Weekly Digests
‹ Back to February

MEGA news in gene editing: using new CRISPR technology for RNA knockdown

February 28, 2024

The CRISPR-Cas9 gene editing platform has revolutionized research and medicine by allowing for precise editing of DNA. However, these systems cannot be dynamically regulated and have limitations in their safety and efficacy. To overcome some of these limitations, Tieu et al. developed multiplexed effector guide arrays (MEGA) – a CRISPR-Cas13d platform that targets sequence-specific RNA transcripts for cleavage and degradation, enabling tunable, reversible, and widespread knockdown of selected genes at the transcriptomic level. The team recently demonstrated the versatility and utility of this platform in various settings related to CAR T cell functionality. Their results were published in Cell.

To evaluate the functionality of MEGA, Tieu et al. used it to target LAG3, PD-1, and TIM3, which are typically upregulated in dysfunctional GD2-targeted HA-28ζ CAR T cells. In an optimized manufacturing workflow, primary human T cells were co-transduced with separate RfxCas13d and HA-28ζ CAR lentiviruses. The next day, cells were transduced with CRISPR-associated RNA lentiviruses carrying RNA guides that target specific RNA transcripts in the cell. Single, double, and triple guides targeting LAG3, PD-1, and TIM3 transcripts in various combinations all effectively knocked down expression of those proteins to near baseline levels, with triple combination guides showing various degrees of robustness depending on the spacer configurations between guides. Off-target or downstream biological effects, such as IFN pathway activation and collateral activity (non-specific RNA degradation) were all either minimal or not observed, even when a highly expressed gene such as B2M was selected as a target.

Next, the researchers hypothesized that a pooled double guide array library could be used to easily screen for gene pairings that could be knocked out in tandem, without the Cas9 requirement for barcoding to identify pairs, as compact RfxCas13d guide arrays can be directly sequenced in a single read. Investigating single and double guide RNAs targeting 24 exhaustion-related genes in all 576 pairwise combinations, the researchers found that co-targeting genes generally had a stronger impact on proliferation than targeting single genes. Top-ranking paired hits were functionally validated with guide- and gene-level data, and were experimentally validated in both bulk and CD8+ CAR T cell cultures, which recapitulated screening results and showed evidence of dysregulation within 48 hours after transduction.

Evaluating the impacts of these screen hits on antitumor immunity, the researchers found that top guide RNAs robustly altered IFNγ secretion and moderately improved IL-2 secretion upon tumor stimulation. This improved tumor cell killing across multiple donors and effector:target cell ratios, particularly upon serial stimulation. Notably, while single targeting of FAS or CBLB (both involved in pathways associated with the regulation of T cell activation) had no impact, simultaneous knockdown potentiated strong, durable antitumor responses across multiple tumor rechallenges, demonstrating the utility of evaluating gene knockdowns in tandem.

In the MEGA system, transcript knockdown is dependent on expression of RfxCas13d. In an effort to enable tunable and reversible control of gene knockdown, Tieu et al. fused a destabilization domain (DD) to the C-terminal of RfxCas13d, resulting in rapid degradation of the complex at steady state. However, with the addition of the FDA-approved antibiotic trimethoprim (TMP), the DD is stabilized, allowing for effective Cas13d-DD-mediated cleavage of target RNA. When this system was tested with CD46 as the target gene, the researchers observed completely reversible regulation of gene expression. Further, titration experiments showed that gene expression could be effectively tuned by altering the dosage of TMP.

Taking advantage of the ability to tune RNA expression, the researchers generated a RfxCas13d-DD to target the CAR proximal signaling genes LCK and ZAP7065, enabling tunable regulation of CAR activation. With increasing amounts of TMP in culture, this targeting array reduced CD69 expression, IL-2 secretion, and exhaustion marker upregulation in various CAR T cell models with different costimulatory and scFv domains, demonstrating that MEGA can be used to tune CAR T cell activation in an receptor-agnostic manner.

Investigating just how many genes can be simultaneously knocked down using the MEGA platform, the researchers tested guides targeting 5 (SURF1) and 10 (SURF2) exhaustion-related genes at once. Without optimization, SURF1 knocked down all 5 target genes to baseline levels or lower, and SURF2 knocked down 9 out of 10 genes at the transcriptional level – though not as fully as with SURF1 – and 8 out of 10 at the protein level. These results suggest that the MEGA platform can allow for many genes to be targeted at once, though the researchers hypothesized that guides may begin to compete for Cas13d when higher numbers of genes are targeted.

Next, the researchers developed a guide array targeting expression of four enzymes (PURI) involved in the purinergic signaling pathway, which converts extracellular inflammatory ATP into immunosuppressive adenosine (ADO). Targeting these genes effectively knocked them down, leading to accumulations of ATP, with less generation of AMP and ADO. This improved effector functions in otherwise dysfunctional CAR T cells, increasing their activation, proliferation, production of IFNγ and IL-2, and capacity for tumor killing upon stimulation in vitro.

Similarly, targeting genes for 4 key glycolytic enzymes (GLY) along the PI3K/Akt axis, disrupted the downstream transition from oxidative phosphorylation to aerobic glycolysis in activated T cells, reducing costimulation, Th1 differentiation, and inhibitory markers, and leading to widespread transcriptional reprogramming. The end result was increased oxidative phosphorylation and a fitter, less exhausted phenotype, suggestive of successful metabolic reprogramming. A similar knockdown using CRISPR-Cas9 was much less effective, and showed evidence of potential genotoxicity, suggesting a benefit to using MEGA over traditional CRISPR techniques. Evaluating antitumor efficacy, MEGA-treated GLY cells showed lower activation and secretion of IFNγ and IL-2, with fewer cells expressing IFNγ. While these cells initially cleared tumors at similar or slower rates than control CAR T cells, they outperformed upon repeated stimulations, and significantly enhanced tumor killing over time. Upon transfer into mice bearing Nalm6-GD2 tumors, MEGA-edited cells were more abundant and cleared tumors significantly better than controls.

Overall, these results show that the MEGA platform, which effectively utilizes CRISPR-Cas13d to knock down gene expression at the transcriptomic level, has numerous applications, from screening knockdowns of multiple genes at once, to tuning gene expression and CAR activation, to metabolically reprogramming cells and improving their antitumor abilities. Offering several advantages over CRISPR-Cas9, including tunability, reversibility, and reduced risk of genotoxicity, MEGA will likely serve as a handy tool in both research in medical toolkits.

Write-up and image by Lauren Hitchings

References:

Tieu V, Sotillo E, Bjelajac JR, Chen C, Malipatlolla M, Guerrero JA, Xu P, Quinn PJ, Fisher C, Klysz D, Mackall CL, Qi LS. A versatile CRISPR-Cas13d platform for multiplexed transcriptomic regulation and metabolic engineering in primary human T cells. Cell. 2024 Feb 14. 

In the Spotlight...

A signature of enhanced proliferation associated with response and survival to anti-PD-L1 therapy in early-stage non-small cell lung cancer

Altorki et al. interrogated RNAseq data from 16 pre-treatment NSCLC tumors and identified a molecular subclass of early-stage NSCLC based on 140 differentially expressed genes associated with a high proliferative index (PI) and a good response to neoadjuvant combination anti-PD-L1 and sub-ablative stereotactic body radiation therapy. High PI tumors showed features of immunosuppressive TMEs, including increased cancer cell PD-L1 expression, reduced MHC class II expression, and increased abundance of Tregs. In 6 of 7 tumors analyzed in TCGA, the 140-gene set identified a high PI subclass, and was associated with worse survival.

Contributed by Shishir Pant

PD-L1-expressing tumor-associated macrophages are immunostimulatory and associate with good clinical outcome in human breast cancer

Addressing inconsistencies about PD-L1 function on TAMs in human breast cancer (BC), Wang and Guo et al. used scRNAseq and functional and spatial cell–cell analyses to compare PD-L1+ TAMs with PD-L1- TAMs. PD-L1+ TAMs were immunostimulatory to T cells (mature and pro-inflammatory), and showed a preference for colocalizing with T cells. In contrast, PD-L1- TAMs were immunosuppressive (anti-inflammatory and pro-tumor) and interacted more frequently with cancer cells. In two independent cohorts of BC patients, those with PD-L1+/high TAMs had significantly better relapse free survival, while PD-L1-/low patients had worse clinical outcomes.

Contributed by Katherine Turner

Engineered T cells secreting anti-BCMA T cell engagers control multiple myeloma and promote immune memory in vivo

To engage bystander T cells and prevent multiple myeloma (MM) relapse, “STAb-T” cells were designed to secrete bispecific T cell engagers (anti-BCMA linked to anti-CD3). Both STAb-T and standard CAR T cells formed an immunological synapse with BCMA+ target cells and differentiated into memory populations in vivo. However, STAb-T cells demonstrated superior cytotoxicity against target cell lines and patient MM samples compared to CAR T cells, especially at low effector:target ratios, and were more effective in vivo. Even when physically separated, STAb-T cells enabled tumor killing by recruiting unactivated T cells.

Contributed by Alex Najibi

IL-23 stabilizes an effector T(reg) cell program in the tumor microenvironment

Wertheimer et al. showed that TAMs were the primary source of IL-23 in tumors, and highly activated Foxp3+ Treg cells comprised a large portion of the IL-23R+ T cells in the TME of solid tumor-bearing mice. Il-23r ablation in only Foxp3+ Treg cells led to reduced tumor growth and, in the TME, to increases of TEFF cells and phenotypically less suppressive myeloid and Treg cells, and to the reduction of Il23r-KO CD44+CD62L- effector (e)Treg cells. Under inflammatory conditions in vitro, IL-23 stabilized Foxp3 expression by Treg cells and induced their differentiation to an eTreg cell suppressive state. IL23Rhigh Treg cells in human tumors had higher expression of the eTreg cell suppressive gene profile.

Contributed by Paula Hochman

Targeting JMJD1C to selectively disrupt tumor T(reg) cell fitness enhances antitumor immunity

Long et al. identified JMJD1C, a histone demethylase, as a tumor Treg-specific epigenetic regulator that promoted Treg fitness in tumors and suppressed the antitumor immune response. JMJD1C deletion activated both AKT (via H3K9me2 methylation and PD-1 downregulation) and STAT3 signaling pathways, leading to robust IFNγ production and tumor Treg fragility. Treatment with a JMJD1C inhibitor (193D7) efficiently reduced the frequency of tumor Tregs with no effect on systemic Tregs, potentiated an antitumor T cell response, and led to tumor growth inhibition.

Contributed by Shishir Pant

High-specificity CRISPR-mediated genome engineering in anti-BCMA allogeneic CAR T cells suppresses allograft rejection in preclinical models

Allogeneic CB-011 CAR T cells were engineered with (1) an anti-BCMA CAR at the TRAC locus, to disrupt endogenous TCR expression and prevent GvHD, and (2) a B2M-HLA-E complex at the B2M locus, to avoid HLA-mismatch allograft rejection and NK cell killing. CB-011 lysed BCMA+ cell lines and patient-derived multiple myeloma (MM) cells in vitro, and treated a MM mouse model without signs of GvHD. Compared to CAR T cells without HLA knockout or B2M-HLA-E insertion, CB-011 resisted HLA-mismatched T cell lysis or NK cell killing, respectively. In NK cell-engrafted mice, CB-011 cells persisted and had lower genomic stress signatures than CAR T cells with B2M KO alone.

Contributed by Alex Najibi

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.