Despite significant progress in the past 20 years, graft-versus-host disease (GVHD) remains a significant cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). T-cell depletion (TCD) of the donor graft offers the potential for prevention of GVHD without the morbidity associated with immunosuppressive drugs such as methotrexate and cyclosporine. Most early trials documented that TCD could substantially limit acute and chronic GVHD. However, this reduction in GVHD did not translate into improved overall survival because of unexpected high rates of graft failure, Epstein-Barr virus–associated lymphoproliferative disorders, and disease recurrence after TCD bone marrow transplantation. Despite the problems associated with TCD, great interest remains in developing and improving this technology, particularly for recipients of HLA-mismatched grafts. If advances in graft engineering can accomplish the goal of GVHD prevention without adversely affecting engraftment, immunocompetence, and antileukemic activity, then substantial improvements in overall transplant outcome can be realized.

GVHD can be classified as acute or chronic based on timing of onset and clinical features. Acute GVHD usually develops within the first 2 months of bone marrow transplantation (BMT) and affects mainly the skin, gastrointestinal tract, and liver. When pharmacologic immunosuppression is used as GVHD prophylaxis after myeloablative transplantation, moderate to severe acute GVHD (grades II-IV) occurs in 25% to 60% of matched related donor transplant recipients, and up to 45% to 70% in unrelated donor recipients.1-5Acute GVHD is also emerging as a major complication after allogeneic nonmyeloablative stem cell transplantation.6 Development of grade II to IV acute GVHD is associated with decreased survival in patients after allogeneic BMT.7 8 

Chronic GVHD has a later onset than acute GVHD and is often clinically distinct. Patients can manifest sclerodermatous skin changes, keratoconjunctivitis, sicca syndrome, lichenoid oral mucosal lesions, esophageal and vaginal strictures, liver disease, and pulmonary insufficiency.9 Despite immunosuppressive agents, approximately 30% to 50% of patients will develop chronic GVHD after myeloablative HLA-identical sibling BMT. The incidence of chronic GVHD may be even higher after allogeneic transplantation using unmanipulated peripheral blood stem cells (PBSCs).10,11 Although limited chronic GVHD often resolves spontaneously with minimal intervention, extensive chronic GVHD requires prolonged immunosuppressive treatment and is associated with significant morbidity and mortality. More than 50% of patients with extensive chronic GVHD will die, mostly secondary to infections resulting from severe immune dysfunction.12 

Acute GVHD is believed to occur in 3 phases: (1) tissue damage from conditioning, (2) donor T-cell activation, and (3) inflammatory effectors. In the earliest phase, inflammatory cytokines are released from host tissue in response to damage by the pretransplantation conditioning regimen.13 These cytokines, including interleukin-1 (IL-1) and tumor necrosis factor-α (TNF-α), up-regulate the expression of adhesion molecules and host major histocompatibility complex (MHC) antigens, and enhance recognition of the host tissue by mature donor T lymphocytes. During the second phase, donor T-helper 1 (Th1) cells are activated on recognition of alloantigens and secrete IL-2 and interferon-γ (IFN-γ), which recruit other T cells, cytotoxic T lymphocytes (CTLs), natural killer (NK) cells, monocytes, and macrophages14 In the last phase, mononuclear cells primed by Th1 cytokines secrete more TNF-α and IL-1, which induce cellular damage or apoptosis, and restart the cycle of inflammation.

Several clinical studies have attempted to interrupt the cytokine cascade as a strategy to prevent GVHD. Unfortunately, a blinded randomized trial of IL-1 receptor antagonist versus placebo did not demonstrate any benefit for IL-1 blockade in acute GVHD prevention.15 Another trial of recombinant human IL-11 to reduce Th1 cytokine production was halted due to unacceptable toxicity.16 Thus, although interruption of the “cytokine storm” may prevent GVHD in murine models, it appears to be insufficient in humans, and T cells remain the prime target for current therapeutic strategies in GVHD prophylaxis.

Current approaches for the prevention and treatment of GVHD involve direct blockade of T-cell function. These include the down-regulation of T lymphocytes by inhibiting cellular proliferation (methotrexate), inhibition of de novo purine synthesis (mycophenolate mofetil), suppression of IL-2 secretion by blocking calcineurin activity (cyclosporine, FK-506), interfering with downstream growth signaling pathways (sirolimus), and reduction of T-cell responsiveness by blocking the IL-2 receptor (daclizumab). However, the most effective means of GVHD prophylaxis is depletion of T cells from the donor inoculum. In many instances, TCD can dramatically reduce the incidence of GVHD even in the absence of posttransplantation immunosuppressive therapy.

In 1968, Dicke and colleagues reported that irradiated mice receiving transplants with spleen cell fractions depleted of small lymphocytes enjoyed 80% to 100% survival without evidence of GVHD, whereas mice given fractions containing increasing numbers of lymphocytes all died from severe GVHD.17 Subsequently, Reisner and coworkers demonstrated similar results with mouse bone marrow and spleen cell suspensions depleted of T cells by soybean or peanut agglutinination.18 In the 1970s, antithymocyte globulin (ATG) was tried as a method of TCD, but with limited success because early ATG preparations were also toxic to hematopoietic cells. This problem was later circumvented through the absorption of ATG with spleen or liver suspensions to remove antibodies against stem cells.19 Using absorbed ATG, animals successfully received transplantations with minimal to no GVHD, across both major and minor histocompatibility barriers.20-25 In the late 1970s, it became clear from murine models that even minor alloantigen differences could stimulate GVHD, and that contamination of the marrow with as few as 0.3% T lymphocytes (3 × 104 cells) could be sufficient to cause lethal GVHD.23 In 1981, Vallera and colleagues reported on the first use of a monoclonal antibody, anti–Thy-1.2, for ex vivo TCD in a murine transplant model.26 In that study, however, many animals still died from complications of GVHD after a few months. This led to the eventual realization that antibody alone is insufficient to abrogate GVHD and that addition of rabbit complement would confer superior protection. The importance of complement would be confirmed in subsequent human studies using monoclonal antibodies for TCD.

Because animal studies had shown that pharmacologic immunosuppression was not necessary after TCD, it was anticipated that TCD transplants in humans without cyclosporine and methotrexate would be associated with less mucositis, renal failure, infections, and other complications of conventional BMT. Furthermore, it was hoped that the lower incidence of GVHD would translate into improved long-term survival. From 1981 to 1986, hundreds of TCD transplantations were performed worldwide using various methods of TCD. Although these trials confirmed a low incidence of acute and chronic GVHD, they also revealed new and important limitations associated with TCD BMT. These included an increased incidence of graft rejection, delayed immune reconstitution, increased posttransplantation lymphoproliferative disorders (LPDs), and increased rate of disease relapse.

Methods of TCD

Methods of TCD in human transplantation have primarily depended on negative selection techniques by physical separation or antibody-based purging (Figure 1). Examples of physical separation techniques include differential agglutination with lectins followed by rosetting with sheep red blood cells,27,28 counterflow centrifugal elutriation,29-31 and fractionation on density gradients.32 The earliest human trials using antibody as T depletion relied on absorbed ATG.33 However, this was soon replaced by monoclonal antibodies directed against specific targets on T cells. Monoclonal antibodies have been used alone,34-36and in conjunction with homologous, heterologous, or rabbit complements,37-49 Many are directed against T-cell antigens with restricted expression, whereas others have broader targets. Some of the most widely used monoclonals are a set of antibodies called CAMPATH-1,40 which is directed against a heterogeneous 23- to 30-kd glycoprotein (CDw52) expressed on lymphocytes and monocytes. CAMPATH-1 antibodies are classified by their immunoglobulin subtype and origin. CAMPATH-1G and CAMPATH-1M are derived from the rat and are IgG2b and IgM, respectively. CAMPATH-1H, in contrast, is a recombinant DNA–derived human IgG1. CAMPATH-1 antibodies have an advantage over other anti–T-cell monoclonal antibodies in that they can fix human as well as rabbit complements. As a result, they can lyse lymphocytes in human donor serum and do not depend on rabbit complement lots that are variable in efficacy and toxicity. This property renders CAMPATH-1 antibodies useful for both in vitro or in vivo TCD. Other antibody-based methods for TCD that do not depend on complement include immunotoxins50-54 and immunomagnetic beads.55 In the 1990s, with the advent of allogeneic PBSC transplantation, CD34+ immunoadsorption columns have been developed that positively select hematopoietic progenitors and permit nonadherent cells to be washed out, effectively reducing the lymphocyte content in the infused PBSC graft.56 

Fig. 1.

Methods of TCD.

T-cell dose in TCD BMT

An average patient receiving a transplant with unmodified marrow receives a T-cell dose on the order of 1 to 5 × 107cells/kg recipient body weight. Initially, it was not known what degree of depletion would be necessary to prevent GVHD, because sufficiently sensitive assays for quantifying T cells were not available. Early efforts at TCD were thus intended to maximally purge lymphocytes without compromising hematopoietic precursors. It was not until the mid-1980s that limiting dilution analysis (LDA) was developed to measure functional T lymphocytes and assess cell dose.57,58 Using counterflow centrifugation elutriation (CCE), Wagner and coworkers demonstrated a 45% incidence of acute GVHD in matched related transplant recipients when the T-cell dose was 1 × 106 cells/kg, but only 22% when the dose was reduced to 0.5 × 106 cells/kg.31,59Recently, Aversa and colleagues reported that reliable engraftment without GVHD could be achieved in full haplotype mismatched transplants using high stem cell numbers plus extensive TCD to a CD3+cell dose of 3 × 104 cells/kg.60 Although these data give a clue as to the relationship between T-cell dose and GVHD, there is likely to be significant variability between donor-recipient pairs depending on minor antigen differences.

The advantages and disadvantages of TCD are listed in Table1 and discussed in more detail below.

Table 1.

Advantages and disadvantages of TCD

AdvantagesDisadvantages
Low incidence of acute and chronic GVHD Higher incidence of graft failure 
Reduced or no requirement for posttransplantation immune suppression
as GVHD prophylaxis 
Loss of GVL activity (higher incidence of disease relapse, especially with CML)
Delayed immune reconstitution  
Decreased pulmonary and hepatic toxicity early after BMT Increased risk for posttransplantation EBV-LPDs  
Decreased early transplant-related mortality  Higher incidence of CMV reactivation  
? Shorter time to engraftment Overall survival not improved compared to non-TCD BMT  
? Decreased cost  
AdvantagesDisadvantages
Low incidence of acute and chronic GVHD Higher incidence of graft failure 
Reduced or no requirement for posttransplantation immune suppression
as GVHD prophylaxis 
Loss of GVL activity (higher incidence of disease relapse, especially with CML)
Delayed immune reconstitution  
Decreased pulmonary and hepatic toxicity early after BMT Increased risk for posttransplantation EBV-LPDs  
Decreased early transplant-related mortality  Higher incidence of CMV reactivation  
? Shorter time to engraftment Overall survival not improved compared to non-TCD BMT  
? Decreased cost  

GVHD prevention

HLA-identical sibling transplantation.

In 1981, Rodt and coworkers reported on the use of absorbed human ATG as TCD in HLA-compatible sibling transplants and demonstrated that only 2 of 12 patients developed mild skin GVHD.33 In contrast, concurrent TCD studies using murine monoclonal antibodies alone reported GVHD rates of approximately 50%.35,36 These results prompted investigators to add complement in hopes of enhancing lymphocytolysis and improving GVHD outcome. Studies in the mid-1980s confirmed that TCD using monoclonal antibodies plus rabbit complement, which removed 2 to 3 logs of T lymphocytes, uniformly decreased the incidence of clinically significant GVHD to 10% to 20% after matched sibling transplantation.37,39,41-43 In the late 1980s and early 1990s, similar results were reported in larger phase II trials of TCD using anti-CD5, -CD6, and -CD8 antibodies plus complement, CAMPATH-1, or immunotoxins.49,54,61-64 In some of these studies, the low incidence of GVHD was achieved even in the absence of immunosuppression after transplantation.49 54 

Studies of HLA-identical sibling PBSC transplantation using CD34+ selection have reported great variation in acute GVHD incidence from 8% to 100%.65-69 This variability was likely influenced by factors such as the number of CD3+cells infused, immunosuppressive agents given after transplantation, and type of CD34+ selection device used. Other TCD methods are also being investigated with PBSC transplantation. Hale and associates have recently reported that HLA-matched sibling PBSC transplantation purged with CAMPATH-1H or CAMPATH-1G is associated with only 4% and 11% incidence of grade II to IV acute GVHD, respectively.70 

Alternative donor transplantation.

Retrospective data from the National Marrow Donor Registry (NMDR) have demonstrated that TCD is a significant negative predictor for the development of grade III to IV acute GVHD after unrelated BMT.71 The International Bone Marrow Transplant Registry (IBMTR) recently reported on 1868 leukemia patients who received allogeneic marrow transplants from donors other than HLA-identical siblings. The incidence of grade II to IV GVHD was between 34% and 38% in the TCD group, as compared to 57% in the non-TCD group (P < .0001).72 HLA-matched unrelated marrow transplants depleted of T cells with anti-CD6 monoclonal antibody plus complement as the sole form of GVHD prophylaxis is associated with an approximately 40% incidence of grade II to IV GVHD.73 A similar result has been reported for TCD using the αβ T-cell receptor antibody T10B9 in unrelated marrow transplants for patients with chronic myelogenous leukemia (CML).74 In another study, the incidence of grade II to IV acute GVHD was only 24% among patients who received unrelated marrow grafts T-cell depleted with CAMPATH-1.75 

T-cell depletion has also been used in HLA-mismatched transplant settings. In single institution studies, grade II to IV GVHD incidence has ranged from 18% to 40% in recipients of HLA-mismatched BMT after TCD using anti-CD6 or T10B9 monoclonal antibody.76-78 In leukemia patients receiving HLA haploidentical marrow from related donors, a low occurrence of GVHD has been observed when the graft was augmented with PBSCs and T-cell depleted using lectin agglutination and E-rosetting.60 79 

Organ dysfunction after TCD BMT

T-cell depletion appears to be associated with less organ toxicity early after transplantation compared to conventional BMT. The incidence of hepatic veno-occlusive disease has been reported to be only 3.1% after TCD BMT, and only 1.2% among patients receiving total body irradiation (TBI) as part of their conditioning.80 This reduction in hepatotoxicity may be due to the fact that the TCD patients are spared from injurious effects of methotrexate or cyclosporine/tacrolimus. Alternatively, decreases in alloreactivity may lead to lower levels of circulating cytokines, which could damage hepatic endothelium. Finally, because TCD patients may have earlier neutrophil recovery in the absence of methotrexate, they are also less likely to require antibiotics such as amphotericin that could have deleterious effects on hepatorenal function.

T-cell depletion has also been associated with decreased pulmonary complications after BMT. Breuer and coworkers have reported that fatal interstitial pneumonitis occurred in only 6 of 80 (7.5%) consecutive allogeneic BMT where TCD was used as GVHD prophylaxis.81In another series, the incidence of life-threatening pulmonary complications within the first 60 days of BMT was 8% among those who received TCD as the sole form of GVHD prophylaxis, but 33% among those who received cyclosporine and methotrexate (P < .0001).82 In this study, the protective effect of TCD against pulmonary complications was independent of acute GVHD in multivariate analysis, perhaps implicating deleterious effects of GVHD prophylactic medications.

Transplant-related mortality

Because TCD protects against GVHD and reduces organ dysfunction after BMT, one would expect these benefits to translate into lower transplant-related mortality (TRM). In a number of series, TRM after TCD BMT has indeed been quite low, ranging from 2% to 15%.49,83-86 However, other TCD studies have reported TRM rates from 20% to 32% even after matched sibling transplantations, with many deaths being secondary to graft failure, infections, and Epstein-Barr virus (EBV)–associated LPDs.54 87-89 This variability highlights the fact that elements other than GVHD, such as the intensity of conditioning, posttransplantation immunosuppression, graft failure, and immune reconstitution, also contribute significantly to TRM. It further reinforces the point that for a TCD regimen to be successful, it must do more than just protect against GVHD. It must also preserve graft-versus-leukemia (GVL) activity and engraftment potential and spare patients from excessive transplant-related toxicity.

Graft failure

Prior to the advent of TCD, graft failure after allogeneic BMT was uncommon, occurring in 1% to 5% of patients having transplantations for leukemia.90,91 In contrast, most TCD BMT series in the 1980s and early 1990s reported higher rates of graft failure.31,32,40,41,43-46,51,52,61,87,92-99 In some of these studies, the incidence of graft failure was as high as 50% to 70%.92,99 In an analysis from the IBMTR of more than 3000 patients who received TCD or non-TCD BMT for leukemia, TCD was associated with a 9-fold increased risk for graft failure compared to unmanipulated marrow transplantation (P < .0001).100 

A number of studies have retrospectively analyzed risk factors associated with graft failure after TCD. Aside from HLA disparity, one important factor appears to be the dose of TBI administered before transplantation. Patients who are conditioned with higher doses of TBI consistently have lower incidence of graft failure compared to those who receive lower TBI doses.92,99,101,102 In addition, donor-recipient sex disparity, male donor gender, donor age over 25 years, and absence of posttransplantation immunosuppression, have also been reported to be significant risk factors for graft failure.95,99 100 

Graft failure after TCD BMT can occur as 3 distinct patterns.92 These include (1) failure of initial engraftment, (2) partial or full initial engraftment followed by graft rejection within 2 weeks of BMT, and (3) delayed graft failure that can occur months after BMT. The pathophysiologic mechanisms behind these different patterns are not well understood, although it is widely accepted that early graft failure after TCD transplantation results primarily from immunologic rejection of donor hematopoietic elements by host lymphoid elements that have survived the conditioning process. Direct evidence for the role of the host immune system have come from the identification of host T lymphocytes from patients at the time of graft rejection, which exhibit donor-specific cytotoxic activity,103-112 and which suppress donor proliferation of granulocyte-macrophage colony-forming units and erythroid colony forming units in vitro.93,104 Clinical evidence further supporting this model includes observations that genetic disparity between donor and recipient correlates with graft rejection and that increased immunosuppression, either through the conditioning regimen or posttransplantation immunosuppressive agents, appears to protect against graft failure.92,95,99-102 113 

It does not appear that failure of initial engraftment is caused by injury to hematopoietic progenitors or auxiliary cells during marrow manipulation, because autologous marrows purged with monoclonal antibodies and complement engraft without difficulty.61,99,114-116 Viral infections, such as cytomegalovirus (CMV) or human herpes virus 6, may contribute to graft failure after BMT.117-121 Although TCD BMT patients may have a higher risk of CMV reactivation after transplantation,122-125 direct clinical evidence implicating this and other viruses to graft failure after TCD transplantation is limited.

Other clinical observations also offer insight into the pathophysiology of graft failure. Many studies have reported that mixed lymphoid and myeloid chimerism is more common after TCD BMT and that such chimerism may be associated with graft failure.99,102,126-130 Viable host-derived hematopoietic cells can often be recovered from patients after TCD marrow transplantation,131 132 and their coexistence with the donor graft implies a state of immune tolerance between the graft and host. This may be especially true for patients who have persistent stable mixed chimerism with fully functional hematopoietic grafts. For patients whose mixed chimerism is transient, however, it is possible that graft failure could result when host lymphoid tolerance of the graft is broken. At the current time, it is unknown whether the mixed chimeric state is a cause of graft rejection or an effect of the graft rejection process itself.

The higher incidence of graft failure associated with donor-patient gender disparity has led researchers to investigate gender-specific antigens as potential targets for graft rejection. Studies suggest that female patients receiving grafts from male donors may be at higher risk for graft rejection because female recipient T lymphocytes can recognize male-specific minor antigens in the donor graft. The H-Y antigens encoded on the Y chromosome have been implicated as targets for this process. During rejection of male stem cell grafts, H-Y–specific CTL clones have been isolated from the peripheral blood of female recipients.110 Multiple H-Y antigens are believed to be involved in graft rejection. Known H-Y epitopes recognized by HLA-A2–restricted CTL clones include peptides derived from the SMCY and DFFRYgenes.133-135 More recently, a third H-Y epitope that is recognized by HLA-B60–restricted CTL clones has been localized to theUTY gene on the Y chromosome.136 The identification of these H-Y antigens could prove useful in the selection of donor recipient pairs for stem cell transplantation and could be especially relevant in female recipients who may have been sensitized to male antigens through prior blood transfusions.

Because graft rejection is felt to be mediated by residual host T cells that have escaped myeloablation, early preventive strategies were focused on intensifying the myeloablative regimen. Some investigators also believed that intensive conditioning regimens would empty out the host marrow and increase “hematopoietic space” for the incoming donor graft. High doses of cytarabine, thiotepa, and anthracyclines have been incorporated into standard ablative regimens and have reduced the rate of graft failure after TCD BMT.89,137-141However, these were also more likely to be associated with increased regimen-related toxicity. Other researchers have investigated the addition of total lymphoid irradiation (TLI) as a strategy against graft failure. Although TLI has been shown to reduce graft failure after conventional BMT for aplastic anemia,142-144 its benefits are less clear after TCD BMT. Although single-arm studies of TCD BMT have reported a low incidence of graft failure when TLI was included in the conditioning regimen,87,145,146 an older prospective randomized trial of TCD transplantation failed to show any benefit.147 

An alternative method of immunosuppression commonly used to reduce graft failure after TCD BMT is the simultaneous depletion of host immune cells using ATG or CAMPATH-1 at the time of transplantation. Studies in animals from the late 1980s showed that in vivo depletion of recipient lymphocytes using anti–T-cell monoclonal antibodies could prevent rejection of the TCD marrow even across major histocompatibility barriers.148,149 The presumed mechanism behind this observation was that the treatment eliminated host lymphocytes that were active in the graft rejection process. In 1994, Hale and his associates adapted this approach to humans and reported that simultaneous donor and host TCD with CAMPATH-1 reduced the incidence of graft failure to less than 10%, without compromising GVHD prophylaxis.87 In their subsequent series of 70 AML patients receiving transplants from HLA-identical sibling donors using this combined TCD approach, the incidence of graft failure was only 6%, significantly lower than that with donor marrow TCD alone.86 

Another widely applied strategy for reducing graft failure involves limited or selective TCD. The premise of this approach arose from the belief that most early methods of TCD had attempted to deplete as many T lymphocytes from the marrow as possible and that the exhaustive depletion nonspecifically removed ancillary marrow elements that were necessary to sustain engraftment. It was thus postulated that by selectively purging only certain subsets of T cells, such as mature T lymphocytes, but sparing NK cells, immature thymocytes, monocytes, and other hematopoietic elements, one could maintain the protective effects against GVHD without increasing the risk for graft failure. Several selective antibody-purging methods have been successfully used. Studies using anti-CD5 immunotoxins, anti-CD6, and anti-TCRαβ (T10B9) antibodies have all demonstrated low graft failure rates without compromising GVHD prophylaxis.49,52-54,83,84 150-152 

T-cell add-back to the marrow inoculum has been studied as a means of reducing graft rejection after TCD BMT. Using centrifugal elutriation technique for T-cell separation, Wagner and coworkers have shown the reintroduction of T lymphocytes to a dose of 0.5 × 106cells/kg protects against GVHD while maintaining engraftment at over 95%.59 Researchers have also added back donor T cells to marrow that had been T depleted using CAMPATH-1. However, these studies are problematic because of an increased risk of severe GVHD as the number of T cells added back is increased.153 154 

Dose escalation of CD34+ stem cells may be an effective strategy for overcoming graft failure after TCD BMT. Preclinical models have shown that mice given “megadoses” of TCD marrow could engraft despite sublethal doses of conditioning irradiation.155 In human studies, the addition of CD34+ cells to TCD marrow to augment stem cell dose has permitted reliable engraftment in leukemia patients despite full HLA haplotype mismatches.60 79 

Delayed immune reconstitution

Regeneration of the immune system after allogeneic BMT is a slow process that is often protracted by the presence of GVHD and its treatment. Peripheral blood T- lymphocyte counts do not normalize until 3 to 12 months after BMT,156 and their functional recovery frequently takes even longer.157-159 In vitro studies of T cells after BMT have demonstrated reduced proliferative response to mitogenic stimuli, abnormal cytotoxic T-cell effector function, and impaired ability to collaborate with B cells in immunoglobulin synthesis.157 160-166 Because of this delayed immune reconstitution, patients after allogeneic BMT remain susceptible to opportunistic infections for months to years.

After allogeneic BMT, regeneration of T-cell populations occurs by both thymic-dependent and thymic-independent pathways. The former pathway involves the positive or negative selection of thymocytes in the thymus and produces a T-cell population with a diverse T-cell receptor (TCR) repertoire. In contrast, the thymicindependent pathway, which reconstitutes the T-cell compartment by expansion of T cells infused along with the graft, produces a T-cell population with limited TCR diversity. Although the thymic- dependent pathway appears to be important in the regeneration of T cells after chemotherapy in children, it may be less active in adults whose thymuses are involuted.167,168 However, even in adults, the host thymus is required after BMT for the reconstitution of CD4+ and CD8+ naı̈ve subsets, and maintenance of TCR diversity.169 

Immune recovery after TCD BMT has been studied in a number of transplant centers using various methods of TCD.163,165,170-175 After allogeneic BMT, NK cells appear to be the first lymphoid subset to emerge, usually within 2 to 3 weeks after transplantation, followed by B cells (3-6 months) and T cells (3-12 months). Phenotypic analyses reveal that total lymphocyte numbers are usually higher early after BMT in recipients of conventional marrow compared to TCD grafts. Furthermore, the reconstituted T-cell compartment is predominantly of the CD8+ subset, and most TCD BMT patients will have a deficit in CD4+ cells, with an inverted CD4+ to CD8+ ratio for up to 2 years.165 The number of CD4+ cells normalizes at 7 to 9 months after conventional BMT, but this process is delayed after TCD BMT.170 

Functional recovery of T cells appears to be impaired after TCD BMT as well. Welte and colleagues reported that in recipients of TCD marrow, the proliferative response of peripheral blood mononuclear cells to exogenous IL-2 stimulation remained abnormal for up to 6 months, compared to only 1 month for recipients of conventional BMT.163 Similarly, the proliferative response of T cells to mitogenic stimulation can be impaired for over 18 months after TCD BMT.165 As another reflection of their decreased function, T lymphocytes from recipients of TCD BMT have significantly restricted variability in their TCR repertoires.173,175 This may be explained in part by the fact that the T-cell compartment after BMT is expanded mainly from lymphocytes cotransfused with the marrow, and therefore recipients of TCD transplants would have many fewer precursors with which to reconstitute their repertoire than recipients of conventional BMT. In support of this hypothesis, TCR analyses have shown the peripheral T-lymphocyte pool after TCD BMT to be the progeny of a limited number of precursors.173 

The delayed reconstitution of CD4+ cells and impaired recovery of T-cell repertoire diversity have led many to speculate that TCD BMT recipients may be at higher risk for opportunistic infections. Although there is little reported evidence to suggest an increased risk of bacterial or fungal infections after TCD transplantation, a number of studies have demonstrated a higher probability of reactivation for viruses such as CMV,122,124,125 and EBV, leading to EBV-associated B-cell lymphoproliferative disorders (EBV-LPDs).54,176 177 

EBV-LPDs

Although EBV-LPDs are well known in immunosuppressed patients after solid organ transplantation, it is surprisingly uncommon after BMT except in the TCD setting, where its incidence has been reported to be between 5% to 30%.54,176,177 Recipients of TCD transplants using HLA-mismatched or unrelated donor marrow appear to be at particularly high risk,138,176,178 as are patients with severe GVHD and those treated with certain anti–T-cell monoclonal antibodies.54,176 179 EBV-LPDs are felt to arise mainly from infected donor B cells that have been cotransplanted with the allograft. However, there have been cases of EBV lymphoma in B cells from EBV-seronegative donors, suggesting that de novo infection in transplant recipients or reactivation of EBV in recipient cells can occur.

Although combinations of IFN-α, intravenous γ-globulin, and high-dose acyclovir have some activity against EBV-LPDs, their response rates have been disappointing.177,180,181 In the past few years, donor lymphocyte infusion (DLI) has emerged as a potential effective treatment of EBV-LPDs.182 The therapeutic effect from DLI stems from the development of EBV-specific immunity mediated by CTLs present in the infusion. As an extension of this strategy, researchers have now demonstrated that administration of EBV-specific CTLs cultivated in vitro from donor lymphocytes is effective against EBV-LPDs.183-186 For patients who are not candidates for DLI or EBV-specific CTL therapy, recent small case series have reported promising responses with the use of the anti-CD20 monoclonal antibody rituximab.187,188 Rituximab is relatively safe and may prove to be as effective as DLI in the treatment of EBV-LPD. Polymerase chain reaction methods to detect EBV DNA may be an effective means of diagnosing patients with EBV-LPDs prior to the onset of clinically evident disease.189 As these and other methods of early detection improve, clinicians may now intervene with DLI or anti-CD20 antibody at a stage of low disease burden, and perhaps alter the poor outcome often associated with this condition.

Spurred by the advances in the understanding and treatment of EBV-LPDs, researchers have in recent years concentrated their efforts on prevention of this disease. B-cell depletion of the donor graft has been investigated as a strategy to reduce EBV-LPDs after TCD BMT. Cavazzana-Calvo and colleagues have shown that combined ex vivo depletion of B and T cells from the graft using monoclonal antibodies effectively prevented EBV-LPDs and improved survival compared to control patients who received TCD grafts without B-cell purging.190 Similarly, Hale and Waldman have reported that TCD BMT using CAMPATH-1 was associated with a minimal risk for EBV-LPDs.191 They attributed this risk reduction to the fact that CAMPATH-1 depletes both T and B cells, thereby removing donor B cells that are potentially infected with EBV from the donor marrow.191 Finally, the prophylactic administration of EBV-specific CTLs has been shown to reduced the incidence of EBV-LPDs after TCD BMT.185 186 

Leukemia relapse

The higher incidence of leukemia relapse associated with TCD BMT was first suggested in a prospective randomized trial which included 40 patients with acute and chronic leukemia.44 Of the 20 patients randomized to the TCD arm, 7 had clinically apparent relapse, compared to only 2 in the control (non-TCD) arm.44Although not statistically significant, this difference prompted researchers to investigate the possibility of higher disease relapse associated with TCD. Multiple retrospective studies have subsequently demonstrated that leukemia relapse is indeed more frequent after TCD BMT, especially for CML.8,100,126 192-196 

The increased rate of leukemia relapse after TCD BMT has been linked, as least in part, to the reduction in GVHD and concomitant loss of the GVL activity. In recipients of T-cell–replete marrow, patients who develop clinically significant GVHD have a lower incidence of leukemia relapse compared to those who do not develop GVHD.197-199Additional evidence linking T cells and GVL activity comes from studies using donor lymphocyte infusions in patients with CML who have relapsed after BMT, where complete response rates of 70% to 80% have been attained.200 201 

TCD BMT for CML.

An increased rate of relapse has been observed in virtually all studies using TCD of HLA-identical sibling bone marrow on patients with CML.100,126,192,193 The risk of relapse after matched related TCD BMT for CML chronic phase has been estimated to be 5- to 6-fold higher than that after conventional BMT.100,192 This relative risk has been reported to be as high as 18 for TCD transplantations done in accelerated phase CML.193 However, the increase in CML relapse is not as apparent after TCD transplantation using matched unrelated marrow.199,202-204 In one series, the 3-year probability of relapse for CML chronic phase was only 8% for recipients of unrelated TCD allografts, compared to 47% for those who received TCD marrow from HLA identical siblings.204 

TCD BMT for acute leukemia.

Unlike CML, TCD appears to have only a modest effect on the relapse rate of patients receiving transplants for acute leukemia. Reported rates of relapse after TCD BMT for acute myelogenous leukemia (AML) in first remission have ranged from 0% to 31% in different centers and are at least partly influenced by factors such as intensity of conditioning, extent and method of TCD, patient selection, and posttransplantation immunosuppression.83,89,205-207Retrospective data from the IBMTR, which included 731 TCD and 2480 non-TCD BMTs for leukemia, have shown that TCD is associated with a 1.7- to 2.0-fold increased risk for recurrence in patients with acute lymphoblastic leukemia (ALL) in any phase and in patients with AML undergoing transplantation in relapse or first remission.100 However, in a small randomized trial comparing TCD with methotrexate and cyclosporine as GVHD prophylaxis for leukemia patients undergoing HLA-matched related BMT, a higher relapse rate was observed after TCD BMT only in patients with CML, but not in patients with acute leukemia.208 

Leukemic relapse after alternative donor TCD BMT.

There have been conflicting reports on whether TCD adversely affects leukemia-free survival after alternative donor transplantation. In an analysis of 462 patients from the NMDP, TCD was associated with a significant increased risk for leukemia relapse at 2 years.71 More recently, Champlin and colleagues reported on the IBMTR experience from 870 patients who underwent T-depleted unrelated or mismatched donor BMT for leukemia.72 They discovered that the leukemia-free survival for patients whose grafts were depleted with “narrow specificity” antibodies (eg, anti-CD5, CD6, anti-TCRαβ) were significantly higher than those whose grafts had been depleted with “broad specificity” antibodies (eg, anti-CD2, ATG, CAMPATH-1, elutriation, or lectin/sheep red blood cell agglutination).72 Furthermore, the 5-year leukemia-free survival among recipients of narrow specificity TCD BMT was similar to that in recipients of non-TCD BMT (29% versus 31%,P = NS). These important results suggest that, at least in the setting of unrelated or mismatched BMT, TCD using narrow specificity antibodies is not associated with loss of GVL effect, but retains the advantage of decreased GVHD compared to conventional BMT.

TCD BMT for lymphoma/myeloma.

Although studies have shown lower lymphoma and myeloma relapse after allogeneic BMT than autologous transplantation,209-214 use of allogeneic BMT has been restricted in these diseases primarily because of disproportionately high rates of transplant-related mortality. Because TCD is associated with less toxicity, it has been suggested that TCD transplants may be particularly beneficial in these conditions. Small studies of matched sibling TCD BMT for non-Hodgkin lymphoma (NHL) and multiple myeloma (MM) have indeed demonstrated low rates of treatment-related toxicity and mortality.151 215However, whether TCD will protect against relapse and improve overall survival remains unknown.

Selective TCD

Although both GVL activity and GVHD are initiated by T cells in the donor graft, evidence suggests that different subsets of T lymphocytes may be involved in these processes and that GVL activity could exist in the absence of GVHD.216,217 In animal models, both donor CD4+ and CD8+ cells play a significant role in GVHD, but donor CD4+ cells in the absence of CD8+ cells can still mediate GVL activity.218 Based in part on these observations, Champlin and colleagues have shown that CD8+ depletion followed by posttransplantation cyclosporine can reduce the incidence and severity of GVHD without compromising GVL activity. In their initial study of 36 leukemia patients given CD8+-depleted matched sibling grafts, 2 (8%) developed grade III to IV GVHD, and only 3 (11%) had leukemia relapse at 2 years.62 In a follow-up double-blind randomized trial, the cyclosporine plus CD8+depletion arm experienced significantly less grade II to IV GVHD compared to the control arm receiving cyclosporine alone (20% versus 80%,P < .004), and the leukemia relapse rates were similar between the 2 groups.219 

Additional evidence to suggest that GVL activity can be separated from GVHD has emerged from studies of DLI given for relapsed disease after BMT. Although many DLI series have demonstrated an association between development of GVHD and antileukemic response, it is clear that in a number of patients, remission can be attained in the absence of GVHD. The GVHD/GVL dichotomy is further evidenced in DLI studies using CD8+ depletion. CD8+-depleted DLI has been shown to significantly reduce the incidence of GVHD, but retain GVL activity and clinical responses in patients with relapsed CML.220 221 Although studies of CD8+ depletion have been encouraging, the pathophysiologic differences between GVHD and GVL are likely more complicated than just CD4 and CD8 cells. Definitive dissection of the anatomic subsets underlying GVHD and GVL remains a crucial but elusive goal for investigators hoping to reduce disease relapse after TCD BMT.

Cytokine stimulation

Another strategy aimed at decreasing disease relapse rates after TCD BMT has been the administration of IL-2 after transplantation to stimulate activity of NK cells. Evidence has emerged to suggest that NK cells may be important mediators of antileukemic activity. Baker and coworkers have recently reported in murine models that CD8+NK/T cells expanded in culture with IL-2, IFN-γ, and anti-CD3 monoclonal antibody exhibit potent cytotoxic activity against syngeneic and allogeneic tumor targets both in vitro and in vivo.222When allogeneic mice across major histocompatibility barriers received these expanded CD8+ NK/T cell transplants, little GVHD was observed despite doses up to 20 × 106 cells. In contrast, when the same donor-recipient pairs were given transplants with 2.5 × 106 splenocytes rich in T cells, all developed acute GVHD and died within 20 days. Evidence for NK cell alloreactivity in GVL has also been suggested in human studies. Addition of IL-2 to peripheral blood lymphocytes collected from CML patients after allogeneic BMT has been shown to induce an NK cell cytolytic response against cryopreserved leukemic cell targets.223,224 In an analysis of 20 patients who received allografts grafts mismatched at the HLA-C or Bw4allele in the direction of GVHD, Ruggeri and coworkers were able to isolate donor versus recipient alloreactive NK cell clones at a high frequency early after transplantation, even though none of these patients had clinical evidence of GVHD.225 They further demonstrated that these alloreactive NK cell clones could lyse the patients' pretransplantation cryopreserved leukemia cells in vitro, suggesting that GVL activity mediated by NK cells exists in these patients without GVHD.

Interleukin-2 is known to stimulate both NK cells and activated T cells at high concentrations. However, IL-2 at low doses appears to stimulate NK cells preferentially. As such, it has been postulated that administration of low doses of IL-2 may enhance GVL activity through NK cell stimulation without inducing GVHD. Prolonged infusion of low-dose recombinant IL-2 following TCD allogeneic BMT has been shown to be well tolerated and results in a marked increase in NK cell numbers.226 Mononuclear cells taken from patients after low-dose IL-2 treatment have also been shown to possess substantially enhanced in vitro cytolytic activity against tumor cell lines.226 In a pilot clinical trial, the prolonged infusion of IL-2 of at least 4 weeks after TCD BMT appeared to lower the incidence of disease relapse relative to historical controls.227 Although promising, these preliminary results have yet to be tested in randomized studies.

DLI

Adoptive immunotherapy using DLI has also emerged as a potential approach to compensate for the higher rate of leukemia relapse after TCD BMT. Sehn and colleagues have retrospectively analyzed the outcomes of 46 CML patients who underwent TCD allogeneic BMT, and compared them to 40 similar patients who received non-TCD allogeneic transplants using cyclosporine and methotrexate.84 They found that, as expected, the TCD group had a lower incidence of GVHD and treatment-related mortality, but a higher probability of hematologic or molecular relapse compared to the non-TCD group. However, most of the patients suffering relapse were successfully salvaged with DLI, compensating for the initial higher relapse rate associated with TCD. Similarly, Drobyski and coworkers reported that despite a high cumulative incidence of CML relapse (49%) at 3 years after TCD BMT, the 5-year overall survival rate in their cohort was 80% because most patients having relapse achieved durable remissions with DLI therapy.85 Taken together, these results suggest that TCD BMT followed by posttransplantation DLI at disease relapse could be a reasonable option for patients with CML, especially for those with concurrent comorbidity or advanced age who would otherwise be suboptimal candidates for non-TCD BMT.

Functional TCD/induction of T-cell anergy

Instead of focusing on removal of anatomic subsets, some investigators have turned their attention to TCD techniques in which only alloreactive T cells are removed from the graft. In one such system, donor T cells are coincubated with recipient mononuclear cells in vitro. After a defined interval, alloreactive cells are identified by expression of activation markers, such as CD25 or CD69, and are physically separated from the remaining cells by immunomagnetic cell sorting.228,229 Using this method, Koh and colleagues have shown that approximately 90% of the alloreactive component could be purged while preserving more than 70% residual immunity as measured by third-party alloantigen response.228 It is postulated that this method would preferentially purge alloreactive lymphocytes from the graft that are responsible for GVHD, but retain nonreactive T cells, which may improve posttransplantation immune reconstitution and enhance engraftment.

As an extension of functional TCD, other investigators have attempted to induce anergy in donor T cells prior to transplantation as a means of reducing GVHD. Murine marrow transplant studies have shown that GVHD could be reduced even across major genetic barriers by treating the recipient with CTLA4-Ig, an agent that blocks the CTLA4-BB1 (also called CD28-B7) interaction between T lymphocytes and antigen-presenting cells.230,231 Blockade of this and other costimulatory pathways (LFA-1/ICAM, CD40-CD40L) have since been shown to deactivate T cells and induce a state of alloimmune tolerance after BMT.231-233 A recent study in pediatric patients has suggested that costimulatory signal blockade using CTLA-Ig may reduce GVHD after HLA-mismatched BMT.234 If differential targets for GVHD and GVL can be identified, it may be possible to expose donor marrow in vitro to GVHD targets in the presence of CTLA4-Ig to induce GVHD specific anergy, while preserving the T-cell response to tumor antigens for a full GVL effect.

Delayed T-cell add-back/prophylactic DLI

The success of DLI in salvaging CML patients after BMT has led many researchers to investigate the potential of using delayed T-cell add-back to eliminate residual disease after TCD BMT. The attraction of this approach is that it allows one to reap the benefits of TCD early after BMT (ie, decreased GVHD and transplant-related toxicity), and yet be able to restore the GVL effect at a later time with DLI. Alyea and colleagues have recently conducted a trial of delayed T-cell add-back in 14 patients with MM after TCD allogeneic BMT.235 Of the 11 patients who had persistent myeloma 6 months after transplantation, 10 responses (6 complete, 4 partial) were observed after DLI, and the 2-year progression-free survival for the 14 patients who received DLI was significantly improved compared to a comparable historical cohort who received TCD BMT without DLI. After nonmyeloablative stem cell transplantation, the GVL effect induced by DLI may be sufficient to prevent relapse in leukemic patients with minimal residual disease.236 

Even though DLI is performed when the patient is further removed from conditioning therapy and the associated inflammatory cytokine milieu, GVHD remains a major complication. Existing data on DLI show a GVHD incidence between 40% and 80%, and an attendant mortality of nearly 20%. The solution to this GVHD problem may lie in the correct timing and dosing of lymphocytes with the T-cell add-back, but neither of these parameters is known at this time. In patients given DLI for relapsed disease, GVHD incidence and severity have been decreased without loss of GVL by lowering the dose of lymphocytes infused,237 or by selectively depleting CD8+cells from the lymphocyte pool.221,238 For prophylactic DLI to be viable, it must reduce relapse rates without inducing GVHD. This may be possible by using DLI with a fixed number of selected T cells. A randomized trial of prophylactic DLI 6 months after TCD BMT has attempted to address this issue. Patients were randomized to receive either unselected or CD8+-depleted lymphocytes, adjusted to a CD4+ dose of 1.0 × 107CD4+ cells/kg. None of 9 patients receiving CD8+-depleted lymphocytes developed GVHD, compared to 6 of 9 patients receiving unselected cells (P = .003). All patients experienced conversion to complete donor chimerism, and relapse rates were similarly low in both groups.244 

In recent years, researchers have introduced the herpes simplex thymidine kinase (HSTK) gene into donor T cells as a novel approach for controlling GVHD after DLI. The insertion of this “suicide” gene into donor T lymphocytes renders them susceptible to destruction with ganciclovir, and therefore provides a reliable means of eliminating these cells should severe GVHD develop after the infusion.239,240 The use of suicide gene therapy may also be applicable in conjunction with TCD BMT. Tiberghien and coworkers have recently shown that HSTK gene-modified T lymphocytes infused along with TCD marrow at the time of transplantation did not interfere with engraftment.241 More significantly, they were able to demonstrate long-lasting circulation of the gene-modified cells after transplantation, and in 2 of 3 patients, GVHD response on treatment with ganciclovir.241 A case of chronic cutaneous GVHD responsive to ganciclovir has also been recently reported in a patient who had received T cells bearing the HSTK gene at the time of BMT.242 

Vaccine strategies

As new leukemia antigens are identified that are potential targets for the GVL response, allogeneic tumor vaccines may be developed to stimulate specific antileukemic activity without GVHD. As an example, a recently identified peptide (PR1) from the serine protease proteinase 3, which is aberrantly expressed in myeloid leukemias, may serve as a specific target for the GVL response.243 Investigators have successfully isolated PR-1–specific CTLs from the peripheral blood of patients with CML. PR-1–specific CTLs were found in 11 of 12 patients who responded to IFN therapy, but none of the 7 patients who did not respond. Furthermore, 6 of 8 patients who responded to allogeneic BMT had PR-1–specific CTLs in their blood, but in the one patient who relapsed after BMT, no PR-1–specific CTL could be detected.243Based on these results, clinical trials of a PR-1 peptide vaccine are currently being conducted in patients with myeloid leukemia, and adoptive cellular therapy using PR-1–specific CTLs may one day replace DLI as a method for eradicating leukemia cells without GVHD after TCD transplantation.

It is frustrating that after 2 decades, we have not been able to establish the role of TCD in transplantation. We still do not have a clear idea of who should receive a TCD BMT, or how marrow or stem cells should be purged. It remains unclear whether additional medications are needed to promote engraftment or control GVHD, or what the nature and timing of immunomodulating manipulations to reduce the risk of relapse should be. There have been no definitive randomized trials to answer these questions to date, partly because researchers have not been able to agree on a single TCD strategy or the best way to engineer a graft. The optimal number of T cells to include in the graft remains unknown and may in fact vary among donor-recipient pairs. It would be ideal to be able to manipulate different lymphoid subgroups responsible for GVHD and GVL. Being able to do so will be critical to the future success of allogeneic stem cell transplantation.

Supported in part by NIH grant AI29530.

R.J.S. is a clinical scholar of the Leukemia Society of America.

© 2001 by The American society of Hematology

1
Gale
RP
Bortin
MM
Van Bekkum
DW
et al
Risk factors for acute graft-versus-host disease.
Br J Haematol.
67
1987
396
406
2
Martin
PJ
Schoch
G
Fisher
L
et al
A retrospective analysis of therapy for acute graft-versus-host disease: initial treatment.
Blood.
76
1990
1464
1472
3
Weisdorf
D
Haake
R
Blazar
B
et al
Treatment of moderate/severe acute graft-versus-host disease after allogeneic bone marrow transplantation: an analysis of clinical risk features and outcome.
Blood.
75
1990
1024
1030
4
Ratanatharathorn
V
Nash
RA
Przepiorka
D
et al
Phase III study comparing methotrexate and tacrolimus (prograf, FK506) with methotrexate and cyclosporine for graft-versus-host disease prophylaxis after HLA-identical sibling bone marrow transplantation.
Blood.
92
1998
2303
2314
5
Nash
RA
Antin
JH
Karanes
C
et al
Phase 3 study comparing methotrexate and tacrolimus with methotrexate and cyclosporine for prophylaxis of acute graft-versus-host disease after marrow transplantation from unrelated donors.
Blood.
96
2000
2062
2068
6
Giralt
S
Khouri
I
Champlin
R
Non myeloablative “mini transplants.”
Cancer Treat Res.
101
1999
97
108
7
Nash
RA
Pepe
MS
Storb
R
et al
Acute graft-versus-host disease: analysis of risk factors after allogeneic marrow transplantation and prophylaxis with cyclosporine and methotrexate.
Blood.
80
1992
1838
1845
8
Horowitz
MM
Gale
RP
Sondel
PM
et al
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood.
75
1990
555
562
9
Sullivan
KM
Shulman
HM
Storb
R
et al
Chronic graft versus host disease in 52 patients: adverse natural course and successful treatment with combination immunosuppression.
Blood.
57
1981
267
276
10
Champlin
RE
Schmitz
N
Horowitz
MM
et al
Blood stem cells compared with bone marrow as a source of hematopoietic cells for allogeneic transplantation: IBMTR Histocompatibility and Stem Cell Sources Working Committee and the European Group for Blood and Marrow Transplantation (EBMT).
Blood.
95
2000
3702
3709
11
Cutler
C
Giri
S
Jeyapalan
S
et al
Incidence of acute and chronic graft-versus-host disease after allogeneic peripheral blood stem cell and bone marrow transplantation: a meta-analysis [abstract].
Blood.
96
2000
205a
12
Atkinson
K
Horowitz
MM
Gale
RP
et al
Risk factors for chronic graft-versus-host disease after HLA-identical bone marrow transplantation.
Blood.
75
1990
2459
2464
13
Ferrera
JLM
Antin
JH
Pathophysiology of graft-versus-host disease.
Hematopoietic Stem Cell Transplantation.
Thomas
ED
Blume
KG
Forman
SJ
1999
305
315
Blackwell Scientific Publications
Boston, MA
14
Via
CS
Finkelman
FD
Critical role of interleukin-2 in the development of acute graft-versus-host disease.
Int Immunol.
5
1993
565
572
15
Antin
J
Weisdorf
D
Neuberg
D
et al
Interleukin-1 blockade does not prevent acute graft versus host disease (GVHD): results of a randomized double blind, placebo-controlled trial of interleukin 1 receptor antagonist (IL-1RA) [abstract].
Blood.
94
1999
152a
16
Antin
J
Lee
S
Harkness
S
et al
Preliminary results of a phase I/II double-blind, placebo-controlled study of recombinant human interleukin-11 (rhIL-11) for mucositis and GVHD prevention in allogeneic transplantation [abstract].
Blood.
96
2000
786a
17
Dicke
KA
van Hoot
JIM
van Bekkum
DW
The selective elimination of immunologically competent cells from bone marrow and lymphatic cell mixtures; II: mouse spleen cell fractionation on a discontinuous albumin gradient.
Transplantation.
6
1968
562
570
18
Reisner
Y
Itzicovitch
L
Meshorer
A
Sharon
N
Hematopoietic stem cell transplantation using mouse bone marrow and spleen cells fractionated by lectins.
Proc Natl Acad Sci U S A.
75
1978
2933
2936
19
Trentin
JJ
Judd
KP
Prevention of acute graft-versus-host (GVH) mortality with spleen-absorbed antithymocyte globulin (ATG).
Transplant Proc.
5
1973
865
868
20
Rodt
H
Theirfelder
S
Eulitz
M
Antilymphocyte antibodies and marrow transplantation; III: effect of heterologous anti-brain antibodies on acute secondary disease in mice.
Eur J Immunol.
4
1974
25
29
21
Rodt
H
Thierfelder
S
Eulitz
M
Antilymphocytic antibodies and marrow transplantation; IV: comparison of the effects of antibody fragments directed against immunoglobulin or lymphocyte antigens on acute secondary disease.
Exp Hematol.
2
1974
195
203
22
Muller-Ruchholtz
W
Wottge
H-U
Muller-Hermelink
HK
Selective grafting of hematopoietic cells.
Transplant Proc.
7
1975
859
862
23
Korngold
R
Sprent
J
Lethal graft-versus-host disease after bone marrow transplantation across minor histocompatibility barriers in mice: prevention by removing mature T cells from marrow.
J Exp Med.
148
1978
1687
1698
24
Kolb
HJ
Rieder
I
Rodt
H
et al
Antilymphocyte antibodies and marrow transplantation; VI: graft-versus-host tolerance in DLA-incompatible dogs after in vitro treatment of bone marrow absorbed with antithymocyte globulin.
Transplantation.
27
1979
242
245
25
Rodt
H
Kolb
HJ
Netzel
B
et al
GVHD suppression by incubation of bone marrow grafts with anti-T-cell globulin: effect in canine model and application to clinical bone marrow transplantation.
Transplant Proc.
11
1979
962
966
26
Vallera
DA
Soderling
CCB
Carlson
GJ
Kersey
JH
Bone marrow transplantation across major histocompatibility barriers in mice.
Transplantation.
31
1981
218
222
27
Reisner
Y
Kapoor
N
Kirkpatrick
D
et al
Transplantation for acute leukemia with HLA-A and B nonidentical parental marrow cells fractionated with soybean agglutinin and sheep red blood cells.
Lancet.
2
1981
327
331
28
Reisner
Y
Kapoor
N
Kirkpatrick
D
et al
Transplantation for severe combined immunodeficiency with HLA-A, B, D, DR incompatibility parental marrow cells fractionated by soybean agglutinin and sheep red blood cells.
Blood.
61
1983
341
348
29
De Witte
T
Hoogenhout
J
de Pauw
B
et al
Depletion of donor lymphocytes by counterflow centrifugation successfully prevents acute graft-versus-host disease in matched allogeneic marrow transplantation.
Blood.
67
1986
1302
1308
30
Noga
SJ
Donnenberg
AD
Schwartz
CL
Strauss
LC
Civin
CI
Santos
GB
Development of a simplified counterflow centrifugation elutriation procedure for depletion of lymphocytes from human bone marrow.
Transplantation.
41
1986
220
228
31
Wagner
JE
Donnenberg
AD
Noga
SJ
et al
Lymphocyte depletion of donor bone marrow by counterflow centrifugal elutriation: results of a phase I clinical trial.
Blood.
72
1988
1168
1176
32
Lowenberg
B
Wagemaker
E
van Bekkum
DW
et al
Graft-versus-host disease following transplantation of “one log” versus “two log” T-lymphocyte depleted bone marrow from HLA-identical donors.
Bone Marrow Transplant.
1
1986
133
140
33
Rodt
H
Kolb
HJ
Netzel
B
et al
Effect of anti-T-cell globulin on GVHD in leukemic patients treated with BMT.
Transplant Proc.
13
1981
257
261
34
Prentice
HG
Blacklock
HA
Janossy
G
et al
Use of anti-T-cell monoclonal antibody OKT3 to prevent acute graft versus host disease in allogeneic bone marrow transplantation for acute leukemia.
Lancet.
1
1982
700
703
35
Filipovich
AH
McGlave
PB
Ramsay
NKC
et al
Pretreatment of donor bone marrow with monoclonal antibody OKT3 for prevention of acute graft versus host disease in allogeneic histocompatible bone marrow transplantation.
Lancet.
1
1982
1266
1269
36
Martin
PJ
Hansen
JA
Thomas
ED
Preincubation of donor bone marrow cells with a combination of murine monoclonal anti-T-cell antibodies without complement does not prevent graft-versus-host disease after allogeneic marrow transplantation.
J Clin Immunol.
4
1984
18
22
37
Reinherz
EL
Geha
R
Rappeport
JM
et al
Reconstitution after transplantation with T-lymphocyte-depleted HLA haplotype-mismatched bone marrow for severe combined immunodeficiency.
Proc Natl Acad Sci U S A.
79
1982
6047
6051
38
Gilmore
MJML
Prentice
HG
Price-Jones
E
et al
Allogeneic bone marrow transplantation: the monitoring of granulocyte macrophage colonies following the collection of bone marrow mononuclear cells and after subsequent in-vitro cytolysis of OKT3 positive lymphocytes.
Br J Haematol.
55
1983
587
593
39
Prentice
HG
Janossy
G
Price-Jones
L
et al
Depletion of T lymphocytes in donor marrow prevents significant graft-versus-host disease in matched allogeneic leukemic marrow transplant recipients.
Lancet.
1
1984
472
475
40
Waldmann
HG
Hale
G
Cividalli
G
et al
Elimination of graft-versus-host disease by in vitro depletion of alloreactive lymphocytes with a monoclonal rat anti-human lymphocyte antibody (Campath-1).
Lancet.
2
1984
483
486
41
Martin
PJ
Hansen
JA
Buckner
CD
et al
Effects of in vitro depletion of T cells in HLA-identical allogeneic marrow grafts.
Blood.
66
1985
664
672
42
Herve
P
Flesch
M
Cahn
JY
et al
Removal of marrow T cells with OKT3-OKT11 monoclonal antibodies and complement to prevent graft-versus-host disease.
Transplantation.
39
1985
138
143
43
Trigg
ME
Billing
R
Sondel
PM
et al
Clinical trial depleting T lymphocytes from donor marrow for matched and mismatched allogeneic bone marrow transplants.
Cancer Treat Reports.
69
1985
377
386
44
Mitsuyasu
RT
Champlin
RE
Gale
RP
et al
Treatment of donor bone marrow with monoclonal anti-T-cell antibody and complement for the prevention of graft versus host disease.
Ann Intern Med.
105
1986
20
26
45
Maraninchi
D
Gluckman
E
Blaise
D
et al
Impact of T-cell depletion on outcome of allogeneic bone-marrow transplantation for standard-risk leukaemia.
Lancet.
2
1987
175
178
46
Maraninchi
D
Mawas
C
Guyotat
D
et al
Selective depletion of marrow-T cytotoxic lymphocytes (CD8) in the prevention of graft-versus-host disease after allogeneic bone marrow transplantation.
Transplant Int.
1
1988
91
94
47
Cahn
JY
Herve
P
Flesch
M
et al
Marrow transplantation from HLA non-identical family donors for the treatment of leukaemias: a pilot study of 15 patients using additional immunosuppression and T-cell depletion.
Br J Haematol.
69
1988
345
349
48
Gabert
J
Thuret
I
Lafage
M
Carcassonne
Y
Maraninchi
D
Mannoni
P
Detection of residual bcr/abl translocation by polymerase chain reaction in chronic myeloid leukaemia patients after bone-marrow transplantation.
Lancet.
2
1989
1125
1128
49
Soiffer
RJ
Murray
C
Mauch
P
et al
Prevention of graft-versus-host disease by selective depletion of CD6-positive T lymphocytes from donor bone marrow.
J Clin Oncol.
10
1992
1191
2000
50
Filipovich
AH
Vallera
DA
Youle
RJ
Neville
DM
Kersey
JH
Ex vivo T cell depletion with immunotoxins in allogeneic bone marrow transplantation: the pilot clinical study for prevention of graft-versus-host disease.
Transplant Proc.
17
1985
442
444
51
Filipovich
AH
Vallera
DA
Youle
RJ
et al
Graft-versus-host disease prevention in allogeneic bone marrow transplantation from histocompatible siblings.
Transplantation.
44
1987
62
69
52
Laurent
G
Maraninchi
D
Gluckman
E
et al
Donor bone marrow treatment with T101 Fab fragment-ricin A-chain immunotoxin prevents graft-versus-host disease.
Bone Marrow Transplant.
4
1989
367
371
53
Filipovich
AH
Vallera
D
McGlave
P
et al
T cell depletion with anti-CD5 immunotoxin in histocompatible bone marrow transplantation.
Transplantation.
50
1990
410
415
54
Antin
JH
Bierer
BE
Smith
BR
et al
Selective depletion of bone marrow T lymphocytes with anti-CD5 monoclonal antibodies: effective prophylaxis for graft-versus-host disease in patients with hematologic malignancies.
Blood.
78
1991
2139
2149
55
Vartdal
F
Albrechtsen
D
Ringden
O
et al
Immunomagnetic treatment of bone marrow allografts.
Bone Marrow Transplant.
2
1987
94
98
56
Dreger
P
Viehmann
K
Steinman
J
et al
G-CSF-mobilized peripheral blood progenitor cells for allogeneic transplantation: comparison of T cell depletion strategies using different CD34+ selection systems or Campath-1.
Exp Hematol.
23
1995
147
154
57
Martin
PJ
Hansen
JA
Quantitative assays for detection of residual T cells in T-depleted human marrow.
Blood.
65
1985
1134
1140
58
Kernan
NA
Flomenberg
N
Collins
NH
O'Reilly
RJ
Dupont
B
Quantification of T-lymphocytes in human bone marrow by limiting assay.
Transplantation.
40
1985
317
322
59
Wagner
JE
Santos
GW
Noga
SJ
et al
Bone marrow graft engineering by counterflow elutriation: results of a phase I-II clinical trial.
Blood.
75
1990
1370
1377
60
Aversa
F
Tabilio
A
Velardi
A
et al
Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype.
N Engl J Med.
339
1998
1186
1193
61
Hale
G
Cobbold
S
Waldmann
H
T-cell depletion with Campath-1 in allogeneic bone marrow transplantation.
Transplantation.
45
1988
753
759
62
Champlin
R
Ho
W
Gajewski
J
et al
Selective depletion of CD8+ T lymphocytes for prevention of graft-versus-host disease after allogeneic bone marrow transplantation.
Blood.
76
1990
418
423
63
Jansen
J
Hanks
S
Akard
L
et al
Selective T cell depletion with CD8-conjugated magnetic beads in the prevention of graft-versus-host disease after allogeneic bone marrow transplantation.
Bone Marrow Transplant.
15
1995
271
278
64
Nimer
SD
Giorgi
J
Gajewski
JL
et al
Selective depletion of CD8+ cells for prevention of graft-versus-host disease after bone marrow transplantation: a randomized controlled trial.
Transplantation.
57
1994
82
87
65
Bensinger
WI
Buckner
CD
Shannon-Dorcy
K
et al
Transplantation of allogeneic CD34+ peripheral blood stem cells in patients with advanced hematologic malignancy.
Blood.
88
1996
4132
4138
66
Finke
J
Brugger
W
Bertz
H
et al
Allogeneic transplantation of positively selected peripheral blood CD34+ progenitor cells from matched related donors.
Bone Marrow Transplant.
18
1996
1081
1086
67
Link
H
Arseniev
L
Bahre
O
Kadar
JG
Diedrich
H
Poliwoda
H
Transplantation of allogeneic CD34+ blood cells.
Blood.
87
1996
4903
4909
68
Urbano-Ispizua
A
Solano
C
Brunet
S
et al
Allogeneic transplantation of selected CD34+ cells from peripheral blood: experience of 62 cases using immunoadsorption or immunomagnetic technique. Spanish Group of Allo-PBT.
Bone Marrow Transplant.
22
1998
519
525
69
Vij
R
Brown
R
Shenoy
S
et al
Allogeneic peripheral blood stem cell transplantation following CD34+ enrichment by density gradient separation.
Bone Marrow Transplant.
25
2000
1223
1228
70
Hale
G
Jacobs
P
Wood
L
et al
CD52 antibodies for prevention of graft-versus-host disease and graft rejection following transplantation of allogeneic peripheral blood stem cells.
Bone Marrow Transplant.
26
2000
69
76
71
Kernan
NA
Bartsch
G
Ash
RC
et al
Analysis of 462 transplantations from unrelated donors facilitated by the National Marrow Donor Program.
N Engl J Med.
328
1993
593
602
72
Champlin
RE
Passweg
JR
Zhang
MJ
et al
T-cell depletion of bone marrow transplants for leukemia from donors other than HLA-identical siblings: advantage of T-cell antibodies with narrow specificities.
Blood.
95
2000
3996
4003
73
Soiffer
RJ
Weller
E
Alyea
EP
et al
CD6+ donor marrow T-cell depletion as the sole form of graft-versus-host disease prophylaxis in patients undergoing allogeneic bone marrow transplant from unrelated donors.
J Clin Oncol.
19
2001
1152
1159
74
Drobyski
WR
Ash
RC
Casper
JT
et al
Effect of T-cell depletion as graft-versus-host disease prophylaxis on engraftment, relapse, and disease-free survival in unrelated marrow transplantation for chronic myelogenous leukemia.
Blood.
83
1994
1980
1987
75
Marks
DI
Bird
JM
Vettenranta
K
et al
T cell-depleted unrelated donor bone marrow transplantation for acute myeloid leukemia.
Biol Blood Marrow Transplant.
6
2000
646
653
76
Soiffer
RJ
Mauch
P
Fairclough
D
et al
CD6+ T cell depleted allogeneic bone marrow transplantation from genotypically HLA nonidentical related donors.
Biol Blood Marrow Transplant.
3
1997
11
17
77
Henslee-Downey
PJ
Parrish
RS
MacDonald
JS
et al
Combined in vitro and in vivo T lymphocyte depletion for the control of graft-versus-host disease following haploidentical marrow transplant.
Transplantation.
61
1996
738
745
78
Henslee-Downey
PJ
Abhyankar
SH
Parrish
RS
et al
Use of partially mismatched related donors extends access to allogeneic marrow transplant.
Blood.
89
1997
3864
3872
79
Aversa
F
Tabilio
A
Terenzi
A
et al
Successful engraftment of T-cell-depleted haploidentical “three-loci” incompatible transplants in leukemia patients by addition of recombinant human granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells to bone marrow inoculum.
Blood.
84
1994
3948
3955
80
Soiffer
RJ
Dear
K
Rabinowe
SN
et al
Hepatic dysfunction following T-cell-depleted allogeneic bone marrow transplantation.
Transplantation.
52
1991
1014
1019
81
Breuer
R
Or
R
Lijovetzky
G
et al
Interstitial pneumonitis in T cell-depleted bone marrow transplantation.
Bone Marrow Transplant.
3
1988
625
630
82
Ho
VT
Weller
E
Lee
SJ
Alyea
EP
Antin
JH
Soiffer
RJ
Prognostic factors for early severe pulmonary complications after hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant.
7
2001
223
229
83
Soiffer
RJ
Fairclough
D
Robertson
M
et al
CD6-depleted allogeneic bone marrow transplantation for acute leukemia in first complete remission.
Blood.
89
1997
3039
3047
84
Sehn
LH
Alyea
EP
Weller
E
et al
Comparative outcomes of T-cell-depleted and non-T-cell-depleted allogeneic bone marrow transplantation for chronic myelogenous leukemia: impact of donor lymphocyte infusion.
J Clin Oncol.
17
1999
561
568
85
Drobyski
WR
Hessner
MJ
Klein
JP
et al
T-cell depletion plus salvage immunotherapy with donor leukocyte infusions as a strategy to treat chronic-phase chronic myelogenous leukemia patients undergoing HLA-identical sibling marrow transplantation.
Blood.
94
1999
434
441
86
Hale
G
Zhang
MJ
Bunjes
D
et al
Improving the outcome of bone marrow transplantation by using CD52 monoclonal antibodies to prevent graft-versus-host disease and graft rejection.
Blood.
92
1998
4581
4590
87
Hale
G
Waldmann
H
Control of graft-versus-host disease and graft rejection by T cell depletion of donor and recipient with Campath-1 antibodies: results of matched sibling transplants for malignant diseases.
Bone Marrow Transplantation.
13
1994
597
611
88
Novitzky
N
Thomas
V
Hale
G
et al
Ex vivo depletion of T cells from bone marrow grafts with CAMPATH-1 in acute leukemia: graft-versus-host disease and graft-versus-leukemia effect.
Transplantation.
67
1999
620
626
89
Papadopoulos
EB
Carabasi
MH
Castro-Malaspina
H
et al
T-cell-depleted allogeneic bone marrow transplantation as postremission therapy for acute myelogenous leukemia: freedom from relapse in the absence of graft-versus-host disease.
Blood.
91
1998
1083
1090
90
Beatty
PG
Clift
RA
Mickelson
EM
et al
Marrow transplantation for related donors other than HLA-identical siblings.
N Engl J Med.
313
1985
765
771
91
Powles
RL
Morgenstern
GR
Kay
HE
et al
Mismatched family donors for bone marrow transplantation as treatment for acute leukemia.
Lancet.
1
1983
612
615
92
Patterson
J
Prentice
HG
Brenner
MK
et al
Graft rejection following HLA matched T-lymphocyte depleted bone marrow transplantation.
Br J Haematol.
63
1986
221
230
93
O'Reilly
R
Collins
NH
Kernan
N
et al
Transplantation of marrow-depleted T cells by soybean lectin agglutination and E-rosette depletion: major histocompatibility complex-related graft resistance in leukemic transplant patients.
Transplant Proc.
17
1985
455
94
Martin
PJ
Hansen
JA
Torok-Storb
B
et al
Effects of treating marrow with a CD3-specific immunotoxin for prevention of acute graft-versus-host disease.
Bone Marrow Transplant.
3
1988
437
444
95
Kernan
NA
Bordignon
C
Heller
G
et al
Graft failure after T-cell-depleted leukocyte antigen identical marrow transplants for leukemia; I: analysis of risk factors and results of secondary transplants.
Blood.
74
1989
2227
2236
96
Champlin
RE
Horowitz
MM
van Bekkum
DW
et al
Graft failure following bone marrow transplantation for severe aplastic anemia: risk factors and treatment results.
Blood.
73
1989
606
613
97
Delain
M
Cahn
JY
Racadot
E
et al
Graft failure after T cell depleted HLA identical allogeneic bone marrow transplantation: risk factors in leukemic patients.
Leuk Lymphoma.
11
1993
359
368
98
Herve
P
Cahn
JG
Flesch
M
et al
Successful graft-versus-host disease prevention without graft failure in 32 HLA-identical allogeneic bone marrow transplantations with marrow depleted of T cells by monoclonal antibodies and complement.
Blood.
69
1987
388
393
99
Martin
PJ
Hansen
JA
Torok-Storb
B
et al
Graft failure in patients receiving T cell-depleted HLA-identical allogeneic marrow transplants.
Bone Marrow Transplant.
3
1988
445
456
100
Marmont
A
Horowitz
MM
Gale
RP
et al
T-cell depletion of HLA-identical transplants in leukemia.
Blood.
78
1991
2120
2130
101
Guyotat
D
Dutou
L
Erhsam
A
et al
Graft rejection after T cell-depleted marrow transplantation: role of fractionated irradiation.
Br J Haematol.
65
1987
499
507
102
Burnett
AK
Hann
IM
Robertson
AG
et al
Prevention of graft-versus-host disease by ex vivo T cell depletion: reduction in raft failure with augmented total body irradiation.
Leukemia.
2
1988
300
303
103
Sondel
PM
Hank
JA
Trigg
ME
et al
Transplantation of HLA-haploidentical T cell-depleted marrow for leukemia: autologous marrow recovery with specific immune sensitization to donor antigens.
Exp Hematol.
14
1986
278
286
104
Bunjes
D
Heit
W
Arnold
R
et al
Evidence for the involvement of host derived OKT8-positive T cells in the rejection of T-depleted, HLA-identical bone marrow grafts.
Transplantation.
43
1987
501
505
105
Bunjes
D
Theobald
M
Wiesneth
M
et al
Graft rejection by a population of primed CDw52-host T cells after in vivo/ex vivo T-depleted bone marrow transplantation.
Bone Marrow Transplant.
12
1993
209
215
106
Kernan
NA
Flomenberg
N
Dupont
B
O'Reilly
RJ
Graft rejection in recipients of T cell depleted HLA-nonidentical marrow transplants for leukemia.
Transplantation.
43
1987
842
847
107
Bierer
BE
Emerson
SG
Antin
J
et al
Regulation of cytotoxic T lymphocyte-mediated graft rejection following bone marrow transplantation.
Transplantation.
49
1990
714
720
108
Bordignon
C
Keever
CA
Small
TN
et al
Graft failure after T-cell-depleted leukocyte antigen identical marrow transplants for leukemia; II: in vitro analysis of host effector mechanisms.
Blood.
74
1989
2237
2243
109
Bosserman
L
Murray
C
Takvorian
T
et al
Mechanism of graft failure in HLA-matched and HLA-mismatched bone marrow transplant recipients.
Bone Marrow Transplant.
4
1989
239
245
110
Voogt
PJ
Fibbe
WE
Marjit
WA
et al
Rejection of bone marrow graft by recipient derived cytotoxic T lymphocytes against minor histocompatibility antigens.
Lancet.
335
1990
135
144
111
Fleischauer
K
Kernan
NA
O'Reilly
RJ
Dupont
B
Tang
SY
Bone marrow-allograft rejection by T lymphocytes recognizing a single amino acid difference on HLA-B44.
N Engl J Med.
323
1990
1818
1822
112
Donohue
J
Homge
M
Kernan
NA
Characterization of cells emerging at the time of graft failure after bone marrow transplantation from an unrelated bone marrow donor.
Blood.
82
1993
1023
1029
113
Bozdech
MJ
Sondel
PM
Trigg
ME
et al
Transplantation of HLA-haploindentical T-cell-depleted marrow for leukemia: addition of cytosine arabinoside to the pretransplant conditioning prevents rejection.
Exp Hematol.
13
1985
1201
1210
114
Gerritsen
WR
Wagemaker
G
Jonker
M
et al
The repopulation capacity of bone marrow grafts following pretreatment with monoclonal antibodies against T lymphocytes in rhesus monkeys.
Transplantation.
45
1988
301
307
115
Anderson
KC
Barut
BA
Ritz
J
et al
Monoclonal antibody-purged autologous bone marrow transplantation therapy for multiple myeloma.
Blood.
77
1991
712
720
116
Soiffer
RJ
Roy
DC
Gonin
R
et al
Monoclonal antibody-purged autologous bone marrow transplantation in adults with acute lymphoblastic leukemia at high risk of relapse.
Bone Marrow Transplant.
12
1993
243
251
117
Reddehase
MJ
Dreher-Stumpp
L
Angele
P
Balthesen
M
Susa
M
Hematopoietic stem cell deficiency resulting from cytomegalovirus infection of bone marrow stroma.
Ann Hematol.
64(suppl A)
1992
125
127
118
Mutter
W
Reddehase
MJ
Busch
FW
Buhring
HJ
Koszinowski
UH
Failure in generating hemopoietic stem cells is the primary cause of death from cytomegalovirus disease in the immunocompromised host.
J Exp Med.
67
1988
1645
1658
119
Steffens
HP
Podlech
J
Kurz
S
Angele
P
Dreis
D
Reddehase
MJ
Cytomegalovirus inhibits the engraftment of donor bone marrow cells by downregulation of hemopoietin gene expression in recipient stroma.
J Virol.
72
1998
5006
5015
120
Johnston
RE
Geretti
AM
Prentice
HG
et al
HHV-6-related secondary graft failure following allogeneic bone marrow transplantation.
Br J Haematol.
105
1999
1041
1043
121
Rosenfeld
CS
Rybka
WB
Weinbaum
D
et al
Late graft failure due to dual bone marrow infection with variants A and B of human herpesvirus-6.
Exp Hematol.
23
1995
626
629
122
Engelhard
D
Or
R
Strauss
N
et al
Cytomegalovirus infection and disease after T cell depleted allogeneic bone marrow transplantation for malignant hematologic diseases.
Transplant Proc.
21
1989
3101
3102
123
Hertenstein
B
Hampl
W
Bunjes
D
et al
In vivo/ex vivo T cell depletion for GVHD prophylaxis influences onset and course of active cytomegalovirus infection and disease after BMT.
Bone Marrow Transplant.
15
1995
387
393
124
Couriel
D
Canosa
J
Engler
H
Collins
A
Dunbar
C
Barrett
AJ
Early reactivation of cytomegalovirus and high risk of interstitial pneumonitis following T-depleted BMT for adults with hematological malignancies.
Bone Marrow Transplant.
18
1996
347
353
125
Maltezou
H
Whimbey
E
Abi-Said
D
et al
Cytomegalovirus disease in adult marrow transplant recipients receiving ganciclovir prophylaxis: a retrospective study.
Bone Marrow Transplant.
24
1999
665
669
126
Apperley
JF
Mauro
FR
Goldman
JM
et al
Bone marrow transplantation for chronic myeloid leukaemia in first chronic phase: importance of a graft-versus-leukaemia effect.
Br J Haematol.
69
1988
239
245
127
Bertheas
MF
Lafage
M
Levy
P
et al
Influence of mixed chimerism on the results of allogeneic bone marrow transplantation for leukemia.
Blood.
78
1991
3103
3106
128
Offit
K
Burns
JP
Cunningham
I
et al
Cytogenetic analysis of chimerism and leukemia relapse in chronic myelogenous leukemia patients after T cell-depleted bone marrow transplantation.
Blood.
75
1990
1346
1355
129
Mackinnon
S
Barnett
L
O'Reilly
R
Minimal residual disease is more common in patients who have mixed T-cell chimerism after bone marrow transplantation for chronic myelogenous leukemia.
Blood.
83
1994
3409
3416
130
van Leeuwen
JEM
van Tol
MJD
Joosten
AM
et al
Mixed T-lymphoid chimerism after allogeneic bone marrow transplantation for hematologic malignancies of children is not correlated with relapse.
Blood.
82
1993
1921
1928
131
Butturini
A
Seeger
RC
Gale
RP
Recipient immune-competent T lymphocytes can survive intensive conditioning for bone marrow transplantation.
Blood.
68
1986
954
956
132
Kedar
E
Or
R
Naparstek
E
Zeira
E
Slavin
S
Preliminary characterization of functional residual host-type T lymphocytes following conditioning for allogeneic HLA-matched bone marrow transplantation (BMT).
Bone Marrow Transplant.
3
1988
129
140
133
Wang
W
Meadows
LR
den Haan
JM
et al
Human H-Y: a male-specific histocompatibility antigen derived from the SMCY protein.
Science.
269
1995
1588
1590
134
Meadows
L
Wang
W
den Haan
JM
et al
The HLA-A*0201-restricted H-Y antigen contains a posttranslationally modified cysteine that significantly affects T cell recognition.
Immunity.
6
1997
273
281
135
Vogt
MH
de Paus
RA
Voogt
PJ
Willemze
R
Falkenburg
JH
DFFRY codes for a new human male-specific minor transplantation antigen involved in bone marrow graft rejection.
Blood.
95
2000
1100
1105
136
Vogt
MH
Goulmy
E
Kloosterboer
FM
et al
UTY gene codes for an HLA-B60-restricted human male-specific minor histocompatibility antigen involved in stem cell graft rejection: characterization of the critical polymorphic amino acid residues for T-cell recognition.
Blood.
96
2000
3126
3132
137
Cahn
JY
Herve
P
Flesch
M
et al
Marrow transplantation from HLA non-identical family donors for the treatment of leukaemia: a pilot study of 15 patients using additional immunosuppression and T-cell depletion.
Br J Haematol.
69
1988
345
349
138
Ash
RC
Casper
JT
Chitambar
CR
et al
Successful allogeneic transplantation of T-cell-depleted bone marrow from closely HLA-matched unrelated donors.
N Engl J Med.
322
1990
485
494
139
Aversa
F
Pelicci
PG
Terenzi
A
et al
Results of T-depleted BMT in chronic myelogenous leukaemia after a conditioning regimen that included thiotepa.
Bone Marrow Transplant.
7(suppl 2)
1991
24
140
Schaap
N
Schattenberg
A
Bar
B
et al
Outcome of transplantation for standard-risk leukaemia with grafts depleted of lymphocytes after conditioning with an intensified regimen.
Br J Haematol.
98
1997
750
759
141
Schattenberg
A
Schaap
N
Preijers
F
van der Maazen
R
de Witte
T
Outcome of T cell-depleted transplantation after conditioning with an intensified regimen in patients aged 50 years or more is comparable with that in younger patients.
Bone Marrow Transplant.
26
2000
17
22
142
Kurisu
K
Hishikawa
Y
Taniguchi
M
et al
Total lymphoid irradiation and total body irradiation for allogeneic bone marrow transplantation in aplastic anemia.
Radiat Med.
9
1991
148
152
143
Castro-Malaspina
H
Childs
B
Laver
J
et al
Hyperfractionated total lymphoid irradiation and cyclophosphamide for preparation of previously transfused patients undergoing HLA-identical marrow transplantation for severe aplastic anemia.
Int J Radiat Oncol Biol Phys.
29
1994
847
854
144
Zapatero
A
Marin
A
Lopez
M
et al
Successful bone marrow transplantation in sensitized aplastic anemia patients using total lymphoid irradiation for conditioning: long-term follow-up.
Hematol Oncol.
14
1996
165
172
145
Soiffer
RJ
Mauch
P
Tarbell
NJ
et al
Total lymphoid irradiation to prevent graft rejection in recipients of HLA non-identical T cell-depleted allogeneic marrow.
Bone Marrow Transplant.
7
1991
23
33
146
Soiffer
RJ
Weller
E
Alyea
EP
et al
CD6+ donor marrow T-cell depletion as the sole form of graft-versus-host disease prophylaxis in patients undergoing allogeneic bone marrow transplant from unrelated donors.
J Clin Oncol.
19
2001
1152
1159
147
Ganem
G
Kuentz
M
Beaujean
F
et al
Additional total lymphoid irradiation in preventing graft failure of T-cell depleted bone marrow transplantation from HLA-identical siblings.
Transplantation.
45
1987
244
248
148
Cobbold
SP
Martin
G
Qin
S
Waldmann
H
Monoclonal antibodies to promote marrow engraftment and tissue graft tolerance.
Nature.
323
1986
164
166
149
Cobbold
S
Martin
G
Waldmann
H
Monoclonal antibodies for the prevention of graft-versus-host disease and marrow graft rejection: the depletion of T cell subsets in vitro and in vivo.
Transplantation.
42
1986
239
247
150
Soiffer
RJ
Ritz
J
Selective T cell depletion of donor allogeneic marrow with anti-CD6 monoclonal antibody: rationale and results.
Bone Marrow Transplant.
12(suppl 3)
1993
S7
10
151
Soiffer
RJ
Freedman
AS
Neuberg
D
et al
CD6+ T cell-depleted allogeneic bone marrow transplantation for non-Hodgkin's lymphoma.
Bone Marrow Transplant.
21
1998
1177
1181
152
Kawanishi
Y
Passweg
J
Drobyski
WR
et al
Effect of T cell subset dose on outcome of T cell-depleted bone marrow transplantation.
Bone Marrow Transplant.
19
1997
1069
1077
153
Poynton
CH
Whittaker
JA
Bailey-Wood
R
et al
Mismatched family and unrelated donors for bone marrow transplantation using fixed low numbers of T cells.
Bone Marrow Transplant.
3(suppl 1)
1988
223
154
Potter
MN
Pamphilon
DH
Cornish
JM
Oakhill
A
Graft-versus-host disease in children receiving HLA-identical allogeneic bone marrow transplants with a low adjusted T lymphocyte dose.
Bone Marrow Transplant.
8
1991
357
361
155
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
1273
156
Witherspoon
RP
Lum
LG
Storb
R
Immunologic reconstitution after marrow grafting.
Sem Hematol.
21
1984
2
10
157
Mori
T
Tsoi
MS
Gillis
S
Santos
E
Thomas
ED
Storb
R
Cellular interactions in marrow-grafted patients; I: impairment of cell-mediated lympholysis associated with graft-vs-host disease and the effect of interleukin 2.
J Immunol.
130
1983
712
716
158
Brkic
S
Tsoi
MS
Mori
T
et al
Cellular interactions in marrow-grafted patients; III: normal interleukin-1 and defective interleukin-2 production in short-term patients and in those with chronic graft-versus-host disease.
Transplantation.
39
1985
30
35
159
Witherspoon
RP
Storb
R
Ochs
HD
et al
Recovery of antibody production in allogeneic marrow graft recipients: influence of time post transplant, the presence or absence of chronic graft versus host disease, and anti-thymocyte globulin treatment.
Blood.
58
1981
360
368
160
Lum
LG
Seigneuret
MC
Storb
R
et al
In vitro regulation of immunoglobulin synthesis after marrow transplantation, I: T and B cell deficiencies in patients with and without chronic graft versus host disease.
Blood.
58
1981
431
439
161
Witherspoon
RP
Lum
LG
Storb
R
Thomas
ED
In vitro regulation of immunoglobulin synthesis after human marrow transplantation; II: deficient T and non-T lymphocyte function within 3-4 months of allogeneic, syngeneic, or autologous marrow grafting for hematologic malignancy.
Blood.
59
1982
844
850
162
Korsmeyer
SJ
Elfenbein
GJ
Goldman
CK
Marshall
SL
Santos
GW
Waldmann
TA
B cell, helper T cell, and suppressor T cell abnormalities contribute to disordered immunoglobulin synthesis in patients following bone marrow transplantation.
Transplantation.
33
1982
184
190
163
Welte
K
Keever
CA
Levick
J
et al
Interleukin-2 production and response to interleukin-2 by peripheral blood mononuclear cells from patients after bone marrow transplantation; II: patients receiving soybean lectin-separated and T cell-depleted bone marrow.
Blood.
70
1987
1595
1603
164
Zander
AR
Reuben
JM
Johnston
D
et al
Immune recovery following allogeneic bone marrow transplantation.
Transplantation.
40
1985
177
183
165
Soiffer
RJ
Bosserman
L
Murray
C
Cochran
K
Daley
J
Ritz
J
Reconstitution of T-cell function after CD6-depleted allogeneic bone marrow transplantation.
Blood.
75
1990
2076
2084
166
Pignata
C
Sanghera
JS
Soiffer
RJ
et al
Defective activation of mitogen-activated protein kinase after allogeneic bone marrow transplantation.
Blood.
88
1996
2334
2341
167
Mackall
CL
Granger
L
Sheard
MA
Cepeda
R
Gress
RE
T-cell regeneration after bone marrow transplantation: differential CD45 isoform expression on thymic-derived versus thymic-independent progeny.
Blood.
82
1993
2585
2594
168
Mackall
CL
Fleisher
TA
Brown
MR
et al
Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy.
N Engl J Med.
332
1995
143
149
169
Pignata
C
Gaetaniello
L
Masci
AM
et al
Human equivalent of the mouse Nude/SCID phenotype: long-term evaluation of immunologic reconstitution after bone marrow transplantation.
Blood.
97
2001
880
885
170
Keever
CA
Small
TN
Flomenberg
N
et al
Immune reconstitution following bone marrow transplantation: comparison of recipients of T-cell depleted marrow with recipients of conventional marrow grafts.
Blood.
73
1989
1340
1350
171
Ault
KA
Antin
JH
Ginsburg
D
et al
Phenotype of recovering lymphoid cell populations after marrow transplantation.
J Exp Med.
161
1985
1483
1502
172
Parreira
A
Smith
J
Hows
JM
et al
Immunological reconstitution after bone marrow transplant with Campath-1 treated bone marrow.
Clin Exp Immunol.
67
1987
142
150
173
Roux
E
Helg
C
Dumont-Girard
F
Chapuis
B
Jeannet
M
Roosnek
E
Analysis of T-cell repopulation after allogeneic bone marrow transplantation: significant differences between recipients of T-cell depleted and unmanipulated grafts.
Blood.
87
1996
3984
3992
174
Roux
E
Dumont-Girard
F
Starobinski
M
et al
Recovery of immune reactivity after T-cell-depleted bone marrow transplantation depends on thymic activity.
Blood.
96
2000
2299
2303
175
Wu
CJ
Chillemi
A
Alyea
EP
et al
Reconstitution of T-cell receptor repertoire diversity following T-cell depleted allogeneic bone marrow transplantation is related to hematopoietic chimerism.
Blood.
95
2000
352
359
176
Zutter
MM
Martin
PJ
Sale
GE
et al
Epstein-Barr virus lymphoproliferation after bone marrow transplantation.
Blood.
72
1988
520
529
177
Shapiro
RS
McClain
K
Frizzera
G
et al
Epstein-Barr virus associated B cell lymphoproliferative disorders following bone marrow transplantation.
Blood.
71
1988
1234
1243
178
Gerritsen
EJ
Stam
ED
Hermans
J
et al
Risk factors for developing EBV-related B cell lymphoproliferative disorders (BLPD) after non-HLA-identical BMT in children.
Bone Marrow Transplant.
18
1996
377
382
179
Martin
P
Shulman
H
Schubach
W
et al
Fatal Epstein-Barr virus associated proliferation of donor B-cells after treatment of acute graft-versus-host disease with a murine anti-T-cell antibody.
Ann Intern Med.
101
1984
310
315
180
Hanto
DW
Najarian
JS
Advances in the diagnosis and treatment of EBV-associated lymphoproliferative diseases in immunocompromised hosts.
J Surg Oncol.
30
1985
215
220
181
Fischer
A
Blanche
S
Le Bidois
J
et al
Anti-B-cell monoclonal antibodies in the treatment of severe B-cell lymphoproliferative syndrome following bone marrow and organ transplantation.
N Engl J Med.
324
1991
1451
1456
182
Papadapoulos
EB
Ladanyi
M
Emmanuel
D
et al
Infusions of donor leukocytes to treat Epstein-Barr-associated lymphoproliferative disorders after allogeneic bone marrow transplantation.
N Engl J Med.
330
1994
1185
1191
183
Heslop
HE
Brenner
MK
Rooney
C
et al
Administration of neomycin-resistance-gene-marked EBV-specific cytotoxic T lymphocytes to recipients of mismatched-related or phenotypically similar unrelated donor marrow grafts.
Hum Gene Ther.
5
1994
381
397
184
Rooney
CM
Smith
CA
Ng
CY
et al
Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr-virus-related lymphoproliferation.
Lancet.
345
1995
9
13
185
Rooney
CM
Smith
CA
Ng
CY
et al
Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients.
Blood.
92
1998
1549
1555
186
Gustafsson
A
Levitsky
V
Zou
JZ
et al
Epstein-Barr virus (EBV) load in bone marrow transplant recipients at risk to develop posttransplant lymphoproliferative disease: prophylactic infusion of EBV-specific cytotoxic T cells.
Blood.
95
2000
807
814
187
McGuirk
JP
Seropian
S
Howe
G
Smith
B
Stoddart
L
Cooper
DL
Use of rituximab and irradiated donor-derived lymphocytes to control Epstein-Barr virus-associated lymphoproliferation in patients undergoing related haplo-identical stem cell transplantation.
Bone Marrow Transplant.
24
1999
1253
1258
188
Kuehnle
I
Huls
MH
Liu
Z
et al
CD20 monoclonal antibody (rituximab) for therapy of Epstein-Barr virus lymphoma after hemopoietic stem-cell transplantation.
Blood.
95
2000
1502
1505
189
Rooney
CM
Loftin
SK
Holladay
MS
Brenner
MK
Krance
RA
Heslop
HE
Early identification of Epstein-Barr virus-associated post-transplantation lymphoproliferative disease.
Br J Haematol.
89
1995
98
103
190
Cavazzana-Calvo
M
Bensoussan
D
Jabado
N
et al
Prevention of EBV-induced B-lymphoproliferative disorder by ex vivo marrow B-cell depletion in HLA-phenoidentical or non-identical T-depleted bone marrow transplantation.
Br J Haematol.
103
1998
543
551
191
Hale
G
Waldmann
H
Risks of developing Epstein-Barr virus-related lymphoproliferative disorders after T-cell-depleted marrow transplants: CAMPATH users.
Blood.
91
1998
3079
3083
192
Goldman
JM
Gale
RP
Horowitz
MM
et al
Bone marrow transplantation for chronic myelogenous leukemia in chronic phase: increased risk for relapse associated with T-cell depletion.
Ann Intern Med.
108
1988
806
814
193
Martin
P
Clift
RA
Fisher
LD
et al
HLA-identical marrow transplantation during accelerated-phase chronic myelogenous leukemia: analysis of survival and remission duration.
Blood.
72
1988
1978
1984
194
Marks
DI
Hughes
TP
Szydlo
R
et al
HLA-identical sibling donor bone marrow transplantation for chronic myeloid leukaemia in first chronic phase: influence of GVHD prophylaxis on outcome.
Br J Haematol.
81
1992
383
390
195
Wagner
JE
Zahurak
M
Piantadosi
S
et al
Bone marrow transplantation of chronic myelogenous leukemia in chronic phase: evaluation of risks and benefits.
J Clin Oncol.
10
1992
779
789
196
Gratwohl
A
Hermans
J
Niderwieser
D
et al
Bone marrow transplantation for chronic myeloid leukemia: long-term results.
Bone Marrow Transplantation.
12
1993
509
516
197
Weiden
PL
Flournoy
N
Thomas
ED
Prentice
R
Buckner
CD
Storb
R
Antileukemic effect of graft-versus-host disease in recipients of allogeneic-marrow grafts.
N Engl J Med.
300
1979
1068
1073
198
Sullivan
KM
Weiden
PL
Storb
R
et al
Influence of acute and chronic graft-versus-host disease on relapse and survival after bone marrow transplantation from HLA-identical siblings as treatment of acute and chronic leukemia.
Blood.
73
1989
1720
1728
199
Enright
H
Davies
SM
DeFor
T
et al
Relapse after non-T-cell-depleted allogeneic bone marrow transplantation for chronic myelogenous leukemia: early transplantation, use of an unrelated donor, and chronic graft-versus-host disease are protective.
Blood.
88
1996
714
720
200
Kolb
HJ
Schattenberg
A
Goldman
JM
et al
Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients.
Blood.
86
1995
2041
2050
201
Collins
R
Shpilberg
O
Drobyski
W
et al
Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation.
J Clin Oncol.
15
1997
433
444
202
McGlave
P
Bartsch
G
Anasetti
C
et al
Unrelated donor marrow transplantation therapy for chronic myelogenous leukemia: initial experience of the National Marrow Donor Program.
Blood.
81
1993
543
550
203
Drobyski
WR
Ash
RC
Casper
JT
et al
Effect of T-cell depletion as graft-versus-host disease prophylaxis on engraftment, relapse, and disease-free survival in unrelated marrow transplantation for chronic myelogenous leukemia.
Blood.
83
1994
1980
1987
204
Hessner
MJ
Endean
DJ
Casper
JT
et al
Use of unrelated marrow grafts compensates for reduced graft-versus-leukemia reactivity after T-cell-depleted allogeneic marrow transplantation for chronic myelogenous leukemia.
Blood.
86
1995
3987
3996
205
Young
JW
Papadopoulos
EB
Cunningham
I
et al
T-cell-depleted allogeneic bone marrow transplantation in adults with acute nonlymphocytic leukemia in first remission.
Blood.
79
1992
3380
3387
206
Bunjes
D
Hertenstein
B
Wiesneth
M
et al
In vivo/ex vivo T cell depletion reduces the morbidity of allogeneic bone marrow transplantation in patients with acute leukaemias in first remission without increasing the risk of treatment failure: comparison with cyclosporin/methotrexate.
Bone Marrow Transplant.
5
1995
563
568
207
Aversa
F
Terenzi
A
Carotti
A
et al
Improved outcome with T-cell-depleted bone marrow transplantation for acute leukemia.
J Clin Oncol.
17
1999
1545
1550
208
Remberger
M
Ringden
O
Aschan
J
Ljungman
P
Lonnquist
B
Markling
L
Long-term follow-up of a randomized trial comparing T-cell depletion with a combination of methotrexate and cyclosporine in adult leukemic marrow transplant recipients.
Transplant Proc.
26
1994
1829
1830
209
Appelbaum
FR
Sullivan
KM
Buckner
CD
et al
Treatment of malignant lymphoma in 100 patients with chemotherapy, total body irradiation, and marrow transplantation.
J Clin Oncol.
5
1987
1340
1347
210
Shepherd
JD
Barnett
MJ
Connors
JM
et al
Allogeneic bone marrow transplantation for poor-prognosis non- Hodgkin's lymphoma.
Bone Marrow Transplant.
12
1993
591
596
211
Jones
RJ
Ambinder
RF
Piantadosi
S
Santos
GW
Evidence of a graft-versus-lymphoma effect associated with allogeneic bone marrow transplantation.
Blood.
77
1991
649
653
212
Chopra
R
Goldstone
AH
Pearce
R
et al
Autologous versus allogeneic bone marrow transplantation for non- Hodgkin's lymphoma: a case-controlled analysis of the European Bone Marrow Transplant Group Registry data.
J Clin Oncol.
10
1992
1690
1695
213
Ratanatharathorn
V
Uberti
J
Karanes
C
et al
Prospective comparative trial of autologous versus allogeneic bone marrow transplantation in patients with non-Hodgkin's lymphoma.
Blood.
84
1994
1050
1055
214
Bjorkstrand
B
Ljungman
P
Svensson
H
et al
Allogeneic bone marrow transplantation versus autologous stem cell transplantation in multiple myeloma: a retrospective case-matched study from the European Group for Blood and Marrow Transplantation.
Blood.
88
1996
4711
4718
215
Anderson
K
Andersen
J
Soiffer
R
et al
Monoclonal antibody-purged bone marrow transplantation therapy for multiple myeloma.
Blood.
82
1993
2568
2576
216
Sykes
M
Dissociating graft-vs-host disease from the graft-vs-leukemia effect of allogeneic T cells: the potential role of IL-2.
Bone Marrow Transplant.
10(suppl 1)
1992
1
4
217
Slavin
S
Or
R
Prighozina
T
et al
Immunotherapy of hematologic malignancies and metastatic solid tumors in experimental animals and man.
Bone Marrow Transplant.
25(suppl 2)
2000
S54
57
218
Korngold
R
Sprent
J
T cell subsets and graft versus host disease.
Transplantation.
44
1987
335
339
219
Nimer
SD
Giorgi
J
Gajewski
JL
et al
Selective depletion of CD8+ cells for prevention of graft-versus-host disease after bone marrow transplantation: a randomized controlled trial.
Transplantation.
57
1994
82
87
220
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.
86
1995
4337
4343
221
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.
91
1998
3671
3680
222
Baker
J
Verneris
MR
Ito
M
et al
Expansion of cytolytic CD8(+) natural killer T cells with limited capacity for graft-versus-host disease induction due to interferon gamma production.
Blood.
97
2001
2923
2931
223
Mackinnon
S
Hows
JM
Goldman
JM
Induction of in vitro graft-versus-leukemia activity following bone marrow transplantation for chronic myeloid leukemia.
Blood.
76
1990
2037
2045
224
Hauch
M
Gazzola
MV
Small
T
et al
Anti-leukemia potential of interleukin-2 activated natural killer cells after bone marrow transplantation for chronic myelogenous leukemia.
Blood.
75
1990
2250
2262
225
Ruggeri
L
Capanni
M
Casucci
M
et al
Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation.
Blood.
94
1999
333
339
226
Soiffer
RJ
Murray
C
Cochran
K
et al
Clinical and immunologic effects of prolonged infusion of low-dose recombinant interleukin-2 after autologous and T-cell-depleted allogeneic bone marrow transplantation.
Blood.
79
1992
517
526
227
Soiffer
RJ
Murray
C
Gonin
R
Ritz
J
Effect of low-dose interleukin-2 on disease relapse after T-cell-depleted allogeneic bone marrow transplantation.
Blood.
84
1994
964
971
228
Koh
MB
Prentice
HG
Lowdell
MW
Selective removal of alloreactive cells from haematopoietic stem cell grafts: graft engineering for GVHD prophylaxis.
Bone Marrow Transplant.
23
1999
1071
1079
229
Fehse
B
Goldmann
M
Frerk
O
Bulduk
M
Zander
AR
Depletion of alloreactive donor T cells using immunomagnetic cell selection.
Bone Marrow Transplant.
26(suppl 2)
2000
S39
42
230
Blazar
BR
Taylor
PA
Linsley
PS
Vallera
DA
In vivo blockade of CD28/CTLA4: B7/BB1 interaction with CTLA4-Ig reduces lethal murine graft-versus-host disease across the major histocompatibility complex barrier in mice.
Blood.
83
1994
3815
3825
231
Blazar
BR
Taylor
PA
Panoskaltsis-Mortari
A
Gray
GS
Vallera
DA
Co-blockade of the LFA1:ICAM and CD28/CTLA4:B7 pathways is a highly effective means of preventing acute lethal graft-versus-host disease induced by fully major histocompatibility complex-disparate donor grafts.
Blood.
85
1995
2607
2618
232
Guinan
EC
Gribben
JG
Boussiotis
VA
Freeman
GJ
Nadler
LM
Pivotal role of the B7:CD28 pathway in transplantation tolerance and tumor immunity.
Blood.
84
1994
3261
3282
233
Blazar
BR
Taylor
PA
Panoskaltsis-Mortari
A
et al
Blockade of CD40 ligand-CD40 interaction impairs CD4+ T cell-mediated alloreactivity by inhibiting mature donor T cell expansion and function after bone marrow transplantation.
J Immunol.
158
1997
29
39
234
Guinan
EC
Boussiotis
VA
Neuberg
D
et al
Transplantation of anergic histoincompatible bone marrow allografts.
N Engl J Med.
340
1999
1704
1714
235
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.
98
2001
934
939
236
Slavin
S
Nagler
A
Naparstek
E
et al
Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases.
Blood.
91
1998
756
763
237
Mackinnon
S
Papadapoulos
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.
86
1995
1261
1268
238
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.
86
1995
4337
4343
239
Munshi
NC
Govindarajan
R
Drake
R
et al
Thymidine kinase (TK) gene-transduced human lymphocytes can be highly purified, remain fully functional, and are killed efficiently with ganciclovir.
Blood.
89
1997
1334
1340
240
Link
CJ
Burt
RK
Traynor
AE
et al
Adoptive immunotherapy for leukemia: donor lymphocytes transduced with the herpes simplex thymidine kinase gene for remission induction: HGTRI 0103.
Hum Gene Ther.
9
1998
115
134
241
Tiberghien
P
Ferrand
C
Lioure
B
et al
Administration of herpes simplex-thymidine kinase-expressing donor T cells with a T-cell-depleted allogeneic marrow graft.
Blood.
97
2001
63
72
242
Aubin
F
Cahn
JY
Ferrand
C
Angonin
R
Humbert
P
Tiberghien
P
Extensive vitiligo after ganciclovir treatment of GvHD in a patient who had received donor T cells expressing herpes simplex virus thymidine kinase.
Lancet.
355
2000
626
627
243
Molldrem
JJ
Lee
PP
Wang
C
et al
Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia.
Nat Med.
6
2000
1018
1023
244
Soiffer RJ, Alyea EP, Canning C, et al. Effects of donor lymphocyte infusions (DLI) on prevention of disease relapse after T-cell depleted (TCD) allogeneic bone marrow transplantation (BMT) [abstract]. Blood. In press.

Author notes

Robert J. Soiffer, Dana-Farber Cancer Institute, 44 Binney St, D1B58, Boston, MA 02115; e-mail:robert_soiffer@dfci.harvard.edu.

Sign in via your Institution