CAR-T Cells RNA Silencing Guide
Enhance persistence, prevent exhaustion, and optimize manufacturing—all without transfection

Why Gene Silencing Enhances CAR-T Cells
Chimeric Antigen Receptor (CAR) T cells represent a revolutionary cancer immunotherapy approach, redirecting T cells to target tumor antigens through engineered receptors. A functional CAR consists of an extracellular single-chain variable fragment (scFv) for antigen recognition, a hinge region, a transmembrane domain, one or more costimulatory domains (CD28 or 4-1BB for second-generation CARs), and the CD3ζ signaling domain for T cell activation.
Despite remarkable clinical successes in hematologic malignancies, CAR-T cells face critical manufacturing and therapeutic challenges: T cell exhaustion during the 7-14 day expansion phase, transfection-induced toxicity when introducing gene modifications, target antigen-mediated fratricide (particularly for CD7-targeting and CD5-targeting CARs), poor persistence in solid tumor microenvironments, and manufacturing costs exceeding $400,000-$500,000 per patient dose.
AUMsilence sdASO is a powerful tool for discovering gene function in CAR-T cells. Researchers use gymnotic ASO delivery to systematically knock down genes and understand their roles in CAR-T exhaustion, persistence, signaling, metabolism, and tumor interactions. This functional genomics approach accelerates target discovery and mechanistic understanding without the complexity and off-target effects of CRISPR screening.
AUMsilence self-delivering ASO technology addresses these challenges without disrupting CAR-T manufacturing workflows. By enabling transfection-free gene silencing, AUMsilence preserves CAR-T cell viability above 95% while enhancing therapeutic properties through checkpoint knockout (PD-1, CTLA-4, LAG-3), exhaustion prevention (TOX, NR4A1 knockdown), fratricide prevention (target antigen silencing), and tumor microenvironment resistance (TGFβR2 knockdown).
The gymnotic delivery mechanism allows simple addition to culture medium—no electroporation, no lipofection, no viral transduction required. This seamless integration with existing activation, transduction, and expansion protocols makes AUMsilence ideal for both research-scale CAR-T optimization and clinical-scale manufacturing enhancement.
Critical Challenges in CAR-T Cell Manufacturing and Function
CAR-T cells face unique biological and manufacturing barriers that limit therapeutic efficacy and scalability:
T Cell Exhaustion During Manufacturing
Repeated stimulation during the 7-14 day expansion phase causes progressive upregulation of inhibitory receptors (PD-1, TIM-3, LAG-3) and exhaustion transcription factors (TOX, NR4A1). This exhausted phenotype reduces proliferative capacity, cytokine production (IFN-γ, TNF-α), and cytotoxic function before CAR-T cells even reach the patient. Studies show 40-60% of CAR-T cells exhibit exhaustion markers by Day 10-14 of manufacturing.
High ImpactTransfection Toxicity in Manufacturing Workflows
Introducing genetic modifications (checkpoint knockout, HLA silencing for universal CAR-T) via electroporation or lipofection causes 25-50% cell death during the critical expansion phase. This toxicity extends manufacturing timelines, reduces yield, and alters T cell phenotype distribution (increased effector memory, reduced central memory). The resulting CAR-T product has compromised persistence potential.
High ImpactTarget Antigen-Mediated Fratricide
CAR-T cells targeting antigens expressed on normal T cells (CD7 for T-cell acute lymphoblastic leukemia, CD5 for T-cell lymphoma, BCMA in rare cases) undergo fratricide—killing each other during manufacturing. This severely reduces or completely prevents manufacturing yield. Current solutions require complex genome editing (CRISPR knockout of target antigen), which introduces regulatory complexity and potential off-target effects.
High ImpactPoor Tumor Persistence and Trafficking in Solid Tumors
CAR-T cells face immunosuppressive solid tumor microenvironments rich in TGF-β, adenosine, and metabolic stressors. Checkpoint receptor upregulation (PD-1, LAG-3, TIM-3) combined with inadequate tumor infiltration limits efficacy. While CAR-T succeeds in hematologic malignancies (where tumor cells are accessible), solid tumor response rates remain below 10-20% in most trials.
High ImpactManufacturing Scalability and Cost
Current autologous CAR-T manufacturing requires individualized processing for each patient: leukapheresis, T cell isolation, activation, viral transduction, expansion (7-14 days), formulation, and cryopreservation. This complex workflow costs $400,000-$500,000 per dose with 10-20% manufacturing failure rates. Allogeneic "off-the-shelf" CAR-T could reduce costs but requires HLA silencing to prevent graft-versus-host disease.
High ImpactCytokine Release Syndrome (CRS) Risk
Rapid CAR-T expansion and activation in vivo triggers massive cytokine release (IL-6, IFN-γ, TNF-α, IL-1), causing potentially fatal systemic inflammation. While manageable with tocilizumab (anti-IL-6R), severe CRS remains a major safety concern limiting CAR-T dosing and accessibility. Reducing CAR-T inflammatory cytokine production without compromising anti-tumor efficacy could improve safety profiles.
Medium ImpactMethod Comparison
Method | Efficiency | Viability | Pros | Cons |
---|---|---|---|---|
Lipofection (Cationic Lipid Reagents) | 20-40% | 30-50% | Simple, commercially available | Severe toxicity in activated CAR-T cells, activation-induced cell death, disrupts manufacturing timeline |
Electroporation (Neon, 4D-Nucleofector) | 50-70% | 50-70% | Moderate efficiency | Significant cell death, phenotypic changes (loss of central memory), alters expansion kinetics, expensive |
CRISPR/Cas9 (Permanent Knockout) | 60-85% | 60-75% | Permanent gene knockout, stable phenotype | Requires electroporation (toxicity), off-target mutagenesis risk, complex regulatory path for clinical use, expensive |
Viral shRNA (Lentiviral Integration) | 70-90% | 75-85% | High efficiency, stable knockdown | Requires additional viral vector (safety concerns), insertional mutagenesis, GMP production complexity, 2-4 week timeline |
AUMsilence sdASO | 70-95% | >95% | No transfection, no manufacturing disruption, preserves central memory phenotype, scalable (add to medium), transient knockdown ideal for optimization, GMP-compatible | Transient knockdown (appropriate for manufacturing optimization; re-dose for sustained effect) |
AUMsilence sdASO
Why This Product?
AUMsilence self-delivering ASOs are uniquely suited for CAR-T cell engineering because they eliminate transfection-induced toxicity that plagues conventional gene editing approaches. CAR-T manufacturing requires maintaining high viability (>80%) throughout the 7-14 day expansion phase—lipofection and electroporation cause 25-50% cell death, extending timelines and reducing therapeutic potential. AUMsilence preserves >95% viability while enabling checkpoint knockout, exhaustion prevention, and fratricide elimination through simple addition to culture medium.
Key Benefits
Maintains >95% CAR-T Viability
Eliminates transfection-induced cell death that reduces manufacturing yield. Critical for meeting clinical dose requirements (typically 1-5 × 10⁸ CAR-T cells per patient).
Compatible with Viral CAR Transduction
AUMsilence does not interfere with lentiviral or retroviral CAR vector transduction. Can be added before, during, or after transduction without affecting CAR expression levels or transduction efficiency.
Prevents Exhaustion During Expansion
Silencing TOX, NR4A1, or checkpoint receptors during the 7-14 day expansion prevents exhaustion programming. Resulting CAR-T cells show enhanced proliferation, cytokine production, and persistence in co-culture assays.
Enables Fratricide-Prone CAR-T Manufacturing
Knockdown of CD7, CD5, or other T-cell-expressed antigens allows successful manufacturing of CAR-T products that would otherwise kill themselves during expansion. No permanent genome editing required.
Rapid Optimization Timeline
Test checkpoint knockout, exhaustion prevention, or other enhancements in 2-3 week experiments. No viral vector cloning, no CRISPR guide RNA optimization. Accelerates translational research.
Regulatory Simplicity vs. CRISPR
Transient ASO-mediated knockdown avoids permanent genome editing and associated regulatory requirements. Ideal for research and manufacturing process optimization.
Ideal For
- CAR-T checkpoint enhancement (PD-1, CTLA-4, LAG-3, TIM-3 knockout)
- Exhaustion prevention during manufacturing (TOX, NR4A1 knockdown)
- Fratricide prevention (CD7, CD5, BCMA knockdown for respective CARs)
- Tumor microenvironment resistance (TGFβR2, IL-10R silencing)
- Universal CAR-T research (HLA Class I knockdown via B2M)
- Cytokine release syndrome mitigation (TNF-α, IL-6 modulation)
- CAR-NK cell enhancement (checkpoint knockout in NK-92 or primary NK cells)
- Translational CAR-T research and manufacturing optimization
Alternative Products
AUMsaver toASO
When to use: For targeting specific RNA structures or splice variants in CAR-T cells. Toe-hold ASOs provide alternative mechanism when standard gapmer ASOs show suboptimal knockdown.
Learn More →Custom ASO Design Service
When to use: For novel CAR-T enhancement targets or multi-gene combinatorial knockdown strategies. AUM scientists design and validate 3-5 ASO candidates per target.
Learn More →AUMsilence Protocols for CAR-T Cell Enhancement
Optimized protocols for integrating gene silencing into CAR-T manufacturing workflows. No transfection reagents or specialized equipment required.
Quick Start Protocol (All CAR-T Applications)
- Activate T cells (anti-CD3/CD28 beads, 1:1 ratio) at 1 × 10⁶ cells/mL in RPMI + 10% FBS + IL-2 (50-100 U/mL)
- At Day 2-3 post-activation, add AUMsilence sdASO directly to culture at 10 μM (no transfection reagent)
- Proceed with lentiviral or retroviral CAR transduction at Day 3-4 (ASO does not interfere)
- Continue expansion for 7-10 days; re-dose ASO every 3-4 days if sustained knockdown needed
- Validate knockdown by qRT-PCR (48-72h) and flow cytometry (72-96h); confirm CAR expression maintained
Cell-Type-Specific Protocols
Silence inhibitory receptors before CAR transduction for enhanced CAR-T persistence
T Cell Isolation and Activation
Isolate CD3+ T cells from PBMCs (negative selection). Activate with anti-CD3/CD28 magnetic beads (1:1 bead-to-cell ratio) in complete RPMI + 10% FBS + IL-2 (50-100 U/mL). Seed at 1 × 10⁶ cells/mL in T-cell culture flasks or G-Rex devices.
AUMsilence Treatment (Pre-Transduction)
At Day 2-3 post-activation, add AUMsilence targeting PD-1 (PDCD1), CTLA-4, or LAG-3 at 10 μM directly to culture. For example: targeting PD-1 to create checkpoint-resistant CAR-T. No media change required. Beads remain in culture.
CAR Transduction
At Day 3-4 (24-48h post-ASO treatment), transduce with lentiviral or retroviral CAR vector at MOI 3-10. AUMsilence does not interfere with viral transduction. Continue culture with IL-2.
CAR-T Expansion
Expand CAR-T cells for 7-10 additional days. Monitor cell density (split to 0.5-1 × 10⁶/mL when exceeding 2 × 10⁶/mL). Re-dose AUMsilence every 3-4 days if sustained knockdown desired.
Validation and Functional Testing
Validate knockdown: qRT-PCR for target mRNA (expect 70-90% reduction), flow cytometry for protein (PD-1, CTLA-4 surface expression). Confirm CAR expression by Protein L or anti-idiotype staining (expect no reduction). Perform cytotoxicity assays against target cells.
Essential Controls for CAR-T Enhancement
Optimization Strategies for CAR-T Applications
Timing in Manufacturing Workflow
Recommendation: Pre-transduction (Day 2-3) for checkpoint knockout and fratricide prevention. Post-transduction (Day 5-7) for exhaustion prevention.
Rationale: Pre-transduction establishes phenotype before CAR introduction. Post-transduction ensures CAR expression established before modifying T cell state.
ASO Concentration
Recommendation: 10 μM standard. Use 5 μM for highly sensitive targets. Higher concentrations (up to 20 μM) for highly stable genes or rapid cell division.
Rationale: CAR-T cells undergo rapid proliferation during expansion (doubling every 24-48h), which dilutes ASO. Higher concentrations or re-dosing compensates.
Duration and Re-Dosing
Recommendation: Single dose sufficient for 3-4 days of knockdown. Re-dose every 3-4 days during expansion (Days 2, 6, 10) for sustained effect throughout manufacturing.
Rationale: Rapid cell division during CAR-T expansion dilutes ASO. Re-dosing maintains knockdown throughout 7-14 day manufacturing timeline.
Combination with CAR Transduction
Recommendation: AUMsilence does NOT interfere with lentiviral or retroviral transduction. Can be added before, during, or after transduction.
Rationale: ASOs target endogenous genes, not viral vectors or CAR transgene. No impact on transduction efficiency or CAR expression.
Multi-Target Knockdown
Recommendation: Can combine multiple ASOs (e.g., PD-1 + LAG-3 dual checkpoint knockout). Use 5-10 μM per ASO. Total concentration should not exceed 20 μM.
Rationale: Combinatorial checkpoint blockade may provide additive benefits. Limit total ASO to avoid non-specific effects.
ASO Sequence Selection
Recommendation: Design and test 3-5 ASOs targeting different regions. Select sequence with highest knockdown and no impact on CAR expression.
Rationale: Knockdown efficiency varies by target site (30-90% range). Multiple sequences ensure optimal efficacy and confirm specificity.
Troubleshooting
Validation Methods for CAR-T Enhancement
Comprehensive validation ensures robust CAR-T optimization with maintained viability and enhanced function.
Purpose: Confirm target gene silencing at mRNA and protein levels
Protocol: For mRNA: Extract RNA at 48-72h post-ASO treatment, perform qRT-PCR with target-specific primers, normalize to GAPDH/ACTB. For protein: Surface markers by flow cytometry at 72-96h (PD-1, CTLA-4, CD7, CD5, etc.), intracellular proteins by fix/perm staining (TOX, NR4A1, etc.). Include viability dye (7-AAD or Live/Dead Aqua).
Expected Results: 70-95% mRNA knockdown by qRT-PCR. 60-85% protein reduction by flow cytometry (MFI shift for surface markers). >95% viability.
Tips: For surface markers, use median fluorescence intensity (MFI) comparison rather than percentage positive (some checkpoints have bimodal expression). For transcription factors (TOX, NR4A1), fix/permeabilize and use validated intracellular antibodies.
Critical Controls for CAR-T Validation
Untreated CAR-T Cells
Purpose: Baseline CAR-T function and expansion
Activate, transduce, and expand CAR-T identically to experimental group without ASO addition. This is the gold standard comparison for all functional assays.
Non-Targeting Control ASO
Purpose: Control for non-specific ASO effects on CAR-T biology
Use AUM non-targeting control ASO at same concentration (10 μM) and timing as experimental ASO. Verifies that phenotypic changes are target-specific, not ASO-related.
Mock-Transduced T Cells + ASO
Purpose: Separate ASO effects on T cells from CAR-specific effects
Treat T cells with ASO but skip CAR transduction. Validates that ASO effects (e.g., checkpoint knockdown) occur in T cells independent of CAR expression.
CAR-T Without ASO, Stimulated with Target
Purpose: Baseline CAR-T exhaustion kinetics
Co-culture untreated CAR-T with tumor targets for extended periods (7-14 days with weekly target replenishment). Measure progressive exhaustion (PD-1, TIM-3, LAG-3 upregulation, loss of killing). Compare to ASO-treated CAR-T in same assay.
Dose-Response Verification
Purpose: Confirm concentration-dependent knockdown and optimal dosing
Test minimum 3 concentrations (e.g., 5, 10, 20 μM AUMsilence). Knockdown efficiency should correlate with concentration. Validate that 10 μM is in linear range (not saturated or insufficient). Essential for ruling out off-target effects and optimizing protocol.
Independent ASO Verification
Purpose: Confirm target specificity with second ASO sequence
Design 3-5 ASOs targeting different regions of same mRNA. Test all sequences and select top 2 performers. Validate that independent ASO sequences produce concordant phenotypes (improved killing, reduced exhaustion, etc.). This is gold standard for confirming on-target effects and eliminating sequence-specific artifacts.
Best Practices
- Use biological triplicates (n=3 independent CAR-T manufacturing runs, ideally different donors)
- Validate knockdown at both mRNA (qRT-PCR, 48-72h) and protein (flow cytometry, 72-96h) levels
- ALWAYS confirm CAR expression is unaffected (Protein L staining)—critical QC checkpoint
- Include serial tumor rechallenge assays to test CAR-T persistence (mimics repeated antigen encounter in vivo)
- For functional validation, use low E:T ratios (1:1 or 3:1) where differences are most apparent
- Monitor exhaustion marker co-expression (PD-1+ TIM-3+ LAG-3+ triple-positive) as sensitive exhaustion metric
- Report CAR-T viability, expansion fold-change, and memory phenotype distribution in all studies
- Compare to literature benchmarks for CAR-T expansion (6-10 population doublings over 14 days is typical)
Research Applications for CAR-T Enhancement
AUMsilence enables diverse strategies to optimize CAR-T cell manufacturing, persistence, and anti-tumor efficacy.
Use AUMsilence to systematically knock down genes and discover their functions in CAR-T cells. Identify regulators of exhaustion, persistence, cytotoxicity, and trafficking through functional genomics approaches.
Systematic Gene Function Discovery in CAR-T Exhaustion
Approach: Design a candidate gene screen targeting transcription factors, signaling molecules, and epigenetic regulators. Knock down each gene individually in CAR-T cells during expansion (Day 5-7 post-transduction). Measure exhaustion markers (PD-1, TIM-3, LAG-3, CD39) by flow cytometry at Day 10-14. Identify genes whose knockdown prevents exhaustion. Validate hits with independent ASO sequences and mechanistic studies (RNA-seq, ATAC-seq to understand how the gene regulates exhaustion programs).
AUM Advantage: Transient knockdown is ideal for functional screens—avoids lethality from permanent knockout while revealing gene function. Can test 50-100 genes in parallel using arrayed format (24-well or 96-well plates). Faster than CRISPR screens and no off-target mutagenesis. Identifies both cell-intrinsic regulators (transcription factors) and cell-extrinsic regulators (secreted factors, receptors).
Mapping CAR Signaling Pathways Through Systematic Knockdown
Approach: Target components of CAR signaling cascades: proximal signaling (LCK, ZAP70, SLP-76, LAT), MAPK pathway (RAS, RAF, MEK, ERK), PI3K/AKT pathway (PI3K subunits, AKT isoforms, mTOR), calcium signaling (STIM1, ORAI1, calcineurin, NFAT family), and costimulatory pathways (CD28 effectors: GRB2, VAV1, PI3K; 4-1BB effectors: TRAF2, NFκB pathway). Knock down each component and measure CAR-T activation (CD69, CD25 upregulation), cytokine production (IFN-γ, TNF-α, IL-2), proliferation (Ki67, CFSE dilution), and cytotoxicity (tumor killing assays). Use phosphoproteomics to map signal flow.
AUM Advantage: Enables pathway mapping without permanent gene disruption. Can perform time-course knockdowns (dose ASO at different timepoints, measure signaling dynamics). Reveals pathway hierarchy and identifies rate-limiting steps. Distinguishes between CAR signaling and TCR signaling by comparing pathways in CAR-T vs conventional T cells. Identifies synthetic lethal combinations (dual knockdowns that completely block CAR function).
Transcription Factor Network Analysis in CAR-T Differentiation
Approach: Target transcription factors controlling T cell fate decisions: exhaustion (TOX, TOX2, NR4A1/2/3, NFAT, BATF), memory (TCF1/TCF7, LEF1, EOMES, ID3), effector (T-bet, BLIMP1), and stemness (KLF2, KLF4, FOXO1). Knock down each transcription factor during CAR-T expansion and track differentiation markers (CD62L, CCR7, CD45RA/RO, CD127, KLRG1, CD39, TOX) by flow cytometry. Perform single-cell RNA-seq to map differentiation trajectories. Use ATAC-seq to understand chromatin remodeling. Build regulatory network maps showing transcription factor hierarchy.
AUM Advantage: Transient knockdown reveals dynamic transcription factor roles without compensatory gene expression from permanent knockout. Can knockdown at different expansion timepoints (Day 3 vs Day 7 vs Day 10) to identify critical windows for differentiation decisions. Enables combinatorial knockdowns (e.g., TOX + NR4A1) to study transcription factor crosstalk. Identifies master regulators that can be targeted to enrich memory CAR-T or prevent exhaustion.
Metabolic Gene Discovery for CAR-T Fitness
Approach: Target metabolic pathways: glycolysis (GLUT1, HK2, LDHA), oxidative phosphorylation (mitochondrial complex components), fatty acid oxidation (CPT1A, ACAD enzymes), amino acid metabolism (glutaminase, serine synthesis pathway), and metabolic checkpoints (mTOR pathway, AMPK, HIF1α, MYC). Knock down each gene and measure CAR-T proliferation, survival, cytokine production, and tumor killing. Profile metabolism using Seahorse (OCR/ECAR), metabolomics (LC-MS), and isotope tracing (13C-glucose/glutamine). Identify metabolic vulnerabilities and fitness genes.
AUM Advantage: Metabolic gene knockdown without permanent disruption avoids selection bias (cells with complete knockout may die or adapt). Can study metabolic transitions during CAR-T expansion (Tscm oxidative → Teff glycolytic → Tex dysfunctional). Test metabolic gene requirements in different nutrient conditions (high glucose vs low glucose, normoxia vs hypoxia). Identifies druggable metabolic targets to enhance CAR-T function.
Comparative CAR-T vs Conventional T Cell Biology
Approach: Perform parallel knockdown screens in CAR-T cells and conventional T cells (anti-CD3/CD28 activated). Target same gene set and compare phenotypes. Identify genes that are CAR-specific (only affect CAR-T function, not conventional T cells) vs T-cell-general (affect both). Study why CAR-T cells exhaust faster than conventional T cells—are exhaustion genes more important in CAR context? Map differences in signaling pathway requirements, transcription factor dependencies, and metabolic needs.
AUM Advantage: Side-by-side comparison reveals what makes CAR-T unique at molecular level. Identifies CAR-specific therapeutic targets (will not affect normal T cell immunity). Explains clinical phenomena (why CAR-T exhausts in tumors but vaccine-induced T cells do not). Can extend to CAR-NK vs conventional NK cells, different CAR designs (CD28 vs 4-1BB), different target antigens (CD19 vs BCMA vs solid tumor targets).
Studying CAR Tonic Signaling Mechanisms
Approach: CAR-T cells can signal in absence of antigen ("tonic signaling") due to CAR aggregation, high scFv affinity, or self-ligands. This causes premature exhaustion. Knock down candidate tonic signaling mediators: CAR clustering proteins, signaling adaptors, transcription factors activated by tonic signaling. Measure basal phosphorylation (phospho-CD3ζ, phospho-ERK, phospho-AKT) in absence of tumor targets. Test if knockdown reduces tonic signaling-induced exhaustion while preserving antigen-specific activation. Use CAR designs with different tonic signaling levels (high affinity vs low affinity scFv) to validate findings.
AUM Advantage: Dissects tonic signaling mechanism without altering CAR structure itself. Can compare tonic signaling across CAR designs (identify which signaling molecules are design-dependent). Reveals whether tonic signaling is beneficial (promotes memory) or detrimental (causes exhaustion). Identifies druggable targets to modulate tonic signaling for optimal CAR-T function.
Tumor-CAR-T Interaction Discovery Through Functional Genomics
Approach: Target genes involved in CAR-T-tumor crosstalk: adhesion molecules (integrins, selectins, ICAMs), chemokine receptors (CXCR3, CCR7, CX3CR1), cytotoxic effectors (perforin, granzymes, FasL, TRAIL), cytokine receptors (IL-2R, IL-7R, IL-15R), and tumor-suppressive receptors (TGFβR, IL-10R, adenosine receptors ADORA2A/2B). Knock down each gene and measure CAR-T tumor infiltration (3D tumor spheroid penetration, transwell migration), tumor killing kinetics (real-time impedance, live-cell imaging), and cytokine production in tumor coculture.
AUM Advantage: Identifies genes required for CAR-T-tumor interaction without bias. Reveals differences between hematologic vs solid tumor requirements. Can study patient-derived xenograft models to identify patient-specific gene dependencies. Discovers combination targets (e.g., knockout TGFβR + enhance CXCR3 expression for better solid tumor trafficking).
miRNA Function Discovery in CAR-T Cells Using AUMantagomir
Approach: Use AUMantagomir sdASO to inhibit microRNAs and discover their roles in CAR-T biology. Target exhaustion-associated miRNAs (miR-155, miR-31, miR-146a), memory-associated miRNAs (miR-150, miR-181a), and cell cycle/proliferation miRNAs (miR-17-92 cluster). Measure effects on CAR-T differentiation, exhaustion markers, proliferation, and tumor killing. Perform mRNA-seq to identify miRNA target genes (genes upregulated upon miRNA inhibition). Build miRNA regulatory networks.
AUM Advantage: AUMantagomir enables functional miRNA studies without genetic manipulation. Reveals post-transcriptional gene regulation in CAR-T cells. Can combine miRNA inhibition with mRNA knockdown (AUMsilence) to study miRNA-target gene relationships. Identifies therapeutic miRNA targets—miRNA inhibitors are clinically viable (several in clinical trials for cancer).
Frequently Asked Questions
Ready to Enhance Your CAR-T Cells?
Discover how AUMsilence sdASO can improve CAR-T persistence, prevent exhaustion, and overcome manufacturing challenges—all without transfection. Our scientists provide expert guidance for CAR-T optimization strategies.