Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Clinical Grade Treg: GMP Isolation, Improvement of Purity by CD127pos Depletion, Treg Expansion, and Treg Cryopreservation

  • Jorieke H. Peters,

    Affiliation Department of Bloodtransfusion and Transplantation Immunology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands

  • Frank W. Preijers,

    Affiliation Central Hematology Laboratory, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands

  • Rob Woestenenk,

    Affiliation Central Hematology Laboratory, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands

  • Luuk B. Hilbrands,

    Affiliation Department of Nephrology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands

  • Hans J. P. M. Koenen,

    Affiliation Department of Bloodtransfusion and Transplantation Immunology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands

  • Irma Joosten

    I.Joosten@abti.umcn.nl

    Affiliation Department of Bloodtransfusion and Transplantation Immunology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands

Abstract

Background

Treg based immunotherapy is of great interest to facilitate tolerance in autoimmunity and transplantation. For clinical trials, it is essential to have a clinical grade Treg isolation protocol in accordance with Good Manufacturing Practice (GMP) guidelines. To obtain sufficient Treg for immunotherapy, subsequent ex vivo expansion might be needed.

Methodology/Principal Findings

Treg were isolated from leukapheresis products by CliniMACS based GMP isolation strategies, using anti-CD25, anti-CD8 and anti-CD19 coated microbeads. CliniMACS isolation procedures led to 40–60% pure CD4posCD25highFoxP3pos Treg populations that were anergic and had moderate suppressive activity. Such CliniMACS isolated Treg populations could be expanded with maintenance of suppressive function. Alloantigen stimulated expansion caused an enrichment of alloantigen-specific Treg. Depletion of unwanted CD19pos cells during CliniMACS Treg isolation proved necessary to prevent B-cell outgrowth during expansion. CD4posCD127pos conventional T cells were the major contaminating cell type in CliniMACS isolated Treg populations. Depletion of CD127pos cells improved the purity of CD4posCD25highFoxP3pos Treg in CliniMACS isolated cell populations to approximately 90%. Expanded CD127neg CliniMACS isolated Treg populations showed very potent suppressive capacity and high FoxP3 expression. Furthermore, our data show that cryopreservation of CliniMACS isolated Treg is feasible, but that activation after thawing is necessary to restore suppressive potential.

Conclusions/Significance

The feasibility of Treg based therapy is widely accepted, provided that tailor-made clinical grade procedures for isolation and ex vivo cell handling are available. We here provide further support for this approach by showing that a high Treg purity can be reached, and that isolated cells can be cryopreserved and expanded successfully.

Introduction

Regulatory T cells (Treg) play a critical role in various immunological processes, particularly in maintaining homeostasis and self-tolerance. Immunotherapy based on Treg infusion is therefore a potential treatment for many immune disorders. This approach has been proven successful in animal models of stem cell transplantation [1][7], solid organ transplantation [8], [9] and auto-immunity [6], [10][13]. Therapeutic application in humans requires large numbers of Treg, that have to be isolated and, if necessary, expanded using clinical grade (Good Manufacturing Practice, GMP) protocols. The CliniMACS system provides a relatively versatile method for GMP cell isolation and is able to manage large quantities of cells, for example from leukapheresis material.

Other authors have recently shown the feasibility of CliniMACS for CD4posCD25high Treg enrichment, but failed to achieve Treg purity of more than 40–60% [14], [15]. For stem cell transplantation recipients, a suboptimal Treg purity is unlikely to be harmful. Currently, many stem cell transplantation recipients are treated with donor lymphocyte infusions to enhance graft-versus-leukemia/tumor responses. The main adverse effect of this treatment is graft-versus-host-disease (GVHD). The aim of Treg immunotherapy in this patient group is to reduce these graft-versus-host responses. Co-infusion of both non-regulatory T cells to ensure graft-versus-leukemia/tumor responses and Treg to prevent excessive GVHD is therefore a logical approach in this setting. However, in other patient groups, such as solid organ graft recipients and patients with auto-immune diseases, high purity of Treg for immunotherapy is crucial. Infusion of non-regulatory cells into patients that already suffer from unwanted immunological activity should be prevented, as these cells can potentially intensify the disease process. The main aggressive cell types in immune responses are cytotoxic CD8pos T cells and CD4pos conventional T cells, and contamination of CliniMACS isolated Treg populations with these cells should be avoided for immunotherapy in patients other than stem cell recipients. Likewise, contamination of the isolate with B cells could lead to the infusion of activated B cells with potential adverse immunological consequences.

Although CliniMACS based Treg isolation from leukapheresis products will yield large numbers of Treg, ex vivo expansion of these cells prior to infusion might still be necessary to obtain sufficient numbers of Treg for effective immunotherapy. Treg can be expanded efficiently using polyclonal stimulation [16][19]. However, antigen-specific Treg have been shown to be far more efficient than polyclonal Treg in animal models [3], [5][7], [12], [13]. In allogeneic transplantation, the target antigens are known (mainly foreign HLA) and human alloantigen-specific Treg can be obtained by expansion using stimulation with alloantigen [20], [21].

For practical reasons, it would be convenient or even necessary to perform the leukapheresis procedure weeks/months before the transplantation or treatment and store the Treg until they are needed. Currently, information on cryopreservation of Treg is lacking.

In the current study, we confirm that GMP Treg isolation using standard CliniMACS procedures results in moderately pure Treg populations. Here, we show that purity of CliniMACS isolated Treg can be improved by depletion of CD127pos cells. In addition, we show that CliniMACS isolated Treg can be expanded with maintenance of suppressive function using polyclonal or alloantigen stimuli in the presence of IL-2. Depletion of CD19pos B cells during CliniMACS Treg isolation appears to be a prerequisite to prevent B cell outgrowth during expansion. Furthermore, we show that cryopreservation of Treg is feasible, but that the cells require activation to restore suppressive potential.

Results

Isolation of Treg using CliniMACS yields 40–60% pure CD4posCD25high Treg populations with suppressive activity

Treg immunotherapy will demand very high numbers of highly pure Treg populations isolated in a GMP manner. To obtain large numbers of Treg with optimal purity, we performed CD4posCD25high T cell isolation from healthy donor leukapheresis products using the GMP CliniMACS system. The most straight-forward strategy to enrich for Treg would be a one step positive selection of CD25pos cells out of leukapheresis products. However, this strategy would unintentionally result in contaminating (activated) cytotoxic CD8pos T cells in the isolated Treg populations. Therefore, our initial CliniMACS isolation strategy consisted of two steps: first a CD8 depletion step to exclude CD8pos effector T cells from the isolated cell populations, followed by a CD25 enrichment step to enrich for CD4posCD25high Treg (referred to as CliniMACS8/25). For comparison, a laboratory scale MiniMACS based Treg isolation protocol was also performed (referred to as MiniMACS).

Leukapheresis products contained on average 3% CD4posCD25high regulatory T cells (Figure 1 and Table 1). Less than 0.1% CD8pos cells were present in the isolated CliniMACS8/25 populations. While 80% of CliniMACS8/25 cells was CD4posCD25pos, only 40–60% was CD4posCD25high (Figure 1A and Table 1). MiniMACS based Treg isolation from the same leukapheresis product yielded a higher Treg purity with 90% CD4posCD25high cells. CliniMACS8/25 populations expressed lower levels of FoxP3 as compared to MiniMACS isolated Treg populations (Figure 1A).

thumbnail
Figure 1. Phenotypic and functional characteristics of freshly CliniMACS isolated Treg.

Treg were isolated from leukapheresis products in GMP CliniMACS-based or non-GMP MiniMACS-based isolation procedures. Data from a typical isolation are shown (N = 3). (A) Cell surface expression of CD4 and CD25 and intracellular expression of FoxP3. (B) Proliferative capacity upon stimulation with allogeneic PBMC in the absence or presence of exogenous IL-2, measured at day 5 of culture. (C) Suppressive capacity in co-cultures of autologous naïve CD4posCD25neg Tresp stimulated with allogeneic PBMC. Significant differences are indicated by asterisks. In figure C, open and filled asterisks refer to Tconv (used as control for the addition of cells) versus MiniMACS or CliniMACS8/25, respectively.

https://doi.org/10.1371/journal.pone.0003161.g001

thumbnail
Table 1. Enrichment of CD4posCD25high Treg by CliniMACS isolation.

https://doi.org/10.1371/journal.pone.0003161.t001

A hallmark of CD4posCD25high Treg is their anergic phenotype, defined as low proliferative capacity upon T cell receptor stimulation alone as compared to Tconv cells, which can be restored by the addition of exogenous IL-2. Both CliniMACS8/25 and MiniMACS populations were anergic (Figure 1B).

Suppressive capacity of Treg populations was determined by titration of Treg into co-cultures of autologous CD4posCD25neg responder T cells stimulated with allogeneic stimulator PBMC. As shown in Figure 1C, Treg populations isolated with the CliniMACS8/25 strategy were moderately suppressive, reaching 50% suppression at a 1∶1 Tresp∶Treg ratio, while MiniMACS isolated Treg populations were more potent, with 50% suppression at Tresp∶Treg ratios of approximately 4∶1.

Expansion of CliniMACS Treg with either polyclonal or alloantigen stimulation increases cell numbers while suppressive activity is retained

CliniMACS Treg isolation strategies yield high numbers of Treg, but these numbers might still be too low for therapeutic purposes. This issue can be overcome by ex vivo expansion of Treg. To analyze expansion potential and effects of expansion on phenotypic and functional characteristics, CliniMACS8/25 isolated Treg populations were activated using either polyclonal stimulation (anti-CD3+anti-CD28 mAb coated microbeads) or alloantigen stimulation (irradiated PBMC from an HLA-mismatched donor), in the presence of exogenous IL-2.

Polyclonal and alloantigen stimulated expansion cultures yielded similar cell numbers (Table 2). CliniMACS8/25 populations increased about 30-fold in cell numbers in ten days. The phenotype of expanded CliniMACS8/25 populations is depicted in Figure 2A. CD25 expression was high in both CliniMACS8/25 and MiniMACS populations, while the expression of FoxP3 was lower in expanded CliniMACS8/25 cells as compared to expanded MiniMACS populations. After expansion, all Treg populations remained anergic (Figure 2B). CliniMACS8/25 and MiniMACS populations suppressed alloantigen driven responder T cell proliferation to a similar extent (Figure 2C). Notably, alloantigen expanded Treg populations showed higher suppressive capacity in alloantigen driven responder T cell responses as compared to polyclonally expanded Treg (>75% suppression at a Tresp∶Treg ratio of 16∶1 versus 50% suppression at Tresp∶Treg ratios a Tresp∶Treg ratio of 8∶1, respectively, Figure 2C). This indicates that the alloantigen expanded cell populations were enriched for alloantigen-specific Treg. As our group has shown previously, the low proliferation observed after addition of alloantigen expanded Tconv can be explained by distinct culture kinetics with an earlier proliferation peak and a net result of lower counts at day 5 [21].

thumbnail
Figure 2. Phenotypic and functional characteristics of expanded CliniMACS isolated Treg.

Cell populations were expanded with polyclonal stimulus or alloantigen stimulus in the presence of exogenous IL-2. Data from a typical experiment are shown (N = 3). (A) Cell surface expression of CD25 and intracellular expression of FoxP3. (B) Proliferative capacity upon restimulation with allogeneic PBMC (same donor as in alloantigen expansion) in the absence or presence of exogenous IL-2, measured at day 3 of culture. (C) Suppressive capacity in co-cultures of autologous naïve CD4posCD25neg Tresp stimulated with allogeneic PBMC (same donor as in alloantigen expansion). Significant differences are indicated by asterisks. In figure C, open and filled asterisks refer to Tconv (expanded with same stimulus as Treg populations) versus MiniMACS or CliniMACS8/25, respectively.

https://doi.org/10.1371/journal.pone.0003161.g002

thumbnail
Table 2. Increase in CliniMACS Treg numbers by expansion.

https://doi.org/10.1371/journal.pone.0003161.t002

It would be convenient for Treg immunotherapy to be able to store Treg cells prior to manipulation and/or infusion. The suppressive capacity of cryopreserved CliniMACS Treg was impaired (Figure 3), and could not be regained by cell resting for up to seven days in culture medium. However, expansion of cryopreserved CliniMACS cells restored suppressive capacity.

thumbnail
Figure 3. Cryopreservation of CliniMACS Treg affects suppressive capacity, which can be restored by activation.

Suppressive capacity of cryopreserved CliniMACS Treg in co-cultures of autologous naïve CD4posCD25neg Tresp stimulated with allogeneic PBMC. Data from a typical experiment are shown (N = 2–4). Prior to co-culture suppression assay, indicated cell populations were expanded for 10 days in the presence of exogenous IL-2 and allogeneic PBMC (same donor as in co-culture suppression assay). Significant differences are indicated by asterisks. Open and filled asterisks refer to cryopreservation versus cryopreservation plus 4 days rest or cryopreservation plus expansion, respectively.

https://doi.org/10.1371/journal.pone.0003161.g003

Depletion of CD19pos cells during CliniMACS Treg isolation is necessary to prevent B cell contamination

After CliniMACS8/25 isolation procedures, less than 3% B cells were present in isolated populations, these numbers increased after expansion to 5 to 10% (Figure 4A). In order to remove B cells from our CliniMACS isolated Treg populations, we tested the efficacy of CD19pos B cell depletion in the CliniMACS isolation procedure. In this strategy, first CD8pos and CD19pos cells were depleted, followed by enrichment of CD25pos cells (referred to as CliniMACS8/19/25). Less than 0.5% B cells were present after these isolations (see Table 1 and Figure 4B) and no substantial B cell contaminations arose after either polyclonal or alloantigen expansion (see Table 2 and Figure 4B). CliniMACS8/19/25 populations were similar to CliniMACS8/25 cells with respect to the phenotype, anergic state and suppressive potential, both directly after isolation as well as after expansion (data not shown). Thus, CD19pos depletion effectively prevents B cell contaminations in CliniMACS Treg populations after isolation and after expansion.

thumbnail
Figure 4. B cell contamination can be prevented by depleting CD19pos cells during CliniMACS Treg isolation.

Percentage of contaminating B cells in freshly isolated or alloantigen expanded CliniMACS populations. Data from a typical experiment are shown (N = 3). (A) CliniMACS8/25 populations. (B) CliniMACS8/19/25 populations.

https://doi.org/10.1371/journal.pone.0003161.g004

Depletion of CD127pos cells improves CliniMACS Treg purity

CD4posCD25neg/lowFoxP3neg/low Tconv cells were the major contaminating cell type in CliniMACS isolated Treg (40–60% of the isolated populations, see Figure 1). Recently, it has been described that Tconv express the IL-7 receptor alpha-chain CD127, while Treg cells do not express CD127 [22], [23]. Indeed, CliniMACS isolated populations contained both CD127neg and CD127pos cells, reflecting the presence of both Treg and Tconv cells (Figure 5A). To improve the purity of Treg isolated with CliniMACS, we hypothesized that depletion of CD127pos cells could be employed to exclude contaminating T cells. Because anti-CD127 CliniMACS beads are currently not available, we depleted CD127pos cells from CliniMACS isolated cell populations by FACS.

thumbnail
Figure 5. CliniMACS Treg isolation can be improved by depleting CD127pos Tconv cells.

CliniMACS populations were stained with anti-CD127 and sorted into CD127neg and CD127pos subsets. CliniMACS cells were cryopreserved prior to cell sorting. Data from one of two similar experiments are shown. (A) Cell surface expression of CD127, CD4 and CD25 and intracellular expression of FoxP3. (B-C-D) CliniMACS populations were expanded with alloantigen stimulation and exogenous IL-2. (B) Cell surface expression of CD25 and intracellular expression of FoxP3. (C) Proliferative capacity upon restimulation with allogeneic PBMC (same donor as in alloantigen expansion) in the absence or presence of exogenous IL-2. (D) Suppressive capacity in co-cultures of autologous naïve CD4posCD25neg Tresp stimulated with allogeneic PBMC (same donor as in alloantigen expansion). Significant differences are indicated by asterisks. In figure D, open and filled asterisks refer to CliniMACS versus CliniMACS CD127pos or CliniMACS CD127neg, respectively.

https://doi.org/10.1371/journal.pone.0003161.g005

As shown in Figure 5A, this procedure indeed enabled separation of Treg from Tconv cells. In contrast to CD127pos cells, the majority of CliniMACS CD127neg cells showed a Treg phenotype: CD4posCD25high and FoxP3pos. Since we used cryopreserved CliniMACS populations for these experiments, we expanded the populations prior to functional characterization. After expansion, CD127neg populations retained a high Treg purity, as evidenced by high expression of CD25 and FoxP3 (Figure 5B). In contrast to CD127pos populations, CD127neg populations were anergic after expansion (Figure 5C). As expected, CD127 depletion enhanced suppressive potential, as shown by the significantly higher suppressive capacity of CD127neg populations as compared to CliniMACS populations that were not depleted of CD127pos cells (Figure 5D).

Discussion

Immunotherapy using ex vivo activated and/or expanded CD4posCD25high regulatory T cells appears promising, not only in stem cell transplantation, but also for a number of autoimmune disorders. To be optimally effective, the availability of tailor-made clinical grade procedures is a prerequisite. As previously reported [14], [15], CliniMACS GMP Treg isolation strategies based on CD25pos enrichment typically result in 40–60% pure Treg with moderate suppressive activity.

The degree of purity of Treg that is required may depend on the actual application. In stem cell transplantation, the first clinical trials on Treg immunotherapy have recently been initiated. In these studies, either CliniMACS isolated CD4posCD25high Treg (with 40–60% purity) or ex vivo manipulated CD4pos T cell lines consisting of 5–10% induced regulatory Tr1 cells and consequently a substantial amount of non-Treg were infused in patients receiving stem cell transplantations [24]. So far, to our knowledge, no adverse effects have been reported. However, caution is necessary when applying Treg for auto-immune disease or solid organ transplant purposes. Here an inadvertently present effector pool might cause serious pathology.

As cytotoxic CD8pos T cells are a potentially aggressive cell type in immune responses, it is highly recommendable to exclude these cells from CliniMACS isolated Treg populations. Here, we show that the CD8pos CliniMACS cell depletion step was effective: less than 0.1% CD8pos cells were present in isolated Treg populations, and no outgrowth of CD8pos cells occurred after expansion of CliniMACS Treg populations. Treg isolated by CD25pos enrichment following CD8pos depletion contained a small contamination of B cells (1–3%), which could increase to up to 10% after T cell expansion. The risks of infusion of expanded and activated autologous B cells are unknown, but could in theory include the development of a B cell lymphoproliferative disorder that can occur after transplantation and is often lethal [25][27]. We were able to prevent the contamination by B cells by including a CD19pos B cell depletion step in the isolation strategy.

Notwithstanding the favorable effects of combined anti-CD25, anti-CD8 and anti-CD19 microbead isolation (the currently available clinical grade tools), the isolated Treg population was of suboptimal purity. Most of the contaminating cells appeared to be CD4posCD25neg/low Tconv cells that could potentially lead to immunopathology. To further improve CliniMACS Treg purity, Wichlan et al. studied titration of anti-CD25 microbeads in the enrichment phase of CliniMACS Treg isolation. While the use of lower amounts of beads indeed led to populations with higher CD25 expression levels [15], the suppressive capacity of the isolated populations did not improve and cell yields were significantly lower, indicating that titration of CD25 microbeads will not provide the solution to achieve higher purity for CliniMACS Treg isolation. These authors also reported that adjustment of CliniMACS flow rates did not lead to a higher Treg purity. Hoffmann et al. used three consecutive CliniMACS selection cycles for CD25pos cells, to preferentially select CD25high Treg and exclude CD25neg/low Tconv [14]. However, the degree of Treg purity obtained with this approach was similar to that achieved with one CD25 enrichment cycle.

Recently, it has been described that Tconv express the IL-7 receptor alpha chain CD127, while Treg do not express this molecule [22], [23]. Thus, in the current study, we hypothesized that the conventional T cells contaminating GMP Treg populations could be excluded from the CliniMACS isolated populations by depleting CD127pos cells. Since anti-CD127-microbeads for use in CliniMACS are currently not available, we depleted CD127pos cells from CliniMACS isolated Treg populations by FACS. This procedure indeed increased Treg purity, as shown by uniformly high expression of FoxP3 in CD127neg populations. After expansion of CliniMACS CD127neg populations, high expression of FoxP3pos was maintained, and the cell populations showed potent suppressive capacity. Notably, suppressive capacity of CliniMACS CD127neg populations was significantly higher than that of non-depleted CliniMACS populations, indicating that CD127 depletion clearly enriched for functional Treg.

The outcome of a particular T cell based immune response is likely to be determined by the balance between the effector and regulatory T cell pools. Consequently, the efficacy of Treg therapy may be determined by the actual number of antigen reactive Treg within the infused cell population. Indeed, antigen-specific Treg were proven to be more efficient than polyclonal Treg in preclinical mouse models of autoimmunity [6], [12], [13] and graft-versus-host disease [3], [5], [7]. Previously, we and others have shown that human Treg isolated by non-clinical grade strategies can be expanded using both polyclonal and alloantigen stimulation methods [16][21]. Importantly, we show in this report that this also holds true for clinical grade isolated Treg. Due to the moderate purity of CliniMACS isolated Treg populations, outgrowth of contaminating cells could be a risk upon expansion. Indeed, expanded CliniMACS8/19/25 Treg populations contained a significant percentage of FoxP3neg cells, however, this could be prevented by depletion of CD127pos cells from the CliniMACS isolated Treg populations.

Clinical implementation of Treg based therapy will be highly facilitated if Treg can be stored prior to infusion, as this will allow a more flexible timing of Treg therapy and/or therapeutic schemes with multiple Treg treatments over time. We studied the feasibility of CliniMACS Treg cryopreservation in liquid nitrogen. Results indicate that Treg can survive cryopreservation, as thawed populations showed 70–80% cell viability. We noted decreased suppressive activity of thawed Treg populations, which could be restored by Treg expansion. An alternative approach would be to expand Treg prior to cryopreservation. This resulted in unaltered suppressive capacity upon thawing, at least in MiniMACS isolated Treg (Supplemental data, Figure S1).

In summary, we here provide further support for clinical implementation of Treg immunotherapy by showing that a high Treg purity can be reached, and that isolated cells can be cryopreserved and expanded successfully.

Materials and Methods

Human cells

Healthy donors were scheduled for leukapheresis procedures to obtain leukocytes for donor lymphocyte infusions in hematopoietic stem cell transplantation patients in the Radboud University Nijmegen Medical Centre. Excess leukapheresis material was used for the current study upon written informed consent with regard to scientific use. Buffy coats from healthy human donors were purchased from Sanquin bloodbank, Nijmegen, The Netherlands, upon written informed consent with regard to scientific use and used as a source of stimulator PBMC. The current study did not require approval from an ethics committee according to the Dutch Medical Research Involving Human Subjects Act.

Treg isolation

Healthy donor leukapheresis products were used for CliniMACS CD4posCD25high Treg isolation. Cells were washed with PBS/EDTA buffer (Miltenyi Biotec, Bergisch-Gladbach, Germany, supplemented with 0.5% HSA (Sanquin bloodbank). Anti-CD8 and/or anti-CD19 coated CliniMACS microbeads (kindly provided by Miltenyi Biotec) were added, incubated for 30 minutes and washed. CliniMACS program 2.1 was run to deplete labeled cells (CliniMACS separation columns were kindly provided by Miltenyi Biotec). The labeling procedure was repeated with anti-CD25 CliniMACS microbeads, and CliniMACS program 1.1 was run to enrich for CD25pos cells.

A non-GMP MiniMACS based CD4posCD25high Treg isolation method was performed on leukocytes from the same leukapheresis products as used for CliniMACS Treg isolation. PBMC were isolated by density gradient centrifugation (Lymphoprep, Nycomed Pharma, Roskilde, Denmark). CD4pos T cells were negatively selected using mAbs directed against CD8 (RPA-T8), CD14 (M5E2), CD16 (3G8), CD19 (4G7), CD33 (P67.6), CD235a (GA-R2(HIR2) (BD Biosciences, San Jose, CA, USA), and CD56 (MOC-1) (Dako, Glostrup, Denmark) combined with sheep-anti-mouse-IgG coated magnetic beads (Dynal Biotech, Oslo, Norway), routinely resulting in a >90% pure CD4pos T cell fraction. CD25high Treg and CD25neg conventional T cells (referred to as Tconv, included in all experiments as control cell population) were separated by MACS-sorting, using 10 µl anti-CD25 magnetic microbeads/107 CD4pos cells (Miltenyi Biotec).

For CD127pos cell depletion, CliniMACS isolated Treg were stained with anti-CD127-AlexaFluor647 (BD Biosciences) and CD127pos cells were depleted on an Elite FACS machine (Beckman Coulter, Fullerton, CA, USA).

Stimulator PBMC were isolated from healthy donor buffy coat by density gradient centrifugation (Lymphoprep, Nycomed Pharma).

HLA typing was performed by serological and DNA based techniques according to international (ASHI/EFI) standards [28].

Treg cryopreservation

Treg cells were suspended in RPMI 1640 supplemented with pyruvate (0,02 mM), penicillin (100 U/ml), streptomycin (100 mg/ml), 20% human pooled serum (HPS) and 15% dimethylsulfoxide, were kept in −80°C for one week, and were subsequently transferred to liquid nitrogen for up to one year. Cells were quickly thawed in a 37°C water bath and washed twice before use.

Treg expansion

Cell cultures were performed in culture medium consisting of RPMI 1640 supplemented with pyruvate (0.02 mM), penicillin (100 U/ml), streptomycin (100 mg/ml), 10% HPS and IL-2 (25 U/ml, Chiron, Amsterdam, the Netherlands) with 5×104 cells per well in 96-well round bottom plates, in a 37°C, 95% humidity, 5% CO2 incubator. Stimulation was provided by either 105 irradiated (30 Gy) fully HLA-mismatched allogeneic PBMC per well (alloantigen expansion) or 1×104 anti-CD3+anti-CD28 mAb coated microbeads per well (Dynal Biotech, polyclonal expansion).

Wells were split and provided with fresh medium containing cytokines every 3 days. After 10 days, the cells were harvested, washed and rested for 2 days in 5% HPS culture medium before functional and phenotypic analyses.

Flow cytometry

The phenotype of cells was analyzed by five-color flow cytometry (FC500, Beckman Coulter). For cell surface staining, the following conjugated mAbs were used: anti-CD4(SFCI12T4D11)-PCy7, anti-CD19(J4.119)-PE (Beckman Coulter), anti-CD25(4E3)-biotin, anti-biotin(Bio3-18E7)-PE (used in Figures 1 and 2), anti-CD20(LT20)-PE (Miltenyi Biotec), anti-CD25(M-A251)-PE (used in Figure 5), and anti-CD127(hIL-7R-M21)-AlexaFluor647 (BD Biosciences). For intracellular staining, Fix and Fix/Perm buffer and anti-FoxP3(PCH101)-FITC were used according to the manufacturer's instructions (eBioscience, San Diego, CA, USA). Isotype controls were used for gate settings.

Stimulation assay to analyze T cell anergy

T cell anergy was examined in (re-)stimulation assays. 2.5×104 cells were stimulated with 105 irradiated allogeneic stimulator PBMC in the presence or absence of IL-2 (12.5 U/ml). Proliferation was measured at day 5 (primary responses) or day 3 (secondary responses). To this end, 0.5 µCi [3H]Thymidine (Amersham Biosciences, Piscataway, NJ) was added to each well. After 8 hours, [3H]Thymidine incorporation was measured using a beta-plate counter (Packard, Canberra, Australia). Tests were set up in triplicate; results were expressed as mean+SD counts per 5 minutes.

Co-culture suppression assays

Suppressive capacity of Treg was studied in co-culture suppression assays. 5×104 autologous responder T cells (CD4posCD25neg, Tresp) were stimulated with 105 irradiated allogeneic stimulator PBMC. Treg were titrated into these cultures. Proliferation was measured at day 5 by determination of [3H]Thymidine incorporation as described above. Tests were set up in triplicate, results were expressed as mean+SD counts per 5 minutes.

Statistical analysis

Proliferative capacities of different cell populations were compared using Student T tests. A P value<0.05 was considered statistically significant.

Supporting Information

Figure S1.

Unaffected suppressive capacity of MiniMACS Treg by expansion prior to cryopreservation. Suppressive capacity of cryopreserved MiniMACS Treg isolated from healthy donor buffy coat in co-cultures of autologous naïve CD4posCD25neg Tresp stimulated with allogeneic PBMC. Data from one of two similar experiments are shown. Prior to co-culture suppression assay, indicated cell populations were expanded for 10 days in the presence of exogenous IL-2 and allogeneic PBMC (same donor as in co-culture suppression assay). Significant differences are indicated by asterisks. Open and filled asterisks refer to cryopreservation versus cryopreservation followed by expansion or expansion followed by cryopreservation, respectively.

https://doi.org/10.1371/journal.pone.0003161.s001

(0.12 MB TIF)

Acknowledgments

We thank Paul Ruijs, Carel Trilsbeek and Jeroen van Velzen for excellent technical assistance.

Author Contributions

Conceived and designed the experiments: JHP HJPMK IJ. Performed the experiments: JHP RW. Analyzed the data: JHP HJPMK IJ. Wrote the paper: JHP FWP LBH HJPMK IJ.

References

  1. 1. Edinger M, Hoffmann P, Ermann J, Drago K, Fathman CG, et al. (2003) CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med 9: 1144–1150.
  2. 2. Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S (2002) Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med 196: 389–399.
  3. 3. Joffre O, Gorsse N, Romagnoli P, Hudrisier D, van Meerwijk JP (2004) Induction of antigen-specific tolerance to bone marrow allografts with CD4+CD25+ T lymphocytes. Blood 103: 4216–4221.
  4. 4. Jones SC, Murphy GF, Korngold R (2003) Post-hematopoietic cell transplantation control of graft-versus-host disease by donor CD425 T cells to allow an effective graft-versus-leukemia response. Biol Blood Marrow Transplant 9: 243–256.
  5. 5. Trenado A, Charlotte F, Fisson S, Yagello M, Klatzmann D, et al. (2003) Recipient-type specific CD4+CD25+ regulatory T cells favor immune reconstitution and control graft-versus-host disease while maintaining graft-versus-leukemia. J Clin Invest 112: 1688–1696.
  6. 6. Tarbell KV, Yamazaki S, Olson K, Toy P, Steinman RM (2004) CD25+ CD4+ T cells, expanded with dendritic cells presenting a single autoantigenic peptide, suppress autoimmune diabetes. J Exp Med 199: 1467–1477.
  7. 7. Yamazaki S, Patel M, Harper A, Bonito A, Fukuyama H, et al. (2006) Effective expansion of alloantigen-specific Foxp3+ CD25+ CD4+ regulatory T cells by dendritic cells during the mixed leukocyte reaction. Proc Natl Acad Sci U S A 103: 2758–2763.
  8. 8. Bushell A, Jones E, Gallimore A, Wood K (2005) The generation of CD25+ CD4+ regulatory T cells that prevent allograft rejection does not compromise immunity to a viral pathogen. J Immunol 174: 3290–3297.
  9. 9. Golshayan D, Jiang S, Tsang J, Garin MI, Mottet C, et al. (2006) In vitro expanded donor alloantigen-specific CD4+CD25+ regulatory T cells promote experimental transplantation tolerance. Blood 6: 1879–1882.
  10. 10. Kohm AP, Carpentier PA, Anger HA, Miller SD (2002) Cutting edge: CD4+CD25+ regulatory T cells suppress antigen-specific autoreactive immune responses and central nervous system inflammation during active experimental autoimmune encephalomyelitis. J Immunol 169: 4712–4716.
  11. 11. Mottet C, Uhlig HH, Powrie F (2003) Cutting edge: cure of colitis by CD4+CD25+ regulatory T cells. J Immunol 170: 3939–3943.
  12. 12. Tang Q, Henriksen KJ, Bi M, Finger EB, Szot G, et al. (2004) In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes. J Exp Med 199: 1455–1465.
  13. 13. Masteller EL, Warner MR, Tang Q, Tarbell KV, McDevitt H, et al. (2005) Expansion of functional endogenous antigen-specific CD4+CD25+ regulatory T cells from nonobese diabetic mice. J Immunol 175: 3053–3059.
  14. 14. Hoffmann P, Boeld TJ, Eder R, Albrecht J, Doser K, et al. (2006) Isolation of CD4+CD25+ regulatory T cells for clinical trials. Biol Blood Marrow Transplant 12: 267–274.
  15. 15. Wichlan DG, Roddam PL, Eldridge P, Handgretinger R, Riberdy JM (2006) Efficient and reproducible large-scale isolation of human CD4(+) CD25(+) regulatory T cells with potent suppressor activity. J Immunol Methods 315: 27–36.
  16. 16. Earle KE, Tang Q, Zhou X, Liu W, Zhu S, et al. (2005) In vitro expanded human CD4+CD25+ regulatory T cells suppress effector T cell proliferation. Clin Immunol 115: 3–9.
  17. 17. Hoffmann P, Eder R, Kunz-Schughart LA, Andreesen R, Edinger M (2004) Large-scale in vitro expansion of polyclonal human CD4(+)CD25high regulatory T cells. Blood 104: 895–903.
  18. 18. Karakhanova S, Munder M, Schneider M, Bonyhadi M, Ho AD, et al. (2006) Highly efficient expansion of human CD4+CD25+ regulatory T cells for cellular immunotherapy in patients with graft-versus-host disease. J Immunother 29: 336–349.
  19. 19. Kreijveld E, Koenen HJ, Hilbrands LB, Joosten I (2006) Ex vivo expansion of human CD4(+)CD25(high) regulatory T cells from transplant recipients permits functional analysis of small blood samples. J Immunol Methods 314: 103–113.
  20. 20. Koenen HJ, Fasse E, Joosten I (2005) CD27/CFSE-based ex vivo selection of highly suppressive alloantigen-specific human regulatory T cells. J Immunol 174: 7573–7583.
  21. 21. Peters JH, Hilbrands LB, Koenen HJ, Joosten I (2008) Ex vivo generation of human alloantigen-specific regulatory T cells from CD4(pos)CD25(high) T cells for immunotherapy. PLoS ONE 3: e2233.
  22. 22. Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, et al. (2006) CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 203: 1701–1711.
  23. 23. Seddiki N, Santner-Nanan B, Martinson J, Zaunders J, Sasson S, et al. (2006) Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med 203: 1693–1700.
  24. 24. Roncarolo MG, Battaglia M (2007) Regulatory T-cell immunotherapy for tolerance to self antigens and alloantigens in humans. Nat Rev Immunol 7: 585–598.
  25. 25. Gross TG, Steinbuch M, DeFor T, Shapiro RS, McGlave P, et al. (1999) B cell lymphoproliferative disorders following hematopoietic stem cell transplantation: risk factors, treatment and outcome. Bone Marrow Transplant 23: 251–258.
  26. 26. Taylor AL, Marcus R, Bradley JA (2005) Post-transplant lymphoproliferative disorders (PTLD) after solid organ transplantation. Crit Rev Oncol Hematol 56: 155–167.
  27. 27. Curtis RE, Travis LB, Rowlings PA, Socie G, Kingma DW, et al. (1999) Risk of lymphoproliferative disorders after bone marrow transplantation: a multi-institutional study. Blood 94: 2208–2216.
  28. 28. Koenen HJ, Fasse E, Joosten I (2003) IL-15 and cognate antigen successfully expand de novo-induced human antigen-specific regulatory CD4+ T cells that require antigen-specific activation for suppression. J Immunol 171: 6431–6441.