The graft-versus-leukemia (GVL) effects and graft-versus-host disease (GVHD)–inducing activity of CD8 T cells was compared in murine recipients of wild-type (WT) or interferon γ (IFN-γ)–deficient (GKO) allogeneic donor cells. CD8 T cells (or CD4-depleted splenocytes) from GKO donor mice induced more severe GVHD in lethally irradiated allogeneic recipients compared to the same cell populations from WT donors. Consistent with GVHD severity, donor CD8 T-cell expansion in allogeneic recipients was augmented in the absence of IFN-γ. These results demonstrate that IFN-γ does not stimulate but instead down-modulates GVHD induced by donor CD8 T cells. Remarkably, antihost lymphohematopoietic reactions, including GVL effects against host leukemia/lymphoma cells, of CD8 T cells correlated inversely with their GVHD-inducing activity, and those of GKO donors were markedly weaker than those mediated by WT donor CD8 T cells. These data show for the first time that GVHD-inducing activity and GVL effects of allogeneic CD8 T cells can be separated by a single cytokine, IFN-γ.

Allogeneic bone marrow transplantation (BMT) is an effective therapeutic approach for the treatment of otherwise fatal hematologic malignancies and nonmalignant hematopoietic disorders. Although reduced leukemic relapse rates resulting from graft-versus-leukemic (GVL) effects have been observed in patients receiving HLA antigen-mismatched marrow compared to HLA-identical transplants,1-5 the high incidences of graft-versus-host disease (GVHD) and GVHD-induced immunodeficiency present an enormous obstacle to HLA-mismatched BMT in humans.6-8 Thus, a major challenge is to separate beneficial GVL effects from GVHD. Although T-cell depletion of donor marrow can inhibit GVHD, and some studies have shown that GVL effects can be induced in recipients of allogeneic natural killer (NK) cells9-11 or T cell–depleted (TCD) allogeneic BMT,12,13 a strong association of leukemic relapse with TCD BMT has been proven in a number of animal studies and clinical trials.14-19 The high incidence of leukemic relapse associated with T-cell depletion indicates that in addition to GVHD, efficient GVL effects against certain leukemias are also largely dependent on donor T cells. Thus, methods that can selectively inhibit the GVHD-promoting activity of allogeneic T cells while preserving allogeneic T cell–mediated GVL effects would be highly beneficial in the use of allogeneic BMT for the treatment of leukemia.

Previous studies have shown that interferon-γ (IFN-γ) is required for the inhibition of CD4-dependent GVHD and preservation of GVL effects of donor CD8 T cells in allogeneic BMT recipients treated with a single dose of interleukin 12 (IL-12) at the time of transplantation.20-22 In the present study, we have investigated the role of IFN-γ in the GVHD- and GVL-associated alloreactivity of the donor CD8 T-cell subset in mice transplanted with CD4-depleted or purified CD8+ splenocytes from WT or IFN-γ–deficient allogeneic donors. Our results demonstrate that allogeneic CD8 T cells induce more severe systemic GVHD but weaker GVL/antihost lymphohematopoietic reactions if they are incapable of producing IFN-γ. Thus, GVHD- and GVL-associated alloresponses of CD8 T cells can be dissociated by an IFN-γ–dependent mechanism.

Mice

Specific pathogen-free wild-type (WT) and IFN-γ knockout (GKO) mice on the BALB/c (BALB/c-Ifngtm1Ts, H-2d, KdIdDd) or C56BL/6 (C57BL/6-Ifngtm1Ts, H-2b, KbIbDb) backgrounds were purchased from the Jackson Laboratory (Bar Harbor, ME). WT C57BL/6 mice (B6, H-2b, KbIbDb) were purchased from the Frederick Cancer Research Facility (National Institutes of Health, Bethesda, MD). The 2C TCR transgenic mice on a C57BL/6 background were generously provided by Dr Dennis Y. Loh23 and were bred to the C57BL/6-Ifngtm1Ts mice to produce GKO 2C TCR transgenic progeny. Expression of the 2C TCR was detected by fluorescence-activated cell sorter (FACS) analysis using 1B2 anti-2C TCR monoclonal antibody (mAb; kindly provided by Dr Herman Eisen).24 IFN-γ genotyping was performed by tail DNA polymerase chain reaction (PCR) using primers for the endogenous (oIMR126 and oIMR127) or the targeted (oIMR128 and oIMR129) IFN-γ allele as described in the Jackson Laboratory Mice Database (www.jax.org). The sequences of these primers are: oIMR126, 5′-AGA AGT AAG TGG AAG GGC CCA GAA G-3′; oIMR127, 5′-AGG GAA ACT GGG AGA GGA GAA ATA T-3′; oIMR128, 5′-TCA GCG CAG GGG CGC CCG GTT CTT T-3′; oIMR129, 5′-ATC GAC AAG ACC GGC TTC CAT CCG A-3′. Mice were housed in sterilized microisolator cages and received autoclaved feed and drinking water.

Bone marrow transplantation

Sex- and age-matched B6 mice were lethally irradiated (9.75 Gy,137Cs source, 0.84 Gy/min) and reconstituted within 4 to 8 hours with 5 × 106 TCD allogeneic bone marrow cells (BMCs) and 7.5 to 10 × 106 CD4-depleted or 1 to 2.5 × 106 purified CD8+ spleen cells from WT or GKO BALB/c mice. Irradiated B6 mice transplanted with 5 × 106 TCD B6 BMCs were used as non-GVHD controls. In some experiments, 5 × 106 TCD B6 BMCs were also given to allogeneic recipients to provide a readout for lymphohematopoietic GVH reactions. In leukemia studies, recipients were additionally injected with 500 EL4 cells, a B6 T-cell leukemia/lymphoma cell line, as previously described.20 TCD was performed by incubating cells with anti-CD4 mAb (GK1.5) and anti-CD8 mAb (2.43) or with anti-CD4 only (for CD4 depletion), followed by rabbit complement, and the completeness of depletion was verified by FACS analysis as previously described.21,22 CD8+ splenocytes were purified using the MACS system (Miltenyi Biotec, Bergisch-Gladbach, Germany) according to the manufacturer's instructions. Briefly, mouse spleen cells were labeled with anti-CD8α (2.43)-coated magnetic microbeads and positively selected on a magnetic separation column. Aliquots of sorted CD8+ splenocytes were restained with fluorescein isothiocyanate (FITC)–conjugated anti-CD8β mAb (53-5.8; Pharmingen, San Diego, CA), and their purity was consistently greater than 97%. To avoid bias from cage-related effects, animals were randomized before and after BMT as described.25 Carcasses were saved in 10% formalin after death or euthanasia for autopsy. Tissues (spleen, liver, kidney, and lung) were embedded in paraffin, sectioned, and stained with hematoxylin and eosin. Necropsies and histologic analyses were performed on randomly chosen samples by observers who were unaware of which treatment group the carcasses belonged to, as previously described.26 

Induction of GVH with 2C T cells was performed by intravenous injection of 10 × 106 BMCs and 12 × 106 splenocytes from WT or GKO 2C TCR transgenic B6 donors into lethally irradiated (8 Gy) BALB/c mice. Non-GVHD controls received similar numbers of BMCs and splenocytes from syngeneic donors. As described above, animals were randomized before and after BMT.

FACS analysis

For the measurement of donor T-cell expansion and chimerism, recipient white blood cells (WBCs) were stained for 30 minutes at 4°C with antihost H-2Kb mAb 5F1-FITC (Pharmingen) and phycoerythrin (PE)–conjugated anti-CD8 mAb (53-6.7, Pharmingen). To block nonspecific FcR binding of labeled antibodies, 10 μL of undiluted culture supernatant of 2.4G2 (rat antimouse FcγR mAb)27 was added to the first incubation. The expression of IFN-γ receptor on EL4 cells was measured by staining cells with rat antimouse CD119-FITC (GR20, Pharmingen). FITC-labeled and biotinylated mouse IgG2a mAb HOPC-1 and PE-labeled rat IgG2a mAb (Pharmingen) were used as nonstaining negative control antibodies. Cells were washed with FACS buffer (Hanks balanced salt solution containing 0.1% bovine serum albumin [BSA] and 0.1% NaN3) between each and following the last stain, and were analyzed on a FACScan (Becton Dickinson, Mountain View, CA).

Proliferation assay

EL4 cells (2 × 103/well) and WEHI-279 cells (1 × 104/well) were incubated in triplicate in 96-well plates with various concentrations of recombinant murine IFN-γ (Pharmingen) in RPMI medium supplemented with 10% fetal calf serum (FCS; Sigma, St Louis, MO), 4% nutrient mixture (l-glutamine, nonessential amino acids, sodium pyruvate, penicillin/streptomycin), 1% Hepes buffer, and 10 μM 2-mercaptoethanol. Cultures were pulsed with 1 μCi (0.037 MBq)3H-thymidine 36 hours after incubation, and harvested 12 hours later. 3H-thymidine uptake was counted on a Betaplate β counter (Wallac, Gaithersburg, MD) and data are presented as the mean ± SD (cpm) of triplicate samples. WEHI-279, a murine lymphoma cell line that is sensitive to an antiproliferative effect of IFN-γ, was used as an assay control.

The Nunc 10-mm tissue culture inserts (0.4 μm polycarbonate membrane, Nalge Nunc International, Roskilde, Denmark) were used to determine the effects of cytokines released by alloreactive T cells (ie, mixed lymphocyte reaction supernatants) on EL4 cell proliferation. BALB/c spleen cells (2 × 106 in 0.5 mL) or 0.5 mL media were added to each well of 24-well culture plates (Nalge Nunc International) containing 30 Gy-irradiated B6 spleen cells (2 × 106 in 0.5 mL). After 10-mm tissue culture inserts were placed into each well, 2.5 × 104 EL4 cells (1 mL) were added into each insert. EL4 cells were harvested on days 1, 2, 3, and 4 after incubation, and the number of viable EL4 cells in each well was counted by trypan blue exclusion. Three wells in each group were harvested at each time point and data are presented as the mean ± SD. The culture insert used in this study allows the permeation into the insert of cytokines produced by spleen cells, but blocks any spleen cell–mediated direct killing of EL4 cells.

Alloreactive cytotoxic T-lymphocyte assay

BALB/c spleen cells were cultured in triplicate in 96-well plates with irradiated B6 spleen cells (30 Gy), at a 1:1 ratio (8 × 105/well) in RPMI supplemented with 10% FCS, 4% nutrient mixture, 1% Hepes buffer, and 10 μM 2-mercaptoethanol for 5 days. Responder cells were mixed with 51Cr-labeled EL4 (target) cells in 96-well plates (8000 cells/well) at various ratios (50:1 to 0.78:1) and incubated for 4 hours. The supernatants were harvested and radioactivity was measured in an automatic gamma counter. The percent specific lysis was determined as follows: specific lysis (%) = [(cpm experimental − cpm background)/(cpm maximum − cpm background)] × 100%. Background cpm was taken as spontaneous release from target cells in the absence of responder cells, and maximum cpm as release by target cells treated with 0.5% Nonidet P-40.

Statistical analysis

Statistical analysis of survival data was performed with the log-rank test. The Student t test was used to determine the level of significance of differences in group means. AP < .05 was considered to be significant in both types of analysis.

IFN-γ down-modulates CD8 T cell–mediated GVHD while mediating GVL effects of the CD8 subset

To determine the role of IFN-γ in GVHD- and GVL-associated alloresponses of allogeneic CD8 T cells, we compared these phenomena in lethally irradiated C57BL/6 (B6) mice receiving CD4-depleted spleen cells from WT or IFN-γ–deficient GKO BALB/c donors. B6 mice were lethally irradiated (9.75 Gy) and reconstituted with 5 × 106 TCD B6 BMCs (syngeneic controls) or with TCD BMCs (5 × 106) and CD4-depleted spleen cells (10 × 106) from WT or GKO BALB/c mice. Some recipients were injected with 500 EL4 cells (a B6 T-cell leukemia/lymphoma cell line) along with the BMT inoculum. It has been demonstrated that the GVL effect against EL4 cells is donor CD8+ cell dependent and CD4+ cell independent.20,28 Consistent with our previous finding that lethal acute GVHD in this strain combination is mostly CD4 dependent,22 most B6 mice injected with WT BALB/c CD4-depleted spleen cells survived long-term (Figure 1A). However, injection of a similar number of GKO BALB/c CD4-depleted spleen cells into B6 mice led to 60% mortality by 20 days (Figure 1A). Nonleukemic recipients of GKO BALB/c cells also showed more severe weight loss compared to nonleukemic recipients of WT BALB/c cells (Figure 1B). The body weight of most recipients of GKO BALB/c cells (4 of 5) decreased to less than 18 g by week 1 after BMT. However, only 1 of 5 mice that received WT BALB/c cells showed severe weight loss (< 18 g) by week 1 and the body weight of all surviving mice in this group recovered by week 2 after BMT. Because GVHD cannot be induced by donor spleen cells if both CD4 and CD8 T cells are depleted in this strain combination, these results indicate that donor-derived IFN-γ down-modulates systemic GVHD induced by allogeneic CD8 T cells.

Fig. 1.

Augmentation of GVHD is associated with reduction of GVL effects in B6 mice transplanted with CD4-depleted splenocytes from IFN-γ–deficient GKO BALB/c mice.

B6 mice were irradiated and transplanted with 5 × 106 B6 TCD BMC (▪, syngeneic), or 5 × 106 TCD BMC and 10 × 106 CD4-depleted splenocytes from WT (○, WT BALB) or GKO (●, GKO BALB) BALB/c donors. Leukemic recipients were also injected with 500 EL4 cells along with the BMT inoculum. Data shown are survival (A) and survivors' body weights (B) of nonleukemic recipients, and survival of leukemic recipients receiving 500 EL4 cells (C).

Fig. 1.

Augmentation of GVHD is associated with reduction of GVL effects in B6 mice transplanted with CD4-depleted splenocytes from IFN-γ–deficient GKO BALB/c mice.

B6 mice were irradiated and transplanted with 5 × 106 B6 TCD BMC (▪, syngeneic), or 5 × 106 TCD BMC and 10 × 106 CD4-depleted splenocytes from WT (○, WT BALB) or GKO (●, GKO BALB) BALB/c donors. Leukemic recipients were also injected with 500 EL4 cells along with the BMT inoculum. Data shown are survival (A) and survivors' body weights (B) of nonleukemic recipients, and survival of leukemic recipients receiving 500 EL4 cells (C).

Close modal

Remarkably, donor-derived IFN-γ had an opposite effect on the GVL reactivity of donor CD8 T cells. In contrast to the exacerbating effect on GVHD, the absence of donor-derived IFN-γ diminished GVL effects of donor CD8 T cells. Administration of CD4-depleted WT but not GKO BALB/c spleen cells led to significant antileukemic effects (Figure 1C;P < .005 for WT BALB/c recipients compared to recipients of GKO BALB/c cells and to syngeneic BMT recipients). Autopsy analysis was performed in randomly selected carcasses without knowledge of which treatment the animals had received.20 Gross evidence for tumor, which was detected in all EL4 recipients receiving transplants with syngeneic cells, was only found in 1 of 7 recipients of WT allogeneic BMCs plus CD4-depleted spleen cells (Experiment [Exp] 1 in Table 1). In contrast, evidence for tumor at autopsy was observed in 4 of 7 leukemic recipients of GKO allogeneic BMCs plus CD4-depleted spleen cells. Furthermore, no histologic evidence for tumor (ie, tissue infiltration of leukemic cells) was detected in the spleen, liver, or kidney of leukemic recipients of WT BALB/c cells that showed lack of tumor at autopsy (data not shown).

Table 1.

Gross evidence for tumor at autopsy in B6 recipients of TCD BMCs plus CD4-depleted spleen cells from WT or GKO BALB/c mice

GroupsTumor at autopsy
(no. with tumor/total evaluated)
Exp 1Exp 2
Syngeneic BMT + 500 EL4 4/4 4/4  
WT BALB/c BMT + 500 EL4 1/7 0/7  
GKO BALB/c BMT + 500 EL4 4/7 3/7 
Nonleukemic recipients 0/5 0/17 
GroupsTumor at autopsy
(no. with tumor/total evaluated)
Exp 1Exp 2
Syngeneic BMT + 500 EL4 4/4 4/4  
WT BALB/c BMT + 500 EL4 1/7 0/7  
GKO BALB/c BMT + 500 EL4 4/7 3/7 
Nonleukemic recipients 0/5 0/17 

Allogeneic BMT recipient mice were transplanted with 5 × 106 TCD BMCs and 10 × 106 (Exp 1) or 7.5 × 106 (Exp 2) CD4-depleted spleen cells from WT or GKO BALB/c mice. B6 mice receiving 5 × 106 TCD B6 BMCs were used as syngeneic BMT controls. Leukemic recipients were additionally injected with 500 EL4 cells. Nonleukemic controls (nonleukemic recipients) are pooled animals from both syngeneic and allogeneic BMT groups.

Similar results were observed in a repeat experiment. To limit the potential for GVHD-associated mortality to interfere with the evaluation of GVL effects, B6 recipients in this experiment were injected with a reduced number (7.5 × 106) of BALB/c CD4-depleted spleen cells. As shown in Figure2A, with the exception of one nonleukemic recipient in each of the WT and GKO allogeneic BMT groups, nonleukemic mice survived for the duration of the experiment. However, the survival advantage against EL4 leukemia conferred by CD4-depleted spleen cells from GKO BALB/c mice was significantly less than that mediated by WT BALB/c CD4-depleted spleen cells (Figure 2B; P < .05 for leukemic recipients of WT BALB/c cells compared to leukemic recipients of GKO BALB/c cells). All recipients of WT BALB/c cells were protected from leukemia-associated lethality, whereas all syngeneic recipients died of leukemia by day 32 after BMT (P < .001). Although leukemic recipients of GKO BALB/c cells were also significantly protected from lethality compared to syngeneic recipients (P < .05), long-term survival was only achieved in less than 50% of these mice. Evidence for tumor at autopsy was found in 3 of 4 leukemic recipients of GKO BALB/c cells that died by 40 days after BMT (Exp 2 in Table 1). No tumor was detected in long-term surviving leukemic recipients of either GKO (3 mice) or WT (7 mice) BALB/c cells. Together, our results indicate that donor-derived IFN-γ contributes to the GVL effect of allogeneic CD8 T cells, while down-modulating CD8 T cell–mediated GVHD.

Fig. 2.

IFN-γ is required for the induction of optimal GVL effects.

B6 mice were irradiated and transplanted with 5 × 106 B6 TCD BMC (▪, syngeneic), or 5 × 106 TCD BMC and 7.5 × 106 CD4-depleted splenocytes from WT (○,WT BALB) or GKO (●, GKO BALB) BALB/c donors. Leukemic recipients were additionally injected with 500 EL4 cells along with the BMT inoculum. Survivals of nonleukemic (A) and leukemic (B) recipients are shown.

Fig. 2.

IFN-γ is required for the induction of optimal GVL effects.

B6 mice were irradiated and transplanted with 5 × 106 B6 TCD BMC (▪, syngeneic), or 5 × 106 TCD BMC and 7.5 × 106 CD4-depleted splenocytes from WT (○,WT BALB) or GKO (●, GKO BALB) BALB/c donors. Leukemic recipients were additionally injected with 500 EL4 cells along with the BMT inoculum. Survivals of nonleukemic (A) and leukemic (B) recipients are shown.

Close modal

IFN-γ–deficient but not WT 2C T cells induce lethal acute GVHD in BALB/c mice

The 2C CTL clone was originally derived from an H-2bmouse immunized with H-2d cells, and the majority of T cells in 2C TCR transgenic mice are CD8+2CTCR+cells that specifically recognize Ld.24 Thus, transplantation of T cells from 2C TCR transgenic mice to H-2Ld+ (eg, BALB/c) mice provides a useful model to study GVH reactivity of CD8 T cells. We have previously shown that injection of 12 × 106 spleen cells along with BMCs from 2C TCR transgenic donors does not induce lethal acute GVHD in BALB/c mice,29 which is consistent with the finding that acute GVHD is largely CD4 dependent in most major histocompatibility complex–mismatched strain combinations in mice.28,30-32Remarkably, IFN-γ seems to be required for the absence of lethal GVHD in this 2C→BALB/c BMT model. As shown in Figure3, administration of 10 × 106 BMCs and 12 × 106 splenocytes from IFN-γ–deficient 2C donors caused severe acute GVHD, with 100% mortality by 20 days, whereas BALB/c mice receiving similar numbers of bone marrow and spleen cells from WT 2C mice survived long-term.

Fig. 3.

IFN-γ–deficient but not WT 2C T cells induce lethal acute GVHD in BALB/c mice.

Survival is shown for irradiated (8 Gy) BALB/c mice that were transplanted with 10 × 106 BMCs and 12 × 106 splenocytes from syngeneic donors (▪, syngeneic), WT (○, WT 2C) or GKO (●, GKO 2C) allogeneic 2C donors.

Fig. 3.

IFN-γ–deficient but not WT 2C T cells induce lethal acute GVHD in BALB/c mice.

Survival is shown for irradiated (8 Gy) BALB/c mice that were transplanted with 10 × 106 BMCs and 12 × 106 splenocytes from syngeneic donors (▪, syngeneic), WT (○, WT 2C) or GKO (●, GKO 2C) allogeneic 2C donors.

Close modal

IFN-γ production by allogeneic CD8 T cells directs their alloreactivity toward GVHD or GVL effects in vivo

Our previous studies have demonstrated that the GVL effects against EL4 leukemia are dependent on donor CD8 T cells.20,28 To determine whether or not IFN-γ produced by the effector CD8 T cells plays a role in dissociating GVL effects from systemic GVHD, we compared GVHD versus GVL effects in lethally irradiated B6 mice receiving transplants with purified CD8+T cells (1 × 106) from GKO or WT BALB/c mice. B6 TCD BMCs (5 × 106) and GKO BALB/c TCD BMCs (5 × 106) were given to all allogeneic BMT recipients and 500 EL4 cells were injected into some groups. Coadministration of TCD B6 BMCs into these lethally irradiated recipients of allogeneic cells provided a marker for the evaluation of lymphohematopoietic GVH reactions (see below). Although GVHD death was not observed (during an observation period of > 100 days) in the recipients of either WT or GKO BALB/c CD8 T cells (Figure 4A), significant loss of body weight was again observed in nonleukemic recipients of GKO BALB/c (19.5 ± 0.9 g), but not in nonleukemic recipients of WT BALB/c (20.7 ± 1.1 g) cells at week 1 after BMT (P < .05). The average body weight of nonleukemic recipients of syngeneic cells at week 1 after BMT was 20.7 ± 0.9 g. However, GVL effects correlated inversely with the degree of GVHD-related weight loss. Despite the more severe GVHD, as indicated by weight loss in nonleukemic recipients of GKO BALB/c CD8 cells, the survival of leukemic recipients of GKO BALB/c CD8 cells was not significantly extended compared to that of syngeneic controls (P = .17). In contrast, the survival of leukemic mice transplanted with WT BALB/c CD8 cells was significantly prolonged (Figure 4A; P < .005 compared to leukemic recipients of syngeneic BMT or GKO BALB/c cells).

Fig. 4.

Allogeneic CD8 T cells induce more severe systemic GVHD but less marked GVL effects if they are incapable of IFN-γ production.

Survival is shown for nonleukemic and leukemic B6 recipients of 5 × 106 TCD B6 BMCs (syngeneic), or 5 × 106 TCD B6 BMCs plus 5 × 106 TCD GKO BALB/c BMCs and 1 × 106 (A) or 2.5 × 106(B) WT BALB/c CD8+ splenocytes (WT BALB), or 5 × 106 TCD B6 BMCs plus 5 × 106 TCD GKO BALB/c BMCs and 1 × 106 (A) or 2.5 × 106(B) GKO BALB/c CD8+ splenocytes (GKO BALB). Leukemic recipients were injected with 500 EL4 cells along with the BMT inoculum. Survival data from 2 independent experiments (panels A and B) are shown and the numbers of mice in each group of the 2 experiments are indicated inside parentheses (a and b are mouse numbers of the indicated group in experiment A and experiment B, respectively).

Fig. 4.

Allogeneic CD8 T cells induce more severe systemic GVHD but less marked GVL effects if they are incapable of IFN-γ production.

Survival is shown for nonleukemic and leukemic B6 recipients of 5 × 106 TCD B6 BMCs (syngeneic), or 5 × 106 TCD B6 BMCs plus 5 × 106 TCD GKO BALB/c BMCs and 1 × 106 (A) or 2.5 × 106(B) WT BALB/c CD8+ splenocytes (WT BALB), or 5 × 106 TCD B6 BMCs plus 5 × 106 TCD GKO BALB/c BMCs and 1 × 106 (A) or 2.5 × 106(B) GKO BALB/c CD8+ splenocytes (GKO BALB). Leukemic recipients were injected with 500 EL4 cells along with the BMT inoculum. Survival data from 2 independent experiments (panels A and B) are shown and the numbers of mice in each group of the 2 experiments are indicated inside parentheses (a and b are mouse numbers of the indicated group in experiment A and experiment B, respectively).

Close modal

Consistent results were observed in a repeat experiment, in which allogeneic recipients were injected with a higher number (2.5 × 106) of WT or GKO BALB/c CD8 T cells. As shown in Figure 4B, 40% of nonleukemic recipients of GKO BALB/c CD8 cells died of GVHD, whereas all nonleukemic recipients of WT BALB/c CD8 cells survived long-term (> 100 days). In addition, the mean body weight of nonleukemic recipients of GKO BALB/c CD8 cells was significantly lower than that of nonleukemic recipients of WT BALB/c CD8 cells (19 ± 1.1 g versus 21 ± 1.2 g at week 1;P < .05). However, unlike GVHD, the potency of GKO BALB/c CD8 T cell–mediated GVL effects was not greater than that of WT BALB/c CD8 T cells (Figure 4B; P = .19). A significant delay in leukemic death was seen in leukemic recipients of both WT BALB/c (P < .0005) and GKO BALB/c (P < .005) CD8 cells compared to leukemic recipients of syngeneic BMT. Because most nonleukemic recipients (8 of 9) were surviving by the time when all leukemic recipients had died in recipients of GKO BALB/c CD8 cells, leukemia, but not GVHD, was the presumed cause of death in the leukemic group. Together, these results indicate that IFN-γ produced by donor CD8 T cells contributes to their GVL effects while mitigating their capacity to cause GVHD.

Discrepancy between systemic GVHD and antihost lymphohematopoietic alloreactivity in recipients of GKO BALB/c CD8 T cells

Previous studies in humans and animal models have shown that lymphohematopoietic GVH reactions that selectively eliminate host lymphohematopoietic cells, including lymphoma cells, can be induced without severe systemic GVHD in allogeneic BMT recipients.33-35 To determine whether or not the loss of GVL effects in recipients of GKO CD8 T cells was due to a reduced lymphohematopoietic GVH reaction, we compared donor CD8 T-cell expansion and levels of residual host hematopoietic cells in recipients of WT or GKO allogeneic BMT along with TCD host-type BMCs. Nonleukemic recipients of WT BALB/c or GKO BALB/c CD8 cells (these are the same mice shown in Figure 4A) were bled at weeks 5 and 9 after BMT, and the levels of donor CD8 T cells and surviving host cells in the recipient WBCs were determined by FACS analysis. Because these mice were injected with 5 × 106 TCD B6 BMCs along with allogeneic cells, lymphohematopoietic GVH reactions were assessed by measuring the preservation of host-type hematopoiesis. Consistent with the increased severity of systemic GVHD as shown by weight loss, the extent of GKO donor CD8 T-cell expansion was significantly greater than that of WT CD8 T cells in B6 recipients (Figure 5A). However, this greater expansion of GKO BALB/c CD8 T cells that was associated with significant loss of recipient body weight was associated with poor alloreactivity against host lymphohematopoietic cells. The levels of host (H-2Kb+) peripheral blood cells in mice receiving transplants with GKO BALB/c CD8 T cells were similar to and higher than those in the recipients of WT BALB/c CD8 T cells at weeks 5 and 9, respectively (Figure 5B). The difference was even more significant when comparing the levels of host-type cells in non–T-cell lineages (ie, when expanded donor T cells were excluded). As shown in Figure 5C, the percentages of host B cells (ie, percent of H-2Kb+CD19+ cells in the CD19+ cell population) in recipients of GKO BLAB/c CD8 T cells were markedly higher than that in recipients of WT CD8 T cells. These results indicate that allogeneic CD8 T cells induce more severe systemic GVHD but weaker antihost hematopoietic alloreactivity (and an associated reduction in GVL effects) if they are incapable of producing IFN-γ.

Fig. 5.

Discrepancy between donor T-cell expansion and antihost lymphohematopoietic alloreactivity in recipients of GKO BALB/c CD8 T cells.

WBCs were prepared from nonleukemic recipients of 5 × 106 TCD B6 BMCs plus 5 × 106 TCD GKO BALB/c BMCs and 1 × 106 purified CD8+splenocytes from WT (▪; n = 10) or GKO (■; n = 10) BALB/c mice at indicated times. Levels of donor CD8 T (5F1CD8+) cells (A) and total host type (5F1+, ie, H-2Kb+) cells (B) in the WBCs, and percentages of host B cells (5F1+CD19+) in the B-cell (CD19+) population (C) were determined by FACS. Data are presented as group means (±SD). These mice are the same nonleukemic WT BALB and GKO BALB recipients shown in Figure 4A. *P < .05, **P < .01, ***P < .0001.

Fig. 5.

Discrepancy between donor T-cell expansion and antihost lymphohematopoietic alloreactivity in recipients of GKO BALB/c CD8 T cells.

WBCs were prepared from nonleukemic recipients of 5 × 106 TCD B6 BMCs plus 5 × 106 TCD GKO BALB/c BMCs and 1 × 106 purified CD8+splenocytes from WT (▪; n = 10) or GKO (■; n = 10) BALB/c mice at indicated times. Levels of donor CD8 T (5F1CD8+) cells (A) and total host type (5F1+, ie, H-2Kb+) cells (B) in the WBCs, and percentages of host B cells (5F1+CD19+) in the B-cell (CD19+) population (C) were determined by FACS. Data are presented as group means (±SD). These mice are the same nonleukemic WT BALB and GKO BALB recipients shown in Figure 4A. *P < .05, **P < .01, ***P < .0001.

Close modal

CD4CD8 donor splenocytes are required for CD8 T cells to express maximal GVL effects

Although the GVL effect against EL4 in this model is donor CD8 T cell dependent, neither WT nor GKO CD8 T cells, when injected with GKO TCD BMCs, led to complete protection from leukemia induced by 500 EL4 cells (Figure 4), whereas complete protection was observed in recipients of this EL4 dose with CD4-depleted WT donor cells (Figures 1and 2). These results suggest that donor CD4CD8 spleen cells or IFN-γ–producing BMCs or both play a role in the anti-EL4 GVL effect, which has been shown to be donor CD8 T-cell dependent.20,28 To address this possibility, we compared GVL effects in B6 recipients of 500 EL4 cells and 5 × 106 TCD WT BALB/c BMCs along with different populations of WT BALB/c splenocytes: (1) 8.5 × 106 CD4-depleted spleen cells (with 16.5% CD8+ cells); (2) 1.4 × 106 CD8+splenocytes; (3) 7.1 × 106 TCD (ie, CD4 and CD8 cell-depleted) splenocytes; or (4) 1.4 × 106CD8+ and 7.1 × 106 TCD splenocytes. Syngeneic controls were injected with 5 × 106 TCD B6 BMCs and 500 EL4 cells. Consistent with previous studies20,28 and the results described above (Figures 1and 2), GVL effects against EL4 cells were completely abolished by depletion of CD4+ and CD8+ splenocytes (P = .1 for the recipients of TCD BALB/c splenocytes compared to syngeneic controls), whereas they were preserved if only CD4+ splenocytes were depleted (P < .0005 for the recipients of CD4-depleted BALB/c splenocytes compared to syngeneic controls; Figure 6). Although leukemic mortality was delayed in mice receiving TCD BALB/c BMCs plus purified CD8+ BALB/c splenocytes compared to syngeneic controls (P < .005), all of these mice eventually succumbed to leukemia (Figure 6). However, the GVL effects were fully restored by adding TCD (CD4CD8) splenocytes back to purified CD8+ splenocytes. The potency of GVL effects in the recipients of a combination of CD8+ and TCD BALB/c splenocytes was significantly greater than that in mice receiving CD8+ BALB/c splenocytes only (P < .005), and was indistinguishable from that in recipients of CD4-depleted BALB/c splenocytes (P = .6; Figure 6). Thus, donor CD4 CD8 splenocytes, which do not mediate GVL effects when injected alone, act synergistically with CD8 T cells to augment the antileukemic alloreactivity of CD8 T cells. Furthermore, these results confirm our previous results20,28 that donor CD4 cells do not contribute to GVL effects in this model.

Fig. 6.

CD4CD8 splenocytes act synergistically with CD8 T cells to augment GVL effects in vivo.

Survival is shown for mice that received 500 EL4 cells and 5 × 106 TCD WT BALB/c BMCs along with 8.5 × 106 CD4-depleted (●; n = 7), 1.4 × 106 CD8+ (♦; n = 7), 7.1 × 106 TCD (▴; n = 7), or 1.4 × 106 CD8+ and 7.1 × 106TCD (▾; n = 7) WT BALB/c splenocytes. Syngeneic controls receiving 5 × 106 TCD B6 BMCs (■; n = 5) or 5 × 106 B6 BMC plus 500 EL4 cells (▪; n = 5) are indicated. FACS analysis revealed that CD4-depleted splenocytes contained 16.5% CD8+ and 83.5% CD4CD8 cells, so the actual numbers of BALB/c CD8+ and CD4CD8splenocytes given to the recipients of 8.5 × 106CD4-depleted BALB/c splenocytes were 1.4 × 106 and 7.1 × 106, respectively.

Fig. 6.

CD4CD8 splenocytes act synergistically with CD8 T cells to augment GVL effects in vivo.

Survival is shown for mice that received 500 EL4 cells and 5 × 106 TCD WT BALB/c BMCs along with 8.5 × 106 CD4-depleted (●; n = 7), 1.4 × 106 CD8+ (♦; n = 7), 7.1 × 106 TCD (▴; n = 7), or 1.4 × 106 CD8+ and 7.1 × 106TCD (▾; n = 7) WT BALB/c splenocytes. Syngeneic controls receiving 5 × 106 TCD B6 BMCs (■; n = 5) or 5 × 106 B6 BMC plus 500 EL4 cells (▪; n = 5) are indicated. FACS analysis revealed that CD4-depleted splenocytes contained 16.5% CD8+ and 83.5% CD4CD8 cells, so the actual numbers of BALB/c CD8+ and CD4CD8splenocytes given to the recipients of 8.5 × 106CD4-depleted BALB/c splenocytes were 1.4 × 106 and 7.1 × 106, respectively.

Close modal

IFN-γ does not directly inhibit the proliferation of EL4 cells

Interferon-γ has been shown to mediate antitumor effects by directly inhibiting tumor cell growth and inducing T cell–mediated antitumor responses.36-41 To determine whether the reduced GVL effect in leukemic recipients of GKO allogeneic cells is due to the loss of direct inhibition of EL4 cell proliferation by donor-derived IFN-γ, we measured the susceptibility of EL4 cells to an IFN-γ–mediated antiproliferative effect. EL4 cells were incubated with varying concentrations of IFN-γ for 48 hours and cell proliferation was assessed by tritiated thymidine incorporation. Despite the expression of IFN-γ receptor on their surface (Figure7A), the proliferation of EL4 cells was not significantly inhibited by IFN-γ, while IFN-γ efficiently inhibited the growth of WEHI-279, an IFN-γ–susceptible murine lymphoma cell line (Figure 7B). Moreover, no suppression of EL4 proliferation was mediated by supernatants of BALB/c–anti-B6 mixed lymphocyte reactions, suggesting that cytokines released by alloreactive T cells are incapable of directly suppressing the growth of EL4 cells (Figure 7C).

Fig. 7.

EL4 cells are not susceptible to an IFN-γ–mediated antiproliferative effect.

(A) A FACS profile showing expression of IFN-γ receptor on EL4 cells. EL4 cells stained with anti-CD119 (IFN-γ receptor α chain) and isotype control mAb are presented as filled and open histograms, respectively. Results from one representative experiment of 3 are shown. (B) Proliferation of EL4 and WEHI-279 cells cultured in medium containing various concentrations of IFN-γ. Data are presented as the mean ± SD (cpm) of triplicate cultures in each culture condition. Results from one representative experiment of 3 are shown. (C) Lack of inhibitory activity on the proliferation of EL4 cells in allogeneic mixed lymphocyte reaction supernatants. EL4 cells were cultured inside tissue culture inserts placed in 24-well plates containing responders (BALB/c splenocytes) and stimulators (irradiated B6 splenocytes) (▪) or stimulators only (■) (see “Materials and methods”). Three wells from each group were harvested at each time point and the number of viable EL4 cells in each well was counted. Data are presented as the mean ± SD (cell number/well) of triplicate samples.

Fig. 7.

EL4 cells are not susceptible to an IFN-γ–mediated antiproliferative effect.

(A) A FACS profile showing expression of IFN-γ receptor on EL4 cells. EL4 cells stained with anti-CD119 (IFN-γ receptor α chain) and isotype control mAb are presented as filled and open histograms, respectively. Results from one representative experiment of 3 are shown. (B) Proliferation of EL4 and WEHI-279 cells cultured in medium containing various concentrations of IFN-γ. Data are presented as the mean ± SD (cpm) of triplicate cultures in each culture condition. Results from one representative experiment of 3 are shown. (C) Lack of inhibitory activity on the proliferation of EL4 cells in allogeneic mixed lymphocyte reaction supernatants. EL4 cells were cultured inside tissue culture inserts placed in 24-well plates containing responders (BALB/c splenocytes) and stimulators (irradiated B6 splenocytes) (▪) or stimulators only (■) (see “Materials and methods”). Three wells from each group were harvested at each time point and the number of viable EL4 cells in each well was counted. Data are presented as the mean ± SD (cell number/well) of triplicate samples.

Close modal

IFN-γ up-regulates expression of Fas and major histocompatibility class I on EL4 cells and moderately increases the susceptibility of EL4 cells to the cytotoxicity of allogeneic CD8 T cells

Interferon-γ has been shown to augment the sensitivity of tumor cells to cytolytic T lymphocyte (CTL) activity by up-regulating surface expression of Fas and major histocompatibility complex (MHC) on tumor cells.42,43 To determine whether or not IFN-γ affects the expression of Fas and class I MHC on EL4 cells, we have analyzed the cell surface expression of these molecules on EL4 cells treated with IFN-γ in comparison with untreated EL4 cells. EL4 cells were incubated with IFN-γ at various concentrations (7 different concentrations from 0.78 to 50 ng/mL) for 12 to 14 hours. Both Fas and MHC class I expression were up-regulated on EL4 cells treated with IFN-γ at all concentrations compared to control EL4 cells incubated in IFN-γ–free medium (Figure 8A and data not shown). The peak expression for both molecules was observed on EL4 cells treated with IFN-γ in a concentration range of 6.25 to 12.5 ng/mL (Figure 8A). CTL assays revealed that EL4 cells are highly sensitive to the killing activity of both WT and GKO BALB/c CTLs (Figure 8B). Indeed, the levels of Fas and MHC class I expression were high even on untreated EL4 cells (Figure 8A). Consistently, the susceptibility of EL4 cells to alloreactive CTLs was only slightly increased by pretreatment with IFN-γ (Figure 8B).

Fig. 8.

Effect of IFN-γ on expression of Fas and MHC class I on EL4 cells and susceptibility of EL4 cells to the cytotoxicity of allogeneic CD8 T cells.

(A) IFN-γ up-regulates Fas and MHC class I expression on EL4 cells. Surface expression of Fas and MHC class I on EL4 cells cultured in the absence (dotted histograms) or the presence of IFN-γ (6.25 ng/mL; solid histograms) is shown. (B) Killing of EL4 cells by alloreactive CTLs. Spleen cells from WT or GKO BALB/c mice were stimulated with irradiated B6 splenocytes for 5 days, and the killing activity against untreated EL4 (○) or EL4 cells that were cultured with IFN-γ (6.25 or 12.5 ng/mL for 12-14 hours) (●) was measured by a 51Cr-release assay. Data are presented as the average (percent specific lysis) of triplicates. Results from one representative experiment of at least 3 independent experiments are shown.

Fig. 8.

Effect of IFN-γ on expression of Fas and MHC class I on EL4 cells and susceptibility of EL4 cells to the cytotoxicity of allogeneic CD8 T cells.

(A) IFN-γ up-regulates Fas and MHC class I expression on EL4 cells. Surface expression of Fas and MHC class I on EL4 cells cultured in the absence (dotted histograms) or the presence of IFN-γ (6.25 ng/mL; solid histograms) is shown. (B) Killing of EL4 cells by alloreactive CTLs. Spleen cells from WT or GKO BALB/c mice were stimulated with irradiated B6 splenocytes for 5 days, and the killing activity against untreated EL4 (○) or EL4 cells that were cultured with IFN-γ (6.25 or 12.5 ng/mL for 12-14 hours) (●) was measured by a 51Cr-release assay. Data are presented as the average (percent specific lysis) of triplicates. Results from one representative experiment of at least 3 independent experiments are shown.

Close modal

The data presented here demonstrate that IFN-γ produced by donor cells controls the alloresponses of donor CD8 T cells and determines whether they will mediate predominantly GVHD or lymphohematopoietic GVH reactions with associated GVL effects. Alloresponses mediated by GKO CD4-depleted (or purified CD8+) BALB/c splenocytes resulted in more severe GVHD (weight loss and mortality), but in reduced GVL effects in B6 recipients, compared to a similar cell population from WT BALB/c mice. In association with the increased severity of GVHD, the expansion of GKO donor CD8 T cells in vivo was significantly greater than that of WT donor CD8 T cells. However, the increased donor CD8 T-cell expansion was paradoxically associated with a reduction of lymphohematopoietic GVH reactions, which is able to mediate GVL effects without GVHD. It has been observed both in man and in animal models that a conversion from mixed chimerism to fully allogeneic donor chimerism can occur without clinical GVHD, demonstrating that lymphohematopoietic GVH reactions can be selectively preserved in allogeneic BMT recipients while the capacity to mediate tissue GVHD is suppressed.33-35 Such GVH reactions directed against host lymphohematopoietic cells can eliminate host leukemic cells and lead to long-term remissions in recipients of allogeneic BMT who have lymphomas.35,78 Results from the present study indicate that IFN-γ production by donor cells plays an important role in the induction of CD8 T cell–mediated lymphohematopoietic GVH reactions. Importantly, GVHD-inducing alloresponses of CD8 T cells display a differential response to IFN-γ, such that IFN-γ acts as an inhibitory cytokine.

The mechanism for the discrepancy between augmented systemic GVHD (identified as weight loss and mortality) and reduced lymphohematopoietic GVH reactions in the recipients of GKO BALB/c cells remains to be defined. It has been proposed that Th1 cytokines are critical for inducing acute GVHD,44 and a number of studies have shown that IFN-γ produced by activated alloreactive T cells plays an important role in the induction of acute GVHD.45-48 In contrast, both exogenously injected and endogenously produced IFN-γ has been shown to paradoxically inhibit GVHD,49-52 and lethal acute GVHD can be induced in the complete absence of IFN-γ.22 IFN-γ has been reported to play an important role in regulating the death of activated CD4 T cells.53-59 Consistently, Fas expression on donor T cells is required for IL-12–mediated inhibition of CD4-mediated GVHD, an IFN-γ–dependent effect.22,60 In the 2C BMT model, we have previously shown that WT 2C CD8 cells expanded greatly (about 15-fold) in BALB/c recipients at early times after transplantation. This expansion was followed by a rapid decline in 2C CD8 cell numbers (7- to 15-fold).29 Importantly, apoptosis was evident in a significant proportion of host antigen–activated 2C CD8 T cells in allogeneic transplant recipients, and the timing of the increase in apoptosis of 2C donor CD8 T cells coincided with the decrease in the numbers of 2C CD8 T cells in lethally irradiated BALB/c recipients of 2C splenocytes.29 It has been recently demonstrated that IFN-γ also contributes to the death phase of activated CD8 T cells.61,62 Thus, it is possible that the increased expansion of donor CD8 T cells and associated augmentation of GVHD in recipients of IFN-γ–deficient donor cells was due to decreased death of alloreactive CD8 T cells.

However, reduced death of alloreactive donor CD8 T cells alone cannot explain the opposing effects of IFN-γ on the 2 types of alloreactivity (ie, GVHD-inducing activity and lymphohematopoietic GVH response/GVL effects) mediated by CD8 T cells. Thus, other mechanisms must be involved. These may include effects of IFN-γ on both the GVL effector cells and the leukemic cells. Results of our ex vivo studies indicate that IFN-γ does not directly suppress the proliferation of EL4 cells (Figure 7), but that this cytokine might slightly increase the sensitivity of EL4 cells to the cytolytic activity of alloreactive CD8 T cells through up-regulation of MHC class I and Fas molecules (Figure 8). However, this small increase in sensitivity of EL4 cells to CTL-mediated killing seems insufficient to explain the observed differences in GVL effects of WT versus GKO CD8 T cells. Differences in the homing of alloreactive donor T cells in the presence and the absence of IFN-γ production might also contribute to the different outcomes observed for GKO and WT allogeneic BMT recipients. IFN-γ plays an important role in regulating chemokine production and thereby directing the tissue infiltration of activated, including alloantigen-primed, T cells.63-67 Studies using an immunogenic tumor model demonstrated that the failure of cytolytic effectors (“tumor-antigen”–specific CD8 T cells) to remain at the site of the tumor is a major limitation in the ability of CD8 T-cell responses to control tumor growth.68 Contact-dependent lysis is also critical for alloreactive CTLs to mediate GVL effects in allogeneic BMT recipients. It has been reported that IFN-γ contributes to alloreactive donor T-cell infiltrates in lymphoid tissues and lymphoid hypoplasia associated with GVHD,69,70suggesting that IFN-γ may direct alloresponses toward the lymphohematopoietic system rather than the parenchymal GVHD target tissues. Consistently, the present study showed that GKO donor CD8 T cells that induce more severe GVHD were less efficient in destroying host hematopoietic cells compared to WT donor CD8 T cells (Figure 5). Thus, it is possible that the reduction of GVL effects in mice receiving IFN-γ–deficient donor cells reflects a lack of sufficient contact between donor CD8 T cells and the leukemic cells within the lymphohematopoietic system in these mice, and that the increased GVHD is due to increased T-cell migration into the parenchymal GVHD target tissues.

Although the differentiation of cytotoxic CD8 T cells has been shown to require help from CD4 cells,71,72 the generation of alloreactive CD8 T cells can also be independent of CD4 help.73,74 The striking increase in GVHD mortality in recipients of GKO CD8 T cells or 2C cells demonstrates that this helper-independent CD8 subset is regulated by cell-autonomous IFN-γ production. Our previous studies have shown that the GVL effects of splenic T cells against EL4 can be completely eliminated by depleting CD8+ splenocytes, but are not affected by depleting CD4+ cells, indicating that such effects are CD8 dependent and CD4 independent.20,28 We have now observed that CD4CD8 splenocytes are required for CD8+ cells to mediate an optimal GVL effect. It has recently been shown that the CD4-independent induction of cytotoxic CD8 T cells against allogeneic tumor cells is dependent on costimulation and can be inhibited by blocking the interaction of either CD40/CD40L or B7/CD28 between antigen-presenting cells (APCs) and CD8 T cells.74 However, it is unlikely that the synergistic effect of CD4CD8 splenocytes observed in the present study was mediated by donor APCs included in the CD4CD8 cell population, because donor BMCs that also contain APCs had no such effect (Figures 4 and 6). It is possible that efficient GVL effects may require the presence of both large quantities of IFN-γ and CD8 T cells, and the synergistic effects of CD4CD8 splenocytes may reflect their capacity to produce IFN-γ. It has been reported that both NK and NKT cells are potential IFN-γ producers and play an important role in regulating alloresponses of T cells.75-77 

The present study demonstrated that allogeneic CD8 T cells lacking the capacity for IFN-γ production induce more severe GVHD, but less potent antilymphohematopoietic GVH reactions and antileukemic effects than WT CD8 T cells. The findings suggest that global suppression of IFN-γ production should be avoided in the development of strategies for controlling GVHD in leukemic patients.

We thank Drs Markus Mapara and Yong-mi Kim for critical reading of the manuscript and Sharon Titus for her expert secretarial assistance.

Supported by National Institutes of Health grant RO1 CA79989, American Cancer Society grant IRG-87-007-13, and American Society for Blood and Marrow Transplantation/Orphan Medical New Investigator Award.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Beatty
PG
Clift
RA
Mickelson
FM
et al
Marrow transplantation from related donors other than HLA-identical siblings.
N Engl J Med.
313
1985
765
771
2
Bortin
MM
Bone marrow transplantation for leukemia using family donors other than HLA-identical siblings: a preliminary report from the International Bone Marrow Transplant Registry.
Transplant Proc.
19
1987
2629
2631
3
Beatty
PG
Anasetti
C
Hansen
JA
et al
Marrow transplantation from unrelated donors for treatment of hematologic malignancies: effect of mismatching for one HLA locus.
Blood.
81
1993
249
253
4
Sierra
J
Storer
B
Hansen
JA
et al
Transplantation marrow cells from unrelated donor for treatment of high-risk acute leukemia: the effect of leukemic burden, donor HLA-matching, and marrow cell dose.
Blood.
89
1997
4226
4235
5
Verdonck
LF
Dekker
AW
Lokhorst
HM
Petersen
EJ
Nieuwenhuis
HK
Allogeneic versus autologous bone marrow transplantation for refractory and recurrent low-grade Non-hodgkin's lymphoma.
Blood.
90
1997
4201
4205
6
Gale
RP
Graft-versus-host disease.
Immunol Rev.
88
1985
193
214
7
Ringden
O
Nilsson
B
Death by graft-versus-host disease associated with HLA mismatch, high recipient age, low marrow cell dose, and splenectomy.
Transplantation.
40
1985
39
44
8
Lokhorst
HM
Schattenberg
A
Cornelissen
JJ
Thomas
LLM
Verdonck
LF
Donor leukocyte infusions are effective in relapsed multiple myeloma after allogeneic bone marrow transplantation.
Blood.
90
1997
4206
4211
9
Uharek
L
Glass
B
Gaska
T
et al
Natural killer cells as effector cells of graft-versus-leukemia activity in a murine transplantation model.
Bone Marrow Transplant.
12
1993
S57
60
10
Zeis
M
Uharek
L
Glass
B
et al
Allogeneic NK cells as potent antileukemic effector cells after allogeneic bone marrow transplantation in mice.
Transplantation.
59
1995
1734
1736
11
Asai
O
Longo
DL
Tian
RL
Taub
DD
Ruscetti
FW
Murphy
WJ
Suppression of graft-versus-host disease and amplification of graft-versus-tumor effects by activated natural killer cells after allogeneic bone marrow transplantation.
J Clin Invest.
101
1998
1835
1842
12
Papadopoulos
EB
Carabasi
MH
Castro-Malaspina
H
et al
T-cell-depleted allogeneic bone marrow transplantation as postremission therapy for acute myelogenous leukemia: freedom from relapse in the absence of graft-versus-host disease.
Blood.
91
1998
1083
1090
13
Aversa
F
Tabilio
A
Velardi
A
et al
Treatment of high risk leukemia with T cell depleted stem cells in patients from related donors with one fully matched haplotype.
N Engl J Med.
339
1998
1186
1193
14
Martin
PJ
Hansen
JA
Torok-Storb
B
et al
Graft failure in patients receiving T cell-depleted HLA-identical allogeneic marrow transplants.
Bone Marrow Transplant.
3
1988
445
456
15
Poynton
CH
T cell depletion in bone marrow transplantation.
Bone Marrow Transplant.
3
1988
265
279
16
Butturini
A
Gale
RP
T cell depletion in bone marrow transplantation for leukemia: current results and future directions.
Bone Marrow Transplant.
3
1988
185
192
17
Slavin
S
Ackerstein
A
Naparstek
E
Or
R
Weiss
L
The graft-versus-leukemia (GVL) phenomenon: is GVL separable from GVHD?
Bone Marrow Transplant.
6
1990
155
161
18
Sykes
M
Novel approaches to the control of GVHD.
Curr Opin Immunol.
5
1993
774
781
19
Uharek
L
Glass
B
Zeis
M
et al
Abrogation of graft-vs.-leukemia activity after depletion of CD3+ T cells in a murine model of MHC-matched peripheral blood progenitor cells.
Exp Hematol.
26
1998
93
99
20
Yang
YG
Sergio
JJ
Pearson
DA
Szot
GL
Shimizu
A
Sykes
M
Interleukin-12 preserves the graft-vs-leukemia effect of allogeneic CD8 T cells while inhibiting CD4-dependent graft-vs-host disease in mice.
Blood.
90
1997
4651
4660
21
Yang
YG
Dey
B
Sergio
JJ
Sykes
M
IL-12 prevents severe acute GVHD and GVHD-associated immune dysfunction in a full MHC haplotype-mismatched murine bone marrow transplantation model.
Transplantation.
64
1997
1343
1352
22
Yang
YG
Dey
B
Sergio
JJ
Pearson
DA
Sykes
M
Donor-derived interferon γ is required for inhibition of acute graft-versus-host disease by interleukin 12.
J Clin Invest.
102
1998
2126
2135
23
Sha
WC
Nelson
CA
Newberry
RD
Kranz
DM
Russell
RD
Loh
DY
Selective expression of an antigen receptor on CD8-bearing T lymphocytes in transgenic mice.
Nature.
335
1988
271
274
24
Kranz
DM
Sherman
DH
Sitkowsky
MV
Pasternack
MS
Eisen
HN
Immunoprecipitation of cell surface structures of cloned cytotoxic T lymphocytes by clone-specific antisera.
Proc Natl Acad Sci U S A.
81
1984
573
577
25
Sykes
M
Romick
ML
Hoyles
KA
Sachs
DH
In vivo administration of interleukin 2 plus T cell-depleted syngeneic marrow prevents graft-versus-host disease mortality and permits alloengraftment.
J Exp Med.
171
1990
645
658
26
Sykes
M
Bukhari
Z
Sachs
DH
Graft-versus-leukemia effect using mixed allogeneic bone marrow transplantation.
Bone Marrow Transplant.
4
1989
465
474
27
Unkeless
JC
Characterization of a monoclonal antibody directed against mouse macrophage and lymphocyte Fc receptors.
J Exp Med.
150
1979
580
596
28
Sykes
M
Abraham
VS
Harty
MW
Pearson
DA
IL-2 reduces graft-vs-host disease and preserves a graft-vs-leukemia effect by selectively inhibiting CD4+ T cell activity.
J Immunol.
150
1993
197
205
29
Dey
B
Yang
YG
Preffer
F
et al
The fate of donor T-cell receptor transgenic T cells with known host antigen specificity in a graft-versus-host disease model.
Transplantation.
68
1999
141
149
30
Korngold
R
Sprent
J
Surface markers of T cells causing lethal graft-vs-host disease to class I vs class II H-2 differences.
J Immunol.
135
1985
3004
3010
31
Vallera
DA
Soderling
CCB
Kersey
JH
Bone marrow transplantation across major histocompatibility barriers in mice, III: treatment of donor grafts with monoclonal antibodies directed against Lyt determinants.
J Immunol.
128
1982
871
875
32
Palathumpat
V
Dejbachsh-Jones
S
Strober
S
The role of purified CD8+ T cells in graft-versus-leukemia activity and engraftment after allogeneic bone marrow transplantation.
Transplantation.
60
1995
355
361
33
Sykes
M
Sheard
MA
Sachs
DH
Graft-versus-host-related immunosuppression is induced in mixed chimeras by alloresponses against either host or donor lymphohematopoietic cells.
J Exp Med.
168
1988
2391
2396
34
Pelot
MR
Pearson
DA
Swenson
K
et al
Lymphohematopoietic graft-vs-host reactions can be induced without graft-vs-host disease in murine mixed chimeras established with a cyclophosphamide-based non-myeloablative conditioning regimen.
Biol Blood Marrow Transplant.
5
1999
133
143
35
Spitzer
TR
McAfee
S
Sackstein
R
et al
Intentional induction of mixed chimerism and achievement of antitumor responses after nonmyeloablative conditioning therapy and HLA-matched donor bone marrow transplantation for refractory hematologic malignancies [in process citation].
Biol Blood Marrow Transplant.
6
2000
309
320
36
Nastala
CL
Edington
HD
McKinney
TG
et al
Recombinant IL-12 administration induces tumor regression in association with IFN-γ production.
J Immunol.
153
1994
1697
1706
37
Brunda
MJ
Sulich
V
Bellantoni
D
The anti-tumor effect of recombinant interferon alpha and gamma is influenced by tumor location.
Int J Cancer.
40
1987
807
810
38
Sayers
TJ
Wiltrout
TA
McCormick
K
Wiltrout
RH
Antitumor effects of alpha-interferon and gamma-interferon on a murine renal cancer (Renca) in vitro and in vivo.
Cancer Res.
50
1990
5414
5420
39
Brunda
MJ
Luistro
L
Hendrzak
JA
Fountoulakis
M
Garotta
G
Gately
MK
Role of interferon-γ in mediating the antitumor efficacy of interleukin-12.
J Immunother.
17
1995
71
77
40
Tannenbaum
CS
Wicker
N
Armstrong
D
et al
Cytokine and chemokine expression in tumors of mice receiving systemic therapy with IL-12.
J Immunol.
156
1996
693
699
41
Tan
J
Crucian
BE
Chang
AE
et al
Interferon-gamma-inducing factor elicits antitumor immunity in association with interferon-gamma production.
J Immunother.
21
1998
48
55
42
Bohm
W
Thoma
S
Leithauser
F
Moller
P
Schirmbeck
R
Reimann
J
T cell-mediated, IFN-γ-facilitated rejection of murine B16 melanomas.
J Immunol.
161
1998
897
908
43
Sayers
TJ
Brooks
AD
Lee
J-K
et al
Molecular mechanisms of immune-mediated lysis of murine renal cancer: differential contributions of perforin-dependent versus Fas-mediated pathways in lysis by NK and T cells.
J Immunol.
161
1998
3957
3965
44
Ferrara
JLM
Levy
R
Chao
NJ
Pathophysiologic mechanisms of acute graft-vs.-host disease.
Biol Blood Marrow Transplant.
5
1999
347
356
45
Allen
RD
Staley
TA
Sidman
CL
Differential cytokine expression in acute and chronic murine graft-versus-host disease.
Eur J Immunol.
23
1993
333
337
46
Guy-Grand
D
Vassalli
P
Gut injury in mouse graft-vs-host reaction: study of its occurrence and mechanism.
J Clin Invest.
77
1986
1584
1595
47
Mowat
AM
Antibodies to IFN-gamma prevent immunologically mediated intestinal damage in murine graft-versus-host reaction.
Immunology.
68
1989
18
23
48
Ellison
CA
Fischer
JMM
HayGlass
KT
Gartner
JG
Murine graft-versus-host disease in an F1-hybrid model using IFN-γ gene knockout donors.
J Immunol.
161
1998
631
640
49
Brok
HP
Heidt
PJ
Van der Meide
PH
Zurcher
C
Vossen
JM
Interferon-gamma prevents graft-versus-host disease after allogeneic bone marrow transplantation in mice.
J Immunol.
151
1993
6451
6459
50
Murphy
WJ
Welniak
LA
Taub
DD
et al
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
1998
1742
1748
51
Baker
J
Verneris
MR
Ito
M
Shizuru
JA
Negrin
RS
Expansion of cytolytic CD8(+) natural killer T cells with limited capacity for graft-versus-host disease induction due to interferon gamma production.
Blood.
97
2001
2923
2931
52
Welniak
LA
Blazar
BR
Anver
MR
Wiltrout
RH
Murphy
WJ
Opposing roles of interferon-gamma on CD4+ T cell-mediated graft-versus-host disease: effects of conditioning.
Biol Blood Marrow Transplant.
6
2000
604
612
53
Liu
Y
Janeway
CA
Interferon-gamma plays a critical role in induced cell death of effector T cell: a possible third mechanism of self-tolerance.
J Exp Med.
172
1990
1735
1739
54
Novelli
F
D'Elios
MM
Bernabei
P
et al
Expression and role in apoptosis of the alpha- and beta-chains of the IFN-gamma receptor on human Th1 and Th2 clones.
J Immunol.
159
1997
206
213
55
Willenborg
DO
Fordham
S
Bernard
CC
Cowden
WB
Ramshaw
IA
IFN-gamma plays a critical down-regulatory role in the induction and effector phase of myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis.
J Immunol.
157
1996
3223
3227
56
Willenborg
DO
Fordham
SA
Staykova
MA
Ramshaw
IA
Cowden
WB
IFN-gamma is critical to the control of murine autoimmune encephalomyelitis and regulates both in the periphery and in the target tissue: a possible role for nitric oxide.
J Immunol.
163
1999
5278
5286
57
Tarrant
TK
Silver
PB
Wahlsten
JL
et al
Interleukin 12 protects from a T helper type 1-mediated autoimmune disease, experimental autoimmune uveitis, through a mechanism involving interferon-γ, nitric oxide, and apoptosis.
J Exp Med.
189
1999
219
230
58
Chu
CQ
Wittmer
S
Dalton
DK
Failure to suppress the expansion of the activated CD4 T cell population in interferon gamma-deficient mice leads to exacerbation of experimental autoimmune encephalomyelitis.
J Exp Med.
192
2000
123
128
59
Dalton
DK
Haynes
L
Chu
CQ
Swain
SL
Wittmer
S
Interferon gamma eliminates responding CD4 T cells during mycobacterial infection by inducing apoptosis of activated CD4 T cells.
J Exp Med.
192
2000
117
122
60
Dey
B
Yang
YG
Szot
GL
Pearson
DA
Sykes
M
IL-12 inhibits GVHD through a Fas-mediated mechanism associated with alterations in donor T cell activation and expansion.
Blood.
91
1998
3315
3322
61
Badovinac
VP
Tvinnereim
AR
Harty
JT
Regulation of antigen-specific CD8(+) T cell homeostasis by perforin and interferon-gamma.
Science.
290
2000
1354
1358
62
Badovinac
VP
Harty
JT
Adaptive immunity and enhanced CD8+ T cell response to Listeria monocytogenes in the absence of perforin and IFN-gamma.
J Immunol.
164
2000
6444
6452
63
Liao
F
Rabin
RL
Yannelli
JR
Koniaris
LG
Vanguri
P
Farber
JM
Human Mig chemokine: biochemical and functional characterization.
J Exp Med.
182
1995
1301
1314
64
Luster
AD
Weinshank
RL
Feinman
R
Ravetch
JV
Molecular and biochemical characterization of a novel gamma-interferon-inducible protein.
J Biol Chem.
263
1988
12036
12043
65
Luster
AD
Ravetch
JV
Biochemical characterization of a gamma interferon-inducible cytokine (IP-10).
J Exp Med.
166
1987
1084
1097
66
Tran
EH
Prince
EN
Owens
T
IFN-γ shapes immune invasion of the central nervous system via regulation of chemokines.
J Immunol.
164
2000
2759
2768
67
Koga
S
Auerbach
MB
Engeman
TM
Novick
AC
Toma
H
Fairchild
RL
T cell infiltration into class II MHC-disparate allografts and acute rejection is dependent on the IFN-γ-induced chemokine Mig.
J Immunol.
163
1999
4878
4885
68
Shrikant
P
Mescher
MF
Control of syngeneic tumor growth by activation of CD8+ T cells: efficacy is limited by migration away from the site and induction of nonresponsiveness.
J Immunol.
162
1999
2858
2866
69
Klimpel
GR
Annable
CR
Cleveland
MG
Jerrells
TR
Patterson
JC
Immunosuppression and lymphoid hypoplasia associated with chronic graft versus host disease is dependent upon IFN-gamma production.
J Immunol.
144
1990
84
93
70
Parfrey
NA
El Sheikh
A
Monckton
EA
Cockfield
SM
Halloran
PF
Linetsky
E
Interferon-gamma gene expression during acute graft-versus-host disease: relationship to MHC induction and tissue injury.
J Pathol.
189
1999
99
104
71
Bennett
SRM
Carbone
FR
Karamalis
F
Flavell
RA
Miller
JFAP
Heath
WR
Help for cytotoxic- T-cell responses is mediated by CD40 signalling.
Nature.
393
1998
478
480
72
Schoenberger
SP
Toes
REM
van der Voort
EIH
Offringa
R
Melief
CJM
T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions.
Nature.
393
1998
480
483
73
Manning
TC
Rund
LA
Gruber
MM
Fallarino
F
Gajewski
TF
Kranz
DM
Antigen recognition and allogeneic tumor rejection in CD8+ TCR transgenic/RAG(−/−) mice.
J Immunol.
159
1997
4665
4675
74
Zhang
Y
Corbett
AJ
Brady
JL
Sutherland
RM
Lew
AM
CD4 help-independent induction of cytotoxic CD8 cells to allogeneic P815 tumor cells is absolutely dependent on costimulation.
J Immunol.
165
2000
3612
3619
75
Zeng
D
Lewis
D
Dejbachsh-Jones
S
et al
Bone marrow NK1.1- and NK1.1+ T cells reciprocally regulate acute graft versus host disease.
J Exp Med.
189
1999
1073
1081
76
Raziuddin
A
Longo
DL
Mason
L
Ortaldo
JR
Bennett
M
Murphy
WJ
Differential effects of the rejection of bone marrow allografts by the depletion of activating versus inhibiting Ly-49 natural killer cell subsets.
J Immunol.
160
1998
87
94
77
Seino
K
Fukao
K
Muramoto
K
et al
Requirement for natural killer T (NKT) cells in the induction of allograft tolerance.
Proc Natl Acad Sci U S A.
98
2001
2577
2581
78
Mapara MY, Kim Y-M, Wang S-P, Bronson R, Sachs DH, Sykes M. Donor lymphocyte infusions (DLI) mediate superior graft-versus-leukemia (GvL) effects in mixed compared to fully allogeneic chimeras: a critical role for host antigen-presenting cells. Blood. In press.

Author notes

Yong-Guang Yang, Bone Marrow Transplantation Section, Transplantation Biology Research Center, Massachusetts General Hospital, MGH East, Bldg 149-5102, 13th St, Boston, MA 02129; e-mail:yongguang.yang@tbrc.mgh.harvard.edu.

Sign in via your Institution