Learn More
Learn More
Learn More
Learn More
News & Blogs » CRISPR News » Multiplex Editing T Cells with BE and Cas12 | GenScript
Author: Dr. Wanjia Zeng
Senior Scientist of Gene Editing
Traditional autologous CAR-T therapies, which rely on patient-derived T cells, face significant limitations, including lengthy manufacturing times, high costs, and variability in cell quality, particularly for patients with compromised immune systems. In contrast, allogeneic CAR-T therapies, derived from healthy donor cells, enable standardized, large-scale production—a single batch can treat hundreds of patients, drastically reducing costs and wait times while improving accessibility.
The emergence of allogeneic, or "off-the-shelf," CAR-T therapies like Sana Biotechnology’s SC291 and Allogene Therapeutics’ ALLO-329, both recently granted the Food and Drug Administration (FDA) Fast Track Designations (FTD) for autoimmune diseases, underscores the critical need for scalable and accessible treatments in this field.1,2
SC291, a CD19-targeted hypoimmune (HIP)-modified CAR-T, leverages gene editing to evade immune rejection, enabling deep B-cell depletion to reset dysregulated immunity in conditions like systemic lupus erythematosus (SLE). Similarly, ALLO-329, a dual-targeted CD19/CD70 CAR-T, incorporates CRISPR-based engineering and Allogene’s Dagger® technology to eliminate dysfunctional cells and minimize lymphodepletion, addressing the major barrier in autoimmune patients.
The FDA’s FTD recognition for these allogeneic CAR-T therapies highlights the urgency to address unmet needs in refractory autoimmune diseases, where current therapies often fail to provide sustained remission. Allogeneic CAR-T platforms not only promise broader patient access but also pave the way for transformative, one-time treatments capable of achieving long-term immune tolerance, reshaping the therapeutic landscape for autoimmune diseases.
Allogeneic cell therapies often require multiplex gene editing to eliminate immune rejection (e.g., disrupting TCR and MHC genes) and enhance therapeutic function. However, traditional CRISPR/Cas9-based methods, which induce double-strand breaks (DSBs), carry risks of chromosomal translocations and large deletions when multiple edits occur simultaneously.3 These genomic aberrations, caused by error-prone DNA repair mechanisms, raise safety concerns such as oncogenic transformation or loss of critical genes.
To mitigate these risks, base editing (BE) has emerged as a precise alternative. Unlike CRISPR/Cas9, base editors directly convert single DNA bases without creating DSBs, minimizing genomic instability. This approach enables efficient gene knockouts or functional corrections while preserving chromosomal integrity.
In cell therapies, base editing enhances safety for applications like hypoimmune CAR-T cells or correcting disease-causing mutations in hematopoietic stem cells.4 An early study demonstrates its potential to streamline allogeneic product development by reducing off-target effects and enabling multiplex editing with lower toxicity, positioning it as a transformative tool for next-generation genetic medicines.5
To replace CRISPR/Cas9 with BE for multiplex gene disruption, we evaluated the editing efficiency of CBEmax mRNA and ABE protein targeting B2M, TRAC, PDCD1, CIITA in Jurkat cells. We electroporated BE and sgRNA in Jurkat cells and detected editing efficiency by Sanger sequencing and BEAT analysis 3 days after electroporation.
Both BE formats achieved efficient base conversion at splice donor sites of all target genes, enabling robust gene knockout. Notably, CBEmax and ABE demonstrated comparable editing rates (~90%), confirming the feasibility of BE for eliminating immune-related antigens and checkpoint molecules without double-strand breaks.
Multiplex Editing with CBEmax and ABE base editors. Editing efficiency of TRAC/B2M/PDCD1/CIITA by CBEmax and ABE base editors. Both base editor formats achieved efficient base conversion and gene knockout.
When combining nCas9-based BE for gene knockout with Cas9-mediated knock-in, unintended gRNA exchange between the two systems may compromise editing efficiency. To circumvent this difficulty, a Cas12-based knock-in system can be paired with BE-mediated knockout. This strategy leverages the orthogonal scaffold sequences required for the ribonucleoprotein (RNP) complex formation by Cas12 versus nCas9, effectively preventing cross-utilization of gRNAs.6 By utilizing distinct CRISPR-Cas enzymes (nCas9 for BE and Cas12 for knock-in), the systems operate independently, eliminating interference between base-editing and knock-in machinery.
Combining multiple Cas protein types in multiplex editing. Using different Cas families, such as Cas12 RNP and nCas9RNP for KI and base editing, prevents unwanted gRNA exchange.6
To evaluate the multiplex editing efficacy of this combinatorial approach, we tested Cas12-mediated knock-in efficiency at the TRAC locus in primary T cells while simultaneously knocking out B2M and PDCD1 using ABE8e mRNA to establish an allogeneic CAR-T model.
We designed a single-stranded GFP donor template flanked by TRAC homology arms and incorporated Cas12-targeting sequences (CTS) at either the 5’ end (5’ only) or both ends (5’+ 3’). These CTS regions were annealed with complementary oligonucleotides to generate localized double-stranded structures, enhancing nuclear delivery and homology-directed repair (HDR) efficiency by facilitating Cas12 RNP binding.7
To achieve B2M/PDCD1/TRAC knockout and GFP knock-in at the TRAC locus, we electroporated T cells with ABE8e mRNA/sgRNA, Cas12/crRNA, and ssCTS template. We also set up a control group with GFP knock-in at the TRAC locus only.
ABE8e mRNA | sgRNA | Cas12 protein | TRAC crRNA | ssCTS HDRT | Notes | |||
---|---|---|---|---|---|---|---|---|
1 µg | B2M | 15 pmol | Cas12-1/2/3/4/5 | 10 pmol | 20 pmol | TRAC_EGFP-ssCTS-5'-only | 2 µg | B2M/PDCD1 double KO by ABE8e TRAC_EGFP KI by Cas12 |
PDCD1 | 15 pmol | TRAC_EGFP-ssCTS-5'+3' | 2 µg | |||||
/ | / | / | Cas12-1/2/3/4/5 | 10 pmol | 20 pmol | TRAC_EGFP-ssCTS-5'-only | 2 µg | TRAC_EGFP KI by Cas12 |
TRAC_EGFP-ssCTS-5'+3' | 2 µg |
Cas12 RNP demonstrated robust knock-in efficiency at the TRAC locus, with multiple edited groups showing comparable performance to TRAC knock-in only controls. This finding confirms that BE-mediated B2M and PDCD1 knockout does not interfere with Cas12-driven integration.
This orthogonal approach enables efficient and precise multiplex genome editing in T cells without cross-system interference. Notably, ssCTS-5’+ 3’ designs significantly improved knock-in efficiency for most Cas12 variants (vs. ssCTS-5’-only), which is different from Cas9-mediated HDR, where the ssCTS-5’only design works better.
Validating multiplex editing efficiency combining Cas12 and ABE8e. Editing efficiency of GFP insertion at TRAC by Cas12 RNP-mediated HDR, with or without gene knockout by ABE8e mRNA. Efficient multiplex genome editing in T cells without cross-system interference.
Flow cytometry further validated the gene knockout efficiency, with ABE8e mRNA achieving>95% depletion of B2M and PD-1 proteins. Most Cas12 variants editing at TRAC eliminated TCR expression in 93% of edited cells. These results underscore the compatibility of combining BE and Cas12 systems to simultaneously disrupt MHC-I and immune checkpoints, deplete endogenous TCR, and integrate therapeutic transgenes via non-viral gene editing—a critical advancement for developing allogeneic CAR-T therapy with enhanced safety and efficacy.
Flow cytometry for validation of gene knock-out efficiency under multiplex gene editing conditions. High gene knockout efficiency of TRAC/B2M/PDCD1 in T cells measured by protein staining and flow cytometry.
Our data demonstrate the feasibility of combining base editing and Cas12-mediated knock-in to achieve efficient multiplex genome editing in primary T cells. This approach enables simultaneous disruption of B2M, PDCD1, and TRAC genes for allogeneic CAR-T engineering, which is ideal for developing “off-the-shelf” cell therapies. We will expand this platform by testing multiple editing strategies and optimizing donor templates. By systematically evaluating editing efficiency, off-target effects, and long-term functional persistence, we aim to refine multiplex editing workflows and accelerate the development of next-generation “off-the-shelf” allogeneic therapies with enhanced safety, potency, and manufacturability.