Treg Cells
Treg Cells
SPECIALTY SECTION
This article was submitted to
Regulatory T (Treg) cells are essential for maintaining peripheral tolerance,
Autoimmune and Autoinflammatory
Disorders : Autoimmune Disorders, preventing autoimmunity, and limiting chronic inflammatory diseases. This
a section of the journal small CD4+ T cell population can develop in the thymus and in the peripheral
Frontiers in Immunology
tissues of the immune system through the expression of an epigenetically
RECEIVED 14 February 2023 stabilized transcription factor, FOXP3. Treg cells mediate their tolerogenic
ACCEPTED 22 March 2023
PUBLISHED 19 April 2023
effects using multiple modes of action, including the production of inhibitory
cytokines, cytokine starvation of T effector (e.g., IL-2), Teff suppression by
CITATION
Bluestone JA, McKenzie BS, Beilke J and metabolic disruption, and modulation of antigen-presenting cell maturation or
Ramsdell F (2023) Opportunities for function. These activities together result in the broad control of various immune
Treg cell therapy for the treatment
of human disease.
cell subsets, leading to the suppression of cell activation/expansion and effector
Front. Immunol. 14:1166135. functions. Moreover, these cells can facilitate tissue repair to complement their
doi: 10.3389/fimmu.2023.1166135 suppressive effects. In recent years, there has been an effort to harness Treg cells
COPYRIGHT as a new therapeutic approach to treat autoimmune and other immunological
© 2023 Bluestone, McKenzie, Beilke and
Ramsdell. This is an open-access article
diseases and, importantly, to re-establish tolerance. Recent synthetic biological
distributed under the terms of the Creative advances have enabled the cells to be genetically engineered to achieve
Commons Attribution License (CC BY). The tolerance and antigen-specific immune suppression by increasing their
use, distribution or reproduction in other
forums is permitted, provided the original specific activity, stability, and efficacy. These cells are now being tested in
author(s) and the copyright owner(s) are clinical trials. In this review, we highlight both the advances and the challenges
credited and that the original publication in
this journal is cited, in accordance with
in this arena, focusing on the efforts to develop this new pillar of medicine to treat
accepted academic practice. No use, and cure a variety of diseases.
distribution or reproduction is permitted
which does not comply with these terms.
KEYWORDS
molecular markers of the suppressor cell population, and proteins of the cell surface of antigen-presenting cells (24). In
conflicting data; yet, studies by Nishizuka and colleagues showed addition, Tregs function indirectly and mediate bystander
that mice thymectomized at 3 days after birth achieved systemic suppression through the production of suppressive cytokines
organ-specific autoimmunity (3). Critically, these diseases could be (including IL-10, IL-35, and TGFb), consumption of T cell
prevented or even reversed with the adoptive transfer of growth factors (including IL-2), and altering the local metabolic
CD4+CD25+ T cells (4). This was reminiscent of the Medawar environment to limit Teff cell activity (11). It is important to note
work demonstrating the importance of tolerance induction early in that Treg activation is dependent on activation through the T cell
life and implicated a small subset of T cells in the biology. receptor and CD28 engagement as a co-stimulatory signal. In fact,
In the late 1990s, studies of a mouse strain with multi-system the ability to respond to IL-2 is also TCR/CD28 dependent,
autoimmunity (scurfy mice) led to the identification of the supporting a critical interplay of these signaling pathways in
transcription factor Foxp3 (5). The scurfy mouse, which was determining the ability of these cells to mediate bystander
originally developed at Oak Ridge National Labs during the suppression (25–27). Finally, Tregs have also been shown to
Manhattan (atomic bomb) project, displayed many of the same mediate what is called infectious tolerance (28–31). Gershon and
attributes of the disease seen in neonatally thymectomized mice (6). Kondo were the first to coin the phrase “infectious tolerance” in the
Furthermore, a disease in male children—immune-mediated, 1970s (2, 28). The term was based on the observation, by this group
polyendocrinopathy, X-linked—resembled the pathology found in and others (29–31), that long-term tolerance to allogeneic skin
scurfy mice, and it was shown that these patients also possessed grafts and other immune settings was induced by adoptively
mutations in FOXP3 (7). Further studies by multiple groups transferred Tregs but, once established, remained durable by the
demonstrated that FOXP3 is expressed primarily in the recruitment and development of other immunosuppressive cell
CD4+CD25+ subset of T cells that were shown to be capable of populations. This ability of Tregs to promote other regulatory
controlling autoimmunity in mice (8–10). Thus, by the mid-2000s, cells is likely to provide more robust and durable tolerance. In
T regulatory (Treg) cells could be defined at the molecular level in addition to inhibition of inflammation, Treg cells also promote the
both mice and humans, and their critical role in maintaining repair of damage by releasing tissue and stem cell factors (32, 33).
immunological tolerance was unequivocally established. The multifaceted activity of Treg cells provides a means to reduce
Over the last 20 years, immunological tolerance has been linked inflammation with one cell type but via many different
to multiple mechanisms (1, 11). However, thymically derived Treg mechanisms, and thus Treg and Treg-friendly immune therapies
cells represent the dominant mechanism for maintaining normal have therapeutic potential in a variety of autoimmune,
immune homeostasis and preventing fatal multi-organ inflammatory, and transplant-related diseases.
autoimmunity. These cells possess a broad repertoire of self- One major concern with any potential tolerogenic therapy is the
reactivity and are found in every tissue of the body. Preclinical durability of the treatment. For most therapies, patients must take
studies have demonstrated that Treg cells from the periphery can their medications on a daily, weekly, or monthly basis. In many
traffic to many tissues and can treat many different models of instances, the inflammatory mechanisms can become resistant to
autoimmune disease, and their persistence within the tissue the pathway modulators, leading to a continued reduction in
depends on the presence of self-antigens and commensal bacteria original efficacy. Living drugs, such as Treg cell therapies, hold
(12–14). Importantly, there is strong evidence that the local tissue the prospect of a one-and-done therapy due to the potential of the
and the infiltrating Tregs have a selective activity for proteins drug to be long-lived—for instance, in the case of CAR-T cell
present in the specific inflamed tissue and, in some instances, the therapies in cancer, June and colleagues have shown that the cells
local microbiota (15–17). While Treg cells from the blood are can survive for more than 10 years (34). However, it is yet to be
primarily naïve-like cells, tissue-resident Tregs are “tuned” to the shown that Tregs have the same longevity, that is, where infectious
specific environment within a given tissue. They can display tolerance may be a distinct advantage. Several pre-clinical studies
different chemokine receptors, transcription factors, and have shown that one consequence of the multifunctional activity of
metabolic properties. Tregs is their ability to recruit and influence other immune cells
In addition to the dominant population of thymically derived (such as myeloid and naive T cells) to differentiate into regulatory
FOXP3+ Treg cells, in the periphery, factors such as TGFb can cell populations (MDSCs, Tr1 cells, and additional Tregs), leading
convert conventional T cells to FOXP3+ Tregs, especially in the gut to an amplification of the immunoregulation such that the long-
(18). Such peripherally “induced” treg cells are not as clearly defined term effects of Treg therapy can be durable. In the end, the depth
at the molecular level as the thymically derived FOXP3+ Treg cells, and the breadth of the therapeutic effect of Tregs will be crucial as
and in pre-clinical models, they do not appear to be as stable (19, new modalities of treatments for people with chronic and
20). In this regard, Dijke et al. performed an examination using debilitating diseases. These therapies must, by necessity, lead
discarded human thymuses as a source of therapeutic Tregs. They ultimately to more than a modest step-wise improvement to the
concluded that thymic Tregs are superior to Tregs derived from standard of care. In addition, a combination of precision medicine
peripheral or cord blood (21). using biomarkers to identify specific patient segments that are most
There are multiple mechanisms by which Treg cells have been likely to respond to the therapy, combined with an ability to take
shown to inhibit immune function (22, 23). Tregs directly suppress advantage of the multiple immune regulatory activities or even
antigen presentation directly through the engagement of checkpoint manipulate the cells to maximize the therapeutic impact, will be
receptors, CTLA-4, and stripping major histocompatibility complex required to fulfil the promise of these new therapies.
2 The tolerogenic potential of Treg inflamed tissues. In contrast, TCR-engineered Treg cells engage
HLA peptide-expressing cells which can be expressed in both the
therapies draining lymph node and the inflammatory sites. Although there
are concerns that broad immunosuppression might occur in the
Treg cells provide unique opportunities for meaningful therapeutic
presence of an acute infection or even in the context of cancer,
differentiation that can be envisioned to tackle the complex
healthy individuals routinely combat pathogens and cancer in the
pathobiologies that underlie most autoimmune and inflammatory
presence of antigen-specific Treg activity. There are examples where
diseases. In contrast to other approaches, Treg therapies have been
the deletion of Tregs in pre-clinical models can improve effector
shown to induce immune tolerance, which means the ability to treat for
immunity, but, in general, Tregs are part of the homeostatic balance
a short period of time resulting in long-term disease-free existence.
of a healthy immune response (48, 49). Data in the NOD mouse
Moreover, Tregs can exert their activities in both lymphoid and non-
model of T1D, in which Treg therapy prevents the autoimmune
lymphoid tissues due to the multiple mechanisms that the cells use to
destruction of pancreatic beta cells, demonstrate that viral
control the immunological responses (35). Indeed, in a variety of
immunity is preserved. In adoptive Treg therapy for clinical bone
preclinical studies, Treg adoptive immunotherapy has been shown to
marrow transplant, immunity toward the cancer is likewise
effectively ameliorate systemic inflammation and specific organ injury.
maintained. In fact, studies have shown that Treg activity at sites
These model systems include a wide variety of autoimmune models,
of pathogenic infection can limit the overall tissue damage, and
including inflammatory bowel diseases (36), type 1 diabetes (T1D)
uncontrolled chronic inflammation can be a driver for cancer in
(37), experimental autoimmune encephalomyelitis (EAE) (38),
several settings (50). Finally, in recent years, low-dose IL-2 and IL-2
collagen-induced arthritis (39), as well as organ transplant rejection
muteins have been tested as therapeutics in a variety of clinical
(40) and non-immune diseases, such as stroke (41) and amyotrophic
settings given their ability to expand Tregs in vivo (51). In many of
lateral sclerosis (ALS) (42), where inflammation plays a role in disease
these studies, there has been two- to fivefold increases in Treg
pathogenesis. Thus, Treg therapies have the potential to significantly
numbers in the circulation, even over a long-term treatment
improve a variety of autoimmune and inflammatory disease activities
regimen, yet there have been no reported increases in pathogen
given the broad applicability of the therapeutic approach.
infections or cancer, suggesting that Tregs are not overly
However, given the bystander properties of Tregs and the
immunosuppressive in the context of infections. Finally, as
recognition that organ-specific Tregs play a critical role in
discussed above, Tregs need to constantly engage antigens to
controlling autoimmunity in the local tissue environment, it was
survive—that is, without ongoing TCR and CD28 engagement,
hypothesized that Tregs with single-antigen specificities might have
Tregs fail to persist. Thus, the current preclinical and human data
an increased specific activity and a greater safety profile. In fact, data
suggest that Treg therapy does not inhibit the natural immunity to
in multiple autoimmune (43) and transplant settings (44) showed
pathogens or cancer.
that Treg cells expressing a single T cell receptor (TCR) suppressed
However, despite the early results suggesting that Tregs will be a
autoreactive, multi-specific Teff cells and led to long-term tolerance.
safe therapy, the potential liability of a broad anti-inflammatory
In fact, preliminary data in humans suggest that alloantigen-specific
activity is also being addressed in a variety of ways. First, several of
Treg cells can be effective in the transplantation setting (45). More
the companies are incorporating tags and kill switches in
recent studies have utilized new synthetic biology approaches to
therapeutic Treg products that might allow the patient to be
provide singular antigen specificity to Tregs, including the
treated with clinically approved monoclonal antibodies and small
introduction of specific TCR (46) and chimeric antigen receptors
molecules to eliminate the adoptively transferred Treg.
(CAR) (47) to target specific inflamed tissues. These studies have
Additionally, efforts are underway to create regulated CARs and
shown not only efficacy in multiple pre-clinical models but also 10–
TCRs that allow the specificity to be modulated in case of adverse
25-fold increased activity when compared with polyclonal Treg
effects of the treatment. In addition, as in the CAR-T space in
populations. Thus, given the polypharmacy nature of Tregs, their
cancer, the use of mRNA or short-lived DNA to deliver the
activity in mechanistically different diseases, and their ability to be
specificity will lead to a transient expression of the CAR or TCR.
engineered, Treg therapeutics represent a new approach to creating
This might be especially useful in conditions in which a severe
deep and durable treatments.
pathology is driven in a large part by inflammation and, once
resolved, does not require ongoing Treg persistence. Examples
include acute respiratory distress syndrome (ARDS) or stroke,
3 Challenges and key features to be among others. Such settings would benefit from or perhaps
addressed in developing Treg require the development of an off-the-shelf Treg cell product that
therapeutics might be short-lived.
several other concerns must be addressed in considering a robust in expansion capabilities and potential phenotype changes, such as
cell therapy. Most importantly, the Tregs must maintain their increased interferon production, when compared with Treg derived
phenotype and function stably over time, even in the harshest from healthy individuals. Thus, investigators employed similar tools
inflammatory settings. There are some studies of mouse Tregs as those used for T conventional cell expansion, namely, the use of
showing that, under certain inflammatory conditions, a subset of anti-CD3/anti-CD28-coated beads in combination with IL-2 to
Treg cells can lose Foxp3 expression and produce proinflammatory expand Tregs. In previously published studies, the cells are
cytokines (52, 53). However, other studies suggest that Tregs are routinely expanded 300- to 500-fold in 14 days. The purity of the
quite stable (54). Similar conflicting results have been seen in expanded Treg populations is usually over 90% FOXP3+, and high
patients with autoimmune diseases, usually at the site of levels of multiple phenotypic and functional markers are expressed,
inflammation. This has raised concerns related to cytokine release including CTLA-4, GITR, LAP, and CD25 (55, 80).
syndrome (CRS) as seen in some cancer cell therapy settings using Most importantly, the expanded Tregs have equal or better
CAR-T cells. However, in these settings, CRS primarily occurs in phenotypic and functional activity when compared with freshly
the context of hematological malignancies that carry large antigen isolated Tregs (55). This is especially evident in Tregs isolated from
loads within the blood. Moreover, in many settings (such as patients with autoimmune diseases, where genetic and/or
transplantation and autoimmunity), the patients are on other environmental influences often result in Tregs that are less
immunosuppressive drugs known to moderate the effector effective in multiple suppressive activities and, in many cases, less
cytokine production. In fact, the human clinical data suggests that stable as read out by reduced FOXP3, CD25, and CTLA-4
Treg therapy does not promote CRS in a fashion like that of CAR expression as well as less ability to respond to growth factors such
Teff cells. The stability of Tregs is maintained, and Treg function as IL-2. However, the process of expansion generally leads to a
has been shown to reduce inflammation and have less infection population expressing higher levels of key functional markers and
events compared with immunosuppression. That said, as “repair” of functional defects. In fact, in early studies of IL-2
highlighted below, there are multiple academic and company therapies, the repair of endogenous Treg’s inability to respond
efforts to ensure the stability of these cells (Table 1). effectively to IL-2 was not only reversed but found to be stable
even a year after the discontinuation of cytokine therapy (81). Most
importantly, the expansion of the Tregs derived from peripheral
4 Lessons learned from clinical trials blood as well as cord blood was highly stable when examined over
time after Treg transfer into patients—for instance, an analysis of
Over the last 15 years, there have been more than 25 early-stage circulating adoptively transferred Treg cells demonstrated the
clinical trials conducted, mostly in an academic setting, to continued expression of key functional and phenotypic markers
investigate the safety and biological activity of Tregs in diseases for at least a year post-transfer (55). The results of an extensive bulk
ranging from autoimmunity, organ transplantation, graft versus and single-cell T cell receptor analysis suggest that the expanded
host, and other inflammatory diseases. An overall summary of a population of Tregs remains broad after expansion. Thus, it is not
small “subset” of clinical trials is found in Table 2. These trials have surprising that the current efforts to develop Treg populations for
led to clear evidence that the therapy is feasible, can be administered therapy have begun to employ novel genetic approaches to
safely, and is tolerated for a long term. To date, there have been no homogenize and maximize Treg phenotype and functional
life-threatening adverse events, including no observed increase in activity. This includes the use of antigen-specific receptors,
infections or evidence of cancer development (either the including CARs or TCRs, to focus the tissue specificity of the
administered cells or in the patients). In fact, as confidence in the Treg product.
therapy has grown, more than 10 companies have initiated Treg
programs using both conventional and engineered Treg cells. The
first industry-sponsored trials are underway, with efficacy studies on 4.2 Experiences in Treg stability in vitro and
the horizon, to assess the therapeutic potential of these novel cell in vivo
therapies. That said, there are key lessons that have been learned
from past studies that inform future efforts and have created Despite the previously described limited animal studies, to date,
potential opportunities to improve the therapies. there is no evidence of Treg instability in any human clinical
studies. This result may be due to multiple reasons. First, the
expansion process is much more robust in human versus mouse,
4.1 Experiences in Treg expansion and the likely preferential expansion of thymus-derived CD45RA
naïve Tregs using current approaches may selectively result in the
Fundamental research on Tregs demonstrated that the cells expansion of the most stable Treg populations. In fact, the literature
signal in a similar fashion as conventional T cells, requiring both a suggests that the majority of “unstable” Tregs are likely derived
TCR and CD28 co-stimulatory signals for maximal expansion. In from induced peripheral Tregs, generated in the periphery in vivo
addition, Tregs have been shown to be exquisitely dependent on the upon encounter with TGFb and other Treg-promoting cytokines
IL-2 growth factor for both expansion and survival. Moreover, (19, 22). Moreover, in vitro studies suggest that human Tregs
Tregs coming from individuals with autoimmune or other isolated and expanded under the conditions noted above are
inflammatory diseases can be dysfunctional, leading to differences “resistant” to certain inflammatory cytokines suspected of
Quell Therapeutics FOXP3+ Treg cells Autologous peripheral blood FOXP3 and HLA-A2-CAR Liver transplant
Sangamo Therapeutics FOXP3+ Treg cells Autologous peripheral blood HLA-A2-CAR Kidney transplant
Coya Therapeutics FOXP3+ Treg cells Autologous peripheral blood NONE ALS
Tract Therapeutics FOXP3+ Treg cells Autologous peripheral blood NONE Kidney transplant
GentiBio CD4+ T cells Autologous peripheral blood FOXP3 and islet-specific TCR T1D
Cabaletta Peripheral blood T cells Autologous and Allogeneic T cells CD19-CAR Pemphigus vulgaris
Kyverna Peripheral blood T cells Autologous and Allogeneic peripheral regulated CD19-CAR B cell autoimmunity
blood T cells
Atara Peripheral blood T cells Adult peripheral blood EBV-reactive T cells PPMS
ORCA Bio FOXP3+ Treg cells Allogeneic peripheral blood NONE GvHD
promoting instability in rodent Tregs—for instance, culturing was seemingly apparent in the first cord blood-derived Treg study
human Tregs with IL-6 or TNFa (both present in highly in GvHD, where the cell product had a shorter duration in vivo
inflamed tissues) routinely results in increased expansion and when compared with autologous cells in other settings (71, 72).
function, which are likely due to the upregulation of the TNFR2 However, there are circumstances where an off-the-shelf allogeneic
receptor on the expanded Tregs (52, 82, 83). Finally, it cannot be product may be required, such as in the treatment of acute ARDS,
ruled out that unstable Tregs are only present in tissues subject to such as COVID, where the cells need to be administered
extreme inflammatory milieu; however, as will be noted below, in immediately (79). As will be highlighted elsewhere, new
several clinical settings, biopsy analysis post-transfer showed that approaches to both genetically engineered Tregs as well as
Tregs in target tissues expressed higher levels of FOXP3 and CTLA- potential stem cell-derived Treg products may enable the
4 at 3 months post-transfer of Treg. Thus, it is likely that current disruption of core allogeneic proteins, such as MHC antigens, and
expansion technologies result in the production of highly purified avoid rejection. Interestingly, the inherent alloreactivity of Tregs has
Treg populations that are stable in disease settings (including been exploited in the organ and islet transplant setting, where
autoimmune lesions and inflamed transplanted organs). investigators have either relied on the resident alloreactive Tregs in
At present, the majority of Treg products generated for clinical the polyclonal Treg population or developed approaches to
application are autologous. For autoimmune diseases, this means selectively expand the alloreactive cells prior to transfer. This
that the expansion protocol needs to consider the possibility of approach has great potential but may be superseded by the
contaminated activated Teff cells, which express high levels of CD25 genetic engineering approaches described below.
like Tregs, as well as genetically and environmentally induced Treg It should be noted that, in some settings, the challenge of high
dysfunction. One approach to avoid these potential issues has been purity and stability has required additional precautions in the
to use allogeneic Tregs derived from healthy individuals or development of Treg products—for instance, Teff cell
umbilical cord blood (UCB) (72, 79). UCB Tregs are contamination has been an issue due to inadequate separation
overwhelmingly naïve and have a very broad repertoire (84). In technology and/or patient cell surface phenotype variation. As
some cases, such as in the suppression of graft versus host disease, noted previously, preclinical studies suggest that peripherally
this has the added advantage of 5%–15% of the Tregs having derived Tregs, especially Helios- Tregs, are less stable than their
alloreactivity and thus recognize the class II MHC of the host. thymus-derived counterparts. Thus, depending on the source of
However, there are multiple risks associated with this approach. Tregs cells, the potential for Teff or Teff-like activity is possible. To
First, should the Treg cells become unstable in patients, there is an avoid some of the challenges to isolating and expanding highly
expanded repertoire that can target the allogeneic tissue, be it host stable and pure Tregs, investigators have taken advantage of the
or transplanted tissues. Secondly, the host immune system can selective ability of rapamycin to suppress effector T cell growth
recognize and destroy the adoptively transferred Treg product. This while preserving Treg expansion. The differential mechanistic role
Kidney transplantation
(Continued)
TABLE 2 Continued
(Continued)
TABLE 2 Continued
Expanded autologous
Liver transplantation with Tregs
donor alloantigen-reactive
NCT03577431 (LITTMUS MGH) 1/2 Terminated NA
CD4+CD127lo/−CD25+
(NIAID/Mass Gen Hosp)
Treg cells (arTregs)
(Continued)
TABLE 2 Continued
Other diseases
of mTORC1 and mTORC2 in Treg versus Teff cells results in the there have been only a limited number of presentations and no
rapamycin-induced increased stability of Tregs, selectively published phase 2 studies using polyclonal or antigen-specific
inhibiting Teff cell expansion (85, 86). In the future, new genetic engineered Tregs. In the case of the T1D study, NCT02691247,
engineering approaches that are now being developed will both the company, Caladrius, released a press release saying that the trial
increase the ability to isolate highly pure populations of Tregs as did not meet its primary endpoint. In the case of the ALS study,
well as provide safety switches and a more stable Treg population in recruitment was impacted by COVID-19, and no meaningful
settings where instability remains a risk. evaluation could be made. The early Treg studies were performed
in patients experiencing acute or chronic GvHD, using either cord
blood allogeneic or adult-derived autologous Tregs, based on
4.3 Experiences in early phase 1/2 trials supportive pre-clinical mouse data. In the first study reported out
of Tregs of Poland, ex vivo-expanded CD4+CD127lo/-CD25+ Treg cells were
infused into patients with active GvHD (87). A similar study was
As noted in Table 2, there have been over 25 phase 1/2 clinical performed with cord blood-derived Tregs by Wagner and
trials to test the efficacy of autologous and allogeneic polyclonal colleagues (71, 72). The studies showed that pure Tregs could be
adult and UCB-derived Tregs in a variety of diseases. However, isolated, expanded, and transferred into patients with the disease. It
is also worth noting that one of the impetuses for using cord blood- liver, and, most recently, islet transplantation (Table 2). Once again,
derived Tregs was the greater ease in purifying the population due the therapies have been deemed safe, and in several instances, there
to the distinctly high levels of CD25 on these cells. However, has been biomarker evidence (such as reduced effector cytokine
although the typical expansion time—14 days—of adult-derived production in the urine of patients treated at the time of protocol
autologous Tregs is sufficient to generate target doses, in the case of biopsy proven inflammation) suggesting biologic activity. One
cord blood, the small number of Treg at the start of culture required biomarker study, conducted in the liver transplant setting,
more extensive expansion, often three to four cycles (71, 72). This showed that the therapy was safe, and there was an altered
can lead to increased instability of the product and, in some cases, effector cytokine profile in the blood. Another example of a small
diversion to a Th2 (IL-4)-producing Treg functional state. In the safety trial was a study conducted by Appel and colleagues looking
cord blood-derived allogeneic Treg study, there was an early at the effect of Treg therapy in patients with ALS. The pre-clinical
suggestion of efficacy. This approach in GvHD is now being results had suggested that, even in this non-immune-mediated
examined, with or without low-dose IL-2, in a formal phase II disease, the control of the associated inflammation could impact
study to determine the efficacy and biomarker activity. disease progression. In the study of Appel et al., three patients with
highly advanced ALS were treated with multiple courses of
autologous Tregs along with low-dose IL-2 and monitored for
4.4 Autologous Tregs have been tested in a disease progression (77, 78). There were some suggestions that
variety of clinical settings the disease progression slowed during immunotherapy; however, it
was not clear what role the IL-2 was playing in the
The first clinical studies of autologous Tregs in autoimmunity therapeutic effects.
were conducted in patients with T1D. These studies included One of the challenges in the early Treg interventional studies in
children and adults recently diagnosed with the disease and were autoimmunity has been the small number of patients enrolled in
based on extensive preclinical data that suggested that Tregs were any given trial and the limited biomarker analyses. Although
able to reverse diabetes in a spontaneous animal model of T1D, the biopsies were possible in the transplant studies, any effects were
NOD mouse, if given soon after the diagnosis. The protocol for Treg difficult to interpret given the concomitant treatment of patients
therapy in T1D patients required the transfer of ex vivo-expanded with multiple immune-suppressive drugs. Furthermore, in many
autologous polyclonal CD4+CD127lo/-CD25+ Treg cells within 6 settings, biopsies were not obtained due to logistical and safety
weeks of diagnosis (55, 58, 62). In these studies, the c-peptide levels, reasons. However, several phase 1 studies in autoimmune settings
as a readout of insulin production, were monitored for up to 2 years. have been performed where serial biopsies could be performed. In
While there was a suggestion of efficacy, the studies were small one published study, a patient with cutaneous lupus erythematosus
and uncontrolled. In the study conducted in Poland, the treated was treated with 1 × 108 ex vivo-expanded CD4+CD127lo/-CD25+
children had higher c-peptide levels and lower insulin requirements Treg cells. The affected skin was biopsied both on day 0 (prior to
than the historical data in untreated children (62). In the first phase Treg injection) and at 12 weeks. The biopsy was assessed for
I clinical trial conducted in the United States, ex vivo-expanded changes in Tregs and Teff cells. As noted above, there was a
polyclonal Treg cells were infused into 14 patients with new-onset significant increase in the number of Tregs when the 12-week
T1D in doses ranging from 5 × 106 to 2.6 × 109 cells. The infusions biopsy was compared with that of day 0. More importantly, the
were well tolerated, and the c-peptide levels in most patients level of FOXP3 was significantly increased in the cells isolated from
remained stable for 1 year, although efficacy could not be the biopsy at week 12. In contrast, at the same time point, there was
conclusively shown (55). Notably, the cell pharmacokinetic an approximately 75% reduction in interferon-producing CD4+
analysis of Treg cells labeled with deuterium during ex vivo Teff cells when compared with the day 0 biopsy. Interestingly, in
expansion showed that a subset of the infused Treg cells persisted some patients, IL-17-producing T cells were preserved. Thus, a shift
in peripheral blood for at least 1 year and that the Treg cells from IFNg to IL-17 may lead to reduced inflammation and
remained phenotypically stable after the infusion. A phase II clinical increased tissue repair. Similar results were observed in a phase 1
trial performed by Caladrius Biosciences, in which 113 newly trial in patients with pemphigus (unpublished results/manuscript in
diagnosed (less than 100 days post-T1D diagnosis) adolescents preparation). In one patient with paired samples, the percentage of
with T1D were either untreated or infused with autologous ex Tregs cells increased in the skin biopsy at week 12, similar to that
vivo polyclonally expanded Treg cells (56). The treated group failed observed in the lupus patient (56). There was a significant decrease
to show a clinical effect of the therapy, i.e., preservation of c-peptide in both the number of Teff cells and their ability to produce
production 1 year after the infusion. While the highly enrolled study interferon at 12 weeks. These results together suggest that the
was found to be safe, with no major serious adverse events reported, Treg therapy had an impact on the microenvironment of the
the study highlighted the need to consider a more targeted antigen- pathogenic lesion, resulting in diminished inflammatory processes.
specific Treg population to gain true efficacy as seen in the pre- There have been several studies looking at alloantigen-reactive
clinical studies. Tregs based on preclinical efficacy studies. Overall, Treg cell
Recent years have seen an increased number of polyclonal Tregs infusions have been safe and well tolerated. A challenge in these
trials in multiple different clinical settings. There have been multiple studies, however, has been in manufacturing the cells, potentially
studies of autologous Treg therapy in patients receiving kidney, because these patients are chronically immunosuppressed, which
alters the Tregs both quantitatively and qualitatively. In addition, it 5.2 Enhancing efficacy
appears that the alloreactive Tregs can be sequestered in the target
tissue—in the cited instance, the liver—resulting in a reduced Most new anti-inflammatory drugs fail because of two main
precursor frequency in the blood (69). reasons: lack of clinical safety and lack of clinical efficacy. If the early
clinical work with polyclonal Tregs continues to be a guide, Treg
therapies are likely to be safe. As such, the lack of clinical efficacy
5 Opportunities to enhance Treg cell remains the biggest unknown in the field today. Harnessing the
natural homing and tissue-specific regulatory mechanisms of Tregs
therapy as well as engineering new mechanisms of immune regulation may
be the key to achieving a meaningful and durable clinical effect.
5.1 Enhanced antigen-specific activation
Here are some key areas of focus for the field.
possible to select for unique tissue homing subsets or to engineer a destabilized Treg product. Nonetheless, due to the lack of relevant
homing capabilities to bulk populations of Tregs to send them to long-term model systems for using human Treg cells, there is
specific locations—for example, in treating inflammatory bowel considerable interest in maximizing Treg product stability. There
diseases, it may be advantageous to select for a4b7-expressing Tregs are several approaches to further enhance Treg stability, ranging from
capable of homing to MAdCAM-1-expressing endothelium in sites overt over-expression of FOXP3 to modification of endogenous
of tissue inflammation and in Peyer’s patches and mesenteric lymph genomic loci. One such approach is epigenetic modification of the
nodes (91). This may enhance the total tissue load of Treg and FOXP3 locus to stabilize Treg phenotype and functional activity (93).
enhance both the anti-inflammatory and the pro-repair By targeting the CAR or TCR specificity to the FOXP3 locus, the
mechanisms. Alternatively, one could select against or modify stability of the Tregs would be linked to the expression of an
homing receptors that aid the transit of Tregs to other tissues and introduced TCR or CAR. Other pathways (including IL-2
sites of cell clearance—for example, in the liver (a major site of T cell modulation) may also be effective at promoting FOXP3 expression
clearance). Homing receptors are interesting targets for logic gated and lineage stability. In all cases, stability will need to be assessed in
systems as described above. A homing receptor-mediated logic gate the context of a pro-inflammatory environment.
could be used to selectively kill cells homing to unwanted sites of
action (for example, the CNS) and avoid any consequences of 5.3.3 Allogenic Treg products
tissue-specific immunosuppression in sites of ongoing infection or There is an increasing interest in the use of allogenic cell
increased tumor risk. In this regard, Hoeppli et al. demonstrated products, including gene-modified mature Tregs and iPSC-
that the migration capacity of human Tregs can be tailored by derived off-the-shelf cell platforms. Beyond the advantages from a
adding cytokines (IL-12 and IFNg) and or/metabolites (retinoic cost-of-goods and deliverability perspective, a key advantage is the
acid) to culture conditions during in vitro expansion (92). ability to ensure product consistency and limit patient-to-patient
variability. Additionally, the ability to use healthy donor cells would
eliminate any patient segment considerations on Treg quality or
5.3 Durability and stability quantity. Finally, as noted earlier, an off-the-shelf product would be
more amenable to rapid dosing for acute indications such as ARDS
5.3.1 Enhanced growth factor independence or stroke in which it would be impossible to make autologous Tregs
As noted above, Treg cells are highly dependent on growth factors quickly enough to impact pathology.
for survival, activation, and proliferation. Although many cytokines Another significant advantage to an allogeneic product,
and growth factors can influence Tregs, IL-2 is a critical target for particularly based on iPSC technology, would be the ability to make
advanced genetic engineering. As discussed above, in proinflammatory several (maybe even dozens) of genetic modifications in a serial
conditions, particularly those mediated by T cells, IL-2 is in abundance, manner. It is conceivable to generate “panels” of engineered cells
and Treg are readily able to harness IL-2 through their high-affinity IL- with, for example, differing specificities and then match a particular
2 receptor complexes. This has a twofold effect: it stimulates Treg antigen-specific Treg with a patient segment. Each modification can be
expansion (and function) while capturing IL-2 and thus prevent Teff thoroughly characterized in isolation or in conjunction with other
cells in the environment from receiving a needed growth factor, leading modifications before clinical studies. Allogenic products may be more
to activation-induced cell death. In those indications where there is amenable to repeat dosing as well as exploring combination therapies.
insufficient IL-2—for example, in non-T cell driven inflammation—it Despite the promise of allogenic products, CAR-T effector clinical
may be necessary to provide IL-2 signals directly. As noted previously, development is just beginning. Both engineering and validation have
in the ALS clinical trials, exogenous IL-2 was provided as support for proven more difficult than expected.
expanded Treg cells following infusion. Tregs may be engineered to
provide a cell-intrinsic IL-2 signal in several ways, including IL-2
secretion, IL-2 tethering, and modulation of intracellular signaling 6 Concluding remarks
pathways. Given the central importance of IL-2 signaling in Tregs,
coupled with the inconsistent presence of IL-2 in inflammatory disease Autoimmune disorders arise from defects in immune tolerance
of interest, it is likely that IL-2 will remain a key focus for Treg growth and affect more than 50 million people in the United States and
and persistence. more than 4% of the world’s population. Autoimmune disorders
have a high impact on an individual’s morbidity and mortality as
5.3.2 Enhancing stability well as their quality of life, given that their chronicity, various organ
Although data surrounding the plasticity of adoptively manifestations, and association with co-morbidities are common
transferred human Tregs is lacking, in certain disease settings, and devastating for many sufferers. Despite several transformative
Tregs can become destabilized (as shown by changes in FOXP3 medicines that have improved many autoimmune diseases, most
expression and genome methylation status). As Treg therapies utilize patients do not adequately respond to existing therapies. Long-term
TCRs and CARs to confer disease and/or tissue specificity, ensuring drug therapy is required to maintain long-term efficacy; thus, there
the stability of any Treg product becomes paramount. As noted remains a major unmet medical need. Importantly, the goal of
previously, existing clinical trials, even those using alloantigen- achieving true immune tolerance by re-establishing immune
specific expanded Treg cells, have proven safe without evidence of homeostasis remains unrealized.
As summarized in this review, Tregs, which constitute only a Treg stability and durability, and the development of novel
small percentage of circulating T cells, play a pivotal role in techniques to build new and enhanced activities. Although the
establishing and maintaining peripheral tolerance, preventing field of Treg cell therapies is only at the “end of the beginning”,
autoimmune diseases, and limiting chronic inflammatory diseases many have suggested that this living drug may finally enable
(94–97). In fact, Treg cells are now widely regarded as the primary achieving immune tolerance—the holy grail of immunotherapy.
cells involved in the persistence of peripheral tolerance. Their
essential role is based on the cells’ multiple functionalities ranging
from the production of key immunosuppressive cytokines and Author contributions
metabolites to the cell surface expression of key checkpoint
molecules to control antigen presentation and metabolism. JAB, BM, JB and FR contributed to the writing, revision,
Importantly, the cells exhibit broad bystander suppression and reading, and approval of the submitted manuscript. All authors
can mediate infectious tolerance amplifying the impact of the contributed to the article and approved the submitted version.
cells, thus resulting in robust and durable efficacy.
As cell therapies have flourished in the cancer space as a new pillar
of medicine, it is not surprising that Treg therapies, which have the Acknowledgments
capacity to control inflammation and autoimmunity, have become the
latest approach to treat these devastating diseases. A wealth of pre- The authors wish to acknowledge the contributions of many
clinical data has shown that adoptive Treg cell therapy can be effective colleagues who have pioneered the work summarized in this review.
in the treatment of diseases ranging from autoimmunity and
prevention of organ transplant rejection to cancer-related GvHD and
neurological diseases such as ALS and stroke. Efforts are underway to Conflict of interest
exploit these cells as immunotherapies to treat and potentially prevent
these diseases (97). Early clinical studies suggest that Treg adoptive All the authors are employed by Sonoma Biotherapeutics.
immunotherapy is safe and can lead to biological and molecular
changes that alter disease biology. In the oncology setting, however,
it is likely that modifications to Treg cells may be required to fully Publisher’s note
achieve durable and long-lasting effects. Making use of synthetic
biology, efforts are underway to genetically manipulate these cells to All claims expressed in this article are solely those of the authors
incorporate novel antigen specificities, altered cell characteristics and do not necessarily represent those of their affiliated
including the reliance of certain growth factors and a carrier of organizations, or those of the publisher, the editors and the
payloads that can modify local inflamed tissues. reviewers. Any product that may be evaluated in this article, or
The future for exploiting Treg therapies will depend on solving claim that may be made by its manufacturer, is not guaranteed or
key questions regarding process development, the assessment of endorsed by the publisher.
References
1. Schwartz RH. Historical overview of immunological tolerance. Cold Spring Harb 9. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and
Perspect Biol (2012) 4:a006908. doi: 10.1101/cshperspect.a006908 function of CD4+CD25+ regulatory T cells. Nat Immunol (2003) 4:330–36. doi:
2. Gershon RK, Kondo K. Cell interactions in the induction of tolerance: the role of 10.1038/ni904
thymic lymphocytes. Immunol (1970) 18:723–37. 10. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the
3. Kojima A, Tanaka-Kojima Y, Sakakura T, Nishizuka Y. Spontaneous transcription factor Foxp3. Science (2003) 299:1057–61. doi: 10.1126/science.1079490
development of autoimmune thyroiditis in neonatally thymectomized mice. Lab 11. Waldmann H. Immunological tolerance. Elsevier Reference Module Biomed Sci
Invest (1976) 34:550–57. (2014) 1–7. doi: 10.1016/B978-0-12-801238-3.00116-1
4. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance 12. Campbell DJ. Control of regulatory T cell migration, function, and homeostasis.
maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). breakdown of J Immunol (2015) 195:2507–13. doi: 10.4049/jimmunol.1500801
a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol (1995) 13. Pearce NW, Spinelli A, Gurley KE, Hall BM. Specific unresponsiveness in rats
155:1151–64. doi: 10.4049/jimmunol.155.3.1151 with prolonged cardiac allograft survival after treatment with cyclosporine. v.
5. Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, et al. dependence of CD4+ suppressor cells on the presence of alloantigen and cytokines,
Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal including interleukin 2. Transplantation (1993) 55:374–80. doi: 10.1097/00007890-
lymphoproliferative disorder of the scurfy mouse. Nat Genet (2001) 27:68–73. doi: 199302000-00027
10.1038/83784 14. Panduro M, Benoist C, Mathis D. Tissue tregs. Annu Rev Immunol (2016)
6. Appleby MW, Ramsdell F. Scurfy, the Foxp3 locus, and the molecular basis of 34:609–33. doi: 10.1146/annurev-immunol-032712-095948
peripheral tolerance. Curr Top Microbiol Immunol (2008) 321:151–68. doi: 10.1007/ 15. Samy ET, Wheeler KM, Roper RJ, Teuscher C, Tung KS. Cutting edge:
978-3-540-75203-5_7 Autoimmune disease in day 3 thymectomized mice is actively controlled by
7. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The endogenous disease- specific regulatory T cells. J Immunol (2008) 180:4366–70. doi:
immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is 10.4049/jimmunol.180.7.4366
caused by mutations of FOXP3. Nat Genet (2001) 27:20–1. doi: 10.1038/83713 16. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al.
8. Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for scurfin in CD4 Induction of colonic regulatory T cells by indigenous clostridium species. Science
+CD25+ T regulatory cells. Nat Immunol (2003) 4:337–42. doi: 10.1038/ni909 (2011) 331:337–41. doi: 10.1126/science.1198469
17. Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, et al. 44. Pilat N, Steiner R, Sprent J. Treg therapy for the induction of immune tolerance
Peripheral education of the immune system by colonic commensal microbiota. Nature in transplantation–not lost in translation? Intl J Mol Sci (2023) 24:1752. doi: 10.3390/
(2011) 478:250–54. doi: 10.1038/nature10434 ijms24021752
18. Schmitt EG, Williams CB. Generation and function of induced regulatory T 45. Todo S, Yamashita K, Goto R, Zaitsu M, Nagatsu A, Oura T, et al. A pilot study
cells. Front Immunol (2013) 4:152. doi: 10.3389/fimmu.2013.00152 of operational tolerance with a regulatory T-cell- based cell therapy in living donor liver
19. Yadav M, Louvet C, Davini D, Gardner JM, Martinez-Llordella M, Bailey- transplantation. Hepatology (2016) 64:632–43. doi: 10.1002/hep.28459
Bucktrout S, et al. Neuropilin-1 distinguishes natural and inducible regulatory T cells 46. Elinav E, Waks T, Eshhar Z. Redirection of regulatory T cells with
among regulatory T cell subsets in vivo. J Exp Med (2012) 209:1713–22. doi: 10.1084/ predetermined specificity for the treatment of experimental colitis in mice.
jem.20120822 Gastroenterology (2008) 134:2014–24. doi: 10.1053/j.gastro.2008.02.060
20. Weiss JM, Bilate AM, Gobert M, Ding Y, Curotto de Lafaille MA, Parkhurst CN, 47. Pierini A, Iliopoulou BP, Peiris H, Pé rez-Cruz M, Baker J, Hsu K, et al. T Cells
et al. Neuropilin 1 is expressed on thymus- derived natural regulatory T cells, but not expressing chimeric antigen receptor promote immune tolerance. JCI Insight (2017) 2:
mucosa-generated induced Foxp3+ treg cells. J Exp Med (2012) 209:1723–42. doi: e92865. doi: 10.1172/jci.insight.92865
10.1084/jem.20120914 48. Veiga-Parga T, Sehrawat S, Rouse BT. Role of regulatory T cells during virus
21. Dijke IE, Hoeppli RE, Ellis T, Pearcey J, Huang Q, McMurchy AN, et al. infection. Immunol Rev (2013) 255:182–96. doi: 10.1111/imr.12085
Discarded human thymus is a novel source of stable and long-lived therapeutic 49. Belkaid Y. Paradoxical roles of Foxp3+ T cells during infection: from regulators
regulatory T cells. Am J Transplant (2016) 16:58–71. doi: 10.1111/ajt.13456 to regulators. Cell Host Microbe (2008) 3:341–43. doi: 10.1016/j.chom.2008.05.011
22. Shevach EM, Thornton AM. tTregs, pTregs, and iTregs: similarities and 50. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development.
differences. Immunol Rev (2014) 259:88–102. doi: 10.1111/imr.12160 Front Immunol (2012) 2:98. doi: 10.3389/fimmu.2011.00098
23. Shevach EM. Foxp3+ T regulatory cells: Still many unanswered questions - a 51. Graßhoff H, Comdühr S, Monne LR, Müller A, Lamprecht P, Riemekasten G,
perspective after 20 years of study. Front Immunol (2018) 9:1048. doi: 10.3389/ et al. Low-dose IL-2 therapy in autoimmune and rheumatic diseases. Front Immunol
fimmu.2018.01048 (2021) 12:648408. doi: 10.3389/fimmu.2021.648408
24. Nakayama M, Hori A, Toyoura S, Yamaguchi SI. Shaping of T cell functions by 52. Chen X, Wu X, Zhou Q, Howard OM, Netea MG, Oppenheim JJ. TNFR2 is critical
trogocytosis. Cells (2021) 10:1155. doi: 10.3390/cells10051155 for the stabilization of the CD4+Foxp3+ regulatory t. cell phenotype in the inflammatory
25. Chinen T, Kannan AK, Levine AG, Fan X, Klein U, Zheng Y, et al. An essential environment. J Immunol (2013) 190:1076–84. doi: 10.4049/jimmunol.1202659
role for the IL-2 receptor in treg cell function. Nat Immunol (2016) 17:1322–33. doi: 53. Komatsu N, Okamoto K, Sawa S, Nakashima T, Oh-hora M, Kodama T, et al.
10.1038/ni.3540 Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat
26. Levine AG, Arvey A, Jin W, Rudensky AY. Continuous requirement for the TCR Med (2014) 20:62–8. doi: 10.1038/nm.3432
in regulatory T cell function. Nat Immunol (2014) 15:1070–78. doi: 10.1038/ni.3004 54. Rubtsov YP, Niec RE, Josefowicz S, Li L, Darce J, Mathis D, et al. Stability of the
27. Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, et al. B7/ regulatory T cell lineage in vivo. Science (2010) 329:1667–71. doi: 10.1126/science.1191996
CD28 costimulation is essential for the homeostasis of the CD4+CD25+ 55. Bluestone JA, Buckner JH, Fitch M, Gitelman SE, Gupta S, Hellerstein MK, et al.
immunoregulatory T cells that control autoimmune diabetes. Immunity (2000) Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med
12:431–40. doi: 10.1016/S1074-7613(00)80195-8 (2015) 7:315ra189. doi: 10.1126/scitranslmed.aad4134
28. Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of foreign 56. Dall'Era M, Pauli ML, Remedios K, Taravati K, Sandova PM, Putnam AL, et al.
cells. Nature (1953) 172(4379):603–6. doi: 10.1038/172603a0 Adoptive treg cell therapy in a patient with systemic lupus erythematosus. Arthritis
29. Qin S, Cobbold SP, Pope H, Elliott J, Kioussis D, Davies J, et al. "Infectious" Rheumatol (2019) 71:431–40. doi: 10.1002/art.40737
transplantation tolerance. Science (1993) 259:974–77. doi: 10.1126/science.8094901 57. Caladrius Biosciences. Caladrius biosciences reports top-line data for the phase 2a
30. Waldmann H, Adams E, Fairchild P, Cobbold S. Infectious tolerance and the Sanford project: T-Rex trial of CLBS03 for recent onset type 1 diabetes (2019). Available
long-term acceptance of transplanted tissue. Immunol Rev (2006) 212:301–13. doi: at: https://www.globenewswire.com/en/news-release/2019/02/13/1725052/18623/en/
10.1111/j.0105-2896.2006.00406.x Caladrius-Biosciences-Reports-Top-Line-Data-for-the-Phase-2a-Sanford-Project-T-
31. Jonuleit H, Schmitt E, Kakirman H, Stassen M, Knop J, Enk AH. Infectious tolerance: Rex-Trial-of-CLBS03-for-Recent-Onset-Type-1-Diabetes.html.
human CD25(+) regulatory T cells convey suppressor activity to conventional CD4(+) T 58. Dong S, Hiam-Galvez KJ, Mowery CT, Herold KC, Gitelman SE, Esensten JH,
helper cells. J Exp Med (2002) 196:255–60. doi: 10.1084/jem.20020394 et al. The effect of low- dose IL-2 and treg adoptive cell therapy in patients with type 1
32. Zhang C, Li L, Feng K, Fan D, Xue W, Lu J. 'Repair' treg cells in tissue injury. Cell diabetes. JCI Insight (2021) 6:e147474. doi: 10.1172/jci.insight.147474
Physiol Biochem (2017) 43:2155–69. doi: 10.1159/000484295 59. Desreumaux P, Foussat A, Allez M, Beaugerie L, Hébuterne X, Bouhnik Y, et al.
33. Arpaia N, Green JA, Moltedo B, Arvey A, Hemmers S, Yuan S, et al. A distinct Safety and efficacy of antigen-specific regulatory T-cell therapy for patients with refractory
function of regulatory T cells in tissue protection. Cell (2015) 162:1078– 89. doi: crohn's disease. Gastroenterology (2012) 143:1207–17. doi: 10.1053/j.gastro.2012.07.116
10.1016/j.cell.2015.08.021 60. Canavan JB, Scotta C, Vossenkamper A, Goldberg R, Elder MJ, Shoval I, et al.
34. Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, et al. Developing in vitro expanded CD45RA+ regulatory T cells as an adoptive cell therapy
Decade-long leukaemia remissions with persistence of CD4+ CAR T cells. Nature for crohn's disease. Gut (2016) 65:584–94. doi: 10.1136/gutjnl-2014-306919
(2022) 602:503–09. doi: 10.1038/s41586-021-04390-6 61. Voskens C, Stoica D, Rosenberg M, Vitali F, Zundler S, Ganslmayer M, et al.
35. Akkaya B, Shevach EM. Regulatory T cells: Master thieves of the immune Autologous regulatory T-cell transfer in refractory ulcerative colitis with concomitant
system. Cell Immunol (2020) 355:104160. doi: 10.1016/j.cellimm.2020.104160 primary sclerosing cholangitis. Gut (2023) 72:49–53. doi: 10.1136/gutjnl-2022-327075
36. Uhlig HH, Coombes J, Mottet C, Izcue A, Thompson C, Fanger A, et al. 62. Marek-Trzonkowska N, Myśliwec M, Siebert J, Trzonkowski P. Clinical
Characterization of Foxp3+CD4+CD25+ and IL-10-secreting CD4+CD25+ T cells during application of regulatory T cells in type 1 diabetes. Pediatr Diabetes (2013) 14:322–
cure of colitis. J Immunol (2006) 177:5852–60. doi: 10.4049/jimmunol.177.9.5852 32. doi: 10.1111/pedi.12029
37. Tang Q, Henriksen KJ, Bi M, Finger EB, Szot G, Ye J, et al. In vitro-expanded 63. Chandran S, Tang Q, Sarwal M, Laszik ZG, Putnam AL, Lee K, et al. Polyclonal
antigen-specific regulatory T cells suppress autoimmune diabetes. J Exp Med (2004) regulatory T cell therapy for control of inflammation in kidney transplants. Am J
199:1455–65. doi: 10.1084/jem.20040139 Transplant (2017) 17:2945–54. doi: 10.1111/ajt.14415
38. Stephens LA, Malpass KH, Anderton SM. Curing CNS autoimmune disease with 64. Sawitzki B, Harden PN, Reinke P, Moreau A, Hutchinson JA, Game DS, et al.
myelin- reactive Foxp3+ treg. Eur J Immunol (2009) 39:1108–17. doi: 10.1002/eji.200839073 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
39. Morgan ME, Flierman R, van Duivenvoorde LM, Witteveen HJ, van Ewijk W, (2020) 395:1627–39. doi: 10.1016/S0140-6736(20)30167-7
van Laar JM, et al. Effective treatment of collagen-induced arthritis by adoptive transfer
of CD25+ regulatory T cells. Arthritis Rheum (2005) 52:2212–21. doi: 10.1002/ 65. Mathew JM, H-Voss J, LeFever A, Konieczna I, Stratton C, He J, et al. A phase I
art.21195 clinical trial with ex vivo expanded recipient regulatory T cells in living donor kidney
transplants. Sci Rep (2018) 8:7428. doi: 10.1038/s41598-018-25574-7
40. Joffre O, Santolaria T, Calise D, Al Saati T, Hudrisier D, Romagnoli P, et al.
Prevention of acute and chronic allograft rejection with CD4+CD25+Foxp3+ 66. Roemhild A, Otto NM, Moll G, Abou-El-Enein M, Kaiser D, Bold G, et al.
regulatory T lymphocytes. Nat Med (2008) 14:88–92. doi: 10.1038/nm1688 Regulatory T cells for minimising immune suppression in kidney transplantation:
phase I/IIa clinical trial. BMJ (2020) 371:m3734. doi: 10.1136/bmj.m3734
41. Liesz A, Suri-Payer E, Veltkamp C, Doerr H, Sommer C, Rivest S, et al.
Regulatory T cells are key cerebroprotective immunomodulators in acute 67. Brook MO, Hester J, Petchey W, Rombach I, Dutton S, Bottomley MJ, et al.
experimental stroke. Nat Med (2009) 15:192–99. doi: 10.1038/nm.1927 Transplantation without overimmunosuppression (TWO) study protocol: A phase 2b
randomised controlled single-centre trial of regulatory T cell therapy to facilitate
42. Banerjee R, Mosley RL, Reynolds AD, Dhar A, Jackson-Lewis V, Gordon PH, immunosuppression reduction in living donor kidney transplant recipients. BMJ Open
et al. Adaptive immune neuroprotection in G93A-SOD1 amyotrophic lateral sclerosis (2022) 12:e061864. doi: 10.1136/bmjopen-2022-061864
mice. PloS One (2008) 3:e2740. doi: 10.1371/journal.pone.0002740
68. Safinia N, Vaikunthanathan T, Fraser H, Thirkell S, Lowe K, Blackmore L, et al.
43. Ferreira LMR, Muller YD, Bluestone JA, Tang Q. Next-generation regulatory T cell Successful expansion of functional and stable regulatory T cells for immunotherapy in
therapy. Nat Rev Drug Discov (2019) 18:749–69. doi: 10.1038/s41573-019-0041-4 liver transplantation. Oncotarget (2016) 7:7563–77. doi: 10.18632/oncotarget.6927
69. Tang Q, Leung J, Peng Y, Sanchez-Fueyo A, Lozano JJ, Lam A, et al. Selective decrease lineage stability or function. Front Immunol (2021) 12:783282. doi: 10.3389/
of donor-reactive tregs after liver transplantation limits treg therapy for promoting allograft fimmu.2021.783282
tolerance in humans. Sci Transl Med (2022) 14:eabo2628. doi: 10.1126/scitranslmed.abo2628 84. Motwani K, Peters LD, Vliegen WH, El-Sayed AG, Seay HR, Lopez MC, et al.
70. Bernaldo-de-Quiró s E, Có zar B, Ló pez-Esteban R, Clemente M, Gil-Jaurena JM, Human regulatory T cells from umbilical cord blood display increased repertoire
Pardo C, et al. A novel GMP protocol to produce high-quality treg cells from the diversity and lineage stability relative to adult peripheral blood. Front Immunol (2020)
pediatric thymic tissue to be employed as cellular therapy. Front Immunol (2022) 11:611. doi: 10.3389/fimmu.2020.00611
13:893576. doi: 10.3389/fimmu.2022.893576 85. Battaglia M, Stabilini A, Migliavacca B, Horejs-Hoeck J, Kaupper T, Roncarolo
71. Brunstein CG, Miller JS, McKenna DH, Hippen KL, DeFor TE, Sumstad D, et al. MG. Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T
Umbilical cord blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, cells of both healthy subjects and type 1 diabetic patients. J Immunol (2006) 177:8338–
and clinical effect. Blood (2016) 127:1044–51. doi: 10.1182/blood-2015-06-653667 47. doi: 10.4049/jimmunol.177.12.8338
72. MacMillan ML, Hippen KL, McKenna DH, Kadidlo D, Sumstad D, DeFor TE, 86. Fraser H, Safinia N, Grageda N, Thirkell S, Lowe K, Fry LJ, et al. A rapamycin-
et al. First-in-human phase 1 trial of induced regulatory T cells for graft-versus-host based GMP-compatible process for the isolation and expansion of regulatory T cells for
disease prophylaxis in HLA- matched siblings. Blood Adv (2021) 5:1425–36. doi: clinical trials. Mol Ther Methods Clin Dev (2018) 8:198–209. doi: 10.1016/
10.1182/bloodadvances.2020003219 j.omtm.2018.01.006
73. Hamilton BK. Current approaches to prevent and treat GVHD after allogeneic 87. Trzonkowski P, Dukat-Mazurek A, Bieniaszewska M, Marek-Trzonkowska N,
stem cell transplantation. Hematol Am Soc Hematol Educ Program (2018) 2018:228–35. Dobyszuk A, Juściń ska J, et al. Treatment of graft-versus-host disease with naturally
doi: 10.1182/asheducation-2018.1.228 occurring T regulatory cells. BioDrugs (2013) 27:605–14. doi: 10.1007/s40259-013-0050-5
74. Meyer EH, Laport G, Xie BJ, MacDonald K, Heydari K, Sahaf B, et al. Transplantation 88. Poorebrahim M, Mohammadkhani N, Mahmoudi R, Gholizadeh M, Fakhr E,
of donor grafts with defined ratio of conventional and regulatory T cells in HLA-matched Cid-Arregui A. TCR-like CARs and TCR-CARs targeting neoepitopes: an emerging
recipients. JCI Insight (2019) 4:e127244. doi: 10.1172/jci.insight.127244 potential. Cancer Gene Ther (2021) 28:581–9. doi: 10.1038/s41417-021-00307-7
75. Andú jar-Sá nchez F, Lopes-Ramos T, Bejarano-Garcı́a JA, Garcı́a-Guerrero E, 89. Allen GM, Frankel NW, Reddy NR, Bhargava HK, Yoshida MA, Stark SR, et al.
Calderó n-Cabrera C, Caballero-Velá zquez T, et al. Combined treatment of graft versus Synthetic cytokine circuits that drive T cells into immune-excluded tumors. Science
host disease using donor regulatory T cells and ruxolitinib. Sci Rep (2022) 12:8348. doi: (2022) 378:eaba1624. doi: 10.1126/science.aba1624
10.1038/s41598-022-12407-x 90. Nirschl CJ, Brodkin HR, Hicklin DJ, Ismail N, Morris K, Seidel-Dugan C, et al.
76. Meyer EORCA team. (2021). Available at: https://www.cancernetwork.com/view/ Discovery of a conditionally activated IL-2 that promotes antitumor immunity and
orca-t-improves-outcomes-over-standard-of-care-for-serious-hematologic-malignancies. induces tumor regression. Cancer Immunol Res (2022) 10:581–96. doi: 10.1158/2326-
77. Beers DR, Thonhoff JR, Faridar A, Thome AD, Zhao W, Wen S, et al. Tregs 6066.CIR-21-0831
attenuate peripheral oxidative stress and acute phase proteins in ALS. Ann Neurol 91. Neurath MF. New targets for mucosal healing and therapy in inflammatory
(2022) 92:195–200. doi: 10.1002/ana.26375 bowel diseases. Mucosal Immunol (2014) 7:6–19. doi: 10.1038/mi.2013.73
78. Thonhoff JR, Beers DR, Zhao W, Pleitez M, Simpson EP, Berry JD, et al. 92. Hoeppli RE, MacDonald KN, Leclair P, Fung VCW, Mojibian M, Gillies J, et al.
Expanded autologous regulatory T-lymphocyte infusions in ALS: A phase I, first-in- Tailoring the homing capacity of human tregs for directed migration to sites of Th1-
human study. Neurol Neuroimmunol Neuroinflamm (2018) 5:e465. doi: 10.1212/ inflammation or intestinal regions. Am J Transplant (2019) 19:62–76. doi: 10.1111/ajt.14936
NXI.0000000000000465 93. Colamatteo A, Carbone F, Bruzzaniti S, Galgani M, Fusco C, Maniscalco GT,
79. Lyu MA, Huang M, Zeng K, Li L, Khoury JD, Nishimoto M, et al. Allogeneic et al. Molecular mechanisms controlling Foxp3 expression in health and autoimmunity:
cord blood regulatory T cells can resolve lung inflammation. Cytotherapy (2023) From epigenetic to post-translational regulation. Front Immunol (2020) 10:3136. doi:
25:245–53. doi: 10.1016/j.jcyt.2022.10.009 10.3389/fimmu.2019.03136
80. Balcerek J, Shy BR, Putnam AL, Masiello LM, Lares A, Dekovic F, et al. 94. Bluestone JA, Buckner JH, Herold KC. Immunotherapy: Building a bridge to a
Polyclonal regulatory T cell manufacturing under cGMP: A decade of experience. cure for type 1 diabetes. Science (2021) 373:510–16. doi: 10.1126/science.abh1654
Front Immunol (2021) 12:744763. doi: 10.3389/fimmu.2021.744763 95. Terry LV, Oo YH. The next frontier of regulatory T cells: Promising
81. Long SA, Rieck M, Sanda S, Bollyky JB, Samuels PL, Goland R, et al. Rapamycin/IL-2 immunotherapy for autoimmune diseases and organ transplantations. Front
combination therapy in patients with type 1 diabetes augments tregs yet transiently impairs Immunol (2020) 11:565518. doi: 10.3389/fimmu.2020.565518
b-cell function. Diabetes (2012) 61:2340–48. doi: 10.2337/db12-0049 96. Sakaguchi S, Mikami N, Wing JB, Tanaka A, Ichiyama K, Ohkura N. Regulatory
82. Leclerc M, Naserian S, Pilon C, Thiolat A, Martin GH, Pouchy C, et al. Control of T cells and human disease. Annu Rev Immunol (2020) 38:541–66. doi: 10.1146/
GVHD by regulatory T cells depends on TNF produced by T cells and TNFR2 expressed by annurev-immunol-042718-041717
regulatory T cells. Blood (2016) 128:1651–59. doi: 10.1182/blood-2016-02-700849 97. Esensten JH, Muller YD, Bluestone JA, Tang Q. Regulatory T-cell therapy for
83. Skartsis N, Peng Y, Ferreira LMR, Nguyen V, Ronin E, Muller YD, et al. IL- 6 autoimmune and autoinflammatory diseases: The next frontier. J Allergy Clin Immunol
and TNFa drive extensive proliferation of human tregs without compromising their (2018) 142:1710–18. doi: 10.1016/j.jaci.2018.10.015