What are the different types of drugs available for CAR-Tr​​eg?

17 March 2025
Introduction to CAR-Treg Therapy
CAR-Treg (chimeric antigen receptor regulatory T cell) therapy represents a novel class of cell-based drugs in which regulatory T cells (Tregs) are genetically engineered to express a chimeric antigen receptor (CAR) that endows them with enhanced antigen specificity and the ability to home to inflamed tissues. These engineered Tregs are designed to modulate and suppress aberrant immune responses, thereby restoring immunological tolerance in conditions such as autoimmune diseases, transplant rejection, and inflammatory disorders.

Definition and Mechanism of CAR-Treg
CAR-Treg therapy involves isolating Tregs from a patient (or a donor), followed by ex vivo genetic modification to introduce a CAR. The CAR construct typically consists of:
• An extracellular antigen-recognition domain—most often a single-chain variable fragment (scFv) that confers target specificity.
• A hinge and transmembrane region that provides flexibility and anchorage on the T cell surface.
• An intracellular signaling domain that frequently includes a costimulatory domain (such as CD28 or 4-1BB) fused to the CD3ζ signaling chain.

For CAR-Tregs, the choice of costimulatory domains is critical; for instance, CD28 is often preferred over 4-1BB because it has been shown to maintain a stable Treg phenotype and promote robust suppressive function. When these engineered cells recognize antigens expressed on target tissues, they are activated to release anti-inflammatory cytokines and mediate contact-dependent suppression, thereby inhibiting autoreactive effector cell activity.

Importance in Immunotherapy
The therapeutic importance of CAR-Tregs lies in their potential to provide antigen-specific immunosuppression, which contrasts with the generalized immunosuppression associated with many current treatments. By targeting defined antigens, CAR-Tregs can localize their effects primarily at the site of immune-mediated pathology, reducing systemic side effects. This precision is particularly valuable in autoimmune diseases, organ transplantation, and inflammatory conditions where restoration of immune tolerance is essential for long-term remission. Moreover, CAR-Treg therapy holds promise for lowering the need for chronic immune-suppressing drugs, which are often accompanied by complications such as increased risk of infections and malignancies.

Types of Drugs in CAR-Treg Therapy
The realm of CAR-Treg drugs is evolving rapidly and can be broadly divided into various classifications based on both the manufacturing approach and the design of the CAR construct. The available types span from early-generation autologous therapies to highly engineered next-generation products with safety switches and other advanced features.

Classification of Drugs
CAR-Treg drugs can be categorized along several dimensions, including:

1. Based on the Source of Tregs:
• Autologous vs. Allogeneic CAR-Tregs:
Autologous CAR-Tregs are derived from the patient’s own cells, minimizing the risk of graft-versus-host disease (GvHD) and immunologic rejection. However, the manufacturing process can be challenging in patients with an underlying immunological disorder. Allogeneic CAR-Tregs, harvested from healthy donors, are being explored to provide off-the-shelf products, though they may require further genetic modifications (e.g., disruption of the TCR or MHC components to reduce immunogenicity).

2. Based on CAR Construct Generation and Design:
• Second-generation CAR-Tregs:
These include CAR constructs with a single costimulatory domain (e.g., CD28), which has been found to maintain Treg stability and potent suppressive function. The second-generation approach has dominated current clinical trials because of its balanced activation and safety profile.
• Next-generation CAR-Tregs (Third/Fourth-generation):
Newer designs integrate additional safety features such as suicide genes or inducible “off switches” that allow for the rapid elimination of CAR-Tregs if adverse events occur. They may also incorporate multiple costimulatory domains or logic gating systems to further refine antigen specificity, persistence, and activity in complex tissue environments.
• CARs with modified co-stimulatory domains:
As noted in multiple studies, the selection of costimulatory domains in CAR-Tregs is critical. For instance, while standard CAR-T designs in cancer therapy might employ 4-1BB for enhanced persistence, in Tregs, CD28-based constructs have been shown to be superior in maintaining immunosuppressive function.

3. Based on Target Antigen Specificity:
• HLA-specific CAR-Tregs:
Some CAR-Treg drugs are designed to recognize major histocompatibility complex (MHC) molecules such as HLA-A2. An example is QEL-001, which is under development by Quell Therapeutics Ltd., and targets HLA-A2—acting through mechanisms such as HLA-A2 inhibition, gene transference, and T lymphocyte replacement.
• Antigen-specific vs. Polyclonal CAR-Tregs:
Antigen-specific CAR-Tregs are engineered to express a receptor that recognizes a single or a limited set of antigens associated with a disease condition, whereas polyclonal Tregs do not possess such specificity. Antigen-specific CAR-Tregs have been shown to have increased potency, a higher safety profile, and less off-target immunosuppression compared to their polyclonal counterparts.

4. Based on Advanced Engineering Features:
• CAR-Tregs with Safety Switches:
Innovative approaches include the incorporation of “kill switches” or “suicide genes” that allow clinicians to rapidly ablate the infused cells if necessary, enhancing safety particularly when severe immunosuppression or off-target effects are observed.
• Gene-edited CAR-Tregs:
Emerging strategies involve using genome-editing tools (e.g., CRISPR-Cas9 or TALENs) to remove endogenous TCR components or to knock down specific genes (such as Tet2) that might otherwise lead to uncontrolled activation. These modifications improve both safety and efficacy by reducing the risk of alloreactivity.

Specific Drugs Used
Based on current references from the Synapse database (noting that these are structured and oftentimes more reliable sources):

1. **QEL-001:**
QEL-001 is noted as a CAR-Treg drug candidate developed by Quell Therapeutics Ltd. It is designed for the treatment of immune system diseases and digestive system disorders. This drug product is in Phase 2 development and primarily functions as a CAR-Treg with mechanisms including HLA-A2 inhibition, gene transference, and T lymphocyte replacement.

2. **PolTREG Pipeline Candidates (e.g., PTG-007):**
PolTREG is a company actively developing next-generation Treg therapies, including CAR-Tregs. Their lead candidate, PTG-007, is designed for multiple indications such as type 1 diabetes (T1D), multiple sclerosis (MS), and even neurodegenerative diseases like amyotrophic lateral sclerosis (ALS). Although PTG-007 is primarily described under a broader regulatory T cell therapy pipeline, the CAR-Treg approach is a significant component of their strategy, further underscoring the heterogeneity in CAR-Treg drugs.

3. **Other Preclinical Candidates:**
There are several investigational drugs that fall under the umbrella of engineered CAR-Treg therapies that have been tested in various autoimmune disease models. For instance:
• Studies have explored anti-insulin CAR-Tregs for type 1 diabetes prevention and treatment.
• CAR-Tregs engineered with specific antigen receptors (such as those targeting myelin oligodendrocyte glycoprotein [MOG] in experimental autoimmune encephalomyelitis) have been evaluated in preclinical models, indicating potential for diseases like multiple sclerosis.
• CAR-Tregs targeting epithelial antigens (such as carcinoembryonic antigen [CEA], used in contexts like inflammatory bowel disease) have also been explored.
While many of these candidates are in the preclinical or early clinical trial stage, they exemplify the rapid diversification in CAR-Treg drug design to optimize tissue specificity, homing, persistence, and overall suppressive activity.

Applications of CAR-Treg Drugs
CAR-Treg drugs are not simply a single therapeutic product; they represent a breakthrough strategy that can be applied across a range of clinical conditions where immune modulation is desired. Their engineered specificity allows them to be used as “living drugs” that provide targeted immunomodulation with fewer systemic side effects.

Disease Areas Targeted
CAR-Treg therapies are being developed for various disease areas, including:

1. **Autoimmune Diseases:**
CAR-Treg drugs have been demonstrated in multiple preclinical studies to reverse or mitigate autoimmune pathology. Examples include:
• **Type 1 Diabetes (T1D):** Anti-insulin or GAD65 CAR-Tregs have been designed to target pancreatic beta cell antigens and promote immune tolerance, thereby preventing beta-cell destruction.
• **Multiple Sclerosis (MS):** CAR-Tregs targeting myelin components such as MOG help curb inflammation in the central nervous system. The antigen-specific suppression by these cells aids in reducing demyelination and neurodegeneration.
• **Inflammatory Bowel Disease (IBD):** CAR-Tregs engineered to recognize epithelial antigens (e.g., CEA) have been shown in animal models to reduce colitis severity and prevent the subsequent development of colitis-associated colorectal cancer.

2. **Transplantation:**
In organ transplantation, the risk of graft rejection remains a significant challenge. CAR-Treg therapies that are designed to recognize donor-specific antigens (such as HLA-A2) can promote allograft tolerance and reduce the need for lifelong immunosuppression. QEL-001, for instance, includes mechanisms that may support transplantation immunotolerance.

3. **Inflammatory and Immune-Mediated Disorders:**
Beyond classical autoimmunity and transplantation, engineered Tregs have potential roles in treating a variety of inflammatory conditions. This includes diseases where inflammation is a driver of pathology—for instance, in certain neurological or hepatic conditions. CAR-Treg therapies might modulate local immune responses to prevent chronic tissue damage.

Clinical Trials and Outcomes
Most clinical data on CAR-Treg therapies are presently emerging from early-phase trials and preclinical studies, and they are assessed alongside other cell-based immunotherapies. Key points include:

• **Early-Stage Clinical Studies:**
Clinical trials for CAR-Treg therapies have focused on safety, tolerability, and early signs of efficacy. For example, QEL-001 is in Phase 2 with a focus on patients with immune and digestive system disorders.
• **Preclinical Successes:**
Many preclinical studies have demonstrated strong immunosuppressive capabilities of CAR-Tregs, with animal models of T1D, MS, and colitis showing promising results. These studies emphasize the importance of antigen-specific Treg activity that exceeds that of bulk polyclonal Tregs.
• **Pipeline Expansion:**
Companies like PolTREG are actively expanding their pipelines not only through CAR-Treg drugs but also by combining multiple approaches (such as TCR-Tregs) for diseases that require nuanced immunomodulation.

The evolving clinical outcomes underscore that while early results are promising, mitigation of side effects and ensuring long-term persistence and stability of the infused cell population remain critical endpoints.

Challenges and Future Directions
CAR-Treg therapies as a drug class present unique challenges in design, manufacturing, and clinical implementation. Their development is intricately linked with advances in genetic engineering, cell manufacturing protocols, regulatory standardization, and safety monitoring. Understanding these challenges is essential to pave the way for broader clinical adoption and eventual commercialization.

Current Challenges in Drug Development
Several key challenges remain in the development and clinical deployment of CAR-Treg drugs:

1. **Manufacturing Complexity and Standardization:**
The process of isolating, genetically engineering, and expanding Tregs demands specialized facilities and expertise. Autologous manufacturing raises issues related to variable cell quality from patients with active disease and the lengthy production timelines needed for personalized cell therapies. Standardizing manufacturing protocols and improving scalability are major priorities, and off-the-shelf allogeneic products may alleviate some production challenges, albeit introducing issues of immune compatibility.

2. **CAR Design and Treg Stability:**
Engineering a CAR that provides adequate activation without compromising the inherent regulatory function of the Treg is a delicate balance. For instance, the choice of co-stimulatory domain is critical: while 4-1BB has been effective in conventional CAR-T cells, it may impair Treg suppressive activity, making CD28 more suitable. Additionally, ensuring that CAR-Tregs maintain a stable FoxP3-positive phenotype over time is essential to avoid potential conversion into effector T cells that may exacerbate autoimmunity.

3. **Safety Concerns and Off-target Effects:**
While CAR-Tregs aim for antigen-specific immunosuppression, unintended targeting of non-diseased cells expressing low levels of the target antigen remains a risk. The incorporation of safety switches (such as suicide genes) is therefore important, but these must be finely tuned so as not to reduce therapeutic efficacy. Excessive persistence might lead to prolonged immunosuppression or increase infection risk, whereas poor persistence could lead to loss of therapeutic effect.

4. **Immunogenicity and Persistence:**
The host immune system may recognize genetically engineered CAR-Tregs as foreign, potentially leading to their premature elimination and reduced therapeutic durability. Techniques such as gene-editing to remove immunogenic markers or to knock out TCR chains have been explored to improve persistence and reduce immunogenicity.

5. **Regulatory and Economic Hurdles:**
As with other advanced therapy medicinal products (ATMPs), CAR-Treg drugs must pass rigorous regulatory scrutiny regarding quality, safety, and efficacy. Furthermore, the high manufacturing costs and complexities of individualized cell-based products raise questions about overall cost-effectiveness. Establishing a sustainable economic model that balances high production costs with patient access is an ongoing challenge.

Future Prospects and Research Directions
Looking ahead, several research strategies and technological advances may help to overcome current limitations and broaden the clinical impact of CAR-Treg therapies:

1. **Advances in Genomic Engineering:**
The use of CRISPR-Cas9 and TALENs to edit genes within Tregs is expected to significantly improve both the safety and efficacy profiles of CAR-Tregs. For example, disruption of endogenous TCR or immunogenic markers and targeted knockdown of genes such as Tet2 can improve cell function and persistence. These modifications can also reduce the need for lifelong immunosuppression by enhancing the intrinsic regulatory capacity of the cells.

2. **Next-generation CAR Constructs:**
Incorporation of multiple costimulatory domains or even logic gates (that allow CAR-Tregs to respond only when multiple antigens are present) may further enhance specificity and safety. The design of inducible safety switches that can rapidly eliminate CAR-Tregs in case of adverse events is also a promising area of development. Furthermore, combining CAR-Treg therapies with additional immunomodulatory agents (e.g., low-dose IL-2) may synergistically enhance their suppressive function and durability in vivo.

3. **Improved Manufacturing Processes:**
Automation, shortened ex vivo expansion times, and optimization of cell culture processes will be crucial in reducing manufacturing times and costs. Innovations in bioreactor technology and process standardization will likely pave the way for more reproducible and scalable manufacturing platforms. Allogeneic “off-the-shelf” products, expanded from healthy donor Tregs, are an actively pursued alternative that may address current logistical challenges.

4. **Enhanced Safety Monitoring and Clinical Protocols:**
Development of robust biomarkers to monitor CAR-Treg persistence, functionality, and phenotypic stability in patients will help tailor treatment protocols and minimize risks. Long-term follow-up studies are critical, especially in autoimmunity, to understand the durability of therapeutic responses and potential late-onset toxicities. Moreover, tighter collaboration between academic research, clinical centers, and industry partners will be essential for advancing CAR-Treg therapies from preclinical models into routine clinical practice.

5. **Expanding Application Areas and Combination Approaches:**
While early trials have mostly centered around autoimmune diseases and transplantation, there is potential to explore CAR-Treg applications in other inflammatory disorders, neurodegeneration, and even in combination with conventional immunosuppressants or biological therapies. Such combined therapeutic regimens may offer synergistic benefits by coupling precise, localized immunosuppression provided by CAR-Tregs with systemic therapies, thereby reducing overall drug dosages and associated side effects.

Conclusion
CAR-Treg therapies represent one of the most exciting and promising fields in cell-based immunotherapy. They are designed to induce antigen-specific immunosuppression by engineering regulatory T cells with chimeric antigen receptors. The diversity in CAR-Treg drug design—ranging from autologous products like QEL-001 (targeting HLA-A2) to emerging pipelines from companies like PolTREG (with candidates such as PTG-007)—reflects an evolving landscape that is addressing a wide spectrum of immune-mediated diseases.

From a classification perspective, CAR-Treg drugs can be segregated based on the cell source (autologous versus allogeneic), the generation and engineering of the CAR construct (second-generation to next-generation with advanced safety features), and the target antigen specificity (HLA-specific, autoantigen-specific, or epitope-directed constructs). Each type has been designed with a specific therapeutic application in mind, whether it be for treating autoimmune diseases such as type 1 diabetes, multiple sclerosis, and inflammatory bowel disease, or for promoting graft tolerance in transplantation. Preclinical studies and early-phase clinical trials have demonstrated promising safety and efficacy profiles. However, challenges remain in terms of manufacturing complexity, ensuring stability and persistence of the regulatory phenotype, mitigating immunogenicity and off-target effects, and establishing sustainable economic models.

Future research directions point towards improved genomic and molecular engineering, including the use of gene-editing tools to optimize CAR design and cell persistence. Next-generation CAR constructs that incorporate multiple safety switches and combinatorial signaling domains are expected to enhance both the potency and the safety profile of these drugs. Additionally, the integration of advanced manufacturing techniques and the development of off-the-shelf products could significantly improve accessibility and reduce costs, thereby broadening the therapeutic impact of CAR-Treg drugs on immune-mediated conditions.

In summary, the different types of drugs available for CAR-Treg therapy encompass a spectrum of engineered products defined by their source, CAR design, and target specificity. They have broad applications in autoimmune diseases, transplant medicine, and inflammatory disorders and will continue to evolve as researchers address current challenges. The overall progress in preclinical investigations and clinical trials underscores the potential of CAR-Treg therapies to transform the management of conditions where precise, localized immunosuppression is needed, ultimately advancing patient care and setting the stage for future breakthroughs in immunotherapy.

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.