The appropriate timing of bone marrow transplantation (BMT) for adults with acute myelogenous leukemia (AML) and acute lymphoblastic leukemia (ALL) is controversial. Although allogeneic transplantation results in a lower risk of disease recurrence than intensive chemotherapy alone, overall outcome following BMT may not be improved due to the higher incidence of therapy-related fatal complications, frequently as a result of the development of graft-versus-host disease (GVHD). Selective T-cell depletion of donor marrow can reduce the incidence of GVHD and thereby limit transplant-related toxicity. Herein we report the risk of GVHD, incidence of transplant related mortality (TRM), likelihood of disease relapse, and overall survival in adult patients undergoing BMT with CD6 depleted allogeneic marrow for acute leukemia in first remission. Forty-one consecutive allogeneic transplants were performed on patients with acute leukemia and high-risk features (28 AML, 13 ALL) using T12 monoclonal antibody and complement to remove CD6+ T cells from donor marrow. No pre- or posttransplant immune suppressive medications for GVHD prophylaxis were administered. The actuarial estimated risk of grade 2 to 4 acute GVHD was 15% in patients receiving HLA identical grafts. Chronic GVHD developed in five patients. The estimated risk of TRM for patients in first complete remission was 5% at Day +100 and 16% at 2 years. Fatalities attributable to infection with cytomegalovirus or Epstein-Barr virus occurred in only three patients. Estimated probabilities of relapse, overall survival, and event-free survival at 4 years were 25%, 71%, and 63%, respectively. No significant differences in GVHD, TRM, relapse rate, or survival was observed for patients with AML compared with those with ALL. Allogeneic transplantation with CD6 depleted bone marrow is effective in consolidating remissions of high-risk patients with acute leukemia in first remission without excessive toxicity.

OVER THE PAST 20 YEARS, bone marrow transplantation (BMT) has been used with increasing frequency in the treatment of patients with acute leukemia.1,2 Reports from many centers indicate that adults undergoing allogeneic BMT for acute myelogenous leukemia (AML) or acute lymphoblastic leukemia (ALL) have a lower risk of relapse than individuals treated with intensive chemotherapy alone.3-6 However, since the toxicities associated with BMT are significant and represent the primary cause of treatment failure for patients with leukemia transplanted in first remission, there is considerable controversy over the appropriate timing of transplantation.7 

Allogeneic BMT would be a far more attractive treatment modality for patients with acute leukemia in first remission if the risks associated with the procedure could be limited. Transplant-related mortality often comes as a direct or indirect result of the development of graft-versus-host disease (GVHD).8-10 GVHD is initiated, in large part, by T lymphocytes in the donor marrow.11 Decreasing the number of donor marrow T cells by a variety of purging methods will decrease the incidence of GVHD.12-15 However, the overall utility of T-cell depletion has been questioned because of reports of higher rates of relapse, graft failure, and Epstein-Barr virus (EBV) related lymphoproliferative disorders post-BMT.16-20 Cellular populations lost in many T-cell depletion procedures appear to play critical roles in promoting donor marrow engraftment, suppressing outgrowth of EBV-transformed lymphocytes, and eliminating residual disease post-BMT.

In vitro purging methods are not uniform, and the precise extent of T cells removed by different techniques vary.21,22 Some depletion approaches eliminate other potentially important cell types as well, such as natural killer (NK) cells, B lymphocytes, monocytes, and myeloid progenitors. We have used T12, an IgM monoclonal antibody (MoAb) that recognizes the CD6 antigen on T cells, to purge donor marrow. T12 reacts with 90% to 95% of mature T cells, but does not affect immature thymocytes, NK cells, monocytes, or other hematopoietic elements.23 Three cycles of incubation with T12 plus rabbit complement removes approximately 1.5 to 2.0 logs of T lymphocytes from normal bone marrow.24 We have previously demonstrated that depletion of CD6+ T cells from donor marrow with T12 and complement can, without the addition of any immune suppressive agents, significantly reduce the incidence of grades 2 to 4 acute GVHD in patients transplanted for hematologic malignancy.25 We now report results of CD6 depletion in BMTs performed for acute leukemia in first remission at our institution through April 1995.

Patient population.Forty-one consecutive adults with a diagnosis of AML or ALL who underwent BMT at our institution between March 1984 and April 1995 are included in this analysis. Results are analyzed as of April 1, 1996. Since allogeneic transplantation had not been included in the routine treatment of patients entered onto cooperative group or institutional studies at our institution, these patients represent a minority of the individuals presenting to Dana-Farber since we initiated our transplant program. These patients were selected for transplantation because they were felt to be at high risk for relapse following intensive chemotherapy based on their clinical characteristics including adverse karyotypic anomalies, FAB classification, antecedent myelodysplastic syndrome, and slow response to induction chemotherapy. Inpatients were cared for at the Brigham and Women's Hospital from 1985 to 1987 (n = 6) and at Dana-Farber Cancer Institute from 1987 to 1995 (n = 35). Treatment protocols were approved by the Human Subjects Protection Committees of DFCI and BWH. Written informed consent was obtained in all cases.

Treatment protocol.Forty of the 41 patients received cyclophosphamide 60 mg/kg intravenously (IV) × 2 on 2 consecutive days followed by total body irradiation (TBI) as ablation. The dose of TBI was sequentially escalated from 1,300 cGy in 1985 to 1,560 cGy in 1995 (Table 1). One patient who had previously received TBI as part of a conditioning regimen for an autologous transplant for non-Hodgkin's lymphoma received busulfan (1 mg/kg × 16 doses) instead of TBI. In all patients who received TBI, the total dose was administered in six to eight equal fractions administered twice daily, 5 to 6 hours apart. All patients were treated at 10 cGy/min on a dedicated linear accelerator. Patients who received marrow from HLA mismatched donors (n = 7) also received total lymphoid irradiation (TLI) at 750 to 1,050 cGy 1 week before admission as previously described to prevent graft rejection.26 

Table 1.

Patient Characteristics

VariableN
Diagnosis 
AML 28 
ALL 13 
Patient/donor sex 
Male/male 
Male/female 13 
Female/female 
Female/male 12 
Age 
Patient 34 (22-54) 
Donor 34 (16-51) 
HLA disparity 
Matched 34 
Mismatched 
1 antigen 
2 antigens 
Ablative regimen 
Cyclophosphamide/TBI (1,300 cGy) 
Cyclophosphamide/TBI (1,400 cGy) 26 
Cyclophosphamide/TBI (1,480 cGy) 
Cyclophosphamide/TBI (1,560 cGy) 
Cyclophosphamide/Busulfan 
Cell dose 
Median BM cells infused 5.6 × 107 cells/kg 
Median CD3+ BM cells infused 1.5 × 106 cells/kg 
VariableN
Diagnosis 
AML 28 
ALL 13 
Patient/donor sex 
Male/male 
Male/female 13 
Female/female 
Female/male 12 
Age 
Patient 34 (22-54) 
Donor 34 (16-51) 
HLA disparity 
Matched 34 
Mismatched 
1 antigen 
2 antigens 
Ablative regimen 
Cyclophosphamide/TBI (1,300 cGy) 
Cyclophosphamide/TBI (1,400 cGy) 26 
Cyclophosphamide/TBI (1,480 cGy) 
Cyclophosphamide/TBI (1,560 cGy) 
Cyclophosphamide/Busulfan 
Cell dose 
Median BM cells infused 5.6 × 107 cells/kg 
Median CD3+ BM cells infused 1.5 × 106 cells/kg 

Donor bone marrow was harvested on the last day of TBI administration. CD6+ T cells were removed from donor marrow by complement-mediated antibody lysis using anti-T12 MoAb as previously described.25 Marrow was infused into the patient through an indwelling central venous catheter the same day it was harvested. No patients received prophylactic immunosuppressive therapy of any type, including corticosteroids, methotrexate, or cyclosporine. Since January 1994, six patients received granulocyte colony stimulating factor (G-CSF ) beginning at day + 1 and continuing until the absolute neutrophil count exceeded 1.0 × 109 cells/L.

All patients were treated in HEPA-filtered rooms using standard reverse isolation procedures. Oral prophylactic antibiotics, either ciprofloxacin or trimethoprim-sulfamethoxazole, were administered to all patients. These were discontinued when patients developed fevers that required initiation of broad spectrum IV antibiotics. Since 1987, all patients have received prophylactic acyclovir. All patients received single donor platelets from cytomegalovirus sero-negative individuals and frozen deglycerolized red blood cells. All blood components were irradiated to prevent transfusion-related GVHD. After BMT, three of the 41 patients participated on trials in which they received low-dose interleukin-2 (IL-2; Hoffmann-LaRoche [Nutley, NJ] or Amgen, Inc) administered either IV by continuous infusion or by subcutaneous bolus at doses of 2 to 6 × 105 U/m2/d for up to 90 days.27 

Immunofluorescence assays.Bone marrow and peripheral blood mononuclear cells (PBMC) were isolated using Ficoll-Hypaque density centrifugation and were analyzed by direct or indirect immunofluorescence for reactivity with a series of MoAbs using standard techniques. Antibodies used included T3 (CD3), T4 (CD4), T8 (CD8), T11 (CD2), T12 (CD6), NKH1 (CD56), B1 (CD20), and My4 (CD14) (Coulter Immunology, Hialeah, FL). Ten thousand cells were analyzed in each sample using automated flow cytometry (EPICS-C; Coulter Electronics, Hialeah, FL). Bone marrow cells were analyzed before and after in vitro treatment with anti-T12 plus complement. PBMC were analyzed at regular intervals post-BMT.

Statistical analysis.Descriptive statistics are reported as proportions, medians, and means. Fisher's exact test was used to compare proportions.28 The duration of survival, event-free survival, time to relapse, GVHD, and transplant-related mortality were computed from the date of marrow infusion. GVHD was graded by standard criteria.29 Patients were considered censored at the time of death for “time to relapse” and censored at the time of relapse for “time to death without relapse” (transplant-related mortality). Survival, event-free survival, time to relapse (risk), and transplant-related mortality curves were constructed using the Kaplan-Meier product limit method.30 The logrank test of survival analysis was used to compare the various subgroups with respect to their survival, event-free survival, time to relapse, and time to death without relapse (transplant mortality) distributions.31 Fisher's exact test and Wilcoxon rank sum tests were used to identify possible predictors of GVHD. Predictors of treatment-related mortality, relapse, and event-free survival and overall survival were examined using the logrank test.

Patient characteristics.Twenty-eight patients had a diagnosis of AML and 13 patients had ALL (Table 1). The median age of patients was 34 years (range, 22 to 54 years). There were 22 males and 19 females. The majority of patients received 1,400 cGy TBI in seven fractions over 4 days. Thirty-four received marrow from sibling donors who were genotypically matched while seven received marrow from HLA-mismatched related donors. The average age of donors was 34 years (16 to 51 years). Patients were sex matched with their donors in 16 cases and mismatched in 25 cases. Of the 20 females donors, 12 were multiparous, 5 nulliparous, and 3 unknown. The median time from diagnosis to transplant for patients grafted in first remission was 4.5 months (range, 2 to 12 months).

The majority of patients reported possessed adverse clinical features that prompted the decision to have them undergo BMT. Cytogenetics conveying a poor prognosis for patients treated with chemotherapy alone were present in 19 patients (Table 2). Normal karyotypes were documented in 11 patients, and two patients had favorable cytogenetics. In nine patients, no cytogenetic data were available. Six patients transplanted for AML had a history of antecedent myelodysplasia. An additional three patients (1 AML, 2 ALL) had been treated with chemotherapy for a prior malignancy. Five of the 28 patients with AML had histologic features of M5, M6, or M7 leukemia at diagnosis. Five patients with AML had initially failed induction with two courses of anthracycline and cytosine arabinoside and had achieved complete remission only after treatment with high dose ara-C. Four patients with ALL required at least 8 weeks to achieve a complete remission and five ALL patients presented with a WBC count in excess of 50,000 × 106/L.

Table 2.

Cytogenetics at Diagnosis (CR1)

DiseaseKaryotypeN
 AML Unfavorable 11 
 Trisomy 8 
 Multiple abnormalities 
 Monosomy 7 
 t(9; 11) 
 t(9; 22) 
 Normal 
 Favorable (inv 16) 
 Unknown 
 ALL Unfavorable 
 t(9; 22) 
 Multiple abnormalities 
 t(4; 11) 
 t(8; 14) 
 Normal 
 Unknown 
DiseaseKaryotypeN
 AML Unfavorable 11 
 Trisomy 8 
 Multiple abnormalities 
 Monosomy 7 
 t(9; 11) 
 t(9; 22) 
 Normal 
 Favorable (inv 16) 
 Unknown 
 ALL Unfavorable 
 t(9; 22) 
 Multiple abnormalities 
 t(4; 11) 
 t(8; 14) 
 Normal 
 Unknown 

Marrow infusion.The median number of nucleated marrow cells infused into patients was 4.0 × 109 (1.4 to 12.7 × 109) which was the equivalent of 5.6 × 107 cells/kg (2.0 to 25.4 × 107). Immunophenotypic analysis was performed on infused marrow following CD6 depletion. The median number of CD3 cells infused was 1.5 × 106 cells/kg. In the majority of patients, no cells expressing CD6 could be detected in the infused marrow by immunophenotypic analysis.

Engraftment.Forty of 41 patients demonstrated successful initial engraftment (>1.0 × 109 WBC/L) within 30 days of marrow infusion. The patient who failed to engraft had received a one-antigen HLA mismatched marrow. This patient was successfully engrafted with previously cryopreserved autologous bone marrow. The median time to achieve a neutrophil count of 0.5 × 109 cells/L was 19 days (11 to 30 d). Multivariable analysis of factors influencing neutrophil engraftment (including patient age, donor age, sex mismatch, number of cells infused/kg, number of CD3+ cells infused/kg, TBI dose, use of G-CSF ) revealed that only use of G-CSF correlated with speed of engraftment (logrank, P = .003). Platelet counts of 50,000 × 106/L were achieved at a median of 25 days (range, 14 to 55 days) post-BMT.

Late graft failure, defined as a drop in absolute neutrophil count to a level <0.1 × 109 cells/L beyond day +30, was noted in two patients who had no evidence of GVHD. It was recognized 54 to 76 days after initial marrow infusion. An active cellular rejection process could not be demonstrated. Both patients were successfully engrafted following reconditioning with cyclophosphamide (30 mg/kg × 4 days), antithymocyte globulin (30 mg/kg for 3 days), and infusion of unmanipulated marrow from the original donor. One patient developed GVHD despite prophylaxis with cyclosporine and corticosteroids.

GVHD.Grade 2 to 4 GVHD occurred in nine patients. It was scored as grade 2 in eight, and grade 3 in one (Fig 1A). Eight of nine patients had cutaneous involvement only. The onset of acute GVHD occurred at a median of day +26. The estimated probability of developing grades 2 to 4 GVHD was 15% in recipients of genotypically identical marrow compared with 42% for those receiving genotypically nonidentical marrow (Fisher's exact, P = .01). Univariate analysis was performed evaluating risk factors including HLA genotypic identity, patient/donor age, patient/donor sex, number of nucleated cells infused (total, CD3+, CD56+, CD6+, CD4+, CD8+), TBI dose, diagnosis, disease stage, and use of G-CSF revealed that only the use of nongenotypically HLA identical donors could predict for the development of acute GVHD. Chronic GVHD was observed in five patients, with an estimated probability for those at risk of 14% (Fig 1B). Chronic GVHD arose in the setting of prior acute GVHD in all cases; no patients developed de novo chronic GVHD. Posttransplant treatment with IL-2 did not induce acute or chronic GVHD in any patients.

Fig. 1.

Incidence of acute and chronic GVHD after CD6 depleted allo-BMT. The estimated probability of developing grades 2 to 4 and 3 to 4 acute GVHD is depicted in (A). The incidence of chronic GVHD over time is shown in (B).

Fig. 1.

Incidence of acute and chronic GVHD after CD6 depleted allo-BMT. The estimated probability of developing grades 2 to 4 and 3 to 4 acute GVHD is depicted in (A). The incidence of chronic GVHD over time is shown in (B).

Close modal

Transplant-related complications.Six patients died as a result of transplant-related complications. Interstitial pneumonitis was the most common direct cause of toxic death, occurring in five patients. In two instances, pneumonitis appeared to be caused by CMV, in one case by toxoplasmosis, and in two cases no organisms could be identified. Fatal EBV lymphoproliferative disease occurred in one patient. Nonfatal veno-occlusive disease of the liver was diagnosed by clinical criteria32 in two patients. Day 100 transplant-related mortality was only 5% due to the low incidence of acute/chronic GVHD and other transplant-related complications such as CMV, EBV, and VOD. Transplant-related mortality at 2 years was 16%. Development of GVHD was the only variable associated with transplant-related mortality (Fig 2, logrank, P = .08). Among patients who did not develop grades 2 to 4 GVHD, overall transplant-related mortality was only 10%. Patient or donor sex, age, diagnosis, use of G-CSF, and dose of TBI did not influence the likelihood of transplant-related death. No patients who received IL-2 suffered a fatal complication posttransplant. Among surviving patients, more than 90% have Karnofsky performance status of 90% or greater.

Fig. 2.

Transplant-related mortality after CD6 depleted allo-BMT. The estimated probability of death while in remission is depicted for patients with grades 0 to 1 GVHD and compared with those who developed grades 2 to 4 GVHD (logrank, P = .08).

Fig. 2.

Transplant-related mortality after CD6 depleted allo-BMT. The estimated probability of death while in remission is depicted for patients with grades 0 to 1 GVHD and compared with those who developed grades 2 to 4 GVHD (logrank, P = .08).

Close modal

Disease relapse.For patients undergoing CD6-depleted allo-BMT for acute leukemia in first remission, the Kaplan-Meier 3-year estimated incidence of relapse was 25%. The majority of relapses occurred between 6 and 24 months posttransplant. Clinical factors including age, sex, interval between diagnosis and transplant, number of T cells or NK cells infused with the marrow, TBI dose, and development of GVHD did not significantly influence the risk of relapse post-BMT. Six of 32 patients (19%) with grades 0 to 1 GVHD relapsed compared to two of nine patients (22%) with grades 2 to 4 GVHD. Although a greater percentage of patients with AML relapsed than patients with ALL, the estimated likelihood of relapse was not significantly different for the two diseases (31% v 10%, logrank P = .25, Fig 3). All three patients who received IL-2 post-BMT remain alive and free of disease. Two of the patients who relapsed 1 year post-BMT are now disease free following either a second CD6 depleted BMT procedure with busulfan and cyclophosphamide conditioning (4 years after relapse) or CD4 donor lymphocyte infusion (1 year after relapse).

Fig. 3.

Relapse rate following CD6-depleted BMT for acute leukemia. Kaplan-Meier estimated risk of disease relapse is depicted for patients with AML and ALL (logrank, P = .25).

Fig. 3.

Relapse rate following CD6-depleted BMT for acute leukemia. Kaplan-Meier estimated risk of disease relapse is depicted for patients with AML and ALL (logrank, P = .25).

Close modal

Event-free and overall survival.Overall survival at 4 years was 71% with an estimated event-free survival of 63% for adults. Survival was similar for patients with AML and ALL (Fig 4). Age, TBI dose, HLA disparity, and cellular composition of the marrow did not significantly influence event-free or overall survival. Because of the higher mortality rate associated with GVHD, event-free survival was somewhat inferior for those patients who developed grades 2 to 4 GVHD compared with that of patients in whom grades 2 to 4 GVHD did not develop (logrank, P = .08, Fig 5).

Fig. 4.

Survival for patients with AML and ALL. Kaplan-Meier estimated probability of survival is shown for patients transplanted in first remission. Results for patients transplanted for AML and ALL are contrasted (P = NS).

Fig. 4.

Survival for patients with AML and ALL. Kaplan-Meier estimated probability of survival is shown for patients transplanted in first remission. Results for patients transplanted for AML and ALL are contrasted (P = NS).

Close modal
Fig. 5.

Influence of GVHD survival and event-free survival. Kaplan-Meier estimated probability of survival (A) and event-free survival (B) is displayed for patients who developed grades 0 to 1 and 2 to 4 GVHD. Event-free survival is somewhat lower for patients with a history of grades 2 to 4 GVHD (logrank, P = .08)

Fig. 5.

Influence of GVHD survival and event-free survival. Kaplan-Meier estimated probability of survival (A) and event-free survival (B) is displayed for patients who developed grades 0 to 1 and 2 to 4 GVHD. Event-free survival is somewhat lower for patients with a history of grades 2 to 4 GVHD (logrank, P = .08)

Close modal

In most series of patients undergoing unpurged marrow transplantation for acute leukemia in first remission, transplant-related toxicity is the primary reason for treatment failure.16,33,34 Most commonly, fatalities are related to the development of acute and chronic GVHD. GVHD, once established, is difficult to successfully treat and often leads to a fatal outcome.8,10 Ringden et al, reporting for the IBMTR, found a 33% incidence of acute GVHD at 100 days for leukemic patients transplanted in first remission.35 Chronic GVHD occurred in 46% of patients at risk. Transplant-related mortality was 37%.

The risk of transplant-related complications we observed following CD6-depleted allo-BMT for acute leukemia was considerably lower than that previously reported by the IBMTR. Among patients undergoing BMT in first remission, the cumulative incidence of fatal complications was 5% at day +100 and 16% at 2 years. The low risk of transplantrelated mortality in our series is likely related to the low incidence of acute and chronic GVHD. Other serious complications, such as hepatic veno-occlusive disease and EBV lymphoproliferative disease, were rare. In addition, in the absence of immune suppression administered for GVHD, infectious complications were uncommon. In particular, parenchymal infection with CMV was diagnosed in less than 5% of patients. The low incidence of CMV in our series contrasts with the higher likelihood of developing invasive CMV disease (10%-50%) reported after BMT using either other T-cell depletion methods or unpurged marrow.36-39 The relative paucity of opportunistic infections in our series may be related to the elimination of the routine use of immune suppressive medications for GVHD prophylaxis and the steady recovery of circulating T cell number during the first 6 months post-BMT. Although we have observed a variety of abnormalities in T-cell activation and responsiveness following CD6-depleted allo-BMT during the first year posttransplant, the clinical relevance of these defects remains uncertain.40-42 

Despite reductions in the incidence of GVHD achieved by several different T-cell depletion methods, this approach has not been widely accepted for use in recipients of HLA-identical sibling marrow because of an increased risk of disease relapse reported by a number of centers.16,17 The observed increase in disease recurrence has been linked, in part, to the reduction in the incidence of GVHD following donor marrow T-cell depletion. The antitumor effect associated with allogeneic marrow and GVHD has been termed the graft-versus-leukemia (GVL) effect.43-45 There is both experimental and clinical evidence, however, that allogeneic marrow can mediate GVL activity in the absence of GVHD.46-49 Data reported by Gale et al suggests that, after adjusting for the development of GVHD, allogeneic BMT results in a lower risk of relapse than syngeneic transplantation.50 Such data imply that effectively preventing GVHD may not eliminate GVL activity in allogeneic transplant recipients. Conversely, induction of GVHD by truncating the prophylactic regimen or by administering donor buffy coat cells at the time of marrow infusion does not necessarily result in GVL activity sufficient to prevent disease relapse.51 It is noteworthy that in our series the development of GVHD did not exert any significant effect on subsequent disease recurrence.

The impact of T-cell depletion on relapse rate depends on the disease for which patients are being transplanted. In virtually all reports of T-cell depletion of donor marrow in patients with chronic myelogenous leukemia (CML), an increased rate of relapse has been observed.16,17 In contrast, the effect of T-cell depletion on recurrence of acute leukemia post-BMT appears less striking. The relapse rate we observed after CD6 depleted allo-BMT was similar to that reported by the IBMTR as well as that of many individual transplant centers for adults with acute leukemia in first remission52-54 even though our patient population was composed of a large fraction of individuals whose leukemia had cytogenetic abnormalities that confer a poor prognosis following transplantation.55 

Despite the encouraging results we observed in patients with early leukemia, relapse remains a problem. It is possible that efforts to restore some of the GVL activity potentially lost by donor marrow T-cell depletion with IL-2 therapy might reduce relapse rates and improve disease-free survival in these patients. At this point, it would be premature to draw any conclusions regarding the efficacy of low doses of IL-2 after CD6 depleted allogeneic BMT for patients with acute leukemia in first remission. Both encouraging56 and disappointing57 results of IL-2 therapy have recently been reported following autologous transplantation for AML and ALL, respectively. In addition to IL-2 administration, other immunotherapeutic strategies should be investigated, such as the delayed infusion of defined numbers of either marrow or blood derived T-cell subsets post-BMT47,58 or the use of more selective T-cell depletion methods.49 There is some evidence in patients with CML that depletion of CD8+ cells from donor marrow can reduce the incidence of GVHD without compromising GVL activity. However, the efficacy of this approach by itself for GVHD prophylaxis remains unclear as patients receiving CD8 depleted marrow also received cyclosporine at the time of BMT. Another potential approach to preventing disease relapse after T-cell depleted allo-BMT might be intensification of the ablative regimen. Although dose escalation of ablative regimens has previously been associated with decreases in relapse rates at the cost of increased morbidity and mortality,59,60 this strategy may merit further exploration in a setting where the incidence of GVHD is low and patients do not require hepatotoxic and nephrotoxic immune suppressive agents.

Reducing the incidence of GVHD and its attendant mortality without significantly increasing relapse rates makes CD6 T-cell depletion an attractive means of GVHD prophylaxis for patients undergoing allogeneic BMT for acute leukemia in first remission. Studies in patients with AML and in those with ALL comparing the use of allogeneic marrow transplantation to intensive chemotherapy have suggested that the potential advantage provided by allogeneic BMT is significantly diminished by complications associated with BMT. If CD6-depleted allo-BMT can decrease toxicity without compromising disease eradication, then it might be reasonable to adopt it or similar strategies in the treatment of adults in first remission. Not only do morbidity and mortality appear to be limited by this approach, but hospitalization and the costs associated with it may be reduced compared with conventional unpurged marrow transplantation.

Supported by Grant No. A129530. R.J.S. is a recipient of the Baruj Benacerraf Clinical Scholar Award.

Address reprint requests to Robert J. Soiffer, MD, Dana-Farber Cancer Institute, 44 Binney St, Boston, MA 02115.

1
Forman
SJ
Blume
KG
Allogeneic bone marrow transplantation for acute leukemia.
Hematol Oncol Clin North Am
4
1990
517
2
Bortin
MM
Horowitz
MM
Rimm
AA
Increasing utilization of allogeneic bone marrow transplantation.
Ann Intern Med
116
1992
505
3
Zhang
MJ
Hoelzer
D
Horowitz
MM
Gale
RP
Messerer
D
Klein
JP
Loffler
H
Sobocinski
KA
Thiel
E
Weisdorf
DJ
Longterm follow-up of adults with acute lymphoblastic leukemia in first remission treated with chemotherapy or bone marrow transplantation.
Ann Intern Med
123
1995
428
4
Zittoun
R
Mandelli
F
Willemze
R
Autologous or allogeneic bone marrow transplantation compared with intensive chemotherapy in acute myelogenous leukemia.
N Engl J Med
331
1995
217
5
Sebban
C
Lepage
E
Vernant
JP
Gluckman
E
Attal
M
Reiffers
J
Sutton
L
Racodot
E
Michallet
M
Maraninchi
D
Dreyfus
F
Fiere
D
Allogeneic bone marrow transplantation in adult acute lymphoblastic leukemia in first complete remission: A comparative study.
J Clin Oncol
12
1994
2580
6
Archimbaud
E
Thomas
X
Michallet
M
Jaubert
J
Troncy
J
Guyotat
D
Fiere
D
Prospective genetically randomized comparison between intensive postinduction chemotherapy and bone marrow transplantation in adults with newly diagnosed acute myeloid leukemia.
J Clin Oncol
12
1994
262
7
Mayer
RJ
Acute leukemias in adults: An overview of recent strategies.
J Cancer Res Clin Oncol
116
1990
94
8
Martin
PJ
Schoch
G
Fisher
L
Byers
V
Anasetti
C
Appelbaum
FR
Beatty
PG
Doney
K
McDonald
GB
Sanders
JE
Sullivan
KM
Storb
R
Thomas
ED
Witherspoon
RP
Lomen
P
Hannigan
J
Hansen
JA
A retrospective analysis of therapy for acute graft-versus-host disease: Initial treatment.
Blood
76
1990
1464
9
Wingard
JR
Piantadosi
S
Vogelsang
GB
Farmer
ER
Jabs
DA
Levin
LS
Beschorner
WE
Cahill
RA
Miller
DF
Harrison
D
Saral
R
Santos
GW
Predictors of death from chronic graft-versus-host disease after bone marrow transplantation.
Blood
74
1989
1428
10
Sullivan
KM
Weiden
PL
Storb
R
Witherspoon
RP
Fefer
A
Fisher
L
Buckner
CD
Anasetti
C
Appelbaum
FR
Badger
C
Beatty
P
Bensinger
W
Berenson
R
Bigelow
C
Cheever
MA
Clift
R
Deeg
HJ
Doney
K
Greenberg
P
Hansen
JA
Hill
R
Loughran
T
Martin
P
Neiman
P
Petersen
FB
Sanders
J
Singer
J
Stewart
P
Thomas
ED
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
11
Korngold
R
Sprent
J
T cell subsets and graft-versus-host disease.
Transplantation
44
1987
335
12
Wagner
JE
Donnenberg
AD
Noga
SJ
Cremo
CA
Gao
IK
Yin
HJ
Vogelsang
GB
Rowley
S
Saral
R
Santos
GW
Lymphocyte depletion of donor bone marrow by counterflow centrifugal elutriation: Results of a phase I clinical trial.
Blood
72
1988
1168
13
Young
JW
Papadopoulos
EB
Cunningham
I
Castro-Malaspina
H
Flomenberg
N
Carabasi
MH
Gulati
SC
Brochstein
JA
Heller
G
Black
P
Collins
NH
Shank
B
Kernan
NA
O'Reilly
RJ
T-cell-depleted allogeneic bone marrow transplantation in adults with acute nonlymphocytic leukemia in first remission.
Blood
79
1992
3380
14
Filipovich
AH
Vallera
D
McGlave
P
Polich
D
Gajl-Peczalska
K
Haake
R
Lasky
L
Blazar
B
Ramsay
NK
Kersey
J
Weisdorf
D
T cell depletion with anti-CD5 immunotoxin in histocompatible bone marrow transplantation. The correlation between residual CD5 negative T cells and subsequent graft-versus-host disease.
Transplantation
50
1990
410
15
Hale
G
Cobbold
S
Waldmann
H
T cell depletion with CAMPATH-1 in allogeneic bone marrow transplantation.
Transplantation
45
1988
753
16
Marmont
AM
Horowitz
MM
Gale
RP
Sobocinski
K
Ash
RC
van Bekkum
DW
Champlin
RE
Dicke
KA
Goldman
JM
Good
RA
Herzig
RH
Hong
R
Masaoka
T
Rimm
AA
Ringden
O
Speck
B
Weiner
RS
Bortin
MM
T-cell depletion of HLA-identical transplants in leukemia.
Blood
78
1991
2120
17
Goldman
JM
Gale
RP
Horowitz
MM
Biggs
JC
Champlin
RE
Gluckman
E
Hoffmann
RG
Jacobsen
SJ
Marmont
AM
McGlave
PB
Messner
HA
Rimm
AA
Rozman
C
Speck
B
Tura
S
Weiner
RS
Bortin
MM
Bone marrow transplantation for chronic myelogenous leukemia in chronic phase: Increased risk for relapse associated with T-cell depletion.
Ann Intern Med
108
1988
806
18
Kernan
NA
Flomenberg
N
Dupont
B
O'Reilly
RJ
Graft rejection in recipients of T-cell-depleted HLA-nonidentical marrow transplants for leukemia. Identification of host-derived antidonor allocytotoxic T lymphocytes.
Transplantation
43
1987
842
19
Zutter
MM
Martin
PJ
Sale
GE
Shulman
HM
Fisher
L
Thomas
ED
Dumam
DM
Epstein-Barr virus lymphoproliferation after bone marrow transplantation.
Blood
72
1988
520
20
Mitsuyasu
RT
Champlin
RE
Gale
RP
Ho
WG
Lenarsky
C
Winston
D
Selch
M
Elashoff
R
Giorgi
JV
Wells
J
Terasaki
P
Billing
R
Feig
S
Treatment of donor bone marrow with monoclonal anti-T-cell antibody and complement for the prevention of graft-versus-host disease. A prospective, randomized, double-blind trial.
Ann Intern Med
105
1986
20
21
Frame
JN
Collins
NH
Cartagena
T
Waldmann
H
O'Reilly
RJ
Dupont
B
Kernan
NA
T cell depletion of human bone marrow. Comparison of Campath-1 plus complement, anti-T cell ricin A chain immunotoxin, and soybean agglutinin alone or in combination with sheep erythrocytes or immunomagnetic beads.
Transplantation
47
1989
984
22
Frame
JN
Sheehy
D
Cartagena
T
Cirrincione
C
O'Reilly
RJ
Dupont
B
Kernan
NA
Optimal conditions for in vitro T cell depletion of human bone marrow by Campath-1a plus complement as demonstrated by limiting dilution analysis.
Bone Marrow Transplant
4
1989
55
23
Reinherz
EL
Geha
R
Rappaport
JM
Wilson
M
Penta
AC
Hussey
RE
Fitzgerald
KA
Daley
JF
Levine
H
Rosen
FS
Schlossman
SF
Reconstitution after transplantation with T-lymphocyte-depleted HLA haplotype-mismatched bone marrow for severe combined immunodeficiency.
Proc Natl Acad Sci USA
79
1982
6047
24
Rohatiner
A
Gelber
R
Schlossman
SF
Ritz
J
Depletion of T cells from human bone marrow using monoclonal antibodies and rabbit complement. A quantitative and functional analysis.
Transplantation
42
1986
73
25
Soiffer
RJ
Murray
C
Mauch
P
Anderson
KC
Freedman
AS
Rabinowe
SN
Takvorian
T
Robertson
MJ
Spector
N
Gonin
R
Miller
KB
Rudders
RA
Freeman
A
Blake
K
Nadler
LM
Ritz
J
Prevention of graft-versus-host disease by selective depletion of CD6 positive T lymphocytes from donor bone marrow.
J Clin Oncol
10
1992
1191
26
Soiffer
RJ
Mauch
P
Tarbell
NJ
Anderson
KC
Freedman
AS
Rabinowe
SN
Takvorian
T
Murrey
CI
Coral
F
Bosserman
L
Dear
K
Nadler
LM
Ritz
J
Total lymphoid irradiation to prevent graft rejection in recipients of HLA non-identical T cell-depleted allogeneic marrow.
Bone Marrow Transplant
7
1991
23
27
Soiffer
RJ
Murray
C
Cochran
K
Cameron
C
Wang
E
Schow
PW
Daley
JF
Ritz
J
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
28
Mehta
C
Patel
N
A network algorithm for the exact treatment of Fisher's exact test in RxC contingency tables.
J Am Stat Assoc
78
1983
427
29
Glucksberg
H
Storb
R
Fefer
A
Buckner
CD
Nesman
PE
Clift
RA
Lerner
KG
Thomas
ED
Clinical manifestations of graft-versus-host disease in human recipients of HLA matched sibling donors.
Transplantation
18
1974
295
30
Kaplan
EL
Meier
P
Nonparametric estimation from incomplete observations.
J Am Stat Assoc
53
1958
457
31
Armitage P, Berry G: Statistical Methods in Medical Research. Oxford, UK, Oxford, 1987
32
Soiffer
RJ
Dear
K
Rabinowe
SN
Anderson
KC
Freedman
AS
Murray
C
Tarbell
NJ
Mauch
P
Nadler
LM
Ritz
J
Hepatic dysfunction following T-cell-depleted allogeneic bone marrow transplantation.
Transplantation
52
1991
1014
33
Nash
RA
Pepe
MS
Storb
R
Longton
G
Pettinger
M
Anasetti
C
Appelbaum
FR
Bowden
RA
Deeg
HJ
Doney
K
Martin
PJ
Sullivan
KM
Sanders
J
Witherspoon
RP
Acute graft-versus-host disease: Analysis of risk factors after allogeneic marrow transplantation and prophylaxis with cyclosporine and methotrexate [see comments].
Blood
80
1992
1838
34
Mitus
AJ
Miller
KB
Schenkein
DP
Ryan
HF
Parsons
SK
Wheeler
C
Antin
JH
Improved survival for patients with acute myelogenous leukemia.
J Clin Oncol
13
1995
560
35
Ringden
O
Horowitz
MM
Sondel
P
Gale
RP
Biggs
JC
Champlin
RE
Deeg
HJ
Dicke
K
Masaoka
T
Powles
RL
Rimm
AA
Rozman
C
Sobocinski
KA
Speck
B
Zwann
F
Bortin
MM
Methotrexate, cyclosporine, or both to prevent graft-versus-host disease after HLA-identical sibling bone marrow transplants for early leukemia?
Blood
81
1993
1094
36
Engelhard
D
Or
R
Strauss
N
Morag
A
Aker
M
Naparstek
E
Breuer
R
Ravid
Z
Sarov
I
Lijovetzky
G
Slavin
S
Cytomegalovirus infection and disease after T cell depleted allogeneic bone marrow transplantation for malignant hematologic diseases.
Transplant Proc
21
1989
3101
37
Bunjes
D
Hertenstein
B
Wiesneth
M
Stefanic
M
Novotny
J
Duncker
C
Heit
W
Arnold
R
Heimpel
H
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
15
1995
563
38
Winston
DJ
Ho
WG
Lin
CH
Bartoni
K
Budinger
MD
Gale
RP
Champlin
RE
Intravenous immune globulin for prevention of cytomegalovirus infection and interstitial pneumonia after bone marrow transplantation.
Ann Intern Med
106
1987
12
39
Meyers
JD
Flournoy
N
Thomas
ED
Risk factors for cytomegalovirus infection after human marrow transplantation.
J Infect Dis
153
1986
478
40
Kameoka
J
Sato
T
Torimoto
Y
Sugita
K
Soiffer
RJ
Schlossman
SF
Ritz
J
Morimoto
C
Differential CD26-mediated activation of the CD3 and CD2 pathways after CD6-depleted allogeneic bone marrow transplantation.
Blood
85
1995
1132
41
Sugita
K
Nojima
Y
Tachibana
K
Soiffer
RJ
Murray
C
Schlossman
SF
Ritz
J
Morimoto
C
Prolonged impairment of VLA-mediated T cell proliferation via the CD3 pathway following T cell depleted allogeneic bone marrow transplantation.
J Clin Invest
94
1994
481
42
Soiffer
R
Bosserman
L
Murray
C
Cochran
K
Daley
J
Ritz
J
Reconstitution of T cell function following CD6-depleted allogeneic bone marrow transplantation.
Blood
76
1990
235
43
Weiden
PL
Flournoy
MS
Thomas
ED
Prentice
R
Fefer
A
Buckner
CD
Storb
R
Anti-leukemic effect of graft-versus-host-disease in human recipients of allogeneic-marrow grafts.
N Engl J Med
300
1979
1068
44
Antin
JH
Graft-versus-leukemia: No longer an epiphenomenon.
Blood
82
1993
2273
45
Horowitz
MM
Gale
RP
Sondel
PM
Goldman
JM
Kersey
J
Kolb
HJ
Rimm
AA
Ringden
O
Rozman
C
Speck
B
Truitt
RL
Zwaan
FE
Bortin
MM
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
75
1990
555
46
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
1992
1
47
Johnson
B
Drobyski
W
Truitt
R
Delayed infusion of normal donor cells after MHC-matched bone marrow transplantation provides an antileukemia reaction without graft-versus-host disease.
Bone Marrow Transplant
11
1993
329
48
Weiss
L
Lubin
I
Factorowich
I
Lapidot
Z
Reich
S
Reisner
Y
Slavin
S
Effective graft-versus-leukemia effects independent of graft-versus-host disease after T cell depleted allogeneic bone marrow transplantation in a murine model of B cell leukemia/lymphoma. Role of cell therapy and recombinant IL-2.
J Immunol
153
1994
2562
49
Nimer
SD
Giorgi
J
Gajewski
JL
Ku
N
Schiller
GJ
Lee
K
Territo
M
Ho
W
Feig
S
Selch
M
Isacescu
V
Reichert
TA
Champlin
RE
Selective depletion of CD8+ cells for prevention of graft-versus-host disease after bone marrow transplantation. A randomized controlled trial.
Transplantation
57
1994
82
50
Gale
RP
Horowitz
MM
Ash
RC
Champlin
RE
Goldman
JM
Rimm
AA
Ringden
O
Stone
JAV
Bortin
MM
Identical-twin bone marrow transplants for leukemia.
Ann Intern Med
120
1994
646
51
Sullivan
KM
Storb
R
Buckner
CD
Fefer
A
Fisher
L
Weiden
PL
Witherspoon
RP
Appelbaum
FR
Banaji
M
Hansen
J
Martin
P
Sanders
JE
Singer
J
Thomas
ED
Graft-versus-host disease as adoptive immunotherapy in patients with advanced hematologic neoplasms.
N Engl J Med
320
1989
828
52
McGlave
PB
Haake
RJ
Bostrom
BC
Brunning
R
Hurd
DD
Kim
TH
Nesbit
ME
Vercellotti
GM
Weisdorf
D
Woods
WG
Ramsay
NKC
Kersey
JH
Allogeneic bone marrow transplantation for acute nonlymphocytic leukemia in first remission.
Blood
72
1988
1512
53
Doney
K
Fisher
LD
Appelbaum
FR
Buckner
CD
Storb
R
Singer
J
Fefer
A
Anasetti
C
Beatty
P
Bensinger
W
Cliff
R
Hansen
J
Hill
R
Loughran
TP Jr
Martin
P
Petersen
FB
Sanders
J
Sullivan
KM
Stewart
P
Weiden
P
Witherspoon
R
Thomas
ED
Treatment of adult acute lymphoblastic leukemia with allogeneic bone marrow transplantation: Multivariate analysis of factors affecting acute graft-versus-host disease, relapse, and relapse-free survival.
Bone Marrow Transplant
7
1991
453
54
Chao
NJ
Forman
SJ
Schmidt
GM
Snyder
DS
Amylon
MD
Konrad
PN
Nademanee
AP
O'Donnell
MR
Parker
PM
Stein
AS
Smith
E
Wong
RM
Hoppe
RT
Blume
KG
Allogeneic bone marrow transplantation for high-risk acute lymphoblastic leukemia during first complete remission.
Blood
78
1991
1923
55
Gale
RP
Horowitz
MM
Weiner
RS
Ash
RC
Atkinson
L
Babu
R
Dicke
KA
Klein
JP
Lowenberg
B
Reiffers
J
Rimm
AA
Rowlings
PA
Sandberg
AA
Sobocinski
KA
Veum-Stone
J
Bortin
MM
Impact of cytogenetic abnormalities on outcome of bone marrow transplants in acute myelogenous leukemia in first remission.
Bone Marrow Transplant
16
1995
203
56
Benyunes
MC
Massumoto
C
York
A
Higuchi
CM
Buckner
CD
Thompson
JA
Petersen
FB
Fefer
A
Interleukin-2 with or without lymphokine-activated killer cells as consolidative immunotherapy after autologous bone marrow transplantation for acute myelogenous leukemia.
Bone Marrow Transplant
12
1993
159
57
Attal
M
Blaise
D
Marit
G
Payen
C
Michallet
M
Vernant
J
Sauvage
C
Troussard
X
Nedellec
G
Pico
J
Huguet
F
Stoppa
A
Broustet
A
Sotto
J
Pris
J
Maraninchi
D
Reiffers
J
Consolidation treatment of adult acute lymphoblastic leukemia: A prospective, randomized trial comparing allogeneic versus autologous bone marrow transplantation and testing the impact of recombinant interleukin-2 after autologous bone marrow transplantation.
Blood
86
1995
1619
58
Naparstek
E
Or
R
Nagler
A
Cividalli
G
Engelhard
D
Aker
M
Gimon
Z
Manny
N
Sacks
T
Tochner
Z
Weiss
L
Samuel
S
Brautbar
C
Hale
G
Waldmann
H
Steinberg
SM
Slavin
S
T-cell depleted allogeneic bone marrow transplantation for acute leukaemia using Campath-1 antibodies and post-transplant administration of donor's peripheral blood lymphocytes for prevention of relapse.
Br J Haematol
89
1995
506
59
Clift
RA
Buckner
CD
Appelbaum
FR
Bryant
E
Bearman
SI
Peterson
FB
Fisher
LD
Anasetti
C
Beatty
P
Bensinger
WI
Doney
K
Hill
RS
McDonald
GB
Martin
P
Meyers
J
Sanders
J
Singer
J
Stewart
P
Sullivan
KM
Witherspoon
R
Storb
R
Hansen
JA
Thomas
ED
Allogeneic marrow transplantation in patients with chronic myeloid leukemia in the chronic phase: A randomized trial of two irradiation regimens.
Blood
77
1991
1660
60
Geller
RB
Myers
S
Devine
S
Larson
RA
Williams
SF
Park
CL
O'Toole
K
Chandler
C
Topper
RL
Phase I study of busulfan, cyclophosphamide, and timed sequential escalating doses of cytarabine followed by bone marrow transplantation.
Bone Marrow Transplant
9
1992
41
Sign in via your Institution