What CAR-Tr​​eg are being developed?

17 March 2025
Introduction to CAR-Treg Therapies
CAR‐Treg therapies represent a cutting‐edge development in adoptive cell therapy, where regulatory T cells (Tregs) are genetically engineered to express chimeric antigen receptors (CARs) to target specific antigens. Unlike conventional CAR‐T cell therapies that are designed to kill target cells such as tumor cells, CAR‐Tregs are designed to home to sites of inflammation or graft tissue and provide localized immunosuppression. These “living drugs” can restore immune tolerance and curb pathogenic immune responses. This innovative approach combines the targeting precision of CAR strategies with the intrinsic immunomodulatory functions of Tregs, potentially overcoming some limitations of both non‐specific immunosuppressive drugs and conventional adoptive T‐cell therapies.

Definition and Basic Concepts of CAR-Treg
CAR-Tregs are regulatory T cells that have been genetically modified to express a chimeric antigen receptor. The CAR itself is a synthetic receptor typically composed of an extracellular antigen-binding domain—usually in the form of a single-chain variable fragment (scFv)—a hinge region, a transmembrane domain, and intracellular signaling domains. In CAR-Tregs, the signaling domain is optimized to maintain the suppressive phenotype of Tregs while enabling their activation in the presence of a specific antigen. This design allows the modified Tregs to recognize antigens in an HLA-independent manner and to traffic to specific tissues where their immunosuppressive functions are needed. Their key role is to suppress effector immune responses (such as T cell–mediated cytotoxicity), secrete anti-inflammatory cytokines, and promote local immune tolerance. Unlike conventional Tregs, which are polyclonal and may require high doses due to limited antigen specificity, CAR-Tregs are engineered to be antigen-specific, greatly enhancing their potency in diseases where targeted suppression is desired.

Comparison with CAR-T Cell Therapies
Although both CAR-T cells and CAR-Tregs utilize the same underlying CAR technology, their mechanisms of action and therapeutic goals differ dramatically. CAR-T cell therapy is primarily intended for the eradication of pathological cells, such as malignant cancer cells; their cytotoxic activity is driven by the activation of killing machinery upon antigen recognition, and they often are associated with inflammatory side effects like cytokine release syndrome (CRS). By contrast, CAR-Tregs do not primarily mediate cytotoxicity. Instead, they suppress immune responses, promote tolerance, and modulate inflammation once they localize at the target site. This immunosuppressive function is critical in applications such as autoimmune disorders and the prevention of organ rejection after transplantation. In addition, while CAR-T cells can sometimes be limited by off-tumor toxicity due to a lack of unique tumor-specific antigens, CAR-Tregs are generally intended to dampen immune responses locally without systemic immunosuppression, potentially reducing the risk of adverse effects like infections and unintended tissue damage. The differences in co-stimulatory domains used (CD28 vs. 4-1BB, for example) and in manufacturing processes also influence their function and persistence, with current evidence suggesting that a CD28 domain may be more favorable for maintaining the suppressive phenotype in CAR-Tregs.

Current Developments in CAR-Treg Therapies
The development of CAR-Treg therapies has accelerated over the past several years with a number of academic and industrial groups working towards constructing, testing, and refining these engineered cells. Ongoing research rather than the mere adaptation of CAR-T cell technology reflects a growing understanding of Treg biology and the need for immune tolerance restoration in diverse clinical settings.

Leading Research and Development Initiatives
A number of initiatives are driving the progress in CAR-Treg development:
- Antigen-Specific CAR-Tregs for Autoimmunity: Several academic studies have engineered CAR-Tregs with antigen-specificity to treat autoimmune conditions. For example, research has explored the use of CAR-Tregs with scFv domains directed towards antigens such as insulin in type 1 diabetes, or carcinoembryonic antigen (CEA) in inflammatory bowel disease—a strategy designed to localize immunosuppression to the affected tissue and reduce systemic side effects.
- CAR-Tregs in Organ Transplantation: One promising avenue is the development of donor-antigen-specific CAR-Tregs for preventing graft rejection. An excellent example is the engineering of CAR-Tregs to recognize donor HLA molecules (typically HLA-A2), which has been shown in preclinical models to enhance graft tolerance while minimizing systemic immunosuppression. Sangamo Therapeutics, for instance, has advanced a candidate—TX200—in a phase 1/2 clinical study aimed at preventing immune-mediated rejection post-kidney transplantation.
- Optimization of Co-Stimulation and Intracellular Signaling: Researchers are also refining the structural components of the CAR to better support Treg survival and function. Several studies have compared co-stimulatory domains, with a recurring finding that including a CD28 costimulatory domain may help preserve Treg phenotype and function better than alternatives, such as 4-1BB, which in some cases may attenuate suppression.
- Comparative Evaluations of CAR Constructs: Investigations into different CAR designs are not limited merely to the extracellular targeting component. Detailed preclinical studies are assessing how intracellular signaling cascades impact Treg expansion, persistence, and immunosuppressive abilities in vivo, thereby helping to shape the next generation of CAR-Treg products. These design alterations are guided by both in vitro functional studies and in vivo animal models, ensuring that the engineered Tregs maintain their regulatory characteristics even when repeatedly stimulated.

Clinical Trials and Preclinical Studies
Preclinical and early clinical studies have begun to shed light on the safety and efficacy of CAR-Treg therapies:
- Preclinical Models in Autoimmunity: Several murine models of autoimmune diseases have been employed to test CAR-Tregs. In one study using a colitis mouse model, CAR-Tregs directed against trinitrophenol demonstrated the capacity to reduce colitis severity without reverting to an inflammatory phenotype. Similar models in type 1 diabetes have been explored, with engineered CAR-Tregs targeting beta cell antigens showing potential to suppress autoreactive T cells, although challenges related to persistence of the cells have also been noted.
- Organ Transplantation Studies: In transplantation, preclinical studies have focused on CAR-Tregs designed to recognize donor-specific antigens, such as HLA-A2. Studies in humanized mouse models have demonstrated that these CAR-Tregs can home effectively to the transplanted organ and mitigate alloreactive T cell responses, thereby prolonging graft survival without the need for high doses of immunosuppressants. The STEADFAST phase 1/2 study, evaluating an autologous HLA-A2-specific CAR-Treg (TX200-TR101 product) in renal transplant recipients, represents one of the earliest clinical applications in humans, marking a significant milestone towards translation.
- Additional Early-Phase Human Studies: Although still in the nascent stages of clinical translation, several phase I/II trials are underway that will test the feasibility, safety, and persistence of CAR-Tregs in various indications. Early signals indicate that these therapies may achieve localized immune suppression with minimal systemic adverse effects compared to traditional immunosuppressive regimens.

Potential Applications of CAR-Treg Therapies
CAR-Treg therapies are being developed for a broad spectrum of clinical applications. Their underlying principle of inducing antigen-specific tolerance makes them attractive across several disease areas.

Autoimmune Diseases
Autoimmune conditions are characterized by the immune system’s aberrant response against self-antigens, and conventional immunosuppressive therapies often lack specificity, leading to generalized immune suppression. CAR-Tregs, however, offer the potential to target only the pathological components of the immune response.
- Multiple Sclerosis, Rheumatoid Arthritis, and Systemic Lupus Erythematosus: Several studies have explored the use of CAR-Tregs for autoimmune disorders. For instance, research in lupus models using anti-CD19 CAR-T cells (and by extension, similar strategies are under investigation with CAR-Tregs) suggests that targeted depletion or suppression of autoreactive B cells can lead to long-lasting remission. CAR-Tregs could be engineered to express receptors targeting specific autoantigens unique to these diseases to re-establish immune homeostasis without the off-target effects typically seen with systemic immunosuppression.
- Inflammatory Bowel Diseases (IBD): In diseases like ulcerative colitis and Crohn’s disease, CAR-Tregs engineered to recognize gut-specific antigens such as CEA or other markers have demonstrated in preclinical models the potential to reduce gut inflammation and restore mucosal integrity. Patient willingness studies toward CAR-Treg therapies in IBD further highlight the clinical need and prospective acceptance of such approaches, underscoring their future role in personalized medicine.
- Type 1 Diabetes: Given that type 1 diabetes (T1D) results from an autoimmune attack on pancreatic beta cells, CAR-Tregs have been studied as a strategy to mitigate this response. Although early studies using anti-insulin CAR-Tregs showed variable results in terms of persistence, these studies underscore the potential of targeted immunotherapy in preventing or mitigating T1D by restoring beta cell tolerance.

Organ Transplantation
A particularly promising application of CAR-Treg therapies is in the field of transplantation. Organ transplantation currently relies on life-long immunosuppression, which carries significant long-term risks.
- Prevention of Allograft Rejection: CAR-Tregs engineered to target donor-specific antigens, such as mismatched HLA molecules (typically HLA-A2), offer a means to induce localized immune tolerance. Preclinical studies have demonstrated that such cells can home to the graft, suppress alloreactive T cell responses, and prolong graft survival.
- Clinical Translation in Renal Transplantation: The STEADFAST study is a landmark trial wherein CAR-Tregs (e.g., TX200) are being evaluated for their efficacy in preventing rejection in kidney transplant recipients. By targeting donor HLA antigens, these cells could significantly reduce or even obviate the need for chronic immunosuppression, minimizing associated complications such as infections, cardiovascular disease, and malignancy.
- Liver Transplantation and Beyond: In addition to renal transplantation, other studies have explored the use of CAR-Tregs in liver transplantation. Early pilot studies using donor-specific Tregs have shown promise in improving graft function and reducing immunosuppressant burden, and adaptations using CAR technology are expected to further enhance these outcomes.

Challenges in CAR-Treg Therapy Development
Despite their tremendous potential, the development of CAR-Treg therapies is fraught with complex challenges that span technical, manufacturing, and safety concerns.

Technical and Manufacturing Challenges
- Cell Source and Isolation: One of the principal challenges in CAR-Treg therapy is obtaining a sufficient number of highly functional Tregs from patients. Treg populations are naturally low in peripheral blood, and isolating high-purity, stable Tregs demands sophisticated protocols. Furthermore, generating an autologous product for each patient is a labor-intensive and expensive process.
- Genetic Engineering and Expansion: The genetic modification process, often involving viral transduction or gene editing technologies, must preserve the regulatory phenotype of Tregs. Studies have shown that even subtle differences in the vector or the insertion method can affect Treg stability and suppressive functions. Additionally, ex vivo expansion of CAR-Tregs—necessary to yield therapeutic doses—requires careful optimization to avoid Treg exhaustion while maintaining their suppressive capabilities.
- CAR Construct Optimization: Designing the CAR itself presents technical hurdles. The choice of antigen-binding domain, the length and composition of the hinge region, the choice of transmembrane domain, and especially the costimulatory domains (with evidence supporting the use of CD28 over 4-1BB for Tregs) all significantly affect CAR-Treg function. Fine-tuning each of these components for optimal performance in Tregs, as opposed to cytotoxic T cells, demands extensive preclinical testing.
- Manufacturing Infrastructure: Current cell therapy manufacturing platforms are largely based on protocols developed for CAR-T cells. Adapting these methods for the production of CAR-Tregs involves additional challenges, given the distinct biology of Tregs. The need for scalable, reproducible, high-purity cell products necessitates the development of specialized bioreactor-based processes and comprehensive quality control standards.

Safety and Efficacy Concerns
- Off-Target Suppression and Systemic Immunosuppression: Although CAR-Tregs are engineered to provide localized immunosuppression, there is a valid concern that they could migrate to off-target sites and induce systemic immunosuppression. Such an event could increase a patient’s susceptibility to infections or even permit the development of malignancies.
- Phenotypic Instability: The maintenance of a stable Treg phenotype after genetic modification is critical. One of the major risks is that CAR-Tregs may lose FOXP3 expression over time, potentially converting into proinflammatory effector T cells, which would be counterproductive and could exacerbate the disease condition.
- Immunogenicity of CAR Constructs: There is also potential for the immune system to recognize the CAR constructs as foreign, particularly the murine-derived scFv components, leading to immune-mediated rejection of the CAR-Tregs. Ongoing efforts in humanizing these components are critical to mitigate such risks.
- In Vivo Persistence and Expansion: While persistence of CAR-T cells is often desirable for sustained tumor suppression, for CAR-Tregs, too prolonged or uncontrolled activity might likewise pose safety risks. The short-term persistence observed in some studies may limit long-term efficacy, while prolonged persistence could pose risks of prolonged immunosuppression.
- Cytokine Release and Interaction with Other Therapies: Although CAR-Tregs typically do not induce the robust cytokine release seen with CAR-T cells, careful monitoring is still required. Some studies suggest that even a modest cytokine induction might affect host immune modulation, and there is ongoing research into combining CAR-Tregs with other agents to optimize safety profiles.

Future Directions and Opportunities
Despite these challenges, numerous opportunities exist to further optimize CAR-Treg therapies, driven by rapid innovations in genetic engineering, manufacturing processes, and clinical trial design.

Innovations in CAR-Treg Technology
- Next-Generation CAR Designs: Future innovations may include the development of “smart” CARs that incorporate regulatory switches allowing external control over CAR-Treg activity. This could involve the integration of inducible expression systems or safety switches that can deactivate or even eliminate the CAR-Tregs in the case of adverse events.
- Genome Editing Approaches: New advancements in genome editing, including CRISPR-Cas9 and TALENs, are being integrated to modify Tregs with greater precision. Such techniques could be used to knock out genes that might otherwise promote instability or conversion to effector cells, or to insert the CAR construct into a defined locus (for instance, the TRAC locus) to ensure more consistent expression and function.
- Optimized Expansion Protocols: New bioreactor-based manufacturing methods are under development to streamline the expansion of CAR-Tregs while preserving their phenotype. Process optimization strategies are being leveraged to produce large numbers of genetically stable and functionally potent Tregs.
- Combination Therapies: The future may see CAR-Treg therapies being combined with other immunomodulatory agents, such as low-dose IL-2 or rapamycin, to enhance Treg activity and persistence while mitigating the risk of phenotypic instability. Combined therapeutic strategies may also help tailor treatments to individual patients’ needs.

Regulatory and Ethical Considerations
- Clinical Trial Design and Regulatory Approvals: As the first phase I/II trials of CAR-Treg therapies progress, regulators and clinicians must collaborate closely to establish robust protocols for patient monitoring and safety assessments. Regulatory agencies are taking note of the unique challenges presented by CAR-Treg products, including discussions on product characterization, dosing strategies, and long-term safety monitoring.
- Ethical Considerations in Genetic Engineering: The use of genetic engineering to modify Tregs raises important ethical questions. Ensuring patient safety while addressing concerns about long-term immune alterations is a critical area of discussion. Transparent reporting of clinical trial results, informed consent procedures, and careful post-treatment monitoring are essential to building public trust in these novel therapies.
- Cost and Accessibility: The complexity of manufacturing CAR-Treg therapies also poses challenges in terms of cost and scalability. Efforts to standardize and streamline the manufacturing process, coupled with innovations in off-the-shelf allogeneic products, could help lower the costs and increase access to these therapies in the future.

Detailed Conclusion
In summary, CAR-Treg therapies are being developed as a novel and promising strategy to restore immune tolerance in a range of clinical settings, including autoimmune diseases and organ transplantation. The fundamental premise is to harness the precision targeting capability of CAR technology while leveraging the potent immunosuppressive functions of regulatory T cells. Researchers have been focusing on developing antigen-specific CAR-Tregs, such as those targeting specific autoantigens in diseases like type 1 diabetes and inflammatory bowel disease, as well as donor-specific antigens (e.g., HLA-A2) to prevent graft rejection in organ transplantation. Preclinical models have demonstrated promising results, and early-phase clinical trials—such as the STEADFAST study evaluating TX200 for renal transplantation—underscore the translational potential of these therapies.

However, significant challenges remain. Technical hurdles such as the efficient isolation, expansion, and genetic modification of Tregs must be overcome to ensure that these cells retain their regulatory phenotype and do not convert to pro-inflammatory cells. Safety concerns regarding off-target immunosuppression, potential immunogenicity of the CAR components, and long-term in vivo persistence require careful evaluation. Yet, innovative solutions such as next-generation CAR designs with built-in safety switches, advanced genome editing techniques, and optimized manufacturing processes are actively being pursued. Moreover, regulatory and ethical aspects, including clinical trial design, long-term safety monitoring, and cost considerations, are paramount to ensure that CAR-Treg therapies are not only effective but also accessible and ethically sound.

The future of CAR-Treg therapy is poised for transformative breakthroughs. As our understanding of Treg biology deepens and manufacturing and engineering techniques continue to evolve, these therapies offer the potential to deliver precise, durable, and safe immunomodulation tailored to individual patient needs. Whether deployed to quell the autoimmune cascade in diseases like lupus, rheumatoid arthritis, inflammatory bowel disease, or to promote long-term graft tolerance in transplant recipients, CAR-Tregs provide a versatile platform that may redefine the landscape of immune therapy. Ultimately, the convergence of technological innovation, rigorous clinical testing, and thoughtful regulatory oversight will be crucial to realizing the promise of CAR-Treg therapies in clinical practice.

In conclusion, CAR-Treg therapies being developed today represent a multi-faceted approach targeting both autoimmune diseases and organ transplantation with high specificity and minimal side effects. The field is rapidly evolving, with significant contributions from academic research and industry-driven innovation reflected in extensive preclinical evaluations and the initiation of early-phase clinical trials. As refinements continue in CAR design, expansion methods, and safety monitoring, the prospect of achieving lasting immune tolerance with CAR-Tregs becomes increasingly attainable. These advances herald a new era in personalized immunotherapy where engineered Tregs could eventually offer effective, targeted, and safer treatments for diseases marked by immune dysregulation, thereby improving long-term patient outcomes while minimizing the burden of chronic immunosuppressive medications.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.