Peripheral blood progenitor cells (PBPCs) are increasingly being used to replace bone marrow cells (BMCs) as a source of hematopoietic stem cells also in the field of allogeneic transplantation. Whereas it is well known that PBPC grafts and BM differ significantly in progenitor cell content and lymphocyte dose, the clinical consequences of these differences with respect to engraftment, graft-versus-host disease (GVHD), and the graft-versus-leukemia (GVL) effect are more difficult to assess. We present a murine model that allows us to evaluate engraftment, GVHD, and GVL effect of allogeneic PBPC transplantation (PBPCT). Balb/c mice (H-2d) served as recipients. Donors were major histocompatibility complex-matched DBA/2 mice or syngeneic Balb/c mice, respectively. Experiments with increasing numbers of BMCs or Filgastrim-mobilized PBPCs showed that the number of progenitor cells in the graft was correlated with the probability to engraft, irrespective of the graft type. With identically high cell numbers transferred (1 × 109 nucleated cells/kg body weight [BW]), the mortality rates due to GVHD (25%) were about the same after allogeneic BM transplantation (BMT) and allogeneic PBPCT, although PBPC grafts contained four times more CD3+ T cells as compared with BM grafts (6.2 × 108v 1.4 × 108/kg BW). For investigation of GVL activity, Balb/c recipients were injected with syngeneic cells of the B-lymphocytic leukemia cell line A20 2 days before transplantation. After total body irradiation to a dose of 7.5 Gy, 1 × 109/kg BW Balb/c PBPCs, DBA BMCs, or DBA PBPCs were infused. The relapse rates observed were 80% after syngeneic PBPCT (n = 22), 60% after allogeneic BMT (n = 23), and 34% after allogeneic PBPCT (n = 26) (allogeneic BMT v PBPCT, P = .032). We conclude that transplantation of allogeneic PBPCs instead of BM may enhance the GVL effect without an increase of GVHD.

ALLOGENEIC bone marrow transplantation (BMT) is the treatment of choice for a variety of hematologic malignancies. Animal experiments performed as early as 19571 suggested that the strong antileukemic effect of BMT resulted not only from the high-dose chemoradiotherapy preceding the transplantation but also was a consequence of adoptive immunotherapy exerted by the graft itself. Numerous experimental and clinical reports2-8 have confirmed these findings and underscored our view that the superior antitumor effect of BMT is primarily due to the destruction of leukemic target cells by an allogeneic immune response mediated by T lymphocytes and natural killer (NK) cells contained in the transplant.

Recently, granulocyte colony-stimulating factor (G-CSF )–mobilized peripheral blood progenitor cells (PBPCs) have been introduced as an alternative source of hematopoietic stem cells for allogeneic transplantation.9-11 Harvesting PBPCs instead of BM avoids the discomfort of marrow harvesting and the risks of general anesthesia in the donor while the high numbers of progenitor cells contained in such a graft lead to rapid and reliable engraftment in the recipient. The large numbers of progenitor cells that can be harvested from the PB12,13 also allow for extensive ex vivo manipulations and make PBPC attractive as target cells for gene therapeutical maneuvers and immunotherapy.

Important questions related to PBPCT remain unanswered at this time. Whereas reports from single institutions14 and the European Group for Blood and Marrow Transplantation15 suggest that the transplantation of allogeneic PBPC does not cause devastating graft-versus-host disease (GVHD), a more precise comparison of the incidence and severity of GVHD after PBPCT as opposed to BMT must await the results of prospective clinical trials. There is even more uncertainty if the high numbers of T cells and NK cells contained in a typical PBPC collection product exert a more vigorous graft-versus-leukemia (GVL) effect, reduce the risk of relapse, and can thus improve disease-free survival after allogeneic PBPCT.

To address these latter questions, we developed a murine model that allows us to investigate the engraftment potential, graft-versus-host reactivity, and GVL activity of allogeneic PBPCs. Experiments presented here show a superior antileukemic activity of allogeneic, major histocompatibility complex (MHC)-matched PBPCs over BM cells (BMCs) in leukemia-bearing mice.

Animals.Balb/c (H-2d) and DBA/2 (H-2d) mice were bred and kept at the animal facilities of our institution. All animals were housed in conventional cages, 7 to 10 animals to a cage, and received nonsterilized food and water ad libitum. Cotrimoxazole was added to the water for 40 days after BMT or PBPCT.

Hematopoietic stem cell transplantation.A Cs137 source was used for total body irradiation (TBI) of the recipients. A lethal dose of 7.5 Gy TBI was chosen for conditioning.16 BM grafts were prepared according to standard procedures as previously described.17 Mice intended to serve as donors of PBPCs had their spleens removed under general anesthesia at least 14 days before the donation of PBPCs. Starting 5 days before the collection of PBPC, 5 μg of rhu-met-G-CSF (Filgastrim; Amgen, Thousand Oaks, CA) was injected twice daily subcutaneously. The last injection of Filgastrim was administered 2 hours before harvesting of PBPC. The mice were then anticoagulated with heparin, anesthetized, and killed by cervical dislocation. The PB was collected under sterile conditions by dissection of both Aa carotidea. Erythrocytes were lysed by incubation of PB in 0.15 mol/L ammoniumchloride buffer at 20°C for 5 minutes. The cell number was adjusted to 6 × 107 nucleated cells (NC)/mL. For smaller grafts, a cell concentration of 1 × 107 NC/mL was used. BM or PBPC grafts were injected intravenously 2 hours after conditioning of the recipients by TBI.

T-cell depletion of PBPC grafts.Immunomagnetic depletion of T cells from the PBPC grafts was used in some experiments. Immunomagnetic beads (Dynal, Oslo, Norway), coated with goat-antimouse Ig, were coupled to an antimurine CD3 monoclonal antibody (MoAb; KT-3; Serotec, Wiesbaden, Germany). The CD3 MoAb was used at a concentration of 11 μg per 4 × 108 beads. Coupling of MoAb and magnetic beads was performed at 4°C for 30 minutes. The ratio of beads/target cell was 6:1. The magnetic beads were coincubated with the PBPC graft for 1 hour at 21°C, and the cells were then removed with a strong permanent-magnetic device (Dynal). The T-depleted grafts were examined for their content of residual T cells by fluorescence-activated cell sorting (FACS) analysis. The content of T cells after depletion was at the threshold of detection by FACS analysis (1% of nucleated cells; range, 0.2% to 2.3%).

Progenitor cell colony assays.For determining the progenitor cell content of the grafts, the Methocult GF M3434 kit was used (Stemcell Technologies, Vancouver, British Columbia, Canada). The assays were performed according to the protocol of the manufacturer. Colony-forming units–granulocyte-macrophage (CFU-GM) were counted 12 days after the plating of 1.5 × 104 nucleated cells per dish. The cell concentration was 1.5 × 105 NC/mL. Experiments were performed in parallel to the transplantation experiments with graded numbers of PBPCs and BMCs.

Leukemia cells.A20 is a B-cell leukemia/lymphoma of Balb/c origin that occurred spontaneously in a 15-month-old mouse.18 The cells were maintained in culture with RPMI 1640 + 5% fetal calf serum at 37°C and 5% CO2. We performed in vivo passages of these cells by intravenous injection into Balb/c animals. After the mice had developed hepatosplenomegaly, they were killed and their spleens were removed. Spleen cell suspensions containing close to 100% of these in vivo passaged leukemia/lymphoma cells were stored in liquid nitrogen and used for further experiments.

Experimental design and definitions.Engraftment and GVHD were investigated in leukemia-free mice of the Balb/c strain. Donors were either animals of Balb/c or of DBA/2 origin. Increasing numbers of BMCs or PBPCs were transferred to the recipients after TBI (7.5 Gy). The animals were examined at least once daily afterwards. Signs of GVHD (weight loss, rough fur, and gibbus) were documented. PB counts were performed every 3 days from day 7 to complete recovery of hematopoiesis or death. In a number of surviving animals, long-term chimerism was investigated by FACS analysis determining the presence/absence of the Lyt 1.1 antigen (CD5, clone H11 86.1; Pharmingen, San Diego, CA) on spleen cells. This antigen exists in two different forms: Balb/c mice express the isoform Lyt 1.2, whereas DBA/2 mice are known to express Lyt 1.1. Animals were killed on day 100 posttransplantation, their spleens were removed, and a single-cell suspension was produced. Untreated animals of Balb/c and DBA/2 origin were used as negative and positive controls, respectively. If the recipient mouse showed the same fraction of splenocytes (±6%) being positive for Lyt 1.1 as for the DBA control, it was named a long-term chimera. Investigation of the GVL effect was performed in Balb/c recipient animals injected intravenously with 1 × 105 A20 cells 2 days before transplantation of either 2 × 107 Balb/c PBPCs, DBA PBPCs, or DBA BMCs, respectively. The animals were examined daily and necropsied after death. The cause of death was determined according to the following definitions. Graft failure or graft rejection was defined as death between day 6 and 30 after transplantation with leukocytes less than 1/nL and granulocytes greater than 0.5/nL. Death due to GVHD was defined as death with recovery of hematopoiesis (leukocytes >3/nL and thrombocytes >50/nL) and macroscopic signs of GVHD such as weight loss, rough fur, hair loss, and gibbus. In some mice with macroscopic GVHD, histologic examination of skin and liver was performed that showed changes compatible with acute GVHD.19,20 Leukemic relapse was defined as death with macroscopic tumor and liver weight more than 1.5 g and spleen weight more than 0.15 g. Histologic examination of liver and spleen was performed for two animals in each group. Animals with hepatosplenomegaly were found to harbor leukemic cells without exception. Healthy mice of the same age have liver weights of 1.3 ± 0.2 g and spleen weights of 0.1 ± 0.02 g, respectively.

Statistics.Survival data and freedom from leukemia were calculated according to the method of Kaplan and Meier. The experimental groups were compared using the log-rank test. The calculations were performed on a PC with Statistica statistical software (StatSoft, Tulsa, OK).

Mobilization of PB progenitor cells and composition of the transplants.To avoid pooling of PBPCs in the spleen, mice were splenectomized before G-CSF administration. This led to an increase of white blood cells from 6.4/nL to 9.9/nL (range, 9.6 to 10.8 cells/nL) in DBA/2 mice. Differential counts were not altered after splenectomy. G-CSF administration induced a substantial increase of the leukocyte count up to 59.6/nL (range, 42.9 to 73.3/nL) in Balb/c mice or 72.9/nL (range, 51.8 to 85.8/nL) in DBA mice. In steady state, blood of DBA mice contained 0.80 CFU-GM/104 NC. After mobilization, the PBPC collection product contained 1.13 CFU-GM/104 NC, resulting in a substantially higher concentration of progenitor cells in PB (8.23 CFU-GM/μL after and 0.79 CFU-GM/μL blood before G-CSF administration). BM of DBA mice gave rise to 4.83 CFU-GM/104 NC. Balb/c mice were shown to have lower concentrations of progenitor cells in BM or PB, but the relationship between the PBPC graft and BM was identical (Table 1). A total of 2 × 107 NC/animal was the standard dose transferred to each animal in GVHD and GVL experiments. With the average weight of a Balb/c mouse being 20 g, this would correspond to a dose of 1 × 109 NC/kg body weight (BW) and 11 × 104 CFU-GM/kg BW administered with an allogeneic PBPC graft. The progenitor cell dose administered with the standard BMT procedure was fourfold higher. The percentage of lymphocytes was significantly higher in the PB as compared with the BM transplants. Allogeneic PBPC grafts contained 62% (range, 50% to 71%) of CD3+ cells; BM grafts contained 14% (range, 6% to 29.8%; Table 1). This corresponded to 6.2 × 108 T cells/kg BW for allogeneic PBPCT and 1.4 × 108 T cells/kg BW for allogeneic BMT, respectively, with the standard transplant.

Table 1.

Cellular Composition of Stem Cell Transplants

StrainSourcenLeukocytes (/nL)Neutrophils (%)MNC (%)T Cells CD3+ (%)NK Cells 5E6+ (%)B Cells CD19+ (%)Progenitor Cells CFU-GM
(/104 cells)
Balb/c PB 4.9 (3.4-8.2) 10 (10-50) 90 21 (11-32) 7 (4-11) ND 0.13 (0.06-0.2) 
 PBPC 59.6 (42.6-73.3) 50 (20-80) 50 32 (15-35) 4 (2-5) 11 (4-13) 0.53 (0.2-0.87) 
DBA/2 PB 9.9 (9.4-10.8) 50 (10-60) 50 20 (12-27) 11 (8-13) ND 0.80 (0.67-3.0) 
 PBPC 72.9 (51.8-85.8) 40 (15-60) 60 62 (50-71) 5 (2-7) 20 (11-24) 1.13 (0.07-3.6) 
 BM ND ND ND 14 (6-30) 4 (3-4) ND 4.83 (1.4-11.47) 
StrainSourcenLeukocytes (/nL)Neutrophils (%)MNC (%)T Cells CD3+ (%)NK Cells 5E6+ (%)B Cells CD19+ (%)Progenitor Cells CFU-GM
(/104 cells)
Balb/c PB 4.9 (3.4-8.2) 10 (10-50) 90 21 (11-32) 7 (4-11) ND 0.13 (0.06-0.2) 
 PBPC 59.6 (42.6-73.3) 50 (20-80) 50 32 (15-35) 4 (2-5) 11 (4-13) 0.53 (0.2-0.87) 
DBA/2 PB 9.9 (9.4-10.8) 50 (10-60) 50 20 (12-27) 11 (8-13) ND 0.80 (0.67-3.0) 
 PBPC 72.9 (51.8-85.8) 40 (15-60) 60 62 (50-71) 5 (2-7) 20 (11-24) 1.13 (0.07-3.6) 
 BM ND ND ND 14 (6-30) 4 (3-4) ND 4.83 (1.4-11.47) 

Characterization of stem cell transplants as administered to leukemia-bearing Balb/c recipients. Medians and ranges are given for data obtained from the pooled grafts derived from 1 or 2 donors. The flow cytometry analysis was performed in parallel in 6 of 10 single transplantation experiments. The percentage of lymphocytes is given as the percentage of nucleated cells. Nucleated cells were also the basis for the calculation of graft size in all experiments. The MoAb 5E6 labels a subpopulation of NK cells. CFU-GM were counted on day +12 after seeding.

Abbreviations: MNC, mononuclear cells; ND, not determined.

Engraftment.In experiments with nonleukemic animals, three parameters possibly determining engraftment were varied: the cell dose of the transplant, the immunogenetic difference between donor and recipient, and the source of hematopoietic stem cells. With PBPCs, the number of nucleated cells transferred had a major impact on engraftment as measured by the rate of graft failure (Fig 1) or the kinetics of hematopoietic recovery in the surviving animals (Fig 2). Transplantation of a higher cell dose was followed by a lower rejection rate and faster hematopoietic recovery after both syngeneic and allogeneic transplantation. Using the same fixed cell dose, the rate of graft failure was higher after allogeneic MHC-matched PBPCT than after syngeneic PBPCT. Complete engraftment was achieved with a minimum cell dose of 5 × 108 cells/kg BW after syngeneic PBPCT; the same cell dose caused a graft failure rate of 19% after allogeneic PBPCT. Thus, the immunogenetic difference was another parameter determining engraftment after PBPCT. On the basis of fixed numbers of nucleated cells transferred, allogeneic BMT was followed by lower rejection rates (Fig 1) than allogeneic PBPCT. The source of hematopoietic stem cells also influenced engraftment kinetics: a cell dose of 5 × 107 cells/kg BW gave a substantial slower engraftment after PBPCT (>2 leukocytes/nL on day 17) as compared with BMT (>2 leukocytes/nL on day 11). Long-term engraftment was investigated by immunofluorescence staining of recipient splenocytes with an anti-Lyt 1.1 antibody (CD5) and subsequent FACS analysis (Fig 3). Staining of the spleen cells of allo-PBPCT recipients 100 days after transplantation showed the majority of T lymphocytes to be of donor origin in all surviving recipients tested (n = 10). High cell numbers of either origin, PBPC and BM, uniformly resulted in complete and very fast engraftment (>2 leukocytes/nL on day 7, Fig 2).

Fig. 1.

Comparison of graft failure rates after syngeneic or allogeneic MHC-matched PBPCT. Recipients were Balb/c mice and donors were Balb/c or DBA/2 mice, respectively. Donors were pretreated by 2 × 5/μg Filgastrim subcutaneously for 5 consecutive days before stem cell harvest. Experiments were performed in a head-to-head manner with six repeated single experiments. The total number of animals in each group ranges from 8 to 27.

Fig. 1.

Comparison of graft failure rates after syngeneic or allogeneic MHC-matched PBPCT. Recipients were Balb/c mice and donors were Balb/c or DBA/2 mice, respectively. Donors were pretreated by 2 × 5/μg Filgastrim subcutaneously for 5 consecutive days before stem cell harvest. Experiments were performed in a head-to-head manner with six repeated single experiments. The total number of animals in each group ranges from 8 to 27.

Close modal
Fig. 2.

Leukocyte recovery in recipients of allogeneic MHC-matched PBPC or BM graft. Data are from the same experiments as described in Fig 1.

Fig. 2.

Leukocyte recovery in recipients of allogeneic MHC-matched PBPC or BM graft. Data are from the same experiments as described in Fig 1.

Close modal
Fig. 3.

Example for analysis of long-term chimerism. Spleen cells of surviving animals from the experiment described in Figs 1 and 2 were collected 100 days after transfer of 1 × 109 DBA PBPC/kg BW in Balb/c mice. The mononuclear cells were prepared by density gradient centrifugation and stained with MoAb anti-Lyt 1.1. Lyt 1.1 is an isoform of murine CD5 found in DBA but not in Balb/c mice. Untreated Balb/c and DBA mice served as negative and positive controls, respectively. The transplant recipients showed the same percentage of Lyt 1.1+ splenocytes as DBA mice, demonstrating long-term chimerism of T cells after transplantation.

Fig. 3.

Example for analysis of long-term chimerism. Spleen cells of surviving animals from the experiment described in Figs 1 and 2 were collected 100 days after transfer of 1 × 109 DBA PBPC/kg BW in Balb/c mice. The mononuclear cells were prepared by density gradient centrifugation and stained with MoAb anti-Lyt 1.1. Lyt 1.1 is an isoform of murine CD5 found in DBA but not in Balb/c mice. Untreated Balb/c and DBA mice served as negative and positive controls, respectively. The transplant recipients showed the same percentage of Lyt 1.1+ splenocytes as DBA mice, demonstrating long-term chimerism of T cells after transplantation.

Close modal

GVHD.The transplantation of 1 × 109 cells/kg BW allogeneic MHC-matched PBPCs resulted in stable engraftment, but 4 of 16 (25%) nonleukemic animals died with signs of GVHD. Histological examination showed GVHD-typical infiltration in the liver and skin in these animals. After allogeneic BMT with 1 × 109 cells/kg BW, 2 of 8 animals died on days 43 and 93 with signs of GVHD (Fig 4). This type of mortality was not observed after syngeneic PBPCT. After depletion of the CD3+ cells from allogeneic PBPC transplants, only 1 of 17 animals (6%) died with macroscopic signs of GVHD. One of the animals died during the period of TBI-induced aplasia.

Fig. 4.

Survival of nonleukemic Balb/c mice after TBI only (n = 12), transfer of 1 × 109 syngeneic PBPCs/kg BW (n = 10), and transfer of the same dose of unmanipulated allogeneic PBPC (n = 16) or CD3-depleted allogeneic PBPCs (n = 16). T-cell depletion was performed by immunomagnetic removal of these cells by anti-CD3–coated magnetic beads, resulting in less than 1% CD3+ cells as shown by flow cytometry.

Fig. 4.

Survival of nonleukemic Balb/c mice after TBI only (n = 12), transfer of 1 × 109 syngeneic PBPCs/kg BW (n = 10), and transfer of the same dose of unmanipulated allogeneic PBPC (n = 16) or CD3-depleted allogeneic PBPCs (n = 16). T-cell depletion was performed by immunomagnetic removal of these cells by anti-CD3–coated magnetic beads, resulting in less than 1% CD3+ cells as shown by flow cytometry.

Close modal

GVL effect.To evaluate the GVL effects after transplanting PBPCs or BM cells, the relapse risk of Balb/c mice bearing the lymphoid leukemia A20 and transplanted with PBPCs or BM cells was assessed. After intravenous injection of 1 × 105 A20 cells, all untreated animals died after a median of 28 days. TBI with a dose of 7.5 Gy and subsequent syngeneic PBPCT with 1 × 109 cells/kg BW 2 days after leukemia cell injection resulted in a prolongation of the time to relapse to a median of 43 days; 20% of the animals remained free from leukemia. Transfer of 1 × 109 cells/kg BW allogeneic MHC-matched PBPCs after identical pretreatment of the recipients caused a significantly lower relapse rate with freedom from leukemia at 66% (P = .0045). Freedom from leukemia is 40% (median time to relapse, 58 days) after allogeneic BMT and thus significantly lower than after allogeneic PBPCT (P = .032; Fig 5).

Fig. 5.

Freedom from leukemia of Balb/c mice injected with 1 × 105 cells of the B-lymphocytic leukemia cell line A20. Recipients received no further treatment (n = 10) or received TBI with a dose of 7.5 Gy followed by transplantation of hematopoietic stem cells 2 days after leukemia cell injection. The transplants were either syngeneic PBPCs (n = 22), allogeneic BM cells (n = 23), or allogeneic PBPCs (n = 26).

Fig. 5.

Freedom from leukemia of Balb/c mice injected with 1 × 105 cells of the B-lymphocytic leukemia cell line A20. Recipients received no further treatment (n = 10) or received TBI with a dose of 7.5 Gy followed by transplantation of hematopoietic stem cells 2 days after leukemia cell injection. The transplants were either syngeneic PBPCs (n = 22), allogeneic BM cells (n = 23), or allogeneic PBPCs (n = 26).

Close modal

The animal data presented here address some of the important questions related to transplantation of allogeneic PBPCs. In particular, and in contrast to other murine21-23 and dog experiments,24 our model allows the investigation of the GVL activity of PBPCs as opposed to BM grafts. Results of animal experiments can seldom be directly translated into the clinic because of inherent differences between the experimental setting and the clinical situation. In our model, recipients (Balb/c) and donors (DBA/2) of hematopoietic stem cells share the MHC complex but differ with respect to the rest of their genetic background. This situation most closely resembles the transplantation of hematopoietic stem cells between HLA-identical donor/recipient pairs, more so in the matched unrelated than in the HLA-identical sibling setting. There are other limitations of the model pertaining to the composition of both marrow and PBPC grafts in mice and humans and their clinical implications. Although other parameters may be more important with respect to certain experimental or clinical endpoints, we chose to primarily characterize marrow and PBPC grafts by their content of nucleated cells and relate comparisons between BMT and PBPCT to fixed numbers of nucleated cells present in either type of graft. This is practical and matches the way how clinical BM and PBPC grafts have been characterized in many instances.10,12,13 We additionally determined the progenitor cell and lymphocyte content of the grafts to enable a comparison of these parameters and their possible relevance with respect to engraftment potential, graft-versus-host reactivity, and GVL effect observed after BMT or PBPCT.

We and others have previously shown that the progenitor cell dose in a graft is the decisive parameter to predict the engraftment potential of a hematopoietic stem cell graft if pretransplantation immunosuppression and the immunogenetic difference between donor and recipient remain unchanged.16,25,26 In this series of experiments we were able to confirme that the transfer of high numbers of marrow or PB cells improve engraftment and engraftment kinetics. However, when the engraftment potential of BM cells and PBPCs was compared with identical numbers of nucleated cells in the grafts, BM cells resulted in faster and more reliable engraftment, a situation contradictory to clinical experience.9 This seemingly surprising result can easily be explained if one keeps in mind that, in the clinic, we aim to transplant the highest possible number of cells and do not transfer fixed numbers of nucleated cells, CD34+ cells, or colony-forming cells. However, the average number of nucleated cells contained in a PBPC collection product is 3 to 4 times higher than in a typical BM graft13 and thus can more than compensate for the relative scarcity of colony-forming cells in PB.

Acute and chronic graft-versus-host disease are less well defined and may even have a partly different pathogenesis in mice27,28 as compared with humans29 or dogs.30 Therefore, sublethal forms of GVHD were not taken into account for this analysis. With these limitations in mind, we did not find substantial differences between BM or PBPC grafts with respect to their GVHD-inducing potential. Roughly the same rates of mortality due to GVHD (25%) were observed after allogeneic BMT and PBPCT. With approximately three times the number of T cells in a PBPC graft than in marrow, this is a surprising finding that is in line, however, with clinical observations reported so far.14,15 The model of the leukemia-bearing mouse allows to investigate the GVL effect conferred by allogeneic PBPCs as compared with allogeneic BMCs. This feature of the model is especially valuable because the heterogeneity of human recipients of allogeneic PBPCs in terms of their underlying disease and stage precludes any conclusive answer to the question if the relatively low relapse rates observed after PBPCT so far are the consequence of a more vigorous GVL effect. Reliable clinical data on the GVL effect after transplantation of unmanipulated or T-cell–depleted allogeneic PB stem cells will not be available for years because this will need long-term follow-up of a homogeneous patient population grafted with either PBPCs or BM. The results presented here suggest an advantage of PBPC over BM grafts with respect to their antileukemic activity. What are the possible explanations for this finding? First, there have been reports that priming with G-CSF can alter T-cell phenotype and function.31,32 It cannot be excluded that these differences not only affect graft-versus-host reactivity but also GVL activity. Second, it has repeatedly been reported that human PBPC grafts contain 10-fold more T cells and 19- to 25-fold more NK cells as compared with BM grafts.11-13 In this experimental system, even with a fixed number of nucleated cells administered, we also transferred more T cells and NK cells with PBPCs than with BM grafts. Both cell types have been shown to be involved in GVL activity.33-39 In the murine model presented here, the increase in GVL reactivity was not accompanied by a substantial increase of GVHD. One possible explanation for this finding could be that GVL and GVHD are stochastic processes. If the frequency of graft-versus-host–reactive cells in the donor is higher than the frequency of GVL-reactive cells, the relatively small lymphocyte dose transferred with a BM graft may already contain the near complete GVHD potential of the donor but may be too small to confer all of the GVL potential. An increase of the lymphocyte dose as found in a PBPC harvest will not add significantly more GVHD-reactive cells but could result in a substantial increase of GVL-reactive cells. Further experiments using the experimental model reported here are underway to further elucidate the relative roles of T cells and NK cells triggering graft-versus-host reactivity and GVL activity. These experiments will hopefully shed further light on the basic biology of graft-versus-host reactivity and GVL effects and give some indication how allogeneic PBPC grafts should best be manipulated to avoid GVHD but preserve their antileukemic potential. The experiments reported here have encouraged us to continue with the clinical evaluation of allogeneic PBPCT.

The authors thank S. Schlemminger for expert technical assistance.

Supported by a grant from Deutsche Krebshilfe (1-0958-Uh I).

Address reprint requests to Bertram Glass, MD, 2nd Department of Internal Medicine, Christian-Albrechts-University of Kiel, Chemnitzstrasse 33, D-24116 Kiel, Germany.

1
Barnes
DWH
Loutit
JF
Treatment of murine leukaemia with X-rays and homologous bone marrow: II.
Br J Haematol
3
1957
241
2
Bortin
MM
Truitt
RL
Rimm
AA
Bach
FH
Graft-versus-leukaemia reactivity induced by alloimmunisation without augmentation of graft-versus-host reactivity.
Nature
281
1979
490
3
Truitt
RL
Atasoylu
AA
Contribution of CD4+ and CD8+ T cells to graft-versus-host disease and graft-versus-leukemia reactivity after transplantation of MHC-compatible bone marrow.
Bone Marrow Transplant
8
1991
51
4
Weiss
L
Weigensberg
M
Morecki
S
Bar
S
Cobbold
S
Waldmann
H
Slavin
S
Characterization of effector cells of graft vs leukemia following allogeneic bone marrow transplantation in mice inoculated with murine B-cell leukemia.
Cancer Immunol Immunother
31
1990
236
5
Aizawa
S
Sado
T
Graft-versus-leukemia effect in MHC-compatible and -incompatible allogeneic bone marrow transplantation of radiation-induced, leukemia-bearing mice.
Transplantation
52
1991
885
6
Weiden
PL
Flournoy
N
Thomas
ED
Prentice
R
Fefer
A
Buckner
CD
Storb
R
Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts.
N Engl J Med
300
1979
1068
7
Horowitz
MM
Gale
RP
Sondel
PM
Goldman
JM
Kersey
J
Kolb
HJ
Rimm
AA
Ringden
O
Rozman
C
Speck
B
Truitt
RL
Zwaan
FE
Bortin
MM
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
75
1990
555
8
Barrett
AJ
Malkovska
V
Graft-versus-leukemia: Understanding and using the alloimmune response to treat haematological malignancies.
Br J Haematol
93
1996
754
9
Schmitz
N
Dreger
P
Suttorp
M
Rohwedder
EB
Haferlach
T
Loeffler
H
Hunter
A
Russell
NH
Primary transplantation of allogeneic peripheral blood progenitor cells mobilized by filgrastim (granulocyte colony-stimulating factor).
Blood
85
1995
1666
10
Bensinger
WI
Weaver
CH
Appelbaum
FR
Rowley
S
Demirer
T
Sanders
J
Storb
R
Buckner
CD
Transplantation of allogeneic peripheral blood stem cells mobilized by recombinant human granulocyte colony-stimulating factor.
Blood
85
1995
1655
11
Koerbling
M
Przepiorka
D
Huh
YO
Engel
H
van Besien
K
Giralt
S
Andersson
BS
Kleine
HD
Seong
D
Deisseroth
AB
Andreeff
M
Champlin
R
Allogeneic blood stem cell transplantation for refractory leukemia and lymphoma: Potential advantage of blood over marrow allografts.
Blood
85
1995
1659
12
Koerbling
M
Huh
YO
Durett
A
Mirza
N
Miller
P
Engel
H
Anderlini
P
van Besien
K
Andreeff
M
Przepiorka
D
Deisseroth
AB
Champlin
RE
Allogeneic blood stem cell transplantation: Peripheralization and yield of donor-derived primitive hematopoietic progenitor cells (CD34+ Thy-1dim) and lymphoid subsets, and possible predictors of engraftment and graft-versus-host disease.
Blood
86
1995
2842
13
Dreger
P
Haferlach
T
Eckstein
V
Jacobs
S
Suttorp
M
Löffler
H
Löffler
M
Schmitz
N
G-CSF-mobilized peripheral blood progenitor cells for allogeneic transplantation: Safety, kinetics of mobilization, and composition of the graft.
Br J Haematol
87
1994
609
14
Bensinger
WI
Clift
R
Martin
P
Appelbaum
F
Demirer
T
Gooley
TA
Lilleby
K
Rowley
S
Sanders
J
Storb
R
Buckner
CD
Allogeneic peripheral blood stem cell transplantation in patients with advanced hematologic malignancies: A retrospective comparison with marrow transplantation.
Blood
88
1996
2794
15
Schmitz
N
Bacigalupo
A
Labopin
M
Majolino
I
Laporte
JP
Brinch
L
Cook
G
Lambertenghi-Deliliers
G
Lange
A
Rozman
C
Garcia-Conde
J
Finke
J
Domingo-Albos
A
Gratwohl
A on behalf of the EBMT
Transplantation of peripheral blood progenitor cells from HLA-identical sibling donors.
Br J Haematol
95
1996
715
16
Uharek
L
Glass
B
Gaska
T
Gassmann
W
Loeffler
H
Mueller
Ruchholtz W
Influence of donor lymphocytes on the incidence of primary graft failure after allogeneic bone marrow transplantation in a murine model.
Br J Haematol
88
1994
79
17
Glass
B
Uharek
L
Gassmann
W
Focks
B
Bolouri
H
Loeffler
H
Mueller-Ruchholtz
W
Graft-versus-leukemia activity after bone marrow transplantation does not require graft-versus-host disease.
Ann Hematol
64
1992
255
18
Kim
KJ
Kannellopoulus-Langevin
C
Mervin
RM
Sachs
DH
Asofsky
R
Establishment and characterization of Balb/c lymphoma lines with B cell properties.
J Immunol
122
1979
549
19
Martin
PJ
Animal experimentation relevant to human marrow transplantation.
Curr Opin Oncol
4
1992
239
20
van Bekkum DW: What is graft versus host disease? Bone Marrow Transplant 7:110, 1991 (suppl 2)
21
Molineux
G
Pojda
Z
Hampson
IN
Lord
BI
Dexter
TM
Transplantation potential of peripheral blood stem cells induced by granulocyte colony-stimulating factor.
Blood
76
1990
2153
22
Yan
XQ
Hartley
C
McElroy
P
Chang
A
McCrea
C
McNiece
I
Peripheral blood progenitor cells mobilized by recombinant human granulocyte colony-stimulating factor plus recombinant rat stem cell factor contain long-term engrafting cells capable of cellular proliferation for more than two years as shown by serial transplantation in mice.
Blood
85
1995
2303
23
Andrews
RG
Briddell
RA
Knitter
GH
Rowley
SD
Appelbaum
FR
McNiece
IK
Rapid engraftment by peripheral blood progenitor cells mobilized by recombinant human stem cell factor and recombinant human granulocyte colony-stimulating factor in nonhuman primates.
Blood
85
1995
15
24
Sandmaier
BM
Storb
R
Santos
EB
Krizanac
Bengez L
Lian
T
McSweeney
PA
Yu
C
Schuening
FG
Deeg
HJ
Graham
T
Allogeneic transplant of canine peripheral blood stem cells mobilized by recombinant canine hematopoietic growth factors.
Blood
87
1996
3508
25
Uharek
L
Gassmann
W
Glass
B
Steinmann
J
Loeffler
H
Mueller-Ruchholtz
W
Influence of cell dose and graft-versus-host reactivity on rejection rates after allogeneic bone marrow transplantation.
Blood
79
1992
1612
26
Bachar
Lustig E
Rachamim
N
Li
HW
Lan
F
Reisner
Y
Megadose of T cell-depleted bone marrow overcomes MHC barriers in sublethally irradiated mice.
Nat Med
1
1995
1268
27
Vallera
DA
Soderling
CC
Carlson
GJ
Kersey
JH
Bone marrow transplantation across major histocompatibility barriers in mice. Effect of elimination of T cells from donor grafts by treatment with monoclonal Thy-1.2 plus complement or antibody alone.
Transplantation
31
1981
218
28
Blazar
BR
Roopenian
DC
Taylor
PA
Christianson
GJ
Panoskaltsis
Mortari A
Vallera
DA
Lack of GVHD across classical, single minor histocompatibility (miH) locus barriers in mice.
Transplantation
61
1996
619
29
Ferrara
JL
Deeg
HJ
Graft-versus-host disease.
N Engl J Med
324
1991
667
30
Storb
R
Thomas
ED
Graft-versus-host disease in dog and man: The Seattle experience.
Immunol Rev
88
1985
215
31
Pan
L
Delmonte
J
Jalonen
CK
Ferrara
JLM
Pretreatment of donor mice with granulocyte colony-stimulating factor polarizes donor T lymphocytes toward type-2 cytokine production and reduces severity of experimental graft-versus-host disease.
Blood
86
1995
4422
32
Krenger
W
Snyder
KM
Byon
JCH
Falzarano
G
Ferrara
JLM
Polarized type 2 alloreactive CD4+ and CD8+ donor cells fail to induce experimental acute graft-versus-host disease.
J Immunol
155
1995
585
33
Goldman
JM
Gale
RP
Horowitz
MM
Biggs
JC
Champlin
RE
Gluckman
E
Hoffmann
RG
Jacobsen
SJ
Marmont
AM
McGlave
PB
Messner
HA
Rimm
AA
Rozman
C
Speck
B
Tura
S
Weiner
RS
Bortin
MM
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
34
Marmont
AM
Horowitz
MM
Gale
RP
Sobocinski
K
Ash
RC
van Bekkum
DW
Champlin
RE
Dicke
KA
Goldman
JM
Good
RA
Herzig
RH
Hong
R
Masaoka
T
Rimm
AA
Ringdén
O
Speck
B
Weiner
RS
Bortin
MM
T-cell depletion of HLA-identical transplants in leukemia.
Blood
78
1991
2120
35
Weiss
L
Weigensberg
M
Morecki
S
Bar
S
Cobbold
S
Waldmann
H
Slavin
S
Characterization of effector cells of graft vs leukemia following allogeneic bone marrow transplantation in mice inoculated with murine B-cell leukemia.
Cancer Immunol Immunother
31
1990
236
36
O'Kunewick
JP
Kociban
DL
Machen
LL
Buffo
MJ
Comparison of the effects of CD3 and CD5 donor T cell depletion on graft-versus-leukemia in a murine model for MHC-matched unrelated-donor transplantation.
Bone Marrow Transplant
13
1994
11
37
Hauch
M
Gazzola
MV
Small
T
Bordignon
C
Barnett
L
Cunningham
I
Castro
Malaspinia H
O'Reilly
RJ
Keever
CA
Anti-leukemia potential of interleukin-2 activated natural killer cells after bone marrow transplantation for chronic myelogenous leukemia.
Blood
75
1990
2250
38
Zeis
M
Uharek
L
Glass
B
Gaska
T
Steinmann
J
Gassmann
W
Loeffler
H
Mueller-Ruchholtz
W
Allogeneic NK cells as potent antileukemic effector cells after allogeneic bone marrow transplantation in mice.
Transplantation
59
1995
1734
39
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
Sign in via your Institution