Allogeneic transplantation of hematopoietic cells is an effective treatment of leukemia, even in advanced stages. Allogeneic lymphocytes produce a strong graft-versus-leukemia (GVL) effect, but the beneficial effect is limited by graft-versus-host disease (GVHD). Depletion of T cells abrogates GVHD and GVL effects. Delayed transfusion of donor lymphocytes into chimeras after T cell–depleted stem cell transplantation produces a GVL effect without necessarily producing GVHD. Chimerism and tolerance provide a platform for immunotherapy using donor lymphocytes. The allogeneic GVL effects vary from one disease to another, the stage of the disease, donor histocompatibility, the degree of chimerism, and additional treatment. Immunosuppressive therapy before donor lymphocyte transfusions may augment the effect as well as concomitant cytokine treatment. Possible target antigens are histocompatibility antigens and tumor-associated antigens. Immune escape of tumor cells and changes in the reactivity of T cells are to be considered. Durable responses may be the result of the elimination of leukemia stem cells or the establishment of a durable immune control on their progeny. Recently, we have learned from adoptive immunotherapy of viral diseases and HLA-haploidentical stem cell transplantation that T-cell memory may be essential for the effective treatment of leukemia and other malignancies.

Treatment of leukemia and other malignancies of the hematopoietic system with total body irradiation (TBI) and transplantation of bone marrow from healthy persons was developed from studies on radiation protection in the 1950s. However, it was evident from bioassays of murine leukemia that radiation could not eliminate leukemia in doses tolerated clinically.1  In contrast, TBI followed by allogeneic marrow transplantation could cure leukemia in some murine models.2  The term adoptive immunotherapy by allogeneic marrow transplantation was coined by Mathé3  in the early 1960s when little was known about donor selection, appropriate conditioning treatment, and prevention of graft-versus-host disease (GVHD). Selection of sibling donors by human leukocyte antigen (HLA) typing, the combination of TBI with cyclophosphamide for conditioning treatment, and postgrafting immunosuppressive treatment with methotrexate provided the basis of success in the treatment of acute leukemia with allogeneic marrow transplantation.4,5  In these patients, Weiden et al described the beneficial effect of GVHD on the incidence of leukemia relapse.6  In patients with chronic GVHD, this reduced relapse was even associated with a survival benefit.7  The role of T cells became evident when depletion of T cells from the graft was introduced into the clinical practice for prevention of GVHD.8,9  In chronic myelogenous leukemia (CML), the beneficial effect of T-cell depletion on GVHD was negated because of an increased relapse rate.10  Increased relapse after T-cell depletion was most pronounced in CML, less in acute myeloid leukemia (AML), and lowest in acute lymphoblastic leukemia (ALL).11 

The challenge of allogeneic stem cell transplantation for treatment of leukemia and other malignancies of the hematopoietic system is the prevention of GVHD without losing the graft-versus-leukemia (GVL) effect. One possible approach has been is to apply a delayed transfusion of T cells after T cell–depleted marrow transplantation. Previous experiments in stable canine chimeras showed that graft-versus-host tolerance was not abrogated by donor lymphocyte transfusions (DLTs).12  In DLA-identical littermate chimeras, we analyzed at which time after T cell–depleted marrow transplantation we could transfuse donor lymphocytes without producing GVHD. In the first days and weeks, we produced fatal GVHD by DLTs, at 2 months, and later we could transfuse large numbers of donor lymphocytes without producing GVHD. Using small numbers of marrow cells, we could induce mixed chimerism, which was converted to complete chimerism after DLT.13  The results of these animal experiments encouraged us in October 1988 to treat a patient with relapse of CML with DLT after interferon-α (IFN-α) treatment had failed.14  This was the first evidence that lymphocytes could induce lasting remissions without chemotherapy or radiotherapy. Another early case was reported by S. Slavin15  involving a patient treated in January 1987 for early relapse of ALL at one month after transplantation. These early successes stimulated research and wide clinical application of DLT for the treatment of leukemia, lymphoma, and myeloma. Moreover, the possibility of controlling residual disease by DLT has encouraged conditioning regimens of reduced intensity16,17  that are better tolerated by elderly and debilitated patients.

In patients with immune deficiency, blood transfusions can produce severe myelosuppression18  by a graft-versus-host reaction of donor blood unit T cells against recipient hematopoiesis. A GVL effect can also be obtained by transfusion-induced suppression of the host's hematopoiesis resulting from sharing of histocompatibility antigens with the leukemia. The primary targets of graft-versus-host reactions are cells of the hematopoietic system of the host. In dogs, mixed chimerism was studied as a model for GVL reactions in the form of a graft-versus-host hematopoiesis reaction.13  Similarly, mixed chimerism could be converted into complete chimerism by DLT in human patients.19  Several minor histocompatibility antigens (mHA) with a tissue distribution restricted to hematopoietic cells have been described20-22  that may elicit strong GVL effects.23  However, the gene frequency of minor histocompatibility antigens with restricted tissue distribution allows the use only in a small proportion of patients.

Donor T cells are stimulated by dendritic cells (DCs) of the host, through presentation of mHA (Figure 1). CD4+ helper T cells are stimulated by HLA class II peptide complexes of DCs. Stimulation of T cells requires additional signals via costimulatory molecules, adhesion molecules, and stimulatory cytokines, which may also produce acute GVHD-like changes by themselves.24  This GVHD-like reaction is seen in syngeneic transplants and some autologous transplantations. Normally, it subsides after several days of treatment with steroids. In contrast, in allogeneic transplantations, the reaction is maintained by the T-cell recognition of mHA. Intensive conditioning treatment with radiation and chemotherapy prematurely activates antigen-presenting cells (APCs) and leads to an accelerated stimulation of T helper cells. Similarly, bacterial and viral infections activate APCs via Toll-like receptors (TLRs).25  Indeed, activation of APCs and release of pro-inflammatory cytokines stimulate T helper cells, and their subsequent release of IFN-γ and granulocyte-macrophage colony-stimulating factor (GM-CSF) results in a vicious circle known as a “cytokine storm.”26  Tempering the cytokine storm by treatment with TNF-α antibody during the conditioning treatment suppressed acute GVHD27  or led to its delay. Similarly, delayed acute GVHD may occur in patients conditioned with reduced intensity to avoid up-regulation of costimulatory molecules on APCs and release of proinflammatory cytokines.

Figure 1

Pathophysiology of GVHD: the donor T cell is encountering a host antigen-presenting cell (APC: monocyte/dendritic cell). The APC is activated by the conditioning treatment with radiation and chemotherapy; kit may also be activated by infections: viral via Toll-like receptor-9 (TLR9), bacterial infection via lipopolysaccharide (LPS), CD14, and TLR4. The interaction is regulated by recognition of mHA in the context with CD4 and HLA class II. The reaction is stimulated by costimulatory molecules CD40-CD40 ligand, B7.1-CD28, and adhesion molecules ICAM and LFA3. Cytokine secretion includes tumor necrosis factor-α and IL-6 by the APC and IFN-γ and GM-CSF by the T cell.

Figure 1

Pathophysiology of GVHD: the donor T cell is encountering a host antigen-presenting cell (APC: monocyte/dendritic cell). The APC is activated by the conditioning treatment with radiation and chemotherapy; kit may also be activated by infections: viral via Toll-like receptor-9 (TLR9), bacterial infection via lipopolysaccharide (LPS), CD14, and TLR4. The interaction is regulated by recognition of mHA in the context with CD4 and HLA class II. The reaction is stimulated by costimulatory molecules CD40-CD40 ligand, B7.1-CD28, and adhesion molecules ICAM and LFA3. Cytokine secretion includes tumor necrosis factor-α and IL-6 by the APC and IFN-γ and GM-CSF by the T cell.

Close modal

Activation of myeloid DCs not only stimulates helper T cells but also stimulates CD8+ cytotoxic T cells through presentation of HLA class I–restricted peptides (Figure 2). Activated DCs provide a “license to kill” to cytotoxic T cells.28  This license is also required for the GVL reaction29  whereby expression of MHC class II by APCs and stimulation of CD4+ cells are necessary for a CD8-mediated GVL response. The role of MHC class I on host DCs in GVHD was stressed by Shlomchik et al.30  Host DCs deficient for MHC class I were unable to initiate CD8-associated GVHD. DCs of the graft that express MHC class I maximize GVHD through cross-presentation of host antigens; however, they could not optimize the GVL reaction in a murine CML model.31 

Figure 2

Pathophysiology of GVHD. The interaction of the donor T cell and the host APC leads to the activation of APC and immunogenic presentation of HLA class I–restricted minor histocompatibility antigens (“license to kill”) to activate CD8 cells of the transplantation that can attack mHA on epithelial tissue or also on hematopoietic tissue.

Figure 2

Pathophysiology of GVHD. The interaction of the donor T cell and the host APC leads to the activation of APC and immunogenic presentation of HLA class I–restricted minor histocompatibility antigens (“license to kill”) to activate CD8 cells of the transplantation that can attack mHA on epithelial tissue or also on hematopoietic tissue.

Close modal

DCs of leukemia origin have been demonstrated in CML,32-34  and they may present mHA23  and leukemia-specific peptides to donor lymphocytes.35  Most probably, the spontaneous differentiation of CML progenitor cells to DCs is one important factor in the good response of CML to DLT. Similarly, blasts of AML and MDS may differentiate spontaneously to DCs,36  or differentiation can usually be induced by stimulation with GM-CSF alone or in combination with other cytokines.37  In the presence of cytokines, differentiation of leukemia blasts toward DCs in vitro has been demonstrated to be independent of the karyotype. This finding indicates that the prognostic role of the karyotype may not apply to immunotherapy.37,38  In CML, the combination of GM-CSF and IFN-α has been better than the combination of GM-CSF and IL-4 for producing DCs presenting HLA class I–restricted peptides.39 

Delayed DLT takes advantage of the transplantation tolerance induced by stem cell transplantation (Figure 3). In long-term canine chimeras, transplantation tolerance could not be abrogated by DLT.12  In these animals, only T cells of donors sensitized against the recipients produced GVHD in some cases. Resistance to the induction of GVHD by DLT may be the result of replacement of host DCs by DCs produced by the graft or to the generation of nonstimulatory, immature DCs of the host that fail to stimulate naive donor T cells. The majority of patients with leukemia are age 40 years and older, and peripheral tolerance is more probable than central tolerance. Host DCs are down-regulated by regulatory T cells of donor origin (Figure 4). Peripheral tolerance is maintained by down-regulation of costimulatory molecules and production of IL-10 and transforming growth factor-β by DCs and IL-4 by T cells. This tolerance can be broken by T-cell depletion before DLT (H. Menzel, H.-J.K., S. Thierfelder, unpublished data, 1994).40,41  CD4,CD25,CD28 positive regulatory T cells of the donor42  as well as residual host T cells able to produce Fas-L, perforin, and IFN-γ have been found to be responsible for the GVHD resistance with delayed DLT.41  Depletion of T cells by prior chemotherapy increases the risk for GVHD caused DLT. In human patients with recurrent acute leukemia, immunosuppressive conditioning with fludarabine and cyclophosphamide before DLT produced significantly more GVHD.43  Patients given T cell–depleted stem cell transplantations have a higher risk of GVHD after DLT than those given unmanipulated transplantations.44  This supports the role of regulatory T cells controlling tolerance via DCs. On the other side, activation of the DCs by conditioning treatment with chemotherapy and radiotherapy before DLT and severe infections may activate the costimulatory mechanisms and produce GVHD after DLT (H.-J.K., C. Schmid, E. Weissinger, E. Holler, unpublished data, 2000).

Figure 3

Adoptive immunotherapy in chimeras.

Figure 3

Adoptive immunotherapy in chimeras.

Close modal
Figure 4

Graft-versus-host tolerance: donor T cells and “immature” host DCs are nonstimulatory. IL-10 and transforming growth factor-β are produced by the DC and IL-4 by the T cell; no costimulatory molecules are expressed by indoleamin 2,3 deoxygenase is released. This peripheral tolerance is maintained as long as there is no conditioning treatment with radiation or chemotherapy and no infection that may activate the DCs.

Figure 4

Graft-versus-host tolerance: donor T cells and “immature” host DCs are nonstimulatory. IL-10 and transforming growth factor-β are produced by the DC and IL-4 by the T cell; no costimulatory molecules are expressed by indoleamin 2,3 deoxygenase is released. This peripheral tolerance is maintained as long as there is no conditioning treatment with radiation or chemotherapy and no infection that may activate the DCs.

Close modal

GVL reactions are maintained by host DCs that stimulate donor T cells (Figure 5). T helper cells type I can themselves become cytotoxic or “license” the DCs to activate CD8+ donor cells. In CML patients, clinical responses have been seen in all patient groups with an allogeneic donor. In syngeneic twins, donor lymphocytes did not induce remissions.44  The best candidates for target antigens of the GVL reactions are mHA expressed on hematopoietic cells, such as HA-1, HA-2, HB-1, BCL2A1, and HB-145  (for review, see Bleakley and Riddell22 ). However, donor-recipient combinations with relevant differences for known mHA are a small minority of cases. Of higher clinical relevance are Y chromosome-coded antigens that are recognized by female donor cells. Unfortunately, most Y chromosome-coded proteins have a broad tissue expression, but UTY is only weakly expressed on nonhematopoietic cells and highly expressed on hematopoietic cells.46 

Figure 5

The GVL reaction is maintained by the interaction of host DCs of leukemia origin that presents mHA restricted to hematopoietic tissue or mHA with general distribution. These can be presented on epithelial cells at a lower concentration, leaving a hematopoiesis-restricted activity.

Figure 5

The GVL reaction is maintained by the interaction of host DCs of leukemia origin that presents mHA restricted to hematopoietic tissue or mHA with general distribution. These can be presented on epithelial cells at a lower concentration, leaving a hematopoiesis-restricted activity.

Close modal

Overexpressed differentiation antigens may also serve as targets for the GVL reaction. Examples of such targets are Wilms' tumor 1,47  the peptide PR-148  of proteinase 3 of granulocytes, and receptor for hyaluronic acid–mediated motility.49  Vaccination studies with Wilms' tumor 1 and PR-1 have shown responses in patients with AML.50  Overexpressed proteins are more antigenic when presented by foreign HLA molecules. The allogeneic situation can be simulated by transfection of allogeneic T-cell receptors against these proteins into autologous T cells.51 

The best results for DLT have been obtained in the treatment of relapse of CML after transplantation15,44,52-57  (Table 1). In most studies, between 70% and 80% of patients treated for hematologic and cytogenetic relapse obtained a complete cytogenetic response. In patients with cytogenetic or molecular relapse, defined as an increase of the quantitative reverse-transcribed polymerase chain reaction for bcr/abl, remissions could be rescued in 80% by early DLT treatment. Most remissions are durable, and the first patients treated with DLT for CML relapse in 1988 are still in remission. In more advanced stages of relapse, the rate of complete cytogenetic remission is lower and responses are not as durable. In the European Blood and Marrow Transplantation (EBMT) experience,44  responses were observed in all patient groups with allogeneic donors, HLA-identical siblings, HLA-mismatched family members, and unrelated HLA-matched donors. However, no response was observed in 4 patients with syngeneic monozygotic twin donors. The GVL effect appeared to be part of the GVH reaction; there was a correlation of GVL with GVHD, and the best responses were seen in patients with GVHD of grade more than or equal to II. However, 50% of patients had a complete cytogenetic remission without any signs of clinical GVHD. Therefore, there may be a GVL reaction separate from GVHD or a GVH reaction severe enough to manifest with clinical symptoms.

Table 1

Donor lymphocyte transfusion for treatment of relapse and persistence of malignancy

Disease/study cohortsNo. of patients responding/no. treated% CCR (y)Reference number/comments
CML molecular/cytogenetic relapse    
    EBMT study 40/50 80% (4 y) 44  
    North American 3/3  56  
Chronic phase    
    EBMT 88/114 60% (4 y) 44  
    North American 25/34  56  
    Japan 11/12 82% (3 y) 158  
Transformed phase    
    EBMT 13/36 20% (4 y) 44  
    North American 5/18  56  
    Japan 3/11 0% (3 y) 158  
AML/MDS    
    EBMT 15/58 15% (4 y) 44  
    North American 8/44  56  
    Prospective US study 25/51 19% (2 y) 88  
    Japan 13/32 7% (2/3 y) 95  
      33% (2/3 y)  
    Korean 10/17 31% (2 y) 162/chemotherapy + G-CSF mobilized DLI 
    Lille, France 2/14 2/14 (4 y) 92  
ALL    
    EBMT 3/20 0% (4 y) 44  
    North American 2/11 ND 56  
    IBMTR 11/44 13% (3 y) 160  
    Japan 6/23 0% (3 y) 95  
    Korean 7/10 10% (2 y) 161/chemotherapy + G-CSF mobilized DLI 
CLL    
    German Multicenter trial on molecular relapse and persistence 7/9 molecular remission 7/9 (> 2 y) 103  
    Bristol multicenter 1/7 19  
    DFCI 6/7 NK 102  
NHL    
    North American study 0/6 NK 56  
    EBMT study 10/14 OR NK 162  
    Progressive and refractory 6/14 CR   
    UC London LG-NHL 6/10 NK 163  
    Relapsed and refractory HG-NHL 3/9   
Myeloma    
    EBMT study 5/17 45% (2 y) 44  
    Relapse/progression    
    North American Study relapse/progression 2/4  56  
    US multicenter study persistent/progressive 7/22 4/7 (> 1 y) 113  
    Dutch multicenter study relapse and progression 14/27 OR 5/27 (> 2.5 y) 164  
 10/27 CR   
    Preemptive in chemosensitive MMY 6/20 CR/PR 30% (2 y) 165  
 → 7/14 CR/PR   
    Relapse/progression 24/63  117  
 12 CR ∼ 45% (3 y)   
 12 PR   
    Relapse/progression Johns Hopkins Hospital 8/16 5 > 2 y 166  
 6 CR 2PR   
Disease/study cohortsNo. of patients responding/no. treated% CCR (y)Reference number/comments
CML molecular/cytogenetic relapse    
    EBMT study 40/50 80% (4 y) 44  
    North American 3/3  56  
Chronic phase    
    EBMT 88/114 60% (4 y) 44  
    North American 25/34  56  
    Japan 11/12 82% (3 y) 158  
Transformed phase    
    EBMT 13/36 20% (4 y) 44  
    North American 5/18  56  
    Japan 3/11 0% (3 y) 158  
AML/MDS    
    EBMT 15/58 15% (4 y) 44  
    North American 8/44  56  
    Prospective US study 25/51 19% (2 y) 88  
    Japan 13/32 7% (2/3 y) 95  
      33% (2/3 y)  
    Korean 10/17 31% (2 y) 162/chemotherapy + G-CSF mobilized DLI 
    Lille, France 2/14 2/14 (4 y) 92  
ALL    
    EBMT 3/20 0% (4 y) 44  
    North American 2/11 ND 56  
    IBMTR 11/44 13% (3 y) 160  
    Japan 6/23 0% (3 y) 95  
    Korean 7/10 10% (2 y) 161/chemotherapy + G-CSF mobilized DLI 
CLL    
    German Multicenter trial on molecular relapse and persistence 7/9 molecular remission 7/9 (> 2 y) 103  
    Bristol multicenter 1/7 19  
    DFCI 6/7 NK 102  
NHL    
    North American study 0/6 NK 56  
    EBMT study 10/14 OR NK 162  
    Progressive and refractory 6/14 CR   
    UC London LG-NHL 6/10 NK 163  
    Relapsed and refractory HG-NHL 3/9   
Myeloma    
    EBMT study 5/17 45% (2 y) 44  
    Relapse/progression    
    North American Study relapse/progression 2/4  56  
    US multicenter study persistent/progressive 7/22 4/7 (> 1 y) 113  
    Dutch multicenter study relapse and progression 14/27 OR 5/27 (> 2.5 y) 164  
 10/27 CR   
    Preemptive in chemosensitive MMY 6/20 CR/PR 30% (2 y) 165  
 → 7/14 CR/PR   
    Relapse/progression 24/63  117  
 12 CR ∼ 45% (3 y)   
 12 PR   
    Relapse/progression Johns Hopkins Hospital 8/16 5 > 2 y 166  
 6 CR 2PR   

CML indicates chronic myelogenous leukemia; EBMT, European Blood and Marrow Transplantation; AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; ALL, acute lymphoblastic leukemia; ND, not done; CLL, chronic lymphocytic leukemia; DFCI, Dana Farber Cancer Institute; NHL, non-Hodgkin lymphoma; LG-NHL, low-grade non-Hodgkin lymphoma; HG-NHL, high-grade non-Hodgkin lymphoma; NK, not known; OR, overall response; CR, complete remission; PR, partial remission; MMY, multiple myeloma; and CCR, continuous complete remission.

It is noteworthy that the GVL effect may not be observed until 4 to 8 weeks after DLT. In some patients, the disease may even progress for a month before counts drop suddenly and remission is induced. Therefore, clinicians must be patient and not give up on DLT at less than 2 months. The time until molecular remission is achieved can be 4 to 6 months, and remissions have been observed more than 1 year after DLT. This delayed type of response could be the result of an ongoing immune reaction of donor T cells against the leukemia or the result of the elimination of an early leukemic stem cell whose progeny survive for months.

Risks of the treatment are the occurrence of GVHD and myelosuppression. The risk of GVHD of moderate and severe degree (≥ II°) was 34%. This risk is lower than that for GVHD after stem cell transplantation, despite the omission of immunosuppressive therapy. Prevention of GVHD has been attempted by depletion of CD8+ T cells.58,59  CD4+ T cells produce a sufficient GVL effect and CD8+ cells can be recruited in vivo.60  Others have transferred a suicide gene into the T cells,61,62  providing an opportunity to ablate T cells if GVHD occurs by treatment with a drug that leads to selective death of transduced T cells. The most widely accepted method for DLT is the transfusion of donor lymphocytes in escalating doses.63  It could be shown that less than or equal to 2 × 107/kg mononuclear blood cells produced less than 5% DLT-related mortality in patients with an HLA-matched donor.64  Recommended initial doses are 106/kg CD3+ T cells in patients with an HLA-identical sibling donor and 0.5 × 106/kg CD3+ T cells in patients with an HLA-matched unrelated donor. Considering the delayed response seen after DLT, escalation of the dose can be delayed for 2 months in patients without rapid progression of the disease. In more progressive forms of relapse, the interval may be shortened to 4 weeks because in most cases GVHD occurs within 4 weeks of DLT, if it occurs.

Myelosuppression was seen in approximately 10% of patients and was most pronounced in patients with hematologic relapse. It may be the consequence of eliminating host-type hematopoiesis without sufficient replacement by donor-type hematopoiesis. Transfusion of donor marrow without conditioning treatment restored hematopoiesis in some patients. In others, stem cell replacement was not effective. In these patients, it has been hypothesized that the stroma may have become deficient in supporting HSCs.

Our preferred method to treat recurrent CML after transplantation is the combination of IFN-α and DLT. Low dose IFN-α (106 U/day) is given daily together with escalating doses of donor lymphocytes, starting with 106/kg CD3+ T cells in HLA-identical sibling transplantations. Escalated doses of 5 × 106/kg and 10 × 106/kg are given at 2-month intervals, according to the clinical course. In patients with unrelated matched donors, the starting dose is 0.5 × 106/kg CD3+ T cells. In patients with HLA-haploidentical transplantations, 105/kg is advocated as the starting dose.65 

Patients not responding to the combination of IFN-α and DLT may still respond to the combination of interferon-α, GM-CSF, and DLT.66  However, it is very important that the disease be treated in a chronic phase. Five of 7 patients in chronic phase relapse responded with complete remission, whereas the combination failed in 2 patients and they were subjected to retransplantations without immunosuppression. One responder died of myelosuppression at 4 months. Of 3 patients with blast phase relapse, only one responded after the combination of chemotherapy with IFN-α, GM-CSF, and DLT, but she died of extensive chronic GVHD.

Remissions induced by DLT are mostly durable, with the longest survivors now alive more than 19 years after treatment. Yet relapses occur, as summarized for outcome of patients treated in Great Britain by Cummins et al.67  Although most patients responded again to the treatment with DLT, the role for imatinib in treatment of relapse is uncertain.

The selective tyrosine kinase inhibitor imatinib has revolutionized the treatment of CML by its high efficiency and low toxicity.68  Therefore, treatment with imatinib was given for relapse of CML in several studies instead of DLT.69,70  The majority of patients with hematologic relapse responded to imatinib with cytogenetic remissions. Several patients developed signs of GVHD and myelosuppression. The majority of patients experienced relapse after discontinuation of imatinib; however, single patients remained in remission. In our own experience, 7 of 10 patients treated with imatinib became negative in reverse-transcribed polymerase chain reaction. Six patients relapsed: 4 on treatment and 2 patients within 2 to 4 months after discontinuation of imatinib. However, 1 patient remained negative for more than 2 years.71 

Application of imatinib before transplantation provided excellent results in patients treated for advanced phase CML. Three of 6 patients with blast crisis who responded to imatinib remain in continued complete remission after transplantation.72 

The combination of DLT and imatinib has been advocated by some investigators73  for the treatment of recurrent CML after transplantation. This combination induced complete remissions in 11 patients, including 4 patients in advanced phase relapse; 7 patients were still on treatment at the time of reporting, whereas imatinib could be discontinued in 4 patients. Controversial results have been reported on the immunosuppressive effects of imatinib on T cells and APCs.74-79  In contrast, there is little doubt that imatinib spares leukemia stem cells.80,81  In a small study of 12 patients where imatinib was discontinued after 2 to 4 years, 6 patients remained in remission, whereas 6 patients relapsed within 6 months.82  It is possible that prior treatment with IFN-α contributed to the long-lasting success in some patients.

Treatment of relapsed AML and MDS with DLT has been less effective than chronic phase CML.44,56  The pace of the disease may be one factor because AML may outpace a GVL reaction that may need several months for effective control of leukemia. Therefore, most centers have used some form of chemotherapy before DLT. We have chosen low-dose cytarabine for 2 to 4 weeks to avoid a cytokine storm released by intensive therapy. However, some patients are progressive during low-dose cytarabine treatment and require more intensive treatment.83  Another factor for less favorable response to DLT is the immune phenotype of AML blasts. In more than 80% of patients, blasts do not express CD86 costimulatory molecules,38,84,85  potentially allowing blasts to escape immune attack. GM-CSF in combination with IFN-α and other cytokines is highly effective to produce DCs of leukemia origin.38,39,86  Therefore, we used GM-CSF in addition to granulocyte-CSF–mobilized blood cells for the treatment of relapses of AML after transplantation. In a retrospective analysis, we found a better response in patients with this combination than with chemotherapy or DLT alone (Figure 6). These results compare favorably with the results of a retrospective evaluation of the EBMT data.87  Levine et al88  performed a prospective multicenter study of treating AML relapse after allogeneic transplantation with chemotherapy and DLT. The best results were obtained in patients with relapse occurring more than 6 months after transplantation and with a response to chemotherapy (Figure 7). In our own study, several patients are alive after more than 4 years, a late death from leukemia was the result of refractory central nervous system (CNS) leukemia,83  and another patient developed testicular leukemia. These observations point to the fact that GVL does not eliminate leukemia in sanctuary sites; therefore, regular control and prophylactic treatment of the CNS are required even in patients with AML. After allogeneic stem cell transplantation, relapses of AML often occur at extramedullary sites without evidence of blasts in marrow and blood. These relapses rarely respond to DLT.

Figure 6

Evaluation of relapses of AML/MDS at the University of Munich Transplant Center. Retrospective analysis of the treatment with chemotherapy, donor lymphocyte transfusion, and the combination of low-dose cytarabine, mobilized blood stem cells, and GM-CSF.

Figure 6

Evaluation of relapses of AML/MDS at the University of Munich Transplant Center. Retrospective analysis of the treatment with chemotherapy, donor lymphocyte transfusion, and the combination of low-dose cytarabine, mobilized blood stem cells, and GM-CSF.

Close modal
Figure 7

Survival after chemotherapy and donor leukocyte infusions (DLIs) for patients with relapsed acute myelogenous leukemia (AML). Survival is significantly improved in recipients of DLI for relapsed AML more than 6 months after bone marrow transplantation (BMT; solid line) compared with recipients of DLI for relapse less than 6 months from BMT (dashed line; P < .001). Reprinted from Levine et al88  with permission.

Figure 7

Survival after chemotherapy and donor leukocyte infusions (DLIs) for patients with relapsed acute myelogenous leukemia (AML). Survival is significantly improved in recipients of DLI for relapsed AML more than 6 months after bone marrow transplantation (BMT; solid line) compared with recipients of DLI for relapse less than 6 months from BMT (dashed line; P < .001). Reprinted from Levine et al88  with permission.

Close modal

In a small series, 16 patients from Seattle with myelodysplastic syndromes (MDSs) were treated with DLT; 2 were not evaluable because of chemotherapy-induced remission. Three of 14 patients had a remission, but no patient survived because of GVHD or relapse.89  In a study of 25 patients with relapse of AML (N = 22) and MDS (N = 3), various strategies of DLT, second transplantations and chemotherapy were studied: 2 patients with MDS and one patient with AML survived more than 2 years or 1 year, respectively, after DLT and second transplantation.90  Positive results were also reported in an earlier study by Porter et al91  and more recently by Depil et al.92  The treatment of AML relapse with low-dose cytarabine, mobilized DLT, and GM-CSF is currently studied in a German multicenter trial. The results of our center were confirmed in a preliminary analysis.

The role of preemptive DLT has been studied in patients with high-risk AML treated with reduced intensity conditioning93  and allogeneic stem cell transplantation. DLT was administered to those patients who had survived 120 days after transplantation and were free of GVHD, immunosuppressive treatment, and infections. Escalating doses of 106/kg, 5 × 106/kg, and 107/kg CD3+ T cells were given at monthly intervals. Forty-six patients were treated. Reasons for not giving the next scheduled DLT were ongoing immunosuppression, presence of GVHD, and early relapse.94  The comparison in a matched-pair analysis showed a significantly better survival of patients given preemptive DLT (Figure 8).

Figure 8

Preemptive donor lymphocyte transfusions in high-risk AML; evaluation in a matched pair analysis. Courtesy of M. Schleuning, EBMT 2007.

Figure 8

Preemptive donor lymphocyte transfusions in high-risk AML; evaluation in a matched pair analysis. Courtesy of M. Schleuning, EBMT 2007.

Close modal

The response of standard B-cell or pre–B-cell ALL to DLT is generally inferior to that of myeloid leukemia,44,56,95  although the first patient treated successfully by DLT for residual leukemia was a child with ALL.15  Hematologic responses to DLT alone are rare96 ; the rare prolonged remissions have been observed primarily in patients treated with intensive chemotherapy or radiotherapy. The response rate of relapsed ALL to chemotherapy and DLT may not be so different from AML, but the duration of remission is generally short. Presumably, the GVL effect has to occur early after transplantation to be effective. In a treatment model of ALL in nonobese diabetic/severe combined immunodeficiency mice, Nijmeijer et al found donor CTL to be turned off after a few weeks resulting in tumor regrowth.97  The GVL effect was strongest if CTL and tumor cells were HLA-incompatible. The limited efficacy of DLT against ALL may be associated with the pre-B lymphocyte phenotype that lacks costimulatory molecules and induces tolerance.98 

In contrast, several cases of T-ALL and T-cell lymphoma have been reported to respond to DLT,99,100  with durable responses. The GVL effect of acute GVHD is strong in B- and T-ALL,101  with the effect being stronger in T-ALL, although not significantly. We have observed several patients with durable responses to DLT in T-ALL and mature T-cell lymphoma.

Chronic lymphocytic leukemia is of B-cell origin in most of the cases. Single cases of a strong response to DLT have been reported in patients with recurrent clinical disease after allogeneic stem cell transplantation.102  However, in most cases, the beneficial effect of DLT has been observed at the molecular level,103  stressing the importance of the measurement of minimal residual disease. In 7 of 9 patients, durable molecular remissions were induced by DLT, whereas molecular remissions were seen in only 6 of 26 patients not given DLT, and these were not durable.

The graft-versus-lymphoma effect has been questioned in a statistical analysis of the International Bone Marrow Transplant Registry and EBMT.104  The results of syngeneic and allogeneic transplantation were compared in low-grade and high-grade lymphoma. In low-grade lymphoma, there was no difference in relapse rate in allogeneic and syngeneic transplantations; these patients had a lower relapse rate than in autologous transplantations. In high-grade lymphoma, the differences in relapse rates after allogeneic, syngeneic, and autologous transplantations were not significant.

The evidence for an allogeneic graft-versus-lymphoma effect has been discussed by Grigg and Ritchie.105  There is good evidence for graft-versus-lymphoma effects in mantle cell lymphoma,106  follicular lymphoma, and chronic lymphocytic leukemia.107  Reduced intensity conditioning and preemptive DLT were studied by Peggs et al, revealing strong evidence for a graft-versus-lymphoma effect in lymphoma, including Hodgkin disease.108  High response rates were also reported in low-grade lymphoma, in some cases even without signs of GVHD (Table 2).109 

Table 2

Evidence for a graft-versus-lymphoma effect

LymphomaNo. of patientsin CCREffectReference (year)
Follicular 8/13 DLI Marks et al19  (2002) 
Mantle cell 15/18 DLI Khouri et al167  (2003) 
High grade 8/9 vs 0/9 T-cell depletion Glass et al168  (2004) 
Hodgkin 5/7 DLI Peggs et al169  (2007) 
T-NHL 12/17 RIC + DLI Corradini et al99  (2004) 
CLL 8/9 MRD Ritgen et al103  (2004) 
LymphomaNo. of patientsin CCREffectReference (year)
Follicular 8/13 DLI Marks et al19  (2002) 
Mantle cell 15/18 DLI Khouri et al167  (2003) 
High grade 8/9 vs 0/9 T-cell depletion Glass et al168  (2004) 
Hodgkin 5/7 DLI Peggs et al169  (2007) 
T-NHL 12/17 RIC + DLI Corradini et al99  (2004) 
CLL 8/9 MRD Ritgen et al103  (2004) 

DLI indicates donor lymphocyte transfusion; RIC, reduced intensity conditioning; MRD, minimal residual disease; T-NHL, T-cell non-Hodgkin lymphoma; CLL, chronic lymphocytic leukemia; and CCR, continuous complete remission.

Multiple myeloma is a disease responsive to the treatment with donor lymphocytes.44,56,110-112  In 20% to 50% of cases, the disease responded to the treatment with donor lymphocytes (Table 1). However, most remissions were not durable,59,113-115  and the strongest prognostic factor predicting response was the occurrence of GVHD.116,117  The best results can be achieved at a stage of minimal residual disease, in patients in remission after chemotherapy.118  Seven of 20 patients with complete remission (N = 4) and partial remission (N = 16) remained in continuous complete remission more than 4 years after partial T cell–depleted stem cell transplantation and preemptive donor lymphocyte transfusions (actuarial 6-year survival, 29%). The response of multiple myeloma to donor lymphocyte transfusion is not predictable and, in advanced disease relapses, often occurs outside the marrow cavity. Myeloma is a tumor with close interaction to the microenvironment that may determine the response to chemotherapy.119  This interaction protects myeloma cells from regular chemotherapy, whereas new drugs may disrupt this protection. It is not known how allogeneic T cells affect this interaction. Recently, evidence has been provided that myeloma relapses from CD20+ and CD27+ memory B cells with distinct properties of stem cells.120  These B lymphoid stem cells may also be the relevant targets of the donor lymphocyte attack. Similar to low-grade lymphoma, long-term remissions are most probably obtained in patients with minimal residual disease.

The largest experience using adoptive immunotherapy for solid tumors has been obtained in renal cell carcinoma.121  A review of allografts for solid tumors performed at EBMT centers lists renal cell cancer as the most frequent indication (335 patients), followed by breast cancer (143 patients), neuroblastoma (70 patients), and ovarian carcinoma (40 patients).122  The original study by Childs et al121  provided unequivocal evidence for a graft-versus-tumor effect of DLT in renal cell cancer. In several patients with clear cell cancer, complete remissions were achieved and maintained.123  Evidence of a graft-versus-tumor effect has been reported in many other solid tumors.124  In general, less advanced disease, good clinical performance status of the patient, and the occurrence of GVHD were prognostic factors for response.125  Animal studies in mice126  and rats127  indicated that CD8+ T cells were effective in tumor control after T cell–depleted marrow transplantation. In human patients with metastatic breast cancer, DLT after T cell–depleted stem cell transplantation produced a graft-versus-tumor effect.128  Solid tumors may express mHA otherwise restricted to hematopoietic cells.129  In patients with renal cell cancer that responded to allografting, CD8 T cells have recently been found that recognize peptides from endogenous human retrovirus specific for renal cell cancer.123  In solid tumors, the role of stroma can be critical; it maintains viability of tumor stem cells by paracrine cytokine stimulation and fostering interaction. On the other hand, it may be relevant in antigen presentation130  and a target of effector mechanisms.131 

Transplantations from HLA-haploidentical donors take advantage of the antileukemic activity of NK cells. NK cells are inhibited by cross-reactive groups of self-HLA-antigens via an inhibitory killer immunoglobulin-like receptor.132  In the case of the patient's cells expressing antigens of a different group from the donor, the transplanted NK cells are not inhibited and exert a strong GVL effect. NK cells can eliminate host hematopoietic cells, including leukemia cells, without producing GVHD. The GVL effect of NK cells is most pronounced in AML and is less evident in ALL. Several groups could not show the beneficial effect of NK cells,133  but these groups did not use the extensive T-cell depletion used by the Perugia group.132 

In our own group of patients with advanced leukemia, we observed a very good response in ALL patients. However, we did not use T-cell depletion of marrow, and we added CD6-depleted mobilized blood cells on day 6. In the CD6-depleted preparation, we retained CD34+ stem cells, NK cells, and a minority of CD8+ T cells. In this way, a large number of cells could be given without an increased risk of GVHD. In this cohort of patients, we observed NK activity of grafts from donors that were heterozygous for the cross-reactive group C KIR-ligand in recipients homozygous for a cross-reactive group C-KIR ligand. Moreover, we observed a better GVL effect for cells from female donors given to male recipients and with cells from mothers compared with fathers, without an increase of GVHD. The advantage of female and maternal donors may be the result of memory T cells in the transplantation recognizing mHA in the patient.

Most experience in the treatment of viral infections after allogeneic transplantation has been accumulated in cytomegalovirus (CMV)134  and Epstein-Barr virus (EBV) infections.135  Both infections can be life-threatening, and reactivation of EBV may produce highly malignant lymphoblastic lymphoma. Virostatic drugs may control lytic infections of CMV and EBV, but posttransplant lymphoproliferative disease and lymphoma are associated with latent EBV infection. CD20 antibody is able to kill B cells immortalized by EBV, but lasting protection is only provided by T cells. Virus-reactive T cells can be selected from seropositive donors by tetramers136  and immunomagnetic bead selection, less than 104/kg T cells are sufficient to protect the patient. These cells proliferate in the patient within 1 to 2 weeks to protective levels. Similarly, EBV infections have responded to EBV-specific T cells selected from the blood of the stem cell donor by the gamma-IFN capture technique137  (Moosmann et al, in preparation). One requirement for these successful attempts at treating viral infections with these techniques is preexisting seropositivity of the stem cell donor, indicating immunologic memory for the virus.

The role of T-cell memory has been stressed by the lower relapse rate of male patients given female transplantations.138  The lower relapse rate was also evident after adjustment for GVHD. Recently, we have found a significantly lower relapse rate in male patients transplanted with marrow and blood cells from HLA-haploidentical female donors96  (H.-J.K., I. Bigalke, D. Ter Meer, A. Hausmann, C. Falk, B. Simoes, R. Buhmann, unpublished data, 2008). Moreover, recipients of maternal transplantations had less relapse than recipients of paternal transplantations. Memory T cells in the blood of mothers may recognize mHA on the leukemia of their children. Recently, the survival of central memory T cells has been shown to be independent of CD4 help; CMV-reactive CD8+, CD28+, Fas high central memory T cells survive in blood, marrow, and lymph node of macaques.139  Therefore, the ideal effector cell of GVL reactions is the central memory T cell.

Patients with CML surviving after allogeneic transplantation more than 10 years may be cured, but relapses after more than 10 years have been reported in single cases.140  Normal stem cells may survive and host-type hematopoiesis may reappear during severe infections without evidence of relapse.141  The reports on the elimination of leukemia stem cells are controversial. mHA-specific CD8+ T cells eliminate progenitor cells forming colonies in vitro142  and stem cells inducing leukemia in nonobese diabetic/severe combined immunodeficiency mice.143,144  CML patients responding to DLT produce antibodies against stem cell antigens CML 28 and CML 66145 ; in others, antigens on progenitor cells are not recognized.146  In myeloma, progression-free survival for many years after allogeneic transplantation has been observed in some patients who were refractory to high-dose chemotherapy and autologous transplantation. Persistence of monoclonal immunoglobulin at a low level indicated that the tumor was not eradicated, but it was controlled for several years until relapse occurred. Presumably DLT established immune control for a prolonged period of time.

Unfortunately, many patients develop a relapse of their leukemia, lymphoma, or myeloma after allogeneic transplantation and DLT (Table 3). It is not infrequent that these relapses occur at extramedullary sites as chloromas or plasmocytomas. These sites can be immunologically privileged sites, including CNS or gonads or other sites, such as skin, kidneys, and any other site outside of the marrow. Therefore, it is necessary to survey spinal fluid and gonads and to teach the patient self-examination. Intrathecal medication should be given in every patient treated by DLT for relapse of acute leukemia. Unfortunately, extramedullary infiltrates do not respond sufficiently to immunotherapy. There are no ideal methods of treatment; some patients respond to acute GVHD on discontinuation of immunosuppressive therapy. The preferred method is local treatment with radiotherapy in limited infiltrates. More extended infiltrations usually require intensive chemotherapy.

Table 3

Mechanisms of immune escape of leukemia

Invasion of immunologically privileged sites: CNS, gonads 
Down-regulation of HLA class I and class II expressions 
Deficient processing and presentation of peptides 
Deficient expression of costimulatory molecules: CD80, CD83, CD86, CD40, LFA-1, ICAM 
Secretion of inhibitory cytokines by leukemia cells: IL-10, TGF-β 
Abnormal secretion of and resistance to pro-inflammatory cytokines: TNF-α, IFN-γ 
Nonfunctional FAS on leukemia cells 
FAS-L expression by leukemia cells 
Regulatory T cells and production of indoleamin 2,3-dioxygenase (IDO) by leukemia cells 
Down-regulation of ζ-chain (lymphoma and CML) and ϵ-chain (CML) of the T-cell receptor 
Down-regulation of CD28 in AML 
Down-regulation of IL-2 receptor 
Low IL-2 
Apoptosis of high-avidity T cells 
Invasion of immunologically privileged sites: CNS, gonads 
Down-regulation of HLA class I and class II expressions 
Deficient processing and presentation of peptides 
Deficient expression of costimulatory molecules: CD80, CD83, CD86, CD40, LFA-1, ICAM 
Secretion of inhibitory cytokines by leukemia cells: IL-10, TGF-β 
Abnormal secretion of and resistance to pro-inflammatory cytokines: TNF-α, IFN-γ 
Nonfunctional FAS on leukemia cells 
FAS-L expression by leukemia cells 
Regulatory T cells and production of indoleamin 2,3-dioxygenase (IDO) by leukemia cells 
Down-regulation of ζ-chain (lymphoma and CML) and ϵ-chain (CML) of the T-cell receptor 
Down-regulation of CD28 in AML 
Down-regulation of IL-2 receptor 
Low IL-2 
Apoptosis of high-avidity T cells 

Different mechanisms can contribute to the failure of DLT to eradicate leukemia. Leukemia cells may persist in immunologically privileged sites, such as the CNS and gonads, or in extramedullary sites, such as the skin or the kidney, while still showing a continued remission in marrow. Malignant cells may escape immune detection and elimination if they have an altered expression of target antigens or costimulatory molecules required for efficient recognition. They may also directly down-regulate effector cells through secretion of inhibitory cytokines. Modulation of expression of FAS or FAS-ligand can also contribute to tumor escape from immune control.

There are many ways leukemia and other tumor cells camouflage themselves from the immune system, including changes in antigen presentation,38,84,85,147-149  production of inhibitory cytokines,150-152  and the production of regulatory T cells by indoleamin 2,3 deoxygenase secretion and local tryptophan depletion.153 

T-cell effector functions may be influenced by the leukemia cells. High avidity T cells disappear during active disease and reappear in interferon-α responders.154  Defective effector functions of T cells, which were associated with diminished expression of the CD3-ζ and CD3-ϵ chains, have also been described.155,156  Decreased expression of CD3-ζ and CD3-ϵ was more probable on lymphocytes of patients with advanced disease. Diminished expression of CD3-ζ and CD28 was also observed in patients with advanced malignancies, chronic viral infections, or autoimmune diseases, occurring as a consequence of chronic lymphocyte stimulation.157  T cells with these phenotypes were prone to apoptosis, but they could be rescued with IFN-α and IL-2.156 

In the past decades, the paradigm of stem cell transplantation has changed. Nowadays confidence in the power of the GVL effect has encouraged the use of donor lymphocytes and mobilized donor cells for adoptive immunotherapy in cases of relapse or preemptively in patients with high relapse risk. The conditions for a GVL effect without severe GVHD have become better defined. Furthermore, it has become clear that myeloid diseases have a better response to DLT than lymphoid diseases and that this response can be improved by simultaneous treatment with IFN-α and/or GM-CSF. The better response to DLT from female donors and maternal donors suggests a role of memory T cells that may also control lymphoid leukemia. Obviously, these memory T cells should not produce vigorous GVHD. In the near future, we will learn more about these cells and the antigens they recognize.

To achieve maximum GVL effects, it will also be necessary to perform stem cell transplantation without immune suppression after transplantation. The intensity of conditioning should only be reduced to a level that allows prompt and permanent engraftment of T cell–depleted transplantations. Learning about the immune repertoire of our donors will allow treatment of patients with hematologic malignancies as well as those with other tumors. In this way, we will be able to select our donors not only according to optimum histocompatibility, but also to the relevant T-cell repertoire.

The author thanks Dolores Schendel, PhD, Helmholtz Zentrum Muenchen, and Jamie Ferrara, MD, PhD, University of Michigan, for critical review of the manuscript.

This work was supported by Deutsche Forschungsgemeinschaft (SFB TRR 36), Deutsche Krebshilfe, Wilhelm Sander Stiftung, Deutsche José Carreras Leukämie Stiftung, and EU-Contract Marie Curie Program.

Contribution: H.-J.K. wrote the paper.

Conflict-of-interest disclosure: The author declares no competing financial interests.

Correspondence: Hans-Jochem Kolb, Hematopoietic Cell Transplantation, Department of Medicine 3, University of Munich & Helmholtz Zentrum Muenchen-National Research Centre for Environmental Health, Munich, Germany; e-mail: Hans.kolb@med.uni-muenchen.de.

1
Burchenal
 
JH
Oettgen
 
HF
Holmberg
 
EA
Hemphill
 
SC
Reppert
 
JA
Effect of total-body irradiation on the transplantability of mouse leukemias.
Cancer Res
1960
, vol. 
20
 (pg. 
425
-
430
)
2
Barnes
 
DHW
Loutit
 
JF
Treatment of murine leukaemia with X-rays and homologous bone marrow.
Br J Haematol
1957
, vol. 
3
 (pg. 
241
-
252
)
3
Mathé
 
G
Amiel
 
JL
Schwarzenberg
 
L
Cattan
 
A
Schneider
 
M
Adoptive immunotherapy of acute leukemia: experimental and clinical results.
Cancer Res
1965
, vol. 
25
 (pg. 
1525
-
1530
)
4
Thomas
 
ED
Storb
 
R
Clift
 
RA
, et al. 
Bone marrow transplantation.
N Engl J Med
1975
, vol. 
292
 (pg. 
832
-
843
)
5
Thomas
 
ED
Buckner
 
CD
Banaji
 
M
, et al. 
One hundred patients with acute leukemia treated by chemotherapy, total body irradiation and allogeneic marrow transplantation.
Blood
1977
, vol. 
49
 (pg. 
511
-
533
)
6
Weiden
 
PL
Flournoy
 
N
Thomas
 
ED
, et al. 
Antileukemic effects of graft versus host disease in human recipients of allogeneic marrow grafts.
N Engl J Med
1979
, vol. 
300
 (pg. 
1068
-
1070
)
7
Weiden
 
PL
Sullivan
 
KM
Flournoy
 
N
, et al. 
Antileukemic effect of chronic graft-versus-host disease: contribution to improved survival after allogeneic marrow transplantation.
N Engl J Med
1981
, vol. 
304
 (pg. 
1529
-
1531
)
8
Rodt
 
H
Kolb
 
HJ
Netzel
 
B
, et al. 
GVHD suppression by incubation of bone marrow grafts with anti-T-cell globulin: effect in the canine model and application to clinical bone marrow transplantation.
Transplant Proc
1979
, vol. 
11
 (pg. 
962
-
966
)
9
Kolb
 
HJ
Rieder
 
I
Rodt
 
H
, et al. 
Antilymphocytic antibodies and marrow transplantation: VI. Graft- versus-host tolerance in DLA-incompatible dogs after in vitro treatment of bone marrow with absorbed antithymocyte globulin.
Transplantation
1979
, vol. 
27
 (pg. 
242
-
245
)
10
Apperley
 
JF
Mauro
 
FR
Goldman
 
JM
, et al. 
Bone marrow transplantation for chronic myeloid leukemia in first chronic phase: importance of a graft-versus-leukemia effect.
Br J Haematol
1988
, vol. 
69
 (pg. 
239
-
245
)
11
Horowitz
 
MM
Gale
 
RP
Sondel
 
PM
, et al. 
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
1990
, vol. 
75
 (pg. 
555
-
562
)
12
Weiden
 
PL
Storb
 
R
Tsoi
 
M-S
, et al. 
Infusion of donor lymphocytes into stable canine radiation chimeras: implications for mechanism of transplantation tolerance.
J Immunol
1976
, vol. 
116
 (pg. 
1212
-
1219
)
13
Kolb
 
HJ
Günther
 
W
Schumm
 
M
, et al. 
Adoptive immunotherapy in canine chimeras.
Transplantation
1997
, vol. 
63
 (pg. 
430
-
436
)
14
Kolb
 
HJ
Mittermuller
 
J
Clemm
 
C
, et al. 
Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients.
Blood
1990
, vol. 
76
 (pg. 
2462
-
2465
)
15
Slavin
 
S
Naparstek
 
E
Nagler
 
A
, et al. 
Allogeneic cell therapy with donor peripheral blood cells and recombinant human interleukin-2 to treat leukemia relapse after allogeneic bone marrow transplantation.
Blood
1996
, vol. 
87
 (pg. 
2195
-
2204
)
16
Giralt
 
S
Cohen
 
A
Mehra
 
R
, et al. 
Preliminary results of fludarabine/melphalan or 2CDA/melphalan as preparative regimens for allogeneic progenitor cell transplantation (ALLO-PCT) in poor candidates for conventional myeloablative conditioning [abstract].
Blood
1997
, vol. 
90
 pg. 
417a
 
17
McSweeney
 
PA
Wagner
 
JL
Maloney
 
DG
, et al. 
Outpatient PBSC allografts using immunosuppression with low dose TBI before, and cyclosporin (CSP) and mycophenolate mofetil (MMF) after transplant [abstract].
Blood
1998
, vol. 
92
 pg. 
519a
  
Abstract 2133
18
Anderson
 
KC
Weinstein
 
HJ
Transfusion-associated graft-versus-host disease.
N Engl J Med
1990
, vol. 
323
 (pg. 
315
-
321
)
19
Marks
 
DI
Lush
 
R
Cavenagh
 
J
, et al. 
The toxicity and efficacy of donor lymphocyte infusions given after reduced-intensity conditioning allogeneic stem cell transplantation.
Blood
2002
, vol. 
100
 (pg. 
3108
-
3114
)
20
Goulmy
 
E
Human minor histocompatibility antigens: new concepts for marrow transplantation and adoptive immunotherapy.
Immunol Rev
1997
, vol. 
157
 (pg. 
125
-
130
)
21
Warren
 
EH
Greenberg
 
PD
Riddell
 
SR
Cytotoxic T-lymphocyte-defined human minor histocompatibility antigens with a restricted tissue distribution.
Blood
1998
, vol. 
91
 (pg. 
2197
-
2207
)
22
Bleakley
 
M
Riddell
 
SR
Molecules and mechanisms of the graft-versus-leukaemia effect.
Nat Rev Cancer
2004
, vol. 
4
 (pg. 
371
-
380
)
23
Marijt
 
WA
Heemskerk
 
MHM
Kloosterboer
 
FM
, et al. 
Hematopoiesis-restricted minor histocompatibility antigens HA-1 or HA-2-specific T cells can induce complete remissions of relapsed acute leukemia.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 (pg. 
2742
-
2747
)
24
Teshima
 
T
Ordemann
 
R
Reddy
 
P
, et al. 
Acute graft-versus-host disease does not require alloantigen expression on host epithelium.
Nat Med
2002
, vol. 
8
 (pg. 
575
-
581
)
25
Antin
 
JH
Ferrara
 
JL
Cytokine dysregulation and acute graft-versus-host disease.
Blood
1992
, vol. 
80
 (pg. 
2964
-
2968
)
26
Ferrara
 
JLM
Deeg
 
HJ
Graft-versus-host disease.
N Engl J Med
1991
, vol. 
324
 (pg. 
667
-
674
)
27
Holler
 
E
Kolb
 
HJ
Mittermüller
 
J
, et al. 
Modulation of acute graft-versus-host disease after allogeneic bone marrow transplantation by tumor necrosis factor (TNF) release in the course of pretransplant conditioning: role of conditioning regimens and prophylactic application of a monoclonal antibody neutralizing human TNF (MAK 195F).
Blood
1995
, vol. 
86
 (pg. 
890
-
899
)
28
Ridge
 
JP
Di Rosa
 
F
Matzinger
 
P
A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell [see comments].
Nature
1998
, vol. 
393
 (pg. 
474
-
478
)
29
Chakraverty
 
R
Eom
 
HS
Sachs
 
J
, et al. 
Host MHC class II+ antigen-presenting cells and CD4 cells are required for CD8-mediated graft-versus-leukemia responses following delayed donor leukocyte infusions.
Blood
2006
, vol. 
108
 (pg. 
2106
-
2113
)
30
Shlomchik
 
WD
Couzens
 
MS
Tang
 
CB
, et al. 
Prevention of graft versus host disease by inactivation of host antigen-presenting cells.
Science
1999
, vol. 
285
 (pg. 
412
-
415
)
31
Matte
 
CC
Liu
 
J
Cormier
 
J
, et al. 
Donor APCs are required for maximal GVHD but not for GVL.
Nat Med
2004
, vol. 
10
 (pg. 
987
-
992
)
32
Eibl
 
B
Schwaighofer
 
H
Nachbaur
 
D
, et al. 
Evidence for a graft-versus-tumor effect in a patient treated with marrow ablative chemotherapy and allogeneic bone marrow transplantation for breast cancer.
Blood
1996
, vol. 
88
 (pg. 
1501
-
1508
)
33
Smit
 
WM
Rijnbeck
 
M
van Bergen
 
CAM
, et al. 
Dendritic cells generated from FACS sorted chronic myeloid leukemia (CML) precursor cells express BCR/ABL, and are potent stimulators for allogeneic T cells [abstract].
Br J Haematol
1996
, vol. 
93
 pg. 
313a
  
Abstract 1186
34
Chen
 
X
Regn
 
S
Kolb
 
H-J
Roskrow
 
M
The generation of leukemic dendritic cells from patients with chronic myeloid leukemia (CML) using the combination of interferon-α and GM-CSF [abstract].
Blood
1999
, vol. 
94
 pg. 
529a
 
35
Clark
 
RE
Dodi
 
IA
Hill
 
SC
, et al. 
Direct evidence that leukemic cells present HLA-associated immunogenic peptides derived from the BCR-ABL b3a2 fusion protein.
Blood
2001
, vol. 
98
 (pg. 
2887
-
2893
)
36
Mittermueller
 
J
Kolb
 
HJ
Gerhartz
 
HH
Wilmanns
 
W
In vivo differentiation of leukemic blasts and effect of low dose ara-c in a marrow grafted patient with leukemic relapse.
Br J Haematol
1986
, vol. 
62
 (pg. 
757
-
762
)
37
Schmetzer
 
HM
Kremser
 
A
Loibl
 
J
Kroell
 
T
Kolb
 
HJ
Quantification of ex vivo generated dendritic cells (DC) and leukemia-derived DC contributes to estimate the quality of DC, to detect optimal DC-generating methods or to optimize DC-mediated T-cell-activation-procedures ex vivo or in vivo.
Leukemia
2007
, vol. 
21
 (pg. 
1338
-
1341
)
38
Woiciechowsky
 
A
Regn
 
S
Kolb
 
H-J
Roskrow
 
M
Leukemic dendritic cells generated in the presence of FLT3 ligand have the capacity to stimulate an autologous leukaemia-specific cytotoxic T cell response from patients with acute myeloid leukaemia.
Leukemia
2001
, vol. 
15
 (pg. 
246
-
255
)
39
Chen
 
X
Regn
 
S
Raffegerst
 
S
Kolb
 
HJ
Roskrow
 
M
Interferon alpha in combination with GM-CSF induces the differentiation of leukaemic antigen-presenting cells that have the capacity to stimulate a specific anti-leukaemic cytotoxic T-cell response from patients with chronic myeloid leukaemia.
Br J Haematol
2000
, vol. 
111
 (pg. 
596
-
607
)
40
Johnson
 
BD
Becker
 
EE
LaBelle
 
JL
Truitt
 
RL
Role of immunoregulatory donor T cells in suppression of graft-versus-host disease following donor leukocyte infusion therapy.
J Immunol
1999
, vol. 
163
 (pg. 
6479
-
6487
)
41
Blazar
 
BR
Lees
 
CJ
Martin
 
PJ
, et al. 
Host T cells resist graft-versus-host disease mediated by donor leukocyte infusions.
J Immunol
2000
, vol. 
165
 (pg. 
4901
-
4909
)
42
Johnson
 
BD
Konkol
 
MC
Truitt
 
RL
CD25+ immunoregulatory T-cells of donor origin suppress alloreactivity after BMT.
Biol Blood Marrow Transplant
2002
, vol. 
8
 (pg. 
525
-
535
)
43
Miller
 
JS
Weisdorf
 
DJ
Burns
 
LJ
, et al. 
Lymphodepletion followed by donor lymphocyte infusion (DLI) causes significantly more acute graft-versus-host disease than DLI alone.
Blood
2007
, vol. 
110
 (pg. 
2761
-
2763
)
44
Kolb
 
HJ
Schattenberg
 
A
Goldman
 
JM
, et al. 
Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients.
Blood
1995
, vol. 
86
 (pg. 
2041
-
2050
)
45
Dolstra
 
H
Fredrix
 
H
Maas
 
F
, et al. 
A human minor histocompatibility antigen specific for B cell acute lymphoblastic leukemia.
J Exp Med
1999
, vol. 
189
 (pg. 
301
-
308
)
46
Warren
 
EH
Gavin
 
MA
Simpson
 
E
, et al. 
The human UTY gene encodes a novel HLA-B8-restricted H-Y antigen.
J Immunol
2000
, vol. 
164
 (pg. 
2807
-
2814
)
47
Gao
 
L
Xue
 
SA
Hasserjian
 
R
, et al. 
Human cytotoxic T lymphocytes specific for Wilms' tumor antigen-1 inhibit engraftment of leukemia-initiating stem cells in non-obese diabetic-severe combined immunodeficient recipients.
Transplantation
2003
, vol. 
75
 (pg. 
1429
-
1436
)
48
Molldrem
 
J
Dermime
 
S
Parker
 
K
, et al. 
Targeted T-cell therapy for human leukemia: cytotoxic T lymphocytes specific for a peptide derived from proteinase 3 preferentially lyse human myeloid leukemia cells.
Blood
1996
(pg. 
2450
-
2457
)
49
Greiner
 
J
Schmitt
 
M
Li
 
L
, et al. 
Expression of tumor-associated antigens in acute myeloid leukemia: implications for specific immunotherapeutic approaches.
Blood
2006
, vol. 
108
 (pg. 
4109
-
4117
)
50
Mailander
 
V
Scheibenbogen
 
C
Thiel
 
E
, et al. 
Complete remission in a patient with recurrent acute myeloid leukemia induced by vaccination with WT1 peptide in the absence of hematological or renal toxicity.
Leukemia
2004
, vol. 
18
 (pg. 
165
-
166
)
51
Stauss
 
HJ
Immunotherapy with CTLs restricted by nonself MHC.
Immunol Today
1999
, vol. 
20
 (pg. 
180
-
183
)
52
Hertenstein
 
B
Wiesneth
 
M
Novotny
 
J
, et al. 
Interferon alpha and donor buffy coat transfusions for treatment of relapsed chronic myeloid leukemia after allogeneic marrow transplantation [abstract].
Transplantation
1993
, vol. 
56
 (pg. 
1114
-
1118
)
53
Helg
 
C
Roux
 
E
Beris
 
P
, et al. 
Adoptive immunotherapy for recurrent CML after BMT.
Bone Marrow Transplant
1993
, vol. 
12
 (pg. 
125
-
129
)
54
Bar
 
BMAM
Schattenberg
 
A
Mensink
 
EJBM
, et al. 
Donor leukocyte infusions for chronic myeloid leukemia relapsed after allogeneic bone marrow transplantation.
J Clin Oncol
1993
, vol. 
11
 (pg. 
513
-
519
)
55
Porter
 
DL
Roth
 
MS
McGarigle
 
C
Ferrara
 
JLM
Antin
 
JH
Induction of graft-versus-host disease as immunotherapy for relapsed chronic myeloid leukemia.
N Engl J Med
1994
, vol. 
330
 (pg. 
100
-
106
)
56
Collins
 
RH
Shpilberg
 
O
Drobyski
 
WR
, et al. 
Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation.
J Clin Oncol
1997
, vol. 
15
 (pg. 
433
-
444
)
57
Bacigalupo
 
A
Soracco
 
M
Vassallo
 
F
, et al. 
Donor lymphocyte infusions (DLI) in patients with chronic myeloid leukemia following allogeneic bone marrow transplantation.
Bone Marrow Transplant
1997
, vol. 
19
 (pg. 
927
-
932
)
58
Giralt
 
S
Hester
 
J
Huh
 
Y
, et al. 
CD8-depleted donor lymphocyte infusion as treatment for relapsed chronic myelogenous leukemia after allogeneic bone marrow transplantation.
Blood
1995
, vol. 
86
 (pg. 
4337
-
4343
)
59
Alyea
 
EP
Soiffer
 
RJ
Canning
 
C
, et al. 
Toxicity and efficacy of defined doses of CD4(+) donor lymphocytes for treatment of relapse after allogeneic bone marrow transplant.
Blood
1998
, vol. 
91
 (pg. 
3671
-
3680
)
60
Orsini
 
E
Bellucci
 
R
Alyea
 
EP
, et al. 
Expansion of tumor-specific CD8+ T cell clones in patients with relapsed myeloma after donor lymphocyte infusion.
Cancer Res
2003
, vol. 
63
 (pg. 
2561
-
2568
)
61
Bonini
 
C
Verzelletti
 
S
Servida
 
P
, et al. 
Transfer of HSV-TK gene into donor peripheral blood lymphocytes for in vivo immunomodulation of donor anti-tumor immunity after allo-BMT [abstract].
Blood
1994
, vol. 
84
 
Suppl
pg. 
110a
 
62
Ciceri
 
F
Bonini
 
C
Gallo-Stampino
 
C
Bordignon
 
C
Modulation of GvHD by suicide-gene transduced donor T lymphocytes: clinical applications in mismatched transplantation.
Cytotherapy
2005
, vol. 
7
 (pg. 
144
-
149
)
63
Mackinnon
 
S
Papadopoulos
 
EB
Carabasi
 
MH
, et al. 
Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease.
Blood
1995
, vol. 
86
 (pg. 
1261
-
1268
)
64
Guglielmi
 
C
Arcese
 
W
Dazzi
 
F
, et al. 
Donor lymphocyte infusion for relapsed chronic myelogenous leukemia: prognostic relevance of the initial cell dose.
Blood
2002
, vol. 
100
 (pg. 
397
-
405
)
65
Lewalle
 
P
Triffet
 
A
Delforge
 
A
, et al. 
Donor lymphocyte infusions in adult haploidentical transplant: a dose finding study.
Bone Marrow Transplant
2003
, vol. 
31
 (pg. 
39
-
44
)
66
Kolb
 
HJ
Rank
 
A
Chen
 
X
, et al. 
In-vivo generation of leukaemia-derived dendritic cells.
Best Pract Res Clin Haematol
2004
, vol. 
17
 (pg. 
439
-
451
)
67
Cummins
 
M
Cwynarski
 
K
Marktel
 
S
, et al. 
Management of chronic myeloid leukaemia in relapse following donor lymphocyte infusion induced remission: a retrospective study of the Clinical Trials Committee of the British Society of Blood & Marrow Transplantation (BSBMT).
Bone Marrow Transplant
2005
, vol. 
36
 (pg. 
1065
-
1069
)
68
O'Brien
 
SG
Guilhot
 
F
Larson
 
RA
, et al. 
Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia.
N Engl J Med
2003
, vol. 
348
 (pg. 
994
-
1004
)
69
Kantarjian
 
HM
O'Brien
 
S
Cortes
 
JE
, et al. 
Imatinib mesylate therapy for relapse after allogeneic stem cell transplantation for chronic myelogenous leukemia.
Blood
2002
, vol. 
100
 (pg. 
1590
-
1595
)
70
Olavarria
 
E
Craddock
 
C
Dazzi
 
F
, et al. 
Imatinib mesylate (STI571) in the treatment of relapse of chronic myeloid leukemia after allogeneic stem cell transplantation.
Blood
2002
, vol. 
99
 (pg. 
3861
-
3862
)
71
Weisser
 
M
Tischer
 
J
Schnittger
 
S
, et al. 
A comparison of donor lymphocyte infusions or imatinib mesylate for patients with chronic myelogenous leukemia who have relapsed after allogeneic stem cell transplantation.
Haematologica
2006
, vol. 
91
 (pg. 
663
-
666
)
72
Weisser
 
M
Schleuning
 
M
Haferlach
 
C
Schwerdtfeger
 
R
Kolb
 
HJ
Allogeneic stem-cell transplantation provides excellent results in advanced stage chronic myeloid leukemia with major cytogenetic response to pre-transplant imatinib therapy.
Leuk Lymphoma
2007
, vol. 
48
 (pg. 
295
-
301
)
73
Savani
 
BN
Montero
 
A
Kurlander
 
R
Childs
 
R
Hensel
 
N
Barrett
 
AJ
Imatinib synergizes with donor lymphocyte infusions to achieve rapid molecular remission of CML relapsing after allogeneic stem cell transplantation.
Bone Marrow Transplant
2005
, vol. 
36
 (pg. 
1009
-
1015
)
74
Burchert
 
A
Wolfl
 
S
Schmidt
 
M
, et al. 
Interferon-alpha, but not the ABL-kinase inhibitor imatinib (STI571), induces expression of myeloblastin and a specific T-cell response in chronic myeloid leukemia.
Blood
2003
, vol. 
101
 (pg. 
259
-
264
)
75
Appel
 
S
Boehmler
 
AM
Grunebach
 
F
, et al. 
Imatinib mesylate affects the development and function of dendritic cells generated from CD34+ peripheral blood progenitor cells.
Blood
2004
, vol. 
103
 (pg. 
538
-
544
)
76
Cwynarski
 
K
Laylor
 
R
Macchiarulo
 
E
, et al. 
Imatinib inhibits the activation and proliferation of normal T lymphocytes in vitro.
Leukemia
2004
, vol. 
18
 (pg. 
1332
-
1339
)
77
Dietz
 
AB
Souan
 
L
Knutson
 
GJ
Bulur
 
PA
Litzow
 
MR
Vuk-Pavlovic
 
S
Imatinib mesylate inhibits T-cell proliferation in vitro and delayed-type hypersensitivity in vivo.
Blood
2004
, vol. 
104
 (pg. 
1094
-
1099
)
78
Wang
 
H
Cheng
 
F
Cuenca
 
A
, et al. 
Imatinib mesylate (STI-571) enhances antigen-presenting cell function and overcomes tumor-induced CD4+ T-cell tolerance.
Blood
2005
, vol. 
105
 (pg. 
1135
-
1143
)
79
Chen
 
J
Schmitt
 
A
Giannopoulos
 
K
, et al. 
Imatinib impairs the proliferation and function of CD4+CD25+ regulatory T cells in a dose-dependent manner.
Int J Oncol
2007
, vol. 
31
 (pg. 
1133
-
1139
)
80
Bhatia
 
R
Holtz
 
M
Niu
 
N
, et al. 
Persistence of malignant hematopoietic progenitors in chronic myelogenous leukemia patients in complete cytogenetic remission following imatinib mesylate treatment.
Blood
2003
, vol. 
101
 (pg. 
4701
-
4707
)
81
Copland
 
M
Hamilton
 
A
Elrick
 
LJ
, et al. 
Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction.
Blood
2006
, vol. 
107
 (pg. 
4532
-
4539
)
82
Rousselot
 
P
Huguet
 
F
Rea
 
D
, et al. 
Imatinib mesylate discontinuation in patients with chronic myelogenous leukemia in complete molecular remission for more than 2 years.
Blood
2007
, vol. 
109
 (pg. 
58
-
60
)
83
Schmid
 
C
Schleuning
 
M
Aschan
 
J
, et al. 
Low dose ara-c, donor cells and GM-CSF for treatment of recurrent acute myeloid leukemia after allogeneic stem cell transplantation: a pilot study.
Leukemia
2004
, vol. 
18
 (pg. 
1430
-
1433
)
84
Dermime
 
S
Mavroudis
 
D
Jiang
 
YZ
, et al. 
Immune escape from a graft-versus-leukemia effect may play a role in the relapse of myeloid leukemias following allogeneic bone marrow transplantation.
Bone Marrow Transplant
1997
, vol. 
19
 (pg. 
989
-
999
)
85
Brouwer
 
RE
Hoefnagel
 
J
Borger van Der
 
BB
, et al. 
Expression of co-stimulatory and adhesion molecules and chemokine or apoptosis receptors on acute myeloid leukaemia: high CD40 and CD11a expression correlates with poor prognosis.
Br J Haematol
2001
, vol. 
115
 (pg. 
298
-
308
)
86
Kufner
 
S
Zitzelsberger
 
H
Kroell
 
T
, et al. 
Leukemia-derived dendritic cells can be generated from blood or bone marrow cells from patients with acute myeloid leukaemia: a methodological approach under serum-free culture conditions.
Scand J Immunol
2005
, vol. 
62
 (pg. 
86
-
98
)
87
Schmid
 
C
Labopin
 
M
Nagler
 
A
, et al. 
Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party.
J Clin Oncol
2007
, vol. 
25
 (pg. 
4938
-
4945
)
88
Levine
 
JE
Braun
 
T
Penza
 
SL
, et al. 
Prospective trial of chemotherapy and donor leukocyte infusions for relapse of advanced myeloid malignancies after allogeneic stem-cell transplantation.
J Clin Oncol
2002
, vol. 
20
 (pg. 
405
-
412
)
89
Campregher
 
PV
Gooley
 
T
Scott
 
BL
, et al. 
Results of donor lymphocyte infusions for relapsed myelodysplastic syndrome after hematopoietic cell transplantation.
Bone Marrow Transplant
2007
, vol. 
40
 (pg. 
965
-
971
)
90
Pollyea
 
DA
Artz
 
AS
Stock
 
W
, et al. 
Outcomes of patients with AML and MDS who relapse or progress after reduced intensity allogeneic hematopoietic cell transplantation.
Bone Marrow Transplant
2007
, vol. 
40
 (pg. 
1027
-
1032
)
91
Porter
 
DL
Roth
 
MS
Lee
 
SJ
, et al. 
Adoptive immunotherapy with donor mononuclear cell infusions to treat relapse of acute leukemia or myelodysplasia after allogeneic bone marrow transplantation.
Bone Marrow Transplant
1996
, vol. 
18
 (pg. 
975
-
980
)
92
Depil
 
S
Deconinck
 
E
Milpied
 
N
, et al. 
Donor lymphocyte infusion to treat relapse after allogeneic bone marrow transplantation for myelodysplastic syndrome.
Bone Marrow Transplant
2004
, vol. 
33
 (pg. 
531
-
534
)
93
Schmid
 
C
Schleuning
 
M
Schwerdtfeger
 
R
, et al. 
Long term survival in refractory acute myeloid leukemia after sequential treatment with chemotherapy and reduced intensity conditioning for allogeneic stem cell transplantation.
Blood
2006
, vol. 
108
 (pg. 
1092
-
1099
)
94
Schmid
 
C
Schleuning
 
M
Ledderose
 
G
Tischer
 
J
Kolb
 
HJ
Sequential regimen of chemotherapy, reduced-intensity conditioning for allogeneic stem-cell transplantation, and prophylactic donor lymphocyte transfusion in high-risk acute myeloid leukemia and myelodysplastic syndrome.
J Clin Oncol
2005
, vol. 
23
 (pg. 
5675
-
5687
)
95
Shiobara
 
S
Nakao
 
S
Ueda
 
M
, et al. 
Donor leukocyte infusion for Japanese patients with relapsed leukemia after allogeneic bone marrow transplantation: lower incidence of acute graft-versus-host disease and improved outcome.
Bone Marrow Transplant
2000
, vol. 
26
 (pg. 
769
-
774
)
96
Kolb
 
HJ
Schmid
 
C
Barrett
 
AJ
Schendel
 
DJ
Graft-versus-leukemia reactions in allogeneic chimeras.
Blood
2004
, vol. 
103
 (pg. 
767
-
776
)
97
Nijmeijer
 
BA
Willemze
 
R
Falkenburg
 
JH
An animal model for human cellular immunotherapy: specific eradication of human acute lymphoblastic leukemia by cytotoxic T lymphocytes in NOD/scid mice.
Blood
2002
, vol. 
100
 (pg. 
654
-
660
)
98
Cardoso
 
AA
Schultze
 
JL
Boussiotis
 
VA
, et al. 
Pre-B acute lymphoblastic leukemia cells may induce T-cell anergy to alloantigen.
Blood
1996
, vol. 
88
 (pg. 
41
-
48
)
99
Corradini
 
P
Tarella
 
C
Olivieri
 
A
, et al. 
Reduced-intensity conditioning followed by allografting of hematopoietic cells can produce clinical and molecular remissions in patients with poor-risk hematologic malignancies.
Blood
2002
, vol. 
99
 (pg. 
75
-
82
)
100
Herbert
 
KE
Spencer
 
A
Grigg
 
A
, et al. 
Graft-versus-lymphoma effect in refractory cutaneous T-cell lymphoma after reduced-intensity HLA-matched sibling allogeneic stem cell transplantation.
Bone Marrow Transplant
2004
, vol. 
34
 (pg. 
521
-
525
)
101
Passweg
 
JR
Tiberghien
 
P
Cahn
 
JY
, et al. 
Graft-versus-leukemia effects in T lineage and B lineage acute lymphoblastic leukemia.
Bone Marrow Transplant
1998
, vol. 
21
 (pg. 
153
-
158
)
102
Gribben
 
JG
Zahrieh
 
D
Stephans
 
K
, et al. 
Autologous and allogeneic stem cell transplantations for poor-risk chronic lymphocytic leukemia.
Blood
2005
, vol. 
106
 (pg. 
4389
-
4396
)
103
Ritgen
 
M
Stilgenbauer
 
S
von Neuhoff
 
N
, et al. 
Graft-versus-leukemia activity may overcome therapeutic resistance of chronic lymphocytic leukemia with unmutated immunoglobulin variable heavy-chain gene status: implications of minimal residual disease measurement with quantitative PCR.
Blood
2004
, vol. 
104
 (pg. 
2600
-
2602
)
104
Bierman
 
PJ
Sweetenham
 
JW
Loberiza
 
FR
, et al. 
Syngeneic hematopoietic stem-cell transplantation for non-Hodgkin's lymphoma: a comparison with allogeneic and autologous transplantation. The Lymphoma Working Committee of the International Bone Marrow Transplant Registry and the European Group for Blood and Marrow Transplantation.
J Clin Oncol
2003
, vol. 
21
 (pg. 
3744
-
3753
)
105
Grigg
 
A
Ritchie
 
D
Graft-versus-lymphoma effects: clinical review, policy proposals, and immunobiology.
Biol Blood Marrow Transplant
2004
, vol. 
10
 (pg. 
579
-
590
)
106
Maris
 
MB
Sandmaier
 
BM
Storer
 
BE
, et al. 
Allogeneic hematopoietic cell transplantation after fludarabine and 2 Gy total body irradiation for relapsed and refractory mantle cell lymphoma.
Blood
2004
, vol. 
104
 (pg. 
3535
-
3542
)
107
Khouri
 
IF
Reduced-intensity regimens in allogeneic stem-cell transplantation for non-Hodgkin lymphoma and chronic lymphocytic leukemia.
Hematology Am Soc Hematol Educ Program
2006
(pg. 
390
-
397
)
108
Peggs
 
KS
Thomson
 
K
Hart
 
DP
, et al. 
Dose escalated donor lymphocyte infusions following reduced intensity transplantation: toxicity, chimerism, and disease responses.
Blood
2004
, vol. 
103
 (pg. 
1548
-
1556
)
109
Bloor
 
AJ
Thomson
 
K
Chowdhry
 
N
, et al. 
High response rate to donor lymphocyte infusion after allogeneic stem cell transplantation for indolent non-Hodgkin lymphoma.
Biol Blood Marrow Transplant
2008
, vol. 
14
 (pg. 
50
-
58
)
110
Tricot
 
G
Vesole
 
DH
Jagannath
 
S
, et al. 
Graft-versus-myeloma effect: proof of principle.
Blood
1996
, vol. 
87
 (pg. 
1196
-
1198
)
111
Verdonck
 
LF
Lokhorst
 
HM
Dekker
 
AW
Nieuwenhuis
 
HK
Petersen
 
EF
Graft-versus-myeloma effect in two cases.
Lancet
1996
, vol. 
347
 (pg. 
800
-
801
)
112
Diez-Martin
 
JL
Penalver
 
FJ
Gonzalez-Hurtado
 
JA
, et al. 
Double infusion of donor leukocytes (DLI) to treat relapsed multiple myeloma after an allogeneic bone marrow transplantation [abstract].
Bone Marrow Transplant
1997
, vol. 
19
 
suppl
pg. 
178
 
113
Salama
 
M
Nevill
 
T
Marcellus
 
D
, et al. 
Donor leukocyte infusions for multiple myeloma.
Bone Marrow Transplant
2000
, vol. 
26
 (pg. 
1179
-
1184
)
114
Alyea
 
E
Weller
 
E
Schlossman
 
R
, et al. 
T-cell-depleted allogeneic bone marrow transplantation followed by donor lymphocyte infusion in patients with multiple myeloma: induction of graft-versus-myeloma effect.
Blood
2001
, vol. 
98
 (pg. 
934
-
939
)
115
Bellucci
 
R
Alyea
 
EP
Weller
 
E
, et al. 
Immunologic effects of prophylactic donor lymphocyte infusion after allogeneic marrow transplantation for multiple myeloma.
Blood
2002
, vol. 
99
 (pg. 
4610
-
4617
)
116
Lokhorst
 
HM
Wu
 
K
Verdonck
 
LF
, et al. 
The occurrence of graft-versus-host disease is the major predictive factor for response to donor lymphocyte infusions in multiple myeloma.
Blood
2004
, vol. 
103
 (pg. 
4362
-
4364
)
117
van de Donk
 
NW
Kroger
 
N
Hegenbart
 
U
, et al. 
Prognostic factors for donor lymphocyte infusions following non-myeloablative allogeneic stem cell transplantation in multiple myeloma.
Bone Marrow Transplant
2006
, vol. 
37
 (pg. 
1135
-
1141
)
118
Levenga
 
H
Levison-Keating
 
S
Schattenberg
 
AV
, et al. 
Multiple myeloma patients receiving pre-emptive donor lymphocyte infusion after partial T-cell-depleted allogeneic stem cell transplantation show a long progression-free survival.
Bone Marrow Transplant
2007
, vol. 
40
 (pg. 
355
-
359
)
119
Mitsiades
 
CS
Mitsiades
 
NS
Richardson
 
PG
Munshi
 
NC
Anderson
 
KC
Multiple myeloma: a prototypic disease model for the characterization and therapeutic targeting of interactions between tumor cells and their local microenvironment.
J Cell Biochem
2007
, vol. 
101
 (pg. 
950
-
968
)
120
Matsui
 
W
Wang
 
Q
Barber
 
JP
, et al. 
Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance.
Cancer Res
2008
, vol. 
68
 (pg. 
190
-
197
)
121
Childs
 
R
Chernoff
 
A
Contentin
 
N
, et al. 
Regression of metastatic renal-cell carcinoma after nonmyeloablative allogeneic peripheral-blood stem-cell transplantation.
N Engl J Med
2000
, vol. 
343
 (pg. 
750
-
758
)
122
Demirer
 
T
Barkholt
 
L
Blaise
 
D
, et al. 
Transplantation of allogeneic hematopoietic stem cells: an emerging treatment modality for solid tumors.
Nat Clin Pract Oncol
2008
, vol. 
5
 (pg. 
256
-
267
)
123
Takahashi
 
Y
Harashima
 
N
Kajigaya
 
S
, et al. 
Regression of human kidney cancer following allogeneic stem cell transplantation is associated with recognition of an HERV-E antigen by T cells.
J Clin Invest
2008
, vol. 
118
 (pg. 
1099
-
1109
)
124
Gratwohl
 
A
Baldomero
 
H
Demirer
 
T
, et al. 
Hematopoetic stem cell transplantation for solid tumors in Europe.
Ann Oncol
2004
, vol. 
15
 (pg. 
653
-
660
)
125
Barkholt
 
L
Bregni
 
M
Remberger
 
M
, et al. 
Allogeneic haematopoietic stem cell transplantation for metastatic renal carcinoma in Europe.
Ann Oncol
2006
, vol. 
17
 (pg. 
1134
-
1140
)
126
Suzuki
 
Y
Adachi
 
Y
Minamino
 
K
, et al. 
A new strategy for treatment of malignant tumor: intra-bone marrow-bone marrow transplantation plus CD4- donor lymphocyte infusion.
Stem Cells
2005
, vol. 
23
 (pg. 
365
-
370
)
127
Koike
 
Y
Adachi
 
Y
Suzuki
 
Y
, et al. 
Allogeneic intrabone marrow-bone marrow transplantation plus donor lymphocyte infusion suppresses growth of colon cancer cells implanted in skin and liver of rats.
Stem Cells
2007
, vol. 
25
 (pg. 
385
-
391
)
128
Bishop
 
MR
Fowler
 
DH
Marchigiani
 
D
, et al. 
Allogeneic lymphocytes induce tumor regression of advanced metastatic breast cancer.
J Clin Oncol
2004
, vol. 
22
 (pg. 
3886
-
3892
)
129
Klein
 
CA
Wilke
 
M
Pool
 
J
, et al. 
The hematopoietic system-specific minor histocompatibility antigen HA-1 shows aberrant expression in epithelial cancer cells.
J Exp Med
2002
, vol. 
196
 (pg. 
359
-
368
)
130
Spiotto
 
MT
Yu
 
P
Rowley
 
DA
, et al. 
Increasing tumor antigen expression overcomes “ignorance” to solid tumors via crosspresentation by bone marrow-derived stromal cells.
Immunity
2002
, vol. 
17
 (pg. 
737
-
747
)
131
Quin
 
Z
Blankenstein
 
T
CD4 T cell-mediated tumor rejection involves inhibition of angiogenesis that is dependent on IFN-g receptor expression by nonhematopoietic cells.
Immunity
2000
, vol. 
12
 (pg. 
677
-
686
)
132
Ruggeri
 
L
Capanni
 
M
Urbani
 
E
, et al. 
Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants.
Science
2002
, vol. 
295
 (pg. 
2097
-
2100
)
133
Davies
 
SM
Ruggieri
 
L
DeFor
 
T
, et al. 
Evaluation of KIR ligand incompatibility in mismatched unrelated donor hematopoietic transplants: killer immunoglobulin-like receptor.
Blood
2002
, vol. 
100
 (pg. 
3825
-
3827
)
134
Riddell
 
SR
Watanabe
 
KS
Goodrich
 
JM
, et al. 
Restoration of viral immunity in immunodeficient humans by adoptive transfer of T-cell clones.
Science
1992
, vol. 
257
 (pg. 
238
-
241
)
135
Rooney
 
CM
Smith
 
CA
Ng
 
CYC
, et al. 
Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr-virus-related lymphoproliferation.
Lancet
1995
, vol. 
345
 (pg. 
9
-
13
)
136
Cobbold
 
M
Khan
 
N
Pourgheysari
 
B
, et al. 
Adoptive transfer of cytomegalovirus-specific CTL to stem cell transplant patients after selection by HLA-peptide tetramers.
J Exp Med
2005
, vol. 
202
 (pg. 
379
-
386
)
137
Becker
 
C
, et al. 
Adoptive tumor therapy with T lymphocytes enriched through an IFN-α capture assay.
Nat Med
2001
, vol. 
7
 (pg. 
1159
-
1162
)
138
Randolph
 
SS
Gooley
 
TA
Warren
 
EH
Appelbaum
 
FR
Riddell
 
SR
Female donors contribute to a selective graft-versus-leukemia effect in male recipients of HLA-matched, related hematopoietic stem cell transplants.
Blood
2004
, vol. 
103
 (pg. 
347
-
352
)
139
Berger
 
C
Jensen
 
MC
Lansdorp
 
PM
, et al. 
Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates.
J Clin Invest
2008
, vol. 
118
 (pg. 
294
-
305
)
140
Duell
 
T
van Lint
 
MT
Ljungman
 
P
, et al. 
Health and functional status of long-term survivors of bone marrow transplantation: EBMT Working Party on Late Effects and EULEP Study Group on Late Effects. European Group for Blood and Marrow Transplantation.
Ann Intern Med
1997
, vol. 
126
 (pg. 
184
-
192
)
141
Kolb
 
HJ
Mittermüller
 
J
Holler
 
E
Thalmeier
 
K
Bartram
 
CR
Graft-versus-host reaction spares normal stem cells in chronic myelogenous leukemia.
Bone Marrow Transplant
1996
, vol. 
17
 (pg. 
449
-
452
)
142
Falkenburg
 
JH
Goselink
 
HM
van der Harst
 
D
, et al. 
Growth inhibition of clonogenic leukemic precursor cells by minor histocompatibility antigen-specific cytotoxic T lymphocytes.
J Exp Med
1991
, vol. 
174
 (pg. 
27
-
33
)
143
Bonnet
 
D
Warren
 
EH
Greenberg
 
PD
Dick
 
JE
Riddell
 
SR
CD8+ minor histocompatibility antigen-specific cytotoxic T lymphocyte clones eliminate human acute myeloid leukemia stem cells.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 (pg. 
8639
-
8644
)
144
Gao
 
L
Bellantuono
 
I
Elsasser
 
A
, et al. 
Selective elimination of leukemic CD34(+) progenitor cells by cytotoxic T lymphocytes specific for WT1.
Blood
2000
, vol. 
95
 (pg. 
2198
-
2203
)
145
Wu
 
CJ
Biernacki
 
M
Kutok
 
JL
, et al. 
Graft-versus-leukemia target antigens in chronic myelogenous leukemia are expressed on myeloid progenitor cells.
Clin Cancer Res
2005
, vol. 
11
 (pg. 
4504
-
4511
)
146
dem Borne
 
PA
Luxemburg-Heijs
 
SA
Heemskerk
 
MH
, et al. 
Molecular persistence of chronic myeloid leukemia caused by donor T cells specific for lineage-restricted maturation antigens not recognizing immature progenitor-cells.
Leukemia
2006
, vol. 
20
 (pg. 
1040
-
1046
)
147
Brouwer
 
RE
van der
 
HP
Schreuder
 
GM
, et al. 
Loss or downregulation of HLA class I expression at the allelic level in acute leukemia is infrequent but functionally relevant, and can be restored by interferon.
Hum Immunol
2002
, vol. 
63
 (pg. 
200
-
210
)
148
Hirano
 
N
Takahashi
 
T
Ohtake
 
S
, et al. 
Expression of costimulatory molecules in human leukemias.
Leukemia
1996
, vol. 
10
 (pg. 
1168
-
1176
)
149
Schneider
 
EM
Chen
 
ZZ
Ellwart
 
J
Wilmanns
 
W
Kolb
 
HJ
Immune phenotype of chronic myelogenous leukemia progenitor cells [abstract].
Bone Marrow Transplant
1996
, vol. 
17
 
Suppl
pg. 
69
 
150
Schulz
 
U
Munker
 
R
Ertl
 
B
Holler
 
E
Kolb
 
HJ
Different types of human leukemias express the message for TNF-alpha and interleukin-10.
Eur J Med Res
2001
, vol. 
6
 (pg. 
359
-
363
)
151
Pawelec
 
G
Rehbein
 
A
Schlotz
 
E
da Silva
 
P
Cellular immune responses to autologous chronic myelogenous leukaemia cells in vitro.
Cancer Immunol Immunother
1996
, vol. 
42
 (pg. 
193
-
199
)
152
Bergmann
 
L
Schui
 
DK
Brieger
 
J
, et al. 
The inhibition of lymphokine-activated killer cells in acute myeloblastic leukemia is mediated by transforming growth factor-beta 1.
Exp Hematol
1995
, vol. 
23
 (pg. 
1574
-
1580
)
153
Munn
 
DH
Mellor
 
AL
Indoleamine 2,3-dioxygenase and tumor-induced tolerance.
J Clin Invest
2007
, vol. 
117
 (pg. 
1147
-
1154
)
154
Molldrem
 
JJ
Lee
 
PP
Kant
 
S
, et al. 
Chronic myelogenous leukemia shapes host immunity by selective deletion of high-avidity leukemia-specific T cells.
J Clin Invest
2003
, vol. 
111
 (pg. 
639
-
647
)
155
Buggins
 
AG
Hirst
 
WJ
Pagliuca
 
A
Mufti
 
GJ
Variable expression of CD3-zeta and associated protein tyrosine kinases in lymphocytes from patients with myeloid malignancies.
Br J Haematol
1998
, vol. 
100
 (pg. 
784
-
792
)
156
Chen
 
X
Woiciechowsky
 
A
Raffegerst
 
S
, et al. 
Impaired expression of the CD3-zeta chain in peripheral blood T cells of patients with chronic myeloid leukaemia results in an increased susceptibility to apoptosis.
Br J Haematol
2000
, vol. 
111
 (pg. 
817
-
825
)
157
Trimble
 
LA
Kam
 
LW
Friedman
 
RS
Xu
 
Z
Lieberman
 
J
CD3zeta and CD28 down-modulation on CD8 T cells during viral infection.
Blood
2000
, vol. 
96
 (pg. 
1021
-
1029
)
158
Shiobara
 
S
Nakao
 
S
Ueda
 
M
, et al. 
Donor leukocyte infusion for Japanese patients with relapsed leukemia after allogeneic bone marrow transplantation: indications and dose escalation.
Ther Apher
2001
, vol. 
5
 (pg. 
40
-
45
)
159
Choi
 
SJ
Lee
 
JH
Lee
 
JH
, et al. 
Treatment of relapsed acute myeloid leukemia after allogeneic bone marrow transplantation with chemotherapy followed by G-CSF-primed donor leukocyte infusion: a high incidence of isolated extramedullary relapse.
Leukemia
2004
, vol. 
18
 (pg. 
1789
-
1797
)
160
Collins
 
RH
Goldstein
 
S
Giralt
 
S
, et al. 
Donor leukocyte infusions in acute lymphocytic leukemia.
Bone Marrow Transplant
2000
, vol. 
26
 (pg. 
511
-
516
)
161
Choi
 
SJ
Lee
 
JH
Lee
 
JH
, et al. 
Treatment of relapsed acute lymphoblastic leukemia after allogeneic bone marrow transplantation with chemotherapy followed by G-CSF-primed donor leukocyte infusion: a prospective study.
Bone Marrow Transplant
2005
, vol. 
36
 (pg. 
163
-
169
)
162
Robinson
 
SP
Goldstone
 
AH
Mackinnon
 
S
, et al. 
Chemoresistant or aggressive lymphoma predicts for a poor outcome following reduced-intensity allogeneic progenitor cell transplantation: an analysis from the Lymphoma Working Party of the European Group for Blood and Bone Marrow Transplantation.
Blood
2002
, vol. 
100
 (pg. 
4310
-
4316
)
163
Morris
 
E
Thomson
 
K
Craddock
 
C
, et al. 
Outcomes after alemtuzumab-containing reduced-intensity allogeneic transplantation regimen for relapsed and refractory non-Hodgkin lymphoma.
Blood
2004
, vol. 
104
 (pg. 
3865
-
3871
)
164
Lokhorst
 
HM
Schattenberg
 
A
Cornelissen
 
JJ
, et al. 
Donor lymphocyte infusions for relapsed multiple myeloma after allogeneic stem-cell transplantation: predictive factors for response and long-term outcome.
J Clin Oncol
2000
, vol. 
18
 (pg. 
3031
-
3037
)
165
Peggs
 
KS
Mackinnon
 
S
Williams
 
CD
, et al. 
Reduced-intensity transplantation with in vivo T-cell depletion and adjuvant dose-escalating donor lymphocyte infusions for chemotherapy-sensitive myeloma: limited efficacy of graft-versus-tumor activity.
Biol Blood Marrow Transplant
2003
, vol. 
9
 (pg. 
257
-
265
)
166
Huff
 
CA
Fuchs
 
EJ
Noga
 
SJ
, et al. 
Long-term follow-up of T cell-depleted allogeneic bone marrow transplantation in refractory multiple myeloma: importance of allogeneic T cells.
Biol Blood Marrow Transplant
2003
, vol. 
9
 (pg. 
312
-
319
)
167
Khouri
 
IF
Lee
 
MS
Saliba
 
RM
, et al. 
Nonablative allogeneic stem-cell transplantation for advanced/recurrent mantle-cell lymphoma.
J Clin Oncol
2003
, vol. 
21
 (pg. 
4407
-
4412
)
168
Glass
 
B
Nickelsen
 
M
Dreger
 
P
, et al. 
Reduced-intensity conditioning before allogeneic transplantation of hematopoietic stem cells: the need for T cells early after transplantation to induce a graft-versus-lymphoma effect.
Bone Marrow Transplant
2004
, vol. 
34
 (pg. 
391
-
397
)
169
Peggs
 
KS
Sureda
 
A
Qian
 
W
, et al. 
Reduced-intensity conditioning for allogeneic haematopoietic stem cell transplantation in relapsed and refractory Hodgkin lymphoma: impact of alemtuzumab and donor lymphocyte infusions on long-term outcomes.
Br J Haematol
2007
, vol. 
139
 (pg. 
70
-
80
)
Sign in via your Institution