Depletion of T lymphocytes from allogeneic bone marrow transplants successfully prevents the development of graft-versus-host disease (GvHD) but is associated with impaired engraftment, immunosuppression, and abrogation of the graft-versus-leukemia effect. We therefore explored the possibility of selectively eliminating alloreactive T cells by CD95/CD95L–mediated activation-induced cell death (AICD) in a major histocompatibility complex allogeneic murine model system. Activation of resting or preactivated T lymphocytes from C3H/HeJ (H-2k) mice was induced with irradiated BALB/cJ (H-2d) mouse-derived stimulators. Substantial decrease (≥ 80%) of proliferative and lytic responses by activated alloreactive T cells was subsequently achieved by incubating the mixed lymphocyte culture with an agonistic monoclonal antibody to CD95, and residual T cells recovered did not elicit alloreactivity upon challenge to H-2d. Depletion of alloreactive T lymphocytes by AICD was specific because reactivity to an I-Ad–restricted ovalbumin (OVA) peptide by OVA-specific CD4+ T cells mixed into the allogeneic T-cell pool and subjected to induction of AICD in the absence of OVA peptide could be preserved. Adoptive transfer of donor-derived allogeneic T lymphocytes, depleted from alloreactive T cells by AICD in vitro, in the parent (C3H/He) to F1 (C3H/He × BALB/c) GvHD model prevented lethal GvHD. The results presented suggest that alloreactive T cells can effectively be depleted from allogeneic T cells by induction of AICD to prevent GvHD and might introduce a new strategy for the separation of GvH-reactive T cells and T cells mediating antiviral and possibly graft-versus-leukemia effects.

Graft-versus-host disease (GvHD) represents a major complication of allogeneic bone marrow transplantation (BMT), which results in significant morbidity and mortality. Donor T lymphocytes present in the BM or peripheral blood stem cell transplant cause GvHD by recognizing major as well as minor histocompatibility antigens (reviewed by Ferrara et al1). Although innovative strategies for the prediction, prevention, or treatment of this caveat in allogeneic BMT have been proposed,2-9 prophylaxis or treatment of GvHD still remains unsatisfactory. Removal of donor-derived T lymphocytes prior to transplantation efficiently ameliorates GvHD. However, ex vivo depletion of T cells has been associated with an increased risk of viral opportunistic infections,10 impaired engraftment,11 and the loss of the graft-versus-leukemia (GvL) effect, resulting in an increased incidence of recurrence of leukemia.12,13 These observations emphasize the clinical importance of allogeneic T cells in conferring a protective antiviral immunity as well as mediating a GvL effect. Several clinical and experimental studies support the idea that GvHD and GvL are not only mediated by the same (host-specific) T cell but also by distinct (leukemia-specific) alloreactive donor T-cell populations.14-16 Moreover, allo–major histocompatibility complex (MHC)–restricted cytotoxic T lymphocytes (CTLs) to antigens frequently expressed at elevated levels on leukemic cells can be cloned from an allorestricted T-cell repertoire and used to mediate a GvL reaction without causing GvHD.17-19 However, immunotherapy with ex vivo–generated specific T cells requires the identification and characterization of tumor-specific antigens to be targeted. To circumvent this prerequisite of cloning leukemia-specific T cells, we aimed to develop a complementing strategy that would allow ex vivo elimination of alloreactive T cells responsible for GvHD while preserving an allorestricted T-cell population able to both mediate GvL and antiviral immunity effects. Because the activation-induced cell death (AICD) mediated by CD95/CD95 ligand (CD95L) (reviewed by Lenardo et al20) represents an important physiologic pathway to control the expansion of antigen-activated T cells at the down phase of an immune response, it therefore might provide a means to selectively deplete activated alloreactive T lymphocytes. Induction of AICD triggered by repetitive T-cell receptor (TCR) re-engagement in the presence of interleukin-2 requires antigen-specific activation of the T cell and appears not to affect bystander T cells.21-23Thus, inducing AICD in alloreactive T lymphocytes by repetitive stimulation with alloantigen ex vivo prior to transfer should allow us to leave T cells unaffected that confer antiviral or GvL effects.

In this study we tested this approach by the induction of apoptosis in activated alloreactive T lymphocytes in an allogeneic mixed lymphocyte culture (MLC) using an agonistic monoclonal antibody (mAb) to CD95 followed by transfer of residual allogeneic T cells in a lethal P to F1(P→F1) GvHD model system (H-2k→H-2dxk). We report here that proliferative and lytic alloreactive responses of donor T cells can be substantially decreased by AICD specifically to a given alloantigen. In addition, depletion of activated alloreactive T cells prior to adoptive transfer of allogeneic T cells into F1 recipients prevents the occurrence of lethal GvHD, which invariably occurred in untreated controls.

The results presented suggest that elimination of alloreactive donor T lymphocytes following activation by CD95 death receptor–mediated depletion might provide one approach to modify the T-cell repertoire in allogeneic transplants and complement strategies to include pathogen-specific T lymphocytes and in vitro–generated tumor-specific T cells for immunotherapy in an allogeneic BMT.

Mice and tumor cell lines

C3H/HeJ (H-2k) mice, BALB/cJ (H-2d) mice, (C3H/HeJ × BALB/cJ) F1 (H-2kxd) hybrid mice, and BALB/cJ OVA323-339-specific TCR transgenic mice originally obtained from Dr Loh and colleagues24 were bred and maintained at the Central Animal Facility of the University of Mainz. All mice strains were subjected to regular health status controls and were free of commonly tested viruses, parasites, and bacteria.

The animals were housed in sterilized microisolator cages with filter tops, and experimental procedures were performed according to German federal and state regulations and the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

The age of mice to be used for experiments ranged from 8 to 14 weeks. For adoptive transfer experiments, only female F1 mice were taken as recipients.

The A20 tumor cell line represents a B-cell lymphoma derived from BALB/c mice. The thymoma cell line EL-4 was derived from C57BL/6 (H-2b) mice.

Media and reagents

The tumor cell lines A20 and EL-4 were propagated in RPMI 1640 medium (Biomed, Berlin, Germany) supplemented with 5% fetal calf serum (FCS) (Gibco, Ichinnan, United Kingdom), 5 mM/Ll-glutamine, 0.1 mM β-mercaptoethanol, 1% antibiotic-antimycotic (Gibco) solution consisting of penicillin (10 000 IU)/streptomycin (10 mg/mL)/amphotericin B (25 μg/mL), 1% sodium bicarbonate, 1 mM/L sodium pyruvate, and 0.8% nonessential amino acids, and maintained in flasks during culture. Preparation of cell suspensions made from ex vivo–isolated cells was conducted in Dulbecco modified Eagle medium (DMEM) (Biomed) containing 2% FCS or horse serum (Gibco) and 1% antibiotic-antimycotic (Gibco). Culture medium for mixed lymphocyte reactions (MLC medium) consisted of RPMI 1640 (Biomed) supplemented with 10% FCS (Gibco) and 5% NCTC (Gibco) in addition to supplements used for cell culture medium.

Biotin-conjugated antibodies to H-2d and H-2k, fluorescein isothiocyanate (FITC)–labeled anti-CD3 and anti-CD4 antibodies, phycoerythrin-conjugated anti-CD8 antibody, and appropriate isotype controls were purchased from BD Biosciences (San Diego, CA). FITC- and phycoerythrin-labeled streptavidin was from Jackson ImmunoResearch (West Grove, PA). MicroBeads used for immunomagnetic separation of T cells were from Miltenyi (Bergisch Gladbach, Germany). The anti CD3 (145-2C11) mAb and the anti-CD95 antibody (Jo2) were obtained in a low endotoxin and culture grade from BD Biosciences; isotype-matched hamster immunoglobulin (Ig) and polyclonal goat anti–hamster Ig were purchased from Serotec (Oxford, United Kingdom). Protein G used for cross-linking of Jo2 mAb was from Sigma (Deisenhofen, Germany). The ovalbumin (OVA) peptide (amino acids 323-339 from chicken ovalbumin) was obtained from the Department of Immunohematology, Leiden University Medical Center, The Netherlands.

Induction of AICD in MLC assays

Single-cell suspensions from spleen or a mixture of spleen and lymph node cells derived from inguinal, axillary, and submandibullary lymph nodes were prepared in complete DMEM followed by red cell lysis using NH4Cl treatment. Purification of CD90+, CD4+, or CD8+ responder T cells from C3H/HeJ or BALB/cJ mice was carried out using a magnetic cell separation system (MACS) according to the manufacturer's instructions (Miltenyi). Cells were incubated with either anti-CD90–, anti-CD4–, or anti-CD8–conjugated MicroBeads in MACS buffer (phosphate-buffered saline [PBS] containing 0.5% FCS) for 20 minutes at 4°C. After removal of excess beads by washing, the cell suspension was passed through a prewashed VS+ separation column placed in a magnetic field, and cells coated with MicroBeads were selected to be transferred into MLC medium. Preparations of T cells were periodically stained with anti-CD90, anti-CD4, or anti-CD8 antibody to be analyzed by flow cytometry. Purity of the enriched T cells always exceeded 95%.

Highly purified ex vivo–isolated naive CD4+, CD8+, or CD90+ responder T cells were mixed with irradiated (30 Gy [3000 cGy]) splenic stimulator cells from BALB/c mice (H-2d) or F1 mice (H-2dxk) at concentrations of 1 × 106/mL and 5 × 106/mL, respectively. They were cultured for 5 to 6 days in MLC medium in 96-well round-bottom plates in the absence or presence of various concentrations of soluble agonistic anti-CD95 mAb (Jo2). Depletion of alloreactive T cells for adoptive transfer studies was performed in a bulk culture in 24-well plates generally culturing 2.5 × 106 responder cells with 7.5 × 106irradiated stimulator cells. To enhance its proapoptotic effect, microtiter plate wells were also coated with anti–hamster Ig at a concentration of 5 μg/mL in PBS prior to addition of anti-CD95 antibody to facilitate cross-linking. Alternatively, protein G was used at 2 μg/mL. In case of a secondary MLC, responder T cells were either prestimulated in vitro with irradiated allogeneic splenic stimulators or immobilized with anti-CD3 antibody in bulk cultures for 3 days or in vivo by intraperitoneal injection of 5 × 106 irradiated BALB/c spleen cells 10 to 14 days prior to isolation. T-cell blasts were then harvested from culture or isolated from spleen and lymph node suspension by immunomagnetic separation using MACS as described above to be restimulated in a bulk culture MLC.

When OVA-specific T cells (H-2d) were used to evaluate the specificity of depletion, CD4+ cells were isolated from OVA-TCR transgenic mice by MACS technology as described above and mixed with BALB/c T-cell responder at ratios of 1:1, 1:5, or 1:10 to be cultured with irradiated allogeneic F1 splenic stimulators in the presence of Jo2 mAb. Residual T cells were then not only challenged with F1 cells but also stimulated with these F1 cells in the presence of OVA peptide (0.5 μg/mL).

Depending on a primary or secondary MLC or T-cell stimulation by peptide antigen, 2 or 3 days after beginning the culture the cells were harvested every day following an overnight pulse with 1 μCi (0.037 MBq) per well of 3H-thymidine. Radioactivity incorporated into DNA was measured on a β-plate liquid scintillation counter (Wallace, Turku, Finland).

Cytolytic assays

A total of 2 × 106 A20 (H-2d) or EL-4 (H-2b) tumor cells was labeled with 100 μCi (3.7 MBq) of51Cr for 4 hours. After washing 3 times in PBS, labeled targets were plated at 1 × 104 cells per well in U-bottom microtiter plates (Nunc, Wiesbaden, Germany). Allogeneic CD90+ responder T cells (H-2k), preactivated in a primary MLC to H-2d spleen cells for 5 days, were washed 3 times and added to triplicate wells at varying effector-target ratios to be incubated for 5 hours. Maximum and spontaneous release were determined by incubation of targets with Triton X-100 (Sigma) or addition of media, respectively. 51Cr activity in the supernatants taken 5 hours later was determined in an auto γ counter (Packard Instruments, Dreieich, Germany), and lysis was expressed as a percentage of maximum release.

Experimental model for induction of lethal GvHD

Acute and lethal GvHD was induced in a classical P→F1 model using C3H/HeJ mice (H-2k) or BALB/cJ mice (H-2d) as donor of T cells and (C3H/He × BALB/c) F1 mice (H-2kxd) as recipients. Following depletion of alloreactive T cells in bulk culture MLC in the presence of agonistic anti-CD95 antibody for 5 to 6 days as described above, remaining T cells were harvested to be purified by immunomagnetic separation on a MACS column. Single suspensions of highly purified in vitro–activated viable (trypan blue–excluding) CD4+ or CD8+ T cells were then injected at various concentrations through the tail vein into female F1 recipients on day 0. Mice were sublethally total body137Cs-irradiated (6.5 Gy [650 cGy]) 4 to 5 hours prior to adoptive transfer of T cells. Control mice consisted of age- and sex-matched F1 mice that received syngeneic spleen cells or untreated allogeneic T cells, respectively.

For BMT studies, BM was obtained aseptically by flushing femurs and tibias of C3H/HeJ donor mice with complete DMEM, and T-cell depletion was carried out by immunomagnetic removal of CD90+ cells using MACS (Miltenyi) as described above. T-cell–depleted BM cells (2 × 107 cells per mouse) were transplanted with highly purified CD4+ or CD8+ donor T cells or T cells depleted of alloreactivity by AICD (2.5 × 106 per animal), respectively, by tail-vein infusion into lethally irradiated F1 recipients on day 0. F1 mice received 13 Gy (1300 cGy) total body irradiation as a split dose (with 3 hours between the 2 doses to reduce gastrointestinal toxicity) 3 to 4 hours prior to BMT.

Recipient mice were monitored for clinical signs of GvHD, including diarrhea, alopecia, and hunched posture, and their weight loss was determined. At least 4 recipient mice were included in each experimental group, and adoptive transfer was repeated at least 3 times.

Histology of GvHD and immunohistochemistry for CD4 and CD8 antigen expression in tissue-infiltrating lymphocytes

Liver, spleen, lymph node, small intestine, kidney, and ear skin sections obtained from mice on various days after adoptive transfer of untreated donor T cells or donor T lymphocytes depleted of alloreactive responders by AICD were either fixed in 10% buffered formalin and embedded in paraffin to be stained with hematoxylin and eosin for microscopic examination or embedded with OCT medium (Tissue-Tek, Zoeterwoude, The Netherlands) to be stored at −80°C for immunohistochemistry. Slides of cryostat-sliced (5 μm) samples were first incubated with rat anti-CD8 mAb at 5 μg/mL (BD Biosciences) or rat anti-CD4 mAb at 2.5 μg/mL (BD Biosciences) overnight at 4°C, followed by rinsing in PBS and incubation with a secondary antibody (0.01 mg/mL biotinylated anti–rat IgG) (Dako, Hamburg, Germany) at room temperature for 30 minutes. The antigen-antibody binding was detected using an avidin-biotin alkaline phosphatase (AP) system (Dako). The slides were immersed briefly in hematoxylin for counterstaining and evaluated under light microscopy.

Flow cytometry analysis

Generally, 1 × 106 viable T cells per sample, isolated from spleen and lymph node preparations by immunomagnetic separation using CD4-, CD8-, or CD90-conjugated MicroBeads (Miltenyi), were incubated for 15 minutes at 4°C with the indicated antibodies. The antibodies were either directly conjugated or biotinylated to be incubated with streptavidin-conjugated FITC or phycoerythrin as second-step reagents, followed by 2 washes (PBS containing 2.5% FCS and 1% sodium azide) between incubations. Viable lymphocytes were gated either by forward and side scatter or on a life gate using 7–aminoactinomycin D (Molecular Probes, Leiden, The Netherlands), and 2-color fluorescence analysis was performed on 10 000 cells using an Epics Altra instrument (Beckman Coulter, Stanford, CA) with Expo 32 software.

Statistics

All values are expressed as mean ± SD. Statistical analysis of survival data was performed by the Mantel-Cox log-rank test.P < .05 was considered statistically significant.

Proliferative responses of alloreactive T cells are strongly reduced after induction of AICD by agonistic mAb to CD95

Because both CD4+ and CD8+ T cells undergo CD95-mediated activation-induced apoptosis after prolonged activation,20-23 we investigated the possibility of deploying AICD as a method for selective depletion of alloreactive T cells in an allogeneic mixed lymphocyte reaction. Naive T cells isolated ex vivo from C3H/He (H-2k) mice were highly purified into CD4+ or CD8+ T-cell subpopulations by immunomagnetic separation using the MACS technology and cultured together with irradiated unfractionated splenic stimulator cells from BALB/c (H-2d) mice in the presence of increasing concentrations of soluble agonistic mAb (Jo2) to CD95 for 5 to 6 days. Jo2 has previously been shown to induce apoptosis in lymphocytes.25 Following stimulation to the H-2d alloantigen, the proliferative responses of both CD4+ or CD8+ alloreactive T lymphocytes were reduced by up to 90% in the presence of anti-CD95 mAb in a dose-dependent fashion when compared with the proliferation of untreated allogeneic controls (Figure 1). Reduction of proliferative responses was specific for anti-CD95–mediated apoptosis of T cells, because responder cells stimulated in the presence of an anti-CD95 mAb-matched hamster Ig isotype control showed comparable responses to untreated allogeneic controls (Figure 1). Flow cytometry analyses of CD90+responders isolated at daily time points during stimulation with T-cell–depleted allogeneic stimulators (H-2d) in a 6-day bulk culture by immunomagnetic separation revealed an increased CD95 expression within the first 36 to 48 hours following activation and an increase of apoptotic CD4+ and CD8+ T lymphocytes upon treatment with agonistic anti-CD95 mAb as measured by annexin V and 7–aminoactinomycin D viability staining.

Fig. 1.

Reduced proliferative responses of alloreactive T cells upon induction of AICD by agonistic anti-CD95 mAb.

Strong reduction of alloreactivity by CD4+ and CD8+ responders upon induction of CD95-mediated AICD by agonistic mAb to CD95 in a dose-dependent fashion. Naive T cells from C3H/He (H-2k) mice were purified into CD4+ and CD8+ subpopulations by immunomagnetic separation using MACS. Following stimulation with irradiated allogeneic splenic stimulators from BALB/c (H-2d) in an MLC over 6 days in the absence (▪) or presence of 1 μg/mL (●) or 5 ng/mL (♦) of agonistic anti-CD95 mAb (Jo2), proliferation of T cells was determined by 3H-thymidine uptake for the last 12 hours. Addition of a 1 μg/mL isotype-matched hamster-Ig control antibody (○) during culture was performed as specificity control. Syngeneic controls (■) are shown without induction of AICD. Results are expressed as mean ± SD of triplicate wells and depict 1 of at least 5 representative experiments. (A) Proliferative response by allogeneic CD4+responders. (B) Proliferative response by allogeneic CD8+T cells.

Fig. 1.

Reduced proliferative responses of alloreactive T cells upon induction of AICD by agonistic anti-CD95 mAb.

Strong reduction of alloreactivity by CD4+ and CD8+ responders upon induction of CD95-mediated AICD by agonistic mAb to CD95 in a dose-dependent fashion. Naive T cells from C3H/He (H-2k) mice were purified into CD4+ and CD8+ subpopulations by immunomagnetic separation using MACS. Following stimulation with irradiated allogeneic splenic stimulators from BALB/c (H-2d) in an MLC over 6 days in the absence (▪) or presence of 1 μg/mL (●) or 5 ng/mL (♦) of agonistic anti-CD95 mAb (Jo2), proliferation of T cells was determined by 3H-thymidine uptake for the last 12 hours. Addition of a 1 μg/mL isotype-matched hamster-Ig control antibody (○) during culture was performed as specificity control. Syngeneic controls (■) are shown without induction of AICD. Results are expressed as mean ± SD of triplicate wells and depict 1 of at least 5 representative experiments. (A) Proliferative response by allogeneic CD4+responders. (B) Proliferative response by allogeneic CD8+T cells.

Close modal

Both naive and preactivated T lymphocytes are equally susceptible to AICD

Because T cells have been reported to be initially resistant to AICD upon primary activation but elicit increasing susceptibility to CD95-mediated induction of apoptosis 4 to 5 days20-23 upon sustained TCR re-engagement, we further investigated the induction of AICD on naive versus preactivated T cells.

Following polyclonal activation in vitro by TCR cross-linking through coated anti-CD3 mAb (145-2C11) for 3 days, CD4+ T-cell blasts as well as freshly isolated resting CD4+ T lymphocyte mice were stimulated with alloantigen (H-2d) in a bulk culture MLC in the presence of agonistic mAb to CD95 for 5 days to be tested for residual response to the stimulating alloantigen in a mixed lymphocyte reaction. Upon induction of AICD, both primary stimulated (Figure 2A) or restimulated (Figure 2B) T-cell responders showed a marked decrease in their capacity to proliferate to allogeneic splenic stimulators regardless of their preactivation state (note the reduced proliferation of 85% both in Figure 2A and 2B on day 3) in contrast to their untreated responder controls, although with an earlier kinetic in the response to H-2d for anti-CD3–preactivated cells.

Fig. 2.

Both resting and preactivated T lymphocytes are susceptible for AICD.

Strongly decreased proliferation of responders after induction of AICD independent of whether alloreactive T cells had been subjected to anti-CD95 treatment during primary or secondary stimulation. Small resting CD4+ T cells (H-2k) or CD4+T cells preactivated by TCR cross-linking using anti-CD3 mAb (145-2C11) for 3 days were stimulated with irradiated allogeneic splenic stimulators (H-2d) in bulk culture in the presence of agonistic anti-CD95 mAb for 5 days. Residual T cells collected from these cultures were then tested for alloreactivity to the H-2d alloantigen in a 5-day MLC. Results are expressed as mean ± SD of triplicate wells and show 1 of at least 3 representative experiments. (A) Alloreactivity of responders primarily stimulated to H-2d alone (▪) or in the presence of anti-CD95 mAb (●). (B) Alloreactivity of anti-CD3–preactivated responders after restimulation to H-2d alone (▪) or in the presence of 1 μg/mL anti-CD95 mAb (●). Syngeneic controls are shown without (■) or with (○) induction of AICD.

Fig. 2.

Both resting and preactivated T lymphocytes are susceptible for AICD.

Strongly decreased proliferation of responders after induction of AICD independent of whether alloreactive T cells had been subjected to anti-CD95 treatment during primary or secondary stimulation. Small resting CD4+ T cells (H-2k) or CD4+T cells preactivated by TCR cross-linking using anti-CD3 mAb (145-2C11) for 3 days were stimulated with irradiated allogeneic splenic stimulators (H-2d) in bulk culture in the presence of agonistic anti-CD95 mAb for 5 days. Residual T cells collected from these cultures were then tested for alloreactivity to the H-2d alloantigen in a 5-day MLC. Results are expressed as mean ± SD of triplicate wells and show 1 of at least 3 representative experiments. (A) Alloreactivity of responders primarily stimulated to H-2d alone (▪) or in the presence of anti-CD95 mAb (●). (B) Alloreactivity of anti-CD3–preactivated responders after restimulation to H-2d alone (▪) or in the presence of 1 μg/mL anti-CD95 mAb (●). Syngeneic controls are shown without (■) or with (○) induction of AICD.

Close modal

These results suggested that induction of AICD at least in an allogeneic situation upon repetitive stimulation of T lymphocytes with MHC alloantigen was not dependent on the preactivation state of the alloreactive T cells.

Induction of AICD in alloreactive T cells is specific for the stimulating alloantigen

To evaluate whether alloantigen-specific T-cell depletion by AICD would retain T-cell specificities to defined nonalloantigens, we chose to add I-Ad-restricted OVA323-339-specific CD4+ T cells isolated from BALB/c OVA-TCR transgenic mice24 to allogeneic responder T cells from BALB/c mice (H-2d) at different ratios. These T cells should not be activated in the absence of OVA and thus not be susceptible to AICD. Following culture with splenic stimulators from F1 mice (H-2dxk) in the presence of cross-linked anti-CD95 mAb, remaining responder T cells were then challenged with F1stimulators as well as with BALB/c-derived spleen cells presenting OVA peptide.

As expected from the above findings, restimulation to H-2kalloantigen in vitro resulted in an impaired proliferative response by these T cells compared with untreated controls (Figure3A, in vitro). The proliferation of untreated T cells to H-2k observed in Figure 3 was truly allospecific because T cells from the OVA transgenic mice were shown not to cross-react with H-2kxd (data not shown). In contrast, proliferation of the responder cells to OVA was preserved in both anti-CD95–treated and untreated T-cell responder populations (Figure 3A, in vitro).

Fig. 3.

Depletion of alloreactive T cells by AICD to the H-2d alloantigen is specific.

Purified CD4+ responder T cells from BALB/c mice (H-2d) mixed with OVA-specific CD4+ T cells derived from BALB/c (H-2d) at a 1:1 ratio were stimulated with spleen cells from F1 mice (H-2dxk) in the absence or presence of anti-CD95 mAb (1 μg/mL) cross-linked by protein G (2 μg/mL) in a bulk MLC for 5 days. Remaining T cells were then challenged in vitro (A) with syngeneic spleen cells (▪), H-2kxd alloantigen (░), or with H-2dstimulators presenting OVA peptide (▨). In addition, remaining T-cell responders from primary cultures were transferred into irradiated F1 recipients. On day 6 after transfer, splenic responders recovered from these recipients ex vivo (B) were tested for reactivity to syngeneic spleen cells (▪), H-2kxd (░), as well as to OVA peptide (▨). Shown are stimulation indices from cultures harvested on day 6. Results are expressed as mean ± SD of triplicate wells and represent 1 of 2 reproducible experiments. (A) In vitro restimulation of responder T cells depleted of alloreactive cells by AICD during primary MLC resulted in a decreased reactivity to H-2dxk compared with allogeneic T cells left untreated. However, proliferative responses of OVA-specific T cells stimulated with OVA peptide were comparable between anti-CD95–pretreated T cells and untreated controls. (B) Alloreactivity of responders isolated ex vivo after adoptive transfer of pretreated T lymphocytes to H-2dxk was strongly impaired in contrast to untreated controls. Again, specific and comparable OVA responses could be measured in both responder populations regardless of initial depletion by AICD or no treatment.

Fig. 3.

Depletion of alloreactive T cells by AICD to the H-2d alloantigen is specific.

Purified CD4+ responder T cells from BALB/c mice (H-2d) mixed with OVA-specific CD4+ T cells derived from BALB/c (H-2d) at a 1:1 ratio were stimulated with spleen cells from F1 mice (H-2dxk) in the absence or presence of anti-CD95 mAb (1 μg/mL) cross-linked by protein G (2 μg/mL) in a bulk MLC for 5 days. Remaining T cells were then challenged in vitro (A) with syngeneic spleen cells (▪), H-2kxd alloantigen (░), or with H-2dstimulators presenting OVA peptide (▨). In addition, remaining T-cell responders from primary cultures were transferred into irradiated F1 recipients. On day 6 after transfer, splenic responders recovered from these recipients ex vivo (B) were tested for reactivity to syngeneic spleen cells (▪), H-2kxd (░), as well as to OVA peptide (▨). Shown are stimulation indices from cultures harvested on day 6. Results are expressed as mean ± SD of triplicate wells and represent 1 of 2 reproducible experiments. (A) In vitro restimulation of responder T cells depleted of alloreactive cells by AICD during primary MLC resulted in a decreased reactivity to H-2dxk compared with allogeneic T cells left untreated. However, proliferative responses of OVA-specific T cells stimulated with OVA peptide were comparable between anti-CD95–pretreated T cells and untreated controls. (B) Alloreactivity of responders isolated ex vivo after adoptive transfer of pretreated T lymphocytes to H-2dxk was strongly impaired in contrast to untreated controls. Again, specific and comparable OVA responses could be measured in both responder populations regardless of initial depletion by AICD or no treatment.

Close modal

In addition, challenge of purified CD4+ and CD8+ T cells isolated ex vivo from F1recipients 6 days after adoptive transfer of a mixture of OVA-specific T cells and allogeneic T-cell responders of BALB/c donors either left untreated or depleted of alloreactive cells by AICD into irradiated F1 mice confirmed the findings obtained in vitro. Again, the response of pretreated responders to H-2k alloantigen was reduced in contrast to untreated controls (Figure 3B, ex vivo), but stimulation of OVA-specific T cells by OVA peptide–presenting BALB/c spleen cells appeared not to be significantly different (Figure 3B, ex vivo) in both responder populations.

These results strongly suggested that induction of AICD using the agonistic mAb Jo2 to CD95 was specific in depleting alloreactive T cells to a given alloantigen while retaining T cells specific for non-MHC antigens like OVA.

Abrogation of cytolytic responses of alloreactive T cells after induction of AICD

We further tested whether induction of AICD by anti-CD95 mAb would also reduce the cytolytic responses of alloreactive T cells. Following stimulation of purified CD90+ responder T cells from C3H/He mice with irradiated allogeneic splenic stimulators from BALB/c mice in the presence of anti-CD95 mAb (1 μg/mL) in a bulk culture MLC for 5 days, remaining T lymphocytes were recovered from culture after treatment and tested in a cytolytic assay using chromium-labeled A20 cells as targets. The capacity of residual CTLs to kill A20 targets was reduced by at least 80% compared with untreated alloreactive control T cells, which exhibited 50% to 60% specific lysis at an effector-target ratio of 3:1 (Figure 4). This reduction in alloreactive cytoxicity after induction of AICD was again specific for the alloantigen H-2d because there was no significant lysis of EL-4 target cells expressing H-2b(Figure 4). Taken together, these findings and the results presented above strongly suggested that the induction of alloantigen-dependent apoptosis in an activated allogeneic T-cell pool by administration of a CD95 agonistic mAb results in marked depletion of alloreactive T cells to a given alloantigen without grossly affecting T-cell specificities that have not previously been activated in an alloantigen-dependent fashion.

Fig. 4.

Cytolytic response of alloreactive T cells after induction of AICD.

A representative experiment showing impaired capacity of allogeneic CTLs to lyse H-2d–expressing A20 target cells after depletion of alloreactive T cells to H-2d by AICD (●) in comparison to untreated activated allogeneic control cells (▪). Lysis of A20 was antigen specific because H-2b EL-4 target cells were not recognized by untreated alloreactive CTLs (■) or anti-CD95–treated CTLs (○). CTLs were generated by allogeneic stimulation of purified CD90+ T cells (H-2k) with splenic stimulator cells (H-2d) in the presence of anti-CD95 in bulk culture MLC for 5 days and recovered cells incubated with chromium-labeled A20 or EL-4 target cells for 5 hours to be analyzed for their residual lytic capacity to H-2d.

Fig. 4.

Cytolytic response of alloreactive T cells after induction of AICD.

A representative experiment showing impaired capacity of allogeneic CTLs to lyse H-2d–expressing A20 target cells after depletion of alloreactive T cells to H-2d by AICD (●) in comparison to untreated activated allogeneic control cells (▪). Lysis of A20 was antigen specific because H-2b EL-4 target cells were not recognized by untreated alloreactive CTLs (■) or anti-CD95–treated CTLs (○). CTLs were generated by allogeneic stimulation of purified CD90+ T cells (H-2k) with splenic stimulator cells (H-2d) in the presence of anti-CD95 in bulk culture MLC for 5 days and recovered cells incubated with chromium-labeled A20 or EL-4 target cells for 5 hours to be analyzed for their residual lytic capacity to H-2d.

Close modal

Recipients of allogeneic T cells depleted from host reactivity by AICD do not develop GvHD

To verify the efficacy of depleting alloreactive T cells by AICD in vitro, we investigated the potential of residual T cells to evoke a GvHD in the parent C3H/He (H-2k)→F1(BALB/c × C3H/He, H-2dxk) murine GvHD model.

Naive allogeneic CD90+ donor T lymphocytes or CD90+ T cells primed to H-2d BALB/c spleen cells in vivo 14 days prior to isolation from donor mice were cultured with irradiated BALB/c-derived splenic stimulators in bulk MLC in the presence of Jo2 mAb for 5 to 6 days. Remaining responders were separated into CD4+ or CD8+ donor T cells by immunomagnetic separation to be injected into the tail vein of F1 recipients irradiated (6.5 Gy [650 cGy]) within 6 to 8 hours prior to transfer. During the preparation of T-cell responders, extensive care was taken to wash out excessive anti-CD95 mAb. Furthermore, viability and purity of cell preparations was assessed by flow cytometry as described in “Materials and methods.” A dose response of donor T cells depleted of alloreactivity or left untreated was performed, and F1 mice that received 1 × 106 to 5 × 106 in vivo–primed and in vitro–restimulated donor T lymphocytes invariably developed an acute GvHD between days 9 to 18 after transfer depending on the amount of T cells injected. GvHD was clinically characterized by rapid weight loss, severe diarrhea, and hunched posture, and all mice died within 20 days after T-cell transfer, as shown for injection of 2.5 × 106 T cells per recipient in Figure5.

Fig. 5.

Depletion of alloreactive T cells by AICD prevents acute GvHD in a murine P→F1 GvHD model.

Depicted are survival curves representing 3 independent experiments in which F1 recipients (n = 10) received 2.5 × 106 purified viable parental donor T lymphocytes. T cells from C3H/He mice (H-2k) previously primed to H-2d alloantigen in vivo were restimulated with H-2d–expressing BALB/c stimulator cells and depleted from alloreactivity by AICD. Following adoptive transfer of treated T lymphocytes into irradiated (6.5 Gy [650 cGy]) female F1hosts, recipients did not develop acute GvHD, in contrast to mice injected with untreated allogeneic control cell blasts. (A) Depletion of transferred CD4+ T cells from host reactivity by induction of AICD into F1 recipients prevents development of acute GvHD (▪, P < .05). In contrast, transfer of activated untreated control cells (▾) or donor T lymphocytes depleted of alloreactive responders by soluble anti-CD95 mAb (●) led to rapid and lethal GvHD within 20 days after transfer (P < .05). Controls shown received equivalent numbers of syngeneic spleen cells (■). (B) The same experiments as described in panel A but performed with allogeneic CD8+ T cells are presented.

Fig. 5.

Depletion of alloreactive T cells by AICD prevents acute GvHD in a murine P→F1 GvHD model.

Depicted are survival curves representing 3 independent experiments in which F1 recipients (n = 10) received 2.5 × 106 purified viable parental donor T lymphocytes. T cells from C3H/He mice (H-2k) previously primed to H-2d alloantigen in vivo were restimulated with H-2d–expressing BALB/c stimulator cells and depleted from alloreactivity by AICD. Following adoptive transfer of treated T lymphocytes into irradiated (6.5 Gy [650 cGy]) female F1hosts, recipients did not develop acute GvHD, in contrast to mice injected with untreated allogeneic control cell blasts. (A) Depletion of transferred CD4+ T cells from host reactivity by induction of AICD into F1 recipients prevents development of acute GvHD (▪, P < .05). In contrast, transfer of activated untreated control cells (▾) or donor T lymphocytes depleted of alloreactive responders by soluble anti-CD95 mAb (●) led to rapid and lethal GvHD within 20 days after transfer (P < .05). Controls shown received equivalent numbers of syngeneic spleen cells (■). (B) The same experiments as described in panel A but performed with allogeneic CD8+ T cells are presented.

Close modal

Surprisingly, adoptive transfer of donor T lymphocytes depleted of alloreactive donor cells by soluble mAb to CD95 alone also led to the development of lethal GvHD (Figure 5). However, depletion of activated alloreactive T cells using anti-CD95 mAb cross-linked by polyclonal anti–hamster Ig prior to adoptive transfer of purified CD4+ (Figure 5A) or CD8+ (Figure 5B) allogeneic T cells resulted in the amelioration of acute GvHD in F1recipients. In this experimental group all recipient mice survived and were free of clinical GvHD, as did control mice that received syngeneic spleen cells. Prevention of GvHD was stable and lasted until day 120, when the monitoring was terminated.

In recent experiments we also used this P→F1 model for allogeneic BMT in which 2 × 107 T-cell–depleted BM cells of H-2k origin were transplanted together with 2.5 × 106 purified allogeneic (H-2k) CD90+ T cells or T cells subjected to anti-CD95 treatment prior to transfer. These studies appear to confirm the results described above because F1 recipients that received pretreated donor T lymphocytes did not develop GvHD, in contrast to F1 BMT controls.

Histopathological examination of sections taken from skin, small intestine, and spleen at different time points after adoptive transfer confirmed the survival data: apoptotic cells in the basal layer of epidermis and inflammation in the cryptic epithelium within the small intestine due to infiltrating lymphocytes were clearly detectable in mice injected with unmodified donor T cells, in contrast to F1 recipients that had received T cells depleted of alloreactive lymphocytes (Figure 6). Moreover, immunohistologic staining of spleen showed expansion of CD4+ T cells and, albeit less extensive, of CD8+ T cells in F1 mice after transfer of untreated donor T cells compared with recipients injected with donor T lymphocytes treated by AICD (Figure 6).

Fig. 6.

Histologic examination of GvHD.

Induction of lethal acute GvH reactions after adoptive transfer of allogeneic donor T lymphocytes or donor T cells depleted of alloreactivity by AICD was examined histologically and by immunohistology as described in “Materials and methods.” On day 6 after transfer, 2 mice from each group were killed and tissue from skin, liver, spleen, small intestine, and lymph nodes was fixed or embedded. Depicted are sections of skin (A) and small intestine (B) (magnifications of 100 μm and 200 μm) stained by hematoxylin and eosin, which show cells in the basal layer of the epidermis undergoing apoptosis and lymphocytic infiltrations into the cryptic epithelium of small intestine following adoptive transfer of allogeneic donor cells. In addition, immunohistology (shown in red) revealed massive expansion of CD4+ T cells and, albeit to a lesser extent, of CD8+ T cells in spleen (C) isolated from F1recipients that had received untreated allogeneic donor T cells. In contrast, no significant GvHD histopathology could be detected in recipients administered donor T lymphocytes subjected to AICD prior to transfer.

Fig. 6.

Histologic examination of GvHD.

Induction of lethal acute GvH reactions after adoptive transfer of allogeneic donor T lymphocytes or donor T cells depleted of alloreactivity by AICD was examined histologically and by immunohistology as described in “Materials and methods.” On day 6 after transfer, 2 mice from each group were killed and tissue from skin, liver, spleen, small intestine, and lymph nodes was fixed or embedded. Depicted are sections of skin (A) and small intestine (B) (magnifications of 100 μm and 200 μm) stained by hematoxylin and eosin, which show cells in the basal layer of the epidermis undergoing apoptosis and lymphocytic infiltrations into the cryptic epithelium of small intestine following adoptive transfer of allogeneic donor cells. In addition, immunohistology (shown in red) revealed massive expansion of CD4+ T cells and, albeit to a lesser extent, of CD8+ T cells in spleen (C) isolated from F1recipients that had received untreated allogeneic donor T cells. In contrast, no significant GvHD histopathology could be detected in recipients administered donor T lymphocytes subjected to AICD prior to transfer.

Close modal

To monitor homing and GvH reactions of alloantigen-activated T cells after adoptive transfer into F1 hosts, the presence and expansion of adoptively transferred donor T cells in vivo was evaluated. Following immunomagnetic separation of CD90+cells from single spleen and lymph node cell suspensions of F1 recipients at various time points after adoptive transfer, the expansion of H-2k–positive T cells versus recipient-derived T lymphocytes (H-2kxd) isolated from the pool of CD90+ cells was analyzed by 2-color flow cytometry (Figure 7). Proliferation of allogeneic T cells (H-2k) depleted from alloreactive responders in vitro by AICD was very low in F1 recipients at day 10 after initial transfer of 2.5 × 106 cells (Figure 7B; 6.4% for CD4+ and 4.5% for CD8+, respectively). In contrast, untreated allogeneic T lymphocytes from C3H/He mice were significantly expanded (Figure 7A; 33.4% for CD4+ and 22.8% for CD8+, respectively) from the same number of cells injected.

Fig. 7.

Reduced in vivo expression of donor T cells depleted from host reactivity by AICD.

Shown by flow cytometry analysis is a very low expansion of allogeneic CD4+ or CD8+ T cells (H-2k) depleted of alloreactive responders (B) in contrast to significantly increased populations of untreated allogeneic donor CD4+and CD8+ T cells (A) in F1 hosts 10 days after transfer. Monitoring the expansion of donor-derived T cells was performed on CD4+ and CD8+ T cells isolated from spleen and lymph node suspension by immunomagnetic separation and stained for H-2kk and H-2Dd to discriminate single-positive donor-derived (H-2k) T cells from F1-specific (H-2kxd) Tlymphocytes.

Fig. 7.

Reduced in vivo expression of donor T cells depleted from host reactivity by AICD.

Shown by flow cytometry analysis is a very low expansion of allogeneic CD4+ or CD8+ T cells (H-2k) depleted of alloreactive responders (B) in contrast to significantly increased populations of untreated allogeneic donor CD4+and CD8+ T cells (A) in F1 hosts 10 days after transfer. Monitoring the expansion of donor-derived T cells was performed on CD4+ and CD8+ T cells isolated from spleen and lymph node suspension by immunomagnetic separation and stained for H-2kk and H-2Dd to discriminate single-positive donor-derived (H-2k) T cells from F1-specific (H-2kxd) Tlymphocytes.

Close modal

The significant incidence of GvHD following transfer of donor T lymphocytes contained in the stem cell graft into recipients remains as a major caveat in allogeneic BMT, although various strategies for the prevention have been proposed.2-9 In particular, T-cell depletion is undoubtedly a very efficient method to prevent GvHD; however, it increases the risk of severe infections caused predominantly by Epstein-Barr virus26 and cytomegalovirus,10 engraftment failure,11 and relapse of leukemia.12,13 An ideal strategy for a specific and individualized immunotherapy would thus aim for selective elimination of host-reactive T lymphocytes but would leave all other T-cell specificities recognizing Epstein-Barr virus and cytomegalovirus or leukemia-restricted (minor) histocompatibility antigens and leukemia antigens unaffected. Various experimental means, including elimination of alloreactive T cells by immunotoxin-conjugated mAbs to CD25 mAb (as first described by Datta et al14 and followed by others8) or via the early T cell activation antigen CD69,27 have been employed. Furthermore, suicide gene transfer into donor T lymphocytes,4,5 induction of anergy by blocking costimulatory signals necessary for optimal T-cell activation,6,7 and the administration of cytokines to modify T-cell effector responses9,28 have been reported. These methods, however, affect either primarily T-cell subopulations6,7 or all T cells regardless of their specificity.4,5 They also appear not to be very effective on the negative selection of alloreactive T cells,14technically quite complex,27 or limited by the development of resistant T lymphocytes transduced with the HSV-Tk suicide gene to ganciclovir.29 Alternatively, the in vitro generation of allorestricted T cells conferring GvL reactivity without causing GvHD has been described.18 19 

We explored the possibility of separating alloreactive T cells from antiviral T-cell specificities and possibly GvL-conferring T lymphocytes using the CD95/CD95L-mediated AICD as a strategy to deplete GVH-reactive T lymphocytes by apoptosis. AICD via the CD95 system has been demonstrated to occur in T-cell hybridomas as well as in human and murine T cells following activation by TCR cross-linking or by antigen and is thought to represent an important mechanism to limit the clonal expansion of activated T cells at the down phase of an immune response.20-22,30,31 AICD also plays a major role in the maintenance of peripheral T cell tolerance.20,32 Mature T cells acquire susceptibility to AICD after prolonged activation.20-22 This process requires repetitive TCR re-engagement by antigen or anti-TCR/CD3 complex antibodies, is dependent on cell cycle progression caused by interleukin-2,23,33 and results in apoptosis of antigen-specific T cells but not of resting T cells.20,22 23 

Using an (MHC) allogeneic model system we first demonstrated the requirements for induction of AICD in vitro. Secondly, we used this physiologic mean to deplete alloreactive T cells from an allogeneic T-cell repertoire. Upon stimulation of purified naive T lymphocytes from C3H/He mice with BALB/c mouse–derived allogeneic splenic stimulators in the presence of an agonistic mAb (Jo2) to CD95 during a primary MLC proliferation of both CD4+- or CD8+-alloreactive T cell was substantially (≥ 80%) decreased in comparison to control allogeneic responses. Reduction of proliferative responses was mediated by the agonistic anti-CD95 mAb Jo2 25 in a dose-dependent fashion. Seemingly, the capacity of CTLs to lyse allogeneic target cells was also strongly impaired after depletion of alloreactive responders by CD95-mediated AICD. Flow cytometry analysis of responder T cells performed at different time points during MLC with T-cell–depleted stimulator cells revealed that activated alloreactive T lymphocytes up-regulated expression of the activation markers CD69 and CD25 as well as CD95 in a time-dependent fashion and became susceptible for AICD when cultured with agonistic anti-CD95 mAb. Induction of AICD upon activation of alloreactive T cells with one particular alloantigen was specific and did not affect I-Ad–restricted T lymphocytes expressing the transgenic αβ-TCR (DO11.10) specific for the OVA323–339 peptide upon titration into the experimental system. Moreover, OVA-specific T cells could be restimulated from F1 recipients after adoptive transfer of a mixture of OVA–T cells and allogeneic responders pretreated by AICD. Thus, depletion of alloreactivity by AICD retained functional T-cell specificities to defined nonalloantigens.

We then established a classical parent (C3H/HeJ or BALB/cJ)→F1 (C3H/HeJ × BALB/c) murine model. P→F1 models have become an invaluable tool in a number of adoptive transfer and BMT studies on GvHD and GvL because recipients cannot evoke any host-versus-graft effects, in contrast to models using 2 different mice strains. Our model also allows us to use CD95L-deficient C3H/gld donor T cells as controls for current studies on the GvL effect of anti-CD95–pretreated T cells.

In line with the results obtained in vitro, we could show by adoptive transfer of donor-derived T lymphocytes that F1 recipients that received allogeneic T cells, depleted from alloreactive responders by AICD in vitro, did not develop a lethal GvHD. In contrast, mice receiving untreated allogeneic T-cell blasts displayed all clinical signs of acute GvHD and died within 20 days after transfer. Recent experiments performed on lethally irradiated F1 mice and receiving BM cells together with pretreated donor T cells also did not yet result in the induction of GvHD compared with controls.

Additionally, histopathological examinations of skin and small intestine sections taken from F1 mice with GvHD revealed apoptotic cells in the basal lamina of epidermis and infiltration of lymphocytes into cryptic epithelium of small intestine. The monitoring of allogeneic donor cells in the host by flow cytometry also supported the findings described, because 5-fold expansion of alloreactive T cells was exclusively seen in mice receiving untreated allogeneic T lymphocytes. In contrast, very low host-reactive cells were detected in mice that had received allogeneic cells depleted of alloreactivity. Moreover, splenic cells isolated from F1 recipients 6 days after adoptive transfer of both anti-CD95–pretreated OVA-specific T cells and donor T cells could respond to OVA peptide, whereas proliferation to H-2k was reduced.

In summary, these results strongly suggest that depletion of alloreactive T lymphocytes by AICD prevents GvHD and retains functional specificities of non-GVH–reactive donor T cells for engraftment.

Although we cannot exclude that a low number of alloreactive T cells might escape induction of AICD because it has been reported that some T cells escape CD95-mediated apoptosis to become memory cells,34 the numbers of remaining alloreactive T cells in our GvHD model appear to be below a clinically relevant threshold. In fact, considering the diverse repertoire of allogeneic T-cell specificities that might confer a GvL effect, it might be conceivable to tolerate a very low number of, for example, defined alloreactive T-cell subpopulations. Because CD4+ T helper 1 (Th-1)–type T cells have been described to be more susceptible to CD95-mediated AICD than their Th-2 type counterparts,35 depletion of alloreactive T cells by AICD also polarizes residual CD4+GVH-reactive T lymphocytes toward a Th-2 type (data not shown), which additionally might protect against clinical manifestation of acute GvHD.36,37 Moreover, because the CD95/CD95L system has been documented by several groups to play an important role in the pathogenesis of GvHD38,39 and blockade of this pathway appears to ameliorate GvHD without impairing GvL effect,40 41 it remains to be elucidated whether the elimination of alloreactive T lymphocytes by the induction of AICD results in the modifications of allogeneic donor T lymphocytes suitable for preserving a GvL effect in the absence of a clinically manifested GvHD.

In conclusion, we have established an (MHC) allogeneic model system that allows elimination of alloreactive T cells by the induction of CD95-mediated AICD. Transfer of a modified allogeneic donor T-lymphocyte repertoire into a P→F1 model demonstrated that depletion of alloreactive T cells was sufficient for preventing a lethal GvHD in F1 recipients. Thus, utilizing AICD for elimination of alloreactivity in allogeneic transplants might represent a suitable strategy for the enrichment of T cells that mediate an antiviral as well as a GvL effect.

Further studies on the phenotypic and functional characterization of the residual allogeneic T lymphocytes as well as their capacity to confer a GvL effect are in progress. They will help us assess the utility of AICD to optimize clinical BMT.

The authors thank K. Kunz, C. Metz, and S. Khan for expert technical assistance and M. Jülch for the fluorescence-activated cell sorter analysis.

Supported by the research grant “Immunological Tumor Therapy by Allogeneic Stem Cell Transplantation” from the German Cancer Society.

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
Ferrara
JL
Cooke
KR
Pan
L
Krenger
W
The immunopathophysiology of acute graft-versus-host-disease.
Stem Cells.
14
1996
473
489
2
Theobald
M
Nierle
T
Bunjes
D
Arnold
R
Heimpel
H
Host-specific interleukin-2-secreting donor T-cell precursors as predictors of acute graft-versus-host disease in bone marrow transplantation between HLA-identical siblings.
N Engl J Med.
327
1992
1613
1617
3
Barrett
AJ
Mavroudis
D
Tisdale
J
et al
T cell-depleted bone marrow transplantation and delayed T cell add-back to control acute GVHD and conserve a graft-versus-leukemia effect.
Bone Marrow Transplant.
21
1998
543
551
4
Cohen
JL
Boyer
O
Salomon
B
et al
Prevention of graft-versus-host disease in mice using a suicide gene expressed in T lymphocytes.
Blood.
89
1997
4636
4645
5
Bonini
C
Ferrari
G
Verzeletti
S
et al
HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia.
Science.
276
1997
1719
1723
6
Guinan
EC
Vasiliki
AB
Neuberg
D
et al
Transplantation of anergic histoincompatible bone marrow allografts.
N Engl J Med.
340
1999
1704
1714
7
Blazar
BR
Taylor
PA
Noelle
RJ
Vallera
DA
CD4+ T cells tolerized ex vivo to host alloantigen by anti-CD40 ligand (CD40L:CD154) antibody lose their graft-versus-host disease lethality capacity but retain nominal antigen responses.
J Clin Invest.
102
1998
473
482
8
Montagna
D
Yvon
E
Calcaterra
V
et al
Depletion of alloreactive T cells by a specific anti-interleukin-2 receptor p55 chain immunotoxin does not impair in vitro antileukemia and antiviral activity.
Blood.
93
1999
3550
3557
9
Yang
YG
Sergio
JJ
Pearson
DA
Szot
GL
Shimizu
A
Sykes
M
Interleukin-12 preserves the graft-versus-leukemia effect of allogeneic CD8+ T cells while inhibiting CD4-dependent graft-versus-host disease in mice.
Blood.
90
1997
4651
4660
10
Li
CR
Greenberg
PD
Gilbert
MJ
et al
Recovery of HLA-restricted cyto-megalovirus (CMV)-specific T cell responses for allogeneic bone marrow transplant: correlation with CMV disease and effect of ganciclovir prophylaxis.
Blood.
83
1994
1971
1979
11
Martin
PJ
Donor CD8+ cells prevent allogeneic marrow graft rejection in mice: potential implications for marrow transplantation in humans.
J Exp Med.
178
1993
703
712
12
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
13
Horowitz
MM
Gale
RP
Sondel
PM
et al
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood.
75
1990
555
562
14
Datta
AR
Barrett
AJ
Jiang
YZ
et al
Distinct T cell populations distinguish chronic myeloid leukemia cells from lymphocytes in the same individual: a model for separating GVHD from GVL reactions.
Bone Marrow Transplant.
14
1994
517
524
15
Jiang
YZ
Mavroudis
DA
Dermine
S
Molldrem
J
Hensel
NF
Barrett
AJ
Preferential usage of T cell receptor (TCR) Vβ by allogeneic T cells recognizing myeloid leukemia cells: implications for separating graft-versus-leukemia effect from graft-versus-host disease.
Bone Marrow Transplant.
19
1997
899
903
16
Falkenburg
JHF
Wafelman
AR
Joosten
P
et al
Complete remission of accelerated phase chronic myeloid leukemia by treatment with leukemia-reactive cytotoxic T lymphocytes.
Blood.
94
1999
1201
1208
17
Molldrem
J
Dermine
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
18
Gao
L
Yang
T-H
Tourdot
S
Sadovnikova
E
Hasserjian
R
Stauss
HJ
Allo-major histocompatibility complex-restricted cytotoxic T lymphocytes engraft in bone marrow transplant recipients without causing graft-versus-host disease.
Blood.
94
1999
2999
3006
19
Gao
L
Bellantuono
I
Elsässer
A
et al
Selective elimination of leukemic CD34+ progenitor cells by cytotoxic T lymphocytes specific for WT-1.
Blood.
95
2000
2198
2203
20
Lenardo
M
Chan
KM
Hornung
F
et al
Mature T lymphocyte apoptosis—immune regulation in a dynamic and unpredictable antigenic environment.
Annu Rev Immunol.
17
1999
221
253
21
Klas
C
Debatin
KM
Jonker
R
Krammer
PH
Activation interferes with the APO-1 pathway in mature human T cells.
Int Immunol.
5
1993
625
630
22
Russell
JH
White
CL
Loh
DY
Meleedy-Rey
P
Receptor-stimulated death pathway is opened by antigen in mature T cells.
Proc Natl Acad Sci U S A.
88
1991
2151
2155
23
Lenardo
MJ
Interleukin-2 programs mouse α β T lymphocytes for apoptosis.
Nature.
353
1991
858
862
24
Murphy
KM
Heimberger
AB
Loh
DY
Induction by antigen of intrathymic apoptosis of CD4+ CD8+ TCRlo thymocytes in vivo.
Science.
250
1990
1720
1723
25
Ogasawara
J
Watanabe-Fukunaga
R
Adachi
M
et al
Lethal effect of the anti-Fas antibody in mice.
Nature.
364
1993
806
809
26
Zutter
MM
Martin
PJ
Sale
GE
et al
Epstein-Barr virus lymphoproliferation after bone marrow transplantation.
Blood.
72
1988
520
529
27
Koh
MBC
Prentice
HG
Lowdell
MW
Selective removal of alloreactive cells from haematopoetic stem cell grafts: graft engineering for GVHD prophylaxis.
Bone Marrow Transplant.
23
1999
1071
1079
28
Hill
GR
Cooke
KR
Teshima
T
et al
Interleukin-11 promotes T cell polarization and prevents acute graft-versus-host disease after allogeneic bone marrow transplantation.
J Clin Invest.
102
1998
115
123
29
Garin
MI
Garrett
E
Tiberghien
P
et al
Molecular mechanism for ganciclovir resistance in human T lymphocytes transduced with retroviral vectors carrying the herpes simplex virus thymidin kinase gene.
Blood.
97
2001
122
129
30
Brunner
T
Mogli
RJ
LaFace
D
et al
Cell-autonomous Fas (CD95)/Fas-ligand interaction mediates activation-induced apoptosis in T cell hybridomas.
Nature.
373
1995
441
444
31
Dhein
J
Walczak
H
Bäumler
C
Debatin
K-M
Krammer
P
Autorine T-cell suicide mediated by APO-1/(Fas/CD95).
Nature.
373
1995
438
441
32
Van Parijs
L
Ibraghimov
A
Abbas
A-K
The roles of costimulation and Fas in T cell apoptosis and peripheral tolerance.
Immunity.
4
1996
321
329
33
Lissy
NA
Van Dyk
LF
Becker-Hapak
M
Vocero-Akbani
A
Mendler
JH
Dowdy
SF
TCR antigen-induced cell death occurs from a late G1 phase cell cycle check point.
Immunity.
8
1998
57
65
34
Kirchhoff
S
Müller
WW
Krueger
A
Schmitz
I
Krammer
PH
TCR-mediated up-regulation of cFLIPshort correlates with resistance toward CD95-mediated apoptosis by blocking death-inducing signalling complex activity.
J Immunol.
165
2000
6293
6300
35
Zhang
X
Brunner
T
Carter
L
et al
Unequal death in T helper cell (Th)1 and Th2 effectors: Th1, but not Th2, effectors undergo rapid Fas/FasL-mediated apoptosis.
J Exp Med.
185
1997
1837
1849
36
Fowler
DH
Kurasawa
K
Smith
R
Eckhaus
M-A
Gress
R-E
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
3549
37
Krenger
W
Snyder
KM
Byon
JCH
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
593
38
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
39
Baker
MB
Riley
RL
Podack
ER
Levy
RB
Graft-versus-host-disease-associated lymphoid hypoplasia and B cell dysfunction is dependent upon T cell-mediated Fas-ligand function, but not perforin function.
Proc Natl Acad Sci U S A.
94
1997
1366
1371
40
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
41
Schmaltz
C
Alpdogan
O
Horndasch
KJ
et al
Differential use of Fas ligand and perforin cytotoxic pathways by donor T cells in graft-versus-host disease and graft-versus-leukemia effect.
Blood.
97
2001
2886
2895

Author notes

Udo F. Hartwig, III Dept of Medicine, University Medical School Mainz, Langenbeckstr 1, D-55101 Mainz, Germany; e-mail: uhartwig@mail.uni-mainz.de.

Sign in via your Institution