Review

Engineering CAR and TCR T-Cells to Overcome Resistance in Cancer Therapy

Abstract

Adoptive cell therapy (ACT) is one of the most transformative advances in modern oncology, using genetically engineered T lymphocytes to achieve targeted and long-lasting tumor elimination. Chimeric antigen receptor (CAR) and T-cell receptor (TCR) therapies have shown remarkable success in treating blood cancers, but their application to solid tumors is still limited by multiple resistance factors, including antigen diversity, immunosuppressive microenvironments, and limited T-cell persistence.

This review summarizes the mechanistic barriers and engineering innovations shaping the next generation of adoptive T-cell therapies. We compare structural and functional differences between CAR- and TCR-based systems, explore major resistance mechanisms such as antigen escape, metabolic restrictions, and T-cell exhaustion, and discuss emerging strategies like dual and logic-gated CARs, armored constructs, and TCR mimic designs. We highlight how systems biology, artificial intelligence, and advanced modeling tools are transforming receptor optimization, preclinical testing, and manufacturing scalability.

Although progress is rapid, key mechanistic questions remain about dynamic antigen evolution, cytokine control over space and time, and the long-term safety of multi-circuit constructs. Future advances will require integrating computational feedback, adaptive signaling, and modular receptor designs to create precise, self-optimizing T-cell therapies. These developments collectively mark a shift from static receptor engineering to intelligent, adaptive immune treatments capable of sustained control across diverse and resistant cancer environments.

Introduction

Adoptive cell therapy (ACT) has become a groundbreaking method in cancer immunotherapy, using ex vivo expanded and genetically modified T lymphocytes to specifically seek out and destroy cancer cells1. Unlike vaccine-based or checkpoint-targeted immunotherapies, which rely on activating existing immune responses, ACT supplies patients with pre-prepared effector T cells capable of attacking tumors even in severely immunosuppressed environments1. This approach bypasses many limitations of natural antitumor immunity, providing a direct and programmable immune intervention. Although promising, ACT still faces significant biological and translational hurdles.

A major obstacle is immune tolerance to self-derived tumor-associated antigens (TAAs), which are common targets in both solid and blood cancers. Since these TAAs are often unmutated and resemble normal tissue antigens, naturally occurring tumor-reactive T cells usually have low-affinity T cell receptors (TCRs), resulting in weak activation, limited proliferation, and low cytotoxicity1. To overcome this, synthetic receptor engineering was developed to bypass natural tolerance mechanisms and improve tumor recognition while balancing efficacy and safety. This led to the development of chimeric antigen receptors (CARs), synthetic molecules that reprogram T-cell specificity toward tumor surface antigens independently of MHC.

CARs combine the extracellular antigen-binding domain of monoclonal antibodies, usually a single-chain variable fragment (scFv), with intracellular T-cell signaling domains from CD3ζ and costimulatory molecules like CD28, 4-1BB, or OX402. This design allows T cells to recognize surface antigens directly, without needing MHC presentation, thus avoiding one of the main tumor immune evasion tactics, loss or downregulation of MHC class I molecules. Clinically, CAR-T cell therapy has transformed treatment for blood cancers. Tisagenlecleucel, the first FDA-approved CD19 CAR-T therapy, demonstrated complete remission rates of 81–90% in pediatric and young adult patients with relapsed/refractory B-cell acute lymphoblastic leukemia (ALL)3. Notable cases, such as the sustained remission of pediatric patient Emily Whitehead4, highlight the curative potential of this approach. However, these advances have also revealed the risks, including severe toxicities like cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), which show how potent immune activation can cause systemic harm4.

The success in hematologic cancers has led to exploring TCR-engineered T cells (TCR-T) as a complementary method that can target intracellular antigens presented on MHC molecules. Unlike CAR-Ts, which target surface epitopes, TCR-T therapies can access a wider range of tumor-related peptides or neoantigens5. Nevertheless, both techniques face shared issues, including antigen heterogeneity, immune escape, and an immunosuppressive tumor microenvironment (TME)2. These challenges have so far limited their effectiveness against solid tumors. Overall, these differences highlight the need for integrated receptor engineering approaches that optimize activation, specificity, and safety.

Understanding the comparative advantages and constraints of CAR-T and TCR-T systems (Table 1), alongside innovations in circuit design, metabolic programming, and safety modulation, will be essential for extending the benefits of ACT beyond hematologic cancers and into the far more complex landscape of solid tumors.

Table 1: Structural, functional, and translational features of CAR-T and TCR-T cell therapies

ABCD
FeaturesCAR T-CellsTCR T-CellsCitations
Antigen recognitionExtracellular surface antigens via scFv; MHC-independentIntracellular peptides via TCR–MHC; MHC-dependent6
Antigen repertoireLimited to surface proteins (e.g., CD19, HER2)Broad; includes intracellular and viral antigens (e.g., NY-ESO-1, KRAS)7
MHC requirementNoneRequires a patient HLA match8
Engineering complexitySynthetic receptor insertionαβ TCR modification; mispairing prevention9
Tumor coverageHematologic cancers; limited solid-tumor efficacyPotential for solid tumors; limited by antigen presentation10
ToxicitiesCRS, ICANS, off-tumor effectsCRS, ICANS, off-tumor effects11
ManufacturingAutologous standard; allogeneic emergingAutologous; limited allogeneic feasibility12
Key advantagesPotent, MHC-independent, proven efficacyAccess to intracellular targets, physiologic signaling13
Key limitationsSurface-only targeting, cytokine toxicityMHC-restriction, complex engineering14

TCR and CAR T-Cells in Adoptive Cell Therapy: Structure, Function, and Mechanisms

ACT encompasses several immune-engineering modalities, among which T-cell receptor (TCR)-engineered T cells and chimeric antigen receptor (CAR)-T cells represent the most advanced and clinically validated platforms1. While both aim to redirect T cells toward tumor antigens, they differ fundamentally in antigen recognition, signaling architecture, and translational potential. Understanding these structural and mechanistic distinctions is essential to explain their respective successes in hematologic malignancies and the persistent barriers faced in solid tumors.

TCR-Engineered T Cells: Structure and Mechanism

T-cell receptors (TCRs) are naturally occurring αβ heterodimeric proteins composed of variable (V) and constant (C) domains. Antigen recognition occurs through the complementarity-determining regions (CDRs) within the V domains, particularly CDR3, which engages peptide fragments presented by major histocompatibility complex (MHC) molecules on target cells. This interaction is central to adaptive immunity, allowing discrimination between self and non-self antigens5. Importantly, the TCR itself lacks intrinsic signaling capacity; instead, activation depends on its association with the CD3 complex, which contains ten immunoreceptor tyrosine-based activation motifs (ITAMs) responsible for downstream signaling5.

Upon binding a peptide-MHC (pepMHC) complex, the TCR initiates a multi-step activation cascade involving phosphorylation of CD3 ITAMs by Lck, recruitment of ZAP70, and subsequent activation of adapter proteins such as LAT. This culminates in transcriptional programs that drive cytokine secretion, proliferation, and cytotoxicity15. The strength of the TCR-pepMHC interaction is relatively low (Kd ~1–100 µM), yet through serial triggering and co-receptor amplification (CD4/CD8), TCRs exhibit remarkable sensitivity, capable of responding to only a few antigenic complexes per cell16. From a therapeutic perspective, TCR engineering enables recognition of intracellular tumor antigens, including mutated neoantigens and cancer-testis antigens, which are inaccessible to antibody-based receptors. This expands the range of targetable malignancies, particularly for tumors lacking unique surface markers.

However, MHC restrictions remain: while it allows exquisite specificity, it also limits the applicability of a given TCR to patients with compatible HLA alleles and renders tumors vulnerable to immune escape via MHC downregulation. Clinically, TCR-T cells have achieved encouraging responses in tumors such as synovial sarcoma and melanoma, yet their translation is constrained by on-target/off-tumor toxicities and HLA-dependent variability, underscoring the need for improved antigen selection and affinity tuning.

CAR-T Cells: Modular Design and Function

Chimeric antigen receptors (CARs) are synthetic, modular constructs that endow T cells with the ability to recognize target antigens independently of MHC presentation. Structurally, a CAR integrates several functional domains: an extracellular single-chain variable fragment (scFv) derived from an antibody, a hinge or spacer providing flexibility, a transmembrane domain ensuring receptor stability, and one or more intracellular signaling modules responsible for activation15.

The evolution of CAR design has dramatically influenced therapeutic outcomes. First-generation CARs, containing only the CD3ζ signaling domain, induced cytotoxicity but lacked robust cytokine secretion and persistence; second-generation CARs, incorporating costimulatory motifs such as CD28 or 4-1BB, achieved enhanced proliferation and survival, marking the foundation for today’s FDA-approved CAR therapies; and third-generation CARs, which combine multiple costimulatory domains (e.g., CD28 and 4-1BB), generate stronger signaling but have shown mixed clinical advantages, suggesting that excessive stimulation can predispose to exhaustion and toxicity rather than improved efficacy17.

Functionally, CAR-T cells operate as “living drugs.” Upon antigen engagement, they activate, proliferate, release proinflammatory cytokines, and exert cytolytic activity through perforin and granzyme release. Some persist as memory-like populations, contributing to long-term immune surveillance2. However, antigen density, tumor microenvironmental factors, and receptor design critically modulate this activity. High antigen expression favors potent killing but also increases the risk of CRS and ICANS, highlighting the delicate balance between potency and safety.

Mechanisms of Resistance to CAR and TCR Therapies

Engineered T-cell therapies have demonstrated remarkable efficacy in hematologic malignancies, yet they face substantial and multifactorial resistance barriers in solid and relapsed cancers18. Resistance arises through a convergence of biological processes that erode cytotoxic function, persistence, and antigen recognition fidelity, ultimately compromising therapeutic durability. Understanding these mechanisms is central to the rational design of next-generation immune engineering strategies capable of overcoming tumor evolution and immune evasion.

Up to 60% of relapses are characterized by CD19 antigen loss after CAR-T therapy19. A major axis of failure lies in antigen-related resistance, which remains one of the most fundamental obstacles to sustained clinical response20. Tumor cells can evade immune recognition through antigen loss, mutation, or transcriptional downregulation of target epitopes21. In CAR-T therapies, epitope masking and alternative splicing of canonical targets such as CD19 have been shown to drive relapse following initially successful treatment22. Similarly, in TCR-T therapies, tumor cells often downregulate MHC class I molecules or disrupt antigen processing and presentation pathways, rendering themselves invisible to TCR-mediated recognition23.

Beyond complete antigen loss, intra- and intertumoral heterogeneity allows for the coexistence of antigen-negative or low-density subclones24. These resistant subpopulations survive immune pressure and later expand, forming a reservoir for tumor relapse. Such antigenic plasticity underscores the need for multi-antigen targeting and adaptive recognition systems rather than reliance on single epitope specificity24. Resistance is further compounded by the immunosuppressive TME, which represents one of the most formidable barriers to effective ACT. Structural and metabolic constraints within solid tumors, including dense extracellular matrix, aberrant vasculature, and hypoxic gradients, physically restrict T-cell infiltration25. At the same time, soluble factors such as TGF-β, IL-10, and VEGF, alongside regulatory immune cells including Tregs, myeloid-derived suppressor cells, and tumor-associated macrophages, collectively reprogram infiltrating T cells toward an exhausted or anergic phenotype. These suppressive interactions diminish cytokine secretion, proliferation, and cytolytic function, creating an immunological “trap” that blunts effector activity even when tumor cells are recognized.

A related but distinct mechanism of dysfunction involves cell-intrinsic exhaustion, a state of progressive dysfunction induced by chronic antigen stimulation and sustained signaling through CAR or TCR constructs26. Prolonged activation triggers epigenetic and transcriptional reprogramming, locking T cells into a terminally exhausted state characterized by high expression of inhibitory receptors such as PD-1, TIM-3, and LAG-3, diminished effector cytokine production, and poor recall responses upon re-encountering tumor antigen. The TME further amplifies this exhaustion through metabolic competition for glucose and amino acids, oxidative stress, and exposure to immunoregulatory metabolites like adenosine and kynurenine. Together, these pressures create a self-reinforcing feedback loop that erodes T-cell persistence and long-term functionality. In addition to immune evasion and exhaustion, deficient trafficking and limited persistence remain key practical barriers to therapeutic success27. Engineered T cells frequently fail to home efficiently to tumor sites, particularly in solid tumors that lack the chemokine gradients required for guided migration. Even when T cells reach the tumor, nutrient deprivation, hypoxia, and oxidative stress compromise their survival, while macrophage-mediated clearance and host immune rejection further limit persistence25.

These findings highlight the importance of engineering strategies that enhance chemokine receptor compatibility, metabolic fitness, and stress resistance to sustain antitumor function in the hostile tumor niche. Finally, manufacturing and scalability limitations introduce a less visible but clinically significant layer of heterogeneity. Autologous CAR- and TCR-T cell products vary widely in transduction efficiency, differentiation state, and metabolic profile, leading to inconsistent expansion and potency across patients28. Such manufacturing variability complicates both clinical outcomes and mechanistic interpretation, as differences in cell composition or exhaustion state can obscure true therapeutic performance.

Current engineering solutions tend to focus on isolated resistance mechanisms, for instance, developing dual-antigen CARs to counter immune escape or armored CARs to resist TGF-β suppression, without fully addressing the dynamic and interdependent feedback loops that tumors exploit to evade immune pressure.  The next frontier in CAR and TCR engineering should therefore embrace systems-level, adaptive designs capable of sensing and responding to evolving tumor signals. Integrative strategies combining antigen multiplexing, metabolic resilience, and programmable regulatory circuits could enable engineered cells to dynamically adjust their behavior in hostile environments. A shift toward such holistic, adaptive frameworks, rather than single-variable modifications, will be essential to achieve durable remissions across genetically heterogeneous and evolutionarily adaptable cancer types.

Engineering Strategies to Overcome Antigen Escape

Antigen escape remains one of the most frequent and devastating mechanisms of relapse following CAR or TCR T-cell therapy29. Quantitatively, CD19 antigen loss accounts for 40–60% of post-CAR-T relapses, underscoring its clinical relevance as a dominant resistance mechanism30. Loss or mutation of the target epitope, antigen downregulation, and heterogeneous expression within the tumor mass all conspire to reduce recognition and killing efficacy22,29. Modern synthetic biology approaches are now enabling the design of multifunctional, logic-gated, and adaptive immune receptors that respond to this complexity with enhanced precision and resilience. These strategies (Table 2) aim to extend the durability and flexibility of engineered T cells beyond the static, single-target paradigm of early CAR constructs.

Table 2: Functional CAR Architectures and Engineering Strategies for Overcoming Tumor Resistance and Enhancing Safety

AbCDE
CAR TypeCore Engineering PrincipleBiological Goal / Targeted LimitationKey Drawbacks or RisksCitations
Tandem (bi-scFv) CARsTwo scFvs in a single receptor chainSimultaneous engagement of two antigens with one receptorSteric hindrance, variable binding affinity31
SynNotch CARsSynthetic Notch receptor triggers the expression of a secondary CAR after detecting Antigen ABoolean “A AND B” logic to enhance tumor selectivityRequires dual-antigen co-expression; complex circuit tuning32
Dual CARsTwo separate CAR constructs expressed in the same T cellExpands recognition spectrum; mitigates antigen escapeSignal imbalance; greater metabolic stress33
Armored CARsIncorporate genes for cytokines (e.g., IL-12, IL-15, IL-18) or costimulatory ligandsReinforce T-cell activation and remodel the immunosuppressive TMEIL-12 gives strong cytotoxicity but systemic toxicity; IL-18 is safer but weaker8, 34
iCasp9 / safety-switch CARsInducible suicide system or reversible small-molecule control (e.g., dasatinib)Rapid termination of severe CRS/ICANS or uncontrolled activationAdds complexity; cannot prevent early cytokine spike35
iCARs (inhibitory CARs)Contain inhibitory domains (PD-1, CTLA-4) triggered by off-target antigen“A NOT B” logic to prevent normal-tissue attackPrecise inhibitory thresholds are hard to calibrate36

Multi-targeting and Logic-Gated CARs

Conventional CAR-T cells are limited by their reliance on a single antigen target, making them vulnerable to clonal escape when that antigen is lost or mutated6. To counter this, multi-targeting and logic-gated CAR architectures such as tandem CARs (TanCARs), DualCARs, and SynNotch systems have been developed to enable combinatorial antigen recognition31,32,33. TanCARs link two single-chain variable fragments (scFvs) within one receptor, allowing simultaneous binding to two antigens, which enhances recognition breadth and reduces relapse probability33. DualCAR designs employ separate CARs expressed within the same T cell, each targeting distinct antigens to create redundant activation pathways33. Antigen heterogeneity is the principal cause of relapse in solid and hematologic malignancies. Dual and TanCAR designs, which encode two or more scFv binders, reduce immune escape but introduce new structural and signaling complexities. Dual-CAR systems enhance the breadth of recognition yet risk tonic signaling and exhaustion when both receptors are co-expressed at high density33. In contrast, TanCARs, which integrate two scFvs within one receptor chain, improve immune synapse stability when both targets are co-expressed but lose potency if one antigen is absent. Recent trispecific and “CAR pool” approaches further extend valency, but their translation is impeded by increased vector size, steric interference, and compounding on-target/off-tumor risks. Universal or switchable CARs, using antibody–CAR bridges (e.g., CD16-based, SpyTag-SpyCatcher, or small-molecule adaptors), offer a modular solution, one cell product for multiple diseases, but hinge on the pharmacologic behavior and immunogenicity of the adaptor molecule. Critically, universal CARs may offer dose-dependent tunability, yet long-term persistence without ligand engagement remains unpredictable, and manufacturing complexity remains a barrier to scalability37.

SynNotch systems, by contrast, introduce Boolean logic into immune signaling: the recognition of one antigen through a synthetic Notch receptor induces the expression of a second CAR targeting another antigen32, 38. Traditional CARs operate on a binary “on–off” paradigm, where antigen engagement directly triggers activation. However, this model lacks contextual sensitivity and often results in off-tumor toxicity. Logic-gated CARs, including split-signal AND, NOT, and synNotch IF/THEN systems, represent the next conceptual leap. Split-signal AND-CARs distribute CD3ζ and co-stimulatory domains across separate receptors, requiring two antigens for full activation, minimizing false-positive engagement.

Yet, their stringency may compromise efficacy in tumors with heterogeneous antigen expression, increasing the risk of escape. The synNotch architecture adds conditional control: recognition of one antigen activates a transcriptional program that induces a second CAR specific for another antigen, functioning as an “IF/THEN” circuit. This approach localizes cytotoxicity to antigen-rich regions, mitigating systemic toxicity. Nevertheless, preclinical data reveal leakage of transcriptional activity and incomplete silencing in healthy tissues, suggesting the need for improved temporal precision and humanized synNotch scaffolds to prevent immunogenicity. The co-LOCKR system further extends this concept by combining AND/OR/NOT logic within modular protein switches, though translational feasibility remains limited by the non-human protein components and complex pharmacokinetics32,38. This logic-gated approach offers conditional activation that minimizes off-tumor toxicity and enhances tumor specificity. However, despite these advances, most existing platforms remain static in their antigen recognition capacity. Tumor antigen profiles evolve dynamically under immune pressure, yet current CAR configurations cannot adapt to those shifts. This gap underscores the need for adaptive receptor systems capable of reprogramming in situ without full re-engineering or reinfusion.

Future directions include developing modular CAR libraries that allow real-time exchange or tuning of scFv domains through switchable docking modules or universal adaptor scaffolds. Adjusting scFv affinity can selectively spare normal tissues expressing low antigen levels, while extended hinge domains and modified CD3ε motifs dampen cytokine release without compromising cytotoxicity. Such “mechanical tuning” of CAR conformation represents an underappreciated but powerful layer of control, one that, when integrated with transcriptional and pharmacologic systems, may achieve multi-tier safety without sacrificing potency. These could enable clinicians to “retarget” existing CAR-T populations as new resistance patterns emerge. Integration of AI-guided epitope prediction could further refine antigen selection by identifying evolutionarily stable, lineage-restricted neoantigens less prone to immune escape. Moreover, CRISPR-based screening platforms can be employed to perform high-throughput testing of antigen-pair combinations to determine optimal co-targeting strategies for individual tumor types39. Collectively, these approaches aim to transform CAR-T therapy from a static therapeutic into a dynamic, evolving immunologic system capable of maintaining surveillance against tumor plasticity.

TCR Engineering to Broaden Recognition

While CAR-T cells are confined to surface-expressed antigens, TCR-engineered T cells extend immune recognition into the intracellular proteome by detecting peptide fragments presented via MHC molecules7. This property allows access to previously “undruggable” intracellular oncoproteins, thereby broadening the therapeutic landscape well beyond surface antigen availability. However, despite this conceptual advantage, TCR-based therapies continue to face significant resistance barriers, including MHC downregulation, defective antigen processing, and cross-reactive alloreactivity, all of which can reduce specificity or lead to severe off-target toxicity40. To address these limitations, several engineering innovations have been explored. Affinity-tuned TCRs aim to strengthen recognition of tumor-specific peptides while preserving tolerance to self-antigens, minimizing autoimmune risk41. Meanwhile, TCR-mimic CARs (TCRm-CARs) have emerged as hybrid constructs that merge the MHC-dependent specificity of TCRs with the robust signaling of CARs, allowing antibody-derived scFv domains to recognize peptide–MHC complexes directly42. These hybrids may represent a practical bridge between traditional TCR and CAR modalities, potentially achieving intracellular antigen targeting with CAR-like kinetics and amplification. In parallel, data-driven and AI-assisted receptor discovery pipelines are rapidly advancing. Deep learning–trained TCR libraries can predict antigen–HLA binding affinities and potential cross-reactivity profiles, accelerating the discovery of safe, high-avidity receptors capable of maintaining recognition across mutating tumor variants.

Yet, despite this progress, key translational challenges persist. Tumor-induced MHC downregulation and disruptions in antigen processing continue to limit the presentation of intracellular peptides, while the high diversity of HLA alleles among human populations hinders universal application43. Therefore, the future of TCR engineering may depend on integrating synthetic biology principles to reprogram signaling modules for partial or complete MHC independence. This shift would blur the functional boundary between CAR and TCR systems, giving rise to hybrid or context-dependent receptors capable of sustaining recognition even in antigenically unstable tumors. Ultimately, the integration of patient-specific immunopeptidome profiling with AI-driven antigen selection may lead to personalized, broad-spectrum T cells that retain adaptability and precision in real time. Together, these emerging strategies signal a paradigm transition, from rigid, single-target constructs to adaptive, evolution-aware immune platforms capable of co-evolving with the tumor ecosystem.

Engineering for Tumor Microenvironment (TME) Resistance

TME forms a major axis of resistance against adoptive T-cell therapies, imposing physical, biochemical, and metabolic constraints that blunt immune activation44. Within this hostile ecosystem, suppressive cytokines, nutrient deprivation, hypoxia, and inhibitory checkpoints converge to induce T-cell dysfunction and exhaustion<26. Overcoming these layered inhibitory signals requires not only persistence against suppression but also the capacity to actively remodel and reprogram the TME. Recent bioengineering advances have led to the development of T cells that secrete immunostimulatory factors, rewire their metabolic machinery, or operate through synthetic circuits capable of sensing and adapting to local environmental cues.

Armored CAR/TCR Cells

CAR-T failure in solid tumors is largely due to metabolic, immunosuppressive, and physical barriers within the TME45. Armored CARs, engineered to secrete cytokines or express resistance modules, represent one of the most intensively studied yet clinically underperforming solutions. Constructs secreting IL-12 or IL-18 enhance macrophage activation and antigen spreading but frequently trigger systemic inflammation, reflecting the narrow therapeutic window of constitutive cytokine expression. Conditional systems, such as NFAT-inducible cytokine expression, reduce baseline toxicity but remain susceptible to “leakiness” and unpredictable kinetics.

Metabolic reprogramming through overexpression of arginine-synthetic enzymes (ASS1, OTC) or resistance to inhibitory cytokines (dominant-negative TGFβRII, A2A receptor knockdown) has shown enhanced persistence in murine models, but human data remain sparse, and the pleiotropic effects of TGFβ or adenosine signaling complicate their safety assessment45. Overall, while armored CARs demonstrate mechanistic rationale, their clinical translation demands more precise spatial and temporal regulation, a role potentially filled by logic-gated or sensor-integrated constructs.

One of the most direct approaches to counteract the immunosuppressive milieu involves engineering “armored” CAR and TCR T cells capable of secreting cytokines or checkpoint-blocking agents directly within the tumor. Cells engineered to release IL-12, IL-15, IL-18, or PD-L1-blocking single-chain antibodies (scFvs) locally can enhance cytotoxicity, recruit endogenous immune effectors, and reshape the TME toward a proinflammatory phenotype34. IL-12–secreting CAR-T cells, for instance, have demonstrated enhanced persistence and resistance to Tregs, while IL-15 promotes memory-like phenotypes and sustains metabolic fitness. Local secretion of checkpoint inhibitors also allows autonomous blockade of PD-1/PD-L1 signaling, avoiding the need for systemic antibody administration34.

However, a persistent gap remains in the spatiotemporal regulation of these immune mediators. Constitutive cytokine expression can trigger systemic inflammation and off-target toxicity, limiting translational feasibility8. Future engineering directions therefore emphasize inducible cytokine expression systems that respond to drug cues or microenvironmental conditions such as hypoxia or high PD-L1 expression. For example, drug-responsive promoters can allow transient activation under physician control, while synthetic gene circuits incorporating AND/NOT logic gates could restrict IL-12 or IL-18 release exclusively to regions exhibiting both immunosuppressive and hypoxic signals. These programmable systems aim to provide localized immunostimulation with minimal systemic burden, transforming T cells into precision-controlled immunologic actuators.

Metabolic and Epigenetic Reprogramming

T cells entering solid tumors encounter severe metabolic stress characterized by glucose and amino acid depletion, high lactate levels, and oxygen scarcity, all of which impair effector function and survival9. Engineering metabolically resilient T cells has thus become a critical focus in overcoming TME-induced dysfunction. Overexpression of peroxisome proliferator–activated receptor gamma coactivator 1-alpha (PGC1α) enhances mitochondrial biogenesis and oxidative phosphorylation, improving persistence under nutrient limitation10. Similarly, HIF-2α overexpression promotes adaptation to hypoxia and sustains cytotoxic activity in oxygen-deprived niches11. Beyond metabolic rewiring, epigenetic engineering offers a means to prevent terminal exhaustion by resetting dysfunctional chromatin landscapes. Targeted editing using dCas9-fused epigenetic modifiers, such as dCas9-TET1 (for demethylation) or dCas9-KRAB (for gene repression), can modulate exhaustion-associated loci and restore transcriptional flexibility.

Yet despite these promising directions, a major knowledge gap persists: there are no robust predictive models linking metabolic or epigenetic modifications to long-term antitumor outcomes. The complexity of metabolic, epigenetic crosstalk and interpatient variability complicate rational design. Moving forward, integration of computational metabolic modeling and multi-omics longitudinal profiling is essential to forecast how engineered pathways influence persistence, proliferation, and exhaustion trajectories in vivo. Such insights could guide the development of rationally tuned metabolic circuits that adapt dynamically to TME stressors while maintaining controlled activation states.

TME-Responsive Sensors

An emerging frontier in T-cell engineering involves embedding synthetic biosensors capable of detecting and responding to tumor-specific molecular cues, thereby allowing conditional activation or adaptive behavior12,13. These biosensors can monitor environmental signals such as reactive oxygen species (ROS), lactate, hypoxia, or immunosuppressive cytokines like TGF-β, and trigger predefined transcriptional responses such as cytokine release, costimulatory activation, or self-regulation13. For example, hypoxia-sensitive CARs activate signaling only in low-oxygen environments, minimizing off-tumor cytotoxicity in healthy tissues.

Real-time sensing CAR-T platforms integrated with microelectronic feedback systems or reporter circuits are needed to enable continuous monitoring and adaptive dosing. Such feedback-controlled designs could modulate therapeutic intensity in response to changing TME conditions, functioning as closed-loop immunotherapy systems. Furthermore, microfluidic tumor-on-a-chip technologies can be employed as preclinical testing platforms to simulate tumor architecture, gradient dynamics, and cellular interactions, allowing the refinement of conditional circuits before advancing to animal or human studies46. These technologies represent a critical translational bridge between synthetic immunology and clinical oncology, enabling safe, programmable, and context-aware immune therapeutics.

Overcoming Trafficking and Persistence Barriers

Effective tumor eradication requires engineered T cells not only to recognize and attack malignant cells but also to efficiently traffic to, infiltrate, and persist within the tumor microenvironment. Many adoptive cell therapies fail due to insufficient homing, limited tissue penetration, or premature exhaustion following infiltration27. Solid tumors often exclude immune cells through disorganized vasculature, dense extracellular matrices, and mismatched chemokine signaling20,21,26. In addition, chronic antigen exposure, metabolic deprivation, and inhibitory checkpoint signaling compromise the longevity of infused cells. Therefore, recent engineering strategies have focused on optimizing both spatial navigation and long-term persistence within the hostile TME.

Chemokine receptor engineering represents one of the most direct approaches to improve tumor-directed migration. Many solid tumors secrete chemokines such as CXCL1, CCL5, and CX3CL1, which are not efficiently recognized by unmodified T-cells14. To exploit these chemotactic gradients, CAR and TCR T cells can be engineered to express corresponding receptors such as CXCR2, CCR5, or CX3CR1, aligning their migratory profiles with the tumor’s chemokine landscape14,47. For example, CXCR2 expression enhances trafficking toward melanoma and ovarian carcinoma, while CCR5 facilitates migration to CCL5-rich environments characteristic of certain breast and pancreatic cancers48,49. This receptor reprogramming allows effector cells to localize more effectively within tumor cores where immunosuppression is most profound.

Once T cells arrive at the tumor site, the extracellular matrix (ECM) imposes a major physical barrier to effective infiltration. The ECM, enriched in collagen, proteoglycans, and fibronectin, restricts T-cell motility and limits cytotoxic interactions with tumor cells50. To overcome this obstacle, engineered lymphocytes have been equipped with degradative enzymes such as heparanase or matrix metalloproteinases (MMPs) to facilitate ECM remodeling and deeper penetration. For instance, heparanase-expressing CAR T-cells demonstrate improved tumor infiltration and clearance in preclinical models50. However, maintaining a delicate balance between matrix degradation and stromal integrity remains crucial, as excessive proteolysis could damage healthy tissues or promote metastasis.

Beyond physical infiltration, sustained T-cell persistence is essential for durable tumor control. Genetic modulation of exhaustion pathways and metabolic reprogramming can reinforce cell survival and effector function. Incorporation of cytokine support systems, such as IL-15 or IL-7 transgenes, enhances memory differentiation and long-term activity, while silencing inhibitory receptors like PD-1 or transcription factors such as TOX mitigates exhaustion. Additionally, metabolic enhancement strategies, such as overexpressing PGC1α to boost mitochondrial biogenesis, improve energy utilization and resilience within nutrient-depleted tumor niches. Together, these approaches promote functional persistence and prevent premature T-cell attrition in the TME.

Despite these promising advances, a key limitation persists: existing preclinical models often fail to replicate the spatial and molecular heterogeneity of human tumors. Conventional two-dimensional cultures oversimplify stromal architecture and do not capture the dynamic gradients that govern chemokine signaling and matrix density51. Moreover, murine models differ from human tumors in vascular organization, stromal stiffness, and chemokine repertoire, limiting translational predictability. Future directions point toward integrating three-dimensional bioprinted tumor constructs that incorporate stromal, vascular, and immune compartments, allowing for high-fidelity evaluation of T-cell infiltration and retention under physiologically relevant conditions. Complementarily, computational agent-based and multiscale models can simulate CAR/TCR cell migration, antigen encounters, and cytotoxic dynamics within patient-specific tumor geometries. These digital reconstructions, or “immune digital twins”, could guide personalized receptor designs and dosing regimens that maximize infiltration while minimizing off-target effects52. By integrating experimental and computational modeling, next-generation adoptive T-cell therapies can be rationally engineered not only for antigen recognition and signal potency but also for efficient spatial navigation and durable persistence within the complex architecture of solid tumors.

Engineering for Controlled Activation and Safety

Achieving a precise balance between potent antitumor efficacy and controlled immune activation remains one of the most formidable challenges in adoptive T-cell therapy. Conventional CAR and TCR architectures are largely governed by binary activation logic; once antigen recognition occurs, the signaling cascade proceeds irreversibly35. While this design has driven remarkable success in hematologic malignancies, it presents considerable safety risks in solid tumors, where heterogeneous antigen expression increases the likelihood of off-tumor reactivity and CRS. The clinical burden of immune-related adverse events, particularly CRS and ICANS, underscores the urgent need for tunable and reversible activation mechanisms42.

To address these limitations, several molecular safety mechanisms have been developed to allow precise pharmacological control of T-cell activity. The inducible caspase-9 (iCasp9) suicide switch remains the most clinically validated, providing a rapid, small-molecule-triggered apoptotic shutdown of infused cells during severe toxicity53. Other strategies, including Tet-inducible CAR systems and protease-cleavable constructs, enable temporal modulation or selective deactivation of CAR function. However, the persistence of resistant subpopulations, variability in transgene expression, and increased vector complexity highlight the need for simplified yet robust safety architectures53.

Despite these innovations, preclinical safety evaluation remains constrained by the poor predictive power of murine and xenograft models, which fail to capture the complexity of human immune and vascular responses. Consequently, cytokine storms and neurotoxic events often emerge only during clinical trials, after preclinical safety benchmarks were met. Future progress depends on humanized preclinical platforms capable of more accurately reflecting systemic immune dynamics. Immune–organ-on-chip systems, integrating human endothelial, neural, and immune cell components, offer a promising solution by reproducing organ-specific phenomena such as blood–brain barrier permeability and pulmonary inflammation46. These models could substantially improve prediction and mitigation of ICANS and CRS before clinical application. At the molecular design level, logic-gated inhibitory CARs (iCARs) represent a paradigm shift toward conditional activation. By applying Boolean logic principles, such as “AND,” “OR,” and “NOT” gates, engineered receptors can selectively activate or suppress cytotoxicity depending on multi-antigen recognition patterns36. For instance, a CAR may trigger full activation only in the presence of a tumor-specific antigen while being inhibited by the recognition of a self-antigen, creating a “therapeutic safe zone” that prevents off-target damage36.

Beyond biochemical circuits, optogenetic CAR platforms enable precise spatiotemporal control of immune activation using light-sensitive signaling domains54,55. These systems can reversibly switch CAR activity on or off through external light stimuli, providing clinicians with the capacity to fine-tune immune engagement within defined anatomical regions. Although currently preclinical, optogenetic control holds the potential to inaugurate a new era of on-demand immunotherapy, allowing real-time modulation of immune responses within delicate or inaccessible tissues.

In summary, next-generation CAR and TCR engineering must evolve from static to adaptive and predictive frameworks that integrate systems biology, bioengineering, and computational modeling. Combining mechanistic modeling, organ-on-chip validation, and dynamic control circuitry will enable not only safer but also smarter immunotherapies, capable of responding autonomously to tumor microenvironmental cues and patient-specific variables.

Modeling, Manufacturing, and AI Integration

As CAR and TCR T-cell therapies advance from experimental success to clinical reality, the next transformative leap will arise from the convergence of experimental modeling, computational prediction, and scalable manufacturing. This integration marks the beginning of a new engineering paradigm in cellular immunotherapy, one that relies not only on molecular innovation but also on data-driven design, automation, and precision modeling of human immune dynamics.

While substantial progress has been achieved in receptor optimization, safety control, and enhancement of functional persistence, these advances remain constrained by a fragmented research ecosystem. Preclinical models, computational frameworks, and manufacturing processes continue to evolve independently, limiting reproducibility and translational predictability17. The next frontier demands the unification of these once-isolated domains into a cohesive, closed-loop system that enables predictive, reproducible, and patient-specific cellular therapy design.

Advanced Experimental Models

Traditional murine and xenograft systems, though indispensable for initial validation, inadequately replicate the complexity of the human tumor–immune interface56,57. Recent innovations seek to overcome these limitations through models that more faithfully emulate human immunobiology. Humanized mouse platforms reconstituted with complete hematopoietic and myeloid lineages now enable more accurate evaluation of T-cell persistence, exhaustion trajectories, and toxicity under a human-like immune context. Complementary to these in vivo systems, perfused microfluidic tumor-on-chip technologies provide dynamic representations of tumor physiology, incorporating vascular flow, oxygen and nutrient gradients, stromal barriers, and cytokine-driven immunosuppression46. These microphysiological systems allow real-time imaging of T-cell infiltration, killing efficiency, and immune evasion within controlled, physiologically relevant environments.

In parallel, long-term co-culture systems combining engineered T cells with tumor organoids and stromal components are emerging as valuable tools for investigating chronic activation, exhaustion kinetics, and the formation of durable memory subsets58. Nevertheless, no existing platform effectively captures the temporal coevolution between tumors and immune effectors. Processes such as tumor immunoediting, antigen escape, and metabolic adaptation remain poorly represented in static or short-term models59. Future efforts should therefore aim to develop integrative, longitudinal frameworks that combine humanized in vivo systems with in vitro microfluidic co-cultures. In effect, these models would evolve from validation tools into predictive simulators of immune adaptation, allowing experimental data to guide next-generation receptor designs with improved translational reliability.

Computational and Systems-Biology Modeling

The synthesis of computational modeling with experimental biology is redefining the conceptual foundation of adoptive cell therapy. Multi-scale systems models now link receptor-level signaling events to cellular decision-making and, ultimately, to tumor-ecosystem behavior, offering a quantitative bridge between molecular design and clinical outcome60. These mechanistic frameworks allow researchers to simulate how variations in co-stimulatory domains, ligand affinity, or signaling thresholds shape cytotoxicity, persistence, and exhaustion across heterogeneous tumor architectures.

Artificial intelligence (AI) adds a powerful predictive dimension by enabling rapid in-silico design of receptor structures using large sequence–function datasets61. Deep learning models trained on experimental libraries can forecast CAR/TCR configurations that optimize affinity, specificity, and safety, compressing years of experimental iteration into computational hours. Parallel developments in digital twins, which are virtual patient avatars constructed from integrated clinical, genomic, and immunologic data, provide a means to simulate personalized therapy outcomes62. By modeling how an individual’s tumor microenvironment and immune repertoire interact with a proposed CAR or TCR design, digital twins could predict both efficacy and toxicity before clinical infusion.

Yet, a gap is the lack of standardized, large-scale datasets that correlate in vitro and in silico predictions with in vivo outcomes. Furthermore, most modeling efforts exist in isolation, either mechanistic or AI-based, without feedback integration. The next stage requires hybrid modeling frameworks that unite mechanistic pathway simulations with machine-learning prediction engines, allowing adaptive refinement as new experimental data emerge. Standardized ontologies, interoperable databases, and cross-platform data-sharing agreements will be essential to close the current divide between computational predictions and biological validation. Ultimately, this fusion will enable a self-improving system in which experimental feedback continuously enhances model accuracy, advancing predictive immunotherapy design from theoretical potential to clinical reliability.

Manufacturing and Scalability

Even the most advanced receptor designs depend on efficient, reliable, and economically viable manufacturing systems to achieve clinical translation. The vein-to-vein manufacturing time for autologous CAR-T products can extend up to 30 days, often exceeding the clinical stability window for patients with rapidly advancing disease63. Conventional production pipelines, dominated by viral transduction, manual handling, and batch variability, remain significant bottlenecks. To overcome these limitations, automated closed-loop biomanufacturing platforms are being developed that integrate real-time monitoring of cell viability, phenotype, and metabolic state under good manufacturing practice (GMP) conditions64. Such automation reduces human error, enhances reproducibility, and ensures consistency across production batches64.

Simultaneously, non-viral gene-delivery technologies, including mRNA electroporation, Sleeping Beauty, and PiggyBac transposon systems, are transforming genetic engineering by minimizing insertional mutagenesis risk, shortening production time, and reducing cost65,66. These technologies allow both transient and stable transgene expression without the biosafety constraints of viral vectors. In addition, CRISPR-mediated editing of MHC molecules is enabling the creation of universal “off-the-shelf” CAR and TCR platforms, bypassing donor-specific limitations and supporting scalable allogeneic therapy production67.

Despite these promising advances, manufacturing pipelines still lack predictive tools capable of assessing product potency, persistence, and exhaustion risk before infusion. Current quality-control assays rely on static surface or cytokine markers that often fail to forecast in vivo performance. Future development should focus on integrating multi-omics analytics and machine-learning algorithms into real-time process monitoring. By correlating metabolic flux, transcriptomic signatures, and bioreactor parameters, AI-driven manufacturing frameworks could autonomously adjust culture conditions to sustain optimal T-cell functionality. Such adaptive manufacturing would not only standardize product potency but also enable sustainable, cost-efficient production across global therapeutic networks.

To synthesize these complex developments, Table 3 summarizes major domains of ongoing innovation in CAR and TCR engineering, highlighting their current limitations and outlining future directions that could enhance translational efficacy.

Table 3: Summary of innovation domains, key gaps, and future directions in CAR and TCR cell engineering

ABCD
DomainInnovationKey GapFuture Direction
Experimental ModelsHumanized mice, microfluidic tumor chips, organoid co-culturesLack of dynamic tumor–immune coevolutionIntegrate longitudinal multi-omics and metabolic profiling
Computational ModelingMulti-scale & AI-driven prediction systemsWeak correlation with in vivo outcomes; poor data standardizationHybrid mechanistic + AI frameworks with interoperable datasets
ManufacturingAutomated closed-loop platforms, non-viral systems, universal chassisNo predictive potency/exhaustion analyticsAI-guided bioprocess monitoring and adaptive production control

Future Perspectives

Despite the remarkable advances achieved in receptor design, signaling optimization, and manufacturing, current engineering strategies in adoptive cell therapy remain largely static and compartmentalized, addressing isolated challenges such as antigen escape or cytokine regulation without integrating their interdependent biological consequences9,22. Tumors, in contrast, are dynamic ecosystems that constantly evolve through antigenic drift, metabolic reprogramming, and immunosuppressive remodeling68. Bridging this asymmetry requires a paradigm shift: engineered T-cells transition from fixed constructs into adaptive, self-optimizing systems capable of sensing, interpreting, and responding to tumor evolution in real time.

A closed-loop engineering framework embodies this next stage. In such a system, high-dimensional clinical, genomic, and immunologic data continuously inform computational models that predict optimal receptor configurations and activation circuits. These designs are iteratively tested in advanced preclinical models, humanized mice, tumor-on-chip systems, and organoid co-cultures, and the resulting biological feedback refines future receptor generations. Over successive design cycles, this feedback loop could produce T cells with increasing specificity, persistence, metabolic resilience, and safety, ultimately achieving the conceptual goal of a self-learning cellular therapeutic. Technological convergence will be the key enabler of this transformation. Synthetic biology contributes modular logic circuits and inducible effector programs, multi-omics profiling reveals tumor vulnerabilities and immune adaptation pathways, and artificial intelligence accelerates receptor optimization and predictive modeling of therapy outcomes61,69. Together, these domains can turn T cells into context-aware biological devices, capable of conditional activation, controlled cytokine release, and autonomous modulation of their own exhaustion or metabolic states.

At the same time, next-generation receptor engineering must expand beyond single-target approaches. Platforms incorporating dual or tandem CARs, logic-gated synNotch systems, and mechanically tuned hinge domains should enable discrimination between malignant and healthy tissues while mitigating off-tumor toxicity. Integration of metabolic reprogramming modules, such as enhanced mitochondrial fitness or resistance to adenosine and TGF-β signaling, could further support T-cell survival within hostile tumor microenvironments31,32,33,70. These multilayered strategies represent a shift from designing “stronger” CARs toward constructing intelligent, adaptable immune agents that adjust activation thresholds according to antigen density, microenvironmental cues, and immune feedback. Yet, the path to translation remains constrained by scalability, regulatory complexity, and unpredictable in vivo behavior of multi-circuit systems. To ensure reproducibility and patient accessibility, manufacturing must evolve in parallel. Automated, closed-loop bioreactors, non-viral gene-delivery platforms, and CRISPR-based universal chassis can collectively shorten production time, reduce cost, and standardize product potency. Embedding real-time analytics, combining transcriptomic, metabolic, and phenotypic data, will allow AI-guided quality control capable of predicting persistence and exhaustion risk before infusion.

Ultimately, the field is moving from unidimensional receptor enhancement toward multi-layered, programmable immunotherapy. Overcoming resistance will not depend solely on inventing additional receptor formats but on rationally integrating existing modules, pairing antigen multiplexing with metabolic resistance, safety switches, and controlled apoptosis, to achieve reproducible, safe, and durable responses within the heterogeneous landscape of solid tumors.

Table 4 summarizes the short- and long-term outlook for CAR and TCR T-cell engineering, highlighting the evolving priorities across experimental modeling, computational design, manufacturing, and translational integration.

Table 4: A summary of the short- and long-term outlook for CAR/TCR T-cell engineering and translation

DimensionShort-Term OutlookLong-Term Outlook
Experimental ModelsExpansion of humanized mice, tumor-on-chip, and organoid co-cultures to validate in-silico predictionsIntegration of longitudinal, multi-omics, and metabolic profiling to capture tumor–immune coevolution.
Computational and AI ModelingEarly use of AI for receptor optimization; limited by small, isolated datasets.Hybrid mechanistic + AI frameworks with feedback loops and digital twin simulations enabling patient-specific therapy design.
Manufacturing and ScalabilityAdoption of automated closed-loop bioreactors, non-viral gene-transfer, and early real-time QC tools.Fully autonomous, AI-driven manufacturing with predictive potency and exhaustion analytics, enabling global scalability.
Translational IntegrationImproved reproducibility and standardized GMP pipelines.Self-learning, adaptive systems combining computational modeling, manufacturing, and clinical feedback.

Conclusion

In conclusion, the future of CAR- and TCR-based therapies lies in intelligence, adaptability, and integration. By embracing closed-loop, data-driven engineering, adoptive cell therapy can progress from static molecular design to living, responsive, and self-optimizing immunologic systems, capable of anticipating tumor evolution, sustaining durable remission, and redefining precision oncology.

Ethical Considerations

This manuscript is a literature-based review and does not involve any new research with human participants or animals.

Conflict of Interest

The authors declare that there are no conflicts of interest relevant to the content of this manuscript.

License

This article is published under the terms of the Creative Commons Attribution 4.0 International License (CC BY 4.0).

© Mane Tavadyan, 2025. This license permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

References

  1. Feins S, Kong W, Williams EF, Milone MC, Fraietta JA. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol. 2019 Feb 18;94(Suppl 1):S3–S9.

  2. Mitra A, Barua A, Huang L, Ganguly S, Feng Q, He B. From bench to bedside: the history and progress of CAR T cell therapy. Front Immunol. 2023 May 15;14:1188049

  3. Schuster SJ, Svoboda J, Chong EA, et al. Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. N Engl J Med. 2017 Dec 28;377(26):2545-54.

  4. Bouzianas D, Bouziana S. First pediatric B-acute lymphoblastic leukemia patient treated with anti-CD19 chimeric antigen receptor T-cell therapy: Long-term remission or early cure? Hum Vaccin Immunother. 2024 Dec 31;20(1):2321678.

  5. Mariuzza RA, Agnihotri P, Orban J. The structural basis of T-cell receptor (TCR) activation: An enduring enigma. J Biol Chem. 2020 Jan 24;295(4):914-925.

  6. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021 Apr 6;11(4):69.

  7. Sanomachi T, Katsuya Y, Nakatsura T, Koyama T. Next-Generation CAR-T and TCR-T Cell Therapies for Solid Tumors: Innovations, Challenges, and Global Development Trends. Cancers (Basel). 2025 Jun 11;17(12):1945.

  8. Nie J, Zhou L, Tian W, et al. Deep insight into cytokine storm: from pathogenesis to treatment. Signal Transduct Target Ther. 2025 Apr 16;10(1):112.

  9. Rangel Rivera GO, Knochelmann HM, Dwyer CJ, et al. Fundamentals of T Cell Metabolism and Strategies to Enhance Cancer Immunotherapy. Front Immunol. 2021 Mar 18;12:645242.

  10. D’Errico I, Salvatore L, Murzilli S, et al. Peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC1alpha) is a metabolic regulator of intestinal epithelial cell fate. Proc Natl Acad Sci U S A. 2011 Apr 19;108(16):6603-8.

  11. Bae T, Hallis SP, Kwak MK. Hypoxia, oxidative stress, and the interplay of HIFs and NRF2 signaling in cancer. Exp Mol Med. 2024 Mar;56(3):501-514.

  12. Lee HN, Lee S, Hong J, et al. Novel FRET-based Immunological Synapse Biosensor for the Prediction of Chimeric Antigen Receptor-T Cell Function. Small Methods. 2025 Mar;9(3):e2401016.

  13. Kosti P, Opzoomer JW, Larios-Martinez KI, et al. Hypoxia-sensing CAR T cells provide safety and efficacy in treating solid tumors. Cell Rep Med. 2021 Apr 9;2(4):100227.

  14. Lazennec G, Richmond A. Chemokines and chemokine receptors: new insights into cancer-related inflammation. Trends Mol Med. 2010 Mar;16(3):133-44.

  15. Gascoigne NR, Casas J, Brzostek J, Rybakin V. Initiation of TCR phosphorylation and signal transduction. Front Immunol. 2011 Dec 7;2:72.

  16. Holler, P. D., Kranz, D. M. Quantitative analysis of the contribution of TCR/pepMHC affinity and CD8 to T cell activation. Immunity. 2003 Feb; 18(2), 255–264.

  17. Alsaieedi AA, Zaher KA. Tracing the development of CAR-T cell design: from concept to next-generation platforms. Front Immunol. 2025 Jul 17;16:1615212.

  18. Dagar G, Gupta A, Masoodi T, et al. Harnessing the potential of CAR-T cell therapy: progress, challenges, and future directions in hematological and solid tumor treatments. J Transl Med. 2023 Jul 7;21:449.

  19. Xu X, Sun Q, Liang X, et al. Mechanisms of relapse after CD19 CAR T-cell therapy for acute lymphoblastic leukemia and its prevention and treatment strategies. Front Immunol. 2019 Nov 12;10:2664.

  20. Nasiri, F., Safarzadeh Kozani, P., Salem, F. et al. Mechanisms of antigen-dependent resistance to chimeric antigen receptor (CAR)-T cell therapies. Cancer Cell Int. 2025 Feb 24;25(1):64.

  21. Kallingal A, Olszewski M, Maciejewska N, Brankiewicz W, Baginski M. Cancer immune escape: the role of antigen presentation machinery. J Cancer Res Clin Oncol. 2023 Aug;149(10):8131-8141.

  22. Lin H, Yang X, Ye S, Huang L, Mu W. Antigen escape in CAR-T cell therapy: Mechanisms and overcoming strategies. Biomed Pharmacother. 2024 Sep;178:117252.

  23. Golikova EA, Alshevskaya AA, Alrhmoun S, Sivitskaya NA, Sennikov SV. TCR-T cell therapy: current development approaches, preclinical evaluation, and perspectives on regulatory challenges. J Transl Med. 2024 Oct 4;22(1):897.

  24. Zhang B, Wu J, Jiang H, Zhou M. Strategies to Overcome Antigen Heterogeneity in CAR-T Cell Therapy. Cells. 2025; 14(5):320.

  25. Anderson KG, Stromnes IM, Greenberg PD. Obstacles Posed by the Tumor Microenvironment to T cell Activity: A Case for Synergistic Therapies. Cancer Cell. 2017 Mar 13;31(3):311-325.

  26. Kouro T, Himuro H, Sasada T. Exhaustion of CAR T cells: potential causes and solutions. J Transl Med. 2022 May 23;20(1):239.

  27. Daei Sorkhabi A, Mohamed Khosroshahi L, Sarkesh A, et al. The current landscape of CAR T-cell therapy for solid tumors: Mechanisms, research progress, challenges, and counterstrategies. Front Immunol. 2023 Mar 20;14:1113882.

  28. Cadinanos-Garai A, Flugel CL, Cheung A, Jiang E, Vaissié A, Abou-El-Enein M. High-dimensional temporal mapping of CAR T cells reveals phenotypic and functional remodeling during manufacturing. Mol Ther. 2025 May 7;33(5):2291-2309.

  29. Bartoszewska E, Tota M, Kisielewska M, et al. Overcoming Antigen Escape and T-Cell Exhaustion in CAR-T Therapy for Leukemia. Cells. 2024 Sep 23;13(18):1596.

  30. Aparicio-Pérez C, Carmona M, Benabdellah K, Herrera C. Failure of ALL recognition by CAR T cells: a review of CD19-negative relapses after anti-CD19 CAR-T treatment in B-ALL. Front Immunol. 2023 Apr 14;14:1165870.

  31. Ellis GI, Sheppard NC, Riley JL. Genetic engineering of T cells for immunotherapy. Nat Rev Genet. 2021 Jul;22(7):427-447.

  32. Choe JH, Watchmaker PB, Simic MS, et al. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med. 2021 Apr 28;13(591):eabe7378.

  33. Hamieh M, Mansilla-Soto J, Rivière I, Sadelain M. Programming CAR T Cell Tumor Recognition: Tuned Antigen Sensing and Logic Gating. Cancer Discov. 2023 Apr 3;13(4):829-843.

  34. Liu Z, Zhou Z, Dang Q, et al. Immunosuppression in tumor immune microenvironment and its optimization from CAR-T cell therapy. Theranostics. 2022 Aug 29;12(14):6273-6290.

  35. Gacerez AT, Arellano B, Sentman CL. How Chimeric Antigen Receptor Design Affects Adoptive T Cell Therapy. J Cell Physiol. 2016 Dec;231(12):2590-8.

  36. Nolan-Stevaux O, Smith R. Logic-gated and contextual control of immunotherapy for solid tumors: contrasting multi-specific T cell engagers and CAR-T cell therapies. Front Immunol. 2024 Nov 13;15:1490911

  37. Nguyen A, Johanning G, Shi Y. Emerging Novel Combined CAR-T Cell Therapies. Cancers (Basel). 2022 Mar 9;14(6):1403.

  38. Lajoie MJ, Boyken SE, Salter AI, et al. Designed protein logic to target cells with precise combinations of surface antigens. Science. 2020 Sep 25;369(6511):1637-1643.

  39. Lei T, Wang Y, Zhang Y, et al. Leveraging CRISPR gene editing technology to optimize the efficacy, safety and accessibility of CAR T-cell therapy. Leukemia. 2024 Dec;38(12):2517-2543.

  40. Lin P, Lin Y, Mai Z, et al. Targeting cancer with precision: strategical insights into TCR-engineered T cell therapies. Theranostics. 2025 Jan 1;15(1):300-323.

  41. Thaxton JE, Li Z. To affinity and beyond: harnessing the T cell receptor for cancer immunotherapy. Hum Vaccin Immunother. 2014;10(11):3313-21.

  42. Chmielewski M, Hombach AA, Abken H. Antigen-Specific T-Cell Activation Independently of the MHC: Chimeric Antigen Receptor-Redirected T Cells. Front Immunol. 2013 Nov 11;4:371.

  43. Cornel AM, Mimpen IL, Nierkens S. MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers (Basel). 2020 Jul 2;12(7):1760.

  44. Yu J, Fu L, Wu R, et al. Immunocytes in the tumor microenvironment: recent updates and interconnections. Front Immunol. 2025 Apr 14;16:1517959.

  45. Harris DT, Kranz DM. Adoptive T Cell Therapies: A Comparison of T Cell Receptors and Chimeric Antigen Receptors. Trends Pharmacol Sci. 2016 Mar;37(3):220-230.

  46. Liu X, Fang J, Huang S, et al. Tumor-on-a-chip: from bioinspired design to biomedical application. Microsyst Nanoeng. 2021 Jun 21;7:50.

  47. Foeng J, Comerford I, McColl SR. Harnessing the chemokine system to home CAR-T cells into solid tumors. Cell Rep Med. 2022 Feb 28;3(3):100543.

  48. Yang J, Bergdorf K, Yan C, et al. CXCR2 expression during melanoma tumorigenesis controls transcriptional programs that facilitate tumor growth. Mol Cancer. 2023 Jun 3;22(1):92.

  49. Aldinucci D, Borghese C, Casagrande N. The CCL5/CCR5 Axis in Cancer Progression. Cancers (Basel). 2020 Jul 2;12(7):1765.

  50. Caruana I, Savoldo B, Hoyos V, et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat Med. 2015 May;21(5):524-9.

  51. Kapałczyńska M, Kolenda T, Przybyła W, et al. 2D and 3D cell cultures – a comparison of different types of cancer cell cultures. Arch Med Sci. 2018 Jun;14(4):910-919.

  52. Saratkar SY, Langote M, Kumar P, Gote P, Weerarathna IN, Mishra GV. Digital twin for personalized medicine development. Front Digit Health. 2025 Aug 7;7:1583466.

  53. Gargett T, Brown MP. The inducible caspase-9 suicide gene system as a “safety switch” to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front Pharmacol. 2014 Oct 28;5:235.

  54. Tan P, He L, Han G, Zhou Y. Optogenetic Immunomodulation: Shedding Light on Antitumor Immunity. Trends Biotechnol. 2017 Mar;35(3):215-226.

  55. Nguyen NT, Huang K, Zeng H, et al. Nano-optogenetic engineering of CAR T cells for precision immunotherapy with enhanced safety. Nat Nanotechnol. 2021 Dec;16(12):1424-1434.

  56. Ahmed EN, Cutmore LC, Marshall JF. Syngeneic Mouse Models for Pre-Clinical Evaluation of CAR T Cells. Cancers (Basel). 2024 Sep 18;16(18):3186.

  57. Andreu-Sanz D, Gregor L, Carlini E, Scarcella D, Marr C, Kobold S. Predictive value of preclinical models for CAR-T cell therapy clinical trials: a systematic review and meta-analysis. J Immunother Cancer. 2025 Jun 12;13(6):e011698

  58. Cattaneo CM, Dijkstra KK, Fanchi LF, et al. Tumor organoid-T-cell coculture systems. Nat Protoc. 2020 Jan;15(1):15-39.

  59. Binnewies M, Roberts EW, Kersten K,et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018 May;24(5):541-550.

  60. Jafari Nivlouei S, Soltani M, Carvalho J, Travasso R, Salimpour MR, Shirani E. Multiscale modeling of tumor growth and angiogenesis: Evaluation of tumor-targeted therapy. PLoS Comput Biol. 2021 Jun 23;17(6):e1009081.

  61. Baena JC, Victoria JS, Toro-Pedroza A, et al. Smart CAR-T Nanosymbionts: archetypes and proto-models. Front Immunol. 2025 Aug 12;16:1635159.

  62. Silva A, Vale N. Digital twins in personalized medicine: bridging innovation and clinical reality. J Pers Med. 2025 Oct;15:503.

  63. Abdo L, Batista-Silva LR, Bonamino MH. Cost-effective strategies for CAR-T cell therapy manufacturing. Mol Ther Oncolytics. 2025 Jun 18;33:200980.

  64. Dias J, Garcia J, Agliardi G, Roddie C. CAR-T cell manufacturing landscape-Lessons from the past decade and considerations for early clinical development. Mol Ther Methods Clin Dev. 2024 Apr 16;32(2):101250.

  65. Holstein M, Mesa-Nuñez C, Miskey C, et al. Efficient Non-viral Gene Delivery into Human Hematopoietic Stem Cells by Minicircle Sleeping Beauty Transposon Vectors. Mol Ther. 2018 Apr 4;26(4):1137-1153.

  66. Manuri PV, Wilson MH, Maiti SN, et al. piggyBac transposon/transposase system to generate CD19-specific T cells for the treatment of B-lineage malignancies. Hum Gene Ther. 2010 Apr;21(4):427-37.

  67. Su J, Zeng Y, Song Z, et al. Genome-edited allogeneic CAR-T cells: the next generation of cancer immunotherapies. J Hematol Oncol. 2025 Oct 24;18(1):90.

  68. Mangani S, Kremmydas S, Karamanos NK. Mimicking the Complexity of Solid Tumors: How Spheroids Could Advance Cancer Preclinical Transformative Approaches. Cancers (Basel). 2025 Mar 30;17(7):1161.

  69. Zhang Y, Liu Z, Wei W, Li Y. TCR engineered T cells for solid tumor immunotherapy. Exp Hematol Oncol. 2022 Jun 20;11(1):38.

  70. Shang D, Zhou Z, Shi R, et al. Nanoparticle-based strategy in CAR-T cell immunotherapy: challenges, implications, and perspectives. Mol Cancer. 2025 Nov 4;24(1):281.