In graft-versus-leukemia (GVL) responses, the cellular subsets and effector mechanisms responsible for cytotoxicity against leukemic cells in vivo remain poorly characterized. A murine model of syngeneic GVL that features CD4+ and CD8+T-cell responses against the MMB3.19 myeloid leukemia cell line has been previously described. MMB3.19 expresses high levels of functional Fas and tumor necrosis factor (TNF) receptors that do not transduce proapoptotic signals. Through the use of perforin- and Fas ligand (FasL)-deficient mice, it was demonstrated that CD4+ T cells mediate anti-MMB3.19 effects in vivo primarily through the use of FasL and secondarily through perforin mechanisms. Conversely, CD8+ T cells induce GVL effects primarily through the use of perforin and minimally through FasL mechanisms. Although the in vivo observations of CD8+ T cells were reflective of their in vitro cytotoxic T lymphocyte (CTL) activity, for CD4+ T cells, in vitro responses were dominated by the perforin pathway. In addition, the diminished capacity of T cells from perforin- and FasL-deficient mice to lyse MMB3.19 target cells appeared directly related to their deficient cytotoxic functions rather than to defects in activation because these cells were fully capable of mounting proliferative responses to the tumor cells. These findings demonstrate that GVL responses of T-cell subsets can involve preferential use of different cytotoxic mechanisms. In particular, these findings identify a role for both FasL-employing CD4+CTLs and the more novel perforin-utilizing CD4+ T-cell subset in responses against a myeloid leukemia.

Graft-versus-leukemia (GVL) responses are antitumor effects exerted by donor cells following hematopoietic stem cell (HSC) transplantation (HCT). GVL responses have been observed in patients with myeloid and lymphoid leukemias,1,2 and similar antitumor effects have been found in other malignancies such as multiple myeloma2 and breast cancer.3 However, myeloid leukemias seem particularly susceptible to GVL effects. T cells play a major role in GVL activity; this is based on findings that T-cell depletion of HSC grafts results in higher rates of leukemic relapse,4-6 the isolation of patient-derived T-cell clones that can kill leukemia cells in vitro,7-10 and the observations of successful reversal of leukemia relapse after delayed infusion of T cells.11 

The target antigens to which a GVL response are directed remain controversial and may likely involve both shared host alloantigens, which can induce graft-versus-host disease (GVHD), and unique tumor-specific antigens. Evidence for the alloreactive responses comes from the clinical observation that leukemic relapse is less frequent in patients suffering from acute or chronic forms of GVHD.12,13 In regard to leukemia antigen-specific responses, investigators have reported cohorts of allogeneic HCT patients who have experienced GVL without apparent GVHD.13In addition, autologous and syngeneic HSC grafts can be manipulated to induce GVL with or without a concomitant autoimmune-like GVHD.14-19 

T cells can mediate GVL effects through several means, depending upon their subset functional capabilities, and can involve either cytokine induction of other effector cells (eg, via inflammatory molecules such as interferon-γ [IFN-γ]) or direct cytolytic activity. In regard to the latter, both CD4+ and CD8+ cytotoxic T lymphocytes (CTLs) can mediate tumor cell apoptosis through several mechanisms including the exocytosis of cytotoxic granules containing perforin and granzymes, the binding of the Fas ligand (FasL) to target cell Fas, and the binding of tumor necrosis factor–alpha (TNF-α) to tumor cell receptors. Although in vitro studies have described the cytotoxic effector functions of antileukemic T-cell clones,20 the cytotoxic mechanisms involved in GVL responses in vivo have only begun to be characterized,21-24particularly with regard to myeloid leukemias.

We have previously established a mouse model of syngeneic and, therefore, tumor antigen-specific GVL activity that features CD4+ and CD8+ T-cell responses against MMB3.19, a myeloid leukemia of C57Bl/6 (B6) origin.25 In the present study, we demonstrate that GVL activity against MMB3.19 is not mediated by tumor necrosis factor–α (TNF-α). Instead, CD4+ T-cell GVL responses rely primarily on FasL and secondarily on perforin mechanisms. In contrast, CD8+T-cell GVL responses are primarily dependent upon perforin and secondarily on Fas-FasL interactions.

Mice

Wild type (wt) C57BL/6J (B6), perforin-deficient C57BL/6-Pfptm1Sdz(pfpo), and FasL-deficient B6Smn.C3H-Faslgld (gld) mice, all expressing the B6 background, were purchased directly or derived from breeding stock (The Jackson Laboratory, Bar Harbor, ME). Male wt andpfpo mice were used as donors between the ages of 7-12 weeks, while male gld mice were used as donors between the ages of 5-6 weeks. Male wt mice were used as recipients between the ages of 9-16 weeks. The mice were kept in a sterile environment in microisolators at all times and were provided with acidified water and autoclaved food ad libitum.

Cell lines

WEHI164 is a methylcholanthrene-induced fibrosarcoma (American Type Culture Collection, Manassas, VA). MMB3.19, amyc-transformed myeloid leukemia line, was cloned from the ascites of a B6 mouse that had been injected with amyc-encoding Moloney murine leukemia virus (MMLV) construct, as previously described.26 Both cell lines were maintained in Roswell Park Memorial Institute medium (RPMI 1640) supplemented with 10% fetal bovine serum (FBS), 5.5 × 10−5 mol/L 2-mercaptoethanol, 2 mmol/L L-glutamine, 50 IU/mL penicillin, and 50 μg/mL streptomycin.

Media

Phosphate-buffered saline (PBS) supplemented with 0.1% bovine serum albumin (BSA) (Sigma Chemical Co, St Louis, MO) was used for all in vitro manipulations of the donor bone marrow and lymphocytes. Immediately prior to injection, the cells were washed and resuspended in PBS alone. For in vitro assays, the cells were cultured in RPMI 1640 supplemented with 10% FBS, 5.5 × 10−5mol/L 2-mercaptoethanol, 2 mmol/L L-glutamine, 50 IU/mL penicillin, and 50 μg/mL streptomycin.

Irradiation

All recipient mice received an 850- or 950-cGy exposure from a Mark-I-68A cesium 137 (137Cs) source (JL Shepherd and Assocs, San Fernando, CA) at 143 cGy/min as indicated. For in vitro assays, MMB3.19 cells received a 30-Gy exposure.

Monoclonal antibodies

Ascitic fluids containing mAbs (clone names in parentheses; American Type Culture Collection, Manassas, VA) specific for Thy-1.2 (J1j), CD4 (RL172), CD8(3.168), natural killer 1.1 (NK1.1) (PK136), and rat immunoglobulin G (IgG) (MAR18.5; gift of Dr Robert Levy, Univ of Miami, Miami, FL) were used for the preparation of cellular grafts and culture supernatants. Other mAbs used included the supernatant of the rat-antimurine B220 14.8 mAb (gift of Dr Levy); affinity-purified goat antimouse IgG (whole molecule) antibodies (Cappel, Cosa Mesa, CA); guinea pig serum, which was prepared in our laboratory or purchased (Rockland, Boyertown, PA) and used as a source of complement for all mAb treatments; and biotinylated anti-Fas mAb (Jo2; PharMingen, San Diego, CA). For the phenotyping of MMB3.19 cells and the monitoring of cellular subset depletions from transplant grafts, we used fluorescein isothiocyanate (FITC)- and phycoerythrin (PE)-conjugated versions (clone names in parentheses, PharMingen) of the following mAbs that were specific for CD3ε(145-2C11), CD4 (RM4-5), CD8α(53-6.7), B220 (RA3-6B2), 2B4, and an isotype control (R35-95). We also used streptavidin (SA)-PE (Caltag, South San Francisco, CA).

Flow cytometry

Appropriate mAbs in volumes of 25 μL were incubated with 2-5 × 105 cells in the wells of a 96-well U-bottom microplate at 4°C for 25 minutes, centrifuged at 1500 rpm for 3 minutes, and washed with PBS containing 0.1% BSA and 0.01% sodium azide (wash buffer). When applicable, SA-PE or a secondary antibody was added in a volume of 25 μL at 4°C for 25 minutes, followed by 2 washes with wash buffer. The fluorescence analysis was performed on an EPICS Profile II analyzer (Coulter, Hialeah, FL) in the Kimmel Cancer Institute Flow Cytometry Facility, Philadelphia, PA.

Preparation of cellular grafts

Bone marrow cells were obtained from the femurs and tibias ofwt mice by flushing with PBS. To prepare anti–Thy-1–treated (T-cell–depleted) bone marrow (ATBM), the cells were incubated with J1j mAb (at 1:100 dilution) and complement (at 1:25 dilution) at 37°C for 45 minutes and were washed extensively. T-cell–enriched donor cell populations were prepared by treating pooled spleen and lymph node cells in 2 steps: (1) Gey's balanced salt lysing solution containing 0.7% ammonium chloride (NH4Cl) was used to remove red blood cells (RBCs), and (2) panning on a plastic petri dish precoated with a 5 μg/mL dilution of goat antimouse IgG for 1 hour at 4°C was used to remove B cells. These treatments resulted in donor populations of 90%-95% CD3+ cells, as quantitated by fluorescent flow cytometry. As required, the suspensions of enriched T cells or unfractionated splenocytes were depleted of CD4+ or CD8+ cells by treatment with the appropriate mAbs and complement at 37°C for 45 minutes. At the same time, B220+ B cells and T cells were depleted by the addition of 14.8 culture supernatant and MAR18.5 ascites to the cellular preparations. These treatments reduced the targeted populations down to background levels on flow cytometric analysis.

Cytotoxic assays

Mice were presensitized with 5 × 106 irradiated (30 Gy) MMB3.19 cells intraperitoneally (i.p.), and splenocytes or lymph node cells were harvested after 2 weeks and restimulated in vitro for 5 days with irradiated MMB3.19 cells and 1:20 of T-STIM culture supplement (Becton Dickinson Labware, Franklin Lakes, NJ). CD4+ or CD8+ T cells were prepared by depleting splenocytes or lymph node cells of the other T-cell subset and NK cells. The JAM assay was used to measure cytotoxicity.27 Briefly, MMB3.19 cells growing in the log phase were subcultured at 5 × 105 cells per mL in media containing 0.0925 Bq/mL (2.5 μCi/mL) of3H-thymidine (TdR) and allowed to label for 4 hours. The cells were then washed with media and cultured in 96-well U-bottom plates at 1 × 104 cells per well, along with effector cells at varying numbers. After 4 or 8 hours as indicated, the degree of target-cell DNA fragmentation was determined by comparing the radioactive incorporation of wells containing only target cells to wells containing both target and effector cells.

Survival assay for GVL activity

One day prior to transplantation, recipient mice were challenged with an i.p. injection of 0.5 mL PBS with or without 105 MMB3.19 cells. On the following day, recipient mice were lethally irradiated with 8.5 or 9.5 Gy, and approximately 6 hours later, they were injected intravenously (i.v.) with either 2 × 106 donor ATBM cells alone as a negative control or a mixture of ATBM cells plus manipulated donor lymphocyte populations. The mice were checked daily for morbidity and mortality until the experiments were terminated. As indicated, the data were pooled from 2 separate experiments, and median survival times (MSTs) were determined. Statistical comparisons between experimental groups were performed by the nonparametric Wilcoxon signed rank test.

RT-PCR

Total cellular RNA was prepared from 106-107MMB3.19 cells by homogenization in 1 mL Ultraspec (Biotecx Laboratories, Houston, TX), separated of cellular DNA and protein by the addition of a 1:5 volume of chloroform, vortexed for 5 seconds, and centrifuged at 12 500 rpm for 15 minutes. The aqueous phase was transferred to a clean Eppendorf tube, and the RNA was precipitated at 4°C by the addition of an equal volume of isopropanol and then centrifuged at 12 500 rpm for 15 minutes. The pellet was washed with 75% ethanol in diethyl pyrocarbonate (DEPC)-treated water and centrifuged again. The RNA pellet was resuspended in 25 μL DEPC water, heated to 55-65°C for 10 minutes, and stored at −20°C. Recovery of RNA was determined by spectrophotometry. For each sample, the reverse transcription and PCR reactions were performed in a 1-tube format using Ready-To-Go RT-PCR Beads (Amersham Pharmacia Biotech, Arlington Heights, IL). The following were added to each reaction: 1 μmol/L sense and antisense primers, 1.5 μg oligo d(T), and 2-4 μg total RNA.

The following TNF-R and glyceraldehyde-3-phosphate dehydrogenase (GADPH) primer sequences (Dr Keith Kelley, University of Illinois, Urbana, IL) have been previously described28: 55-kd TNF-R (expected product length, 368 base pairs [bp]): 5′-CAG TTG CAA GAC ATG TCG GA-3′ and 3′-GAC CTA GCA AGA TAA CCA GG-5′; 75-kd TNF-R (expected product length, 383 bp): 5′-GAG TGT GTG CTT GCG AAG CT-3′ and 3′-CGA TGT AAG GAT GCT TGG AG-5′; and GADPH (expected product length, 225 bp): 5′-GGA AGC TTG TCA TCA ATG G-3′ and 3′-AGA TCT CGT GGT TCA CAC C-5′. The reactions were performed using a Perkin-Elmer Applied Biosystems GeneAmp PCR System 9700 (Perkin-Elmer, Foster City, CA). Cycling conditions were as follows: 30 minutes at 42°C; 2 minutes at 94°C; and 30 cycles each of 30 seconds at 94°C, 45 seconds at 55°C, and 45 seconds at 72°C. The final extension was then performed for 7 minutes at 72°C. The product size was determined by electrophoresing the samples on an agarose gel and staining them with ethidium bromide.

Fas sensitivity assays

MMB3.19 cells were cultured in 40 mL medium (RPMI 1640 supplemented with 10% FBS, 5.5 × 10−5 mol/L 2-mercaptoethanol, 2 mmol/L L-glutamine, 50 IU/mL penicillin, and 50 μg/mL streptomycin) in tissue culture flasks at 1 × 105 cells per mL with either 500 ng/mL anti-Fas mAb (Jo2) or hamster anti-trinitrophenol (TNP) mAb as an isotype control. Both antibodies (PharMingen) were obtained in a low-endotoxin, sodium azide–free format. After 24 hours, the MMB3.19 cells were harvested, and an aliquot was analyzed for viability by trypan blue staining. The remainder of the cells were stained and analyzed using the APO-DIRECT Kit (PharMingen), a flow cytometry–based TUNEL method (terminal deoxynucleotidyl transferase–mediated dUTP nickend-labeling method).

Proliferation assays

Pfpo, gld, and wt mice were injected with 5 × 106 irradiated (30 Gy) MMB3.19 cells, and their splenocytes were harvested 2 weeks later. The cells were cultured in triplicate at 2 × 105 cells per well in 96-well flat-bottom plates, and irradiated (30 Gy) MMB3.19 cells were added at 100 cells per well to half the samples. After 3, 4, and 5 days, the cells were harvested following an overnight pulse with 0.037 MBq (1 μCi) per well of TdR, and the radioactivity incorporation into DNA was measured. Stimulation indices were calculated by dividing the mean cpm of splenocytes cultured with MMB3.19 cells by the mean cpm of splenocytes cultured alone.

TNF sensitivity assays

Assay conditions to determine the sensitivity of MMB3.19 cells to exogenous TNF-mediated cytotoxicity were based on previous studies.28,29 Recombinant murine TNF-α was used (Peprotech, Rocky Hill, NJ). MMB3.19 and WEHI164 cells were cultured overnight in quadruplicate in 96-well flat-bottom plates at 2 × 104 cells per well in a total volume of 100 μL. We then added 100 μL media, containing either actinomycin D alone (0.5 μg/mL final concentration) or actinomycin D and titrated concentrations of TNF-α, to each well. After 22 hours, 70 μL was aspirated from each well prior to the addition of 50 μL 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) (final concentration 1 mg/mL). After a 3.5- to 4-hour incubation at 37°C, 50 μL media was aspirated, and 150 μL acidified isopropanol (0.04 N hydrochloric acid [HCl]) was added to each well. The media in each well was then extensively run through a pipette to dissolve crystals prior to the measurement of each well's absorbance at 540 nm.

MMB3.19 cells express high levels of functional Fas

Since FasL-Fas interactions are a major mechanism by which T cells mediate target cell death, it was first determined whether MMB3.19 cells had Fas molecules on their surface. Flow cytometric analysis revealed a high level of Fas expression, with 86.6% of the cells staining positive for the marker (relative mean fluorescence intensity of 14.85; Figure 1). Next, to determine whether the Fas molecules expressed by MMB3.19 cells were competent to receive death signals, the cells were incubated with anti-Fas mAb for 24 hours, and viability was measured by trypan blue exclusion. Anti-Fas mAb-treated tumor cells exhibited 22.2% nonviability compared to only 3.2% nonviability of control mAb-treated cells (Figure 2A). Furthermore, there were approximately 42% fewer viable cells remaining in the anti-Fas mAb-treated group (4.14 × 106 cells vs 7.13 × 106 cells). TUNEL analysis of the same cultures revealed that anti-Fas mAb treatment increased the percentage of apoptotic cells from 10.6% to 38.7% at the 24-hour time-point (Figure2B). Collectively, this data indicated that MMB3.19 cells displayed sufficient levels of cell-surface Fas molecules that were capable of transducing apoptotic signals.

Fig. 1.

MMB3.19 expression of cell-surface Fas.

MMB3.19 cells were stained with a biotinylated anti-Fas mAb or a biotinylated isotype control antibody, followed in both cases by staining with SA-PE.

Fig. 1.

MMB3.19 expression of cell-surface Fas.

MMB3.19 cells were stained with a biotinylated anti-Fas mAb or a biotinylated isotype control antibody, followed in both cases by staining with SA-PE.

Close modal
Fig. 2.

Cell death of MMB3.19 cells induced by anti-Fas antibody.

MMB3.19 cells were incubated with anti-Fas mAb for 24 hours. Then (A) half the cells were stained with trypan blue and (B) the remaining cells were stained for TUNEL analysis.

Fig. 2.

Cell death of MMB3.19 cells induced by anti-Fas antibody.

MMB3.19 cells were incubated with anti-Fas mAb for 24 hours. Then (A) half the cells were stained with trypan blue and (B) the remaining cells were stained for TUNEL analysis.

Close modal

MMB3.19 expression of TNF receptors

To determine whether MMB3.19 cells expressed TNF receptors, which could potentially be used as death-signaling molecules and are often expressed at low levels, RT-PCR analyses of both TNF receptor type I and II were performed (Figure 3). Synthesis of both TNF receptors was detectable in the MMB3.19 cells, and expression of each was comparable to that found for the TNF-sensitive cell line, WEHI164. However, despite the presence of the TNF receptors, the MMB3.19 cells were resistant to exogenous TNF-mediated cytotoxicity throughout a concentration range of 0.02-20 ng/mL. This is in contrast to WEHI164 cells, which were highly susceptible at all concentrations (Figure 4). In addition, RT-PCR analysis also revealed that MMB3.19 cells produced endogenous TNF-α (data not shown), which is consistent with the function of this cytokine as a growth and/or activation factor. These combined results suggested that MMB3.19 cells do not receive death signals via ligation of their TNF receptors with exogenous TNF-α.

Fig. 3.

TNF receptor messenger RNA expression by MMB3.19 cells.

Messenger RNA (mRNA) was isolated from MMB3.19 (lanes 5-7) and WEHI164 (lanes 2-4) cells and reverse transcribed to produce complimentary DNA (cDNA). Primers for GADPH (lanes 2 and 5) and TNF receptors type I (lanes 3 and 6) and type II (lanes 4 and 7) were used for PCR, and the products were resolved on an agarose gel.

Fig. 3.

TNF receptor messenger RNA expression by MMB3.19 cells.

Messenger RNA (mRNA) was isolated from MMB3.19 (lanes 5-7) and WEHI164 (lanes 2-4) cells and reverse transcribed to produce complimentary DNA (cDNA). Primers for GADPH (lanes 2 and 5) and TNF receptors type I (lanes 3 and 6) and type II (lanes 4 and 7) were used for PCR, and the products were resolved on an agarose gel.

Close modal
Fig. 4.

MMB3.19 cell resistance to TNF-mediated cytotoxicity.

We cultured MMB3.19 (solid line) or WEHI164 (hatched line) cells in the presence of actinomycin D alone or actinomycin D and titrated amounts of TNF.

Fig. 4.

MMB3.19 cell resistance to TNF-mediated cytotoxicity.

We cultured MMB3.19 (solid line) or WEHI164 (hatched line) cells in the presence of actinomycin D alone or actinomycin D and titrated amounts of TNF.

Close modal

GVL-mediating T-cell subsets rely differentially on FasL and perforin

The cytotoxic effector mechanisms by which T-cell subsets mediate syngeneic GVL responses to MMB3.19 leukemia challenge were investigated. Donor T cells from FasL- and perforin-deficient, mice along with 2 × 106 ATBM cells, were transplanted into lethally irradiated (8.5 Gy) B6 mice that were preinoculated with 1 × 105 leukemia cells. Mice infused with 2 × 106 CD4+ T cells from MMB3.19-presensitized B6 wt donors exhibited GVL activity with a significant (P ≤ .03) extension of MST to 43 days. This contrasts with the 22-day MST for ATBM control mice that were challenged with MMB3.19 cells but did not receive any donor T cells (Figure 5A). Animals that were administered 2 × 106 CD4+ T cells from MMB3.19-presensitized B6 pfpo donor mice exhibited intermediate and marginally significant (P ≤ .075) GVL activity, with an MST of 33 days. Furthermore, the survival pattern of the pfpo group was significantly less (P ≤ .02) than the wt control. In contrast, 2 × 106 CD4+ T cells from MMB3.19-presensitized B6 gld donors did not appear to mediate any GVL activity because recipients of these cells had an MST of 22 days, which is equivalent to the recipients of ATBM plus leukemia cell challenge alone (P ≥ .38). The levels of GVL activity also correlated with the proportion of surviving mice at the termination of experiments on day 46; ie, 40% for mice receiving wt CD4+ T cells, 20% for recipients ofpfpo cells, and 0% for mice administered gld cells.

Fig. 5.

Role of perforin and FasL in CD4+T-cell–mediated GVL.

We injected B6 mice with MMB3.19 cells and lethally irradiated them the next day. (A) The recipient mice received ATBM cells alone (n = 8) or ATBM cells in combination with 2 × 106MMB3.19-primed CD4+ T cells from either wt(n = 10), pfpo (n = 10), orgld (n = 9) donor B6 mice. (B) The mice received ATBM cells alone (n = 7) or ATBM cells in combination with 1 × 107MMB3.19-primed CD4-enriched wt splenocytes (n = 7) or 5 × 107gld CD4-enriched splenocytes (n = 6). The survival of mice was monitored daily until termination of the experiment. The results in panels A and B represent pooled data from 2 similar experiments and a single experiment, respectively.

Fig. 5.

Role of perforin and FasL in CD4+T-cell–mediated GVL.

We injected B6 mice with MMB3.19 cells and lethally irradiated them the next day. (A) The recipient mice received ATBM cells alone (n = 8) or ATBM cells in combination with 2 × 106MMB3.19-primed CD4+ T cells from either wt(n = 10), pfpo (n = 10), orgld (n = 9) donor B6 mice. (B) The mice received ATBM cells alone (n = 7) or ATBM cells in combination with 1 × 107MMB3.19-primed CD4-enriched wt splenocytes (n = 7) or 5 × 107gld CD4-enriched splenocytes (n = 6). The survival of mice was monitored daily until termination of the experiment. The results in panels A and B represent pooled data from 2 similar experiments and a single experiment, respectively.

Close modal

In these experiments, pfpo CD4+T cells mediated GVL activity at a level that was less effective thanwt donor cells, which could still suggest a role for perforin cytolytic mechanisms. Because the initial experiment with 2 × 106 donor gld CD4+ T cells failed to demonstrate any GVL activity mediated by non-FasL pathways, it was imperative to test this notion with higher donor cell doses. The B6 mice were challenged with an i.p. dose of 1 × 105MMB3.19 cells, lethally irradiated with 9.5 Gy the next day, and administered either 2 × 106 ATBM cells alone or in combination with CD4+ T-cell–enriched (CD8+T-cell–depleted) splenocytes (Figure 5B). One group of mice received 107 CD4-enriched splenocytes from MMB3.19-primed wtdonors, while another group received 5 × 107CD4-enriched splenocytes from MMB3.19-primed gld donors. Flow cytometric analysis demonstrated that CD4+ T cells represented approximately 25% of the splenocyte populations (data not shown), which corresponds to a CD4+ T-cell dose of 2.5 × 106 cells and 1.25 × 107cells for recipients of wt and gld cells, respectively.

As expected, the mice that were challenged with MMB3.19 cells and received ATBM alone died rapidly, with only 14% surviving long-term (MST of 30 days). When the mice were challenged with MMB3.19 cells and administered wt CD4+ T-cell–enriched splenocytes, 57% survived beyond 60 days (P ≤ .03), with an MST of more than 60 days. Of most importance, 100% of the MMB3.19-challenged mice receiving gld CD4-enriched splenocytes survived for longer than 60 days (P ≤ .03). This demonstrated that a 5-fold increase of the dose of gld cells relative to wt cells could result in comparable if not superior GVL activity (P ≤ .10). Considered together with the earlier lack of GVL activity mediated by low-dose gld cells (Figure 5A), these results suggested that given enough effector cells, the perforin-mediated cytotoxic pathway could participate in the CD4+ T-cell GVL response, but that the FasL-Fas pathway was clearly dominant.

We infused 5 × 106 CD8+ T cells from MMB3.19-presensitized wt donors into lethally irradiated (8.5 Gy) mice challenged with 1 × 105leukemia cells. After this infusion, GVL activity was clearly demonstrated by the survival of 80% of the recipients for more than 46 days (at which time the experiments were terminated). This compares (P ≤ .02) to 100% lethality with a MST of 22 days for MMB3.19-challenged mice that received only ATBM cells (Figure6). The administration of 5 × 106 CD8+ T cells from MMB3.19-presensitized gld donors to leukemia-challenged recipients resulted in intermediate GVL activity, with 54.5% long-term survival and an MST of 34 days. This compared (P ≤ .02) to the ATBM plus MMB3.19 control, but it was not significantly different (P > .20) from the wt group. However, cells from the pfpo donors mediated very weak responses, with 13.3% long-term survival and an MST of 24 days. This was significantly greater (P ≤ .02) than the ATBM plus MMB3.19 control, but also significantly less (P ≤ .02) than the gld donors. Thus, CD8+ T cells mediating GVL activity seemed to rely heavily upon the use of perforin, with only minimal dependence upon FasL-mediated functions.

Fig. 6.

Role of perforin and FasL in CD8+T-cell–mediated GVL.

We injected B6 mice with MMB3.19 cells and lethally irradiated them the next day. These recipient mice received ATBM cells alone (n = 10) or in combination with 5 × 106 MMB3.19-primed CD8+ T cells from either wt (n = 10), pfpo (n = 15), or gld (n = 11) donor B6 mice. Survival of the mice was monitored daily until termination of the experiment. The results represent pooled data from 2 similar experiments.

Fig. 6.

Role of perforin and FasL in CD8+T-cell–mediated GVL.

We injected B6 mice with MMB3.19 cells and lethally irradiated them the next day. These recipient mice received ATBM cells alone (n = 10) or in combination with 5 × 106 MMB3.19-primed CD8+ T cells from either wt (n = 10), pfpo (n = 15), or gld (n = 11) donor B6 mice. Survival of the mice was monitored daily until termination of the experiment. The results represent pooled data from 2 similar experiments.

Close modal

MMB3.19-directed CTL activity in vitro reflects GVL activity in vivo

To determine the capacity of the CD4+ T-cell subset to mediate in vitro cytotoxic activity against MMB3.19 target cells, CD4+ T cells were prepared from CD8-cell– and NK-cell–depleted lymph nodes of MMB3.19-presensitized mice. The cells were then restimulated with leukemia cells in vitro and used as effectors in an 8-hour JAM assay using 3H-TdR–labeled MMB3.19 target cells (Figure 7A). As reflected in vivo, wt cells exhibited the best, albeit modest, cytotoxic activity at all effector:target (E:T) ratios tested, with a maximum specific killing of 15% at a 50:1 ratio. The gld cells yielded less cytolysis, 8.3%, although they were statistically insignificant from the wt group (P ≥ .12). However, the pfpo cells were significantly less capable of lysing MMB3.19 cells (1.43%; P ≤ .01).

Fig. 7.

Lysis of MMB3.19 target cells by in vitro restimulated CTLs.

(A) CD4-enriched lymph node cells from MMB3.19-primed wt,pfpo, and gld mice were restimulated in vitro with irradiated MMB3.19 cells in the presence of T-STIM. They were then used as effectors in a JAM assay with3H-TdR–labeled MMB3.19 target cells. (B) CD8-enriched splenocytes from MMB3.19-challenged wt,pfpo, and gld mice were restimulated in vitro with irradiated MMB3.19 cells in the presence of T-STIM. They were depleted of NK cells and CD4+ T cells prior to use as CTL effectors against 3H-TdR labeled MMB3.19 targets. The data are presented as the percentage of specific lysis plus or minus one standard deviation.

Fig. 7.

Lysis of MMB3.19 target cells by in vitro restimulated CTLs.

(A) CD4-enriched lymph node cells from MMB3.19-primed wt,pfpo, and gld mice were restimulated in vitro with irradiated MMB3.19 cells in the presence of T-STIM. They were then used as effectors in a JAM assay with3H-TdR–labeled MMB3.19 target cells. (B) CD8-enriched splenocytes from MMB3.19-challenged wt,pfpo, and gld mice were restimulated in vitro with irradiated MMB3.19 cells in the presence of T-STIM. They were depleted of NK cells and CD4+ T cells prior to use as CTL effectors against 3H-TdR labeled MMB3.19 targets. The data are presented as the percentage of specific lysis plus or minus one standard deviation.

Close modal

In a similar experiment to examine the antileukemic activity of CD8+ CTLs, splenocytes from MMB3.19-challenged wt,pfpo, and gld mice were depleted of CD4+ and NK cells prior to use as effectors against3H-TdR–labeled MMB3.19 target cells (Figure 7B). CD8+ CTLs exerted the most cytotoxic activity when derived from wt or gld mice (for both groups, peak specific killing of 54% at an E:T of 10:1), whilepfpo CTLs mediated virtually no lysis of MMB3.19 target cells (4.4% peak killing).

Splenocytes from wt, gld, andpfpomice can proliferate comparably in response to MMB3.19 stimulation

To ensure that pfpo and gld T cells still responded to the MMB3.19 cells, despite their cytotoxic defects, MMB3.19-primed wt, pfpo, and gld splenocytes were cultured with irradiated (30 Gy) MMB3.19 cells and assayed for their proliferative capability (Figure8). The stimulation indices of all 3 types of splenocytes were not significantly different from each other at the peak of the response on day 5 (P ≥ .20 for all comparisons). This data implied that T cells from pfpoand gld mice can respond to MMB3.19 antigens, even though they are incapable of killing the tumor cells via their individual deficient cytotoxic functions.

Fig. 8.

Proliferation of leukemia-primed splenocytes in response to in vitro restimulation with MMB3.19 cells.

Tumor-primed wt, pfpo, andgld splenocytes were cultured with irradiated (30 Gy) MMB3.19 cells and assayed for their ability to incorporate 3H-TdR. Stimulation indices plus or minus SD were calculated based on the mean cpm of wells containing splenocytes and MMB3.19 cells to the mean cpm of background wells containing responder cells alone (day 5 cpm:gld, 7186; wt, 11 494; andpfpo, 3636). Wells with only irradiated MMB3.19 cells had incorporation of between 100 and 150 cpm.

Fig. 8.

Proliferation of leukemia-primed splenocytes in response to in vitro restimulation with MMB3.19 cells.

Tumor-primed wt, pfpo, andgld splenocytes were cultured with irradiated (30 Gy) MMB3.19 cells and assayed for their ability to incorporate 3H-TdR. Stimulation indices plus or minus SD were calculated based on the mean cpm of wells containing splenocytes and MMB3.19 cells to the mean cpm of background wells containing responder cells alone (day 5 cpm:gld, 7186; wt, 11 494; andpfpo, 3636). Wells with only irradiated MMB3.19 cells had incorporation of between 100 and 150 cpm.

Close modal

GVL responses can potentially be mediated by T cells present in both autologous and allogeneic HSC grafts. Interestingly, the myeloid leukemias seem most sensitive to GVL activity, although very little is known about these responses. Investigations into murine GVL myeloid models have been hampered in the past by the limited availability of myeloid cell lines, particularly those that do not involve the expression of retroviral antigens. Our model for GVL responses features a myeloid leukemia that was transformed through transduction with a c-myc construct that does not result in any detectable virus-related protein expression.25 In addition, c-myc is a frequently mutated gene in hematologic malignancies, and therefore in contrast to other cell lines,30-32 the leukemia-specific antigen(s) expressed by the MMB3.19 cells may reflect the consequences of oncogenesis rather than retroviral infection.

Although our syngeneic model is highly suited for the study of leukemia antigen-restricted GVL responses, clinical GVL has been noted to occur more frequently in patients receiving allogeneic rather than autologous or syngeneic HSC grafts.12,13 However, it is unclear to what degree GVL reflects genuine leukemia antigen-specific responses versus overlapping GVHD responses mounted against alloantigens shared by the host tissues and leukemia cells. Yet, isolated clinical studies have described allogeneic HSC patients who have experienced GVL without apparent GVHD and vice versa.13 Similar separation of allogeneic GVHD and GVL has also been achieved in several animal models.22-25,33-35 Furthermore, multiple experimental systems and clinical protocols have demonstrated that syngeneic or autologous GVL can be induced without a concomitant autoimmune-like GVHD.14-19,25 Finally, several studies have successfully used leukemia-specific antigens, such as bcr-abl and proteinase-3, to stimulate T-cell clonal responses both in vitro and in vivo.8,36-39 

The dependence of GVL on T cells has been inferred through observations that leukemic relapse occurs less frequently in patients receiving T-cell–replete HSC grafts than in patients receiving grafts that have been T-cell depleted.4-6 In addition to these clinical data, reports that human T-cell clones can kill leukemic cells in vitro7-10 strongly suggest that T cells are the primary mediators of GVL activity.

In addition to T cells playing a crucial role, studies have also identified NK cells as mediators of GVL activity, either by themselves or as an effector arm dependent upon CD4+ T-cell cytokine induction.40,41 However, in the MMB3.19 model, there was no evidence of NK-cell involvement in GVL activity; weekly injections of depleting anti-NK1.1 mAb did not alter the survival of MMB3.19-challenged recipients of naive wt CD4+ T cells. In addition, in vitro NK-cell–mediated cytotoxicity by naive splenocytes could be demonstrated against susceptible YAC–1 cells but not against MMB3.19 cells (R.K. and Townsend, unpublished observations, 1996).

The findings presented here constitute one of the few reports that CD4+ T cells are capable of mediating GVL effects in vivo (Figure 5A). Specifically, we found that this subset of CD4+ T cells relied primarily on the use of FasL and, surprisingly, secondarily on perforin in order to mediate GVL effects against the MMB3.19 leukemia. The finding that higher doses of gldCD4+ T cells could mount effective GVL activity (Figure5B) was consistent with perforin acting as a secondary mechanism involved in this particular CD4+ T-cell–mediated GVL response. Interestingly, despite the obvious dominance of the FasL GVL pathways in vivo, in the in vitro CD4+ CTL response to MMB3.19 target cells (albeit a weak response, with only 15% specific lysis at a 50:1 E:T ratio using CTLs from wt mice; Figure 7A), CTLs from pfpo mice displayed only minimal cytolysis of MMB3.19 cells. By comparison, CD4+ CTLs fromgld mice proved much more effective in vitro. This paradox may be accounted for by the observations made elsewhere that restimulating CD4+ T cells in the absence of CD8+ T cells allows development of CD4+ CTLs, but the restimulation may give rise to a skewed population that is more reliant on perforin than cells restimulated in vivo.42 

Clearly, CD8+ CTLs dominate the cytolytic response after in vivo priming and in vitro restimulation with MMB3.19 cells. The proportion of cytolytic precursor cells within the CD8+population is normally very high to begin with (90%-95%), whereas only a relatively small portion (less than 10%) of the CD4+ T cells have perforin granules and cytolytic capability.42 Of course, these proportions will vary with individual responses to antigens, but in any event, the findings are consistent with a lower frequency of CD4+ effector cells. This proportional difference in vitro is also consistent with the in vivo result, whereby the CD4+gld population had to be increased significantly to exhibit GVL activity.

In contrast to the CD4+ responses, the CD8+ T cells involved in GVL responses to MMB3.19 challenge relied primarily on perforin and only secondarily on FasL (Figure 6). CTL assays using CD8+ effector cells were consistent with the GVL experiments (Figure 7B) in that CD8+ CTLs from wtor gld mice could efficiently lyse the MMB3.19 cells, whereas the pfpo CTLs were ineffective.

Of note, a previous study43 cautioned that FasL-deficientgld T cells may be unable to mediate cytotoxicity because of an inability to expand in vivo and not just because of a lack of functional Fas-FasL interactions between T cells and their targets. To exclude this possibility, we used gld mice at 5-6 weeks of age and depleted B220+ T cells from inocula derived from these animals.44 Both of these measures have been shown to exclude the anergized B220+CD4CD8T-cell subset that develops in older gld mice and features aberrant functions.45,46 With regard to similar concerns in the use of pfpo mice, careful studies have confirmed that these animals are appropriate for examining antigen-specific responses.47 We have also demonstrated that tumor-primed wt, gld, andpfpo splenocytes were all capable of comparable proliferation in response to restimulation with irradiated MMB3.19 cells (Figure 8). This strongly suggested that at least on a gross level, T cells from all 3 types of mice were equivalent by one classic definition of T-lymphocyte function.

In another study of cytotoxic effector mechanisms involved in GVL, Tsukada and colleagues23 concluded that CD8+GVL activity could be preserved while alleviating GVH through the use of gld donors but not through the use ofpfpo donors. These experiments were performed using the MHC Class I+II L1210 T-cell leukemia and P815 mastocytoma cell lines. Because L1210 is impervious to TNF- and Fas-mediated apoptosis in vitro, it is not surprising that perforin, the only other significant cytotoxic molecule available for antileukemic effects, expressed by glddonors is sufficient to induce GVL effects. It is more interesting that gld lymphocytes can induce GVL against P815 because this leukemia is sensitive to apoptosis induced by FasL-transfected effector cells, but it is resistant to TNF-mediated cell death.23Analogous to GVL activity against L1210 and to MMB3.19 cells in our own study, this result implies that the perforin expressed by gldCD8+ T cells is adequate to eliminate P815 cells.

Investigators have reported that in an allogeneic GVL model, CD8+ T cells rely equally on perforin and FasL to mediate antitumor activity.21 Based on the criteria of body weight, spleen size, and the appearance of mice, the authors also reported that the CD8+ T cells did not mediate GVHD. This study is very interesting in light of our findings that CD8+ T cells mediated GVL activity primarily through perforin and only minimally through FasL (Figure 6). In another system, T cells present in granulocyte colony-stimulating factor (GCSF)–mobilized peripheral blood stem cell transplantation grafts mediated a perforin-dependent allogeneic GVL effect against the P815 mastocytoma.22 As stated above, the reliance of T cells on perforin is particularly intriguing because P815 expresses functional cell-surface Fas. In this model, the exact T- cell subsets responsible for GVL are as of yet unknown.

MMB3.19 cytolysis by CD4+ and CD8+ CTLs is not likely to be induced through TNF-α–TNF receptor interactions because the MMB3.19 cells were resistant to high concentrations of exogenous TNF-α (Figure 4) and produced endogenous cytokine (data not shown). This may reflect an autocrine growth loop that merits future study. The perforin-utilizing CD4+ T cells that are effective at high doses in our model (Figure 5B) are especially intriguing because such cells have only been rarely characterized in vivo.48 This unusual cellular subset has been described in vitro more extensively.49-52 In addition, the importance of the perforin pathway in antitumor responses other than GVL has been underscored by other in vivo models.53 

Cytotoxic effector functions involved in GVL are best discussed in the context of GVHD. Perforin-deficient, unfractionated T cells from fully allogeneic mice or MHC-matched mice differing at minor histocompatibility loci can induce acute GVHD, but with delayed kinetics.43,44 However, FasL-deficient, unfractionated T cells can induce cachexia without cutaneous or hepatic pathology.44 Anti-FasL antibodies administered in vivo have confirmed that FasL on unfractionated T cells seems to mediate cutaneous and hepatic GVHD.54 In addition, through use of FasL-deficient donors, GVHD-associated lymphoid hypoplasia and dysfunction have also been attributed to FasL rather than to perforin expression.43,44 

For other fully allogeneic models, both CD4+ and CD8+ T cells depend on perforin for induction of optimal GVHD.48 In contrast, other studies have reported that CD4+ T-cell–mediated GVHD is heavily dependent on FasL, whereas CD8+ T-cell–induced disease is more reliant on perforin-mediated pathways.55 Furthermore, granzyme B has been found to be important for GVHD mediated by CD8+ MHC class I–mismatched T cells, but it is not important for GVHD induced by CD4+ T cells.56 In addition, through use of neutralizing anti-TNF antibodies in various models of GVHD, several investigations have demonstrated that GVHD can be highly dependent on TNF activity.54,57-59 

Because GVL activity against MMB3.19 cells is as equally affected by an absence of FasL on CD4+ T cells as by an absence of perforin on CD8+ T cells, we do not believe that MMB3.19 cells are inherently more sensitive to FasL-mediated versus perforin-mediated apoptosis. Instead, we hypothesize that GVL-conferring T-cell subsets use different patterns of effector mechanisms whose development is determined by MHC class I- and class II-restricted tumor antigens. Investigators have characterized several systems in which antigens influence the cytotoxic effector functions used by T cells.60-63 Final proof of this hypothesis will require, at the minimum, identification of at least one leukemia antigen that preferentially induces FasL responses and another antigen that skews T cells to use perforin-containing cytotoxic granules. Another set of tenable hypotheses to account for the particular cytolytic mechanisms used by GVL-mediating T cells in our model include those involving the influence of cytokines.

As shown earlier by others, the balance between Tc1- and Tc2-associated cytokines present during CTL development may dictate the armamentarium employed by antileukemic effector cells.24Regardless of what determines the effector mechanisms used by GVL-mediating T cells, the patterns of cytotoxic functions employed against tumors may differ from those brought to bear against host tissues in GVHD.23 This possibility could form the basis for future therapeutic approaches to minimize GVHD while preserving GVL activity.

We express our gratitude to Jeffrey Vakil and Kristin Naper for technical assistance and to David Dicker for his expertise in flow cytometric analyses.

Supported by research grants R01 HL55593, R01 CA60630, and T32 CA09683 from the U.S. Public Health Service.

Reprints:Robert Korngold, Kimmel Cancer Institute, Jefferson Medical College, 233 South 10th Street, Philadelphia, PA 19107; e-mail:r_korngold@lac.jci.tju.edu.

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
Rondon
G
Giralt
S
Huh
Y
et al
Graft-versus-leukemia effect after allogeneic bone marrow transplantation for chronic lymphocytic leukemia.
Bone Marrow Transplant.
18
1996
669
672
2
Tricot
G
Vesole
DH
Jagannath
S
Hilton
J
Munshi
N
Barlogie
B
Graft-versus-myeloma effect: proof of principle.
Blood.
87
1996
1196
1198
3
Eibl
B
Schwaighofer
H
Nachbaur
D
et al
Evidence for a graft-versus-tumor effect in a patient treated with marrow ablative chemotherapy and allogeneic bone marrow transplantation for breast cancer.
Blood.
88
1996
1501
1508
4
Mitsuyasu
RT
Champlin
RE
Gale
RP
et al
Treatment of donor bone marrow with monoclonal anti-T-cell antibody and complement for the prevention of graft-versus-host disease. A prospective, randomized, double-blind trial.
Ann Intern Med.
105
1986
20
26
5
Goldman
JM
Gale
RP
Horowitz
MM
et al
Bone marrow transplantation for chronic myelogenous leukemia in chronic phase. Increased risk for relapse associated with T-cell depletion.
Ann Intern Med.
108
1988
806
814
6
Apperley
JF
Jones
L
Hale
G
et al
Bone marrow transplantation for patients with chronic myeloid leukaemia: T-cell depletion with Campath-1 reduces the incidence of graft-versus-host disease but may increase the risk of leukaemic relapse.
Bone Marrow Transplant.
1
1986
53
66
7
Sosman
JA
Oettel
KR
Smith
SD
Hank
JA
Fisch
P
Sondel
PM
Specific recognition of human leukemic cells by allogeneic T cells, II: evidence for HLA-D restricted determinants on leukemic cells that are crossreactive with determinants present on unrelated nonleukemic cells.
Blood.
75
1990
2005
2016
8
Nieda
M
Nicol
A
Kikuchi
A
et al
Dendritic cells stimulate the expansion of bcr-abl specific CD8+ T cells with cytotoxic activity against leukemic cells from patients with chronic myeloid leukemia.
Blood.
91
1998
977
983
9
Faber
LM
van Luxemburg-Heijs
SA
Willemze
R
Falkenburg
JH
Generation of leukemia-reactive cytotoxic T lymphocyte clones from the HLA-identical bone marrow donor of a patient with leukemia.
J Exp Med.
176
1992
1283
1289
10
van Lochem
E
de Gast
B
Goulmy
E
In vitro separation of host specific graft-versus-host and graft-versus-leukemia cytotoxic T cell activities.
Bone Marrow Transplant.
10
1992
181
183
11
Drobyski
WR
Hessner
MJ
Klein
JP
Kabler-Babbitt
C
Vesole
DH
Keever-Taylor
CA
T-cell depletion plus salvage immunotherapy with donor leukocyte infusions as a strategy to treat chronic-phase chronic myelogenous leukemia patients undergoing HLA-identical sibling marrow transplantation.
Blood.
94
1999
434
441
12
Weiden
PL
Flournoy
N
Thomas
ED
et al
Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts.
N Engl J Med.
300
1979
1068
1073
13
Horowitz
MM
Gale
RP
Sondel
PM
et al
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood.
75
1990
555
562
14
Klingemann
HG
Eaves
CJ
Barnett
MJ
et al
Transplantation of patients with high risk acute myeloid leukemia in first remission with autologous marrow cultured in interleukin-2 followed by interleukin-2 administration.
Bone Marrow Transplant.
14
1994
389
396
15
Charak
BS
Brynes
RK
Katsuda
S
Groshen
S
Chen
SC
Mazumder
A
Induction of graft versus leukemia effect in bone marrow transplantation: dosage and time schedule dependency of interleukin 2 therapy.
Cancer Res.
51
1991
2015
2020
16
Charak
BS
Brynes
RK
Chogyoji
M
Kortes
V
Tefft
M
Mazumder
A
Graft versus leukemia effect after transplantation with interleukin-2-activated bone marrow: correlation with eradication of residual disease.
Transplantation.
56
1993
31
37
17
Byrne
JL
Carter
GI
Ellis
I
Haynes
AP
Russell
NH
Autologous GVHD following PBSCT, with evidence for a graft-versus-myeloma effect.
Bone Marrow Transplant.
20
1997
517
520
18
Weiss
L
Nusair
S
Reich
S
Sidi
H
Slavin
S
Induction of graft versus leukemia effects by cell-mediated lymphokine-activated immunotherapy after syngeneic bone marrow transplantation in murine B cell leukemia.
Cancer Immunol Immunother.
43
1996
103
108
19
Slavin
S
Nagler
A
Cytokine-mediated immunotherapy following autologous bone marrow transplantation in lymphoma and evidence of interleukin-2-induced immunomodulation in allogeneic transplants [see comments].
Cancer J Sci Am.
3(suppl 1)
1997
S59
67
20
Jiang
YZ
Mavroudis
D
Dermime
S
et al
Alloreactive CD4+ T lymphocytes can exert cytotoxicity to chronic myeloid leukaemia cells processing and presenting exogenous antigen.
Br J Haematol.
93
1996
606
612
21
Ito
M
Shizuru
JA
Graft-vs.-lymphoma effect in an allogeneic hematopoietic stem cell transplantation model.
Biol Blood Marrow Tranplant.
5
1999
357
368
22
Pan
L
Teshima
T
Hill
GR
et al
Granulocyte colony-stimulating factor-mobilized allogeneic stem cell transplantation maintains GVL effects through a perforin-dependent pathway while preventing graft-versus-host disease.
Blood.
93
1999
4071
4078
23
Tsukada
N
Kobata
T
Aizawa
Y
Yagita
H
Okumura
K
Graft-versus-leukemia effect and graft-versus-host disease can be differentiated by cytotoxic mechanisms in a murine model of allogeneic bone marrow transplantation.
Blood.
93
1999
2738
2747
24
Fowler
DH
Gress
RE
CD8+ T cells of Tc2 phenotype mediate a GVL effect and prevent marrow rejection.
Vox Sang.
74
1998
331
340
25
Korngold
R
Leighton
C
Manser
T
Graft-versus-myeloid leukemia responses following syngeneic and allogeneic bone marrow transplantation.
Transplantation.
58
1994
278
287
26
Baumbach
WR
Keath
EJ
Cole
MD
A mouse c-myc retrovirus transforms established fibroblast lines in vitro and induces monocyte-macrophage tumors in vivo.
J Virol.
59
1986
276
283
27
Matzinger
P
The JAM test. A simple assay for DNA fragmentation and cell death.
J Immunol Methods.
145
1991
185
192
28
Sipe
KJ
Srisawasdi
D
Dantzer
R
Kelley
KW
Weyhenmeyer
JA
An endogenous 55 kDa TNF receptor mediates cell death in a neural cell line.
Brain Res Mol Brain Res.
38
1996
222
232
29
Vanhaesebroeck
B
Decoster
E
Van Ostade
X
et al
Expression of an exogenous tumor necrosis factor (TNF) gene in TNF-sensitive cell lines confers resistance to TNF-mediated cell lysis.
J Immunol.
148
1992
2785
2794
30
LeFever
AV
Truitt
RL
Shih
CC
Reactivity of in-vitro-expanded alloimmune cytotoxic T lymphocytes and Qa-1-specific cytotoxic T lymphocytes against AKR leukemia in vivo.
Transplantation.
40
1985
531
537
31
Bortin
MM
Rimm
AA
Saltzstein
EC
Rodey
GE
Graft versus leukemia, 3: apparent independent antihost and antileukemia activity of transplanted immunocompetent cells.
Transplantation.
16
1973
182
188
32
Slavin
S
Eckerstein
A
Weiss
L
Adoptive immunotherapy in conjunction with bone marrow transplantation: amplification of natural host defense mechanisms against cancer by recombinant IL-2.
Nat Immun Cell Growth Regul.
7
1988
180
184
33
Glass
B
Uharek
L
Gassmann
W
et al
Graft-versus-leukemia activity after bone marrow transplantation does not require graft-versus-host disease.
Ann Hematol.
64
1992
255
259
34
de la Selle
V
Miconnet
I
Gilbert
D
Bruley-Rosset
M
Peripheral tolerance to host minor histocompatibility antigens in radiation bone marrow chimeras abrogates lethal GVHD while preserving GVL effect.
Bone Marrow Transplant.
16
1995
111
118
35
Sykes
M
Harty
MW
Szot
GL
Pearson
DA
Interleukin-2 inhibits graft-versus-host disease-promoting activity of CD4+ cells while preserving CD4- and CD8-mediated graft-versus-leukemia effects.
Blood.
83
1994
2560
2569
36
ten Bosch
GJ
Toornvliet
AC
Friede
T
Melief
CJ
Leeksma
OC
Recognition of peptides corresponding to the joining region of p210BCR-ABL protein by human T cells.
Leukemia.
9
1995
1344
1348
37
Greco
G
Fruci
D
Accapezzato
D
et al
Two brc-abl junction peptides bind HLA-A3 molecules and allow specific induction of human cytotoxic T lymphocytes.
Leukemia.
10
1996
693
699
38
Pawelec
G
Max
H
Halder
T
et al
BCR/ABL leukemia oncogene fusion peptides selectively bind to certain HLA-DR alleles and can be recognized by T cells found at low frequency in the repertoire of normal donors.
Blood.
88
1996
2118
2124
39
Molldrem
J
Dermime
S
Parker
K
et al
Targeted T-cell therapy for human leukemia: cytotoxic T lymphocytes specific for a peptide derived from proteinase 3 preferentially lyse human myeloid leukemia cells.
Blood.
88
1996
2450
2457
40
Jiang
YZ
Barrett
AJ
Goldman
JM
Mavroudis
DA
Association of natural killer cell immune recovery with a graft-versus-leukemia effect independent of graft-versus-host disease following allogeneic bone marrow transplantation.
Ann Hematol.
74
1997
1
6
41
Glass
B
Uharek
L
Zeis
M
Loeffler
H
Mueller-Ruchholtz
W
Gassmann
W
Graft-versus-leukaemia activity can be predicted by natural cytotoxicity against leukaemia cells.
Br J Haematol.
93
1996
412
420
42
Williams
NS
Engelhard
VH
Perforin-dependent cytotoxic activity and lymphokine secretion by CD4+ T cells are regulated by CD8+ T cells.
J Immunol.
159
1997
2091
2099
43
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
5393
44
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
2656
45
Davidson
WF
Roths
JB
Holmes
KL
Rudikoff
E
Morse
HC
III
Dissociation of severe lupus-like disease from polyclonal B cell activation and IL 2 deficiency in C3H-lpr/lpr mice.
J Immunol.
133
1984
1048
1056
46
Cohen
PL
Eisenberg
RA
Lpr and gld: single gene models of systemic autoimmunity and lymphoproliferative disease.
An Rev Immunol.
9
1991
243
269
47
White
DW
MacNeil
A
Busch
DH
Pilip
IM
Pamer
EG
Harty
JT
Perforin-deficient CD8+ T cells: in vivo priming and antigen-specific immunity against Listeria monocytogenes.
J Immunol.
162
1999
980
988
48
Blazar
BR
Taylor
PA
Vallera
DA
CD4+ and CD8+ T cells each can utilize a perforin-dependent pathway to mediate lethal graft-versus-host disease in major histocompatibility complex-disparate recipients.
Transplantation.
64
1997
571
576
49
Williams
NS
Engelhard
VH
Identification of a population of CD4+ CTL that utilizes a perforin rather than a Fas ligand-dependent cytotoxic mechanism.
J Immunol.
156
1996
153
159
50
Lancki
DW
Hsieh
CS
Fitch
FW
Mechanisms of lysis by cytotoxic T lymphocyte clones: lytic activity and gene expression in cloned antigen-specific CD4+ and CD8+ T lymphocytes.
J Immunol.
146
1991
3242
3249
51
Yakushijin
Y
Yasukawa
M
Kobayashi
Y
Establishment and functional characterization of human herpesvirus 6: specific CD4+ human T-cell clones.
J Virol.
66
1992
2773
2779
52
Miskovsky
EP
Liu
AY
Pavlat
W
et al
Studies of the mechanism of cytolysis by HIV-1-specific CD4+ human CTL clones induced by candidate AIDS vaccines.
J Immunol.
153
1994
2787
2799
53
van den Broek
ME
Kagi
D
Ossendorp
F
et al
Decreased tumor surveillance in perforin-deficient mice.
J Exp Med.
184
1996
1781
1790
54
Hattori
K
Hirano
T
Miyajima
H
et al
Differential effects of anti-Fas ligand and anti-tumor necrosis factor alpha antibodies on acute graft-versus-host disease pathologies.
Blood.
91
1998
4051
4055
55
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
911
56
Graubert
TA
Russell
JH
Ley
TJ
The role of granzyme B in murine models of acute graft-versus-host disease and graft rejection.
Blood.
87
1996
1232
1237
57
Wall
DA
Sheehan
KC
The role of tumor necrosis factor and interferon gamma in graft-versus-host disease and related immunodeficiency.
Transplantation.
57
1994
273
279
58
Piguet
PF
Grau
GE
Allet
B
Vassalli
P
Tumor necrosis factor/cachectin is an effector of skin and gut lesions of the acute phase of graft-vs-host disease.
J Exp Med.
166
1987
1280
1289
59
Murphy
GF
Sueki
H
Teuscher
C
Whitaker
D
Korngold
R
Role of mast cells in early epithelial target cell injury in experimental acute graft-versus-host disease.
J Invest Dermatol.
102
1994
451
461
60
Brossart
P
Bevan
MJ
Selective activation of Fas/Fas ligand-mediated cytotoxicity by a self peptide.
J Exp Med.
183
1996
2449
2458
61
Esser
MT
Krishnamurthy
B
Braciale
VL
Distinct T cell receptor signaling requirements for perforin- or FasL-mediated cytotoxicity.
J Exp Med.
183
1996
1697
1706
62
Kessler
BM
Bassanini
P
Cerottini
JC
Luescher
IF
Effects of epitope modification on T cell receptor-ligand binding and antigen recognition by seven H-2Kd-restricted cytotoxic T lymphocyte clones specific for a photoreactive peptide derivative.
J Exp Med.
185
1997
629
640
63
Bachmann
MF
Ohteki
T
Faienza
KM
et al
Altered peptide ligands trigger perforin rather than Fas-dependent cell lysis.
J Immunol.
159
1997
4165
4170
Sign in via your Institution