In vivo HSPC gene therapy with base editors allows for efficient reactivation of fetal γ-globin in β-YAC mice

C Li, A Georgakopoulou, A Mishra, S Gil… - Blood …, 2021 - ashpublications.org
C Li, A Georgakopoulou, A Mishra, S Gil, RD Hawkins, E Yannaki, A Lieber
Blood advances, 2021ashpublications.org
Base editors are capable of installing precise genomic alterations without creating double-
strand DNA breaks. In this study, we targeted critical motifs regulating γ-globin reactivation
with base editors delivered via HDAd5/35++ vectors. Through optimized design, we
successfully produced a panel of cytidine and adenine base editor (ABE) vectors targeting
the erythroid BCL11A enhancer or recreating naturally occurring hereditary persistence of
fetal hemoglobin (HPFH) mutations in the HBG1/2 promoter. All 5 tested vectors efficiently …
Abstract
Base editors are capable of installing precise genomic alterations without creating double-strand DNA breaks. In this study, we targeted critical motifs regulating γ-globin reactivation with base editors delivered via HDAd5/35++ vectors. Through optimized design, we successfully produced a panel of cytidine and adenine base editor (ABE) vectors targeting the erythroid BCL11A enhancer or recreating naturally occurring hereditary persistence of fetal hemoglobin (HPFH) mutations in the HBG1/2 promoter. All 5 tested vectors efficiently installed target base conversion and led to γ-globin reactivation in human erythroid progenitor cells. We observed ~23% γ-globin protein production over β-globin, when using an ABE vector (HDAd-ABE-sgHBG-2) specific to the –113A>G HPFH mutation. In a β-YAC mouse model, in vivo hematopoietic progenitor/stem cell (HSPC) transduction with HDAd-ABE-sgHBG-2 followed by in vivo selection resulted in >40% γ-globin+ erythrocytes in the peripheral blood. This result corresponded to 21% γ-globin production over human β-globin. The average –113A>G conversion in total bone marrow cells was 20%. No alterations in hematological parameters, erythropoiesis, and bone marrow cellular composition were observed after treatment. No detectable editing was found at top-scoring, off-target genomic sites. Bone marrow lineage–negative cells from primary mice were capable of reconstituting secondary transplant-recipient mice with stable γ-globin expression. Importantly, the advantage of base editing over CRISPR/Cas9 was reflected by the markedly lower rates of intergenic HBG1/2 deletion and the absence of detectable toxicity in human CD34+ cells. Our observations suggest that HDAd-vectorized base editors represent a promising strategy for precise in vivo genome engineering for the treatment of β-hemoglobinopathies.
ashpublications.org