Introduction
Regulatory T cells (Tregs) are a population of T cells that are specialized for suppressing the activation and expansion of aberrant or overreactive lymphocytes1. Tregs comprise 5 - 10% of the CD4+ T cell population2 and are characterized mainly by the transcription factor (and lineage marker) forkhead box protein P3 (FOXP3)3,4. Other important markers include high surface expression of CD25 (the IL-2 receptor α-chain) and low expression of CD127 (the IL-7 receptor α-chain)5. Therefore, phenotypically, Tregs can be defined as CD4+CD25+CD127low T cells and this combination of markers can be used to identify them from other cells6.
The role of Tregs in immune regulation and autoimmune diseases became clear after the discovery of intracellular FOXP3 and certain Treg surface markers (e.g., CD25, or IL-2 receptor α). The importance of FOXP3, in particular, was better understood after it was discovered that mutations in the gene encoding this transcription factor led to a severe autoimmune polyendrocrine syndrome7. Mutations in the gene encoding FOXP3 lead to immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX) - a syndrome in which patients manifest features of autoimmunity such as enteropathy, endocrinopathy, and dermatitis as well as other noninfectious, autoimmune features such as autoimmune hepatitis8. This discovery established the importance of Tregs in immune tolerance and additionally opened the door for the idea of adoptive transfer of Tregs as therapy for various autoimmune diseases.
In vivo, Tregs originate either through direct differentiation in the thymus or they develop from conventional CD4+ T cells in the periphery9. In the thymus, immature CD4 single-positive thymocytes that receive T cell antigen receptor (TCR) signals and have high affinity for self-peptides differentiate into FOXP3+ Treg cells (thymic Tregs or tTregs, also known as natural Tregs or nTregs)10. In the periphery, conventional CD4+ T cells encounter antigens (mostly non-self antigens such as allergens, food, and the commensal microbiota) in the presence of specific factors such as TGF-β, which leads to development of peripheral Treg cells (pTregs)9,10. Despite the difference in origin, tTregs and pTregs are similar in function and there is currently no protein marker to differentiate them in humans9.
Tregs suppress immune function through both cell-contact mechanisms and the secretion of inhibitory cytokines9. They target T cells directly or indirectly by modulating antigen presenting cells (APCs)11. Direct cell-contact mechanisms involve the expression of negative regulatory cell surface receptors such as cytotoxic T lymphocyte antigen 4 (CTLA-4)11. Another mechanism of action for Tregs is through the release of anti-inflammatory soluble mediators released such as IL-10, TGFβ and IL-35 as well as through consumption of IL-2 (acting as an IL-2 sink)9–11. Tregs also act by promoting the emergence of other immunosuppressive cell populations such as Tregs with different specificities (such as type 1 T helper cell (TH1 cell)-like Tregs) and T regulatory 1 (Tr1) cells11. For a detailed overview of how Tregs function and act, we direct the reader to an excellent review by Josefowicz et al10. Understanding these mechanisms of action has inspired several approaches to harness the potential of Tregs for adoptive cell therapy.
Given their immune suppressive functions and therapeutic potential, there has been an increasing interest in adoptive transfer of Tregs for treatment of autoimmune diseases and prevention of graft-versus-host disease (GvHD) and transplant rejection12–16. Key factors to consider for successful use of Tregs as a cell therapy include their ability to cause immunosuppression and their survival, stability, and specificity once administered to the patient6. These are all essential factors to consider during the pre-clinical studies and scale-up and optimization of clinical manufacturing of Tregs, as well as while testing them in clinical trials. While progress has been made in manufacturing Tregs over the past decade with promising clinical trial results, a successful Treg cell therapy product has yet to undergo biologics license application and reach the market. In this review, we discuss the clinical-grade manufacturing strategies that have been explored, highlight the key clinical trials that have taken place and provide a brief outlook of future technologies.
Clinical-Grade Manufacturing of Tregs
Many studies have demonstrated the potential for Tregs in clinical practice while using current good manufacturing processes (cGMP) and clinical-grade reagents when possible (Table 1). These various manufacturing techniques have primarily used peripheral blood mononuclear cells (PBMC) or umbilical cord blood (UCB) as the source of Tregs. The technologies to isolate, purify and expand Tregs have advanced over the past decade and newer approaches continue to be explored. Here, we will review the key cGMP methods employed thus far by discussing the details of the starting materials and the various tools employed in isolating, purifying, and expanding the Tregs.
Table 1:
Summary of Treg cell therapy manufacturing methods
| Source of cells; type of Tregs | Summary of protocol | Product specification/ Release criteria | Product purity (method: flow cytometry) | Reference |
|---|---|---|---|---|
| PBMC ➔ iTregs | Apheresis product was used to obtain CD4+25− T cells with immunomagnetic selection; cells were stimulated with anti-CD3 mAb-loaded, irradiated transduced K562 (KT64/86) in IL-2, rapamycin and TGFß. Tregs were restimulated with KT64/86 cells (t = 7 days). Cultures were harvested and infused (t = 14 days). | Viability ≥70%, CD4+CD25+ ≥60%, CD4−/CD8+ cells ≤10%, Gram stain with “no organisms seen,” and endotoxin <5 EU/kg. | CD4+CD25+: 95% [86-99%] Foxp3+ ~ 60% |
MacMillan et al.18 |
| PBMC ➔ tTregs | Cryopreserved leukapheresis products were used to isolate and expand Tregs using immunomagnetic beads and IL-2, TGFβ, and rapamycin (t = 0 and 7 days). Cultures were evaluated (t = 14 days) and final testing completed (t = 21 days). The cellular product was harvested (t = 21 days) and expansion beads were removed before infusion. | Negative aerobic, anaerobic and fungal sterility culture, negative mycoplasma and negative gram stain; <5.0 EU/kg endotoxin; >70% viable; >70% CD4+ CD25+; <10% CD8+ and CD19+; <3000 Exp-Act beads/10E8 cells. | CD4+CD25+: ≥ 99 % Foxp3+ > 80% |
Mathew et al.20 |
| PBMC ➔ tTregs | Apheresis product was used to isolate and purify Tregs using FACS. Purified Tregs stimulated with anti-CD3 and anti-CD28 beads (t = 0 and 9 days) in IL-2 and deuterated glucose. Cells were harvested (t = 14 days) after culture initiation and stimulating beads were removed. | FOXP3% >60%; CD4% >95%; CD8% <5%; Viability >85%. | Foxp3+ > 93% | Chandran et al.22 |
| PBMC ➔ iTregs | Leukapheresis was used to obtain donor and recipient lymphocytes. Recipient lymphocytes were co-cultured with irradiated donor lymphocytes in the presence of anti-CD80/86 antibodies (no IL-2) to obtain inducible donor antigen-specific monoclonal Tregs. | Cell viability >80% and CD4+CD25+Foxp3+ cell count >1 x 106/kg. Absence of bacteria, fungi, mycoplasma, and endotoxins. | CD4+CD25+Foxp3+ cell count: 3.39 ± 2.12 x 106/kg (range: 0.23 to 6.37 x 106/kg) | Todo et al.26 |
| PBMC ➔ tTregs | Apheresis products were used to isolate Tregs with CD8+ reduction and CD25+ enrichment with immunomagnetic separation. Enriched cells were activated with anti-CD3/CD28-coated beads in rapamycin and IL-2 (t = 4-6 days; replenished every 2-3 days). At final harvest, beads were removed from pooled cells. | Positive for CD4, CD25 and FOXP3; ≥ 60% CD4+CD25+ FOXP3; ≤ 10% CD8+; ≤100 beads per 3x106 cells; ≥ 70% viability; ≥ 70% recovery; ≥ 60% suppression; Sterility-no growth; Endotoxin ≤175IU/mL; Mycoplasma - not detected. | CD4+CD25+: 91.3% ± 2.33 CD4+CD25+Foxp3+: 90.7% ± 2.19 |
Safinia et al.21 |
| PBMC ➔ tTegs | Leukapheresis product was used to isolate Tregs using immunomagnetic separation; stimulated with aniti-CD3/anti-CD28-coated beads. Cells were expanded (t = 7 or 12 days) with IL-2 and rapamycin (t = 0 or 2 days after isolation; replenished every 2-3 days). Beads removed at the end. | CD4+CD25highCD127low FOXP3+ | CD4+CD25highCD127low FOXP3+: 84.1% [77.7-91.8] | Thiel et al.24 |
| PBMC ➔ tTregs | Apheresis products were used to isolate Tregs using FACS. Tregs were activated with anti-CD3/anti-CD28–coated beads in the presence of IL-2 and rapamycin (t = 7 days). Cells were resuspended and fresh media and IL-2 were added every 2-3 days. Cells were restimulated (t = 9 days) with fresh anti-CD3/anti-CD28–coated beads. | Viability ≥ 85%; ≥ 60% FOXP3+ and ≥ 95% CD4+ cells; ≤ 5% CD8+ cells, <100 beads per 3 x 106 cells, and endotoxin ≤3.5 EU/ml; negative for mycoplasma, anaerobic and aerobic bacteria, gram stain, fungal culture and KOH exam. | CD4+ CD127low/−CD25+ > 95% Foxp3+: ≥ 76 % |
Bluestone et al.23 |
| PBMC ➔ tTregs | Apheresis products were used to isolate Tregs using combined immunomagnetic/FACS technique. Isolated cells were activated and expanded in the presence of IL-2, and anti-CD3/anti-CD28 beads. Tregs were cultured until the required number was achieved, but for no longer than 2 weeks (median t =10 days; range t = 7-14 days). | FoxP3 expression > 90%, passed interferon (IFN)-g suppression assay, and negative microbiological tests (no genetic material of hepatitis B virus, hepatitis C virus, or HIV and no microbial contamination in the culture supernatants) | Foxp3+: 93% [90-97] | Marek-Trzonkowska et al.28 |
| UCB ➔ tTregs | HLA matched UCB units were thawed, processed, and enriched for CD25+ using immunomagnetic selection. CD25+ cells were co-cultured with CD3/28 co-expressing beads in the presence of IL-2 before culture was harvested (t = 14 days). | Viability ≥70%, Endotoxin <5EU/kg, Gram Stain: No organism seen, Mycoplasma: Negative, Sterility: No organism at time of infusion, CD4−CD8+ cells: <10 %, CD4+CD25+ cells > 60% and <100 beads per 3x106 cells |
CD4+CD25+CD127lo: 90% [86-93%] | Kellner et al.27 |
| UCB ➔ tTregs | HLA matched UCB units were enriched for CD25+ cells with immunomagnetic selection; then stimulated with anti-CD3 mAb loaded KT64/86 artificial APCs (aAPCs). Cultures were supplemented with IL-2 (t = 3 days). Cultures were washed and restimulated with frozen/thawed, anti-CD3 mAb-loaded KT64/86 aAPCs (t = 12 days). | Viability ≥ 70%, CD4+CD25+ purity ≥ 60%, < 10% CD4−/CD8+ cells, negative gram stain, and low endotoxin (≤5 EU/kg) | CD4+CD25+: 86% [62-97] CD4+FoxP3+CD127−: 64% [31%−96] |
Brunstein et al.25 |
| UCB ➔ tTregs | HLA matched UCB units were enriched for CD25+ cells with immunomagnetic selection; isolated cells were cultured with anti-CD3/anti-CD28 monoclonal antibody-coated beads (t = 18 days) and cultures were supplemented with IL-2 (t = 3 days). | Viability ≥ 70%, CD4+CD25+ purity ≥ 60%, < 10% CD4−/CD8+ cells, anti-CD3/anti-CD28 mAB bead count < 100 per 3 x 106 cells, gram stain with “no organisms,” and endotoxin < 5 EU/kg | CD4+CD25+: 97% [88-99] CD4+FoxP3+CD127−: 87% [78-95] |
Brunstein et al.17 |
The major source of Treg products generated under cGMP is PBMCs collected by apheresis (Table 1). PBMCs are particularly important in manufacturing autologous products as they are easily accessible from patients. Apheresis-derived PBMCs are collected aseptically using a closed system and are easy to further manipulate in downstream processing. As Tregs comprise only 5 - 10% of the CD4+ T cell population3,4, obtaining a pure population of Tregs from PBMCs requires multiple steps of isolation and expansion. To achieve this, various techniques have been developed and improved over the years – some of these will be discussed below.
UCB is the other major source of Tregs manufactured under cGMP for clinical trials (Table 1). While relatively fewer clinical trials have used UCB as the source of Tregs, the potential of third-party UCB units to be used as an allogeneic source makes them very attractive. In addition, UCB is an attractive source because of the relatively high proportion of circulating tTregs and paucity of T memory cells in the term fetus compared with adult peripheral blood17. Regardless, UCB also requires advanced techniques for isolating and expanding Tregs to achieve high enough dose for clinical applications. Our group and others have shown that Tregs must undergo multiple rounds of expansion in vitro to generate enough cells for a clinically relevant dose. These techniques of isolation and expansion will be discussed below.
The majority of Tregs derived from PBMCs and UCBs that have entered clinical trial have been tTregs. However, the need for high Treg doses for reliable disease control coupled with the low tTreg frequency and relatively poor proliferation of tTregs compared to T-conventional cells (Tcons) in ex vivo culture, have inspired exploring other options. CD4+ Tcons derived from PBMCs can be driven to a Treg phenotype and function in vitro in the context of TGFβ, IL-2 and rapamycin, leading to the development of iTregs. Our group has recently published the results of the first-in-human phase 1 trial of iTregs for GvHD prophylaxis in HLA-matched siblings18. The unique methods employed in inducing and expanding these cGMP grade iTregs will also be discussed below.
The initial important step in manufacturing Tregs from both PBMCs and UCB is cell selection or sorting. While various methods have been explored in the past19, currently the most common method of isolating Tregs is using immunomagnetic selection performed in a closed system. CD4+CD25+ Tregs are isolated using CD25hi as the main surface marker for positive selection20. This involves incubation of the starting cell product with a cGMP-grade anti-CD25 conjugated to a magnetic nanoparticle followed by exposure to a strong magnetic field, allowing retention of CD25 rich cells that will be separated from the rest of the product. Some groups also perform CD8+ reduction through negative selection21. In addition to immunomagnetic selection, cGMP-grade FACS has also been used to isolate Tregs22. Cell surface markers such as CD4 and CD25 are used for positive selection of CD4+CD25+ Tregs; some groups also use CD127 for negative selection22,23. While direct comparison of immunomagnetic selection versus FACS based sorting in clinical trials have not been performed yet, cGMP-grade FACS sorting is still in its early stages compared to established immunomagnetic selection.
Immunomagnetic selection is also used for iTreg manufacturing from PBMCs, but the process is modified from the above steps to enrich CD4+CD25− cells (the starting cells that are induced into iTregs). The method involves depleting CD25+ cells (including tTregs) from PBMCs with anti-CD25 magnetic beads followed by purifying CD4+ cells from CD25− PBMCs using anti-CD4 magnetic microbeads18. The CD4+CD25− cells are then stimulated/activated before they go through the process that converts them into iTregs.
Once tTregs or CD4+CD25− cells are isolated from the starting material, they are expanded in culture by stimulating through TCR and co-stimulatory signals. The two most common approaches for stimulating/activating these cells have been antibodies bound to beads or cell-based artificial antigen-presenting cells (aAPCs). Anti-CD3/anti-CD28–coated microbeads are widely used despite the requirement of removing the magnetic beads prior to infusion21–24. The most widely used aAPCs to activate Tregs are K562 cells expressing CD86 and CD64 (KT86/64) that are loaded with anti-CD3 mAb18,25. These cells are irradiated prior to use and disappear from culture over time, hence do not require a removal step prior to infusion.
Expansion culture is critical to attain a higher number of cells, as the clinical dose required is much higher than the number of cells derived through standard isolation procedures. As in the expansion of other cell types used in cell therapy, IL-2 is one of the important cytokines used to expand Tregs20,24. The timing and frequency of IL-2 addition is variable among various groups, but in most cases, it is added in the first few days (0-3 days) and replenished as needed either every 2-3 days or on day 7 (Table 1). The immunosuppressive drug rapamycin is the other commonly used agent in expanding Tregs and is added to the media along with IL-223. Rapamycin, which is an inhibitor of cytotoxic T cells, works by suppressing proliferation of effector T cells while sparing Treg cells, resulting in the relative selective expansion of Tregs. For iTregs, the process of inducing the cells from CD4+25− T cells and then expanding them happens essentially simultaneously. The CD4+25− T cells are induced into Tregs (iTregs) through incubation with IL-2, rapamycin and TGFβ, as mentioned above18. While there are other cytokines and small molecules that may induce Treg phenotype, the combination of IL-2/rapamycin/TGFβ is the unique cocktail commonly used in both inducing and expanding iTregs.
In addition to manufacturing tTregs and iTregs through the process mentioned above, there is another unique approach to manufacturing Tregs. In this process, leukapheresis is used to obtain donor and recipient lymphocytes from PBMCs, and then the recipient lymphocytes are co-cultured with irradiated donor lymphocytes in the presence of anti-CD80/86 monoclonal antibodies without IL-2 or rapamycin26. Todo et al call these types of Tregs inducible donor antigen-specific monoclonal Tregs (different than iTregs mentioned above). In addition to the difficulty of obtaining sufficient number of recipient cells and possible contamination with leukemia cells, the final product has very low purity (the mean number of CD4+CD25+Foxp3+ T cells was only 24.8% of the CD4+ cells infused).
The cell culture media used in expanding Tregs have also been variable among groups, but X-VIVO 15 has been the most consistently used22–25,27. The extensive experience of using X-VIVO 15 in other cell therapy applications in a cGMP-compliant setting makes it a safe and reliable option. Other commonly used cGMP-compliant media include the TexMACS GMP media and the CellGro media21,24,28. While serum free X-VIVO 15 exists and is used in some cases, the supplementation of these various media with serum is one additional factor to consider while ensuring cGMP-compliance. Supplementation of the media with 2.5 to 10% human AB serum results in enhanced Treg function. The culture conditions for expanding Tregs in these media have consistently been the standard, 37°C and 5% CO2. Major limitations of current Treg manufacturing procedures using magnetic bead based approaches include less than 100% purity of the cellular therapy product and potential toxicities that might occur due to contaminating effector T cells. As shown in Table 1, the purity of the products is determined by measuring the percentage of CD4+CD25+ and/or Foxp3+ cells. While the percentage of CD4+CD25+ cells is mostly above 90%, the percentage of Foxp3+ cells could be as low as 60%. Another limitation of current manufacturing process is difficulty to meet target cell dose. While manufacturing Tregs with a dose ranging from 0.5-10 x 106/kg and as high as 3 x 108/kg have been reported, some studies report failure to meet the target dose17,23,25.
In comparing the manufacturing approaches employed, one of the big differences is the source material used to make the Tregs. As discussed above, using PBMCs and UCBs as source material have their own advantages and disadvantages. Tregs from PBMCs requires multiple depletion steps, although they have the advantage of being easily accessible from donors while the relatively high proportion of circulating Tregs in UCBs make them an attractive source. Additionally, the potential of third-party UCB units as an allogeneic source of Tregs is another advantage. Another difference in manufacturing steps is the type of agents used in the activation and expansion stage. While IL-2 is used in almost all approaches (except in the approach used to make inducible Tregs26), the use of rapamycin is not consistent across all approaches. Although rapamycin is known to suppress proliferation of contaminating effector T cells and hence improve purity of the product, it is not yet clear whether the use of rapamycin leads to a clinically significant improvement.
In addition to IL-2 and rapamycin, the process of manufacturing iTregs adds TGFβ which induces a regulatory phenotype in CD4+CD25− T cells. iTregs manufactured through this process had an average CD4+CD25+ expression of 95%, but the average Foxp3+ expression was close to 60%18. Manufacturing tTregs through the activation/expansion of CD4+CD25+ has been reported to produce Tregs with Foxp3+ expression as high as 93% (Table 1). While the iTreg manufacturing approach produces Tregs with high dose, this lower Foxp3+ expression will need improvement. Whether there will be significant differences in the clinical effectiveness of iTregs versus tTregs remains to be seen.
Several of the methods involved in manufacturing Tregs are similar to those involved with other immune cell therapies. However, the need to activate/stimulate Tregs through TCR and co-stimulatory signals such as anti-CD3/anti-CD28 microbeads or aAPCs adds to the complexity of the process. While the basic aspects of methods for expanding Tregs have been delineated, the ideal culture/cytokine cocktail and the frequency/timing of when these are added continues to be optimized. With further studies and clinical trials, the important variables that will lead to a highly potent, pure, and safe Treg product will continue to be identified.
Clinical Trials
There are many ongoing clinical trials that are trying to utilize the benefits of Tregs both in adoptive cellular therapy as well as through in vivo expansion strategies with low dose IL-2. According to clinicaltrials.gov, there are currently about 10 active clinical trials utilizing Tregs in adoptive cell therapy for various autoimmune diseases. A summary of the major clinical trials involving Tregs for adoptive cell therapy over the past decade is provided in Table 2. These clinical trials have focused on using Treg cell therapy in patients with a range of conditions including GvHD, transplant rejection, and autoimmune disorders. Most of these trials have been early stage trials (mainly phase 1) recruiting limited number of patients (<20). As noted with the variations in the approach to manufacturing, the clinical protocols for Treg cell therapy have also varied with differences, for example, in dosing and the end points investigated (Table 2). Despite these variations, the one common focus of these early clinical trials has been demonstrating the safety and tolerability of Treg adoptive immunotherapy. As shown in Table 2, most clinical trials included product specifications designed to ensure a safe product, including sterility testing, viability, Gram stain, Mycoplasma testing and cell phenotype determined by flow cytometry.
Table 2:
Summary of Clinical Trials using cGMP-grade Treg cells for therapy
| Patient characteristics | Treg cell product | Outcome | Reference (Listed by year) |
|---|---|---|---|
| 14 adults with high risk malignancy treated with reduced intensity conditioning and mobilized peripheral blood stem cells from HLA-identical sibling donors | Induced regulatory T cells (iTregs) (PBMC derived, prepared from human CD4+25− T cells cultured in IL-2, rapamycin and TGFß along with anti-CD3 mAb-loaded artificial antigen-presenting cells); dose of 3.0 x 106/kg, 3.0 x 107/kg, and 3.0 x 108 cells/kg. | No severe infusion toxicities with all patients achieving neutrophil recovery (median day 13). Circulating iTregs were detectable through day 14. iTregs can be delivered safely at doses as high as 3 x 108 cells/kg. | MacMillan et al.18 (2021) |
| 14 patients treated with relapsing-remitting multiple sclerosis | Autologous Tregs (PBMC derived, activated, and stimulated with anti-CD3/anti-CD28 beads and IL-2), either delivered intravenously (IV), dose 40 x 106 cells/kg or intrathecally (IT), dose 1.0 x 106 cells. | No severe adverse events were observed. Relapses were noted in five IV treated patients, while no patients in the IT group experienced a relapse. | Chwojnicki et al.32 (2021) |
| 104 adult patients who received living-donor kidney transplants; 38 of them received Treg cell therapy | Four out of the six clinical trials pooled in this study used Tregs manufactured at four different sites using various methods. | Treg cell therapy is achievable and safe in living-donor kidney transplant recipients; associated with fewer infectious complications, but similar rejection rates in the first year. | Sawitzki et al.29 (2020) |
| 11 recipients of living donor kidney transplant (compared with nine control patients from a corresponding reference group trial) | Autologous Tregs (PBMC derived, activated, and stimulated with anti-CD3/anti-CD28 beads, rapamycin, and IL-2); given seven days after kidney transplant, dose 0.5, 1.0, or 2.5-3.0 x 106 cells/kg. | No dose limiting toxicity observed. Stable monotherapy immunosuppression was achieved in 73% of patients, while reference group remained on standard dual or triple therapy. | Roemhild et al.33 (2020) |
| Nine adult patients who were enrolled while awaiting liver transplantation or 6-12 months posttransplant | Autologous Tregs (PBMC derived, activated, and stimulated with anti-CD3/CD28 beads, IL-2, and rapamycin); administered intravenously at either 0.5-1 x 106 cells/kg or 3-4.5 x 106 cells/kg. | Treg transfer was safe, transiently increased the pool of circulating Tregs and reduced anti-donor T cell responses. | Sanchez-Fueyo et al.30 (2020) |
| Nine adult recipients of living donor renal allografts that would not require hemodialysis during the first week following renal transplantation | Autologous Tregs (PBMC derived, activated, and stimulated with anti-CD3/CD28 beads, IL-2, TGFβ, and rapamycin); infused at doses of 0.5 x 109, 1.0 x 109, and 5 x 109 cells/recipient. | All doses of Treg therapy tested were safe with no adverse infusion related side effects, infections, or rejection events up to two years post-transplant. | Matthew et al.20 (2018) |
| Three kidney transplant recipients with inflammation on their 6-month surveillance kidney transplant biopsy | Autologous Tregs (PBMC derived, stimulated with anti-CD3 and anti-CD28 beads in media containing IL-2), delivered at doses of ~320 x 106 cells/recipient. | There were no infusion reactions or serious therapy related adverse events. The infused cells demonstrated persistence and stability. | Chandran et al.22 (2017) |
| 11 patients with advanced or high-risk lympho-hematopoietic malignancy: 22 identically treated controls | Allogeneic Tregs (derived from HLA matched UCB, stimulated with anti-CD3 mAb loaded KT64/86 artificial APCs in the presence of IL-2), at doses of 3-100 x 106 cells/kg. | No adverse events observed. Incidence of grade II-IV acute GVHD at 100 days was significantly lower (9% vs 45%, P = 0.05). Chronic GVHD at 1 year was zero in Tregs and 14% in controls. | Brunstein et al.25 (2016) |
| 10 adult patients early post-liver transplantation | Inducible donor antigen-specific monoclonal Tregs (PBMC derived, co-cultured with irradiated donor lymphocytes in the presence of anti-CD80/86 monoclonal antibodies) infused at a dose of 3.39 ± 2.12 x 106/kg, ranging from 0.23-6.37 x 106 cells/kg. | No significant adverse events were observed. All 10 recipients maintained stable graft function and seven patients successfully achieved weaning and completed cessation of immunosuppressive agents. | Todo et al.26 (2016) |
| 14 adult subjects with Type 1 Diabetes | Autologous Tregs (PBMC derived using FACS, activated with anti-CD3/anti-CD28–coated microbeads and cultured in the presence of IL-2 and rapamycin); dose of 0.05 x 108 to 26 x 108 cells. | No infusion reactions or cell therapy–related high-grade adverse events. A subset of the Tregs was long-lived, with up to 25% of the peak level remaining in the circulation at 1 year after transfer. | Bluestone et al.23 (2015) |
| Five adult patients with treatment-refractory cGvHD | Allogeneic Tregs (derived from PBMC, expanded in the presence of aniti-CD3/anti-CD28 beads, IL-2, and rapamycin); delivered at a mean dose of 2.4 x 106 cells/kg. | Two patients showed clinical response with improvement of symptoms, the others showed stable symptoms for up to 21 months. In four patients, increased counts of Treg were detectable and immunosuppressionwas reduced. | Thiel et al.24 (2015) |
| 10 children (aged 8-16 years) with recent-onset Type 1 diabetes (2 months since diagnosis) | Autologous Tregs (derived from PBMC, activated and stimulated in the presence of IL-2 and anti-CD3/anti-CD28 beads); delivered at a dose of 20 x 106 cells/kg. | Administration of Tregs was safe and tolerable. No toxicity was noted. A significant increase in percentage of Tregs in peripheral blood was observed. | Marek-Trzonkowska et al.28 (2012) |
| The first two patients with GvHD who received the treatment with ex vivo expanded Tregs (first-in-man clinical trial for Tregs) | Allogeneic Tregs (derived from PBMC, sorted with immunomagnetic separation and FACS, activated with anti-CD3/anti-CD28 beads and IL-2); delivered at a dose of 1 x 105 cells/kg. | Tregs led to significant alleviation of the symptoms and reduction of pharmacologic immunosuppression in the case of chronic GvHD; in the case of grade IV acute GvHD, the improvement was transient. | Trzonkowski et al.34 (2009) |
In general, the studies so far show that Treg adoptive cell therapy is safe and well-tolerated among the multiple types of patient groups studied. These studies have shown safe delivery of Tregs with doses ranging from 0.5-10 x 106/kg and as high as 3 x 108/kg, with no serious infusion related adverse events documented (Table 2). While the optimal Treg dose for clinical application is yet to be determined, the early safety studies with the high dose of Tregs pave the way for future efficacy studies. In addition, infused cell therapy products were shown to be persistent in circulation up to 1 year retaining their Foxp3+CD25+ Treg phenotype23. However, this study also showed that there was a rapid decline in the percentage of infused Tregs in circulation, with the majority of the Tregs being undetectable within 90 days. Moreover, long-term persistence has not been observed in many studies and this has been a limitation of other clinical trials which mostly report in vivo persistence of 14-30 days25,30. A possible solution for this could be combining Tregs with treatments that promote Treg expansion and persistence in vivo. An example of this is combining low-dose IL-2 with Tregs, which has shown promise in early clinical studies35.
While safety has been the main focus of these early trials, some of the results have indicated promising efficacy as Treg therapy resulted in low risk of GvHD25 and fewer infectious complications or transplant rejections post-kidney transplant20,29. The fact that some of these early trials showed efficacy is encouraging but phase 2 trials with larger number of patients will be required before broader conclusions can be made regarding the wider clinical applications of Treg immunotherapy.
In addition to the use of Tregs as stand-alone cell therapy agents, the role that Treg cell therapy could play in combination with other immunosuppressive agents is an exciting possibility. This is particularly important in patients receiving immunosuppressive drugs after organ transplantation. Current standard of care in organ transplantation such as liver or kidney requires putting patients on immunosuppressive drugs for many years and in some cases for lifetime. Tregs have the potential to be combined with existing immunosuppressive drugs to enhance immunosuppression with a goal to one day either reduce or completely avoid the need for immunosuppressive drugs post organ transplantation. One recent clinical trial has shown that patients receiving Treg cell therapy, and minimization of immunosuppression, can be successfully weaned within the first-year post-transplantation to monotherapy29. The results from other studies26,30 also indicate that there is enough evidence to encourage future trials that investigate the use of Tregs to facilitate the reduction or even the complete discontinuation of immunosuppressive medications following organ transplantation.
Overall, the clinical studies so far show that Treg cellular therapy products have a favorable safety profile along with early signs of therapeutic benefit. With early focus on GvHD and transplant rejection, these areas may be where the first successful implementation of Treg cellular therapy occurs. Factors influencing the long-term efficacy of Treg therapy are still under investigation31. Turning advances in the field into clinical reality in the later phases of clinical trials will require overcoming some remaining challenges. These challenges include further understanding Treg biology to ensure efficient and targeted approaches are used to isolate, induce, and expand Tregs, as well as optimizing the variables involved in manufacturing and administering the Tregs. In addition to optimizing the manufacturing methods, well-designed clinical trials will be crucial. Finally, given the complexity of the patients and the diseases targeted by Tregs, the timing and frequency of administration of the Treg cell therapy products may play a role in enhancing efficacy. Therefore, the success of Treg cellular immunotherapy will depend on the effective collaboration of various teams to design and implement future clinical trials.
Future Directions
As the manufacturing methods and clinical trials discussed above are moving forward, there are also exciting preclinical studies with novel ideas that are ongoing and promise even more advanced ways of using Tregs in clinical applications. Most of these novel ideas involve improvements in manufacturing Tregs, including utilizing new sources as well as engineering Tregs to make them more specific and target-focused. While new Treg sources such as discarded human thymus tissue, routinely removed during pediatric cardiac surgery36,37, provide additional options for large-scale Treg manufacturing, it is the engineering of Tregs using chimeric antigen receptors (CARs) and genome editing to enhance their specificity and functionality that may provide significant contributions38. These advances make the manufacturing process more complex but have the potential to make Tregs more versatile and applicable.
The success and approval of the CD19 CAR T cell therapy have inspired the use of the CAR T technology for a wide range of applications including the engineering of CAR Tregs39. The main advantage of using CAR Tregs is to achieve antigen specificity by redirecting Tregs toward a target antigen. Preclinical studies have shown that CAR can be used to redirect Treg specificity40 and to generate alloantigen specific Tregs for induction of transplant tolerance41. The generation of CAR specific Tregs in autoimmune diseases has been achieved42 and MHC molecule specific-CAR-Tregs have also been shown to have the capacity to prevent transplant rejection43,44. Despite these exciting preclinical studies and the extensive experience with manufacturing CAR T cells, we are still awaiting the first CAR Treg clinical trial.
The next decade promises to lead to significant advances in Treg clinical applications as more phase II and possibly phase III clinical trials based on existing and advancing technology take place. Moreover, current preclinical studies, particularly with CAR Tregs, may also enter phase I trials paving the way for more potentially dramatic advances in treatment of autoimmune disease and transplant rejection. These clinical trials and additional possible clinical applications will depend on continued progress in both the basic science and manufacturing technologies.
Figure 1:

An overview of the regulatory T (Treg) cell manufacturing process. Tregs are isolated from peripheral blood mononuclear cells (PBMC) or umbilical cord blood (UCB) by immunomagnetic selection or fluorescence activated cell sorting (FACS). They are then activated/stimulated by incubation with anti-CD3/CD28 coated beads or artificial antigen-presenting cells (aAPCs) consisting of K562 which express CD86 and CD64 (FcR1). This is followed by the expansion of the Tregs using interleukin (IL)-2 and in some cases rapamycin. In the case of induced Tregs (iTregs), TGFβ is also added. After expansion, Tregs are purified (by removing the beads and other agents) followed by flow cytometric analysis to ensure their purity. The final Treg product is then infused to the patient.
Acknowledgements:
This work was supported by the Production Assistance for Cellular Therapies (PACT) program from NIH/NHLBI at University of Minnesota Molecular and Cellular Therapeutics Facility, PACT Contract # HHSN268201000008C. This publication was also supported by the NCI grant P01 CA065493. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Conflict of Interest:
B.R.B receives remuneration as an advisor to Magenta Therapeutics and Blue Rock Therapeutics; Research funding from BlueRock Therapeutics, Rheos Medicines, Equilibre, Carisma and is a co-founder of Tmunity Therapeutics. C.G.B and J.E.W received research funding from BlueRock Therapeutics. The rest of the authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.
References
- 1.Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell 2008;133: 775–87. [DOI] [PubMed] [Google Scholar]
- 2.Itoh M, Takahashi T, Sakaguchi N, Kuniyasu Y, Shimizu J, Otsuka F, Sakaguchi S. Thymus and autoimmunity: production of CD25+CD4+ naturally anergic and suppressive T cells as a key function of the thymus in maintaining immunologic self-tolerance. J Immunol 1999;162: 5317–26. [PubMed] [Google Scholar]
- 3.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003;4: 330–6. [DOI] [PubMed] [Google Scholar]
- 4.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003;299: 1057–61. [DOI] [PubMed] [Google Scholar]
- 5.Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P, Gingeras TR, Fazekas de St Groth B, Clayberger C, Soper DM, Ziegler SF, Bluestone JA. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 2006;203: 1701–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ferreira LMR, Muller YD, Bluestone JA, Tang Q. Next-generation regulatory T cell therapy. Nat Rev Drug Discov 2019;18: 749–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Husebye ES, Anderson MS, Kampe O. Autoimmune Polyendocrine Syndromes. N Engl J Med 2018;378: 1132–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, Kelly TE, Saulsbury FT, Chance PF, Ochs HD. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 2001;27: 20–1. [DOI] [PubMed] [Google Scholar]
- 9.Dominguez-Villar M, Hafler DA. Regulatory T cells in autoimmune disease. Nat Immunol 2018;19: 665–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 2012;30: 531–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Raffin C, Vo LT, Bluestone JA. Treg cell-based therapies: challenges and perspectives. Nat Rev Immunol 2020;20: 158–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bluestone JA, Tang Q. Treg cells-the next frontier of cell therapy. Science 2018;362: 154–5. [DOI] [PubMed] [Google Scholar]
- 13.Zeiser R, Blazar BR. Acute Graft-versus-Host Disease - Biologic Process, Prevention, and Therapy. N Engl J Med 2017;377: 2167–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Blazar BR, MacDonald KPA, Hill GR. Immune regulatory cell infusion for graft-versus-host disease prevention and therapy. Blood 2018;131: 2651–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zwang NA, Leventhal JR. Cell Therapy in Kidney Transplantation: Focus on Regulatory T Cells. J Am Soc Nephrol 2017;28: 1960–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Riley JL, June CH, Blazar BR. Human T regulatory cell therapy: take a billion or so and call me in the morning. Immunity 2009;30: 656–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Brunstein CG, Miller JS, Cao Q, McKenna DH, Hippen KL, Curtsinger J, Defor T, Levine BL, June CH, Rubinstein P, McGlave PB, Blazar BR, Wagner JE. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood 2011;117: 1061–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.MacMillan ML, Hippen KL, McKenna DH, Kadidlo D, Sumstad D, DeFor TE, Brunstein CG, Holtan SG, Miller JS, Warlick ED, Weisdorf DJ, Wagner JE, Blazar BR. First-in-human phase 1 trial of induced regulatory T cells for graft-versus-host disease prophylaxis in HLA-matched siblings. Blood Adv 2021;5: 1425–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pahwa R, Jaggaiahgari S, Pahwa S, Inverardi L, Tzakis A, Ricordi C. Isolation and expansion of human natural T regulatory cells for cellular therapy. J Immunol Methods 2010;363: 67–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Mathew JM, J HV, LeFever A, Konieczna I, Stratton C, He J, Huang X, Gallon L, Skaro A, Ansari MJ, Leventhal JR. A Phase I Clinical Trial with Ex Vivo Expanded Recipient Regulatory T cells in Living Donor Kidney Transplants. Sci Rep 2018;8: 7428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Safinia N, Vaikunthanathan T, Fraser H, Thirkell S, Lowe K, Blackmore L, Whitehouse G, Martinez-Llordella M, Jassem W, Sanchez-Fueyo A, Lechler RI, Lombardi G. Successful expansion of functional and stable regulatory T cells for immunotherapy in liver transplantation. Oncotarget 2016;7: 7563–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chandran S, Tang Q, Sarwal M, Laszik ZG, Putnam AL, Lee K, Leung J, Nguyen V, Sigdel T, Tavares EC, Yang JYC, Hellerstein M, Fitch M, Bluestone JA, Vincenti F. Polyclonal Regulatory T Cell Therapy for Control of Inflammation in Kidney Transplants. Am J Transplant 2017;17: 2945–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bluestone JA, Buckner JH, Fitch M, Gitelman SE, Gupta S, Hellerstein MK, Herold KC, Lares A, Lee MR, Li K, Liu W, Long SA, Masiello LM, Nguyen V, Putnam AL, Rieck M, Sayre PH, Tang Q. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med 2015;7: 315ra189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Theil A, Tuve S, Oelschlagel U, Maiwald A, Dohler D, Ossmann D, Zenkel A, Wilhelm C, Middeke JM, Shayegi N, Trautmann-Grill K, von Bonin M, Platzbecker U, Ehninger G, Bonifacio E, Bornhauser M. Adoptive transfer of allogeneic regulatory T cells into patients with chronic graft-versus-host disease. Cytotherapy 2015;17: 473–86. [DOI] [PubMed] [Google Scholar]
- 25.Brunstein CG, Miller JS, McKenna DH, Hippen KL, DeFor TE, Sumstad D, Curtsinger J, Verneris MR, MacMillan ML, Levine BL, Riley JL, June CH, Le C, Weisdorf DJ, McGlave PB, Blazar BR, Wagner JE. Umbilical cord blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, and clinical effect. Blood 2016;127: 1044–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Todo S, Yamashita K, Goto R, Zaitsu M, Nagatsu A, Oura T, Watanabe M, Aoyagi T, Suzuki T, Shimamura T, Kamiyama T, Sato N, Sugita J, Hatanaka K, Bashuda H, Habu S, Demetris AJ, Okumura K. A pilot study of operational tolerance with a regulatory T-cell-based cell therapy in living donor liver transplantation. Hepatology 2016;64: 632–43. [DOI] [PubMed] [Google Scholar]
- 27.Kellner JN, Delemarre EM, Yvon E, Nierkens S, Boelens JJ, McNiece I, Olson A, Nieto Y, Ciurea S, Popat U, Ahmed S, Champlin R, Ramos J, Nishimoto M, Ma H, Ke Z, Thall P, Khoury JD, Negrin R, Andersson B, Parmar S. Third party, umbilical cord blood derived regulatory T-cells for prevention of graft versus host disease in allogeneic hematopoietic stem cell transplantation: feasibility, safety and immune reconstitution. Oncotarget 2018;9: 35611–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Marek-Trzonkowska N, Mysliwiec M, Dobyszuk A, Grabowska M, Techmanska I, Juscinska J, Wujtewicz MA, Witkowski P, Mlynarski W, Balcerska A, Mysliwska J, Trzonkowski P. Administration of CD4+CD25highCD127− regulatory T cells preserves beta-cell function in type 1 diabetes in children. Diabetes Care 2012;35: 1817–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Sawitzki B, Harden PN, Reinke P, Moreau A, Hutchinson JA, Game DS, Tang Q, Guinan EC, Battaglia M, Burlingham WJ, Roberts ISD, Streitz M, Josien R, Boger CA, Scotta C, Markmann JF, Hester JL, Juerchott K, Braudeau C, James B, Contreras-Ruiz L, van der Net JB, Bergler T, Caldara R, Petchey W, Edinger M, Dupas N, Kapinsky M, Mutzbauer I, Otto NM, Ollinger R, Hernandez-Fuentes MP, Issa F, Ahrens N, Meyenberg C, Karitzky S, Kunzendorf U, Knechtle SJ, Grinyo J, Morris PJ, Brent L, Bushell A, Turka LA, Bluestone JA, Lechler RI, Schlitt HJ, Cuturi MC, Schlickeiser S, Friend PJ, Miloud T, Scheffold A, Secchi A, Crisalli K, Kang SM, Hilton R, Banas B, Blancho G, Volk HD, Lombardi G, Wood KJ, Geissler EK. Regulatory cell therapy in kidney transplantation (The ONE Study): a harmonised design and analysis of seven non-randomised, single-arm, phase 1/2A trials. Lancet 2020;395: 1627–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sanchez-Fueyo A, Whitehouse G, Grageda N, Cramp ME, Lim TY, Romano M, Thirkell S, Lowe K, Fry L, Heward J, Kerr A, Ali J, Fisher C, Lewis G, Hope A, Kodela E, Lyne M, Farzaneh F, Kordasti S, Rebollo-Mesa I, Jose Lozano J, Safinia N, Heaton N, Lechler R, Martinez-Llordella M, Lombardi G. Applicability, safety, and biological activity of regulatory T cell therapy in liver transplantation. Am J Transplant 2020;20: 1125–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Marek-Trzonkowska N, Mysliwiec M, Iwaszkiewicz-Grzes D, Gliwinski M, Derkowska I, Zalinska M, Zielinski M, Grabowska M, Zielinska H, Piekarska K, Jazwinska-Curyllo A, Owczuk R, Szadkowska A, Wyka K, Witkowski P, Mlynarski W, Jarosz-Chobot P, Bossowski A, Siebert J, Trzonkowski P. Factors affecting long-term efficacy of T regulatory cell-based therapy in type 1 diabetes. J Transl Med 2016;14: 332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chwojnicki K, Iwaszkiewicz-Grzes D, Jankowska A, Zielinski M, Lowiec P, Gliwinski M, Grzywinska M, Kowalczyk K, Konarzewska A, Glasner P, Sakowska J, Kulczycka J, Jazwinska-Curyllo A, Kubach M, Karaszewski B, Nyka W, Szurowska E, Trzonkowski P. Administration of CD4(+)CD25(high)CD127(−)FoxP3(+) Regulatory T Cells for Relapsing-Remitting Multiple Sclerosis: A Phase 1 Study. BioDrugs 2021;35: 47–60. [DOI] [PubMed] [Google Scholar]
- 33.Roemhild A, Otto NM, Moll G, Abou-El-Enein M, Kaiser D, Bold G, Schachtner T, Choi M, Oellinger R, Landwehr-Kenzel S, Juerchott K, Sawitzki B, Giesler C, Sefrin A, Beier C, Wagner DL, Schlickeiser S, Streitz M, Schmueck-Henneresse M, Amini L, Stervbo U, Babel N, Volk HD, Reinke P. Regulatory T cells for minimising immune suppression in kidney transplantation: phase I/IIa clinical trial. BMJ 2020;371: m3734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Trzonkowski P, Bieniaszewska M, Juscinska J, Dobyszuk A, Krzystyniak A, Marek N, Mysliwska J, Hellmann A. First-in-man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+CD25+CD127− T regulatory cells. Clin Immunol 2009;133: 22–6. [DOI] [PubMed] [Google Scholar]
- 35.Dong S, Hiam-Galvez KJ, Mowery CT, Herold KC, Gitelman SE, Esensten JH, Liu W, Lares AP, Leinbach AS, Lee M, Nguyen V, Tamaki SJ, Tamaki W, Tamaki CM, Mehdizadeh M, Putnam AL, Spitzer MH, Ye CJ, Tang Q, Bluestone JA. The effect of low-dose IL-2 and Treg adoptive cell therapy in patients with type 1 diabetes. JCI Insight 2021;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Dijke IE, Hoeppli RE, Ellis T, Pearcey J, Huang Q, McMurchy AN, Boer K, Peeters AM, Aubert G, Larsen I, Ross DB, Rebeyka I, Campbell A, Baan CC, Levings MK, West LJ. Discarded Human Thymus Is a Novel Source of Stable and Long-Lived Therapeutic Regulatory T Cells. Am J Transplant 2016;16: 58–71. [DOI] [PubMed] [Google Scholar]
- 37.MacDonald KN, Ivison S, Hippen KL, Hoeppli RE, Hall M, Zheng G, Dijke IE, Aklabi MA, Freed DH, Rebeyka I, Gandhi S, West LJ, Piret JM, Blazar BR, Levings MK. Cryopreservation timing is a critical process parameter in a thymic regulatory T-cell therapy manufacturing protocol. Cytotherapy 2019;21: 1216–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Dawson NAJ, Levings MK. Antigen-specific regulatory T cells: are police CARs the answer? Transl Res 2017;187: 53–8. [DOI] [PubMed] [Google Scholar]
- 39.Rosado-Sanchez I, Levings MK. Building a CAR-Treg: Going from the basic to the luxury model. Cell Immunol 2020;358: 104220. [DOI] [PubMed] [Google Scholar]
- 40.Elinav E, Adam N, Waks T, Eshhar Z. Amelioration of colitis by genetically engineered murine regulatory T cells redirected by antigen-specific chimeric receptor. Gastroenterology 2009;136: 1721–31. [DOI] [PubMed] [Google Scholar]
- 41.Noyan F, Zimmermann K, Hardtke-Wolenski M, Knoefel A, Schulde E, Geffers R, Hust M, Huehn J, Galla M, Morgan M, Jokuszies A, Manns MP, Jaeckel E. Prevention of Allograft Rejection by Use of Regulatory T Cells With an MHC-Specific Chimeric Antigen Receptor. Am J Transplant 2017;17: 917–30. [DOI] [PubMed] [Google Scholar]
- 42.Tenspolde M, Zimmermann K, Weber LC, Hapke M, Lieber M, Dywicki J, Frenzel A, Hust M, Galla M, Buitrago-Molina LE, Manns MP, Jaeckel E, Hardtke-Wolenski M. Regulatory T cells engineered with a novel insulin-specific chimeric antigen receptor as a candidate immunotherapy for type 1 diabetes. J Autoimmun 2019;103: 102289. [DOI] [PubMed] [Google Scholar]
- 43.Boardman DA, Philippeos C, Fruhwirth GO, Ibrahim MA, Hannen RF, Cooper D, Marelli-Berg FM, Watt FM, Lechler RI, Maher J, Smyth LA, Lombardi G. Expression of a Chimeric Antigen Receptor Specific for Donor HLA Class I Enhances the Potency of Human Regulatory T Cells in Preventing Human Skin Transplant Rejection. Am J Transplant 2017;17: 931–43. [DOI] [PubMed] [Google Scholar]
- 44.MacDonald KG, Hoeppli RE, Huang Q, Gillies J, Luciani DS, Orban PC, Broady R, Levings MK. Alloantigen-specific regulatory T cells generated with a chimeric antigen receptor. J Clin Invest 2016;126: 1413–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
