A major goal of the transplant field is to selectively tolerize only those donor T cells recognizing host alloantigen and mediating graft-vs-host disease (GVHD). Recently, we described an ex vivo approach in which the blockade of the CD40 ligand (CD40L):CD40 costimulatory pathway in bulk MLR cultures induces donor CD4+ T cells to become specifically tolerant to MHC class II-disparate alloantigenic-bearing stimulators, resulting in a profound reduction in GVHD generation in vivo. In studies presented in this work, we investigated the ex vivo requirements for tolerance induction. We found that CD4+ T cells become profoundly more hyporesponsive to alloantigen restimulation with prolonged culture duration such that 7 to 10 but not 4 days is needed to achieve maximum alloantigen hyporesponsiveness as assessed in secondary MLR cultures and GVHD generation. By day 7, both primed and tolerized cells had substantially increased blastogenesis and CD25 expression. Primed but not tolerized cells substantially down-regulated L-selectin expression, indicating that the tolerized cells do not become fully Ag experienced. Both Th1 and Th2 cytokine production is severely impaired by CD40L:CD40 blockade. Analysis of culture supernatants and results from IL-4 and IL-10 knockout mice indicated that GVHD prevention was not mediated by a skewing toward a Th2 phenotype. The addition of IL-4 to the cultures as a survival factor precluded the induction of tolerance in the anti-CD40L-cultured cells. These data provide further impetus for the ex vivo use of anti-CD40L mAb to block GVHD generation.

Bone marrow transplantation (BMT)3 is being utilized as the standard of care for the treatment of a number of malignant and nonmalignant disorders of both hematologic and nonhematologic origin. Graft-vs-host disease (GVHD) remains a major cause of morbidity and mortality after sibling, matched unrelated, haploidentical, or mismatched bone marrow transplantion (1). Although GVHD can be prevented by rigorous T cell depletion ex vivo of the donor graft or prolonged, global immunosuppression in vivo in the recipient, these strategies have associated risks of increased rates of leukemic relapse, infectious complications, or graft failure (1). A strategy that would selectively target only that small fraction of donor T cells capable of mediating GVHD but preserve those T cells that might mediate antitumor, anti-infectious effects and promote engraftment would be desirable. An in vitro blockade strategy that would not involve global in vivo immune suppression or nonspecific multiorgan toxicity also would be advantageous.

Productive T cell activation and proliferation require two signaling events. The first signal is the engagement of the TCR with the MHC-peptide ligand complex on the surface of the APC. Additional costimulatory signals are required for the full activation of the intracellular signaling cascade, IL-2 production, and ultimate T cell proliferation (2, 3, 4, 5, 6, 7). Multiple pathways capable of costimulation have been described, including CD40L:CD40, CD28/CTLA4:B7-1/B7-2, OX40:OX40L, 41BB:41BBL, CD30:CD30L, LFA-1/ICAM-1, CD27:CD70, and CD2:CD48. CD40L is a member of the TNF family and is transiently expressed on activated CD4+ T cells (7, 8). The counter-receptor, CD40, is a member of the TNFR family and is found on APCs, including B cells, bone marrow-dendritic cells, follicular dendritic cells, and activated macrophages (7, 8). The CD40L:CD40 pathway was initially described for its importance in B and T cell interactions, B cell activation, Ig production, and isotype switching. Subsequently, CD40L:CD40 interactions were found to be essential for the initiation and activation of Ag-specific T cell effector functions and the activation of costimulatory activity (including the up-regulation of B7 molecules) on APCs, including B cells, macrophages, and dendritic cells (7). CD40L is expressed early in activation subsequent to the engagement of the TCR with the MHC-peptide ligand complex on the surface of the APC. CD40L transduces a signal to its counter-receptor, CD40, constitutively expressed on APCs, resulting in the up-regulation of additional molecules involved in further T cell costimulation (7, 9).

Administration of anti-CD40L mAb in vivo has been effective in preventing collagen-induced arthritis and ameliorating GVHD in murine models (10, 11). The in vivo administration of B cells from CD40-deficient mice or the simultaneous administration of donor splenocytes with anti-CD40L mAb has conferred alloantigen-specific tolerance to recipient mice (12, 13). In primates, administration of anti-CD40L mAb in vivo has been shown to prevent acute renal and intrahepatic islet allograft rejection (14, 15, 16). Although the effect of in vivo administration of anti-CD40L mAb has been impressive in preventing the rejection of solid organ allografts, we found that the ex vivo tolerization of CD4+ T cells led to a more profound reduction in GVHD lethality than did the in vivo administration of anti-CD40L mAb in the same setting (11, 17). In vivo tolerance may be more difficult to achieve after BMT due to the 1) induction of proinflammatory cytokines and increased expression of costimulatory molecules on APCs as a result of the intense conditioning protocols used for BMT (18, 19), and 2) difficulty of achieving complete in vivo blockade at all possible sites of allorecognition. As an undesired side effect, the in vivo blockade of costimulation could result in the induction of tolerance to tumor Ags as well as alloantigens, preventing an effective graft vs tumor effect.

We have previously described an ex vivo approach in which the blockade of CD40L:CD40 interactions during a 10-day culture induces donor CD4+ T cells to become tolerant to host alloantigens, resulting in a ≥30-fold reduction in GVHD lethality with no additional in vivo immunosuppression (17). Additionally, the tolerized bulk T cell population retained intact responses to Ags not present during tolerization. Tolerance was long-lived and not readily reversible in vivo. In these studies, we sought to determine the prerequisites for tolerance induction in this ex vivo culture system.

B6.C-H2bm12/KhEg (termed bm12), B10.D2 (H2d), and inbred C57BL/6 IL-10−/− mice were purchased from The Jackson Laboratory (Bar Harbor, ME). C57BL/6 (termed B6) were purchased from the National Institutes of Health (Bethesda, MD). (B6 × 129SV)F1 IL-4−/− mice, backcrossed six generations onto a B6 background, were obtained from Dr. Manfred Kopf (Basel, Switzerland). bm12 and B6 (both H2b) mice differ at three amino acids due to mutations in the IA region. Mice were used at 9–10 wk of age. All mice were housed in a specific pathogen-free facility in microisolator cages.

To purify B6 CD4+ T cells, axillary, mesenteric, and inguinal lymph nodes were mashed, and single cell suspensions were passed through a wire mesh and collected into PBS containing 2% FBS. Cell preparations were depleted of NK cells (hybridoma PK136, rat IgG2a, provided by Dr. Gloria Koo, Rahway, NJ) and CD8+ T cells (hybridoma 2.43, rat IgG2b, provided by Dr. David Sachs, Charlestown, MA) by coating with mAb, followed by passage through a goat anti-mouse and goat anti-rat Ig-coated column (Biotex, Edmonton, Canada). The final composition of purified T cells was determined by flow-cytometric analysis to be ≥94% CD4+ T cells. Responder B6 CD4+ T cells were mixed with irradiated (30 Gy), anti-Thy-1.2 mAb (hybridoma 30H-12, rat IgG2b, provided by Dr. David Sachs), and anti-NK1.1 mAb plus baby rabbit complement (Nieffenegger, Woodland, CA)-depleted bm12 splenic stimulators. Responder and stimulator cells were suspended at a final concentration of 0.5 × 106/ml in 24-well plates (Costar, Acton, MA) containing DMEM (BioWhittaker, Walkersville, MD) with 10% FBS (HyClone, Logan, UT), 50 mM 2-ME (Sigma, St. Louis, MO), 10 mM HEPES buffer, 1 mM sodium pyruvate (Life Technologies, Grand Island, NY), and amino acid supplements (1.5 mM l-glutamine, l-arginine, and l-asparagine) (Sigma) and antibiotics (penicillin, 100 U/ml; streptomycin, 100 mg/ml) (Sigma). Anti-CD40L mAb (hybridoma MR1, hamster IgG) was obtained by culturing the hybridoma in 10% FBS/DMEM in a hollow fiber bioreactor (AccucystJr, Cellex Biosciences, Minneapolis, MN). Supernatant was purified by ammonium sulfate precipitation. Anti-CD40L mAb was added at a final concentration of 50 μg/ml to primary MLR cultures. Plates were incubated at 37°C and 10% CO2 for 4, 7, or 10 days. On day 5, the culture was fed 1:1 with new media including mAb. To monitor primary MLR proliferation, 96-well round-bottom microtiter plates (Costar) were set up to contain 105 responders and 105 stimulators cells per well in the presence or absence of exogenous IL-2 (50 IU/ml) (Amgen, Thousand Oaks, CA). In some experiments, murine IL-4 (Schering-Plough, Kenilworth, NJ) (sp. act., 1.5 × 107 U/mg) was added at the indicated doses at the initiation of culture and readded at the day 5 refeed.

To monitor secondary MLR proliferation, 3 × 104 washed, adjusted responders and 105 irradiated (30 Gy) non-T cell-depleted stimulators were plated in the presence or absence of IL-2 (50 IU/ml). Anti-CD40L mAb was not present in the secondary MLR. Microtiter wells were pulsed with tritiated thymidine (1 μCi/well; Amersham Life Science, Buckinghamshire, U.K.) on the indicated days for 16–18 h before harvesting and counted in the absence of scintillation fluid on a β plate reader (Packard Instrument Company, Meriden, CT). Six wells were analyzed for each data point.

Proliferating T lymphocyte precursor frequency analysis was accomplished by setting up eight 3-fold serial dilutions of responder B6 CD4+ T cells at 30 replicates in 96-well round-bottom plates and incubating for 7 days with irradiated (30 Gy), non-T cell-depleted bm12 splenic stimulators in the absence of IL-2. Anti-CD40L mAb was added to the primary LDA at a final concentration of 50 μg/ml. Secondary LDA were established with washed, 10-day cultured control-primed or anti-CD40L-tolerized responder cells and bm12 or third-party B10.D2 splenic stimulators. Anti-CD40L mAb was not present in the secondary LDA. Wells were pulsed with tritiated thymidine for 18 h before harvesting and counted in the absence of scintillation fluid on a β plate reader. Wells were scored positive if their cpm exceeded the average cpm plus 3 SDs of the stimulators plated without responders. Using Poisson distribution statistics according to the method of Taswell and with the aid of a computer program, the likelihood of a single hit was confirmed and a frequency estimate calculated.

Washed 10-day cultured control-primed or anti-CD40L-tolerized cells were incubated 4 h with 104 tritiated thymidine-labeled syngeneic or class II-disparate Con A (Sigma)-stimulated splenocyte blasts at various E:T ratios according to the JAM CTL assay (20). Wells, plated in triplicate, were harvested and counted in the absence of scintillation fluid on a β plate reader. Percent lysis was calculated according to the equation: {[(cpm of targets in absence of killers) − (cpm of targets in presence of killers)]/(cpm of targets in absence of killers)} × 100.

bm12 recipients were sublethally irradiated by exposing mice to 6 Gy total body irradiation from a 137Cesium source at a dose rate of 85 cGy/min. Day 4, 7, or 10 MLR-cultured cells were injected i.v. at the doses indicated. Peripheral blood was obtained by retroorbital venipuncture for measurement of day 14 and 28 hematocrit (HCT) values as an indicator of the bone marrow-destructive effects of infused T cells.

Freshly purified and day 4, 7, and 10 MLR-cultured cells were assessed for evidence of activation by forward scatter (FSC) and side scatter (SSC) profiles and the coexpression of CD4 and activation Ags, including CD25, L-selectin (CD62L), and CD40L. All studies were performed with two-color flow cytometry using fluorescein- and PE-conjugated mAb (PharMingen, San Diego, CA). All results were obtained using a FACSCalibur (Becton Dickinson, San Jose, CA). FSC and SSC settings were gated to exclude debris. A total of 10,000 cells were analyzed for each determination.

Murine cytokine levels in the supernatant of MLR cultures were quantitated by ELISA (R&D Systems, Minneapolis, MN). Sensitivity of the assays was between 1 and 10 pg/ml for each assay. A standard curve using recombinant protein was generated with each assay.

Survival data were analyzed by life-table methods, and actuarial survival rates are shown. Group comparisons were made by log-rank test statistics. For other data, group comparisons were made by Student’s t test. Values of p ≤ 0.05 were considered significant.

Studies by other investigators have indicated that the induction of anergy in mouse T cell clones, human T cell clones, and primary T cells requires from 16 h up to 10 days of primary culture (5, 21, 22, 23, 24, 25, 26). We have previously reported a 10-day ex vivo tolerization approach of primary murine CD4+ T cells, which results in profound secondary in vitro hyporesponsiveness and a ≥30-fold protection from GVHD mortality (17). In the studies presented in this work, we sought to determine the minimum culture duration required for the induction of alloantigen hyporesponsiveness in vitro and inhibition of GVHD in vivo. Purified B6 CD4+ lymph node T cells were mixed with irradiated, T cell-depleted, MHC class II-disparate bm12 splenic stimulators in the presence or absence of anti-CD40L mAb. On days 4, 7, and 10, aliquots of cells were washed to remove Ab and cytokines, and cells were either infused in vivo into irradiated bm12 recipients or reexposed in vitro to irradiated bm12 splenic stimulators in the absence of anti-CD40L mAb. Fig. 1,A illustrates the proliferative responses in the primary culture. The addition of anti-CD40L mAb to the cultures progressively reduced proliferation by 21% (day 2), 91% (day 4), and 99% (day 6) as compared with the control cultures. The addition of exogenous IL-2 (50 U/ml) to the cultures prevented inhibition of proliferation by anti-CD40L mAb, with both groups having equivalent proliferative responses (Fig. 1,B). Fig. 2,A–C illustrates the secondary proliferative responses of the cultures established after 4, 7, or 10 days in primary culture in the presence of anti-CD40L mAb. All secondary cultures were established in the absence of anti-CD40L mAb and were derived from the same primary culture. Because our unpublished data have indicated that a 1- to 3-day primary culture has an impaired capacity to mediate lethal GVHD in vivo (P. A. Taylor and B. R. Blazar, unpublished observation), we chose 4 days as the shortest culture period to study. Upon reexposure to alloantigen restimulation in the secondary MLR, the day 4 control culture peaked on day 3 instead of day 6 as in the primary culture indicative of priming (Fig. 2,A). Responses of cells exposed to anti-CD40L mAb in the primary culture for 4 days were inhibited by 45–74% during the first 3 days of the secondary culture. In secondary MLRs established after 7 days of primary culture, the control group had an early vigorous response upon reexposure to alloantigen, peaking on day 1 and gradually declining (Fig. 2,B). This rapid secondary response by day 7 control-primed cells has been reproducibly seen in these cultures. Responses of cells exposed to anti-CD40L mAb in the primary culture for 7 days were inhibited by 71–81% during the first 3 days of the secondary culture. Seven-day anti-CD40L cultures were more hyporesponsive upon alloantigen restimulation than 4-day anti-CD40L cultures. The peak magnitude of the 7-day anti-CD40L-cultured cells was 5400 cpm compared with 10,000 cpm for the 4-day anti-CD40L-cultured cells. As compared with cultures established from 4 or 7 days of primary culture, secondary MLR cultures established from 10-day anti-CD40L-cultured cells were more profoundly hyporesponsive to alloantigen restimulation. At the time of peak secondary response, the proliferative response of the 10-day anti-CD40L-cultured cells was inhibited by 95% as compared with the control-primed cells (Fig. 2 C). The peak magnitude of the 10-day anti-CD40L-cultured cells was 2700 cpm. These data indicate that hyporesponsiveness upon alloantigen restimulation becomes more profound with prolonged ex vivo blockade of the CD40:CD40L pathway. As in the primary MLR, the addition of IL-2 fully restored secondary responses to alloantigen restimulation (data not shown).

FIGURE 1.

Anti-CD40L mAb induces alloantigen hyporesponsiveness in primary MLR culture, which is restored by exogenous IL-2. Primary MLR culture consisted of B6 CD4+ lymph node T cell responders and bm12 splenic stimulators plated in the absence (A) and presence (B) of exogenous IL-2 (50 U/ml). Anti-CD40L mAb was added at 50 μg/ml at the initiation of culture. On the y-axis are mean cpm ± 1 SD of the mean. On the x-axis are days in primary culture. A representative experiment is shown.

FIGURE 1.

Anti-CD40L mAb induces alloantigen hyporesponsiveness in primary MLR culture, which is restored by exogenous IL-2. Primary MLR culture consisted of B6 CD4+ lymph node T cell responders and bm12 splenic stimulators plated in the absence (A) and presence (B) of exogenous IL-2 (50 U/ml). Anti-CD40L mAb was added at 50 μg/ml at the initiation of culture. On the y-axis are mean cpm ± 1 SD of the mean. On the x-axis are days in primary culture. A representative experiment is shown.

Close modal
FIGURE 2.

Anti-CD40L mAb-cultured CD4+ T cells become profoundly more hyporesponsive to alloantigen restimulation with prolonged primary MLR culture duration. Secondary MLRs were established in the absence of mAb after a 4 (A)-, 7 (B)-, or 10 (C)-day primary MLR culture in the presence of anti-CD40L mAb. On the y-axis are mean cpm ± 1 SD of the mean. On the x-axis are days in secondary culture. A representative experiment is shown.

FIGURE 2.

Anti-CD40L mAb-cultured CD4+ T cells become profoundly more hyporesponsive to alloantigen restimulation with prolonged primary MLR culture duration. Secondary MLRs were established in the absence of mAb after a 4 (A)-, 7 (B)-, or 10 (C)-day primary MLR culture in the presence of anti-CD40L mAb. On the y-axis are mean cpm ± 1 SD of the mean. On the x-axis are days in secondary culture. A representative experiment is shown.

Close modal

As other measures of in vitro function, we determined the frequency of B6 CD4+ cells proliferating in response to bm12 alloantigen and the CTL killing of bm12 targets after 10 days of primary culture with anti-CD40L mAb. Analysis of a 7-day LDA established on day 0 of primary culture in the absence of IL-2 showed that the frequency of B6 CD4+ cells responding to bm12 alloantigen was 1:5,887 in the absence of mAb and 1:22,352 in the presence of mAb, a 4-fold reduction. When LDAs were established with washed, 10-day cultured cells, the frequency increased about 15-fold in the control group to 1:404. In contrast, the frequency in the anti-CD40L-tolerized cells remained the same as in the primary culture (1:24,602). Although the frequency of anti-CD40L mAb-tolerized B6 CD4+ responding to bm12 alloantigen restimulation was decreased about 60-fold compared with the control-primed cells, the frequency of third party B10.D2 allo-responses was only decreased by less than 2-fold, indicating a high degree of specificity (data not shown).

Because other investigators have reported a state of split anergy in which anergic murine T cell clones can be rendered hyporesponsive to Ag reexposure in terms of proliferation and cytokine production but have intact CTL (27, 28), we placed washed 10-day control-primed cells and anti-CD40L-tolerized cells in a 4-h CTL assay against bm12 Con A splenocyte blasts at various E:T ratios. The percent specific lysis of bm12 targets by the 10-day anti-CD40L-tolerized cells was approximately one-fourth that of the control-primed cells (Fig. 3). Consistent with these data, there was an 8–10-fold reduction in the percentage of anti-CD40L-tolerized cells staining positive for granzymes A and B mRNA (data not shown). Granzymes, serine proteases stored in secretory CTL granules, induce DNA fragmentation and CTL lysis of target cells (29, 30). Therefore, a prolonged culture with anti-CD40L mAb prevented the generation of optimal CTL effector function.

FIGURE 3.

Anti-CD40L-tolerized B6 CD4+ T cells have reduced CTL killing of bm12 targets. Ten-day control-primed or anti-CD40L-tolerized B6 CD4+ T cells were washed and plated in a 4-h CTL assay with bm12 targets at various E:T ratios. On the y-axis is percent specific lysis ± 1 SD. On the x-axis is the E:T ratio.

FIGURE 3.

Anti-CD40L-tolerized B6 CD4+ T cells have reduced CTL killing of bm12 targets. Ten-day control-primed or anti-CD40L-tolerized B6 CD4+ T cells were washed and plated in a 4-h CTL assay with bm12 targets at various E:T ratios. On the y-axis is percent specific lysis ± 1 SD. On the x-axis is the E:T ratio.

Close modal

To determine whether anti-CD40L mAb treatment prevented T cell activation, control- and anti-CD40L-cultured cells were evaluated by flow cytometry for their degree of blastogenesis and activation (Fig. 4). Although there was up-regulation of CD40L in the control cultures by day 4, there was very little increase in cell size (FSC) or internal complexity (SSC) at this time. Additionally, CD25 expression was low in both groups. L-selectin (CD62L), a homing receptor lost during activation (31), was marginally down-regulated in only 10% of cells in the control cultures on day 4. By day 7, both primed and tolerized cells had substantially increased FSC, SSC (although values were higher in the primed cells as compared with the tolerized cells), and CD25 expression (more than 50% positive in both groups), indicating a greater degree of activation than day 4 MLR cells (Fig. 4). Primed cells had substantially down-regulated or lost expression of L-selectin by day 7. CD25 expression remained high on day 10 in both groups, with 62% positive in the anti-CD40L-cultured cells compared with 32% positive in the control-primed cells. Notably, L-selectin remained high in 85% of the tolerized cells, while more than one-half of the primed cells lost expression (Fig. 4). We conclude from these data that although both control- and anti-CD40L-cultured cells exhibit marked evidence of blastogenesis and activation by day 7, the tolerized cells do not become fully Ag experienced based upon high L-selectin expression.

FIGURE 4.

Seven- and ten-day MLR cultures exhibit blastogenesis and activation. Primary MLR cultured cells were analyzed by flow cytometry on days 0, 4, 7, and 10 for evidence of T cell activation. FSC and SSC overlay histograms are shown in the first two rows. The thin solid line represents the control-cultured cells, and the bold line represents the anti-CD40L-cultured cells. The mean values are shown in the upper right of the histogram, with the value of the control-cultured cells followed by that of the anti-CD40L-cultured cells. The last three rows illustrate overlay histograms of CD25, L-selectin, and CD40L expression. The dotted line represents the negative isotype control; the thin solid line, the control-cultured cells; and the bold line, the anti-CD40L-cultured cells.

FIGURE 4.

Seven- and ten-day MLR cultures exhibit blastogenesis and activation. Primary MLR cultured cells were analyzed by flow cytometry on days 0, 4, 7, and 10 for evidence of T cell activation. FSC and SSC overlay histograms are shown in the first two rows. The thin solid line represents the control-cultured cells, and the bold line represents the anti-CD40L-cultured cells. The mean values are shown in the upper right of the histogram, with the value of the control-cultured cells followed by that of the anti-CD40L-cultured cells. The last three rows illustrate overlay histograms of CD25, L-selectin, and CD40L expression. The dotted line represents the negative isotype control; the thin solid line, the control-cultured cells; and the bold line, the anti-CD40L-cultured cells.

Close modal

To determine the length of time in culture required to induce tolerance induction in vivo, aliquots on days 4, 7, and 10 of the same primary cultures used to establish the above secondary MLRs were washed and infused into sublethally irradiated recipients bearing the same alloantigen (bm12) used as stimulator cells in the primary MLR culture. Data from our laboratory suggest cells cultured for shorter periods (1–4 days) have a reduced capacity to mediate GVHD lethality (P. A. Taylor and B. R. Blazar, unpublished observation). Therefore, we infused 106, a higher number of cells, or 105 cells, which is typically lethal to all recipients. In contrast to our previous data demonstrating no GVHD lethality with the infusion of 106 10-day anti-CD40L mAb-tolerized cells (17), a 4-day culture period was insufficient to confer uniform GVHD protection. All recipients of 106 control-primed or anti-CD40L-cultured cells died of GVHD-induced BM aplasia by day 25 after infusion of cells (Fig. 5,A). Sixty percent of mice receiving 105 anti-CD40L-cultured cells vs 20% of mice receiving 105 control-primed cells survived the 65-day observation period (p = 0.078 vs control). As an indicator of donor CD4+ T cell-mediated GVHD-induced bone marrow aplasia, HCT values were assessed in all mice on day 14 posttransfer of cells (Table I). There were no statistically signifi- cant differences in HCT values between groups at either cell dose from 4-day cultured cells. Collectively, these data indicate that 4 days of primary culture with anti-CD40L mAb are insufficient to confer a high level of tolerance in vitro. Additionally, the relatively moderate degree of hyporesponsiveness seen in secondary MLR established from 4-day anti-CD40L-cultured cells did not translate to protection from GVHD lethality in vivo.

FIGURE 5.

A 7-, but not 4-day primary MLR culture with anti-CD40L mAb is sufficient to confer protection from GVHD lethality. Four-, 7-, or 10-day MLR cells cultured in the presence of anti-CD40L mAb were washed and infused into sublethally irradiated bm12 recipients at the indicated cell doses (n = 5/group). On the x-axis are days posttransfer of T cells. On the y-axis is the proportion of recipients surviving posttransfer. (Four-day MLR, 106 cells, p = 0.209; 105 cells, p = 0.078; 7-day MLR, 3 × 105 cells, p = 0.0037; 105 cells, p = 0.0018; 10-day MLR, 3 × 105 cells, p = 0.0019; 105 cells, p = 0.0016.)

FIGURE 5.

A 7-, but not 4-day primary MLR culture with anti-CD40L mAb is sufficient to confer protection from GVHD lethality. Four-, 7-, or 10-day MLR cells cultured in the presence of anti-CD40L mAb were washed and infused into sublethally irradiated bm12 recipients at the indicated cell doses (n = 5/group). On the x-axis are days posttransfer of T cells. On the y-axis is the proportion of recipients surviving posttransfer. (Four-day MLR, 106 cells, p = 0.209; 105 cells, p = 0.078; 7-day MLR, 3 × 105 cells, p = 0.0037; 105 cells, p = 0.0018; 10-day MLR, 3 × 105 cells, p = 0.0019; 105 cells, p = 0.0016.)

Close modal
Table I.

Sublethally irradiated bm12 recipients of 7- or 10-day anti-CD40L tolerized cells are protected from GVHD-induced BM aplasiaa

Culture DayGroupNo. of Cells InfusedHCT (%)
Control 106 24.4 ± 4.3 
Anti-CD40L 106 26.6 ± 4.5 
Control 105 30.9 ± 9.9 
Anti-CD40L 105 35.9 ± 7.8 
Control 3 × 105 17.1 ± 4.5 
Anti-CD40L 3 × 105 27.8 ± 3.0b 
Control 105 23.5 ± 7.2 
Anti-CD40L 105 27.8 ± 2.0 
10 Control 3 × 105 14.0 ± 2.8 
10 Anti-CD40L 3 × 105 28.1 ± 5.9b 
10 Control 105 15.3 ± 3.7 
10 Anti-CD40L 105 27.9 ± 2.4b 
Culture DayGroupNo. of Cells InfusedHCT (%)
Control 106 24.4 ± 4.3 
Anti-CD40L 106 26.6 ± 4.5 
Control 105 30.9 ± 9.9 
Anti-CD40L 105 35.9 ± 7.8 
Control 3 × 105 17.1 ± 4.5 
Anti-CD40L 3 × 105 27.8 ± 3.0b 
Control 105 23.5 ± 7.2 
Anti-CD40L 105 27.8 ± 2.0 
10 Control 3 × 105 14.0 ± 2.8 
10 Anti-CD40L 3 × 105 28.1 ± 5.9b 
10 Control 105 15.3 ± 3.7 
10 Anti-CD40L 105 27.9 ± 2.4b 
a

Sublethally irradiated bm12 recipients (n = 5/group) were infused with the indicated number of cells from 4-, 7-, or 10-day control or anti-CD40L-treated cultures. On day 14 postinfusion, hematocrit (HCT) values were determined in all surviving mice. Average percent HCT is shown as mean ± 1 SD.

b

, p < 0.05, control vs. anti-CD40L cultures.

In contrast, a 7-day MLR culture in the presence of anti-CD40L mAb was sufficient to confer GVHD protection at the cell doses used in these studies. All recipients of both cell doses of control-cultured cells (105 or 106) died of GVHD-induced BM aplasia by day 23 (Fig. 5,B). With the exception of one unusually late death at the higher cell dose, all recipients of anti-CD40L-tolerized cells survived the observation period (Fig. 5,B) and had normal HCT values at the time of elective sacrifice on day 65 (data not shown). As expected, all recipients of 10-day anti-CD40L-tolerized cells survived long term (Fig. 5,C) and had normal HCT values at the time of elective sacrifice (data not shown), while all recipients of control-cultured cells died by day 16 (Fig. 5 C). Although there were no significant differences in GVHD mortality at the cell doses used in this study between a 7- and a 10-day primary culture with anti-CD40L mAb, the time to death and day 14 HCT values suggest that 10-day control-primed cells may have a greater GVHD capacity than those cultured for 7 days. Mice receiving 105 10-day control cells succumbed to GVHD mortality 1 wk earlier than those receiving 105 7-day control cells (p = 0.050). Additionally, the day 14 HCT values in mice receiving 105 control cells suggest a greater degree of aplasia with increasing culture duration (4-day MLR HCT, 30.9; 7-day MLR HCT, 23.5; 10-day MLR HCT, 15.3). These data suggest that although a 7-day ex vivo tolerization period confers protection from GVHD lethality, a 10-day ex vivo tolerization period may confer even greater protection.

Fowler et al. (32) and Krenger et al. (40) have found that in vitro generated, alloreactive Th2 (IL-4, IL-5, and IL-10) populations have reduced GVHD capacity. To determine whether the GVHD protection of the anti-CD40L-cultured cells was mediated by a skewing toward CD4+ Th2 cells, supernatants were taken from the primary (days 2, 4, 7, and 10) and secondary cultures (days 1, 3, and 5) and evaluated for the presence of Th1 (IL-2, IFN-γ) and Th2 cytokines. Both control-cultured and anti-CD40L-cultured cells produced IL-2 by day 2 of the primary MLR (Table II). IL-2 continued to accumulate in the control culture, peaking at 312 pg/ml on day 7. Cultures containing anti-CD40L mAb produced 12-fold less IL-2 (27 pg/ml vs 312 pg/ml) during the primary culture than the control. Peak amounts of IFN-γ were detected at the end of the primary culture in the control cells, with very low accumulation in the anti-CD40L cultures. The Th2 cytokines, IL-4 and IL-10, were not detected in the supernatants of either culture in the primary MLR.

Table II.

ELISA analysis of cytokine levels in supernatants from primary MLR culturea

Primary (day)GroupTh1Th2
IL-2IFN-γIL-4IL-10
Control 48 
Anti-CD40L 14 
Control 286 23 
Anti-CD40L 27 
Control 312 187 
Anti-CD40L 17 
10 Control 111 281 
10 Anti-CD40L 
Primary (day)GroupTh1Th2
IL-2IFN-γIL-4IL-10
Control 48 
Anti-CD40L 14 
Control 286 23 
Anti-CD40L 27 
Control 312 187 
Anti-CD40L 17 
10 Control 111 281 
10 Anti-CD40L 
a

Primary MLR cultures were established with B6 CD4+ T cells as responders and irradiated, T cell-depleted bm12 splenic stimulators. Supernatants were harvested on day 2, 4, 7, and 10 of primary culture. Final concentration of responders was 0.5 × 106 cells/ml. Values are in pg/ml, and sensitivity is 1–10 pg/ml.

Secondary cultures established from both groups of 4-day primary cultures produced large amounts of IL-2 and IFN-γ as early as day 1 of culture (Table III). On a per cell basis, the cells exposed to anti-CD40L mAb in the primary culture produced 10-fold more IL-2 (915 pg/0.15 × 106 cells vs 312 pg/0.5 × 106 cells) and 6-fold more IFN-γ (523 pg/0.15 × 106 cells vs 281 pg/0.5 × 106 cells) in the secondary MLR than the control group produced in the primary MLR, suggesting significant Ag priming. In contrast to the primary MLR, the Th2 cytokines, IL-4 and IL-10, were produced by the 4-day control-primed cells upon in vitro alloantigen restimulation, indicating that the control bulk culture consisted of a mixed population of Th1- and Th2-primed cells (Table III). Although in secondary MLRs, Th2 cytokines by the anti-CD40L-cultured cells were reduced in comparison with the control-primed cells, there was still substantial production. These data indicated that although a 4- day culture in the presence of anti-CD40L mAb reduced cytokine production upon reexposure to alloantigen in comparison with the control cultures, there was still a vigorous Th1 and Th2 cytokine response to alloantigen restimulation. Given the high levels of IL-2 production by the 4-day anti-CD40L-cultured cells, it is perhaps not surprising that they did mediate GVHD lethality in vivo (Fig. 5 A).

Table III.

ELISA analysis of cytokine levels in supernatants from secondary MLR culturea

Primary (day)GroupSecondary (day)Th1Th2
IL-2IFN-γIL-4IL-10
Control 1171 390 25 
Anti-CD40L 560 71 
Control 1238 2592 86 237 
Anti-CD40L 915 414 23 57 
Control 295 2465 38 641 
Anti-CD40L 371 523 17 108 
Control 1193 2729 103 98 
Anti-CD40L 286 10 
Control 279 3310 59 1586 
Anti-CD40L 53 48 35 
Control 154 3268 30 1061 
Anti-CD40L 16 25 47 
10 Control 1167 3253 87 53 
10 Anti-CD40L 33 22 
10 Control 698 3368 125 879 
10 Anti-CD40L 
10 Control 133 3190 46 1308 
10 Anti-CD40L 16 
Primary (day)GroupSecondary (day)Th1Th2
IL-2IFN-γIL-4IL-10
Control 1171 390 25 
Anti-CD40L 560 71 
Control 1238 2592 86 237 
Anti-CD40L 915 414 23 57 
Control 295 2465 38 641 
Anti-CD40L 371 523 17 108 
Control 1193 2729 103 98 
Anti-CD40L 286 10 
Control 279 3310 59 1586 
Anti-CD40L 53 48 35 
Control 154 3268 30 1061 
Anti-CD40L 16 25 47 
10 Control 1167 3253 87 53 
10 Anti-CD40L 33 22 
10 Control 698 3368 125 879 
10 Anti-CD40L 
10 Control 133 3190 46 1308 
10 Anti-CD40L 16 
a

Secondary MLR cultures were established in the absence of anti-CD40L mAb with 4-, 7-, or 10-day cultured B6 CD4+ T cells as responders and irradiated, non-T cell-depleted bm12 splenic stimulators. Supernatants were harvested day 1, 3, and 5 of secondary culture. Final concentration of responders was 0.15 × 106 cells/ml. Values are in pg/ml and sensitivity is 1–10 pg/ml.

IL-2 and IFN-γ production in secondary cultures established from 7-day anti-CD40L cultures was reduced 3-fold (286 pg/ml vs 915 pg/ml) and 11-fold (48 pg/ml vs 523 pg/ml), respectively, as compared with that established from 4-day anti-CD40L cultures. It is interesting to note that although the IL-2 production was reduced 75% as compared with the 7-day control group, there was more IL-2 produced on a per cell basis by the 7-day anti-CD40L cultures in secondary MLR than in the primary MLR control group. Surprisingly, in spite of this substantial IL-2 production in vitro upon Ag restimulation by the anti-CD40L-cultured cells, these cells did not mediate GVHD in vivo (Fig. 5,B). IFN-γ production by the 7-day anti-CD40L cultures was reduced by 99% compared with the control-primed cultures. As with the Th1 cytokines, there were lower levels of Th2 cytokines produced in secondary MLRs by 7-day anti-CD40L-cultured cells than by 4-day cultures. Th1 and Th2 cytokine production in secondary cultures established from 10-day anti-CD40L cultures were the most profoundly suppressed (Table III). Most notably, IL-2 production in secondary MLR established from 10-day anti-CD40L cultures was 9-fold (33 pg/ml vs 286 pg/ml) lower than that from 7-day anti-CD40L cultures. Overall, we found a pronounced and progressive dimunition of Th1 and Th2 cytokine production in the secondary MLRs of the anti-CD40L cultures with increasing primary culture duration.

Because ELISA measures the total amount of immunologically reactive protein present in the supernatants, but doesn’t take into account cytokine consumption, Th2 skewing may have been present, but not at a level detectable by ELISA. As additional evidence that GVHD protection was not due to a skewing toward a Th2 phenotype, we used CD4+ cells from IL-4 or IL-10 knockout mice as responders in our 10-day ex vivo tolerization MLR cultures. Ex vivo blockade of the CD40L:CD40 pathway via anti-CD40L mAb in IL-4 knockout CD4+ cells resulted in decreased proliferation in the primary MLR (Fig. 6,A) and profound hyporesponsiveness upon alloantigen restimulation in vitro (data not shown). Mice receiving 105 control-primed IL-4 knockout CD4+ cells died of BM aplasia by 3 wk. Mice receiving 105 anti-CD40L-cultured IL-4 knockout CD4+ cells survived the 65-day observation period (Fig. 6,B) and had normal HCT at time of elective sacrifice (data not shown). Similar results were found with IL-10 knockout CD4+ cells. Proliferation in primary and secondary MLRs was profoundly reduced in anti-CD40L-cultured IL-10 knockout CD4+ cells (data not shown). Mice receiving 105 anti-CD40-cultured IL-10 knockout CD4+ cells survived the observation period (Fig. 7). In contrast, mice receiving 105 control-primed IL-10 knockout CD4+ cells died of BM aplasia by 2 wk posttransfer. These data indicate that production of the Th2 cytokines, IL-4 or IL-10, was not required for anergy induction or GVHD protection.

FIGURE 6.

B6 IL-4 knockout CD4+ T cells are susceptible to tolerance induction via a 10-day ex vivo tolerization culture with anti-CD40L mAb. A, Primary MLR culture consisted of B6 IL-4 knockout CD4+ T cells and bm12 splenic stimulators. Anti-CD40L was added at 50 μg/ml at the initiation of culture. On the y-axis are mean cpm ± 1 SD of the mean. On the x-axis are days in primary culture. B, On day 10 of primary culture, the above MLR was washed and infused into sublethally irradiated bm12 recipients at 105 cells/mouse (n = 8/group). On the x-axis are days posttransfer of T cells. On the y-axis is the proportion of recipients surviving posttransfer.

FIGURE 6.

B6 IL-4 knockout CD4+ T cells are susceptible to tolerance induction via a 10-day ex vivo tolerization culture with anti-CD40L mAb. A, Primary MLR culture consisted of B6 IL-4 knockout CD4+ T cells and bm12 splenic stimulators. Anti-CD40L was added at 50 μg/ml at the initiation of culture. On the y-axis are mean cpm ± 1 SD of the mean. On the x-axis are days in primary culture. B, On day 10 of primary culture, the above MLR was washed and infused into sublethally irradiated bm12 recipients at 105 cells/mouse (n = 8/group). On the x-axis are days posttransfer of T cells. On the y-axis is the proportion of recipients surviving posttransfer.

Close modal
FIGURE 7.

B6 IL-10 knockout CD4+ T cells are susceptible to tolerance induction via a 10-day ex vivo tolerization culture with anti-CD40L mAb. Ten-day MLR cultures were washed and infused into sublethally irradiated bm12 recipients at 105 cells/mouse (n = 8/group). On the x-axis are days posttransfer of T cells. On the y-axis is the proportion of recipients surviving posttransfer.

FIGURE 7.

B6 IL-10 knockout CD4+ T cells are susceptible to tolerance induction via a 10-day ex vivo tolerization culture with anti-CD40L mAb. Ten-day MLR cultures were washed and infused into sublethally irradiated bm12 recipients at 105 cells/mouse (n = 8/group). On the x-axis are days posttransfer of T cells. On the y-axis is the proportion of recipients surviving posttransfer.

Close modal

IL-4 has been reported to preclude anergy induction by signaling through the common γ-chain of the IL-2R, IL-4R, IL-7R, and IL-15R (21, 33, 34). IL-4 has also been found to prevent cell death without cell division and proliferation by preventing the decay of bcl-2 in activated T cells (35). Because cell recovery in the 10-day anti-CD40L cultures is relatively low (≤10% of starting cell number), we examined the effect of a day 0 addition of a low concentration IL-4 as a survival factor to determine whether we could improve recovery without precluding the induction of anergy. The addition of 0.1 ng/ml of IL-4 did not reproducibly improve recovery. However, the addition of 1 ng/ml or 10 ng/ml of IL-4 added to the cultures on day 0 did reproducibly and favorably increase recovery from <10% to 32–39% of the starting cell number (data not shown). The addition of 1 ng/ml of IL-4 added to cultures on day 0 did not preclude the inhibition of proliferation by anti-CD40L mAb in the primary MLR (Fig. 8,A). Peak proliferation in the primary culture was inhibited by anti-CD40L mAb by 96% even in the presence of 1 ng/ml IL-4. Cell recovery on day 10 was 8% in the anti-CD40L cultures in the absence of IL-4 and 48% in the presence of IL-4. Despite a high degree of blockade of proliferation in the primary MLR cultures containing both anti-CD40L mAb and IL-4, these cells were only modestly hyporesponsive upon in vitro alloantigen restimulation in the secondary MLR (Fig. 8,B). This modest reduction (<40%) in secondary response as compared with the control-primed cells did not translate to GVHD protection in vivo. All recipients of either 105 control-primed/IL-4 or anti-CD40L/IL-4-cultured cells died of GVHD-induced BM aplasia by day 18 after cell transfer (Fig. 8 C). In contrast, all recipients of 105 anti-CD40L-cultured cells in the absence of supplemental IL-4 survived the 2-mo observation period. These data indicate that IL-4 (1 ng/ml) added as a survival factor on day 0 precluded the induction of anergy.

FIGURE 8.

Although the addition of IL-4 at 1 ng/ml at the initiation of culture did not interfere with the inhibition of primary MLR by anti-CD40L mAb, the cells were only modestly hyporesponsive upon alloantigen restimulation in the secondary MLR and were not tolerant in vivo. A, Primary MLR culture consisted of B6 CD4+ lymph node T cell responders and bm12 splenic stimulators in the presence of IL-4 (1 ng/ml). Anti-CD40L mAb was added at 50 μg/ml at the initiation of culture. B, Secondary MLRs were established from the above 10-day primary MLR cultures in the absence of IL-4 and anti-CD40L mAb. On the y-axis are mean cpm ± 1 SD of the mean. On the x-axis are days in secondary culture. C, Ten-day MLR cultures established in the presence or absence of IL-4 were washed and infused into sublethally irradiated bm12 recipients at 105 cells/mouse (n = 5/group). On the x-axis are days posttransfer of T cells. On the y-axis is the proportion of recipients surviving posttransfer.

FIGURE 8.

Although the addition of IL-4 at 1 ng/ml at the initiation of culture did not interfere with the inhibition of primary MLR by anti-CD40L mAb, the cells were only modestly hyporesponsive upon alloantigen restimulation in the secondary MLR and were not tolerant in vivo. A, Primary MLR culture consisted of B6 CD4+ lymph node T cell responders and bm12 splenic stimulators in the presence of IL-4 (1 ng/ml). Anti-CD40L mAb was added at 50 μg/ml at the initiation of culture. B, Secondary MLRs were established from the above 10-day primary MLR cultures in the absence of IL-4 and anti-CD40L mAb. On the y-axis are mean cpm ± 1 SD of the mean. On the x-axis are days in secondary culture. C, Ten-day MLR cultures established in the presence or absence of IL-4 were washed and infused into sublethally irradiated bm12 recipients at 105 cells/mouse (n = 5/group). On the x-axis are days posttransfer of T cells. On the y-axis is the proportion of recipients surviving posttransfer.

Close modal

In this study, we have demonstrated that anergy induction via in vitro blockade of the CD40:CD40L costimulatory pathway became more profound with increasing duration in primary culture as measured by in vitro secondary MLR proliferative responses, cytokine production, and in vivo GVHD protection. Th1 and Th2 cytokine production were both reduced in the primary and secondary MLRs of the anti-CD40L-cultured cells, indicating that GVHD was not mediated by a skewing toward a Th2 phenotype. Additionally, IL-4 and IL-10 knockout CD4+ cells were susceptible to the induction of alloantigen hyporesponsiveness in our model, further indicating that Th2 cytokines were not obligatory. IL-4 added to the cultures at doses that improved cell recovery precluded the induction of anergy, indicating that IL-4 does not mediate but rather can prevent the induction of tolerance.

We have previously demonstrated a profound reduction in GVHD lethality with a 10-day ex vivo tolerization culture duration (17). Because studies by others have indicated that the time to optimal tolerance induction can vary between 16 h and 10 days, we wished to determine whether this relatively prolonged duration was essential for GVHD protection (5, 21, 22, 23, 24, 25, 26). Our data indicated a 4-day MLR culture in the presence of anti-CD40L mAb was insufficient to induce tolerance. Although CD40L expression was up-regulated by day 4 of control cultures, cells had not substantially increased in size and internal complexity, CD25 expression was not increased, and L-selectin had not yet significantly down-regulated. Together these data may suggest that T cells require a certain level of activation for tolerance induction to occur.

In contrast to the partial inhibitory effect of 4-day anti-CD40L MLR cultures, 7 days of primary culture with mAb were sufficient to induce hyporesponsiveness in vitro and tolerance in vivo, while 10 days of culture resulted in a more profoundly tolerized population of cells. Although the cell doses used for in vivo GVHD generation did not allow us to conclude that 10 days of primary culture led to greater in vivo GVHD prevention than a 7-day primary culture, the secondary MLR and cytokine data indicated that the secondary in vitro hyporesponsiveness was more profound with a 10-day primary tolerization culture duration. In all experiments, there was a small burst of proliferation on day 1 of secondary MLR by these cultures. This slight degree of proliferation in the hyporesponsive cells in the secondary MLR was accompanied by substantial IL-2 production especially from secondary cultures established from 7-day tolerized MLR cultures. Nine-fold more IL-2 was produced in response to alloantigen restimulation by 7-day tolerized cultures as compared with 10-day tolerized cultures. Based upon the high levels of IL-2 detected in the supernatants of secondary cultures of tolerized cells established from 7-day MLR cultures, one might have predicted greater GVHD mortality. However, this early proliferation and IL-2 production by the tolerized population were not sustained and rapidly declined. Although we do not yet know whether the Ag-specific cells in the tolerized cultures were deleted, we hypothesize that deletion is probably not the sole mechanism involved, as the bulk population had a partial early proliferation and produced IL-2 in response to alloantigen restimulation.

The degree of hyporesponsiveness seen in the secondary MLR upon reexposure to alloantigen correlated well with GVHD protection in vivo, suggesting that the in vitro assays could potentially be used as a monitoring device, which might be predictive of the clinical outcome. The primary MLR may be less predictive of successful tolerance induction than the secondary MLR (see Fig. 8 A). Although the degree of blockade of proliferation was very high by anti-CD40L mAb plus IL-4 in the primary MLR, there was only modest secondary hyporesponsiveness and no GVHD protection.

The requirement for a relatively long primary culture duration to induce alloantigen hyporesponsiveness is in contrast to studies by other investigators. Taub et al. found that a 3-day incubation of the phosphatidylinositol-3-kinase inhibitor, wortmanin, was sufficient to induce alloantigen-specific tolerance in a murine GVHD model (22). However, as the degree of GVHD protection was not quantified, it is unknown whether a longer incubation would have resulted in a more profound resistance to GVHD induction. Although it is possible that tolerance induction via small intracellular signaling inhibitors may occur faster than that via Ab blockade of cell surface costimulatory molecules, not all surface-mediated tolerance-inducing events require prolonged ex vivo culture. For example, Jenkins and Schwartz found that only a 16-h incubation of pigeon cytochrome c-specific T cell clones with Ag and chemically modified splenocytes was sufficient to induce unresponsiveness to restimulation with normal APC and Ag (5). Although these studies used T cell clones and a peptide Ag and not bulk CD4+ lymph node and alloantigen as ours do, it is tempting to speculate that the relatively short culture duration needed to tolerize these cells may be due to the fact that APCs in their culture system were completely incapable of up-regulating the cell surface expression of all potential costimulatory molecules. In our system, costimulatory molecules other than those entirely dependent upon induction via CD40L:CD40 interaction potentially could be induced to be expressed and therefore may have transduced some costimulatory signal. Alternatively, their model may involve a higher frequency of responding T cells and a more rapid response requiring a shorter culture duration to induce anergy. Optimal anergy induction via blockade of costimulation may require a culture duration of sufficient length to ensure that all potentially alloreactive cells have had maximal opportunity for TCR engagement, initial activation and up-regulation, and subsequent blockade of CD40L. Although CD40L was up-regulated by day 2, peak proliferative responses occurred on day 5 or 6 of primary culture. Additionally, maximal up-regulation of the activation Ag, CD25, was not seen until day 7 of primary culture, with very low expression by day 4. Blockade of costimulation as a strategy to induce anergy induction may be required throughout the duration of these events. The minimum culture duration required to obtain optimal anergy may depend as much on the kinetics of the response as on the strategy used to induce anergy.

In a model more analogous to ours, Gribben et al. found that maximal hyporesponsiveness of human alloreactive T cells to alloantigen restimulation was achieved with 36 h via blockade of the CD28:B7-1/B7-2 pathway (21). Other studies have indicated that a longer culture duration is necessary or at least beneficial. Two studies found that a minimum of 5- to 7-day culture of human T cells with stimulator cells in the presence of anti-B7-1 and cyclosporine, followed by 1- to 2-day rest was needed to yield a hyporesponsive state (23, 26). In a system in which human alloreactive T cells were rendered hyporesponsive by IL-10, T cells were noted to become progressively more hyporesponsive during a 3- to 10-day culture period (25). These data and ours suggest that hyporesponsiveness becomes increasingly profound with increasing culture duration in bulk cultures consisting of naive, potentially alloreactive T cells.

We have previously demonstrated that OVA-specific T cells exposed to anti-CD40L mAb during tolerization to alloantigen have intact OVA responses at optimal Ag concentrations (17). Consistent with these data, although the frequency of responding T cells to relevant alloantigen was decreased by 60-fold in anti-CD40L-cultured cells as compared with the control-cultured cells, the frequency of the tolerized cells to third party alloantigen was decreased only 2-fold. This suggests that a bulk T cell population that is tolerized ex vivo to alloantigen may still be capable of responding to viral or tumor Ags encountered in vivo.

In contrast to other groups reporting split anergy in which proliferation and cytokine production were diminished, but cytotoxicity was not impaired (27, 28), we found CTL killing by the anti-CD40L-cultured cells was approximately one-fourth that of the control-primed cells. Perhaps more importantly, anti-CD40L-cultured cells failed to induce mortality in mice, while mice receiving control-primed cells died of BM aplasia. Consistent with this, IFN-γ required for CTL generation was profoundly reduced in both the primary and secondary MLR of the anti-CD40L-cultured cells. We and others have found that an intact CD40L:CD40 pathway is required for optimal in vivo CTL generation, and that the administration of anti-CD40L mAb abrogates a GVL effect (36, 37, 38, 39) (P. A. Taylor and B. R. Blazar, unpublished observation). Collectively, our data indicate that the anti-CD40L-cultured cells are hypofunctional in terms of proliferation, cytokine production, and cytotoxicity.

Although Th2 cells have been reported in some (32, 40) but not all (41) studies to result in reduced GVHD as compared with Th1 cells, our hyporesponsive cells were not skewed toward a Th2 phenotype. The anti-CD40L-cultured cells made far less of the Th2 cytokines, IL-4 and IL-10, upon alloantigen restimulation than did the control-primed cells. As well as being a Th2 cytokine, IL-10 is an immunosuppressive cytokine that has been shown to induce tolerance in human alloreactive T cells (25). In another study, endogenously produced IL-10 and TGF-β were found to mediate superantigen-induced tolerance in a murine in vivo model (42). However, both IL-4 and IL-10 knockout CD4+ cells were susceptible to anergy induction in our model, indicating there is not an absolute requirement for either of these cytokines for hyporesponsiveness in vitro and GVHD protection in vivo. Additionally, we did not find free TGF-β, another immunosuppressive cytokine, in the supernatants of either culture group in either the primary or the secondary MLR (data not shown). We did find the chemokines, RANTES and macrophage-inflammatory protein 1-α, present in the supernatants of the anti-CD40L cultures in both the primary and the secondary MLR, indicating that the tolerized cells are not globally, metabolically disabled (data not shown).

Because cell recovery is typically low (≤10%) in the anergic cultures, we examined the effect of the addition of IL-4 as a survival factor to the primary cultures in an attempt to increase our cell recovery. We did not uncover a concentration of IL-4 that both increased recovery and permitted the induction of secondary hyporesponsiveness. As IL-7 also has been reported to be involved with T cell survival in vivo and in vitro (35), we examined the addition of IL-7 on cell recovery and induction of secondary hyporesponsiveness and discovered that a dose or schedule that improved cell recovery also precluded anergy induction (P. A. Taylor and B. R. Blazar, unpublished data). We surmise that the addition of IL-4 or IL-7 at concentrations high enough to increase recovery results in sufficient signaling through the IL-2R common γ-chain to preclude the induction of anergy.

These studies indicate that ex vivo anti-CD40L tolerization becomes more profound with increasing primary culture duration, highlighting the importance of kinetics studies in tolerance-induction protocols. Our data indicate that a combination of flow cytometry and in vitro secondary proliferative, cytotoxic, and cytokine responses may be useful indicators of the efficacy of a tolerizing procedure in preventing GVHD generation in vivo. Anti-CD40L mAb ex vivo tolerization warrants consideration as a potential therapeutic modality for the prevention of GVHD.

We thank Dr. Arlene Sharpe for critical review of the manuscript.

1

This work was supported in part by National Institutes of Health Grants RO1 AI-34495, R37 HL-56067, and PO1 AI-35296.

3

Abbreviations used in this paper: BMT, bone marrow transplantation; FSC, forward scatter; GVHD, graft-vs-host disease; HCT, hematocrit; LDA, limiting dilution assay; SSC, side scatter.

1
Blazar, B. R., R. Korngold, D. A. Vallera.
1997
. Recent advances in graft-versus-host disease (GVHD) prevention.
Immunol. Rev.
157
:
79
2
Schwartz, R. H..
1990
. A cell culture model for T lymphocyte clonal anergy.
Science
248
:
1349
3
Schwartz, R. H..
1996
. Models of T cell anergy: is there a common molecular mechanism?.
J. Exp. Med.
184
:
1
4
Schwartz, R. H..
1992
. Costimulation of T lymphocytes: the role of CD28, CTLA-4, and B7/BB1 in interleukin-2 production and immunotherapy.
Cell
71
:
1065
5
Jenkins, M. K., R. H. Schwartz.
1987
. Antigen presentation by chemically modified splenocytes induces antigen-specific T cell unresponsiveness in vitro and in vivo.
J. Exp. Med.
165
:
302
6
Jenkins, M. K..
1992
. The role of cell division in the induction of clonal anergy.
Immunol. Today
13
:
69
7
Grewal, I. S., R. A. Flavell.
1996
. A central role of CD40 ligand in the regulation of CD4+ T-cell responses.
Immunol. Today
17
:
410
8
Buhlmann, J. E., T. M. Foy, A. Aruffo, K. M. Crassi, J. A. Ledbetter, W. R. Green, J. C. Xu, L. D. Shultz, D. Roopesian, R. A. Flavell, et al
1995
. In the absence of a CD40 signal, B cells are tolerogenic.
Immunity
2
:
645
9
Van Gool, S. W., P. Vandenberghe, M. de Boer, J. L. Ceuppens.
1996
. CD80, CD86 and CD40 provide accessory signals in a multiple-step T-cell activation model.
Immunol. Rev.
153
:
47
10
Durie, F. H., R. A. Fava, R. J. Noelle.
1994
. Collagen-induced arthritis as a model of rheumatoid arthritis.
Clin. Immunol. Immunopathol.
73
:
11
11
Blazar, B. R., P. A. Taylor, A. Panoskaltsis-Mortari, J. Buhlman, J. Xu, R. A. Flavell, R. Korngold, R. Noelle, D. A. Vallera.
1997
. Blockade of CD40 ligand-CD40 interaction impairs CD4+ T cell-mediated alloreactivity by inhibiting mature donor T cell expansion and function after bone marrow transplantation.
J. Immunol.
158
:
29
12
Hollander, G. A., E. Castigli, R. Kulbacki, M. Su, S. J. Burakoff, J. C. Gutierrez-Ramos, R. S. Geha.
1996
. Induction of alloantigen-specific tolerance by B cells from CD40-deficient mice.
Proc. Natl. Acad. Sci. USA
93
:
4994
13
Niimi, M., T. C. Pearson, C. P. Larsen, D. Z. Alexander, D. Hollenbaugh, A. Aruffo, P. S. Linsley, E. Thomas, K. Campbell, W. C. Fanslow, et al
1998
. The role of the CD40 pathway in alloantigen-induced hyporesponsiveness in vivo.
J. Immunol.
161
:
5331
14
Kirk, A. D., D. M. Harlan, N. N. Armstrong, T. A. Davis, Y. Dong, G. S. Gray, X. Hong, D. Thomas, J. H. Fechner, Jr, S. J. Knechtle.
1997
. CTLA4-Ig and anti-CD40 ligand prevent renal allograft rejection in primates.
Proc. Natl. Acad. Sci. USA
94
:
8789
15
Kirk, A. D., L. C. Burkly, D. S. Batty, R. E. Baumgartner, J. D. Berning, K. Buchanan, J. H. Fechner, Jr, R. L. Germond, R. L. Kampen, N. B. Patterson, et al
1999
. Treatment with humanized monoclonal antibody against CD154 prevents acute renal allograft rejection in nonhuman primates.
Nat. Med.
5
:
686
16
Kenyon, N. S., M. Chatzipetrou, M. Masetti, A. Ranuncoli, M. Oliveira, J. L. Wagner, A. D. Kirk, D. M. Harlan, L. C. Burkly, C. Ricordi.
1999
. Long-term survival and function of intrahepatic islet allografts in rhesus monkeys treated with humanized anti-CD154.
Proc. Natl. Acad. Sci. USA
96
:
8132
17
Blazar, B. R., P. A. Taylor, R. J. Noelle, D. A. Vallera.
1998
. CD4+ T cells tolerized ex vivo to host alloantigen by anti-CD40 ligand (CD40L:CD154) antibody lose their graft-versus-host disease lethality capacity but retain nominal antigen responses.
J. Clin. Invest.
102
:
473
18
Xun, C. Q., J. S. Thompson, C. D. Jennings, S. A. Brown, M. B. Widmer.
1994
. 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
83
:
2360
19
Krenger, W., G. R. Hill, J. L. Ferrara.
1997
. Cytokine cascades in acute graft-versus-host disease.
Transplantation
64
:
553
20
Matzinger, P..
1991
. The JAM test: a simple assay for DNA fragmentation and cell death.
J. Immunol. Methods
145
:
185
21
Gribben, J. G., E. C. Guinan, V. A. Boussiotis, X. Y. Ke, L. Linsley, C. Sieff, G. S. Gray, G. J. Freeman, L. M. Nadler.
1996
. Complete blockade of B7 family-mediated costimulation is necessary to induce human alloantigen-specific anergy: a method to ameliorate graft-versus-host disease and extend the donor pool.
Blood
87
:
4887
22
Taub, D. D., W. J. Murphy, O. Asai, R. G. Fenton, G. Peltz, M. L. Key, S. Turcovski-Corrales, D. L. Longo.
1997
. Induction of alloantigen-specific T cell tolerance through the treatment of human T lymphocytes with wortmannin.
J. Immunol.
158
:
2745
23
Van Gool, S. W., M. de Boer, J. L. Ceuppens.
1994
. The combination of anti-B7 monoclonal antibody and cyclosporin A induces alloantigen-specific anergy during a primary mixed lymphocyte reaction.
J. Exp. Med.
179
:
715
24
Tan, P., C. Anasetti, J. A. Hansen, J. Melrose, M. Brunvand, J. Bradshaw, J. A. Ledbetter, P. S. Linsley.
1993
. Induction of alloantigen-specific hyporesponsiveness in human T lymphocytes by blocking interaction of CD28 with its natural ligand B7/BB1.
J. Exp. Med.
177
:
165
25
Groux, H., M. Bigler, J. E. de Vries, M. G. Roncarolo.
1996
. Interleukin-10 induces a long-term antigen-specific anergic state in human CD4+ T cells.
J. Exp. Med.
184
:
19
26
Comoli, P., D. Montagna, A. Moretta, M. Zecca, F. Locatelli, R. Maccario.
1995
. Alloantigen-induced human lymphocytes rendered nonresponsive by a combination of anti-CD80 monoclonal antibodies and cyclosporin-A suppress mixed lymphocyte reaction in vitro.
J. Immunol.
155
:
5506
27
Go, C., D. W. Lancki, F. W. Fitch, J. Miller.
1993
. Anergized T cell clones retain their cytolytic ability.
J. Immunol.
150
:
367
28
Otten, G. R., R. N. Germain.
1991
. Split anergy in a CD8+ T cell: receptor-dependent cytolysis in the absence of interleukin-2 production.
Science
251
:
1228
29
Berke, G..
1995
. The CTL’s kiss of death.
Cell
81
:
9
30
Kojima, H., N. Shinohara, S. Hanaoka, Y. Someya-Shirota, Y. Takagaki, H. Ohno, T. Saito, T. Katayama, H. Yagita, K. Okumura, et al
1994
. Two distinct pathways of specific killing revealed by perforin mutant cytotoxic T lymphocytes.
Immunity
1
:
357
31
Jung, T. M., W. M. Gallatin, I. L. Weissman, M. O. Dailey.
1988
. Down-regulation of homing receptors after T cell activation.
J. Immunol.
141
:
4110
32
Fowler, D. H., J. Breglio, G. Nagel, C. Hirose, R. E. Gress.
1996
. Allospecific CD4+, Th1/Th2 and CD8+, Tc1/Tc2 populations in murine GVL: type I cells generate GVL and type II cells abrogate GVL.
Biol. Blood Marrow Transplant
2
:
118
33
He, Y. W., B. Adkins, R. K. Furse, T. R. Malek.
1995
. Expression and function of the γ c subunit of the IL-2, IL-4, and IL-7 receptors: distinct interaction of γ c in the IL-4 receptor.
J. Immunol.
154
:
1596
34
Boussiotis, V. A., D. L. Barber, T. Nakarai, G. J. Freeman, J. G. Gribben, G. M. Bernstein, A. D. D’Andrea, J. Ritz, L. M. Nadler.
1994
. Prevention of T cell anergy by signaling through the γ c chain of the IL-2 receptor.
Science
266
:
1039
35
Vella, A. T., S. Dow, T. A. Potter, J. Kappler, P. Marrack.
1998
. Cytokine-induced survival of activated T cells in vitro and in vivo.
Proc. Natl. Acad. Sci. USA
95
:
3810
36
Diehl, L., A. T. den Boer, S. P. Schoenberger, E. I. van der Voort, T. N. Schumacher, C. J. Melief, R. Offringa, R. E. Toes.
1999
. CD40 activation in vivo overcomes peptide-induced peripheral cytotoxic T-lymphocyte tolerance and augments anti-tumor vaccine efficacy.
Nat. Med.
5
:
774
37
Sotomayor, E. M., I. Borrello, E. Tubb, F. M. Rattis, H. Bien, Z. Lu, S. Fein, S. Schoenberger, H. I. Levitsky.
1999
. Conversion of tumor-specific CD4+ T-cell tolerance to T-cell priming through in vivo ligation of CD40.
Nat. Med.
5
:
780
38
Mackey, M. F., J. R. Gunn, C. Maliszewsky, H. Kikutani, R. J. Noelle, R. J. Barth, Jr.
1998
. Dendritic cells require maturation via CD40 to generate protective antitumor immunity.
J. Immunol.
161
:
2094
39
Mackey, M. F., R. J. Barth, Jr, R. J. Noelle.
1998
. The role of CD40/CD154 interactions in the priming, differentiation, and effector function of helper and cytotoxic T cells.
J. Leukocyte Biol.
63
:
418
40
Krenger, W., K. M. Snyder, J. C. Byon, G. Falzarano, J. L. Ferrara.
1995
. Polarized type 2 alloreactive CD4+ and CD8+ donor T cells fail to induce experimental acute graft-versus-host disease.
J. Immunol.
155
:
585
41
Murphy, W. J., L. A. Welniak, D. D. Taub, R. H. Wiltrout, P. A. Taylor, D. A. Vallera, M. Kopf, H. Young, D. L. Longo, B. R. Blazar.
1998
. Differential effects of the absence of interferon-γ and IL-4 in acute graft-versus-host disease after allogeneic bone marrow transplantation in mice.
J. Clin. Invest.
102
:
1742
42
Miller, C., J. A. Ragheb, R. H. Schwartz.
1999
. Anergy and cytokine-mediated suppression as distinct superantigen-induced tolerance mechanisms in vivo.
J. Exp. Med.
190
:
53