What are the different types of drugs available for Tregs cell therapy?

17 March 2025
Introduction to Tregs Cell Therapy
T regulatory cells (Tregs) are a distinct subset of CD4⁺ T lymphocytes that play a vital role in maintaining immune homeostasis. They help in suppressing excessive immune responses against self‐antigens and non‐harmful foreign antigens, ensuring that the immune system does not attack normal tissues. Tregs are characterized by high levels of CD25, low levels of CD127, and the expression of key transcription factors such as FOXP3, which is critical for their development and suppressive function. Their immunosuppressive capacity makes them an appealing target for therapeutic intervention in a variety of immune-mediated conditions, ranging from autoimmune disorders to transplant rejection and even neurodegenerative diseases.

Definition and Role of Tregs
Tregs are defined by their unique phenotypic and functional characteristics: they regulate immune tolerance by directly suppressing the proliferation of effector cells and by modulating the function of antigen-presenting cells (APCs). They act via multiple mechanisms including cell–cell contact-dependent inhibition, secretion of anti-inflammatory cytokines (e.g., IL-10, TGF-β, and IL-35), induction of cytolysis via granzyme-perforin pathways, and metabolic disruption by consuming IL-2 or generating adenosine through enzymes CD39 and CD73. Their ability to “dampen” overactive immune responses is crucial not only in preventing autoimmunity but also in reducing inflammatory damage during organ transplantation and in chronic inflammatory conditions.

Importance in Immunotherapy
The potential to harness Tregs for therapeutic purposes has spurred substantial interest in immunotherapy. While initial therapies using traditional immunosuppressive drugs broadly suppressed the immune system, Treg-based approaches offer the promise of highly targeted control of detrimental immune responses with fewer off-target effects. This is particularly significant for diseases where conventional therapies fall short, either by causing systemic immunosuppression or by failing to provide long-lasting benefit. In recent years, several innovative strategies have been developed to expand, stabilize, and direct the activity of Tregs in vivo or ex vivo, thus positioning them as “living drugs” in the field of adoptive cell therapy.

Types of Drugs Used in Tregs Cell Therapy
A variety of drug types have been explored for Treg cell therapy. These drugs are designed to either enhance the generation, activation, stability, or suppressive functionality of Tregs, or in some cases, to inhibit their suppressive activity when this is undesirable (for example, in cancer immunotherapy). The three major categories include small molecules, biologics, and gene therapy approaches. Each class offers distinct advantages, mechanisms of action, and potential applications in clinical settings.

Small Molecules
Small molecule drugs are low molecular weight compounds with high chemical flexibility, enabling them to cross cellular barriers and act on intracellular targets. They are frequently considered for Treg therapy because they are typically easier and more cost-effective to manufacture, can be administered orally, and often show favorable pharmacokinetic profiles.

1. Treg-Inducing Agents:
Several small molecules have been identified to enhance Treg induction or expansion. For example, high throughput screening strategies using cell-based assays have identified a number of FDA-approved drugs that selectively increase the number or activity of Tregs. Agents like rapamycin (sirolimus) have been widely studied for their ability not only to inhibit conventional T-cell proliferation but also to promote the expansion and stability of Tregs. Rapamycin works by inhibiting the mTOR pathway, which is crucial for cell growth and metabolism, thus favoring the differentiation of Tregs over effector T cells. Furthermore, retinoic acid is another compound that has shown promise in increasing Treg numbers, potentially by modulating TGF-β activity and influencing the transcription of FOXP3. Statins, typically used for lowering cholesterol, glucocorticoids, and even certain antioxidants (for example, curcumin and quercetin) have also been reported to have Treg-inducing capabilities through conditioning of antigen-presenting cells toward tolerogenic profiles.

2. Controlled Release Formulations:
To overcome limitations of short half-lives and off-target effects, controlled release microparticles have been under development. These formulations are designed to locally deliver cytokines and drugs such as IL-2, TGF-β, and rapamycin directly to the site where Treg expansion is desired. Controlled release approaches enable a predictable and sustained release that maximizes the recruitment and function of Tregs while limiting systemic side effects. Additionally, recent research into microparticle-assisted Treg therapy incorporates the use of IL-33 matrix-bound vesicles to enhance Treg proliferation and tissue repair during inflammatory events, such as tissue injury and fibrosis.

3. Metabolic Modulators:
Tregs have distinct metabolic demands compared to conventional effector T cells. Some small molecules can modulate metabolic pathways to favor Treg survival and function. For example, agents that modify glycolysis and fatty acid oxidation can impact the energy metabolism of Tregs, thereby promoting their maintenance under inflammatory conditions. D-mannose and 2-deoxy-D-glucose (2-DG) have been investigated for their roles in influencing the balance between Treg survival and Teff cell proliferation by modulating glucose metabolism.

4. Other Novel Small Molecules:
Emerging studies are investigating additional small molecules that interface with signals specific to Treg cell receptors such as TNF receptor 2 (TNFR2). Antibodies and small-molecule agonists targeting TNFR2 have been shown to stimulate Treg expansion with minimal systemic toxicity due to the restricted expression pattern of TNFR2. Moreover, compounds that inhibit negative regulators (such as MEK, ERK, and AKT1/2 inhibitors) might be used to modulate Treg activation in contexts where excessive suppression is deleterious, such as in cancer therapies.

Overall, small molecules offer the advantage of ease of delivery and the potential for oral administration, rapid onset of action, and cost-effectiveness compared to more complex biologics. Their flexibility in design allows for fine-tuning of Treg functions and targeted modulation of specific pathways critical to Treg biology.

Biologics
Biologic drugs are large, complex molecules, typically proteins or monoclonal antibodies, designed to precisely modulate immune pathways. They are becoming increasingly important in either enhancing Treg activity or, conversely, in limiting their suppressive function in diseases such as cancer.

1. Cytokine-Based Biologics:
Cytokines such as interleukin-2 (IL-2) are fundamental in the biology of Tregs. Low-dose IL-2 therapy has emerged as a promising biologic approach for Treg expansion because Tregs constitutively express high levels of CD25, the IL-2 receptor α-chain. Several clinical trials have demonstrated that low doses of IL-2 can selectively expand Treg populations, enhancing their suppressive function without undesired activation of effector T cells. Additionally, IL-2 muteins—genetically modified forms of IL-2 with reduced affinity for non-Treg cells—are under development to further enhance Treg specificity and reduce adverse effects. Furthermore, combinations of cytokines with other agents (such as TGF-β) in biologic formulations have been used to drive Treg differentiation ex vivo before adoptive transfer.

2. Monoclonal Antibodies:
Monoclonal antibodies targeting immunomodulatory molecules are now central to many immunotherapy approaches. For Treg cell therapies, antibodies that modify the activity of co-stimulatory or co-inhibitory receptors, such as CTLA-4, PD-1, and ICOS, can enhance or stabilize Treg function. For example, CTLA-4 is crucial for Treg-mediated suppression, and biologics designed to mimic or enhance CTLA-4 function (such as abatacept) may bolster the regulatory capacity of Tregs. Moreover, certain patents describe using antibodies directed against CD80/CD86 to encourage the generation of Treg cells in vitro as well as in vivo. Some biologics are also designed to deplete Tregs from the tumor microenvironment by targeting surface markers selectively expressed on Tregs (e.g., CCR8) as a means to counteract their immunosuppressive effect in cancer treatments.

3. Fusion Proteins and Antibody-Drug Conjugates (ADCs):
Fusion proteins that combine active cytokine domains with targeting moieties—such as antibody-cytokine fusion proteins—are a novel class of biologics being studied to deliver stimulatory signals directly to Tregs. In the tumor microenvironment, these fusion proteins can selectively target Tregs or modulate their function to enhance anti-tumor immunity. Additionally, antibody-drug conjugates have been formulated to deliver cytotoxic payloads specifically to Tregs in the context of cancer, with the aim of depleting them selectively in order to restore anti-tumor immune responses.

4. Treg-Enhancing Biologics for Non-Immunosuppressive Applications:
Some biologics are specifically being designed to enhance Treg function in diseases beyond traditional autoimmune conditions. For example, the biotechnology company Coya Therapeutics is developing Treg-enhancing biologics for neurodegenerative diseases. Their approach involves modulating Treg function to suppress neuroinflammation and protect against disease progression, as seen in their Phase I clinical trial initiatives for Alzheimer's Disease and Amyotrophic Lateral Sclerosis (ALS). These biologics may be formulated as cytokine combinations or fusion proteins that simultaneously promote Treg expansion and inhibit pro-inflammatory pathways in the central nervous system.

Biologics, being highly specific and generally exhibiting favorable pharmacodynamics, can provide precise modulation of the immune response. However, due to their large size, they are usually administered via injection and require complex manufacturing processes, which can increase overall costs.

Gene Therapy Approaches
Gene therapy represents a powerful set of techniques that allow for the modification of Tregs to improve their function, specificity, and stability. By directly altering the genome of Tregs, it is possible to create cells with enhanced therapeutic properties that can be used in adoptive cell therapy. In Treg cell therapy, gene therapy approaches include both the genetic engineering of autologous Tregs and the transduction of conventional T cells to induce a regulatory phenotype.

1. Viral Vector-Mediated Gene Transfer:
One of the earliest strategies in gene therapy with Tregs involved using retroviral or lentiviral vectors to overexpress FOXP3 in conventional T cells, thereby converting them into Treg-like cells with suppressive function. Overexpression of FOXP3 must be controlled in a way that is independent of T-cell activation states; for instance, using the EF1α promoter provides a stable expression level that does not fluctuate with activation, which is important for maintaining Treg phenotype and function. In addition to FOXP3, viral vectors have been used to express chimeric antigen receptors (CARs) in Tregs. CAR-Tregs are engineered Tregs that express antigen-specific receptors that redirect their trafficking and functionality to target tissues, such as transplanted organs or inflamed tissues in autoimmune diseases.

2. CRISPR/Cas9 and Gene Editing:
More recently, gene editing tools like CRISPR/Cas9 have been applied to Treg therapy to precisely knock out or knock in genes that modulate Treg function. This allows for the removal of endogenous T cell receptors (TCRs) to prevent graft-versus-host disease or to eliminate immunogenic elements such as HLA molecules to create off-the-shelf Treg products. Moreover, CRISPR/Cas9 can be used to knock out receptors for proinflammatory cytokines (e.g., IL-6 or IL-12 receptors) to stabilize the suppressive phenotype of Tregs in inflammatory environments. These gene editing strategies offer the possibility of tailoring Treg cell products that are more resilient, functionally stable, and specific to their targets. Gene-edited Tregs not only promise improved persistence in vivo but also reduce the risk of converting into pro-inflammatory cellular phenotypes.

3. Synthetic Biology and Chimeric Receptors:
Advances in synthetic biology have allowed the development of next-generation CAR Tregs. These engineered cells express synthetic receptors that combine antigen recognition domains with intracellular signaling motifs optimized for Treg activation and suppressive function. CAR-Tregs have been designed to target specific antigens, such as those expressed on donor grafts in transplantation or on inflamed cells in autoimmune diseases, thereby ensuring that their suppressive activity is localized and antigen-specific. Additionally, gene modification techniques allow for the integration of suicide genes into Tregs, which provide a safety switch to eliminate the cells if adverse events occur. This adds an extra layer of control in clinical applications, enhancing the overall safety profile of gene-modified Treg therapies.

4. Novel Gene Editing Platforms:
In addition to CRISPR/Cas9, other gene editing platforms such as TALENs and adenoviral vectors are being explored to modify Tregs. These platforms aim to increase the efficiency and precision of gene delivery, while minimizing off-target effects and potential immunogenicity. The choice of gene editing platform often depends on the desired modification and the clinical application in question, whether it is for creating antigen-specific Tregs or achieving a broader enhancement of Treg stability and function.

Gene therapy approaches to Treg modulation are among the most exciting areas of current research, with the potential to overcome many of the limitations of traditional drug therapies. Despite challenges in manufacturing and regulatory approval, these strategies hold promise for producing long-lasting, highly specific, and robust Treg cell products for diverse therapeutic applications.

Mechanisms of Action
Understanding the mechanisms of action is crucial for developing drugs that either upregulate or downregulate Treg function. In Treg cell therapy, drugs can be aimed at modulating Treg activity, enhancing their proliferation, or—in cases where Tregs are undesirable, such as in some cancers—inhibiting their suppressive function. The intricate interplay of these mechanisms is key to achieving therapeutic efficacy.

Modulation of Tregs Activity
Drugs in Treg cell therapy often work by modulating the activity of Tregs, ensuring they are sufficiently activated to exert their immunosuppressive functions. For example, low-dose IL-2 biologics specifically target CD25-expressing Tregs, leading to an increase in their activity without significantly affecting other immune cells. This targeted approach enhances the regulatory function of Tregs, thereby attenuating autoimmune inflammation. Similarly, gene therapy approaches that involve overexpression of FOXP3 help in “locking in” the Treg phenotype, thereby ensuring that the cells persist in their suppressive role over time.

Enhancement of Tregs Proliferation
Many drugs work primarily by enhancing Treg proliferation. Small molecules such as rapamycin, retinoic acid, and statins prime Treg expansion by influencing intracellular signaling pathways like mTOR, which is critical for cell growth. Such molecules not only promote Treg proliferation but often also stabilize their suppressive phenotype. In addition, controlled-release formulations delivering cytokines such as IL-2 or TGF-β locally can create microenvironments that are conducive to robust Treg expansion. Viral vector-mediated gene therapies and CRISPR-based modifications further enhance proliferation by allowing for genetic constructs that favor cell cycle progression in Tregs without leading to uncontrolled expansion.

Inhibition of Tregs Suppression
In some clinical settings, such as cancer immunotherapy, it is desirable to inhibit Treg suppression to allow effective anti-tumor responses. Here, small molecules that interfere with Treg signaling pathways can be used to block their suppressive effects. For instance, inhibitors of MEK, ERK, or AKT1/2 can suppress the activation of resting Tregs, thereby reducing their ability to inhibit effector T cells. In a tumor setting, strategies to block Treg-associated suppressive molecules, such as CTLA-4 or PD-1, have been investigated using monoclonal antibodies, leading to decreased Treg function and enhanced anti-tumor immunity. Moreover, antibody-drug conjugates targeting markers unique to Tregs in the tumor microenvironment (like CCR8) aim to selectively deplete these cells, thereby lifting their brake on effector cell activity.

Collectively, the mechanisms by which drugs operate in Treg therapy are multifaceted. Some agents modulate intrinsic signaling pathways, others deliver external cytokine signals, and still others alter the cell-surface receptor expression profile—all ultimately aiming to recalibrate the immune response in favor of tolerance or robust immunity.

Clinical Applications and Research
The translation of Treg-based drugs into clinical practice spans a wide range of indications. Treatments that enhance Treg function have been mainly focused on autoimmune diseases and transplantation tolerance, while strategies that inhibit Treg activity are prominent in the oncology field. The following sections discuss the current clinical trials, approved therapies, and the future directions driving further research into Treg modulation.

Current Clinical Trials
Current clinical trials have primarily focused on adoptive Treg cell therapies in conditions such as type 1 diabetes, graft-versus-host disease (GVHD), and solid organ transplantation. For example, several phase I and phase II studies are investigating the therapeutic potential of low-dose IL-2 formulations for the expansion of Tregs in vivo, which has shown promising results in terms of safety and efficacy. In parallel, CAR-Treg strategies are under development, especially targeting transplant antigens in kidney and liver transplantation, to promote localized immune tolerance. Cell therapy trials using autologous Tregs have also begun in the context of neurodegenerative diseases. Coya Therapeutics, for instance, is initiating a Phase I clinical trial for Treg-enhancing biologics aimed at treating neurodegenerative conditions like Alzheimer’s disease and ALS, indicating a broader potential application beyond conventional autoimmune indications.

Moreover, clinical trials are exploring the combination of adoptive Treg therapy with gene-modified Tregs to address limitations such as cell persistence and antigen specificity. These trials are drawing on the successes noted in early-phase studies that demonstrated the safety of adoptively transferred Tregs, alongside encouraging signals of efficacy such as improved immune tolerance and reduced inflammatory markers in patients.

Approved Therapies
Although many Treg-based therapies remain experimental, certain therapies that involve cytokine-based biologics have seen regulatory approval. Low-dose IL-2, for instance, has been approved for use in specific auto-inflammatory conditions, reflecting its role in selectively expanding Tregs. Additionally, the success of cell-based immunotherapies in oncology (e.g., checkpoint inhibitors) has paved the way for exploring Treg-targeted interventions, although this approach tends to aim for the depletion rather than the expansion of Tregs. At present, while several biologic and gene therapy strategies are in late-stage clinical development, the direct use of Treg-based cell therapies in routine clinical practice has not yet been widely adopted. Ongoing trials and regulatory assessments continue to evaluate safety, dosing, and long-term outcomes to ensure a balance between efficacy and the risk of systemic immunosuppression.

Future Research Directions
Future research in Treg cell therapy is multifaceted. On one front, there is significant promise in the areas of engineered Tregs via gene therapy; advances in CRISPR/Cas9 and viral vectors promise to create Tregs with precise antigen-specificity and improved stability. In the field of small molecules, continued high-throughput screenings and controlled release formulations will likely yield next-generation compounds that can either enhance or inhibit Treg activity as needed in various diseases. Biologics development is expected to focus on multifunctional fusion proteins and refined monoclonal antibodies that can modulate Treg function with heightened precision.

Researchers are also investigating combinatorial approaches in which adoptive cell therapy is complemented by small molecule adjuvants or cytokine-based biologics to improve the in vivo expansion, persistence, and homing efficiency of Tregs. These combinations are particularly attractive for conditions like autoimmune diseases or transplant rejection, where durable immune tolerance is required over long periods. The exploration of biomarkers and TCR repertoire analysis in clinical trials will help refine dosing regimens and optimize patient selection for Treg-based treatments.

Furthermore, advanced process development techniques, including automated, GMP-compliant manufacturing of Treg products, are continuously evolving to overcome the challenges associated with scaling up cell therapies while maintaining quality and reproducibility. Future research may also harness the synergy between Treg therapies and other forms of immunomodulation, such as checkpoint inhibitors or antigen-specific vaccination strategies, to fine-tune immune responses in a personalized manner. This integrated approach holds particular promise for diseases with complex immunopathogenesis, where a single therapeutic intervention may not be sufficient.

Conclusion
In summary, the landscape of drugs available for Treg cell therapy is richly varied and continually expanding. Beginning with small molecules, a new generation of compounds—such as rapamycin, retinoic acid, statins, and metabolic modulators—has been shown to enhance Treg induction, proliferation, and functional stability. These small molecules offer the advantages of oral bioavailability, ease of manufacture, and cost-effectiveness, while controlled-release formulations further optimize their localized action. Biological agents, on the other hand, represent a more targeted approach; intravenously administered cytokines like low-dose IL-2, IL-2 muteins, and monoclonal antibodies that either stimulate or deplete Tregs have provided promising clinical signals. Fusion proteins and antibody-drug conjugates extend the reach of biologics further by delivering their therapeutic payloads directly to the desired cell populations. Gene therapy approaches, including viral vector-mediated gene transfer and advanced genome-editing techniques such as CRISPR/Cas9, are paving the way for next-generation adoptive Treg cell therapies. These innovative strategies allow for the creation of antigen-specific CAR-Tregs, stable FOXP3-expressing regulatory cells, and even “off-the-shelf” universal Treg products, which hold immense promise for both enhancing tolerance in transplantation and mitigating autoimmune disease activity.

From a mechanistic standpoint, drugs modulate Tregs by directly altering their activation pathways, encouraging proliferation through cytokine and metabolic support, or by inhibiting their suppressive functions when necessary. This mechanistic diversity ensures that various diseases, whether they are autoimmunity, transplant rejection, or cancer, can potentially be addressed by fine-tuning the Treg population in vivo. The clinical trials conducted so far have confirmed the safety and feasibility of both biologic and cell therapy approaches focused on Tregs. Although no Treg therapy has yet been universally approved, ongoing studies and regulatory trials continue to refine dosing, administration routes, and manufacturing techniques to eventually bring these therapies into routine clinical use.

Overall, universal challenges such as cell expansion, stability, and specificity remain active areas of investigation. Future research efforts will likely converge on combinatorial treatment regimens that integrate small molecule drugs, biologics, and gene therapy modifications, thereby creating a synergistic effect that optimizes Treg function in both the induction of immune tolerance and the restoration of homeostasis. This multi-pronged, general-specific-general approach to Treg cell therapy emphasizes the promise of these modalities across the spectrum of immune-mediated conditions and marks an exciting frontier in clinical immunotherapy.

In conclusion, the different types of drugs available for Tregs cell therapy emerge from a diverse portfolio of small molecules, biologics, and gene therapy approaches. Each category offers unique advantages in terms of specificity, administration, and mechanism of action. Small molecules are valued for their ease of use and potential for oral administration, biologics for their targeted and robust immunomodulatory effects, and gene therapy for its ability to engineer highly specialized and stable Treg populations. The overall therapeutic strategy revolves around either enhancing Treg expansion and activity to promote immune tolerance or, conversely, inhibiting Treg suppression to allow effective anti-tumor immunity. As the field continues to advance through both preclinical innovations and clinical trials, it is expected that these diverse drug classes will be integrated into personalized treatment regimens designed to restore immune balance with minimal side effects, thereby heralding a new era in immunotherapy.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成