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

Authors:
Annalisa Lattanzi , Joab Camarena , Premanjali Lahiri , Helen Segal , Waracharee Srifa , Christopher A Vakulskas 4, Richard L Frock 5, Josefin Kenrick 5, Ciaran Lee 6, Narae Talbott , Jason Skowronski , M Kyle Cromer , Carsten T Charlesworth , Rasmus O Bak , Sruthi Mantri , Gang Bao , David DiGiusto , John Tisdale , J Fraser Wright , Neehar Bhatia , Maria Grazia Roncarolo , Daniel P Dever , Matthew H Porteus 
In:
Source:
Publication Date: (2021)
Issue: 13: 598
Research Area:
Immunotherapy / Hematology
Stem Cells
Regenerative medicine
Cells used in publication:
CD34+ cell, human
Species: human
Tissue Origin: blood
Platform:
4D-Nucleofector® X-Unit
4D-Nucleofector® LV-Unit
Experiment

RNP electroporation (24–26, 30) was performed using the Lonza 4D-Nucleofector in (micro)Nucleocuvette and 4D-Nucleofector LV Unit in 1-ml Nucleocuvette Cartridge (medium-scale) (program DZ-100). Electroporated cells were then plated at 2.5 × 105 cells/ml in the cytokine-supplemented medium. Immediately after, rAAV6 was dispensed onto cells at multiplicities of infection of 2.5 × 103 to 1.0 × 104 vector genomes per cell on the basis of titers determined by ddPCR and incubated for 24 hours. After, a medium addition or medium exchange was performed to dilute or remove any residual gcSCD-AAV6, and the CD34+ HSPCs were cultured for an additional 12 to 24 hours.

Abstract

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 ß-globin gene (HBB). Ex vivo ß-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.