAM, protospacer adjacent motif; PDE3A, phosphodiesterase 3A gene; PDGFA, platelet-derived growth factor subunit A gene; SMAD3, SMAD family member 3 gene; TFCP2L1, transcription factor CP2-like 1 gene; TNFRSF25; TNF receptor superfamily member 25 gene; TP53I11; tumor protein p53 inducible protein 11 gene.

Figure 1.

Editing the HBG1/2 promoters or the BCL11A erythroid enhancer in CD34+ cells with Cas9 RNP robustly increases HbF expression but leads to different levels of transcriptomic perturbation in erythroid progeny in vitro. (A) Developmentally regulated hemoglobin switch during human ontogenesis. (B) BCL11A is a transcription factor that mediates the HbF to adult hemoglobin (HbA) switch shortly after birth by repressing the expression of γ-globin, a component of HbF, via the distal BCL11A binding sites at the γ-globin gene (HBG1/2) promoters. (C) Single-guide RNAs (sgRNAs) were designed to target the +58 GATA1 binding site at the BCL11A erythroid enhancer on chromosome 2 or the −110 distal BCL11A binding sites at the HBG1/2 promoters on chromosome 11. (D) Peripheral blood–mobilized CD34+ cells from healthy donors were prestimulated for 2 days and then electroporated with 1-8 μM of CRISPR-associated protein 9 (Cas9) complexed with guide RNA (gRNA) targeting either the HBG1/2 promoters (HBG1/2) or the BCL11A erythroid enhancer (BCL11A) at a gRNA-to-protein ratio of 4:1 using a Maxcyte GT electroporator, program HBM34-4. The editing rate, expressed as the frequency of sequencing reads with indels at the HBG1 promoter, HBG2 promoter, or hybrid HBG1-2 promoter (the result of simultaneous cleavage of HBG1 and HBG2 promoters and the deletion of the intervening sequence) was determined by next-generation sequencing, 1 day after electroporation. (E) CD34+ cells were placed in erythroid culture conditions for 18 days and the level of HbF expression was evaluated via reverse phase ultrahigh performance liquid chromatography in which individual globin chains (α, β, Aγ, and Gγ) were separated. HbF level was calculated as (Aγ + Gγ)/(β + Aγ + Gγ) × 100%. Data from CD34+ cells electroporated with 4 μM or 8 μM RNP are shown, with maximum levels of editing (typically >95%) achieved in erythroid cells. (F) Day-11 erythroid cells in triplicates from CD34+ cells transfected with 8 μM RNP were collected and the transcriptome profiled via RNA sequencing. High levels of editing were achieved in erythroid cells (mean ± standard deviation: HBG1/2, 98.68% ± 0.09%; BCL11A, 99.22% ± 0.08%). Transcripts expressed at levels of more than or equal to twofold difference from control samples, and FDR of <0.05 were identified (red, blue [BCL11A], and orange [HBG1/2] dots). (G) Differentially expressed genes (DEGs) involved in the cell processes of apoptosis, differentiation, proliferation, or erythropoiesis in BCL11A-edited samples are listed. For panels D-E, each circle represents an independent sample, and the horizontal bar represents the mean value of the respective treatment group. n = 4 to 7 for panel D and n = 6 to 11 for panel E. Mobilized peripheral blood CD34+ cells from multiple donors were used; n = 9 for HBG1/2 and n = 6 for BCL11A. Ordinary 1-way analysis of variance (ANOVA) with Tukey multiple comparisons test was performed to evaluate the statistical significance of the mean values for panel D. ∗P < .05; ∗∗P < .001; ∗∗∗∗P < .0001. BGLT3, β-globin locus transcript 3; BMPR1A, bone morphogenetic protein receptor type 1A gene; CARD9, caspase recruitment domain family member 9 gene; chr, chromosome; ex, exon; FC, fold change; FDR, false discovery rate; FOXC1, forkhead box C1 gene; FZD4, frizzled class receptor 4 gene; GATA5, GATA-binding protein 5 gene; G0S2, G0/G1 switch 2 gene; HBB, β-globin gene; HBD, δ-globin gene; HBE, ε-globin gene; HES4, hes family BHLH transcription factor 4; HS, hypersensitive site; IGF2, insulin-like growth factor 2 gene; IGFL2, IGF-like family member 2 gene; IRS4, insulin receptor substrate 4 gene; JAG1, jagged canonical notch ligand 1 gene; KLF15, KLF transcription factor 15 gene; LCR, locus control region; PAM, protospacer adjacent motif; PDE3A, phosphodiesterase 3A gene; PDGFA, platelet-derived growth factor subunit A gene; SMAD3, SMAD family member 3 gene; TFCP2L1, transcription factor CP2-like 1 gene; TNFRSF25; TNF receptor superfamily member 25 gene; TP53I11; tumor protein p53 inducible protein 11 gene.

Close modal

Silencing γ-globin is partially mediated by the transcription factor B-cell lymphoma/leukemia 11A (BCL11A), which binds to the promoters of the γ-globin genes, HBG1 and HBG2 (denoted HBG1/2; Figure 1B).3 Editing patient CD34+ hematopoietic stem and progenitor cells (HSPCs) at the BCL11A erythroid enhancer +58 GATAA motif or the distal BCL11A binding site at the HBG1/2 promoters’ −110 regions can permanently disrupt this pathway and reactivate γ-globin expression.4-8 Both approaches have demonstrated clinically meaningful HbF induction and improved clinical outcomes in patients with SCD and TDT.5-8 In clinical studies in which patients received BCL11A erythroid enhancer–edited HSPCs, 97% of patients with SCD were free of vaso-occlusive crisis for ≥12 consecutive months;6 and 91% of patients with TDT achieved transfusion independence.8 These studies validate the safety and effectiveness of this approach and supported the approval of exagamglogene autotemcel for treating SCD and TDT, with long-term studies ongoing.9 Notwithstanding, there is an opportunity to explore other mechanisms of HbF reactivation for optimal patient outcomes. Clinical evaluation of an investigational HBG1/2 promoter-edited CD34+ cell therapy, renizgamglogene autogedtemcel, is ongoing, with rapid and sustained increases in HbF and early correction of anemia having been observed in treated patients with SCD and TDT.7,10,11 Additionally, renizgamglogene autogedtemcel has demonstrated early engraftment and a favorable safety profile to date. In this nonclinical study, we evaluated whether editing the HBG1/2 promoters or the BCL11A erythroid enhancer led to biological consequences beyond HbF induction based on target site.

Mobilized CD34+ cells from healthy donors were edited with CRISPR-associated protein 9 (Cas9) ribonucleoproteins (RNPs) at either the HBG1/2 promoters’ −110 region in which multiple genetic variants of hereditary persistence of HbF reside3,12 or the BCL11A erythroid enhancer5,13,14 (Figure 1C; supplemental Table 1; see supplemental Data for detailed methods). BCL11A-targeting RNPs were more efficient than HBG1/2-targeting RNPs in creating insertions and/or deletions (indels) at the target site (Figure 1D). Noteworthy, simultaneous cleavage of the HBG1 and HBG2 promoters may lead to a 4.9-kilobase deletion of the intervening sequence, creating an HBG1-2 hybrid promoter with or without indels. This 4.9-kilobase deletion is estimated to occur in ∼30% to 40% of the loci.4,12 When differentiated into erythroid cells in vitro, both targeting approaches led to robust HbF expression at ∼35% (Figure 1E), which is consistent with previous publications5,12,14 and above the 30% HbF threshold for therapeutic relevance.15 

Transcriptome analysis of day-11 in vitro–differentiated erythroblasts from unedited (control) or edited CD34+ cells (mean ± standard deviation editing rate: HBG1/2, 98.68% ± 0.09%; BCL11A, 99.22% ± 0.08%) revealed 5 differentially expressed genes (DEGs) in HBG1/2-edited samples and 110 DEGs in BCL11A-edited samples (Figure 1F). In the HBG1/2-edited samples, 4 DEGs were globin related: δ-globin, β-globin locus transcript 3, HBG1, and HBG2. In contrast, most DEGs in BCL11A-edited samples were not globin related. Consistent with BCL11A’s transcriptional repressor role,16 most of the 110 DEGs in BCL11A-edited samples were upregulated, including BCL11A primary targets17 (Figure 1F; supplemental Figure 1; supplemental Data). Multiple DEGs were related to cell processes such as apoptosis, differentiation, and proliferation; and 2 DEGs have been implicated in dysregulated erythropoiesis (Figure 1G). These data contradict previous studies showing minimal transcriptomic perturbation after BCL11A erythroid enhancer editing.4,17 Differences in methodologies may explain this discrepancy. We evaluated day-11 immature erythroblasts, whereas Lamsfus-Calle et al evaluated day-21 erythroid cells, likely reticulocytes.4 Rajendiran et al used base editing, which led to a low BCL11A erythroid enhancer disruption and modest HbF induction.17 Interestingly, when greater BCL11A downregulation was achieved via base editing of the BCL11A initiation codon, they observed a similar pattern of perturbation to BCL11A primary targets17 as we did (supplemental Figure 1).

These data suggest that editing the BCL11A erythroid enhancer, which leads to BCL11A downregulation in erythroid cells,14 causes broader transcriptome perturbation in in vitro–derived erythroblasts vs editing the HBG1/2 promoters. Although no overt differences in erythroid maturation in culture were observed (supplemental Figure 2), as previously found with BCL11A exon 2 editing,17,18 we cannot dismiss that the breadth of transcriptome perturbation may have biological consequences in vitro or in vivo.

To evaluate the engraftment potential of CD34+ cells edited at the 2 different target sites, immunodeficient NBSGW mice were infused with edited and control CD34+ cells.19,20 At 16 to 18 weeks after infusion, successful and robust engraftment of human cells was observed in all groups (Figure 2A; supplemental Figures 3A and 4A). Comparable levels of human chimerism and HSPC, B-cell, and neutrophil engraftment in the bone marrow (BM) were observed regardless of the editing approach. However, although long-term engrafted HBG1/2-edited CD34+ cells produced similar levels of erythroid cells as unedited CD34+ cells, reduced human erythroid output was observed with BCL11A-edited CD34+ cells (Figure 2A; supplemental Figure 3A). Normalization of human erythroid output to HSPC frequency in the BM of corresponding animals corroborates that BCL11A-edited HSPCs produced fewer erythroid cells than unedited HSPCs in vivo (Figure 2B). In a study by Wu et al using the same animal model, deficient erythroid production was not observed after BCL11A erythroid enhancer editing.14 However, the administration route, number of CD34+ cells infused, and gating strategies were different, and relatively few animals were evaluated. Furthermore, although no erythroid reduction occurred in 1 rhesus monkey transplanted with BCL11A erythroid enhancer–edited rhesus CD34+ cells,21 Bcl11a conditional knockout mice, in which BCL11A is reduced by 70% in erythroid cells, do develop mild anemia.22 

Figure 2.

Editing the HBG1/2 promoters is compatible with multilineage engraftment and robust HbF induction, whereas editing the BCL11A erythroid enhancer affects erythroid production in a xenotransplantation model. Mobilized healthy donor CD34+ cells were electroporated with 1 to 8 μM of ribonucleoprotein complexed at a 2:1 gRNA-to-protein ratio using a Maxcyte GT electroporator, program HBM34-4. Cells were cryopreserved 1 day after electroporation. Cells with comparable levels of editing were later thawed and infused intravenously into NBSGW mice at 1 × 106 cells per mouse. (A) At 18 weeks after infusion, animals were euthanized to determine the levels of human engraftment in the BM by flow cytometry. Chimerism was calculated as hCD45+/hCD45++mCD45+; human HSPC frequency was calculated as hCD34+/hCD45+; human B-cell frequency was calculated as hCD19+/hCD45+; human neutrophil frequency was calculated as hCD15+/hCD45+; and human erythroid frequency was calculated as hCD235a+/total viable cells. Human T-cell engraftment was at the background level and therefore not reported. (B) Erythroid output (hCD235a+ per total viable cells) was adjusted to HSPC frequency (hCD34+ per total viable cells) in corresponding animals to evaluate the ability of HSPCs to generate erythroid cells in vivo. (C) HbF expression was determined by reverse phase ultrahigh performance liquid chromatography in human erythroid cells isolated by flow-activated cell sorting from mouse BM. (D) Editing levels of infused CD34+ cells (before) and unfractionated BM were determined by next-generation sequencing. (E) Human HSPCs (hLinhCD34+hCD45+), B cells (hCD19+hCD45+), neutrophils (hCD15+hCD45+), and erythroid cells (hCD235a+) were flow sorted and analyzed for editing levels by next-generation sequencing. (F) Editing data of flow-sorted human cells from 3 independent in vivo studies were compiled, and correlation analyses were performed for B cells, neutrophils, or erythroid cells against HSPCs. The black angled line represents the identity line (y = x). (G) Editing data of flow-sorted HSPCs from BM, and erythroid output from 3 independent in vivo studies were compiled, and correlation analysis was performed. To adjust for the differential erythroid output observed in each study, erythroid output was normalized to the output observed in their respective control samples. Each circle represents 1 sample from 1 animal. n = 7 to 8 per treatment group for panels A-E; n = 33 to 39 for panel F; and n = 41 to 52 for panel G including controls. For panels E-G, samples with reads of <5000 mapped to the target were excluded. For panel G, samples with human chimerism levels of <50% were excluded. For panels A-C, ordinary 1-way ANOVA with Tukey multiple comparisons test was performed. For panel E, mixed-effects analysis with Tukey multiple comparisons test was performed to compare indel levels of samples sorted from the same animals. For panel G, simple linear regression analysis was performed, and the slope was tested for nonzero significance. ∗P < .05; ∗∗P < .001; ∗∗∗∗P < .0001.

Figure 2.

Editing the HBG1/2 promoters is compatible with multilineage engraftment and robust HbF induction, whereas editing the BCL11A erythroid enhancer affects erythroid production in a xenotransplantation model. Mobilized healthy donor CD34+ cells were electroporated with 1 to 8 μM of ribonucleoprotein complexed at a 2:1 gRNA-to-protein ratio using a Maxcyte GT electroporator, program HBM34-4. Cells were cryopreserved 1 day after electroporation. Cells with comparable levels of editing were later thawed and infused intravenously into NBSGW mice at 1 × 106 cells per mouse. (A) At 18 weeks after infusion, animals were euthanized to determine the levels of human engraftment in the BM by flow cytometry. Chimerism was calculated as hCD45+/hCD45++mCD45+; human HSPC frequency was calculated as hCD34+/hCD45+; human B-cell frequency was calculated as hCD19+/hCD45+; human neutrophil frequency was calculated as hCD15+/hCD45+; and human erythroid frequency was calculated as hCD235a+/total viable cells. Human T-cell engraftment was at the background level and therefore not reported. (B) Erythroid output (hCD235a+ per total viable cells) was adjusted to HSPC frequency (hCD34+ per total viable cells) in corresponding animals to evaluate the ability of HSPCs to generate erythroid cells in vivo. (C) HbF expression was determined by reverse phase ultrahigh performance liquid chromatography in human erythroid cells isolated by flow-activated cell sorting from mouse BM. (D) Editing levels of infused CD34+ cells (before) and unfractionated BM were determined by next-generation sequencing. (E) Human HSPCs (hLinhCD34+hCD45+), B cells (hCD19+hCD45+), neutrophils (hCD15+hCD45+), and erythroid cells (hCD235a+) were flow sorted and analyzed for editing levels by next-generation sequencing. (F) Editing data of flow-sorted human cells from 3 independent in vivo studies were compiled, and correlation analyses were performed for B cells, neutrophils, or erythroid cells against HSPCs. The black angled line represents the identity line (y = x). (G) Editing data of flow-sorted HSPCs from BM, and erythroid output from 3 independent in vivo studies were compiled, and correlation analysis was performed. To adjust for the differential erythroid output observed in each study, erythroid output was normalized to the output observed in their respective control samples. Each circle represents 1 sample from 1 animal. n = 7 to 8 per treatment group for panels A-E; n = 33 to 39 for panel F; and n = 41 to 52 for panel G including controls. For panels E-G, samples with reads of <5000 mapped to the target were excluded. For panel G, samples with human chimerism levels of <50% were excluded. For panels A-C, ordinary 1-way ANOVA with Tukey multiple comparisons test was performed. For panel E, mixed-effects analysis with Tukey multiple comparisons test was performed to compare indel levels of samples sorted from the same animals. For panel G, simple linear regression analysis was performed, and the slope was tested for nonzero significance. ∗P < .05; ∗∗P < .001; ∗∗∗∗P < .0001.

Close modal

Erythroid progeny of CD34+ cells edited at either target site expressed more HbF in vivo than unedited controls (Figure 2C). Interestingly, HBG1/2-edited erythroid cells showed stronger HbF induction than BCL11A-edited erythroid cells in vivo. Lower HbF induction in BCL11A-edited erythroid cells is potentially because of residual BCL11A14 suppressing γ-globin and/or the lack of sufficient erythropoietic stress21 driving optimal HbF expression in vivo.

Indel evaluation showed that CD34+ cells edited at either target site successfully engrafted in the mouse BM. The editing levels in BM-engrafted human cells were equivalent to, or higher than, those in infused CD34+ cells (Figure 2D; supplemental Figure 3B). Mice that received HBG1/2-edited CD34+ cells showed similar editing levels across BM-isolated human HSPCs, B cells, neutrophils, and erythroid cells (Figure 2E). In contrast, in mice receiving BCL11A-edited CD34+ cells, human erythroid cells had lower editing levels than HSPCs and other blood lineages. Similar trends were observed in 2 independent experiments (supplemental Figures 3C and 4B).

Data from the 3 independent experiments were combined for correlation analyses between editing levels in isolated HSPCs, B cells, neutrophils, or erythroid cells (Figure 2E). Because HSPCs produce downstream lineages, editing levels in the differentiated progeny should closely resemble the levels in the HSPCs if unedited and edited CD34+ cells have similar downstream progeny output capability. Indeed, editing levels in B cells and neutrophils were highly similar to those in corresponding HSPCs, regardless of editing site. Editing levels in erythroid cells derived from HBG1/2-edited CD34+ cells also closely resembled the corresponding HSPCs. In contrast, erythroid cells derived from BCL11A-edited CD34+ cells had lower editing levels than corresponding HSPCs, except when editing levels were very high (∼100%) with almost no unedited CD34+ cells present. This suggests that BCL11A-edited CD34+ cells generate fewer erythroid cells than unedited CD34+ cells, causing an underrepresentation of edited erythroid cells in the mouse BM. The erythroid-specific nature of this observation reflects the strict lineage specificity of this enhancer element.13,23 

An additional correlation analysis on the compiled data set evaluated the impact of HSPC editing levels on erythroid production. Although the editing levels in HSPCs derived from HBG1/2-edited CD34+ cells did not affect overall erythroid output, a negative correlation was observed in HSPCs derived from BCL11A-edited CD34+ cells (Figure 2F; supplemental Figure 4C). This corroborates that BCL11A erythroid enhancer editing adversely affects the ability of CD34+ cells to generate erythroid cells in a xenotransplantation setting. Whether the DEGs observed in in vitro–derived erythroblasts could account for such observations in vivo requires additional investigation. Because both editing approaches use Cas9, the different in vivo erythroid output potential of HBG1/2-edited vs BCL11A-edited HSPCs is likely a biological consequence of target site selection; this should be broadly applicable to other editing modalities.24,25 

These data suggest that BCL11A is involved in erythroid development and that editing the BCL11A erythroid enhancer in CD34+ cells negatively affects erythroid production in a xenotransplantation setting. In contrast, editing BCL11A binding sites at the HBG1/2 promoters enabled robust HbF induction without adversely affecting erythropoiesis. The ability of edited CD34+ cells to generate erythroid cells efficiently may have implications for sustained correction of anemia in a clinical setting, but this remains to be substantiated. Collectively, these preclinical data suggest that editing the HBG1/2 promoters is a promising alternative to editing BCL11A for the durable treatment of β-hemoglobinopathies.

Acknowledgments: The authors thank Georgia Giannoukos, James Bochicchio, Jack Heath, Patricia Sousa, Gregory Gotta, and Edouard de Dreuzy for experimental and sequencing support. The authors acknowledge useful discussions with Stephen Sherman, Haiyan Jiang, Kate Zhang, Charles Albright, Mark Shearman, Tanya M. Teslovich, and Linda Burkly. Medical writing support was provided by Bertha Jane Vandegrift of Porterhouse Medical US and was funded by Editas Medicine Inc, Cambridge, MA, in accordance with good publication practice guidelines.

This study was funded by Editas Medicine, Inc.

Contribution: K.-H.C. conceived the study; T.J., M.J., S.R.B., L.L., P.Z., Y.S., and K.-H.C. performed experiments; T.J., M.J., S.R.B., L.L., P.Z., E.M., M.W., S.G., and K.-H.C. analyzed results; K.-H.C. and E.M. interpreted the results; all authors wrote and revised the manuscript.

Conflict-of-interest disclosure: T.J., M.J., S.R.B., L.L., P.Z., Y.S., K.-H.C, E.M., M.W., and S.G. are current employees and shareholders of Editas Medicine, Inc.

Correspondence: Kai-Hsin Chang, Editas Medicine Inc, 11 Hurley St, Cambridge, MA 02141; email: kaihsin.chang@editasmed.com.

1.
Khandros
E
,
Blobel
GA
.
Elevating fetal hemoglobin: recently discovered regulators and mechanisms
.
Blood
.
2024
;
144
(
8
):
845
-
852
.
2.
Demirci
S
,
Leonard
A
,
Tisdale
JF
.
Genome editing strategies for fetal hemoglobin induction in beta-hemoglobinopathies
.
Hum Mol Genet
.
2020
;
29
(
R1
):
R100
-
R106
.
3.
Liu
N
,
Hargreaves
VV
,
Zhu
Q
, et al
.
Direct promoter repression by BCL11A controls the fetal to adult hemoglobin switch
.
Cell
.
2018
;
173
(
2
):
430
-
442.e17e417
.
4.
Lamsfus-Calle
A
,
Daniel-Moreno
A
,
Antony
JS
, et al
.
Comparative targeting analysis of KLF1, BCL11A, and HBG1/2 in CD34(+) HSPCs by CRISPR/Cas9 for the induction of fetal hemoglobin
.
Sci Rep
.
2020
;
10
(
1
):
10133
.
5.
Frangoul
H
,
Altshuler
D
,
Cappellini
MD
, et al
.
CRISPR-Cas9 gene editing for sickle cell disease and beta-thalassemia
.
N Engl J Med
.
2021
;
384
(
3
):
252
-
260
.
6.
Frangoul
H
,
Locatelli
F
,
Sharma
A
, et al
.
Exagamglogene autotemcel for severe sickle cell disease
.
N Engl J Med
.
2024
;
390
(
18
):
1649
-
1662
.
7.
Hanna
R
,
Frangoul
H
,
McKinney
C
, et al
.
AsCas12a gene editing of HBG1/2 promoters with EDIT-301 results in rapid and sustained normalization of hemoglobin and increased fetal hemoglobin in patients with severe sickle cell disease and transfusion-dependent beta-thalassemia
.
Blood
.
2023
;
142
(
suppl 1
):
4996
.
8.
Locatelli
F
,
Lang
P
,
Wall
D
, et al
.
Exagamglogene autotemcel for transfusion-dependent beta-thalassemia
.
N Engl J Med
.
2024
;
390
(
18
):
1663
-
1676
.
9.
Hoy
SM
.
Exagamglogene autotemcel: first approval
.
Mol Diagn Ther
.
2024
;
28
(
2
):
133
-
139
.
10.
Hanna
R
,
Frangoul
H
,
McKinney
C
, et al
.
Reni-cel, the first AsCas12a gene-edited cell therapy, led to hemoglobin normalization and increased fetal hemoglobin in severe sickle cell disease patients in an interim analysis of the RUBY trial
.
HemaSphere
.
2024
;
8
(
suppl 1
):
404
-
405
.
11.
Frangoul
H
,
Hanna
R
,
Walters
MC
, et al
.
Reni-cel, the first AsCas12a gene-edited cell therapy, shows promising preliminary results in key clinical outcomes in transfusion-dependent beta-thalassemia patients treated in the EdiThal trial
.
HemaSphere
.
2024
;
8
(
suppl 1
):
2727
-
2728
.
12.
Metais
JY
,
Doerfler
PA
,
Mayuranathan
T
, et al
.
Genome editing of HBG1 and HBG2 to induce fetal hemoglobin
.
Blood Adv
.
2019
;
3
(
21
):
3379
-
3392
.
13.
Bauer
DE
,
Kamran
SC
,
Lessard
S
, et al
.
An erythroid enhancer of BCL11A subject to genetic variation determines fetal hemoglobin level
.
Science
.
2013
;
342
(
6155
):
253
-
257
.
14.
Wu
Y
,
Zeng
J
,
Roscoe
BP
, et al
.
Highly efficient therapeutic gene editing of human hematopoietic stem cells
.
Nat Med
.
2019
;
25
(
5
):
776
-
783
.
15.
Steinberg
MH
,
Chui
DH
,
Dover
GJ
,
Sebastiani
P
,
Alsultan
A
.
Fetal hemoglobin in sickle cell anemia: a glass half full?
.
Blood
.
2014
;
123
(
4
):
481
-
485
.
16.
Yin
J
,
Xie
X
,
Ye
Y
,
Wang
L
,
Che
F
.
BCL11A: a potential diagnostic biomarker and therapeutic target in human diseases
.
Biosci Rep
.
2019
;
39
(
11
). BSR20190604.
17.
Rajendiran
V
,
Devaraju
N
,
Haddad
M
, et al
.
Base editing of key residues in the BCL11A-XL-specific zinc finger domains derepresses fetal globin expression
.
Mol Ther
.
2024
;
32
(
3
):
663
-
677
.
18.
Chang
KH
,
Smith
SE
,
Sullivan
T
, et al
.
Long-term engraftment and fetal globin induction upon BCL11A gene editing in bone-marrow-derived CD34(+) hematopoietic stem and progenitor cells
.
Mol Ther Methods Clin Dev
.
2017
;
4
:
137
-
148
.
19.
Rahmig
S
,
Kronstein-Wiedemann
R
,
Fohgrub
J
, et al
.
Improved human erythropoiesis and platelet formation in humanized NSGW41 mice
.
Stem Cell Rep
.
2016
;
7
(
4
):
591
-
601
.
20.
Fiorini
C
,
Abdulhay
NJ
,
McFarland
SK
, et al
.
Developmentally-faithful and effective human erythropoiesis in immunodeficient and Kit mutant mice
.
Am J Hematol
.
2017
;
92
(
9
):
E513
-
E519
.
21.
Demirci
S
,
Zeng
J
,
Wu
Y
, et al
.
BCL11A enhancer-edited hematopoietic stem cells persist in rhesus monkeys without toxicity
.
J Clin Invest
.
2020
;
130
(
12
):
6677
-
6687
.
22.
Esteghamat
F
,
Gillemans
N
,
Bilic
I
, et al
.
Erythropoiesis and globin switching in compound Klf1::Bcl11a mutant mice
.
Blood
.
2013
;
121
(
13
):
2553
-
2562
.
23.
Smith
EC
,
Luc
S
,
Croney
DM
, et al
.
Strict in vivo specificity of the Bcl11a erythroid enhancer
.
Blood
.
2016
;
128
(
19
):
2338
-
2342
.
24.
Han
W
,
Qiu
HY
,
Sun
S
, et al
.
Base editing of the HBG promoter induces potent fetal hemoglobin expression with no detectable off-target mutations in human HSCs
.
Cell Stem Cell
.
2023
;
30
(
12
):
1624
-
1639.e8
.
25.
Liao
J
,
Chen
S
,
Hsiao
S
, et al
.
Therapeutic adenine base editing of human hematopoietic stem cells
.
Nat Commun
.
2023
;
14
(
1
):
207
.

Author notes

The transcriptome data reported in this article have been deposited in the Gene Expression Omnibus database (accession number GSE264491).

A request for access to original data may be submitted to info@editasmed.com.

The full-text version of this article contains a data supplement.