Minimization of graft-versus-host disease (GVHD) with preservation of the graft-versus-leukemia (GVL) effect is a crucial step to improve the overall survival of allogeneic bone marrow transplantation (BMT) for patients with hematological malignancies. We and other investigators have shown that granulocyte colony-stimulating factor (G-CSF)–mobilized allogeneic peripheral stem cell transplantation (PBSCT) reduces the severity of acute GVHD in murine models. In this study, we investigated whether G-CSF–mobilized PBSC maintain their GVL effect in a murine allogeneic transplant model (B6 → B6D2F1). B6 mice (H-2b) were injected subcutaneously with human G-CSF (100 μg/kg/d) for 6 days and their splenocytes were harvested on day 7 as a source of PBSC. G-CSF mobilization dramatically improved transplant survival compared with nonmobilized controls (95% v0%, P < .001). Systemic levels of lipopolysaccharide and tumor necrosis factor- were markedly reduced in recipients of allogeneic G-CSF–mobilized donors, but cytolytic T lymphocyte (CTL) activity against host tumor target cells p815 was retained in those recipients. When leukemia was induced in recipients by coinjection of p815 tumor cells (H-2d) at the time of transplantation, all surviving recipients of G-CSF–mobilized B6 donors were leukemia-free at day 70 after transplant, whereas all mice who received T-cell–depleted (TCD) splenocytes from G-CSF–mobilized B6 donors died of leukemia. When splenocytes from G-CSF–mobilized perforin-deficient (pfp−/−) mice were used for transplantation, 90% of recipients died of leukemia, demonstrating that perforin is a crucial pathway mediating GVL effects after G-CSF–mobilized PBSCT. These data illustrate that G-CSF–mobilized allogeneic PBSCT separate GVL from GVHD by preserving perforin-dependent donor CTL activity while reducing systemic inflammation.

ALLOGENEIC BONE MARROW transplantation (BMT) is a standard therapy for hematological malignancies. An important benefit of allogeneic BMT is the graft-versus-leukemia (GVL) effect, a process of tumor eradication by donor cells after BMT.1-3 However, GVL effects are closely linked with graft-versus-host disease (GVHD), a major cause of morbidity and mortality after allogeneic BMT.1,4 Results from a series of clinical trials demonstrated that donor T cells play a vital role in both GVL and GVHD, because T-cell depletion (TCD) of the bone marrow reduced the incidence and severity of GVHD, but increased leukemia relapse.2,5-7 It is also well recognized that leukemia relapse is inversely linked to the severity of GVHD after BMT.1 5 Therefore, separation of GVL and GVHD is a crucial step to improve the overall survival of allogeneic BMT for hematologic malignancy.

Recently, there is increased enthusiasm for the use of granulocyte colony-stimulating factor (G-CSF)–mobilized peripheral blood stem cell transplantation (PBSCT). Comparison of G-CSF–mobilized PBSCT (containing a 10- to 20-fold increase in donor CD3+ cells) and traditional bone marrow grafts demonstrate a surprisingly similar incidence and severity of acute GVHD.8-11 This relative reduction of acute GVHD may be attributable to immunomodulation of cells in the donor graft. A decrease in interleukin-2 (IL-2) and interferon-γ (IFN-γ) production to allo-antigen stimulation has been reported both in human and animal studies.12-15Monocytes from G-CSF–mobilized human donors have also been reported to suppress allo-reactivity of T cells in mixed lymphocyte culture,16-18 perhaps through an IL-10–dependent mechanism.19 However, recent studies suggest increased risks of chronic GVHD after G-CSF–mobilized allogeneic PBSCT.20 21 Because donor T cells are major effectors of the GVL effect, it is important to investigate whether G-CSF–mobilized PBSC grafts can maintain GVL effects. In this study of a murine PBSCT-leukemia model, we show that T cells from G-CSF–mobilized PBSC have a markedly diminished capacity to induce acute GVHD, but maintain their GVL function through a perforin-dependent pathway.

Mice.

Female Ly-5 congenic B6.Ly-5a (H-2b, CD45.1+) mice were obtained from the Frederick Cancer Research Facility (Frederick, MD), and female B6D2F1(H-2bxd, CD45.2+) mice were purchased from the Jackson Laboratories (Bar Harbor, ME). Female C57BL/6 (B6, H-2b, CD45.2+) and perforin-deficient mice (pfp−/−, B6x129/SvEv, H-2b, CD45.2+) were purchased from Toconic Laboratory (Germantown, NY). Ly-5 (CD45) alleles are described according to the nomenclature of Morse et al.22 Mice were housed in sterilized microisolator cages and received tap water and normal chow. Mice used for experiments were between the ages of 10 and 14 weeks and received autoclaved hyper-chlorinated drinking water during the first 3 weeks posttransplantation.

G-CSF treatment.

Donor mice were injected subcutaneously with recombinant human G-CSF (Amgen Inc, Thousand Oaks, CA) daily at 100 μg/kg body weight or saline (control diluent) for 6 days, and splenocytes were harvested on day 7.

PBSCT.

This protocol has been described previously.13 14 Briefly, B6D2F1 recipients received 1,100 rad total body irradiation, which was split into two doses separated by 3 hours to minimize gastrointestinal (GI) toxicity. Splenocytes (10 × 106) from B6 donors were injected intravenously into B6D2F1 recipients. Recipients of 5 × 106 TCD B6 splenocytes (treated with 2 cycles of anti-Thy1.2 and rabbit complement) or 10 × 106 B6D2F1 splenocytes served as non-GVHD controls. For GVL experiments, 5,000 to 25,000 p815 leukemic cells (H-2d, CD45.2+) were injected together with donor splenocytes. Survival was monitored daily and recipient body weight was measured weekly. Tumor burden was determined either by detection of tumor cells in peripheral blood or at autopsy at the end of experiments. The criteria for tumor-induced death were defined as either hepatosplenomegaly with macroscopic tumor nodules in liver and/or spleen or evidence of spinal cord involvement (hind leg paralysis or pathological demonstration of p815 tumor cells in the spinal cord). Leukemia-free survival was defined as (1) ≤0.5% tumor cells (H-2d+/b− + CD45.2+)/CD45.1 in peripheral blood and (2) no macroscopic tumor nodules in liver, spleen, and spinal cord at the end of experiments.

Mixed lymphocyte culture.

Splenic T cells were obtained by passage of splenocytes through nylon wool columns and were cultured with 1 × 105irradiated B6D2F1 peritoneal cells (2,000 rad) in completed Dulbecco’s modified Eagle’s medium (DMEM) media in a 96-well flat-bottomed plate at 37°C in a humidified incubator supplemented with 7% CO2. All culture media reagents were purchased from GIBCO BRL (Gaithersburg, MD). Completed DMEM media was supplemented with 10% fetal calf serum, 50 U/mL penicillin, 50 μg/mL streptomycin, 2 mmol/L L-glutamine, 1 mmol/L sodium pyruvate, 0.1 mmol/L nonessential amino acid, 0.02 mmol/L β-mercaptoethanol, and 10 mmol/L HEPES, pH 7.75. At 48 hours, supernatants were collected for cytokine levels. For cytokine determination during 2° MLR, splenic T cells were incubated with irradiated (2,000 rad) B6D2F1 splenocytes at a 1:2 ratio in a 24-well plate for 6 days and then restimulated with fresh irradiated (2,000 rad) B6D2F1 peritoneal cells in completed DMEM media in a 96-well flat-bottom plate for 48 hours.

Fluorescence-activated cell sorting (FACS) analysis.

Fluorescein isothiocyanate (FITC)- or R-phycoerythrin (PE)–conjugated monoclonal antibodies (MoAbs) were purchased from PharMingen (San Diego, CA). Cells (5 × 105/sample) were first incubated with MoAb 2.4G2 for 10 minutes at 4°C to block nonspecific binding to Fc receptors and then with FITC- or PE-conjugated specific MoAbs for 30 minutes at 4°C. Cells were then washed twice with phosphate-buffered saline (PBS)/0.2% bovine serum albumin (BSA) and fixed with PBS/1% paraformaldehyde. Two-color flow cytometric analysis was performed using a FACScan (Becton Dickson, Mountain View, CA). Two methods of staining were used to determining the tumor burden in peripheral blood. Cells were either double-stained with FITC-conjugated anti–H-2Dd (Cedarlane Lab, Hornby, Ontario, Canada) and PE-conjugated H-2Kb or with FITC-conjugated anti-CD45.1 and anti-CD45.2 MoAb. In control experiments, Peripheral blood cells (PBC) from donor B6 Ly-5a mice were 99.8% CD45.1+ and H-2b+/d− and PBC of B6D2F1 were 99.8% CD45.2+ and H-2b+/d+, whereas p815 cells were 99.7% CD45.2+ and H-2Kb−/H-2Dd+ (data not shown).

Enzyme-linked immunosorbent assay (ELISA).

The antibodies used in the assays in the IFN-γ, IL-2, IL-4, IL-10, and IL-12 p40 assays were purchased from PharMingen (San Diego, CA), and antibodies used in the tumor necrosis factor-α (TNF-α) assay were purchased from Genzyme Corp (Cambridge, MA). All assays were performed according to the manufacturer’s protocol. Briefly, cytokines were captured by the specific primary MoAb and detected by horseradish peroxidase-labeled anti–TNF-α or by the biotin-labeled anti–IFN-γ, anti–IL-2, anti–IL-4, anti–IL-10, or anti–IL-12 followed by strepavidin-horseradish peroxidase. The color reaction was developed by TMB microwell peroxidase substrate (KPL, Gaithersburg, MD) and stopped by the addition of an equal volume of 1 mol/L H2SO4. The absorbance of the assay plate was read at 450 nm using a microplate reader (Model 3550; Bio-Rad Labs, Hercules, CA). Recombinant murine TNF-α (mTNF-α), mIFN-γ, mIL-2, mIL-4, mIL-10, and mIL-12 p40 were used as standards for ELISAs. The low limit of sensitivity is 0.1 U/mL for IFN-γ, IL-2, and IL-4; 15 pg/mL for IL-10 and TNF-α; and 1 pg/mL for IL-12 p40.

Limulus amebocyte lysate (LAL) assay.

The serum endotoxin levels were determined by the LAL assay using the QCL-1000 test kit (BioWhittaker, Walkersville, MD). Assays were performed according to the manufacturer’s protocol. Briefly, serum was diluted 10-fold with LAL reagent water and heated to 70°C for 5 minutes to remove any nonspecific inhibition to the assay. Samples were then incubated with equal volumes of LAL for 10 minutes at 37°C and developed with equal volumes of substrate solution for 6 minutes. The absorbance of the assay plate was read at 405 nm using a microplate reader (Model 3550; Bio-Rad Labs). Samples and standards were run in duplicate and the lower limit of detection was 0.15 U/mL. All units expressed are relative to the US reference standard EC-6.

51Cr release assay.

Responder cells from day-6 primary MLR or fresh splenocytes harvested on day 7 posttransplant were used as effector cells. Two million target cells were labeled with 100 μCi 51Cr for 2 hours at 37°C and washed 3 times afterwards. Effector cells were incubated with 10,000 labeled target cells at 37°C for 4 hours at various effector/target ratios, and 51Cr in supernatant was determined by a γ-scintillation counter. p815 cells (H-2d) were used as allogeneic tumor targets; EL-4 cells (H-2b) were used as targets syngeneic to the donor. Maximal and background release was determined by addition to the target cells of 2% Triton-X (Sigma Chemical Co, St Louis, MO) or media, respectively. The percentage of specific lysis (%) = 100 × (sample count − background count)/(maximal count − background count).

Statistical analysis.

The Mann-Whitney U test was used for the statistical analysis of weight loss, whereas the Mantel-Cox log rank-test was used to analyzed survival data. The two-tailed Student’s t-test was used to analyze cytokine and lipopolysaccharide (LPS) data. P = .05 was considered statistically significant.

G-CSF mobilization reduces the severity of acute GVHD.

We first examined the effects of G-CSF mobilization in a murine BMT model (B6 Ly-5a → B6D2F1) that induces acute GVHD to both major and minor histocompatibility antigens. Injections of human G-CSF for 6 days at a dose of 100 μg/kg/d increased the yield of splenocytes by approximately 25%. As shown in Fig 1, the percentages of T cells (CD4+, CD8+), B cells (B220+), and natural killer (NK) cells (NK1.1+) were similar in control and G-CSF–reated donors, whereas myeloid cells (Gr-1+) were significantly increased in splenocytes from G-CSF–treated donors. The percentage of myeloid cells in the bone marrow doubled in G-CSF–treated donors. B6D2F1 recipient mice were irradiated with 1100 cGy and transplanted with 10 × 106 splenocytes from either control or G-CSF–treated B6 Ly-5a mice. GVHD induced in this model was severe and usually lethal. As shown in Fig 2A, all animals receiving control allogeneic splenocytes died within 2 weeks, with clinical evidence of GVHD (hunched posture, inactivity, and weight loss), whereas 95% of mice receiving splenocytes from G-CSF–mobilized donors survived at day 70 posttransplant. This survival was markedly superior to that seen in our previous study when splenocytes were mobilized with a lower dose of human G-CSF.13,14 The optimal dose of hG-CSF for PBSC mobilization is approximately 10 times higher in mice than in humans,15 23-25 making the doses used in this study more clinically relevant. The mortality of acute GVHD seen in control allogeneic recipients was mediated by donor T cells, because all mice receiving allogeneic TCD-splenocytes survived until the end of experiment. Although the severity of acute GVHD in mice receiving allogeneic G-CSF–mobilized splenocytes was dramatically reduced, these mice did show signs of moderate GVHD, as measured by weight loss compared with recipients of allogeneic TCD-splenocytes or syngeneic splenocytes (Fig 2B).

Fig. 1.

Effect of G-CSF on granulocyte population and T-cell phenotype. B6 Ly-5a mice were injected with G-CSF (100 μg/kg/d) or saline for 6 days. BM (n = 15 per group) and splenocytes (n = 20 per group) were harvested the day after the last injection. After lysis of red blood cells, cells were stained with specific Abs and analyzed by FACS. The results represent the mean ± SD from nine experiments. *P < .001 v control mice.

Fig. 1.

Effect of G-CSF on granulocyte population and T-cell phenotype. B6 Ly-5a mice were injected with G-CSF (100 μg/kg/d) or saline for 6 days. BM (n = 15 per group) and splenocytes (n = 20 per group) were harvested the day after the last injection. After lysis of red blood cells, cells were stained with specific Abs and analyzed by FACS. The results represent the mean ± SD from nine experiments. *P < .001 v control mice.

Close modal
Fig. 2.

Survival and weight loss after splenocyte transplant (B6 Ly-5a → B6D2F1). B6 Ly-5a donors were injected with or without G-CSF for 6 days. Total body irradiated B6D2F1 recipients received 1 × 107 splenocytes from control B6 donors (n = 20), G-CSF–mobilized donors (n = 20), or control B6D2F1 donors (n = 15) or 5 × 106 TCD-splenocytes from control B6 donors (n = 10). Survival was monitored daily up to day 70 posttransplantation (A). Body weights were measured weekly (B). *P < .001 v recipients of splenocytes from control B6 donors (A). *P < .001 v recipients of TCD-splenocytes from control B6 donors or splenocytes from control B6D2F1 donors (B).

Fig. 2.

Survival and weight loss after splenocyte transplant (B6 Ly-5a → B6D2F1). B6 Ly-5a donors were injected with or without G-CSF for 6 days. Total body irradiated B6D2F1 recipients received 1 × 107 splenocytes from control B6 donors (n = 20), G-CSF–mobilized donors (n = 20), or control B6D2F1 donors (n = 15) or 5 × 106 TCD-splenocytes from control B6 donors (n = 10). Survival was monitored daily up to day 70 posttransplantation (A). Body weights were measured weekly (B). *P < .001 v recipients of splenocytes from control B6 donors (A). *P < .001 v recipients of TCD-splenocytes from control B6 donors or splenocytes from control B6D2F1 donors (B).

Close modal
G-CSF mobilization reduces systemic levels of LPS and TNF-α.

Both LPS and TNF-α are known to be important mediators of acute GVHD severity.26-28 Consistent with the severe clinical GVHD in animals receiving control allogeneic splenocytes, the serum LPS levels in these animals were markedly elevated compared with syngeneic controls on day 7 posttransplant, a time of maximal elevation (Fig 3A). By contrast, serum LPS levels in animals transplanted with G-CSF–mobilized allogeneic splenocytes were reduced to levels of syngeneic non-GVHD controls (Fig 3A). Serum TNF-α levels were also significantly reduced in the recipients of G-CSF–mobilized donors, although they remained higher than that seen in syngeneic non-GVHD controls (Fig 3B).

Fig. 3.

Serum levels of LPS and TNF- after splenocyte transplant. Total body irradiated B6D2F1 mice received 1 × 107 splenocytes from control B6 donors, G-CSF–mobilized donors, or control B6D2F1 donors (n = 5/group). Serum was collected on day 7 posttransplant, LPS levels were determined by LAL assay (A), and the TNF- level was determined by ELISA (B). UD, under limit of detection. *P < .001 v recipients of control B6 donors.

Fig. 3.

Serum levels of LPS and TNF- after splenocyte transplant. Total body irradiated B6D2F1 mice received 1 × 107 splenocytes from control B6 donors, G-CSF–mobilized donors, or control B6D2F1 donors (n = 5/group). Serum was collected on day 7 posttransplant, LPS levels were determined by LAL assay (A), and the TNF- level was determined by ELISA (B). UD, under limit of detection. *P < .001 v recipients of control B6 donors.

Close modal
G-CSF mobilization induces a type 2 cytokine profile with preservation of CTL activity.

We next examined the effects of G-CSF mobilization on donor T-cell functions. Consistent with previous reports using a lower G-CSF dose,13 14 G-CSF treatment led to an increased production of type 2 cytokines (IL-4 and IL-10) with a decreased production of type 1 cytokines (IL-2 and IFN-γ) in response to host antigen stimulation. This polarization towards a type 2 cytokine profile was maintained in 2° MLR despite the absence of exogenous G-CSF at all times in culture (Table 1). G-CSF also decreased production of IL-12, reflecting its action on the antigen-presenting cells (Table 1). T cells from 1° MLR were then used as effector cells against host type (H-2d) p815 targets or donor type (H-2b) EL4 targets. As shown in Fig 4, G-CSF mobilization did not reduce the CTL activity of splenocytes, despite the shift in cytokine profile.

Fig. 4.

CTL activity in vitro. CTL activity was determined by51Cr release assay. Equal numbers of splenic T cells from a 6-day primary MLR (B6 anti-B6D2F1) were used as effector cells. p815 cells (H-2d) and EL4 cells (H-2b) were labeled with 51Cr and used as targets. After 4 hours of coincubation with effector cells, 51Cr in the supernatants was determined by a γ-scintillation counter. One of three representative experiments is presented.

Fig. 4.

CTL activity in vitro. CTL activity was determined by51Cr release assay. Equal numbers of splenic T cells from a 6-day primary MLR (B6 anti-B6D2F1) were used as effector cells. p815 cells (H-2d) and EL4 cells (H-2b) were labeled with 51Cr and used as targets. After 4 hours of coincubation with effector cells, 51Cr in the supernatants was determined by a γ-scintillation counter. One of three representative experiments is presented.

Close modal
G-CSF mobilization preserves GVL effects.

To examine the effects of G-CSF mobilization on GVL, animals were transplanted as described above and 5,000 p815 tumor cells were injected intravenously together with the donor inoculum. As shown in Fig 5A, syngeneic recipients all died of leukemia by 4 weeks posttransplant with macroscopic evidence of tumor in the liver and spleen. Recipients of allogeneic control donors died within 2 weeks due to severe GVHD, but necropsy showed no evidence of tumor. In contrast, 95% of allogeneic recipients of G-CSF–mobilized donor cells were still alive at day 70 posttransplantation. Eradication of leukemia was confirmed by absence of CD45.2+ cells in peripheral blood and lack of tumor in liver and spleen by histology. The importance of donor T cells in mediating the GVL effect was confirmed by transplantation of TCD-splenocytes from B6 donors and 5,000 p815 tumor cells. None of recipients showed evidence of GVHD (Fig2), but they all died by 5 weeks after transplantation with macroscopic evidence of leukemia (Fig 5B).

Fig. 5.

Survival after leukemia induction (B6 Ly-5a→ B6D2F1). B6 Ly-5a donors were injected with or without G-CSF for 6 days. (A) Total body irradiated B6D2F1 mice received 1 × 107 splenocytes plus 5,000 p815 tumor cells from control B6 donors (n = 15), G-CSF–mobilized donors (n = 20), or control B6D2F1 donors (n = 13). *P < .001 vrecipients of splenocytes from control B6 donors and control B6D2F1 donors. (B) Total body irradiated B6D2F1 recipients were injected with 5 × 106 TCD-splenocytes plus 5,000 p815 tumor cells from control or G-CSF B6 donors (n = 8/group). Survival was monitored daily up to day 70 posttransplantation.

Fig. 5.

Survival after leukemia induction (B6 Ly-5a→ B6D2F1). B6 Ly-5a donors were injected with or without G-CSF for 6 days. (A) Total body irradiated B6D2F1 mice received 1 × 107 splenocytes plus 5,000 p815 tumor cells from control B6 donors (n = 15), G-CSF–mobilized donors (n = 20), or control B6D2F1 donors (n = 13). *P < .001 vrecipients of splenocytes from control B6 donors and control B6D2F1 donors. (B) Total body irradiated B6D2F1 recipients were injected with 5 × 106 TCD-splenocytes plus 5,000 p815 tumor cells from control or G-CSF B6 donors (n = 8/group). Survival was monitored daily up to day 70 posttransplantation.

Close modal
GVL is mediated through a perforin-dependent pathway.

To further delineate the mechanism of GVL after G-CSF–mobilized PBSCT, perforin-deficient (pfp−/−) mice were used as donors, because perforin has been shown to be an important effector of Tc2 cytotoxic functions.29 30 CTL activity from pfp−/− was substantially reduced both in vitro and ex vivo (Fig 6). Splenic T cells after 1° MLR (Fig 6A) or splenocytes from recipients 7 days after transplantation of 10 × 106 allogeneic splenocytes (Fig 6B) showed significant decrease in lysis of host-type p815 tumor targets, and this was unaffected by G-CSF mobilization (Fig 6A and B). We then examined GVL effects in a murine PSCT-leukemia model. Lethal irradiated B6D2F1 recipient mice received 10 × 106splenocytes from G-CSF–mobilized B6 or pfp−/−donors with 25,000 p815 tumor cells. As shown in Fig 7, all syngeneic recipients died with macroscopic evidence of tumor within 3 weeks. All recipients of allogeneic G-CSF donors were leukemia-free at day 70 as determining by FACS staining of peripheral blood cells and macroscopic examination of tumor-targeted organs. By contrast, 90% of recipients transplanted with splenocytes from pfp−/− donors died with gross evidence of leukemia. Engraftment of pfp−/− donor cells was complete by day 60 and expansion of pfp−/− donor T cells on day 7 after transplantation was equivalent to wild-type controls (Table 2). Therefore, the loss of a GVL effect was not due to diminished donor T-cell expansion or engraftment, but rather lack of perforin activity.

Fig. 6.

Abolition of CTL activity in pfp−/− mice. CTL activity was determined by 51Cr release assay. (A) Equal numbers of splenic T cells from a 6-day primary MLR (B6 anti-B6D2F1) were used as effectors. (B) Splenocytes from day 7 posttransplant (n = 5/group) were counted, and equal numbers of T cells (CD4+ plus CD8+ cells adjusted according to FACS analysis) were used as effectors.51Cr-labeled p815 targets (H-2d) were coincubated with effectors for 4 hours, and 51Cr in the supernatants was determined by a γ-scintillation counter.

Fig. 6.

Abolition of CTL activity in pfp−/− mice. CTL activity was determined by 51Cr release assay. (A) Equal numbers of splenic T cells from a 6-day primary MLR (B6 anti-B6D2F1) were used as effectors. (B) Splenocytes from day 7 posttransplant (n = 5/group) were counted, and equal numbers of T cells (CD4+ plus CD8+ cells adjusted according to FACS analysis) were used as effectors.51Cr-labeled p815 targets (H-2d) were coincubated with effectors for 4 hours, and 51Cr in the supernatants was determined by a γ-scintillation counter.

Close modal
Fig. 7.

Survival after leukemia induction (B6 → B6D2F1). Wild-type B6 or pfp−/− donors were injected with G-CSF for 6 days. Total body irradiated B6D2F1 recipients received 1 × 107 splenocytes plus 25,000 p815 tumor cells from G-CSF–mobilized B6 donors (n = 10) or from G-CSF–mobilized pfp−/− donors (n = 10) or control B6D2F1 donors (n = 5). Survival was monitored daily until day 70 posttransplantation. *P < .001 v recipients of splenocytes from G-CSF–mobilized pfp−/− donors and control B6D2F1 donors.

Fig. 7.

Survival after leukemia induction (B6 → B6D2F1). Wild-type B6 or pfp−/− donors were injected with G-CSF for 6 days. Total body irradiated B6D2F1 recipients received 1 × 107 splenocytes plus 25,000 p815 tumor cells from G-CSF–mobilized B6 donors (n = 10) or from G-CSF–mobilized pfp−/− donors (n = 10) or control B6D2F1 donors (n = 5). Survival was monitored daily until day 70 posttransplantation. *P < .001 v recipients of splenocytes from G-CSF–mobilized pfp−/− donors and control B6D2F1 donors.

Close modal

In this study, we demonstrate that G-CSF–mobilized allogeneic PBSCT dramatically reduced the severity of acute GVHD while maintaining perforin-dependent GVL effects in a murine PBSCT-leukemia model. CTL activity against host antigens in G-CSF–mobilized donor PBSCT is preserved, although the inflammatory cytokine response is significantly diminished.

The important balance between cytokines derived from type 1 and type 2 T cells in inducing acute GVHD was first demonstrated in the experimental BMT models.31-33 Elevated levels of type 1 cytokines (IL-12, IL-2, and IFN-γ) are associated with severe acute GVHD,34-39 whereas elevated levels of a type 2 cytokine profile (increased IL-4 and IL-10 production) are not.31-33,40,41 A correlation of type 1 and type 2 cytokine profile with the severity of acute GVHD was also reported by Tanaka et al42 in a clinical study of allogeneic BMT. We and other investigators have reported that G-CSF mobilization skews T-cell cytokines toward a type 2 profile upon allo-antigen stimulation and after experimental allogeneic PBSCT.12-15 G-CSF mobilization also causes a decreased production of type 1 cytokines from PBMC upon allo-antigen stimulation compared with before mobilization,43,44 and increased expression of IL-4 mRNA has also been reported.45 

Type 1 cytokines are known to prime mononuclear cells to secrete TNF-α during GVHD.31,33,46,47 Clinical studies have shown that elevated serum TNF-α levels precede clinical symptoms of acute GVHD,28,48 and anti–TNF-α therapy significantly reduced the severity of acute GVHD.49,50 It has also been reported that G-CSF–mobilized human PBMC produced less TNF-α in mixed lymphocyte cultures than PBMC from same donor pretreated with G-CSF.17 In this study, serum TNF-α levels were significantly reduced in recipients of G-CSF–mobilized splenocytes compared with GVHD controls. TNF-α has been shown to cause necrosis in the GI tract during GVHD.51 The endotoxin that translocates across damaged intestinal mucosa acts as a stimulus to further TNF-α production and may also amplify target organ damage by enhancing the in vivo clonal expansion and differentiation of antigen-activated T cells, as shown in other experimental systems.52 

The reduction in severe GVHD using high-dose G-CSF mobilization in this study was markedly superior to that of previous studies in which a 10-fold lower dose of G-CSF was used.13,14 T-cell responses toward a type 2 cytokine profile were similar after both high-dose and low-dose mobilization, suggesting that improved protection from high-dose mobilization was not due to changes in T-cell cytokine secretion. However, a reduction in proliferation to host antigens was observed using enriched splenic T cells from donors mobilized with high-dose G-CSF (data not shown). In addition, the percentage of myeloid cells in splenocytes after high-dose G-CSF mobilization was significantly greater (Fig 1) than that seen after low-dose G-CSF mobilization.13 These observations are consistent with studies of G-CSF–mobilized human PBSC that show myeloid components may play a role in hypo-responsiveness of donor T cells to allo-antigen stimulation.16-18 53 

Donor T cells play a vital role in mediating GVL effects, as demonstrated by the effectiveness of donor leukocyte infusion to induce remission after leukemia relapse.54-58 In this study, we have showed that G-CSF–mobilized donor T cells maintain their CTL activity against leukemic targets and preserve GVL effects. An improved GVL effect using G-CSF–mobilized allogeneic PBSCT has been reported in another murine leukemia model.59 G-CSF–mobilized PBPC have also been used successfully to treat relapse after allogeneic BMT.60,61 Apoptosis of target cells induced by CTL could be mediated by perforin and/or Fas/FasL pathways, and both pathways may be involved in the development of GVHD.50,62-69 CTL can be divided into Tc1 and Tc2 subpopulations according to their cytokine secretion pattern. Apoptosis mediated by Tc1 cells depends primarily on Fas/Fas ligand pathway.29,70,71 IFN-γ secreted by type 1 T cells has been reported to increase expression of Fas and FasL and may thereby enhance apoptosis mediated by the Fas/FasL pathway.72,73 However, apoptosis mediated by Tc2 cells is more dependent on perforin pathway.29 71 Such mechanisms are consistent with the present study, in which G-CSF mobilization amplifies a Tc2 response, reduces acute GVHD, and maintains GVL through a perforin-dependent pathway.

In this study, we demonstrated that G-CSF–mobilized grafts reduce severity of acute GVHD by disruption of cytokine cascade involved in development of acute GVHD. More importantly, G-CSF–mobilized grafts maintain their GVL effects through a perforin-dependent pathway. Therefore, G-CSF mobilization may offers a novel approach to the separation of GVL effects from GVHD. Studies are currently in progress to determine the effects of G-CSF mobilization of donor cells in chronic GVHD and immune reconstitution.

The authors thank Dr Anastasia Skandalis for her valuable discussions and Scott Bressler and Vicki Mosher for their technical support.

Supported in part by National Institutes of Health Grants No. CA 39542 and HL 55709.

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

1
Truitt
RL
Johnson
BD
McCabe
C
Weiler
MB
Graft versus leukemia
Graft Versus Host Disease: Immunology, Pathophysiology, and Treatment.
Ferrara
JLM
Deeg
HJ
Burakoff
SJ
1997
385
Marcel Dekker
New York, NY
2
Weiden
PL
Horowitz
MM
Graft-vs-leukemia effect in clinical bone marrow transplantation
Graft-versus-Host Disease.
Burakoff
SJ
Deeg
HJ
Ferrara
JLM
Atkinson
K
1990
691
Marcel Dekker
New York, NY
3
Barrett
J
Malkovska
V
The graft-versus-leukemia effect.
Curr Opin Oncol
8
1996
89
4
Champlin
R
Giralt
S
Gajewski
J
T cells, graft-versus-host disease and graft-versus-leukemia: Innovative approaches for blood and marrow transplantation.
Acta Haematol
95
1996
157
5
Apperley
JF
Jones
L
Hale
G
Goldman
JM
Bone marrow transplantation for chronic myeloid leukemia: T cell depletion with Campath-1 reduces the incidence of acute graft-versus-host disease but may increase the risk of leukemia relapse.
Bone Marrow Transplant
1
1986
53
6
Prentice
HG
Blacklock
HA
Janossy
G
Gilmore
MJ
Price-Jones
L
Tidman
N
Trejdosiewicz
LK
Skeggs
DB
Panjwani
D
Ball
S
Depletion of T-lymphocytes in donor marrow prevents significant graft-versus-host disease in matched allogeneic leukemic marrow transplant recipients.
Lancet
1
1984
472
7
Horowitz
MM
Gale
RP
Sondel
PM
Goldman
JM
Kersey
J
Kolb
HJ
Rimm
AA
Ringden
O
Rozman
C
Speck
B
Truitt
RL
Zwaan
FE
Bortin
MM
Graft-verus-leukemia reactions after bone marrow transplantation.
Blood
75
1990
555
8
Bensinger
WI
Clift
R
Martin
P
Appelbaum
FR
Demirer
T
Gooley
T
Lilleby
K
Rowley
S
Sanders
J
Storb
R
Buckner
CD
Allogeneic peripheral blood stem cell transplantation in patients with advanced hematologic malignancies: A retrospective comparison with marrow transplantation.
Blood
88
1996
2794
9
Gratwohl
A
Hermans
J
Baldomero
H
Blood and marrow transplantation activity in Europe 1995.
Bone Marrow Transplant
19
1997
407
10
Harada
M
Shinagawa
K
Kawano
T
Kasai
M
Sawada
H
Nakao
S
Hyodo
H
Aotsuka
N
Furukawa
T
Hirai
H
Eto
T
Imai
Y
Shimazaki
C
Matsue
K
Ogawa
M
Takaku
F
Allogeneic peripheral blood stem cell transplantation for standard-risk leukemia. A multicenter pilot study: Japanese experience. Japan Blood Cell Transplantation Study Group.
Bone Marrow Transplant
21
1998
S54
(suppl 3)
11
Cornelissen
JJ
Fibbe
WE
Schattenberg
AV
Petersen
EJ
Willemze
R
de Witte
TJ
Lowenberg
B
Verdonck
LF
vd Biezen
A
Brand
R
A retrospective Dutch study comparing T cell-depleted allogeneic blood stem cell transplantation vs T cell-depleted allogeneic bone marrow transplantation.
Bone Marrow Transplant
21
1998
S66
(suppl 3)
12
Hartung
T
Docke
WD
Grabtner
F
Krieger
G
Sauer
A
Stevens
P
Volk
HD
Wendel
A
Effect of granulocyte colony-stimulating factor treatment on ex vivo blood cytokine response in human volunteers.
Blood
85
1995
2482
13
Pan
L
Delmonte
J
Jalonen
CK
Ferrara
JLM
Pretreatment of donors with granulocyte colony-stimulating factor polarizes donor T lymphocytes toward type 2 cytokine production and reduces severity of experimental graft versus host disease.
Blood
86
1995
4422
14
Pan
L
Bressler
S
Cooke
KR
Krenger
W
Karandikar
M
Ferrara
JLM
Long-term engraftment, graft-versus-host disease, and immunologic reconstitution following experimental transplantation of allogeneic peripheral blood cells from G-CSF treated donors.
Biol Blood Marrow Transplant
2
1996
126
15
Zeng
D
Dejbakhsh-Jones
S
Strober
S
Granulocyte colony-stimulating factor reduces the capacity of blood mononuclear cells to induce graft-versus-host disease: Impact on blood progenitor cell transportation.
Blood
90
1997
453
16
Ino
K
Singh
R
Talmadge
J
Monocytes from mobilized stem cells inhibit T cell function.
J Leukoc Biol
61
1997
583
17
Kitabayashi
A
Hirokawa
M
Hatano
Y
Lee
M
Kuroki
J
Niitsu
H
Miura
AB
Granulocyte colony stimulating factor downregulates allogeneic immune response by posttranscriptional inhibition of tumor necrosis factor-α production.
Blood
86
1995
2220
18
Mielcarek
M
Martin
PJ
Torok-Storb
B
Suppression of alloantigen-induced T-cell proliferation by CD14+ cells derived from granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cells.
Blood
89
1997
1629
19
Mielcarek
M
Graf
L
Johnson
G
Torok-Storb
B
Production of interleukin-10 by granulocyte colony-stimulating factor-mobilized blood products: A mechanism for monocyte-mediated suppression of T-cell proliferation.
Blood
92
1998
215
20
Storeck
J
Gooley
T
Siadak
M
Besinger
WI
Maloney
DG
Chauncey
TR
Flowers
M
Sullivan
KM
Witherspoon
RP
Rowley
SD
Hansen
JA
Storb
R
Appelbaum
FR
Allogeneic peripheral blood stem cell transplantation may be associated with a high risk of chronic graft-versus-host disease.
Blood
90
1997
4705
21
Majolino
I
Saglio
G
Scime
R
Serra
A
Cavallaro
AM
Fiandaca
T
Vasta
S
Pampinella
M
Catania
P
Indovina
A
Marceno
R
Santoro
A
High incidence of chronic GVHD after primary allogeneic peripheral blood stem cell transplantation in patients with hematologic malignancies.
Bone Marrow Transplant
17
1996
555
22
Morse
HC
Shen
FW
Hammerling
U
Genetic nomenclature for loci controlling mouse lymphocyte antigens.
Immunogenetics
25
1987
71
23
Yan
XQ
Hartley
C
McElroy
P
Chang
A
McCrea
C
McNiece
I
Peripheral blood progenitor cells mobilized by recombinant human granulocyte colony-stimulating factor plus recombinant rat stem cell factor contain long-term engrafting cells capable of cellular proliferation for more than two years as shown by serial transplantation in mice.
Blood
85
1995
2303
24
Bungart
B
Loeffler
M
Goris
H
Dontje
B
Diehl
V
Nijhof
W
Differential effects of recombinant human colony stimulating factor (rh G-CSF) on stem cells in marrow, spleen and peripheral blood in mice.
Br J Haematol
76
1990
174
25
Pojda
Z
Molineux
G
Dexter
TM
Hemopoietic effects of short-term in vivo treatment of mice with various doses of rhG-CSF.
Exp Hematol
18
1990
27
26
Nestel
F
Kichian
K
You-Ten
K
Desbarats
J
Price
K
Lapp
WS
The role of endotoxin in the pathogenesis of acute graft-versus-host disease
Graft-vs-Host Disease
ed 2
Ferrara
JLM
Deeg
HJ
Burakoff
SJ
1997
501
Marcel Dekker
New York, NY
27
Hill
GR
Crawford
JM
Cooke
KJ
Brinson
YS
Pan
L
Ferrara
JLM
Total body irradiation effects on acute graft versus host disease. The role of gastrointestinal damage and inflammatory cytokines.
Blood
90
1997
3204
28
Holler
E
Kolb
HJ
Moller
A
Kempeni
J
Lisenfeld
S
Pechumer
H
Lehmacher
W
Ruckdeschel
G
Gleixner
B
Riedner
C
Ledderose
G
Brehm
G
Mittermuller
J
Wilmanns
W
Increased serum levels of tumor necrosis factor alpha precede major complications of bone marrow transplantation.
Blood
75
1990
1011
29
Carter
LL
Dutton
RW
Relative perforin- and fas-mediated lysis in T1 and T2 CD8 effector populations.
J Immunol
155
1995
1028
30
Graubert
TA
Russell
JH
Ley
T
The role of granzyme B in murine models of acute graft-versus-host disease and graft rejection.
Blood
87
1996
1232
31
Fowler
DH
Kurasawa
K
Husebekk
A
Cohen
PA
Gress
RE
Cells of the Th2 cytokine phenotype prevent LPS-induced lethality during murine graft-versus-host reaction.
J Immunol
152
1994
1004
32
Fowler
DH
Kurasawa
K
Smith
R
Eckhaus
MA
Gress
RE
Donor CD4-enriched cells of Th2 cytokine phenotype regulate graft-versus-host disease without impairing allogeneic engraftment in sublethally irradiated mice.
Blood
84
1994
3540
33
Krenger
W
Snyder
KM
Byon
CH
Falzarano
G
Ferrara
JLM
Polarized type 2 alloreactive CD4+ and CD8+ donor T cells fail to induce experimental acute graft-versus-host disease.
J Immunol
155
1995
585
34
Kichian
K
Nestel
FP
Kim
D
Ponka
P
Lapp
WS
IL-12 p40 messenger RNA expression in target organs during acute graft-versus-host disease.
J Immunol
157
1996
2851
35
Via
CS
Rus
V
Gately
MK
Finkelman
FD
IL-12 stimulates the development of acute graft-versus-host disease in mice that would normally develop chronic, autoimmune graft-versus-host disease.
J Immunol
153
1994
4040
36
Anasetti
C
Hansen
JA
Waldmann
TA
Applebaum
FR
Davis
J
Deeg
HJ
Doney
K
Martin
PJ
Nash
R
Storb
R
Sullivan
KM
Witherspoon
RP
Binger
M
Chizzonite
R
Hakimi
J
Mould
D
Satoh
H
Light
SE
Treatment of acute graft versus host disease with humanized anti-Tac: An antibody that binds to the interleukin-2 receptor.
Blood
84
1994
1320
37
Ferrara
JLM
Marion
A
McIntyre
JF
Murphy
GF
Burakoff
SJ
Amelioration of acute graft-versus-host disease due to minor histocompatibility antigens by in vivo administration of anti-interleukin 2 receptor antibody.
J Immunol
137
1986
1874
38
Allen
RD
Staley
TA
Sidman
CL
Differential cytokine expression in acute and chronic murine graft-versus-host disease.
Eur J Immunol
23
1993
333
39
Kelso
A
Frequency analysis of lymphokine-secreting CD4+ and CD8+ T cells activated in a graft-versus-host reaction.
J Immunol
145
1990
2167
40
Rus
V
Svetic
A
Nguyen
P
Gause
WC
Via
CS
Kinetics of Th1 and Th2 cytokine production during the early course of acute and chronic murine graft-versus-host disease.
J Immunol
155
1995
2396
41
Blazar
BR
Taylor
PA
Panoskaltsis-Mortari
A
Narula
SK
Smith
SR
Roncarolo
MG
Vallera
DA
Interleukin-10 dose-dependent regulation of CD4+ and CD8+ T cell-mediated graft-versus-host disease.
Transplantation
66
1998
1220
42
Tanaka
J
Imamura
M
Kasai
M
Hashino
S
Kobayashi
S
The important balance between cytokines derived from type 1 and type 2 helper T cells in the control of graft-versus-host disease.
Bone Marrow Transplant
19
1997
571
43
Teshima
T
Harada
M
Mobilization of peripheral blood progenitor cells for allogeneic transplantation.
Cytokines Cell Mol Ther
3
1997
101
44
Nawa
Y
Teshima
T
Sunami
K
Hiramatsu
Y
Yano
T
Shinagawa
K
Omoto
E
Harada
M
Responses of granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cells to alloantigen stimulation.
Blood
90
1997
1716
45
Shenoy
S
Brown
R
Adkins
D
Dipersio
J
Mohanakumar
T
Constitutive peripheral blood cytokine mRNA expression in growth factor-mobilized normal donors and allogeneic peripheral blood stem cell transplant recipients.
Blood
90
1997
562a
(abstr, suppl 1)
46
Nestel
FP
Price
KS
Seemayer
TA
Lapp
WS
Macrophage priming and lipopolysaccharide-triggered release of tumor necrosis factor alpha during graft-versus-host disease.
J Exp Med
175
1992
405
47
Fowler
DH
Gress
RE
Graft-versus-host disease as a Th1-type process: Regulation by donor cells of Th2 cytokine phenotype
Graft-vs.-Host Disease.
Ferrara
JLM
Deeg
HJ
Burakoff
SJ
1997
479
Marcel Dekker
New York, NY
48
Remberger
M
Ringden
O
Markling
L
TNFα levels are increased during bone marrow transplantation conditioning in patients who develop acute GVHD.
Bone Marrow Transplant
15
1995
99
49
Holler
E
Kolb
HJ
Mittermueller
J
Kaul
M
Ledderose
G
Duell
T
Seeber
B
Schleuning
M
Hintermeier-Knabe
R
Ertl
B
Kempeni
J
Wilmanns
W
Modulation of acute graft-versus-host disease after allogeneic bone marrow transplantation by tumor necrosis factor α (TNFα) release in the course of pretransplant conditioning: Role of conditioning regimens and prophylactic application of a monoclonal antibody neutralizing human TNFa (MAK 195F).
Blood
86
1995
890
50
Hattori
K
Hirano
T
Tateno
M
Oshimi
K
Kayagaki
N
Yagita
H
Okumura
K
The synergistic effects of anti-Fas ligand and TNF-α antibody on the prevention of lethal acute graft-versus-host disease in mice.
Blood
90
1997
206a
(abstr, suppl 1)
51
Piguet
PF
Grau
GE
Allet
B
Vassalli
PJ
Tumor necrosis factor/cachectin is an effector of skin and gut lesions of the acute phase of graft-versus-host disease.
J Exp Med
166
1987
1280
52
Pape
KA
Khoruts
A
Mondino
A
Jenkins
MK
Inflammatory cytokines enhance the in vivo clonal expansion and differentiation of antigen-activated CD4+ T cells.
J Immunol
159
1997
591
53
Mielcarek
M
Roecklein
BA
Torok-Storb
B
CD14+ cells in granulocyte colony-stimulating factor (GCSF) mobilized peripheral blood mononuclear cells induce secretion of interleukin-6 and GCSF by marrow stroma.
Blood
87
1996
574
54
Porter
DL
Roth
MS
Lee
SJ
McGarigle
C
Ferrara
JL
Antin
JH
Adoptive immunotherapy with donor mononuclear cell infusions to treat relapse of acute leukemia or myelodysplasia after allogeneic bone marrow transplantation.
Bone Marrow Transplant
18
1996
975
55
Collins
RH
Shpilberg
WR
Drobyski
WR
Porter
DL
Giralt
S
Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation.
J Clin Oncol
15
1997
433
56
Dazzi
F
Goldman
JM
Adoptive immunotherapy following allogeneic bone marrow transplantation.
Annu Rev Med
49
1998
329
57
Helg
C
Starobinski
M
Jeannet
M
Chapuis
B
Donor lymphocyte infusion for the treatment of relapse after allogeneic hematopoietic stem cell transplantation.
Leuk Lymphoma
29
1998
301
58
Russell
LA
Jacobsen
N
Heilmann
C
Simonsen
AC
Christensen
LD
Vindelov
LL
Treatment of relapse after allogeneic BMT with donor leukocyte infusions in 16 patients.
Bone Marrow Transplant
18
1996
411
59
Glass
B
Uharek
L
Zeis
M
Dreger
P
Löffler
H
Steinmann
J
Schmitz
N
Allogeneic peripheral blood progenitor cell transplantation in a murine model: Evidence for an improved graft-versus-leukemia effect.
Blood
90
1997
1694
60
Glass
B
Majolino
I
Dreger
P
Scime
R
Santoro
A
Vasta
S
Suttorp
M
Haferlach
T
Schmitz
N
Allogeneic peripheral blood progenitor cells for treatment of relapse after bone marrow transplantation.
Bone Marrow Transplant
20
1997
533
61
Siegert
W
Beyer
J
Kingreen
D
Blasczyk
R
Baurmann
H
Schwella
N
Schleicher
J
Kirsch
A
Huhn
D
Treatment of relapse after allogeneic bone marrow transplantation with unmanipulated G-CSF-mobilized peripheral blood stem cell preparation.
Bone Marrow Transplant
22
1998
579
62
Lowin
B
Hahne
M
Mattmann
C
Tschopp
J
Cytolytic T-cell cytotoxicity is mediated through perforin and Fas lytic pathways.
Nature
370
1994
650
63
Kagi
D
Vignaux
F
Ledermann
B
Burki
K
Depraetere
V
Nagata
S
Hengartner
H
Golstein
P
Fas and perforin pathways as major mechanisms of T cell-mediated cytotoxicity.
Science
265
1994
528
64
Levy
RB
Baker
M
Podack
ER
Perforin deficiency delays the onset of GVHD following bone marrow transplantation across major and minor histocompatibility barriers.
Ann NY Acad Sci
770
1995
366
65
Baker
MB
Riley
RL
Podack
ER
Levy
RB
GVHD-associated lymphoid hypoplasia and B cell dysfunction is dependent upon donor T cell-mediated Fas-ligand function, but not perforin function.
Proc Natl Acad Sci USA
94
1997
1366
66
Via
CS
Nguyen
P
Shustov
A
Drappa
J
Elkon
KB
A major role for the Fas pathway in acute graft-versus-host disease.
J Immunol
157
1996
5387
67
Graubert
TA
DiPersio
JF
Russell
JH
Ley
TJ
Perforin/granzyme-dependent and independent mechanisms are both important for the development of graft-versus-host disease after murine bone marrow transplantation.
J Clin Invest
100
1997
904
68
Baker
MB
Altman
NH
Podack
ER
Levy
RB
The role of cell-mediated cytotoxicity in acute GVHD after MHC-matched allogeneic bone marrow transplantation in mice.
J Exp Med
183
1996
2645
69
Lin
T
Brunner
T
Tietz
B
Madsen
J
Bonfoco
E
Reaves
M
Huflejt
M
Green
DR
Fas ligand-mediated killing by intestinal intraepithelial lymphocytes. Participation in intestinal graft-versus-host disease.
J Clin Invest
101
1998
570
70
Yagita
H
Hanabuchi
S
Asano
Y
Tamura
T
Nariuchi
H
Okumura
K
Fas-mediated cytotoxicity—A new immunoregulatory and pathogenic function of Th1 CD4+ T cells.
Immunol Rev
146
1995
223
71
Shresta
S
Pham
CT
Thomas
DA
Graubert
TA
Ley
TJ
How do cytotoxic lymphocytes kill their targets?
Curr Opin Immunol
10
1998
581
72
Shustov
A
Nguyen
P
Finkelman
F
Elkon
KB
Via
CS
Differential expression of Fas and Fas ligand in acute and chronic graft-versus-host disease: Up-regulation of Fas and Fas ligand requires CD8+ T cell activation and IFN-gamma production.
J Immunol
161
1998
2848
73
Matsue
H
Kobayashi
H
Hosokawa
T
Akitaya
T
Ohkawara
A
Keratinocytes constitutively express the Fas antigen that mediates apoptosis in IFN-γ-treated cultured keratinocytes.
Arch Dermatol Res
287
1995
315

Author notes

Address reprint requests to James L.M. Ferrara, MD, Bone Marrow Transplant Program, University of Michigan Cancer Center, 1500 E Medical Center Dr, Ann Arbor, MI 48109; e-mail: ferrara@umich.edu.

Sign in via your Institution