Abstract
Background The clinical use of ex vivo-expanded T-regulatory cells for the treatment of T-cell-mediated diseases has gained increasing momentum. However, the recent demonstration that FOXP3+ T-regulatory cells may contain interleukin-17–producing cells and that they can convert into effector cells once transferred in vivo raises significant doubts about their safety. We previously showed that rapamycin permits the ex vivo expansion of FOXP3+ T-regulatory cells while impairing the proliferation of non-T-regulatory cells. Here we investigated the Th17-cell content and the in vivo stability of rapamycin-expanded T-regulatory cells as pertinent aspects of cell-based therapy.Design and Methods T-regulatory-enriched cells were isolated from healthy volunteers and were expanded ex vivo with rapamycin with a pre-clinical applicable protocol. T-regulatory cells cultured with and without rapamycin were compared for their regulatory activity, content of pro-inflammatory cells and stability.Results We found that CD4+CCR6+CD161+ T cells (i.e., precursor/committed Th17 cells) contaminate the T-regulatory cells cultured ex vivo in the absence of rapamycin. In addition, Th17 cells do not expand when rapamycin-treated T-regulatory cells are exposed to a “Th17-favorable” environment. Rapamycin-expanded T-regulatory cells maintain their in vitro regulatory phenotype even after in vivo transfer into immunodeficient NOD-SCID mice despite being exposed to the irradiation-induced pro-inflammatory environment. Importantly, no additional rapamycin treatment, either in vitro or in vivo, is required to keep their phenotype fixed.Conclusions These data demonstrate that rapamycin secures ex vivo-expanded human T-regulatory cells and provide additional justification for their clinical use in future cell therapy-based trials.Introduction
In recent years, there has been growing recognition of the ability of human FOXP3 T-regulatory (Treg) cells to suppress adverse immune responses. Much research effort has been devoted to implementing Treg-cell therapy in the clinic1,2 and results of the first clinical trials have been recently reported.3,4 Given their reduced circulating frequency (i.e., 5–10% of the CD4 T cells) FOXP3 Treg cells, to be of any therapeutic use in cell-therapy trials, need to be expanded ex vivo. The intrinsic reduced ability of Treg cells to proliferate in vitro can be reversed by potent stimulation through the T-cell receptor (TCR) in the presence of high doses of exogenous interleukin (IL)-2.5 Protocols for the expansion of human FOXP3 Treg cells performed under conditions of good manufacturing practice (GMP) have been shown to be feasible.3,6–8 Importantly, these culture conditions are also highly advantageous for the expansion of effector CD25 T cells, which may contaminate the starting peripheral FOXP3 Treg cells. This obstacle can be bypassed by: (i) the use of cord blood Treg cells,4 (ii) isolation of very pure peripheral Treg cells through flow-based cell sorters3,9,10 or (iii) the addition of rapamycin to the culture, which preferentially inhibits proliferation of effector T cells while sparing Treg cells.11,12
IL-17 is a pro-inflammatory cytokine with non-redundant functions in the clearance of extracellular pathogens.13 In humans, IL-17 is associated with many inflammatory disorders such as rheumatoid arthritis, asthma, multiple sclerosis, Crohn’s disease and allograft rejection.14 The majority of IL-17 derives from a population of CD4 T cells known as Th17.13 The combination of cytokines which stimulates the development and stabilizes human Th17 cells is a subject of much debate.15 It appears that transforming growth factor (TGF)-β, IL-21, IL-23, IL-6, and IL-1β at various dosages and combinations, depending on the experimental conditions and the starting T-cell subset, are essential for Th17-cell generation/expansion.15
It has recently been shown that a memory fraction of CD25FOXP3 T cells contains a significant number of IL-17-producing cells which lack regulatory activity.16,17 In the context of concerted efforts to use expanded populations of Treg cells for adoptive therapy in human T-cell-mediated diseases, the risk of Th17-cell contamination is a key, as transfer of contaminant pro-inflammatory cells may exacerbate rather than ameliorate disease. Thus, developing methods of securing ex vivo-expanded Treg cells intended for clinical use and inhibiting their pro-inflammatory potential in vivo have become all-important aspects of Treg cell-mediated immunotherapy.18
We set up a clinically applicable protocol for the expansion of human peripheral Treg cells using rapamycin to set the basis for an investigator-initiated clinical trial. Th17-cell content and the in vivo stability of the rapamycin-expanded human FOXP3 Treg cells were investigated as critical pertinent aspects of cell-based therapy.
Design and Methods
Blood samples
Peripheral blood was obtained from healthy donors’ buffy coats obtained after written informed consent in accordance with the Declaration of Helsinki under the protocol approved by the San Raffaele Scientific Institute Ethics Committee (protocol TIGET PERIBLOOD).
T-regulatory cell purification
Peripheral blood mononuclear cells (PBMC) were separated by density-gradient centrifugation over Lymphoprep (Axis-shield, Oslo, Norway). Some PBMC were cryopreserved for later use. The remaining PBMC were processed using CD8 MicroBeads (Miltenyi Biotec, Bergisch Gladbach, Germany) according to the manufacturer’s instructions. Treg cells were then isolated from the negative cell fractions (CD8 cells) using CD25 MicroBeads (Miltenyi Biotec) following the manufacturer’s instructions.
Flow cytometry
The cells were stained for surface antigens with the following monoclonal antibodies: anti-CD4 PerCP (clone SK3, BD, Franklin Lakes, NJ, USA), anti-CD25 APC (clone 2A3, BD), CD161 PE (clone HP3G10, eBioscience), and CCR6-biotin (clone 11A9, BD) and streptavidin APC7 (BD). Intracellular staining for human FOXP3 and Helios was performed using the anti FOXP3-Alexa 488 and anti-Helios-PE monoclonal antibodies (clone 259D and clone 22F6, respectively, BioLegend, San Diego, CA, USA) according to the manufacturer’s instructions. Samples were acquired on a FACSCanto (BD) and analyzed with FCS Express V3 software (DE Novo Software, Los Angeles, CA, USA).
Cell cultures
CD8CD25 T cells isolated from PBMC of healthy subjects were plated at 0.1×10/mL in X-vivo 15 medium (Lonza, Verviers, Belgium) supplemented with 5% pooled AB human serum (Lonza) and 100 U/mL penicillin/streptomycin (Bristol-Myers Squibb, Sermoneta, Italy) in the presence or absence of 100 nM rapamycin (i.e. 100 ng/mL) (Rapamune, Whyet, Maidenhead, Berkshire, UK).12 The cells were activated with GMP grade beads coated with anti-CD3 and anti-CD28 antibodies (T Cell Expander from Dynal Biotech ASA, Oslo, Norway) at a 1:3 ratio (cells:beads). At day 2 post-activation, IL-2 (500 U/mL) (Proleukin, Chiron, Amsterdam, the Netherlands) was added to the culture and replenished as shown in Figure 1. Beads were removed after 14 days of culture by magnetic adherence and the expanded cells were used for further analyses.
Suppression assay with TMED and TRPM cells
Previously frozen PBMC were thawed and stained with CFSE (Molecular Probes, Eugene, OR, USA)11 and a suppression assay was performed as described elsewhere by our group.12 The percentage of CFSE divided cells was calculated using the formula: [(n. of precursors that proliferated1-6/n. of total precursors0-6)×100]. The percentage of CFSE cells divided in the presence of expanded cells was compared to the percentage of CFSE divided cells in the absence of any added cells.12
Cytokine detection in TMED and TRPM cells
The 14-day cultured T cells were plated at a density of 0.5×10 cells in 500 μL in 48-well plates and stimulated for 6 h with latex-beads (Molecular Probes) pre-coated with anti-CD3 (OKT3 clone) and anti-CD28 (clone CD28.2, BD Pharmingen) monoclonal antibodies at 500 ng and 50 ng for 10 beads, respectively at a 1:2 ratio (cells:beads) in the presence of 10 μg/mL brefeldin A (Sigma-Aldrich). At the end of the 6-h activation, the cell surface was stained with anti-CD4 PerCp monoclonal antibody (clone SK3, BD). Subsequently, intracellular staining was performed with the following monoclonal antibodies: anti-IL-2 PE (clone MQ1-17H12), anti-IL-4 PE (clone MP4-25D2, anti-IL-10 PE (clone JES3-9D7), anti-IFNγ APC (clone B27), and anti-TNFα PE (clone MaB11) (all from BD Pharmingen), and anti-IL-17 PE (clone eBio64CAP17, from eBioscence). Each stain was performed in the presence of anti-FOXP3 Alexa488 monoclonal antibody (clone 259D, Biolegend) according to the manufacturer’s protocol.
TH17-cell differentiation/expansion
The TMED and TRPM cells were plated at a density of 0.5×10/mL and cultured with 5 ng/mL TGF-β (R&D System, Minneapolis, MN, USA), 25 ng/mL IL-21 (Cell Sciences, Canton, MA, USA) and 25 ng/mL IL-23 (R&D System) as previously described.19 On day 3, IL-23 was replenished. On day 7 the cells (i.e., TMED-17, and TRPM-17) were collected and used for further analysis.
Suppression assay with TMED-17 and TRPM-17 cells
Previously frozen PBMC were thawed and stained with CFSE (Molecular Probes) as previously described.11 TMED-17 and TRPM-17 cells were added at the ratio of 0.3:1 (T cells:responder cells) and stimulated with Treg Suppression Inspector beads (Miltenyi Biotec) at a ratio of 1:0.3 (cells:beads). Seven days later the cells were collected and stained with anti-CD4 and anti-CD8 monoclonal antibodies and analyzed by flow activated cell sorting (FACS).
Cytokine detection in TMED-17, TRPM-17 cells
TMED-17, TRPM-17 cells were stimulated for 6 h with TPA (CalBiochem, Lucerne, Switzerland) and ionomycin (Sigma-Aldrich) at 10 ng/mL and 500 ng/mL, respectively, in the presence of 10 μg/mL brefeldin A (Sigma-Aldrich). At the end of the 6-h activation, the cell surface was stained with anti-CD4 and anti-CD25 monoclonal antibodies, while intracellular staining was performed with anti-IL-17 or anti-IL-2, and anti-IFNγ APC monoclonal antibodies.
Non-obese diabetic/severe combined immunodeficiency mouse model
Six- to 8-week old female NOD/scid mice were obtained from Charles-River Italia (Calco, Italy). The experimental protocol was approved by the San Raffaele Scientific Animal Care and Use Committee (IACUC#387). The day before human cell transfer, mice were treated with 1 mg of anti-CD122 (TMβ-1) monoclonal antibody (Santa Cruz, Tebubio, Le Perray-en-Yuelines, France) i.p. as previously described.20 On day 0, the mice received total body irradiation (TBI) with a single dose of 350 cGy (γ irradiation from a linear accelerator) and 8×10 cells/mouse were transferred i.p.. Three times a week the general appearance, body weight and mobility of the mice were evaluated and once a week peripheral blood was collected from the tail vein and chimerism analysis was performed. Human chimerism was determined weekly by flow cytometry, after bleeding from the tail vein, using the monoclonal antibodies anti-human-CD45 APC (clone HI30) and anti-mouse-CD45 PerCp (clone 30F11) (both from BD-Pharmingen). The percentage of chimerism was calculated using the formula: [h-CD45/(h-CD45+m-CD45)x100] as described elsewhere.21 To improve human chimerism, some of the mice were treated with 2×10 IU/mouse/day i.p. of recombinant human IL2 (Proleukin, Chiron) for 7 days after cell transfer.21
Phenotype, cytokine production profile and suppressive activity of T cells recovered from non-obese diabetic/severe combined immunodeficiency mice
Thirteen or 19 days after human cell transfer into NOD/scid mice, the animals were sacrificed and peripheral blood, thymus, spleen and lymph nodes were collected and pooled after smashing (TMED-mouse and TRPM-mouse). The total cells recovered from each mouse varied accordingly to the cell-type injected. Total cells recovered from mice injected with CD3 T cells: 4.5±1.2×10, with TMED: 0.2±0.5×10, and with TRPM: 0.5±0.1×10 (average ± SD, n=7). Total cells from mice injected with CD25TRPM cells were analyzed by flow cytometry or were stimulated for 6 h with TPA (CalBiochem) and ionomycin (Sigma-Aldrich) in the presence of brefeldin A (Sigma-Aldrich). At the end of the 6-h activation, the cell surface was stained with anti-human CD4 PerCp (clone SK3, BD) and anti-human-CD45 APC (clone HI30, BD-Pharmingen) monoclonal antibodies. Subsequently, the intracellular staining was performed with anti-IL-17 or anti-IL-2, and anti-IFNγ monoclonal antibodies.
Alternatively, T cells were purified by using human-CD3 MicroBeads (Miltenyi Biotec) according to the manufacturer’s instructions and used as suppressor T cells in a suppression assay. Previously frozen PBMC (responder cells) were thawed and stained with CFSE (Molecular Probes) as described elsewhere.11 TMED-mouse and TRPM-mouse cells were added at the ratio of 0.3:1 and 1:1 (T cells:responder cells) and stimulated with Treg Suppression Inspector beads (Miltenyi Biotec) at a ratio of 1:0.3 (cells:beads). Seven days later the cells were collected and stained with anti-CD4 and anti-CD8 monoclonal antibodies and analyzed by FACS.
Statistical analysis
All statistical analyses were performed using a two-tailed Student’s t-test. P values less than 0.05 were deemed statistically significant.
Results
Human Treg cells were enriched from PBMC using GMP-grade available magnetic beads, which are the best alternative to clinical grade flow-based cell sorters.8 We first depleted CD8 T cells and then positively selected CD25 cells. The purified cell subset (termed CD25) contained a median of 80±10% CD4CD25 cells and 61±5% of CD25FOXP3 cells (data not shown). CD25 cells were cultured ex vivo to obtain the best Treg-cell expansion in the shortest time possible. Increasing doses of GMP-grade anti-CD3/CD28–coated beads (from 1:1 to 1:5 cells:beads) and IL-2 (from 100 to 1000 U/mL), and different periods of culture (from 7 to 21 days) in the presence or absence of rapamycin (TRPM and TMED cells, respectively) were tested. The optimal expansion protocol was 14 days of culture in the presence of three beads per cell and 500 U/mL of exogenous IL-2 (scheme depicted in Figure 1A).
TRPM cells expanded 1 log less than TMED cells with a 7-day time-lag (Figure 1B), as we previously observed upon expansion of unfractionated human CD4 T cells.12 TRPM cells contained an average of 51% FOXP3-expressing cells (range: 32–80%). Helios, a member of the Ikaros transcription factor family, is thought to be a specific marker of thymic-derived Treg cells.22 We found an average of 73% (range, 60–85%) of Helios cells within the FOXP3-expressing TRPM cells, suggesting a selective expansion of thymic-derived Treg cells in the presence of rapamycin. In addition, TRPM cells displayed a reproducible in vitro suppressive capacity towards both autologous and allogeneic responder T cells in a dose-dependent manner. In contrast, CD25 T cells expanded in the absence of rapamycin (i.e., TMED cells) almost completely lost FOXP3-expressing cells (average: 14%; range, 3–30%) and the few cells remaining FOXP3 expressed low levels of Helios (average: 28%; range, 20–35%) and retained no in vitro suppressive function in any condition tested (Figure 2 and Online Supplementary Figure S1). The TCR Vβ repertoire remained polyclonal in both TMED and TRPM cells demonstrating the absence of a specific T-cell clone expansion in the 14-day culture (Online Supplementary Table S1). We previously showed that rapamycin can be considered as a “selective Treg cell growth-survival” factor since it inhibits proliferation of non-Treg cells,12 which follow different molecular signaling pathways than Treg cells.23,24 Analysis of the cytokine production profile of TMED and TRPM cells confirmed that the presence of rapamycin in the culture reduced the frequency of T cells releasing pro-inflammatory cytokines (Figure 3).
Recent findings demonstrate that FOXP3 T cells contain some IL-17-producing non-Treg cells16,17 and that naïve FOXP3 Treg cells can differentiate into Th17 cells in the presence of IL-2 and lineage-specific polarizing factors.25 As a consequence, we wondered whether the CD25 T-cell starting population contained significant numbers of Th17-cells or Th17-cell precursors, recently found to be highly enriched within the CCR6CD161 cell fraction,26,27 which might expand upon the 14-day culture. The starting CD25 T-cell subset was slightly enriched in CCR6CD161 T cells, as compared to the unfractionated PBMC (Figure 4A). We, therefore, tested the IL-17-producing cell content in TMED and TRPM cells. Low amounts of IL-17IFN-γ cells were present in the expanded CD25 T cells irrespectively of the presence of rapamycin (Figure 4B). Nevertheless, TMED cells contained a higher frequency of CCR6CD161 cells as compared to TRPM cells (Figure 4C). Rapamycin, therefore, inhibits the differentiation and growth of CCR6CD161 T cells during ex vivo expansion of CD25-enriched T cells. In spite of this, we could not rule out that a subsequent exposure of TRPM cells to a “Th17-favorable environment” in the absence of exogenous rapamycin would lead to Th17-cell differentiation and/or expansion. To resolve this issue, TMED and TRPM cells were cultured in the presence of IL-23, IL-21 and TGF-β (Figure 4D) (termed TMED-17 and TRPM-17), a cytokine-cocktail known to expand and stabilize Th17 cells.13,19 Seven days of in vitro culture in this “Th17-favorable environment” and in the absence of rapamycin led to the expansion of IL-17-producing cells (at high or low frequency depending on the donor) in TMED but not in TRPM cells (Figure 4E). These data were also confirmed by a high frequency of CCR6CD161 cells in TMED-17 but not in TRPM-17 cells (data not shown). In addition, the FOXP3 cell-content (Figure 5A) and regulatory activity of TRPM-17 cells were as good as those prior to the Th17-cell condition exposure (Figure 5B).
One major concern regarding the use of ex vivo-expanded Treg cells in immunotherapy trials is the risk of their in vivo conversion into effector cells and loss of suppressive ability. We, therefore, decided to test the stability of TRPM cells in vivo upon transfer into NOD/scid mice (protocol depicted in Figure 6A). TMED, TRPM, or freshly isolated CD3 T cells were injected into total body irradiated (TBI)-NOD/scid mice in the absence of any additional rapamycin treatment. T cells previously cultured ex vivo (i.e., TMED and TRPM cells) did not expand in vivo as efficiently as freshly isolated T cells (Figure 6B), despite the fact that the frequency of circulating human CD3CD25 cells was almost identical in the mice receiving either freshly isolated T cells or TMED and TRPM cells 13 days following in vivo transfer (Figure 6C). This is likely due to T-cell exhaustion given by the powerful ex vivo TCR-mediated expansion, as previously demonstrated by others.28 Additional treatment of recipient mice with recombinant human IL-2 did not improve human T-cell chimerism in any of the treated mice (data not shown). Despite the lack of evident circulation of human T cells in the peripheral blood of NOD/scid mice injected with TMED and TRPM cells, human cells were found in their spleens and lymph nodes. We, therefore, isolated human T cells from secondary lymphoid organs of NOD/scid mice 19 days after adoptive cell transfer. Interestingly, the recovered TRPM (i.e., TRPM-MOUSE) cells remained FOXP3 and almost all the FOXP3 cells were Helios–expressing cells and contained no CCR6CD161 precursor/committed Th17 cells (Figure 6D). The cytokine production profile of TRPM-MOUSE cells changed slightly towards more IL-2 and IFN-γ producing cells (average 11% and 8%, respectively). Conversely, IL-17 cells were almost undetectable in all recovered TRPM-MOUSE cells (Figure 6E). Unfortunately, the number of TMED cells recovered from the injected mice was too low to perform the above-mentioned tests. Importantly, TRPM cells retained their suppressive activity even 19 days after in vivo “parking” in TBI-treated host mice (Figure 6F) while neither unexpanded T cells nor TMED cells recovered from injected mice displayed any suppressive activity (data not shown).
Overall, these in vitro and in vivo data strongly suggest that rapamycin not only allows the ex vivo expansion of Treg cells voided of inflammatory-producing cells but also fixes their phenotype.
Discussion
Clinical trials with ex vivo-expanded human FOXP3 Treg cells isolated from peripheral blood are currently under question mainly because purified CD25 T cells may contain contaminant non-Treg cells.16,17 The current study shows that a pre-clinical grade protocol for the expansion of functional peripheral Treg cells with rapamycin is feasible. In addition, we demonstrate that rapamycin-expanded human Treg cells are not contaminated by Th17 cells and that they maintain a stable phenotype even upon in vivo transfer with no need for additional rapamycin treatment.
We previously reported that rapamycin permits the preferential enrichment and subsequent expansion of mouse11 and human12 FOXP3 Treg cells from a population of unfractionated CD4 T cells. In these previous studies we showed that three rounds of powerful in vitro stimulation were required to expand an average of 30% FOXP3 cells with sustained suppressive activity. In this work we aimed to start with a purer Treg-cell fraction using GMP-grade available magnetic beads and to shorten the cell culture. Interestingly, the purity of our starting cell population was very similar to that recently reported by Brunstein et al. who have just performed a clinical trial in humans with ex vivo purified and expanded cord-blood derived CD25 T cells.4 This further supports the validity of our approach for expanding clinical-grade peripheral CD25 T cells. In addition, it is clear that defining Treg cell dosing strategies in new clinical trials requires large-scale Treg-cell expansion capacity. The protocol described here led to an average 45-fold expansion of our starting Treg-cell fraction. This increase is limited as compared to that of other protocols performed in the absence of rapamycin2 but is absolutely sufficient to obtain, for instance, 3×10/kg of expanded cells (for an average patient of 60 kg), which is the highest dose shown to be effective and safe in cord blood-transplanted patients.4
We believe that the novelty of our work lies in two major aspects. First, Th17 cells do not differentiate and expand upon ex vivo culture of magnetically-isolated CD25 Treg cells only in the presence of rapamycin. The recent findings demonstrating that memory FOXP3 T cells contain some IL-17-producing non-Treg cells16,17 and that naïve FOXP3 Treg cells differentiate into Th17 cells in the presence of IL-2 and lineage-specific polarizing factors25 raise significant doubts about the safety of the protocols commonly used for Treg-cell expansion. The experimental procedure must in fact prevent the growth and development of IL-17-producing or IL-17-precursor cells, which could contaminate the starting T-cell fraction. In our experimental conditions, the starting CD25 T-cell subset did indeed contain a certain number of already committed Th17 cells or their precursors26,27 which expanded efficiently over the 14-day culture period only in the absence of rapamycin. Importantly, the subsequent exposure of TRPM cells to a Th17-favorable environment in the absence of new addition of rapamycin to the culture did not lead to Th17-cell differentiation or expansion. These data demonstrate that rapamycin imprints a fixed phenotype, not dependent upon the chronic presence of rapamycin, to the expanded Treg cells. The molecular mechanism underlying such a phenomenon is currently under active investigation.
It has been shown that rapamycin reduces de novo, in vitro generation of IL-17-producing cells by TGF-β+IL-6 while it permits TGF-β-induced development of FOXP3 cells starting from murine CD4 T cells.29 This could be ascribed to blockade of the biological activity of IL-6 by rapamycin.29 Our new findings demonstrate that rapamcyin has an inhibitory effect on human Th17-cell growth and development starting from a Treg-cell–enriched fraction and, importantly, it enables the expansion of Treg cells rich in Helios cells and void of Th17-cell contaminants.
The second novel aspect of our work is the demonstration that TRPM cells preserve their regulatory function even upon in vivo transfer into TBI-treated immunodeficient mice in the absence of any additional rapamycin treatment, thus further supporting their fixed phenotype. The in vivo suppressive potential of FOXP3 Treg cells, freshly isolated30 or previously expanded in the presence31 or absence32,33 of rapamycin, has already been shown in xeno-graft-versus-host disease (GvHD) murine models. However, to our knowledge, to date no experiments have been reported in which only ex vivo-expanded Treg cells have been transferred into TBI-treated NOD/scid mice and subsequently recovered. It is well known that pre-transplant irradiation of immunodeficient mice prompts a massive cytokine storm, which promotes human T-cell engraftment, activation and development of lethal xeno-GvHD.34 The TBI-induced release of TNF-α, IL-1, and IL-6 promotes the activation of host antigen-presenting cells and up-regulation of MHC antigens,35 which are crucial for the induction of lethal xeno-GvHD by human T cells.36 We found that expanded Treg cells have a reduced in vivo proliferative capacity as compared to the same number of freshly isolated T cells, confirming data from the past performed with total T cells.28 However, the transferred T cells did survive for as long as 19 days after in vivo transfer (latest time point analyzed) and despite being exposed to the critical TBI-induced cytokine storm their phenotype and suppressive activity remained stable.
It is evident that many issues need to be resolved prior to using ex vivo-expanded human peripheral FOXP3 Treg cells in clinical trials.2 A common point of discussion among scientists with a strong interest in Treg-cell therapies is the limit of cytokine-producing expanded Treg cells that can be safely infused into a patient. Our data show that, upon in vivo transfer, Treg cells skew their cytokine production profile a bit more towards INF-γ/IL-2-producing cells but, importantly, this profile does not lead to a reduced regulatory function and the mere transfer of these cells in vivo does not cause xeno-GvHD. We, therefore, believe that rather than setting a “cytokine-producing Treg cell threshold” one should set up appropriate tests to monitor the regulatory stability of the transferred cells.
In summary, the demonstration that Treg-enriched cells expanded in the presence of rapamycin lack contaminant Th17 cells and are stable upon in vivo transfer in a murine model of TBI-induced inflammation in the absence of rapamycin administration provides additional justification for the clinical use of these cells in future cell therapy-based trials.
Acknowledgments
We thank all Manuela Battaglia’s and Maria-Grazia Roncarolo’s laboratory staff (San Raffaele Scientific Institute, Milano, Italy) for helpful and continuous discussion. We also thank Prof. Toshiyuki Tanaka (Osaka University, Osaka, Japan) for kindly providing the TMβ-1 hybridoma.
Footnotes
- Authorship and Disclosures The information provided by the authors about contributions from persons listed as authors and in acknowledgments is available with the full text of this paper at www.haematologica.org.
- Financial and other disclosures provided by the authors using the ICMJE (www.icmje.org) Uniform Format for Disclosure of Competing Interests are also available at www.haematologica.org.
- The online version of this article has a Supplementary Appendix.
- Received January 28, 2011.
- Revision received April 15, 2011.
- Accepted May 5, 2011.
References
- Roncarolo MG, Battaglia M. Regulatory T-cell immunotherapy for tolerance to self antigens and alloantigens in humans. Nat Rev Immunol. 2007; 7(8):585-98. PubMedhttps://doi.org/10.1038/nri2138Google Scholar
- 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(5):656-65. PubMedhttps://doi.org/10.1016/j.immuni.2009.04.006Google Scholar
- Trzonkowski P, Szarynska M, Mysliwska J, Mysliwski A. Ex vivo expansion of CD4(+)CD25(+) T regulatory cells for immunosuppressive therapy. Cytometry A. 2009; 75(3):175-88. PubMedGoogle Scholar
- Brunstein CG, Miller JS, Cao Q, McKenna DH, Hippen KL, Curtsinger J. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood. 2011; 117(3):1061-70. PubMedhttps://doi.org/10.1182/blood-2010-07-293795Google Scholar
- Li L, Godfrey WR, Porter SB, Ge Y, June CH, Blazar BR. CD4+CD25+ regulatory T-cell lines from human cord blood have functional and molecular properties of T-cell anergy. Blood. 2005; 106(9):3068-73. PubMedhttps://doi.org/10.1182/blood-2005-04-1531Google Scholar
- Hoffmann P, Boeld TJ, Eder R, Albrecht J, Doser K, Piseshka B. Isolation of CD4+CD25+ regulatory T cells for clinical trials. Biol Blood Marrow Transplant. 2006; 12(3):267-74. PubMedhttps://doi.org/10.1016/j.bbmt.2006.01.005Google Scholar
- Guillot-Delost M, Cherai M, Hamel Y, Rosenzwajg M, Baillou C, Simonin G. Clinical-grade preparation of human natural regulatory T-cells encoding the thymidine kinase suicide gene as a safety gene. J Gene Med. 2008; 10(8):834-46. PubMedhttps://doi.org/10.1002/jgm.1220Google Scholar
- Peters JH, Preijers FW, Woestenenk R, Hilbrands LB, Koenen HJ, Joosten I. Clinical grade Treg: GMP isolation, improvement of purity by CD127 Depletion, Treg expansion, and Treg cryopreservation. PLoS One. 2008; 3(9):e3161. PubMedhttps://doi.org/10.1371/journal.pone.0003161Google Scholar
- Hoffmann P, Boeld TJ, Eder R, Huehn J, Floess S, Wieczorek G. Loss of FOXP3 expression in natural human CD4+CD25+ regulatory T cells upon repetitive in vitro stimulation. Eur J Immunol. 2009; 39(4):1088-97. PubMedhttps://doi.org/10.1002/eji.200838904Google Scholar
- Putnam AL, Brusko TM, Lee MR, Liu W, Szot GL, Ghosh T. Expansion of human regulatory T-cells from patients with type 1 diabetes. Diabetes. 2009; 58(3):652-62. PubMedhttps://doi.org/10.2337/db08-1168Google Scholar
- Battaglia M, Stabilini A, Roncarolo MG. Rapamycin selectively expands CD4+CD25+FoxP3+ regulatory T cells. Blood. 2005; 105(12):4743-8. PubMedhttps://doi.org/10.1182/blood-2004-10-3932Google Scholar
- Battaglia M, Stabilini A, Migliavacca B, Horejs-Hoeck J, Kaupper T, Roncarolo MG. Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients. J Immunol. 2006; 177(12):8338-47. PubMedhttps://doi.org/10.4049/jimmunol.177.12.8338Google Scholar
- Miossec P, Korn T, Kuchroo VK. Interleukin-17 and type 17 helper T cells. N Engl J Med. 2009; 361(9):888-98. PubMedhttps://doi.org/10.1056/NEJMra0707449Google Scholar
- Crome SQ, Wang AY, Levings MK. Translational mini-review series on Th17 cells: function and regulation of human T helper 17 cells in health and disease. Clin Exp Immunol. 2010; 159(2):109-19. PubMedhttps://doi.org/10.1111/j.1365-2249.2009.04037.xGoogle Scholar
- de Jong E, Suddason T, Lord GM. Translational mini-review series on Th17 cells: development of mouse and human T helper 17 cells. Clin Exp Immunol. 2009; 159(2):148-58. PubMedGoogle Scholar
- Battaglia M, Roncarolo MG. The fate of human Treg cells. Immunity. 2009; 30(6):763-5. PubMedhttps://doi.org/10.1016/j.immuni.2009.06.006Google Scholar
- Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009; 30(6):899-911. PubMedhttps://doi.org/10.1016/j.immuni.2009.03.019Google Scholar
- Afzali B, Mitchell P, Lechler RI, John S, Lombardi G. Translational mini-review series on Th17 cells: induction of interleukin-17 production by regulatory T cells. Clin Exp Immunol. 2009; 159(2):120-30. PubMedhttps://doi.org/10.1111/j.1365-2249.2009.04038.xGoogle Scholar
- Yang L, Anderson DE, Baecher-Allan C, Hastings WD, Bettelli E, Oukka M. IL-21 and TGF-beta are required for differentiation of human T(H)17 cells. Nature. 2008; 454(7202):350-2. PubMedhttps://doi.org/10.1038/nature07021Google Scholar
- Tanaka T, Kitamura F, Nagasaka Y, Kuida K, Suwa H, Miyasaka M. Selective long-term elimination of natural killer cells in vivo by an anti-interleukin 2 receptor beta chain monoclonal antibody in mice. J Exp Med. 1993; 178(3):1103-7. PubMedhttps://doi.org/10.1084/jem.178.3.1103Google Scholar
- Bondanza A, Valtolina V, Magnani Z, Ponzoni M, Fleischhauer K, Bonyhadi M. Suicide gene therapy of graft-versus-host disease induced by central memory human T lymphocytes. Blood. 2006; 107(5):1828-36. PubMedhttps://doi.org/10.1182/blood-2005-09-3716Google Scholar
- Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, Belkaid Y. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol. 2010; 184(7):3433-41. PubMedhttps://doi.org/10.4049/jimmunol.0904028Google Scholar
- Bensinger SJ, Walsh PT, Zhang J, Carroll M, Parsons R, Rathmell JC. Distinct IL-2 receptor signaling pattern in CD4+CD25+ regulatory T cells. J Immunol. 2004; 172(9):5287-96. PubMedhttps://doi.org/10.4049/jimmunol.172.9.5287Google Scholar
- Crellin NK, Garcia RV, Levings MK. Altered activation of AKT is required for the suppressive function of human CD4+CD25+ T regulatory cells. Blood. 2007; 109(5):2014-22. PubMedhttps://doi.org/10.1182/blood-2006-07-035279Google Scholar
- Valmori D, Raffin C, Raimbaud I, Ayyoub M. Human ROR{gamma}t+ TH17 cells preferentially differentiate from naive FOXP3+Treg in the presence of lineagespecific polarizing factors. Proc Natl Acad Sci USA. 2010; 107(45):19402-7. PubMedhttps://doi.org/10.1073/pnas.1008247107Google Scholar
- Cosmi L, Maggi L, Santarlasci V, Capone M, Cardilicchia E, Frosali F. Identification of a novel subset of human circulating memory CD4(+) T cells that produce both IL-17A and IL-4. J Allergy Clin Immunol. 2010; 125(1):e1-4. https://doi.org/10.1016/j.jaci.2009.10.012Google Scholar
- Maggi L, Santarlasci V, Capone M, Peired A, Frosali F, Crome SQ. CD161 is a marker of all human IL-17-producing T-cell subsets and is induced by RORC. Eur J Immunol. 2010; 40(8):2174-81. PubMedhttps://doi.org/10.1002/eji.200940257Google Scholar
- Humeau L, Namikawa R, Bardin F, Mannoni P, Roncarolo MG, Chabannon C. Ex vivo manipulations alter the reconstitution potential of mobilized human CD34+ peripheral blood progenitors. Leukemia. 1999; 13(3):438-52. PubMedhttps://doi.org/10.1038/sj/leu/2401329Google Scholar
- Kopf H, de la Rosa GM, Howard OM, Chen X. Rapamycin inhibits differentiation of Th17 cells and promotes generation of FoxP3+ T regulatory cells. Int Immunopharmacol. 2007; 7(13):1819-24. PubMedhttps://doi.org/10.1016/j.intimp.2007.08.027Google Scholar
- Mutis T, van Rijn RS, Simonetti ER, Aarts-Riemens T, Emmelot ME, van Bloois L. Human regulatory T cells control xenogeneic graft-versus-host disease induced by autologous T cells in RAG2−/−gammac−/−immunodeficient mice. Clin Cancer Res. 2006; 12(18):5520-5. PubMedhttps://doi.org/10.1158/1078-0432.CCR-06-0035Google Scholar
- Golovina TN, Mikheeva T, Suhoski MM, Aqui NA, Tai VC, Shan X. CD28 costimulation is essential for human T regulatory expansion and function. J Immunol. 2008; 181(4):2855-68. PubMedhttps://doi.org/10.4049/jimmunol.181.4.2855Google Scholar
- Cao T, Soto A, Zhou W, Wang W, Eck S, Walker M. Ex vivo expanded human CD4+CD25+Foxp3+ regulatory T cells prevent lethal xenogenic graft versus host disease (GVHD). Cell Immunol. 2009; 258(1):65-71. PubMedhttps://doi.org/10.1016/j.cellimm.2009.03.013Google Scholar
- Kleinewietfeld M, Starke M, Di Mitri D, Borsellino G, Battistini L, Rotzschke O. CD49d provides access to “untouched” human Foxp3+ Treg free of contaminating effector cells. Blood. 2009; 113(4):827-36. PubMedhttps://doi.org/10.1182/blood-2008-04-150524Google Scholar
- Reddy P. Pathophysiology of acute graft-versus-host disease. Hematol Oncol. 2003; 21(4):149-61. PubMedhttps://doi.org/10.1002/hon.716Google Scholar
- Xun CQ, Thompson JS, Jennings CD, Brown SA, Widmer MB. Effect of total body irradiation, busulfan-cyclophosphamide, or cyclophosphamide conditioning on inflammatory cytokine release and development of acute and chronic graft-versus-host disease in H-2-incompatible transplanted SCID mice. Blood. 1994; 83(8):2360-7. PubMedGoogle Scholar
- Lucas PJ, Bare CV, Gress RE. The human anti-murine xenogeneic cytotoxic response. II. Activated murine antigen-presenting cells directly stimulate human T helper cells. J Immunol. 1995; 154(8):3761-70. PubMedGoogle Scholar