Epitope base editing CD45 in hematopoietic cells enables universal blood cancer immune therapy

Authors:
Wellhausen N, O'Connell RP, Lesch S, Engel NW, Rennels AK, Gonzales D, Herbst F, Young RM, Garcia KC, Weiner D, June CH, Gill SI.
In:
Source: Science
Publication Date: (2023)
Issue: eadi: 1145
Research Area:
Cancer Research/Cell Biology
Immunotherapy / Hematology
Basic Research
Molecular Biology
Cells used in publication:
T cell, human stim.
Species: human
Tissue Origin: blood
CD34+ cell, human
Species: human
Tissue Origin: blood
Monocyte, human
Species: human
Tissue Origin: blood
Platform:
4D-Nucleofector® X-Unit
Experiment

T cell nucleofection: T cells were activated using Dynabeads CD3/CD28 CTS (Gibco, Life Technologies) at a 3:1 bead-to-cell ratio. 24 hrs after bead activation, T cells were transduced with lentivirus at an MOI of 3. 48 hrs after stimulation, beads were removed and T cells were spun down at 300 xg, washed with PBS once and resuspended at a concentration of 5e6 cells/100 µL in P3 Solution with Supplement (Lonza). Primary T cells were electroporated using the Lonza 4D-Nucleofector Core/X Unit and the P3 Primary Cell 4-D Kit (Lonza). For CRISPR/Cas knockouts, the ribonucleoprotein (RNP) complex was formed by incubating 20 µg of Spy Fi Cas9 (Aldevron) with 10 µg of sgRNA (Table S3) (IDT) for 15 mins at room temperature (RT). The RNP complex and 100 µL of resuspended cells were combined and electroporated in a cuvette. For base-editing, 10 µg mRNA (Trilink, N1-Methylpseudouridine, CleanCap) encoding the base-editor (ABE8e) were mixed with 10 µg sgRNA (IDT) and combined with 100 µL of resuspended cells. Pulse code EO-115 were used for primary T cells. After electroporation, the cells were recovered in CTS Optimizer at 2e6 cells/mL at 37°C for 24 hrs. Cells were counted daily using the Multisizer 4 Coulter Counter (Beckman Coulter). T cells were grown for 8–10 days in Optimizer media containing 5 ng/ml of IL-7 and IL-15 and maintained at 1e6 cells/ml prior to cryopreservation. T cells were thawed and rested at 37°C for 16 hours before experiments.

Primary human CD34+ cells isolation and electroporation: G-CSF mobilized peripheral blood samples were obtained healthy volunteers enrolled on an IRB approved protocol (University of Pennsylvania IRB protocol # 832307). Human subjects were deidentified and thus age and sex information are not available. CD34+ selection was performed using the CD34 Microbead Kit (Miltenyi Biotec, 130–046-702), and purity was confirmed by flow cytometry to be >95%. Cells were cryopreserved in FBS with 10% DMSO until use. Prior to all experiments, CD34+ cells were thawed and cultured in StemSpan SFEM (Stem Cell Technologies, 09650) supplemented with human cytokines (SCF 100 ng/µl, Flt3 ligand 100 ng/µl, TPO 50 ng/µl, IL-6 50 ng/µl, all purchased from Peprotech) for 24 hrs. Nucleofection was performed as described for T cells using pulse code DZ-100. Cells were recovered at 2 x 10^6 cells/mL in StemSpan SFEM (Stem Cell Technologies, 09650) supplemented with human cytokines (SCF 100 ng/µl, Flt3 ligand 100 ng/µl, TPO 50 ng/µl, IL-6 50 ng/µl, all purchased from Peprotech) for 24 hrs at 37°C.

Primary human monocyte base editing: Human monocytes were obtained from PBMCs of healthy donors through the Human Immunology Core at the University of Pennsylvania. Monocytes were cultured in RPMI 1640 + 5% human serum, 1% penicillin/streptomycin (50 IU/ml), 1% Glutamax) (Gibco, Life Technologies) supplemented with 50 ng/mL of MCSF (Peprotech) at 37°C in 5% CO2. After 24 hrs in culture, monocytes were nucleofected with ABE8e mRNA and sgRNA as described for T cells using pulse code CM-137. Cells were recovered at 2 x 10^6 cells/mL in RPMI 1640 + 5% human serum, 1% penicillin/ streptomycin (50 IU/ml), 1% Glutamax) (Gibco, Life Technologies) supplemented with 50 ng/mL of M-CSF (Peprotech) and cultured for 5 days prior to experimental use to allow for sufficient protein turnover.

Abstract

In the absence of cell-surface cancer-specific antigens, immunotherapies such as chimeric antigen receptor (CAR) T cells, monoclonal antibodies, or bispecific T cell engagers typically target lineage antigens. Currently, such immunotherapies are individually designed and tested for each disease. This approach is inefficient and limited to a few lineage antigens for which the on-target/off-tumor toxicities are clinically tolerated. Here, we sought to develop a universal CAR T cell therapy for blood cancers directed against the pan-leukocyte marker CD45. To protect healthy hematopoietic cells, including CAR T cells, from CD45-directed on-target/off-tumor toxicity while preserving the essential functions of CD45, we mapped the epitope on CD45 that is targeted by the CAR and used CRISPR adenine base-editing to install a function-preserving mutation sufficient to evade CAR T cell recognition. Epitope edited CD45 CAR T cell were fratricide-resistant and effective against patient-derived acute myeloid leukemia, B cell lymphoma, and acute T cell leukemia. Epitope edited hematopoietic stem cells (HSCs) were protected from CAR T cells and, unlike CD45 knockout cells, could engraft, persist, and differentiate in vivo. Ex vivo epitope editing in HSCs and T cells enables the safe and effective use of CD45- directed CAR-T cells and bispecific T cell engagers for the universal treatment of hematologic malignancies and might be exploited for other diseases requiring intensive hematopoietic ablation.