Abstract
Blockade with B7 antagonists interferes with CD28:B7 and CTLA-4:B7 interactions, which may have opposing effects. We have examined the roles of CD28:B7 and CTLA-4:B7 on in vivo alloresponses. A critical role of B7:CD28 was demonstrated by markedly compromised expansion of CD28-deficient T cells and diminished graft-versus-host disease lethality of limited numbers of purified CD4+ or CD8+ T cells. When high numbers of T cells were infused, the requirement for CD28:B7 interaction was lessened. In lethally irradiated recipients, anti-CTLA-4 mAb enhanced in vivo donor T cell expansion, but did not affect, on a per cell basis, anti-host proliferative or CTL responses of donor T cells. Graft-versus-host lethality was accelerated by anti-CTLA-4 mAb infusion given early post-bone marrow transplantation (BMT), mostly in a CD28-dependent fashion. Donor T cells obtained from anti-CTLA-4 mAb-treated recipients were skewed toward a Th2 phenotype. Enhanced T cell expansion in mAb-treated recipients was strikingly advantageous in the graft-versus-leukemia effects of delayed donor lymphocyte infusion. In two different systems, anti-CTLA-4 mAb enhanced the rejection of allogeneic T cell-depleted marrow infused into sublethally irradiated recipients. We conclude that blockade of the selective CD28-B7 interactions early post-BMT, which preserve CTLA-4:B7 interactions, would be preferable to blocking both pathways. For later post-BMT, the selective blockade of CTLA-4:B7 interactions provides a potent and previously unidentified means for augmenting the GVL effect of delayed donor lymphocyte infusion.
Optimal T cell expansion requires signaling via the Ag-specific TCR along with a second, costimulatory signal (1, 2), which is necessary to stabilize cytokine mRNA and induce antiapoptotic proteins. The most well-studied costimulatory pathway involves the binding of two distinct B7 ligands (B7-1 (CD80); B7-2 (CD86)) present on APC to two homologous counter-receptors (CD28, CTLA-4)3 expressed on T cells (3). CD28 and CTLA-4 compete for the same B7 ligands, although CTLA-4 has higher avidity for B7.1 as compared with B7.2 (4). The majority of the evidence to date indicates that CD28:B7 interaction provides a positive signal to supporting T cell responses. The role of CTLA-4:B7 has been less well defined. Early studies using anti-CTLA-4 mAb suggested that CTLA-4:B7 interactions were stimulatory (4). The striking lymphoproliferation observed in mice that have been engineered to lack CTLA-4 expression provides evidence that CTLA-4 can function as a negative regulator of T cell activation (5, 6). However, the regulatory role of CTLA-4, as defined by results of studies using mAb rather than knockout mice, has been controversial with contradictory reports, indicating that CTLA-4 can down-regulate or can act as a costimulatory molecule in CD28-deficient mice (7, 8). Studies using anti-CTLA-4 mAb have shown that CTLA-4 regulates cell cycle entry and IL-2 secretion, and is needed for the induction of tolerance (9, 10, 11, 12, 13, 14). Reports using CD28-deficient mice show that under some circumstances, CTLA-4:B7 interactions can provide a positive signal (15). Conversely, Lin et al. recently have shown that blockade of CTLA-4:B7 interaction in CD28-deficient mice accelerated the course of cardiac allograft rejection, consistent with the inhibition of a negative regulatory role of CTLA-4:B7 in CD28-deficient mice (16). To clarify the role of CD28:B7 and CTLA-4:B7 interactions in regulating in vivo alloresponses, we separately analyzed the contribution of these two pathways in systems relevant to humans undergoing bone marrow transplantation (BMT).
Graft-versus-host disease (GVHD) is caused by donor T cells that are alloactivated in vivo upon contact with host alloantigen-bearing target cells. Previously, we have shown that the administration of either CTLA4-Ig fusion protein or anti-B7.1 + anti-B7.2 (anti-B7) mAb can down-regulate GVHD-induced mortality and inhibit donor T cell expansion in vivo (17, 18, 19, 20). We also have observed that the in vivo administration of anti-B7 mAb completely eliminates the graft-versus-leukemia (GVL) effect of delayed donor lymphocyte infusion (DLI) given to allogeneic BMT recipients that were subsequently challenged with acute myeloid leukemia tumor cells 1 wk after splenocyte infusion (21). Because CD28:B7 and CTLA-4:B7 interactions may counterbalance each other, we sought to determine the separate and potentially interrelated roles of each pathway in regulating donor T cell expansion, activation, and function in vitro and in vivo. In this study, we demonstrate that CD28:B7 and CTLA-4:B7 interactions have contrasting effects on the expansion of donor anti-host (GVHD-causing) and host anti-donor (graft-rejecting) T cell responses. These data suggest that the optimal exploitation of competitive antagonists to these pathways will depend upon the circumstances and timing of administration post-BMT.
Materials and Methods
Mice
B10.BR/SgSnJ (H2k), C57BL/6 (termed B6:H2b,CD45.2), CD28-deficient B6 (CD28−/−), C.H2bm1 (termed bm1; CD45.2), and C.H2bm12 (termed bm12; CD45.2) mice were purchased from The Jackson Laboratory (Bar Harbor, ME). BALB/c-SCID (H2d) and B6-CD45.1 congenic mice were purchased from the National Institutes of Health (Bethesda, MD). Mice were housed in a specific pathogen-free facility in microisolator cages. Donors and recipients were used at 8–10 wk of age.
GVHD generation
To determine the effect of CD28/CTLA-4:B7 interactions on the GVHD capacity of CD4+ or CD8+ T cells, we used a system in which purified T cell subsets are given to MHC disparate sublethally irradiated recipients, as initially described by Sprent (22). This system permits highly accurate quantification of the degree of GVHD responses as related to T cell dose. For this purpose, MHC class II (bm12) or class I (bm1) disparate recipients were irradiated with 6 Gray (Gy) TBI on day 0 from a 137Cesium source at a dose rate of 85 cGy/min (20, 22). Four to six hours after TBI, sublethally irradiated bm12 recipients were given purified lymph node (LN) B6CD4+ T cells at doses of 0.3–3 × 105 per recipient, while bm1 recipients were injected with CD8+ T cells at doses of 0.3, 1, or 3 × 106 per recipient. To purify LN cells, single cell suspensions of axillary, mesenteric, and inguinal LN cells were obtained (as a source of GVHD-causing effector cells), depleted of NK cells, and enriched for CD4+ or CD8+ T cells by depletions with anti-CD8 (hybridoma 2.43, rat IgG2b, provided by Dr. David Sachs, Charlestown, MA) or anti-CD4 (hybridoma GK1.5, rat IgG2b, provided by Dr. Frank Fitch, Chicago, IL), respectively. Rat mAb-coated T cells were passaged through a goat anti-mouse and goat anti-rat Ig-coated column (Biotex, Edmonton, Canada). The final composition of T cells in the donor graft was determined by flow cytometry and was always found to be ≥94% T cells of the desired phenotype. Hematocrit values were obtained at periodic intervals as an indicator of the possible BM destructive effects of infused T cells (20, 22). For all GVHD and engraftment systems, mice were monitored daily for survival and clinical appearance and weighed twice weekly.
To avoid a host anti-donor response present in the sublethal irradiation systems, we employed two different types of strategies. In the first, mice were lethally irradiated with 8 Gy TBI by x-ray (39 cGy/min) on day −1, followed on day 0 by the i.v. infusion of T cell-depleted (TCD) BM by anti-Thy-1.2 (clone 30-H-12, provided by Dr. David Sachs, Cambridge, MA) + complement treatment. To measure CD4+ and CD8+ T cell GVHD responses, irradiated B10.BR recipients were infused i.v. with 20 × 106 TCD BM along with 0 or 15 × 106 splenocytes from B6 or CD28−/− donors. To measure CD4+ T cell GVHD responses, irradiated bm12 recipients were infused with 8 × 106 TCD BM along with 0 or 0.3 × 106 purified T cells from B6 or CD28−/− donors. In a second system designed to analyze the effect of CD28−/− T cells in the complete absence of tissue damage induced by high dose TBI, BALB/c-SCID recipients were treated with anti-asialo GM1 antisera (25 μl on days −4 and −2) to deplete endogenous NK activity and then infused with 2 × 106 LN T cells from B6 or CD28−/− donors without further conditioning of the recipient.
Thoracic duct cannulation
For thoracic duct lymphocyte (TDL) isolation, cannulae were inserted in the thoracic duct of recipients at the time of peak proliferation (day 6) post-BMT. TDL were collected over a period of 18 h (20).
GVL studies
Our GVL induction procedure has been described in detail (21). In brief, mice undergoing BMT for GVL assessment were conditioned with 800 cGy irradiation from an x-ray source on day −1 and infused with 8 × 106 B10.BR TCD BM on day 0. Donor B10.BR splenocytes were administered i.v. at a dose of 25 × 106 on day 21 post-BMT. On day 28 post-BMT, mice were challenged with an MHC class I+II− acute myeloid leukemia line, C1498 (21). C1498 (American Type Culture Collection, Manassas, VA), originally derived as a spontaneous tumor line from a B6 mouse, was grown in RPMI (Life Technologies, Grand Island, NY) with 10% heat-inactivated FBS (HyClone, Ogden, UT), 2 mmol/L l-glutamine, 50 mmol/L 2-ME, and penicillin/streptomycin. C1498 cells (2 × 105) were given via the i.v. route. All available animals were examined for evidence of GVHD and tumor by necropsy (21).
Engraftment studies
B6-CD45.1 mice were irradiated with 6 Gy TBI by x-ray on day −1 and then given bm1 or bm12 TCD BM (10 × 106) cells (23). Peripheral blood cells were phenotyped for chimerism twice post-BMT. In addition, the presence of donor or host origin cells in individual lineages was analyzed in detail at least once post-BMT.
Anti-CTLA-4 mAb preparation
Clone UC10-4F10-11 (hamster anti-murine CTLA-4) was generously provided by Dr. Jeffrey Bluestone (University of Chicago, Chicago, IL) (9). mAb for injection was generated as ascites fluid and purified by ammonium sulfate precipitation, followed by dialysis. Control hamster IgG was purchased from Rockland Laboratories (Gilbertsville, PA). Cohorts of mice were given anti-CTLA-4 mAb or irrelevant hamster IgG mAb. In initial GVHD studies, anti-CTLA-4 mAb or control mAb was given at a dose of 250 μg/dose on days −1 and 0, and then 100 μg/dose three times per week until day 10. In a subsequent study, a more intensive regimen was used consisting of anti-CTLA-4 mAb or control mAb at 400 μg/dose from days −1 to +5, then 200 μg/dose three times a week until day 14 (for engraftment studies) or day 21 (for GVHD studies) or day 24 after delayed lymphocyte infusion (for GVL studies).
Flow cytometry
The T cell, B cell, and granulocyte/macrophage constituency of splenocytes, peripheral blood cells, or TDL effluent were measured using mAb directed toward CD4 or CD8, CD19, and Mac1, respectively. All fluorochrome-labeled mAb, unless indicated, were obtained from PharMingen (San Diego, CA). For bm1 recpients of B6-CD45.1 CD8+ T cells, splenocyte suspensions were monitored with αCD45.2 (clone 104-2, rat IgG2a) and αCD45.1 (clone A20-1.7, rat IgG2a), both provided by Dr. U. Hammerling (New York, NY). For splenic flow cytometry studies, cells were first incubated with 2.4G2 to block Fc receptors, and then incubated with an optimal concentration of fluorochrome-labeled mAb for 45 min at 4°C. Cells were washed three times and resuspended for analysis by two- or three-color flow cytometry using FITC-, PE-, or biotin (along with streptavidin-PerCP)-conjugated mAb purchased from PharMingen or Becton Dickinson (Mountain View, CA). Irrelevant mAb control values were subtracted from values obtained with relevant mAbs. All results were obtained using a FACScalibur (Becton Dickinson). Forward and side scatter settings were gated to exclude red cells and debris, and 1 × 104 cells were analyzed for each determination.
In vitro MLR cultures and CTL assessment
For quantifying MLR responses, CD4+ LN T cells or TDL were mixed with irradiated (30 Gy) TCD splenocyte stimulators from host strain mice. Cells were suspended in MLR media consisting of Dulbecco’s MEM (Bio Whittaker, Walkersville, MD), 10% FCS (HyClone, Logan, UT), 2-ME (5 × 10−5 M) (Sigma, St. Louis, MO), 10 mM HEPES buffer, 1 mM sodium pyruvate (Life Technologies, Grand Island, NY), amino acid supplements (1.5 mM l-glutamine, l-arginine, and l-asparagine) (Sigma), and antibiotics (penicillin, 100 U/ml; streptomycin, 100 μg/ml) (Sigma). A total of 0.3–1 × 105 cell responders and 1 × 105 irradiated stimulators were plated into 96-well round-bottom (Costar) plates and placed at 37°C and 10% C02 for 1 to 6 days. In some wells, as indicated, anti-CTLA-4 mAb was added at a final concentration of 50 μg/ml. Δ cpm were calculated by subtracting the syngeneic proliferative response from the allogeneic proliferative response. No scintillant was used to amplify cpm detection. For CTL analysis, splenocytes were activated with Con A (2 μg/ml) for 3 days in MLR media and then labeled overnight with [3H]thymidine (4 μCi/ml) in 24-well plates containing 2 × 106 cells in a 2 ml vol. B6 or CD28−/− TDL T cells were spun at 500 rpm (Beckman TJ-6) for 5 min with targets (104 targets/well) at E:T ratios of 12.5:1–100:1 in MLR media and then incubated together for a period of 4 h. Cell pellets were harvested and then counted as described above for the MLR culture. The percent specific lysis was based upon the retention of [3H]thymidine in the experimental groups as compared with targets not exposed to effector cells (24).
In situ antisense:sense mRNA hybridization of TDL
TDL cytospins were hybridized with riboprobes specific for IL-2 237–837(237–837), IL-4 1–393(1–393), IL-5 1–1534(1–1534), IFN-γ 1–270(1–270), IL-10 392–937(392–937), granzyme B 239–775(239–775), and perforin 1–1700(1–1700) mRNA. The mRNA:RNA probe hybrids were detected using an alkaline phosphatase α-digoxigenin F(ab′)2 Ab and BCIP/NBT (5-bromo-4-chloro-3-indolyl phosphate/nitroblue tetrazolium) color substrate. The in situ hybridization technique has been described elsewhere (25).
Statistical analyses
Group comparisons of continuous data were made by Student’s t test. Survival data were analyzed by lifetable methods using the Mantel-Peto-Cox summary of χ2 (26). Actuarial survival and relapse rates are shown. p values < 0.05 were considered significant.
Results
Role of CD28:B7 interaction on regulating GVHD lethality early post-BMT
Blockade of CD28/CTLA-4:B7 interactions by CTLA4-Ig or anti-B7 mAb has been shown to diminish GVH alloresponses, resulting in a decrease in GVHD-induced mortality in some studies (17, 18, 19, 20, 27, 28, 29, 30). Because CTLA4-Ig and anti-B7 mAb inhibit both the CD28:B7 and CTLA-4:B7 interactions, the analysis of the contributions of each pathway individually in GVHD generation has required alternative approaches, including the use of CD28−/− mice. To determine whether precluding CD28:B7 interactions would have a beneficial effect on preventing GVH lethality, highly purified wild-type or CD28−/− T cells were infused into sublethally irradiated wild-type or MHC disparate recipients. In this setting, alloreactive donor T cells cause multiorgan system GVH tissue destruction (20), and because the hemopoietic compartment is a target of alloresponsive donor T cells, recipients frequently experience lethal BM aplasia. A major advantage of this type of system includes the close correlation between T cell dose and lethality rates.
To quantify the effect of CD28:B7 interactions on GVH lethality of CD4+ T cells, CD28−/− CD4+ T cells were infused at doses of 0.3–3 × 105 into sublethally irradiated bm12 (MHC class II only disparate) recipients. At all cell doses tested, CD28−/− CD4+ T cells were found to be incapable of causing GVHD lethality (Fig. 1⇓A). In contrast, recipients of as few as 0.3 × 105 wild-type cells had a 70% lethality rate, while recipients of 1 or 3 × 105 wild-type cells uniformly succumbed to GVHD. Weight loss was observed in recipients of wild-type, but not CD28−/− cells, reflective of survival results. Day 14 mean hematocrit values were ≤17% in recipients of ≥105 wild-type T cells that were significantly lower than the mean values of 25% in recipients of 0.3 × 105 wild-type T cells or ≥32% obtained from recipients of CD28−/− T cells.
The GVHD lethality capacity of CD28−/− T cells. Sublethally irradiated bm12 (A) or bm1 (B) or heavily irradiated B10.BR (C) recipients were given the indicated numbers of highly purified CD4+ T cells, CD8+ T cells, or nonpurified splenocytes, respectively, from CD28−/− or wild-type B6 donors on day 0. On the x-axis are the days posttransfer, and on the y-axis is the proportion of mice surviving. The numbers of recipients per group are listed. Data in the sublethal systems were pooled from two replicate experiments with similar results. A significant (p ≤ 0.01) decrease in GVHD-induced lethality with CD28−/− as compared with wild-type cells given at the same cell dose was noted in the bm12 and bm1 systems and at the highest splenocyte dose in the B10.BR system.
To quantify the effect of CD28:B7 interactions on GVH lethality of CD8+ T cells, CD28−/− CD8+ T cells were infused at doses of 0.3 or 1 × 106 into sublethally irradiated bm1 (MHC class I only disparate) recipients. The infusion of CD28−/− CD8+ T cells at a dose of 106 resulted in the mortality of 31% of recipients, while 0.3 × 106 cells were not lethal (Fig. 1⇑B). As few as 0.3 × 106 wild-type cells resulted in 69% lethality and 106 cells were 100% lethal. Weight curves were reflective of survival. Day 14 hematocrit values were significantly lower (≤17%) in recipients of wild-type cells as compared with recipients of CD28−/− T cells (≥27%). Based upon these data, there was at least a 10-fold reduction in the GVH lethality capacity of CD28−/− CD4+ T cells and >3- but <10-fold reduction in the GVH lethality of CD28−/− CD8+ T cells.
To determine whether CD4+ and CD8+ T cell-containing inocula from CD28−/− donors were capable of causing GVHD, we used a different model system in which the B6 (H2b) donor and B10.BR (H2k) recipient were mismatched at both MHC class I and class II loci. For this purpose and to simulate a clinical BMT setting in which there is no host anti-donor response as would be present in sublethally irradiated recipients, we used a model system in which the recipient is lethally irradiated and then given 5 or 15 × 106 B6 splenocytes along with TCD BM (Fig. 1⇑C). Recipients of the higher numbers of CD28−/− had a significantly higher actuarial survival rate as compared with wild-type controls, although both groups ultimately succumbed to GVHD lethality. The degree of GVHD protection was <3-fold since recipients of 15 × 106 CD28−/− splenocytes had a significantly lower survival rate than recipients of 5 × 106 wild-type cells. Recipients of as few as 5 × 106 CD28−/− cells had a high survival rate (80% at 100 days), but also had clinical and weight evidence of sublethal GVHD.
The lower degree of GVHD protection could have been due to the use of more irradiation in the B6→B10.BR model than in the sublethal system. To investigate this possibility, we chose a different system in which donor CD4+ and CD8+ T cells are infused into an MHC class I and class II disparate recipient (BALB/c-SCID) that does not require any TBI conditioning of the recipient to permit donor T cell-mediated GVHD lethality. NK-depleted BALB/c-SCID mice were given 2 × 106 T cells from B6 or CD28−/− donors. Ninety percent of recipients of either CD28−/− or wild-type cells succumbed to GVHD mortality within 1 mo after transfer, with both groups averaging >30% loss in mean body weights (data not shown). Together, these data suggest that the reduced GVHD response observed when low numbers of CD28−/− CD4+ or CD8+ T cells are infused into sublethally irradiated recipients can be overwhelmed when larger numbers of donor CD28−/− CD4+ and CD8+ T cells are coinfused into MHC disparate recipients.
Blockade of CTLA-4:B7 interactions increases donor T cell expansion and GVHD lethality, as well as host-mediated BM graft rejection early post-BMT
The reduced GVHD lethality observed with CD28−/− T cells in sublethally irradiated recipients could be due to reduced expansion of CD28−/− T cells due to the absence of a positive regulatory signal or to unperturbed CTLA-4:B7 interactions due to the presence of a negative regulatory signal. In the latter instance, CTLA-4:B7 interactions might be expected to down-regulate donor T cell responses. To directly investigate the latter possibility, anti-CTLA-4 mAb was given in vivo to block CTLA-4:B7 interactions. To address the possibility that anti-CTLA-4 mAb could directly affect donor anti-host T cell alloresponses, in vivo donor T cell expansion and function and GVHD mortality were quantified.
To quantify donor T cell expansion unencumbered by a host anti-donor T cell response, BALB/c-SCID recipients were depleted of NK cells and then given 0.2 × 106 or 2 × 106 T cells from (B6 × 129)F2 (H2b) donors. On days 4 and 6 posttransfer, the number of TDL was quantified in recipients of 2 × 106 cells as an indicator of donor T cell expansion in vivo. We chose to isolate TDL rather than study splenic T cells since more donor T cells are collectable in the lymphatics over an 18-h period than present in the spleen and because splenic but not TDL T cells reside in an environment that inhibits the generation of an immune response. At both time periods, recipients of anti-CTLA-4 mAb had a 2-fold increase in TDL T cell number as compared with those that received irrelevant mAb (day 4, 0.295 × 106/ml vs 0.155 × 106/ml; day 6, 8.14 × 106/ml vs 4.17 × 106/ml, respectively) (data not shown). In a different experiment, recipients were given lower numbers of T cells (0.2 × 106) and cannulated on day 9 posttransfer. Once again, the number of TDL was 2-fold higher in anti-CTLA-4 mAb-treated as compared with irrelevant mAb-treated controls (1.09 × 106/ml vs 0.49 × 106/ml, respectively) (data not shown). Phenotypic analysis of days 4, 6, and 9 TDL cells showed no substantial differences in either the proportion of CD4+ and CD8+ T cells or the expression of a broad panel of activation Ags (data not shown). Proliferative responses to host stimulator cells were measured using day 6 posttransfer TDL from irrelevant or anti-CTLA-4 mAb-treated recipients. In both experiments, peak responses to host stimulator cells were seen after 1 day of culture using TDL obtained from both cohorts as compared with the typical 5 days required for peak response using TDL obtained from non-BMT donor-strain controls. Results from one of these two replicate experiments (Fig. 2⇓) are consistent with an in vivo priming effect. In situ hybridization analysis for cytokine mRNA in fresh day 6 TDL obtained from a pool of four to five mice in each group indicated that anti-CTLA-4 mAb led to a skewed Th2 response, as indicated by an increase in the percentages of TDL cells expressing IL-4 mRNA (1.2% vs 16.4%, respectively), although the percentage of IL-10 mRNA-expressing cells remained similar. The percentage of TDL cells expressing Th1 cytokines (IL-2, IFN-γ) was low in both the control and anti-CTLA-4 mAb groups (<0.3%). The frequency of TDL cells expressing granzyme B was comparable (12.3% vs 11.4%, respectively).
Anti-CTLA-4 mAb infusion increases the expansion of allogeneic donor T cells without affecting the in vitro proliferative responses to alloantigen-bearing stimulators. BALB/c-SCID recipients were depleted of NK cells and given (B6 × 129)F2 T cells (2 × 106) with either hamster IgG (hIgG) irrelevant mAb or anti-CTLA-4 mAb, as described in Materials and Methods. TDL were isolated from cohorts of recipients (n = 6–8 mice/group) given 2 × 106 donor T cells. On day 6 posttransfer, a mean of 4.17 or 8.14 × 106 TDL T cells/ml was collected from recipients of irrelevant mAb or anti-CTLA-4 mAb, respectively. The in vitro proliferative response of these TDL to host alloantigen-bearing stimulator cells is shown. On the y-axis is mean cpm using a log10 scale, and on the x-axis are days in culture. The error bars shown are 1 SD. cpm for syngeneic responses were <10% of cpm for allogeneic responses. These findings were replicated in a second experiment.
To determine whether the increased T cell expansion observed in anti-CTLA-4 mAb-treated recipients would increase mortality rates, we used our well-studied lethal TBI system to simulate a clinical BMT setting. B10.BR recipients were irradiated, reconstituted with high numbers of B6 donor BM along with donor splenocytes, and given either irrelevant or anti-CTLA-4 mAb. Anti-CTLA-4 mAb-treated recipients had a significantly (p = 0.001) lower actuarial survival rate as compared with controls, with the vast majority of mAb-treated recipients dying by day 9 post-BMT (Fig. 3⇓). Weight curves (data not shown) and clinical appearance of recipients were reflective of survival curves, consistent with a fulminant GVHD process in anti-CTLA-4 mAb-treated recipients. These findings were reproduced in a second experiment.
The roles of CTLA-4:B7 interaction on GVHD induced lethality in heavily irradiated recipients of allogeneic donor splenocytes. B10.BR recipients were irradiated (8 Gy TBI) and then given B6 TCD BM plus supplemental splenocytes (15 × 106) and either hamster IgG irrelevant mAb or anti-CTLA-4 mAb, as described in Materials and Methods. On the x-axis are the days post-BMT, and on the y-axis are the proportion of mice surviving. Data from two replicate experiments with similar results are pooled.
Our data clearly indicate that anti-CTLA-4 mAb infusion enhances the expansion of donor T cells exposed to host alloantigens in vivo. To determine whether anti-CTLA-4 mAb would increase host anti-donor T cell-mediated BM rejection, we needed to analyze sublethally irradiated recipients reconstituted with allogeneic BM cells. To facilitate graft rejection, BM was depleted of T cells to preclude a graft-enhancing effect of donor T cells. The separate effects of host CD4+ and CD8+ T cell-mediated graft rejection were analyzed using the same two strain combinations as for quantifying the GVH responses of purified donor CD4+ and CD8+ T cells.
Donor TCD BM was infused in sublethally irradiated recipients of MHC class I (bm1→B6) or II (bm12→B6) disparate recipients. Peripheral blood chimerism of T, B, and myeloid lineages was assessed at time periods up to 145 days post-BMT. In both strain combinations, recipients given anti-CTLA-4 mAb had significantly lower proportions of donor cells and higher proportions of host cells, with all lineages being affected (Table I⇓). Particularly striking were the increases in the proportion of host CD4+ and CD8+ T cells in the anti-CTLA-4 mAb-treated recipients, indicating that anti-CTLA-4 mAb stimulates a host anti-donor response culminating in a higher degree of graft resistance. These data provide a likely explanation for the improved survival rate in sublethally irradiated recipients of allogeneic T cells and anti-CTLA-4 mAb (data not shown), which is presumably due to an augmented host anti-donor resistance process, limiting the GVH lethality effect of donor T cells. Together with the GVH results, these data indicate that the selective targeting of CD28:B7 rather than targeting both CD28:B7 and CTLA-4:B7 interactions would be desirable in preventing GVH lethality and preserving alloengraftment early post-BMT.
Decreased long-term chimerism in anti-CTLA-4 mAb-treated allogeneic recipients of MHC class I or II disparate T cell-depleted donor BM graftsa
Anti-CTLA-4 mAb augments the GVL effect of delayed donor splenocyte infusion
The enhanced donor T cell expansion caused by anti-CTLA-4 mAb could be advantageous in augmenting the GVL effect of donor T cells given post-BMT. Because anti-CTLA-4 mAb increases GVHD lethality when given early post-BMT, we elected to study the potential GVL effect of anti-CTLA-4 mAb given later post-BMT when GVHD lethality of donor T cells is markedly reduced. For this purpose, we utilized a model of delayed splenocyte infusion in which lethally irradiated B6 recipients are reconstituted with B10.BR TCD BM for a period of 3 wk before donor splenocyte infusion (21). Donor T cells given at this later time period post-BMT have a markedly lower capacity to cause GVHD lethality than when given on day 0 of BMT (21). One week after donor splenocyte infusion, recipients are challenged with a supralethal dose of AML cells of the B6 strain. In previous studies, we have shown that the GVL effects of delayed splenocyte infusion against AML cells in this model are dependent upon the binding of B7 ligand(s) to T cell counter-receptor(s) (21). Donor CD8+ T cells and, to a lesser extent, CD4+ T cells present in the donor spleen cells are responsible for the GVL effect of DLI in this setting, while NK cells do not substantially contribute to this response (21).
To discern whether blockade of CTLA-4:B7 binding would enhance the GVL effect against AML cells, anti-CTLA-4 mAb was given to reconstituted recipients of delayed post-BMT donor splenocyte infusion, which are challenged with AML cells. Donor splenocytes provided a GVL effect that delayed the occurrence of leukemia as compared with recipients not given donor splenocytes (Fig. 4⇓A), although all mice treated with donor splenocytes eventually succumbed with leukemia by day 75 post-BMT. In striking contrast, none of the anti-CTLA-4 mAb-treated recipients of donor splenocytes and AML cells examined during the entire 220-day risk period had evidence of leukemia, despite the fact that donor splenocyte infusion alone did not prevent leukemia occurrence in any of the recipients analyzed 75 days post-BMT.
Anti-CTLA-4 mAb infusion augments the GVL capacity of delayed donor splenocyte infusion in recipients challenged with acute myeloid leukemia cells. B6 mice were irradiated with 8 Gy TBI on day −1 and then given B10.BR T and NK cell-depleted BM (8 × 106) on day 0 of BMT. On day 21, cohorts of mice, as indicated, were given B10.BR splenocytes (25 × 106). Anti-CTLA-4 mAb or irrelevant hamster IgG was administered as described in Materials and Methods. On day 28, C1498 acute myeloid leukemia cells were given i.v. at a supralethal dose (2 × 105). Ten mice per group were transplanted. A, Leukemia incidence. All mice dying post-BMT that had received C1498 cells were examined for the presence of tumor. The proportion of mice with identifiable leukemia is shown on the y-axis, and days post-BMT on the x-axis. Eight to nine mice per group were analyzed for relapse. Differences in actuarial rates of leukemia are as follows: C1498 vs spleen/C1498 (p = 0.00094); C1498 vs spleen/C1498/anti-CTLA-4 mAb (p = 0.0015). B, On the x-axis are the days post-BMT, and on the y-axis is the proportion of mice surviving. The actuarial survival is plotted. As compared with control recipients of no spleen/C1498, differences in actuarial survival rates were as follows: spleen/no C1498 (p = 0.0079); spleen/C1498 (p = 0.00069); spleen/C1498/anti-CTLA-4 mAb (p = 0.014). There was no significant survival difference (p = 0.28) between groups of recipients that received spleen/C1498 with or without anti-CTLA-4 mAb.
The overall survival of recipients of donor splenocytes, AML cells, and anti-CTLA-4 mAb was modestly, but not significantly (p = 0.28), decreased, as compared with a group of mice receiving donor splenocytes, no AML cells, and no mAb treatment (Fig. 4⇑B). Anti-CTLA-4 mAb-treated recipients had a tendency toward lower weights than controls that received neither AML cells nor anti-CTLA-4 mAb (data not shown). Of note, the 100-day GVHD mortality risk, presumably the greatest risk period for observing mAb-induced GVHD acceleration of donor splenocytes, was highly comparable with survival rates of 44% or 50% in these two groups, respectively. Taken together, these data indicate that anti-CTLA-4 mAb given later post-BMT in the context of DLI mediated a potent GVL effect, while at most only slightly increasing the GVH side effects of DLI.
Role of CTLA-4:B7 interactions on modulating CD28-dependent and CD28-independent T cell responses
CTLA-4:B7 interaction may serve to regulate CD28:B7 responses. Therefore, experiments were performed to examine the role of CTLA-4:B7 in modifying the responses of CD28−/− T cells to alloantigens. We first examined CD4+ T cell responses obtained from wild-type or CD28−/− donors on in vitro proliferative responses (Fig. 5⇓A) and in vivo GVH lethality responses (Fig. 5⇓B) to MHC class II disparate bm12 strain mice in the presence or absence of anti-CTLA-4 mAb. In vitro proliferative responses of CD28−/− CD4+ T cells to bm12 stimulator cells were lower than observed using CD4+ T cells from wild-type controls beginning on day 2 of culture (Fig. 5⇓A). Anti-CTLA-4 mAb increased proliferation of wild-type but not CD28−/− T cells.
Anti-CTLA-4 mAb increases the in vitro proliferation and in vivo GVHD capacity of wild-type but not CD28−/− CD4+ T cells responsive to MHC class II disparate stimulator cells. In A, results of proliferation studies, using purified B6 wild-type or CD28−/− CD4+ T cells incubated with bm12 TCD splenic stimulators in the presence of anti-CTLA-4 mAb (50 μg/ml), are shown. On the x-axis are the days of cell culture. The mean cpm are shown on the y-axis using a log10 scale. Error bars represent 1 SD of the mean. cpm for syngeneic responses were <10% of cpm for allogeneic responses. The in vivo GVHD results for 0.3 × 106 (B) or 1 or 3 × 106 wild-type or 1 × 106 CD28−/− T cells (C), infused along with TCD BM into heavily irradiated recipients, are shown. As indicated, recipients were given either irrelevant or anti-CTLA-4 mAb. On the x-axis are the days post-BMT, and on the y-axis is the proportion of mice surviving. All groups had five mice, except for the groups receiving 106 CD28−/− CD4+ T cells, which had 10 mice per group. In B, recipients of 0.3 × 106 wild-type but not CD28−/− T cells and anti-CTLA-4 mAb had a significantly (p = 0.001) lower survival than those given irrelevant mAb. In C, recipients given 1 × 106 wild-type or 1 × 106 CD28−/− T cells and anti-CTLA-4 mAb each had a trend (p = 0.08) toward a lower survival rate than recipients given irrelevant mAb. <226>, p <0.05 vs CD28+/+ group; #, p <0.05 vs CD28-/- group that did not receive mAb.
To determine whether the absence of CD28:B7 interactions would affect the accelerated in vivo GVH effects of anti-CTLA-4 mAb infusion, bm12 recipients were given a uniformly sublethal number of B6 wild-type or CD28−/− CD4+ T cells (0.3 × 106) and either irrelevant or anti-CTLA-4 mAb. In recipients of wild-type T cells, survival was 100%, significantly higher than the 0% observed in recipients that were also given anti-CTLA-4 mAb (Fig. 5⇑B). In contrast, recipients given CD4+ T cells from CD28−/− donors had 100% survival regardless as to whether these mice received irrelevant or anti-CTLA-4 mAb. To attempt to uncover a more modest effect of anti-CTLA-4 mAb on accelerating GVHD lethality of CD28−/− CD4+ T cells, the number of donor T cells was increased to 106 per recipient. Under these conditions, acceleration of GVHD by anti-CTLA-4 mAb given to recipients of wild-type CD4+ T cells was difficult to assess since these recipients all succumbed by day 10 post-BMT. Anti-CTLA-4 mAb-treated recipients uniformly died by day 9 post-BMT. Recipients (n = 10/group) given 106 CD4+ T cells from CD28−/− donors and irrelevant mAb had an 80% long-term (>2.5-mo post-BMT) survival rate (Fig. 5⇑C). Anti-CTLA-4 mAb-treated recipients had a 50% survival rate that was of borderline (p = 0.08) significance. Because recipients of a 3-fold higher number of CD28−/− CD4+ T cells (3 × 106) had 0% survival, these data would indicate that anti-CTLA-4 mAb infusion resulted in <3-fold increase in CD28−/− CD4+ T cell-mediated lethality.
To determine whether alloreactive CD28−/− CD4+ and CD8+ T cells would be stimulated to expand in the presence of anti-CTLA-4 mAb, TDL were obtained from cohorts of lethally irradiated B10.BR recipients of B6 or CD28−/− donor splenocytes on days 6 and 9 post-BMT. At the time of peak expansion (day 6), donor T cell expansion was significantly higher in recipients of wild-type as compared with CD28−/− donor cells (Fig. 6⇓A). The increase in TDL T cell number in anti-CTLA-4 mAb-treated BMT recipients was CD28 dependent since no increases in TDL T cells were observed in mAb-treated recipients of CD28−/− T cells. Thus, under these conditions, the negative regulatory role of CTLA-4:B7 interaction is CD28 dependent. Flow cytometry analysis of TDL obtained on days 6 and 9 did not reveal any obvious differences in activation profiles (data not shown). In recipients of wild-type and CD28−/− donor splenocytes, donor CD4+ T cells comprised a higher proportion of the TDL on days 6 and 9 post-BMT in recipients given anti-CTLA-4 mAb as compared with irrelevant hamster IgG mAb (≥50% increase over baseline percentages at both time points). In situ hybridization analysis for cytokine mRNA was performed using fresh TDL obtained from a pool of four mice per group. In recipients of CD28−/− T cells, TDL were skewed toward a Th2 phenotype, as evidenced especially by increases in the frequency of IL-5 (5.7-fold) and IL-10 (5.5-fold), with a concomitant decrease in IFN-γ mRNA-expressing cells (Table II⇓). In addition, as observed in the (B6 × 129)F2→BALB/c-SCID system, anti-CTLA-4 mAb administration led to a skewed Th2 response in TDL obtained from recipients of wild-type cells, and further skewed the Th2 response of CD28−/− T cells increasing the frequency of TDL cells expressing IL-4, IL-5, and IL-10 (≥2.5-fold), while decreasing the frequency of IFN-γ-expressing cells to frequencies that were 3-fold to 6-fold lower than controls. Also as previously noted, anti-CTLA-4 mAb did not substantially alter the percentage of granzyme B and perforin mRNA-expressing TDL T cells (Table II⇓).
The increased expansion of allogeneic donor T cells induced by anti-CTLA-4 mAb is CD28 dependent. B10.BR recipients were irradiated (8 Gy TBI) and then given B6 or CD28−/− TCD BM plus supplemental splenocytes (15 × 106), and either hamster IgG irrelevant mAb or anti-CTLA-4 mAb, as described in Materials and Methods. TDL were isolated from each group on days 6 (n = 6/group) and 9 (n = 6–7 per group, except for recipients of wild-type splenocytes and anti-CTLA-4 mAb that had a high early post-BMT mortality rate (n = 3/group)). A, Quantification of TDL cell number. On the y-axis are the number of TDL per ml quantified in individual mice, and on the x-axis are the days post-BMT. Groupwise comparisons were made. B, TDL proliferation. On the y-axis is mean cpm using a log10 scale, and on the x-axis are days in culture. Error bars represent 1 SD. cpm for syngeneic responses were <10% of cpm for allogeneic responses. C, TDL CTL function. On the y-axis is percent specific cytolysis. E:T ratios are indicated on the x-axis. Error bars represent 1 SD. Significant differences between CD28−/− and wild-type recipients of irrelevant mAb are designated by ∗. Significant differences between recipients that received anti-CTLA-4 mAb as compared with irrelevant mAb are designated by #.
Role of CD28 and CTLA-4 in supporting the in vivo generation of cytokine-producing cells in irradiated B10.BR recipients of B6 allogeneic T cellsa
TDL T cells were examined for in vitro proliferative responses to host alloantigen-bearing stimulator cells (Fig. 6⇑B). Peak responses in all groups on day 1 were indicative of in vivo priming. Wild-type TDL had greater proliferation to stimulator cells than CD28−/− TDL. Proliferative responses of TDL obtained from anti-CTLA-4 mAb-treated recipients were not higher than observed with TDL from irrelevant mAb-treated recipients. Cytolysis against host target cells was higher when using CD28−/− T cells as effector cells (Fig. 6⇑C). Anti-CTLA-4 mAb did not increase the cytolytic capacity of TDL obtained from either irrelevant or anti-CTLA-4 mAb-treated recipients, despite the fact that the proportion of CD8+ T cells was lower in TDL obtained from anti-CTLA-4 mAb versus irrelevant mAb-treated recipients. Therefore, although CTLA-4:B7 regulates the expansion of wild-type but not CD28−/− T cells, neither the in vitro anti-host proliferative nor cytolytic responses of TDL T cells from recipients of wild-type or CD28−/− appear to be substantially increased by anti-CTLA-4 mAb infusion.
Discussion
We demonstrate that the CD28:B7 interaction is required for optimal donor T cell expansion in vivo and GVHD lethality in systems in which limited numbers of cells are infused. When high numbers of T cells are infused, the requirements for CD28:B7 interaction are lessened. CTLA-4:B7 interaction is required to down-regulate the CD28-dependent stimulatory effect on donor T cell expansion. In the presence of anti-CTLA-4 mAb, which prohibits CTLA-4:B7 interaction, GVH lethality is worsened due to the dominant effect of the CD28:B7 pathway. Under conditions in which there is minimal donor anti-host T cell responses and more maximal host anti-donor T cell responses, CTLA-4:B7 interaction is required to down-regulate host T cell-mediated allogeneic BM graft rejection. In the later post-BMT period, BM engraftment already has occurred and the GVH responses of donor T cells are less severe than on the day of BMT. Under these conditions, the delayed administration of anti-CTLA-4 mAb along with donor splenocytes markedly augmented the GVL capacity of these donor T cells without substantially increasing GVH-related side effects. These data have important implications for the selective targeting of these two pathways post-BMT.
Blockade of CD28/CTLA-4:B7 interactions by CTLA4-Ig or anti-B7 mAb has been shown to diminish GVH alloresponses, resulting in a decrease in GVHD-induced mortality in some studies (14, 15, 16, 17, 27, 28, 29, 30, 31, 32). Because CTLA4-Ig and anti-B7 mAb preclude both the CD28:B7 and CTLA-4:B7 interaction, the separate contributions of these pathways in GVHD generation have required alternative approaches, including the use of CD28−/− mice. In vitro, alloresponses of CD28−/− T cells are markedly lower than controls. Indeed, CD28−/− CD4+ or CD8+ T cells have a reduced GVHD lethality capacity or >10-fold and >3-fold, respectively, when infused into sublethally irradiated recipients, confirming and extending studies of Anasetti and coworkers (32). In some experiments in the strain combinations used for our studies, differences in donor T cell expansion were sufficient to provide a significant degree of protection from GVHD lethality, especially under situations in which the onset of lethal GVHD was not rapid. However, in other strain combinations in which recipients uniformly succumbed within 10–22 days following allogeneic donor T cell infusion, CD28:B7 interaction was not necessary for optimal GVH lethality. Others have shown either a slight or no delay in GVHD-induced lethality in sublethally irradiated recipients given high numbers of allogeneic CD28−/− donor splenocytes (30) or a markedly diminished mortality rate, but with significant histologic GVHD in heavily irradiated F1 recipients of CD28−/− parental T cells (31). As measured by thoracic duct cannulation, allogeneic CD28−/− T cells will expand in vivo, albeit to a far lesser extent than wild-type cells. Similar conclusions were reached by Anasetti and coworkers, who analyzed the spleens of unirradiated F1 recipients of CD28−/− donor spleen cells (32). At high T cell doses infused, there could be sufficient numbers of CD28−/− T cells available to cause GVHD. Although CD28−/− T cells may respond to peptide and alloantigen, the response is not sustained, and therefore full expansion may not be achieved (33). The relative importance of CD28:B7 costimulation on GVHD induction will most likely depend upon the number and subset of T cells infused, threshold number of activated T cells required to induce GVHD, and rapidity and extent of GVHD-induced lethality in the control group.
In recipients of wild-type T cells, anti-CTLA-4 mAb administration led to 2–3-fold increase in the number of TDL T cells, especially CD4+ T cells, similar to results in a CD4+ T cell transgenic system (10). The preferential and CD28-dependent expansion of CD4+ T cells in response to blockade of CTLA-4:B7 interaction by anti-CTLA-4 mAb in our experiments has been observed in CTLA-4−/− mice that develop a lymphoproliferative disorder initiated by CTLA-4−/− CD4+ T cells (34). These data also are consistent with those of Abbas and colleagues who have shown that anti-CTLA-4 mAb block the induction of tolerance in vivo (14). The increase in TDL T cell number in anti-CTLA-4 mAb-treated BMT recipients was CD28 dependent. Wild-type and CD28−/− TDL cells generated CTLs capable of lysing host alloantigen, the magnitude of which was at least as high when using CD28−/− as was observed with wild-type TDL cells, consistent with other investigators (30, 31, 35) and our own data on granzyme B mRNA expression in CD28−/− TDL T cells. We interpret the data of Anasetti, who noted a marked reduction in the anti-host-reactive proliferative and CTL capacity of posttransplant spleen cells obtained from unirradiated F1 recipients of CD28−/− donor spleen cells to be reflective of the compromised alloengraftment capacity of CD28−/− T cells in that setting (32). When incubated with host-type stimulator cells, wild-type and CD28−/− TDL had a peak proliferative response on day 1 of culture, indicative of an in vivo priming against host alloantigens, the magnitude of which was greater with wild-type TDL. Anti-CTLA-4 mAb in vivo did not increase the magnitude of the proliferative response of wild-type or CD28−/− donor TDL T cells obtained at the height of peak in vivo alloresponsiveness, although the total number of donor TDL T cells was significantly increased. Even though the function of donor TDL T cells was not affected by mAb infusion, the net effect of anti-CTLA-4 mAb administration in wild-type recipients is to increase the number of donor TDL T cells with anti-host reactivity.
Our data indicate that the majority of anti-CTLA-4 mAb effects was CD28 dependent. Similar to our observations that CTLA-4:B7 interaction did not have a positive regulatory role of T cell proliferation, Green et al. showed that the in vitro alloresponses of CD28−/− T cells were not inhibitable by the addition of CTLA4-Ig fusion protein to the MLR culture (35). In contrast, others have shown that CTLA-4:B7 interaction can have a negative regulatory role on the capacity of CD28−/− recipients to respond to tumor Ags or alloantigen-bearing cardiac grafts (16, 36). The apparent conflicting results between these studies and our data may be related to the type of T cell, APC, Ag, murine strain, or exact type of immune response being measured, as well as the requirement for CD28:B7 interaction in that response. Nonetheless, our data clearly indicate that, under some conditions, the negative regulatory role of CTLA-4:B7 interaction is CD28 dependent.
An interesting observation in the present study is the finding that the percentage of TDL cells expressing Th2 cytokine mRNA was increased in recipients receiving allogeneic T cells and anti-CTLA-4 mAb infusion. Th2 skewing was independent of the presence of CD28. A similar skewing toward a Th2 cytokine phenotype was observed when CD4+CTLA-4−/− TCR transgenic T cells were reprimed with Ag in vitro (A.H.S., unpublished observations). Consistent with our results, CD28 costimulation has been shown to promote Th2 cytokine production (37). Recent data from our group have shown that GVHD lethality can be significantly reduced in heavily irradiated B10.BR recipients of allogeneic donor T cells from B6 Th2-defective (IL-4-deficient) donors (38), suggesting that the accelerated GVHD responses observed in anti-CTLA-4 mAb-treated recipients were due to Th2 skewing. However, the effect of Th1 or Th2 cytokine-producing donor T cells in regulating GVHD responses is most likely dependent upon the particular transplantation setting analyzed. For example, other data from our group have shown that T cells from IL-10-deficient donors cause an increase in GVHD lethality when infused into sublethally irradiated recipients (39). Other investigators have shown that CD4+ and CD8+ Th2 cells can be associated with a lower risk for GVH and less potent GVL than T cells with a Th1 phenotype (40, 41, 42). Thus, we do not know whether the anti-CTLA-4-facilitated induction of Th2 cells contributed to the observed increases in the GVH and GVL effects of wild-type T cells. Nonetheless, collectively, these data indicate that CTLA-4:B7 interaction can regulate Th2 differentiation.
We have shown that anti-CTLA-4 mAb administration had a profound effect on stimulating the GVL response of delayed splenocyte infusion in recipients challenged with MHC class I+II− AML cells, consistent with data from others that have shown that anti-CTLA-4 mAb administration stimulates a potent anti-tumor response in naive mice (43, 44). Because GVHD mortality is known to be lower when donor T cells are given later as contrasted to earlier post-BMT (21, 45), the later post-BMT infusion of anti-CTLA-4 mAb may be more tolerable in terms of GVH side effects than when given early post-BMT. The fact that anti-CTLA-4 mAb infusion did significantly increase GVL without GVHD in this setting does not indicate that GVL and GVHD are separable events. For example, if fewer donor T cells are required to mediate GVL than GVHD lethality, then the increase in donor T cell expansion or function induced by mAb infusion may be of a sufficient magnitude to support a potent GVL effect without an obvious increase in GVHD risk. In our GVL system, anti-B7 mAb infusion eliminates, while transduction of the tumor line with B7-1 markedly increases the GVL effect (21). Because AML B7-1 transduction is cumbersome and the GVH response of DLI was not significantly augmented by anti-CTLA-4 mAb infusion, the inclusion of anti-CTLA-4 mAb along with delayed lymphocyte infusion could represent a new approach to increasing the GVL efficacy in patients relapsing post-BMT.
In summary, our data would indicate that, for GVHD prevention, competitive antagonists of CD28:B7 interaction, which preserve CTLA-4:B7 binding, would be preferable to inhibitors of both pathways. Such an approach could inhibit GVHD generation without adversely affecting the engraftment of TCD BM. Because GVHD was observed when high numbers of CD28−/− CD4+ and CD8+ T cells were infused together, the coblockade of additional pathways such as the CD40 ligand:CD40 costimulatory pathway (29, 46) or an adhesion pathway (19) will be necessary to further reduce GVHD lethality. Anti-CTLA-4 mAb infusion was a potent means of augmenting the GVL effect of donor splenocytes given later post-BMT at a time period when the risks of graft rejection and GVHD are lower. The inclusion of anti-CTLA-4 mAb with DLI in patients that relapse post-BMT warrants investigation. The enhanced GVHD, graft rejection, and GVL responses in anti-CTLA-4 mAb-treated recipients are consistent with the hypothesis that CTLA-4:B7 interaction results in the delivery of a negative signal (5, 6, 7, 11, 12, 13), although Linsley and Liu have proposed that CTLA-4:B7 delivers a positive signal to the T cell (4, 8, 15). Whatever the true nature of the CTLA-4:B7 signaling of T cells, anti-CTLA-4 mAb could be exploited to have a beneficial effect on post-BMT outcome. Together, the data presented in this work have significant potential implications to modifying the CD28/CTLA-4:B7 pathways for humans undergoing BMT.
Acknowledgments
We thank Dr. Jeffrey Bluestone (University of Chicago, Chicago, IL) for providing the anti-CTLA-4 mAb hybridoma used in these studies.
Footnotes
-
↵1 This work was supported in part by grants from the National Institutes of Health (RO1 Grants AI-34495, HL-56067, and CA-72669; and P01 AI-35296).
-
↵2 Address correspondence and reprint requests to Dr. Bruce R. Blazar, University of Minnesota Hospital, Box 109, Mayo Building, 420 S.E. Delaware Street, Minneapolis, MN 55455. E-mail address: blaza001{at}maroon.tc.umn.edu
-
↵3 Abbreviations used in this paper: CTLA, cytolytic T lymphocyte-associated Ag; BM, bone marrow; BMT, BM transplantation; DLI, delayed donor lymphocyte infusion; GVH, graft versus host; GVHD, GVH disease; GVL, graft versus leukemia; Gy, Gray; LN, lymph node; TBI, total body irradiation; TCD, T cell-depleted; TDL, thoracic duct lymphocytes.
- Received December 2, 1998.
- Accepted March 11, 1999.
- Copyright © 1999 by The American Association of Immunologists