-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

1540 Robust Pre-Clinical Results and Large-Scale Manufacturing Process for EDIT-301: An Autologous Cell Therapy for the Potential Treatment of SCD

Program: Oral and Poster Abstracts
Session: 801. Gene Editing, Therapy and Transfer: Poster I
Hematology Disease Topics & Pathways:
sickle cell disease, Biological, HSCs, Diseases, Therapies, red blood cells, Hemoglobinopathies, gene therapy, Technology and Procedures, Cell Lineage, gene editing, transplantation, stem cells
Saturday, December 5, 2020, 7:00 AM-3:30 PM

Edouard De Dreuzy, PhD1, Jack Heath1*, Patricia Sousa1*, Tusneem Janoudi1*, Harry An, PhD1*, Scott Hansen2*, David K. Wood, PhD2*, Charles F Albright, PhD1*, Sandra Teixeira, PhD1*, Tamara Monesmith1*, Kate Zhang, PhD1 and Kai-Hsin Chang, PhD1

1Editas Medicine, Cambridge, MA
2Biomedical Engineering, University of Minnesota, Minneapolis, MN

Sickle cell disease (SCD) is an inherited blood disorder affecting approximately 100,000 individuals in the United States. As fetal hemoglobin (HbF) has been shown to be protective against clinical manifestation of SCD, we are developing EDIT-301, an autologous cell therapy comprising CD34+ cells genetically modified using a Cas12a ribonucleoprotein (RNP) to promote HbF expression to treat SCD.

Fetal hemoglobin induction for EDIT-301 is achieved by disrupting the HBG1 and HBG2 promoter distal CCAAT-box region where naturally occurring mutations are found to be associated with elevated HbF expression. Cas12a was selected over Cas9 due to the more productive and sustainable (NHEJ derived) indel profile, as well as high specificity. Using Cas12a RNP, on-target editing of ~90% was achieved in mobilized peripheral blood CD34+ cells (mPB-CD34+ cells) from both healthy and SCD donors at research scale with no detectable off targets.

Editing of CD34+ cells led to an average of 43% and 54% of HbF expression in the erythroid progeny of normal donor and SCD donor cells respectively in a pancellular fashion (~93% population). The robust HbF induction in SCD red blood cells (RBCs) resulted in significant phenotypic and functional improvement including reduced sickling and increased deformability under hypoxia ex vivo. Using a microfluidic assay that replicated blood flow in microvasculature under varying oxygen conditions, SCD RBCs derived from RNP electroporated CD34+ cells showed improved rheological behavior. The rheology improvement under hypoxia was strongly correlated with the increased levels of HbF in each sample.

Infusion of the modified CD34+ cells from normal donors into NBSGW mice resulted in long-term multi lineage and polyclonal reconstitution. Editing levels at 16 weeks post infusion were > 90% in all human lineages tested, demonstrating the efficient editing of SCID-repopulating hematopoietic stem cells (HSCs). Consistent with the high editing levels, human erythroid cells from the bone marrow of mice that received Cas12a-RNP treated cells demonstrated pancellular (~90% F+ RBCs) HbF expression averaging 40-50% of total hemoglobin compared to ~5% HbF observed in the control group.

We have developed a consistent large-scale process using functionally closed, semi-automated systems suitable for use in clinical manufacturing. We have shown robust editing of normal donor CD34+ cells and SCID-repopulating HSCs with the clinical scale process. Editing levels of >90% were detected after long term engraftment in mice.

In summary, we have demonstrated successful on-target editing of mPB CD34+ cells derived from both normal and SCD donors using a Cas12a RNP, which coincided with robust HbF induction and a phenotypic reduction of sickling in the SCD erythroid progeny, as well as improved rheological behavior. Editing of the HBG1 and HBG2 promoters using this RNP was highly specific with no measurable off-target. In vivo, cells from normal donors readily engrafted and reconstituted all blood cell lineages at levels comparable to unedited cells. Finally, a robust large-scale manufacturing process has been developed to supply material for the clinical setting. Based on these results, we are completing the activities required to assess EDIT-301 in the clinic as treatment for SCD.

Disclosures: De Dreuzy: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company. Heath: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company. Sousa: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company. Janoudi: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company. An: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company. Albright: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company. Teixeira: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company. Monesmith: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company. Zhang: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company. Chang: Editas Medicine Inc.: Current Employment, Current equity holder in publicly-traded company.

*signifies non-member of ASH