Login / Signup

Development of β-globin gene correction in human hematopoietic stem cells as a potential durable treatment for sickle cell disease.

Annalisa LattanziJoab CamarenaPremanjali LahiriHelen SegalWaracharee SrifaChristopher A VakulskasRichard L FrockJosefin KenrickCiaran M LeeNarae TalbottJason SkowronskiM Kyle CromerCarsten T CharlesworthRasmus O BakSruthi MantriGang BaoDavid DiGiustoJohn F TisdaleJ Fraser WrightNeehar BhatiaMaria Grazia RoncaroloDaniel P DeverMatthew H Porteus
Published in: Science translational medicine (2021)
Sickle cell disease (SCD) is the most common serious monogenic disease with 300,000 births annually worldwide. SCD is an autosomal recessive disease resulting from a single point mutation in codon six of the β-globin gene (HBB). Ex vivo β-globin gene correction in autologous patient-derived hematopoietic stem and progenitor cells (HSPCs) may potentially provide a curative treatment for SCD. We previously developed a CRISPR-Cas9 gene targeting strategy that uses high-fidelity Cas9 precomplexed with chemically modified guide RNAs to induce recombinant adeno-associated virus serotype 6 (rAAV6)-mediated HBB gene correction of the SCD-causing mutation in HSPCs. Here, we demonstrate the preclinical feasibility, efficacy, and toxicology of HBB gene correction in plerixafor-mobilized CD34+ cells from healthy and SCD patient donors (gcHBB-SCD). We achieved up to 60% HBB allelic correction in clinical-scale gcHBB-SCD manufacturing. After transplant into immunodeficient NSG mice, 20% gene correction was achieved with multilineage engraftment. The long-term safety, tumorigenicity, and toxicology study demonstrated no evidence of abnormal hematopoiesis, genotoxicity, or tumorigenicity from the engrafted gcHBB-SCD drug product. Together, these preclinical data support the safety, efficacy, and reproducibility of this gene correction strategy for initiation of a phase 1/2 clinical trial in patients with SCD.
Keyphrases