Graft-versus-host disease (GVHD) is a major cause of mortality and morbidity after allogeneic bone marrow transplantation, but can be avoided by removing T lymphocytes from the donor bone marrow. However, T-cell depletion increases the risk of graft rejection. This study examined the use of CD52 monoclonal antibodies to eliminate T cells from both donor marrow and recipient to prevent both GVHD and rejection. Seventy patients receiving HLA-identical sibling transplants for acute myelogenous leukemia (AML) in first remission were studied. An IgM (CAMPATH-1M) was used for in vitro depletion of the graft and an IgG (CAMPATH-1G) for in vivo depletion of the recipient before graft infusion. No posttransplant immunosuppression was given. Results were compared with two control groups: (1) 50 patients who received bone marrow depleted with CAMPATH-1M, but no CAMPATH-1G in vivo; and (2) 459 patients reported to the International Bone Marrow Transplant Registry (IBMTR) who received nondepleted grafts and conventional GVHD prophylaxis with cyclosporin A (CyA) and methotrexate (MTX). The incidence of acute GVHD was 4% in the treatment group compared with 35% in the CyA/MTX group (P < .001). Chronic GVHD was also exceptionally low in the treatment group (3% v 36%; P< .001). The problem of graft rejection, which had been frequent in the historic CAMPATH-1M group (31%), was largely overcome in the treatment group (6%). Thus, transplant-related mortality of the treatment group (15% at 5 years) was lower than for the CyA/MTX group (26%; P = .04). There was little difference in the risk of leukemia relapse between the treatment group (30% at 5 years) and the CyA/MTX group (29%). Survival of the treatment group at 6 months was better than the CyA/MTX group (92% v 78%), although at 5 years the difference was not significant (62% v 58%) and neither was the difference in leukemia-free survival (60% v52%). We conclude that T-cell depletion is a useful strategy to prevent GVHD, provided that measures are taken to ensure engraftment. Using CAMPATH-1G to deplete residual host lymphocytes is a simple and practical method to do this. At least in AML, the beneficial reduction in GVHD can be achieved without an increased risk of relapse.

HIGH-DOSE CHEMOTHERAPY and radiotherapy followed by transplantation of allogeneic hematopoietic stem cells can cure patients with leukemia. However, allografts have several adverse effects, the most serious of which is graft-versus-host disease (GVHD). It can result in severe damage to skin, liver, and gut, frequently leading to death or chronic disability. To control GVHD, immunosuppressive drugs such as cyclosporin A (CyA), methotrexate (MTX), and corticosteroids are administered posttransplant.1,2 However, even with combined cyclosporin and methotrexate, GVHD remains the single most common cause of death after allogeneic transplants.

For many years it has been known that GVHD can be prevented by depleting T lymphocytes from the donor bone marrow and a variety of methods were developed to accomplish this.3-8 One of the most widely used has been the monoclonal antibody (MoAb) CAMPATH-1M, a rat IgM antibody that recognizes the CD52 antigen.9 CD52 is abundantly expressed on all human lymphocytes and is an exceptionally good target for cell lysis by antibody with human complement; this provided a simple method for purging the donor T cells.10Prior studies demonstrated the efficacy of T-cell depletion and consequent reduction in GVHD.6,11-14 However the benefit was offset by an increased risk of graft rejection by residual host T cells,15,16 and some patients suffered an increased risk of leukemia relapse due to the loss of graft-versus-leukemia effects contributed by donor lymphocytes.12,17 Animal models18,19 and clinical experience20,21 showed that graft rejection might be overcome by increasing the immunosuppression of the recipient before transplantation. One way of delivering this, without adding to the toxicity of the conditioning regimen, is to use MoAbs to deplete residual host T cells.22 A rat IgG2b CD52 antibody, CAMPATH-1G, effectively depletes human lymphocytes in vivo.23 Like CAMPATH-1M, it can activate human complement, although this is not sufficient for systemic T-cell depletion. Rat IgG2b also binds human Fc receptors and engages cellular killing mechanisms.24 

A combined strategy using CAMPATH-1M to T-cell–deplete donor bone marrow and intravenous CAMPATH-1G to ablate residual host immunity has been used in more than 600 transplants worldwide, primarily in transplants from unrelated donors.13,14,25,26 We report here results of 70 HLA-identical sibling transplants for acute myelogenous leukemia (AML). We compare the results first to a historical group receiving CAMPATH-1M-treated marrow but no intravenous CAMPATH-1G and second to a matched group of concurrently treated patients reported to the International Bone Marrow Transplant Registry (IBMTR) who received unmanipulated transplants and posttransplant cyclosporin plus methotrexate for GVHD prophylaxis.

MoAbs.

CAMPATH-1G27 was prepared from the culture supernatant of hybrid myeloma cell culture in a hollow-fiber fermentor (Acusyst-Jr; Cellex, Minneapolis, MN). It was purified by affinity chromatography on protein A sepharose, followed by ion exchange chromatography on S-sepharose, and formulated in phosphate-buffered saline. CAMPATH-1M10 was prepared from hybrid myeloma cells using three methods: (1) ascitic fluid fractionated with ammonium sulphate, (2) hollow-fiber culture supernatant fractionated with ammonium sulphate, and (3) culture supernatant from stirred fermentors purified by affinity chromatography on protein A sepharose (this was performed by Wellcome Biotech [Beckenham, UK], who provided some of the antibody for this study). Batches prepared by each method were tested in a variety of analytical systems. All had comparable potency for complement-mediated cell lysis and insignificant effect on colony-forming cells.28,29 Process (1) was used for the historic controls, process (2) was used for the study patients at London and Riyadh, and process (3) was used for the study patients at Ulm.

In vitro T-cell depletion of bone marrow.

A similar depletion procedure was used for all transplants, as described previously.11,25 Donor bone marrow was harvested in the usual way and processed using a cell separator to prepare a cell concentrate in balanced salt solution (containing Ca2+) that was free from plasma and depleted of red blood cells and granulocytes. The volume of the mononuclear cell suspension was adjusted so that the cell density did not exceed 5 × 107/mL, and CAMPATH-1M was added to give a final concentration of 0.1 mg/mL. The mixture was incubated for 10 to 20 minutes at room temperature, and then donor serum was added to a final concentration of 25% (vol/vol). It was then incubated for a further 20 to 45 minutes at 37°C. The treated bone marrow was washed once before infusion. Experiments had shown that differences in antibody batch or incubation timing did not materially affect the efficacy of T-cell depletion (G.H., unpublished work). The fraction of residual T cells was measured using standard methods, according to the practice in each center, either by E-rosettes or by fluorescence-activated cell sorting (FACS) analysis using appropriate T-cell–specific mouse MoAbs. (All of the study patients were assessed by FACS analysis using CD3 antibodies to enumerate T cells.)

In vivo administration of CAMPATH-1G.

Patients were treated with 20 mg/d of CAMPATH-1G over a period of 5 days at the beginning of the pretransplant conditioning therapy. Each dose was diluted in 250 mL normal saline and infused intravenously over 3 hours. To minimize the expected systemic first dose reaction, most patients received medication before the first antibody infusion, either 0.5 to 1 g prednisolone (Ulm) or 100 mg hydrocortisone plus 50 mg diphenhydramine (Royal Free, Riyadh), followed by 3× 1 g paracetamol daily.

Patients: Study group.

Three transplant centers participated in the study: Ulm University Hospital (Ulm, Germany), Royal Free Hospital (London, UK), and King Faisal Hospital (Riyadh, Saudi Arabia). The original plan was to include all patients over 13 years with acute leukemia in first remission transplanted from HLA-matched siblings with the intent of comparing results in acute lymphoblastic leukemia (ALL) and AML. Because very few patients with ALL were recruited, this analysis focuses on patients with AML. (Inclusion of the ALL patients would not significantly affect the results.) The conditioning regimen was determined according to the standard protocol of each center. All included cyclophosphamide plus total body irradiation (TBI), which was administered as a single dose in 23% patients (at the Royal Free) and multiple fractions in the others (at Ulm and Riyadh). Additional chemotherapy (busulphan) was administered in 6% of patients (Royal Free only). None of the patients received any additional lymphoid irradiation and none received posttransplant immunosuppression. There was no selection of patients according to cytogenetic risk group and data on risk groups were not reported systematically. Each center recruited consecutive patients provided that they gave informed consent. The numbers of patients and conditioning regimens at each center are shown in Table 1.

Table 1.

Conditioning Regimens Used at the Three Study Centers

Ulm Royal Free Riyadh
No. of patients  41 16  13  
TBI fractions (no.)  6  1  6  
TBI total dose (rad)  1,200  750  1,200  
TBI dose rate (rad/min) 12  16  8  
Time between last dose of antibody and transplant  5 d  1-3 d  5 d 
Additional chemotherapy (no. of patients)  0  4  
Ulm Royal Free Riyadh
No. of patients  41 16  13  
TBI fractions (no.)  6  1  6  
TBI total dose (rad)  1,200  750  1,200  
TBI dose rate (rad/min) 12  16  8  
Time between last dose of antibody and transplant  5 d  1-3 d  5 d 
Additional chemotherapy (no. of patients)  0  4  
Patients: Historic CAMPATH-1M controls.

A database is maintained by GH of all transplants using CAMPATH-1 antibodies. It contains information on the patient and donor characteristics and transplant outcomes. It is a condition of antibody supply that data are regularly reported to the CAMPATH users database. Controls were selected using the following criteria: (1) patients more than 13 years of age, (2) transplants for AML in first remission, (3) HLA-identical sibling donors, (4) conditioning regimens based on cyclophosphamide and TBI, (5) no lymphoid irradiation, (6) no intravenous CAMPATH-1G or other antibody, (7) T-cell depletion with CAMPATH-1M, and (8) no posttransplant immunosuppression. Fifty patients meeting these criteria were identified; 26 were transplanted at the same three centers as the 70 study patients.

Patients: IBMTR controls.

A second control group of patients receiving non–T-cell–depleted transplants between 1984 and 1995 was selected from the IBMTR database using the same criteria described above, except that patients from the three study centers were excluded and GVHD prophylaxis was with combined CyA and MTX.30 IBMTR is a voluntary study group of over 300 transplant centers worldwide that contribute detailed data to a Statistical Center at the Medical College of Wisconsin. Participants are required to report all consecutive transplants. The IBMTR database includes 40% to 45% of allogeneic transplant recipients since 1970. Computerized error checks, physician review of submitted data, and on-site audits of participating centers ensure data quality. Selection of the control group proceeded as follows: patients with AML, transplanted in CR1 from 1984 through 1995 (inclusive), N = 2,940; exclude the three study centers, N = 2,759; select patients transplanted from HLA-identical siblings, N = 2,429; select cyclophosphamide + TBI with no ATG for conditioning, N = 1,307; select MTX plus CyA for GVHD prophylaxis, N = 512; and select patients more than 13 years of age at transplant, N = 459.

These 459 cases were reported to the IBMTR by 95 transplant centers.

Statistical analysis.

Characteristics of the treatment groups were compared using the χ2 test for categorical variables and the Wilcoxon two-sample test for continuous variables. Comparing outcomes between the treatment groups required adjustment for the differences in baseline characteristics of the patients. First, associations between outcomes and potential prognostic variables were evaluated in each group separately using Cox proportional hazards regression with a forward stepwise approach.31 The outcomes considered were rate of engraftment (days to reach 0.5 × 109neutrophils/L), graft failure, transplant-related mortality (TRM; defined as death in continuous complete remission), relapse, survival, and leukemia-free survival. Variables considered were age at transplant, recipient gender, donor gender, and year of transplant. Variables significantly associated with the outcome in any treatment group were included as covariates in subsequent comparisons. Possible interactions between significant covariates and the type of treatment were tested. The proportionality assumption of the Cox model was tested by adding a time-dependent covariate for each covariate.32The proportionality assumption did not hold for treatment effects on survival, indicating that the relationship between treatment and survival outcomes differed over time. To determine regions of the treatment period where the relative risk of mortality between different treatments was a constant, a series of Cox models with different cut-off time points for time-dependent treatment effects were fitted.32 The final model chosen was the one giving the largest partial likelihood. In this model, treatment was considered as a time-dependent covariate with different coefficients from 0 to 6 and greater than 6 months since transplantation. The adjusted relative risks (95% confidence intervals and P values) of the study group versus historic group and study group versus MTX/CyA group were calculated for each outcome. Where appropriate, confidence intervals were calculated based on a log transformation.

T-cell depletion in vitro by CAMPATH-1M.

Several methodologies were used over the years to estimate the fraction of residual T-cells in treated marrow (Table 2). The measured fraction of residual T cells in the study group (median, 0.4%) was less than in the historic control group (median, 1.0%; P = .024), but the result must be treated with caution owing to the potential variability in the measurement of small numbers of cells. The total number of mononuclear cells infused varied significantly between each of the three study centers and between the study group (median, 1.0 × 108/kg) and the historic control group (median, 2.15 × 108/kg; P < .001). Consequently, the calculated numbers of T cells infused differed significantly between the study group (median, 0.2 × 106/kg) and the historic group (median, 1.7 × 106/kg; P < .001).

Table 2.

Measured Extent of T-Cell Depletion In Vitro and Numbers of T Cells Infused

Ulm Royal Free Riyadh Study Group Historic Group
No. of patients measured  40  15  13  68  40 
Mononuclear cells (×108/kg)  1.4 (0.5-4.0) 0.5 (0.2-0.8)  0.1 (0.1-2.4)  1.0 (0.1-4.0) 2.2 (0.1-6.0)  
T cells (%)  0.8 (0.0-3.0) 0.2 (0.0-7.5)  0.2 (0.0-2.7)  0.4 (0.0-7.5) 1.0 (0.0-10.0)  
T cells infused (×106/kg) 0.8 (0.0-5.4)  0.12 (0.0-3.7)  0.04 (0.0-2.2) 0.2 (0.0-5.4)  1.7 (0.0-25.0) 
Ulm Royal Free Riyadh Study Group Historic Group
No. of patients measured  40  15  13  68  40 
Mononuclear cells (×108/kg)  1.4 (0.5-4.0) 0.5 (0.2-0.8)  0.1 (0.1-2.4)  1.0 (0.1-4.0) 2.2 (0.1-6.0)  
T cells (%)  0.8 (0.0-3.0) 0.2 (0.0-7.5)  0.2 (0.0-2.7)  0.4 (0.0-7.5) 1.0 (0.0-10.0)  
T cells infused (×106/kg) 0.8 (0.0-5.4)  0.12 (0.0-3.7)  0.04 (0.0-2.2) 0.2 (0.0-5.4)  1.7 (0.0-25.0) 

Cell numbers are reported as medians, with the range in parentheses. There was a statistically significant difference between each study center in the total numbers of mononuclear cells (P < .005), but there was no significant difference between centers in the percentage of residual T cells or the total numbers of T cells infused. However, there was a significant difference between the study group and the historic control group with regard to each of these parameters (P = .024 for percentage of T cells and P < .001 for total T cells).

Lymphocyte depletion in vivo by CAMPATH-1G.

CAMPATH1G treatment was started before other components of the conditioning regimen, so that antibody effects could be determined. All patients had rapid and profound depletion of blood lymphocytes. The number of clonable T cells that could be recovered from recipient blood samples obtained 1 to 2 days after the CAMPATH-1G infusions was reduced by 2.5 to 3 logs compared with pretreatment samples.33As in other studies with CAMPATH-1G or CAMPATH-1H, there was generally a first-dose effect of fever, often with rigors and nausea, which is related to a release of cytokines.34,35 In two patients the reaction was severe, and it was decided to discontinue CAMPATH-1G. (These patients are still included in the analysis.) All other patients had diminished reactions to the second and subsequent doses.

Effect of year of transplant.

To determine whether patients transplanted before 1990 could be included in comparative analyses, we examined the IBMTR dataset for differences between patients transplanted in 1984 through 1989 compared with 1990 through 1995. There were no statistically significant differences in the actuarial risks of transplant-related mortality (26% v 27% at 4 years), relapse (26% v 33% at 5 years), survival (57% v 58% at 5 years), or leukemia-free survival (54% v 49% at 5 years). Therefore, all data from 1984 through 1995 were pooled for subsequent analyses.

Comparison of prognostic features of the study and control groups.

Characteristics of the study and control groups are shown in Table 3. The most significant difference was in age. The median age of the study group (36 years) was significantly higher than the historic control (30 years; P = .003) or the CyA/MTX group (31 years; P = .03). There was also a difference in the gender of patients between the study group (57% male) and the historic control (38% male; P = .04), but not the CyA/MTX group (50% male). The potentially confounding effect of patient age on the outcome has been adjusted in a multifactorial model (see below).

Table 3.

Features of Study and Control Groups

Study (1) Historic (2)CyA/MTX (3) Significance
P12P13
No. of patients  70  50  459  
Median year of transplant 92 (87-96)  86 (84-94)  90 (84-95) <.001  <.001  
Median follow-up (months)  44  116 58  <.001  .01  
Male patient  57% 38%  50%  .04  .24  
Male donor 61%  52%  54%  .34  .29 
Median age at transplant  36 (14-50) 30 (14-47)  31 (14-56)  .003  .03 
Study (1) Historic (2)CyA/MTX (3) Significance
P12P13
No. of patients  70  50  459  
Median year of transplant 92 (87-96)  86 (84-94)  90 (84-95) <.001  <.001  
Median follow-up (months)  44  116 58  <.001  .01  
Male patient  57% 38%  50%  .04  .24  
Male donor 61%  52%  54%  .34  .29 
Median age at transplant  36 (14-50) 30 (14-47)  31 (14-56)  .003  .03 

The extreme range of the data is shown in parentheses. Probabilities (P) are calculated by the χ2 test or Wilcoxon two-sample test as appropriate.

Comparison of outcome for study and control groups.

Univariate analyses of outcome are shown in Table 4. Engraftment was significantly slower in both CAMPATH groups compared with the CyA/MTX group (Fig 1). Even disregarding patients who did not engraft at all, there was a delay of 1 day in the median time to reach 0.5 × 109 neutrophils/L for the historic CAMPATH group and 5 days for the study group. There was a significant variation in time to engraftment among the three study centers. The median time to reach 0.5 × 109 neutrophils/L at Ulm was 23 days, whereas at the Royal Free and Riyadh, the median was 30 days (P = .001). Platelet engraftment was not reported in this study.

Table 4.

Outcome According to Treatment Group

% Probability (95% confidence interval) Significance
Study (1) Historic (2)CyA/MTX (3) P12P13
0.5 × 109 neutrophils/L     .84 <.0001  
 By day 21  23 (14, 34)  50 (36, 64) 59 (55, 64)  
 By day 30  78 (67, 87)  70 (57, 82) 93 (90, 95)  
 By day 60  94 (87, 98)  83 (71, 92) 99 (97, 100)  
Graft failure     .0003  .03 
 At 1 mo  4 (1, 11)  20 (10, 32)  1 (0, 3)  
 At 12 mo  6 (2, 13)  31 (19, 45)  2 (1, 3)  
Acute GVHD 
 Grade II-IV  4  20  35  .01 <.001  
Chronic GVHD  
 Grade I-IV (limited-extensive) 3  21  36  .005  <.001 
Transplant-related mortality     <.0001  .06 
 At 3 yrs  15 (7, 25)  53 (38, 67)  25 (21, 29) 
 At 5 yrs  15 (7, 25)  58 (43, 72)  26 (22, 31) 
Relapse     .54  .67  
 At 3 yrs 27 (17, 39)  22 (9, 38)  24 (20, 29)  
 At 5 yrs 30 (19, 42)  22 (9, 38)  29 (24, 35)  
Survival    .001  .27  
 At 3 yrs  65 (53, 76) 39 (26, 53)  59 (55, 64)  
 At 5 yrs  62 (50, 74) 35 (22, 49)  58 (53, 62)  
Leukemia-free survival    .002  .24  
 At 3 yrs  62 (50, 73) 37 (24, 51)  57 (52, 62)  
 At 5 yrs  60 (47, 71) 33 (21, 47)  52 (47, 57) 
% Probability (95% confidence interval) Significance
Study (1) Historic (2)CyA/MTX (3) P12P13
0.5 × 109 neutrophils/L     .84 <.0001  
 By day 21  23 (14, 34)  50 (36, 64) 59 (55, 64)  
 By day 30  78 (67, 87)  70 (57, 82) 93 (90, 95)  
 By day 60  94 (87, 98)  83 (71, 92) 99 (97, 100)  
Graft failure     .0003  .03 
 At 1 mo  4 (1, 11)  20 (10, 32)  1 (0, 3)  
 At 12 mo  6 (2, 13)  31 (19, 45)  2 (1, 3)  
Acute GVHD 
 Grade II-IV  4  20  35  .01 <.001  
Chronic GVHD  
 Grade I-IV (limited-extensive) 3  21  36  .005  <.001 
Transplant-related mortality     <.0001  .06 
 At 3 yrs  15 (7, 25)  53 (38, 67)  25 (21, 29) 
 At 5 yrs  15 (7, 25)  58 (43, 72)  26 (22, 31) 
Relapse     .54  .67  
 At 3 yrs 27 (17, 39)  22 (9, 38)  24 (20, 29)  
 At 5 yrs 30 (19, 42)  22 (9, 38)  29 (24, 35)  
Survival    .001  .27  
 At 3 yrs  65 (53, 76) 39 (26, 53)  59 (55, 64)  
 At 5 yrs  62 (50, 74) 35 (22, 49)  58 (53, 62)  
Leukemia-free survival    .002  .24  
 At 3 yrs  62 (50, 73) 37 (24, 51)  57 (52, 62)  
 At 5 yrs  60 (47, 71) 33 (21, 47)  52 (47, 57) 

The outcome is reported, together with the 95% confidence interval. Significance (P) values in this table (except for GVHD) were calculated from a univariate long-rank test (ie, they do not take into account any potential covariates).

Fig. 1.

Speed of engraftment. Note the cross-over between study and control groups. The study group engrafts more slowly, but a higher proportion of patients eventually engraft.

Fig. 1.

Speed of engraftment. Note the cross-over between study and control groups. The study group engrafts more slowly, but a higher proportion of patients eventually engraft.

Close modal

The study group had a significantly lower risk of rejection than the historic group (6% v 31%; P = .0003) but a higher risk than the CyA/MTX group (2%; P = .03; Fig 2). The incidence of both acute and chronic GVHD was significantly lower in the study group compared with either of the two control groups. Only 3 of the 70 study patients developed grade 2 acute GVHD and 2 developed mild/moderate chronic GVHD; there were no more severe cases.

Fig. 2.

Risk of graft failure.

Fig. 2.

Risk of graft failure.

Close modal

Transplant-related mortality was significantly lower in the study group than in the historic group (15% v 58%; P < .0001) and also less than in the CyA/MTX group (26%; P = .04; Fig 3). Relapse was similar in the three groups. Survival and leukemia-free survival were significantly higher in the study group than in the historic group, but similar to that in the CyA/MTX group (Figs 4,5, and 6).

Fig. 3.

Probability of transplant-related mortality (death in continuous complete remission). Note the significantly reduced mortality of the study group in the first 6 months.

Fig. 3.

Probability of transplant-related mortality (death in continuous complete remission). Note the significantly reduced mortality of the study group in the first 6 months.

Close modal
Fig. 4.

Probability of relapse.

Fig. 4.

Probability of relapse.

Close modal
Fig. 5.

Probability of survival.

Fig. 5.

Probability of survival.

Close modal
Fig. 6.

Probability of leukemia-free survival.

Fig. 6.

Probability of leukemia-free survival.

Close modal
Outcome after graft failure/rejection.

Primary failure of engraftment was documented in 9 of the historic control group (18%) and 3 of the study group (4%). Late graft rejection occurred in 6 of the historic group (12%) and 1 of the study group (1%). It was not possible to determine the cause of graft failure in every case, but it is likely that most, if not all, were due to immunological rejection. Therefore, both early and late graft failure were analyzed together. Patients were treated by reinfusion of stored autologous marrow (5 historic control and 1 study) or by a second transplant of unfractionated allogeneic marrow from the original donor (6 historic control and 3 study). In 11 cases, these rescue procedures resulted in successful engraftment, but many of the patients suffered further complications and eventually 12 of 15 patients died in the historic group (9 from graft failure) and 4 of 4 patients died in the study group (all from relapse).

Multivariate analysis.

The results were further analyzed using the Cox proportional regression model to test for interactions with prognostic factors that might have affected the results of univariate analyses (Table 5). Variables used in building the model were age, year of transplant, patient gender, donor gender, and treatment group. Outcomes were rate of engraftment, graft failure, transplant-related mortality, relapse, survival, and leukemia-free survival. There was a significant association between age and transplant-related mortality. The relative risk of transplant-related mortality for patients more than 30 years of age was 1.53 times that for patients under 30 years (95% confidence interval, 1.08 to 2.15;P = .02). However, the relationship between patient groups and outcome was similar for young and old patients. In the final model, allowing for the effect of age, there was less than half the risk of transplant-related mortality in the study group compared with the CyA/MTX control (relative risk, .45; P = .02). Tests for proportionality indicated that the effect of the study treatment on survival differed at different times after the transplant. In the first 6 months after the transplant, survival of patients in the study group was significantly higher than the control groups; among patients surviving 6 months, subsequent survival was similar in the study group versus the two control groups. It is not surprising that no significant difference in long-term survival could be shown, because the size of the study did not give it sufficient power to demonstrate even a 10% difference.

Table 5.

Analysis of Outcome According to a Multivariate Model

Outcome Studyv Historic Control Studyv CyA/MTX Control
RR (95% CI) PRR (95% CI) P
Graft failure .17 (.06, .51)  .002  3.53 (1.03, 12.05)  .04 
Transplant-related mortality  .20 (.09, .43) <.0001  .45 (.23, .90)  .02  
Relapse 1.33 (.58, 3.07)  .50  1.11 (.67, 1.84)  .68  
Death 
 <6 mo  .30 (.12, .75)  .01  .38 (.18, .82)  .01 
 >6 mo  .55 (.28, 1.07)  .08  1.37 (.82, 2.30) .23  
Death and/or relapse .48 (.29, .80)  .005  .78 (.52, 1.67)  .23 
Outcome Studyv Historic Control Studyv CyA/MTX Control
RR (95% CI) PRR (95% CI) P
Graft failure .17 (.06, .51)  .002  3.53 (1.03, 12.05)  .04 
Transplant-related mortality  .20 (.09, .43) <.0001  .45 (.23, .90)  .02  
Relapse 1.33 (.58, 3.07)  .50  1.11 (.67, 1.84)  .68  
Death 
 <6 mo  .30 (.12, .75)  .01  .38 (.18, .82)  .01 
 >6 mo  .55 (.28, 1.07)  .08  1.37 (.82, 2.30) .23  
Death and/or relapse .48 (.29, .80)  .005  .78 (.52, 1.67)  .23 

The Cox proportional regression model was used to allow for factors that might influence the outcome other than the treatment group. The only relevant factor was patient age. The adjusted relative risks (RR) for transplant outcome according to treatment group are shown, together with the 95% confidence interval (CI) and the significance (P). A relative risk less than 1.0 means the outcome was less likely in the study group compared with the control group; a figure more than 1.0 means that the outcome was more likely.

Causes of death.

The underlying causes of death in each patient group are reported in Table 6. Because of the small numbers in the study and historic control group and the difficulties inherent in assigning a single cause of death to some patients, we did not attempt formal statistical analysis. The most frequent cause of death in the study group was relapse (15 patients), followed by infection (7 patients). There were 3 deaths from infection in the study group after 6 months: 1 varicella zoster + aspergillus (day 191), 1 varicella zoster + hepatitis C (day 858), and 1 unknown organism (day 387).

Table 6.

Causes of Death According to Treatment Group

Study Historic CyA/MTX
Graft failure/rejection 1  9  1  
GVHD  0  2  36  
Relapse  15  56  
Infection  7  8  26  
Other (eg, cardiovascular, conditioning toxicity)  2  4  58  
Secondary malignancy 0  1  1  
Unknown  0  2  5  
Total  25  32 183 
Study Historic CyA/MTX
Graft failure/rejection 1  9  1  
GVHD  0  2  36  
Relapse  15  56  
Infection  7  8  26  
Other (eg, cardiovascular, conditioning toxicity)  2  4  58  
Secondary malignancy 0  1  1  
Unknown  0  2  5  
Total  25  32 183 

A single underlying cause of death was assigned to each patient. Because of the small numbers, the differences in follow-up times (see Table 3), and the difficulty in assigning a unique cause to each case, statistical analysis is deemed to be impracticable.

Since the early clinical trials of T-cell depletion, it was realized that graft rejection by residual host T cells was a major problem that negated much of the clinical benefit of avoiding GVHD. Graft rejection could be reduced, but not eliminated, by giving extra whole body20 or total lymphoid irradiation21 or by administering posttransplant CyA.13 Animal experiments showed that anti–T-cell MoAbs could be used to escalate the immunosuppression without toxicity, and this prompted the collaborative CAMPATH users group to begin a number of pilot studies using CAMPATH-1G to deplete residual host T cells.13,14,25,26 These studies gave encouraging results, but it was possible that other improvements in transplant protocols over time might have influenced the outcome. Therefore, we performed the present comparison with a large contemporaneous control group of patients selected from the IBMTR database. A second problem associated with T-cell depletion is the loss of beneficial graft-versus-leukemia effects. This is well documented for patients with chronic myeloid leukemia (CML), but it is not clear whether there is a significant effect in patients with acute leukemia. In this analysis, we evaluated relapse in patients transplanted for AML in first complete remission.

This study convincingly demonstrates the effectiveness of the combined CD52 antibodies in dramatically reducing the risk of acute and chronic GVHD, without posttransplant immunosuppression. There were no cases of severe acute or chronic GVHD in the study group. Similar results have been reported when the same antibody protocol was used in other transplant indications.14,26 The addition of CAMPATH-1G in vivo pretransplant resulted in lower GVHD rates than those seen in the historic controls, which might be due to additional depletion of donor T cells in vivo by residual CAMPATH-1G at the time of transplant. The measured extent of T-cell depletion by CAMPATH-1M was greater for the study patients than the historic controls, but the significance of this is hard to assess due to the technical difficulties in accurate measurement of small numbers of T cells and the fact that actual depletion is likely to have been greater than measured, because residual T cells would be coated with CAMPATH-1M antibody and lysed when they encounter fresh complement after infusion of the bone marrow.

Importantly, the study group also had a significantly lower risk of graft failure than the historic control (6% at 12 months). This is still higher than in the non–T-cell–depleted group (2%), but acceptable, given the much lower risk of GVHD. Possibly graft failure would be reduced still further if larger numbers of stem cells were infused, as is now possible using peripheral blood harvests. Despite the improvement in overall engraftment, the speed of engraftment, as measured by the time to reach 0.5 × 109neutrophils/L, was significantly slower in the study group compared with either control group (after excluding graft failure). The most likely explanation for this delay is the comparatively small number of mononuclear cells infused in the study group, especially because the greatest delay was observed at the two centers where the smaller numbers of cells were infused (Table 2). Experimental models have shown that speed of engraftment is related to total stem cell dose,36 and this has been confirmed in a recent multifactorial analysis of the whole CAMPATH users database (G.H. and S.P. Cobbold, unpublished work). Rapid engraftment has been reported with CAMPATH-1–treated stem cells from peripheral blood.37 

The relative risk of transplant-related mortality was significantly lower in the study group compared with the CyA/MTX group (15%v 26%). This is most likely due to reducing the incidence and severity of GVHD, although we cannot rule out a benefit due to the avoidance of toxicity of the immunosuppressive drugs themselves or consequent infection risks. One of the most important parameters in the long-term follow-up was the risk of relapse, because it has been shown that T-cell depletion increases relapse risk substantially for CML,17,38 and it is suggested that there could be a modest increase in risk for acute leukemias. In contrast, we found no significant difference between the study group (30% risk of relapse at 5 years) and the CyA/MTX group (29%), supporting the concept that the impact of T-cell–mediated graft-versus-leukemia reactions is minimal in patients transplanted for acute leukemia in first remission.13 Overall survival was significantly better in the study group compared with the CyA/MTX group up to 6 months; subsequent survival and leukemia-free survival were slightly, but not significantly better.

Prospective randomized studies are often thought to be the gold standard in evaluating new treatments, but their application in transplantation is hindered by the relative infrequency of the diseases treated and the fact that only a minority of patients have suitable donors. This makes accrual of sufficient patients difficult, if not impossible. We were primarily interested in measuring differences in the risk of graft failure, where the expected results were in the range of 2% to 15%. Hundreds of patients would be required to give an adequate power. Fortunately, a better alternative is available. Large clinical databases, such as the one maintained by the IBMTR, contain data on a large proportion of transplant recipients worldwide with details of prognostic and treatment factors that allow application of sophisticated statistical techniques to adjust for differences between groups and exploit the power of large numbers.39 In the current study, we identified 459 suitable controls receiving the most common approach to GVHD prophylaxis against which the combined antibody strategy could be compared. Unlike many prospective randomized trials, in which significant differences are sometimes attributed to unusually poor performance of the control group, we can be sure that our CyA/MTX control group is truly representative. The accuracy of the control data are confirmed by published results from the European Transplant Registry on an overlapping set of patients, where the outcomes are superimposable.40 However, it might be argued that the three study centers shared some favorable factor in common other than the treatment protocol. In fact, the three centers were more remarkable for their diversity in approaches to transplantation. Furthermore, this idea is negated by the published comparison from one center (Ulm) of study patients compared with their own CyA/MTX control group.25 The results are very similar to those presented here, except of course for the smaller numbers of control patients. The limitations of our analysis should be recognized, particularly the difficulty of allowing for possible differences in relapse risk according to AML subtype, prior therapy, and conditioning regimen, but we can be confident that the control group is representative of contemporary clinical practice.

Immune reconstitution was not specifically studied here, but results for marrow transplants depleted with CAMPATH-1M have been reported previously.41-43 All of these reports agree that lymphocyte recovery, particularly of CD4+ cells, is slow compared with T-replete transplants. There does not appear to be a substantial long-term mortality as a result of opportunistic infections, but this requires continued surveillance. Some groups report an early increase in the frequency of cytomegalovirus (CMV) reactivation, although this did not necessarily lead to severe clinical disease.44 The cellular distribution of the CD52 antigen may be fortuitous in this regard. NK cells are relatively spared by CAMPATH-1,41,45 and it has been suggested that they may play a role in control of CMV disease and have an antileukemia effect.46-51 However, B cells are efficiently depleted. Elsewhere, we have reported that T-cell depletion with CAMPATH-1 antibodies does not give rise to an excess of B-cell lymphoproliferative disorders, in contrast to some other methods of T-cell depletion.52 We believe this is because depletion of donor B cells removes both a potential reservoir of virus and its major target.

In this trial, CAMPATH-1G was used as a form of monoclonal antilymphocyte globulin to achieve the additional depletion of recipient lymphocytes required to permit engraftment of T-cell–depleted bone marrow. The positive outcome is in accord with animal experiments, confirming that graft failure was caused by lymphocyte-mediated rejection. The logical development is to use the IgG antibody CAMPATH-1G (or its humanized equivalent CAMPATH-1H) for depletion of both donor and recipient T cells, and current trials are aimed at developing the simplest and most effective way to administer it—either in vivo before and after the transplant14,53,54or as a single dose, mixed and infused with the donor bone marrow.13,55 This study confirms that T-cell depletion is the best way to prevent GVHD. Our approach largely avoids graft rejection and does not result in an increased risk of relapse, at least in AML patients. The disease-free survival achieved is at least as good as with conventional regimens, but with the following important advantages: (1) posttransplant immunosuppression is no longer needed and (2) almost complete elimination of acute and chronic GVHD should translate into a substantially better quality of life for the survivors.

The authors are indebted to many colleagues who played an important part in the production of antibodies, care of patients, data collection, and analysis, including Jenny Phillips and the staff of the Therapeutic Antibody Centre (Cambridge, UK), Marcus Wiesneth, Bernd Hertenstein, Renate Arnold (Ulm, Germany), Mike Hamon, Ian MacDonald, Grazyna Galatowicz, Chris Collins (Royal Free, London, UK), Hugh Clink, Andrew Padmos, Peter Ernst, and Kirtikant Sheth (Riyadh, Saudi Arabia) as well as the following transplant teams throughout the world who contributed to the registry data. Australia: Royal Prince Alfred Hospital, Camperdown; Alfred Hospital, Prahran; Westmead Hospital, Westmead; Austria: Univ. Klinik fur Innere Medizin I, Vienna; Belgium: A.Z. Sint-Jan, Brugge; Cliniques Universitaires Saint-Luc, Brussels; University Hospital Gasthuisberg, Leuven; University of Liege, Liege; Brazil: Hospital de Clinicas, Curitiba; Centro Nacional de Transplate de Medula Ossea-CEMO, Rio de Janeiro; Canada: Ontario Cancer Instute, Princess Margaret Hospital, Toronto; China: Bei Tai Ping Lu Hospital, Beijing; Croatia: Centar za Transplataciju Kostane Srzi, Zagreb; Cuba: Institute de Hematologia e Immunologia, Ciudad de la Havana; Czech Republic: Institute of Hematology and Blood Transfusion, Praha 2; Finland: Helsinki University Central Hospital, Helsinki 29; Turku University Central Hospital, Turka 52;France: CHRU, Angers; Chu Clemenceau, Caen; Hopital A. Michallon, Grenoble; Centre Hospitalier Regional de Lille, Lille; Institut J. Paoli I. Calmettes, Marseille; Hopital Saint Louis, Paris; Hopital Saint-Antoine, Paris; Hotel Dieu de Paris, Paris; Groupe Hospitalier du haut Leveque, Pessac; Hopital Jean Bernard, Poiters; Center Henri Becquerel (C.R.L.C.C.), Rouen; Hopital de Purpan, Toulouse; Hopital D’Enfants, Vandoeuvre Les Nancy; Germany: Universitatsklinikum Rudolf Virchow, Berlin; Zentrum fur Knochenmark Transplantation, Hamburg; Medizinische Hochschule Hannover, Hannover; Christian-Albrechts-University, Kiel, Kiel; Universitat Munchen, Munchen; Ulm University Hospital, Ulm; Hungary: National Institute of Hematology, Budapest; Ireland: St. James’s Hospital, Dublin; Italy: Spedali Civili-Brescia, Brescia; Ospedale San Martino, Genoa; Centro Trapianti Midollo Osseo via fonte Romana, Pescara; Ospedale San Camillo, Roma; Ospedale Molinette, Torino; Japan: Chiba University School of Medicine, Chuo-Ku; Osaka Medical Centre for Cancer & Cardiovascular Diseases, Osaka:Korea: Catholic University Medical College, Yongdungpo-Gu;New Zealand: University of Auckland Hospital, Auckland; Canterbury Health Laboratory, Christchurch-NZ; Russia: Clinical Hospital Number 6, Moscow; Saudi Arabia: King Faisal Hospital, Riyadh; Spain: Hospital General Vall d’Hebron, Barcelona; Hospital Sant Pau, Barcelona; Postgraduate School of Haematology, Barcelona; Hospital de la Princesa, Madrid; Hospital Puerta de Hierro, Madrid; Hospital Marques de Valdecilla, Santander; Hospital La Fe, Valencia; Sweden: Huddinge Hospital, Huddinge; University Hospital of Lund, Lund; Switzerland: Kantonsspital, Basel; Kantonsspital Zurich, Zurich; Taiwan: Veteran’s General Hospital-Taipei, Taipei; The Netherlands: Leiden University Hospital, Leiden; Turkey: Ankara University Medical School, Ankara; United Kingdom: Birmingham Heartlands Hospital, Birmingham; The Queen Elizabeth Hospital, Birmingham; The Royal Infirmary of Edinburgh, Edinburgh; Glasgow Royal Infirmary, Glasgow; St. James University Hospital, Leeds; Royal Infirmary, Leicester; Imperial College School of Medicine, London; Royal Free Hospital, London; Royal Marsden Hospital, London; The London Clinic, London; Westminster Hospital, London; The Royal Victoria Infirmary, Newcastle Upon Tyne; Nottingham City Hospital, Nottingham; United States: Egleston Hospital for Children at Emory University, Atlanta, GA; Roswell Park Cancer Institute, Buffalo, NY; Medical University of South Carolina, Charleston, SC; University of Virginia Charlottesville, Charlottesville, VA; Children’s Hospital Medical Center, Cincinnati, OH; De University Medical Centre, Durham, NC; University of Florida, Gainesville, FL; St. Francis Medical Centre, Honolulu, HI; MD Anderson Cancer Center, Houston, TX; Indiana University Medical Center & Outpatient Center, Indianapolis, IN; University of Kansas Medical Center, Kansas City, KS; UCLA Center for Health Sciences, Los Angeles, CA; University of Louisville, Louisville, KY; University of Wisconsin Hospital & Clinics, Madison, WI; University of Minnesota Hospital & Clinics, Minneapolis, MN; Louisiana State University Medical Center, New Orleans, LA; University of Oklahoma Health Sciences Center, Oklahoma City, OK; University of Nebraska Medical Center, Omaha, NE; St. Joseph’s Hospital & Medical Center, Paterson, NJ; Intermountain Health Care, Inc, LDS Hospital, Salt Lake City, UT; Wilford Hall Medical Center, San Antonio, TX; Mayo Clinic Scottsdale, Scottsdale, AZ; George Washington University Medical Center, Washington, DC; Georgetown University Medical Center, Washington, DC.

1
Gluckman
E
Immunosuppressive drugs for the prevention of acute graft-vs-host disease
Graft-vs-Host Disease Immunology, Pathophysiology and Treatment.
Burakoff
SJ
Deeg
HJ
Ferrara
J
Atkinson
K
1990
389
Marcel Dekker
New York, NY
2
Atkinson
K
Treatment of extensive human chronic graft-vs-host disease
Graft-vs-Host Disease Immunology, Pathophysiology and Treatment.
Burakoff
SJ
Deeg
HJ
Ferrara
J
Atkinson
K
1990
681
Marcel Dekker
New York, NY
3
Reisner
Y
Kapoor
N
Kirkpatrick
D
Pollack
MS
Dupont
B
Good
RA
O’Reilly
RJ
Transplantation for acute leukaemia with HLA-A and B nonidentical parental marrow cells fractionated with soybean agglutinin and sheep red blood cells.
Lancet
2
1981
327
4
Prentice
HG
Blacklock
HA
Janossy
G
Bradstock
KF
Skeggs
D
Goldstein
G
Hoffbrand AV: 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
5
Prentice
HG
Janossy
G
Price-Jones
L
Trejdosiewicz
LK
Panjwani
D
Graphakos
S
Ivory
K
Blacklock
HA
Gilmore
MJML
Tidman
N
Skeggs
DBL
Bail
S
Patterson
J
Hoffbrand
AV
Depletion of T lymphocytes in donor marrow prevents significant graft-versus-host disease in matched allogeneic leukaemic marrow transplant recipients.
Lancet
1
1984
472
6
Waldmann
H
Or
R
Hale
G
Weiss
L
Cividalli
G
Samuel
S
Manor
D
Brautbar
C
Polliack
A
Rachmilewitz
EA
Slavin
S
Elimination of graft versus host disease by in vitro depletion of alloreactive lymphocytes using a monoclonal rat anti-human lymphocyte antibody (CAMPATH-1).
Lancet
2
1984
483
7
Noga
SJ
Donnenberg
AP
Schwartz
CL
Strauss
LC
Civin
CI
Santos
GW
Development of a simplified centrifugation elutriation procedure for depletion of lymphocytes from human bone marrow.
Transplantation
41
1986
214
8
Filipovitch
AH
Vallera
DA
Youle
RJ
Haake
R
Blazar
BR
Arthur
D
Neville Ramsay
NK
McGlave
P
Kersey
JH
Graft-versus-host disease prevention in allogeneic bone marrow transplantation from histocompatible siblings. A pilot study using immunotoxins for T cell depletion of donor bone marrow.
Transplantation
44
1987
62
9
Hale
G
Campath-1 Antigen (CD52)
Encyclopedia of Immunology London, UK, Academic
Delves
PJ
Roitt
I
1998
402
10
Hale
G
Bright
S
Chumbley
G
Hoang
T
Metcalf
D
Munro
AJ
Waldmann
H
Removal of T cells from bone marrow for transplantation: A monoclonal antilymphocyte antibody that fixes human complement.
Blood
62
1983
873
11
Heit
W
Bunjes
D
Weisneth
M
Schmeiser
T
Arnold
R
Hale
G
Waldmann
H
Heimpel
H
Ex vivo T-cell depletion with the monoclonal antibody Campath-1 plus human complement effectively prevents acute GvHD in allogeneic bone marrow transplantation.
Br J Haematol
64
1986
479
12
Hale
G
Cobbold
S
Waldmann
H
T cell depletion with CAMPATH-1 in allogeneic bone marrow transplantation.
Transplantation
45
1988
753
13
Hale
G
Waldmann
H
for CAMPATH users
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
14
Hale
G
Waldmann
H
for CAMPATH users
CAMPATH-1 monoclonal antibodies in bone marrow transplantation.
Hematotherapy
3
1994
15
15
Bunjes
D
Heit
W
Arnold
R
Schmeiser
T
Wiesneth
M
Carbonell
F
Porzsolt
F
Raghavachar
A
Heimpel
H
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
16
Kernan
NA
Flomenberg
N
Dupont
B
O’Reilly
RJ
Graft rejection in recipients of T-cell depleted HLA-identical marrow transplants for leukemia. Identification of host-derived anti-donor allocytotoxic T lymphocytes.
Transplantation
43
1987
842
17
Apperley
JF
Mauro
FR
Goldman
JM
Gregory
W
Arthur
CK
Hows
J
Arcese
W
Papa
G
Mandelli
F
Wardle
D
Gravett
P
Franklin
I
Bandini
G
Ricci
P
Tura
S
Iacone
A
Torlontano
G
Heit
W
Champlin
R
Gale
RP
Bone marrow transplantation for chronic myeloid leukaemia in first chronic phase: Importance of a graft-versus-leukaemia effect.
Br J Haematol
9
1988
239
18
Wagemaker
G
Vriesendorp
HM
van Bekkum
DW
Successful bone marrow transplantation across major histocompatibility barriers in rhesus monkeys.
Transplant Proc
13
1981
875
19
Vriesendorp
HM
Klapwijk
WM
Heidt
DJ
Hogeweg
B
Zurcher
C
van Bekkum
DW
Factors controlling the engraftment of transplanted dog bone marrow cells.
Tissue Antigens
20
1982
63
20
Patterson
J
Prentice
HG
Brenner
MK
Gilmore
M
Janossy
G
Ivory-K
Skeggs
D
Morgan
H
Lord
J
Blacklock
HA
Hoffbrand
AV
Apperley
JF
Goldman
JM
Burnett
A
Gribben
J
Alcorn
M
Pearson
C
McVickers
I
Hann
IM
Reid
C
Wardle
D
Gravett
PJ
Bacigalupo
A
Robertson
AG
Graft rejection following HLA matched T-lymphocyte depleted bone marrow transplantation.
Br J Haematol
63
1986
221
21
Slavin
S
Or
R
Naparstek
E
Cividalli
G
Weshler
Z
Weiss
L
Mumcuoglu
M
Engelhard
D
Aker
M
Pollack
A
Ben-Yehuda
A
Brautbar
C
Hale
G
Waldmann
H
New approaches for the prevention of rejection and graft-vs.-host disease in clinical bone marrow transplantation.
Isr J Med Sci
22
1986
264
22
Cobbold
SP
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
23
Dyer
MJS
Hale
G
Hayhoe
FGJ
Waldmann
H
Effects of CAMPATH-1 antibodies in vivo in patients with lymphoid malignancies: Influence of antibody isotype.
Blood
73
1989
1431
24
Hale
G
Clark
M
Waldmann
H
Therapeutic potential of rat monoclonal antibodies: Isotype specificity of antibody-dependant cell-mediated cytotoxicity with human lymphocytes.
J Immunol
134
1985
3056
25
Bunjes
D
Hertenstein
B
Wiesneth
M
Stefanic
M
Novotny
J
Danoker
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 leukaemia in first remission without increasing the risk of treatment failure: Comparison with cyclosporin/methotrexate.
Bone Marrow Transplant
15
1995
563
26
Oakhill
A
Pamphilon
DH
Potter
MN
Steward
CG
Goodman
S
Green
A
Goulden
P
Hale
G
Cornish
JMM
Unrelated donor bone marrow transplantation for children with relapsed acute lymphoblastic leukemia in second complete remission.
Br J Haematol
94
1996
574
27
Hale
G
Cobbold
SP
Waldmann
H
Easter
G
Matejtschuk
P
Coombs
RRA
Isolation of low-frequency class-switch variants from rat hybrid myelomas.
J Immunol Methods
103
1987
59
28
Bridges
J
Dowding
C
Galton
J
Hows
J
Comparison of in vitro cytolytic activity of two preparations of CAMPATH-1M.
XIV Annual Meeting of the European Cooperative Group for Bone Marrow Transplantation. Chamonix, France, April 10-13
1988
163
(abstr)
29
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-1M plus complement as demonstrated by limiting dilution analysis.
Bone Marrow Transplant
4
1989
55
30
Nash
RA
Pepe
MS
Storb
R
Longton
G
Pettinger
M
Anasetti
C
Appelbaum
FR
Bowden
RA
Deeg
HJ
Doney
K
Martin
PJ
Sullivan
K
Sanders
J
Witherspoon
RP
Acute graft-versus-host disease: Analysis of risk factors after allogeneic marrow transplantation and prophylaxis with cyclosporine and methotrexate.
Blood
80
1992
1838
31
Cox
DR
Regression analysis and life tables (with discussion).
J R Stat Soc B
34
1972
187
32
Klein
JP
Moeschberger
ML
Survival Analysis: Techniques for Censored and Truncated Data.
1997
Springer Verlag
New York, NY
33
Theobald
M
Hoffmann
T
Bunjes
D
Heit
W
Comparative analysis of in vivo T cell depletion with radiotherapy, combination chemotherapy and the monoclonal antibody CAMPATH-1G, using limiting dilution methodology.
Transplantation
49
1990
553
34
Moreau
T
Coles
A
Wing
MG
Isaacs
J
Hale
G
Waldmann
H
Compston
A
Transient increase in symptoms associated with cytokine release in patients with multiple sclerosis.
Brain
119
1996
225
35
Wing
MG
Moreau
T
Greenwood
J
Smith
R
Hale
G
Isaacs
J
Waldmann
H
Lachmann
PJ
Compston
DAS
Mechanism of first-dose cytokine release syndrome by CAMPATH-1H: Involvement of CD16 (FcγRIII) and CD11a (LFA-1) on NK cells.
J Clin Invest
98
1996
2819
36
Gerritsen
WR
Wagemaker
G
Jonker
M
Kenter
MJH
Wielenga
JJ
Hale
G
Waldmann
H
van Bekkum
DW
The repopulation capacity of bone marrow grafts following pretreatment with monoclonal antibodies against T lymphocytes in rhesus monkeys.
Transplantation
45
1988
301
37
Starobinski
M
Roosnek
E
Hale
G
Roux
E
Helg
C
Chapuis
B
T-cell depletion of allogeneic peripheral blood stem cells (letter).
Bone Marrow Transplant
21
1998
429
38
Horowitz
MM
Gale
RP
Sondel
P
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
39
Gale
RP
Horowitz
MM
How best to analyze new strategies in bone marrow transplantation.
Bone Marrow Transplant
6
1990
357
40
Frassoni
F
Labopin
M
Gluckman
E
Prentice
HG
Vernant
JP
Zwaan
F
Granena
A
Gahrton
G
De-Witte
T
Grathohl
A
Reiffers
J
Gorin
NC
Results of allogeneic bone marrow transplantation for acute leukemia have improved in Europe with time—A report of the acute leukemia working party of the European group for blood and marrow transplantation (EBMT).
Bone Marrow Transplant
17
1996
13
41
Parreira
A
Smith
J
Hows
JM
Smithers
SA
Apperley
J
Rombos
Y
Goldman
JM
Gordon-Smith
EC
Catovsky
D
Immunological reconstitution after bone marrow transplant with Campath-1 treated bone marrow.
Clin Exp Immunol
67
1987
142
42
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
43
Lowdell
MW
Craston
R
Ray
N
Koh
M
Galatowicz
G
Prentice
HG
The effect of T cell depletion with Campath-1M on immune reconstitution after chemotherapy and allogeneic bone marrow transplant as treatment for leukaemia.
Bone Marrow Transplant
21
1998
679
44
Hertenstein
B
Hampl
W
Bunjes
D
Wiesneth
M
Duncker
C
Koszinowski
U
Heimpel
H
Arnold
R
Mertens T: 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
38
45
Condiotti
R
Nagler
A
Campath-1G impairs human natural killer (NK) cell-mediated cytotoxicity.
Bone Marrow Transplant
18
1996
713
46
Hokland
M
Jacobsen
N
Ellegaard
J
Hokland
P
Natural killer function following allogeneic bone marrow transplantation. Very early re-emergence but strong dependence of cytomegalovirus infection.
Transplantation
45
1988
1080
47
Zeis
M
Uharek
L
Glass
B
Steinmann
J
Dreger
P
Gassmann
W
Schmitz
N
Allogeneic MHC-mismatched activated natural killer cells administered after bone marrow transplantation provide a strong graft-versus-leukaemia effect in mice.
Br J Haematol
96
1997
757
48
Rooney
CM
Wimperis
JZ
Brenner
MK
Patterson
J
Hoffbrand
AV
Prentice
HG
Natural killer cell activity following T-cell depleted allogeneic bone marrow transplantation.
Br J Haematol
62
1986
413
49
Reittie
JE
Gottlieb
D
Heslop
HE
Leger
O
Hazlehurst
G
Drexler
HG
Hoffbrand
AV
Prentice
HG
Brenner
MK
Endogenously generated activated killer cells circulate after autologous and allogeneic marrow transplantation but not after chemotherapy.
Blood
73
1989
1351
50
Lowdell
MW
Ray
N
Craston
R
Corbett
T
Deane
M
Prentice
HG
The in vitro detection of anti-leukaemia-specific cytotoxicity after autologous bone marrow transplantation for acute leukemia.
Bone Marrow Transplant
19
1997
891
51
Jiang
YZ
Barrett
AJ
Goldman
JM
Mavroudis
DA
Association of natural killer cell immune recovery with a graft-versus-leukemia effect independent of graft-versus-host disease following allogeneic bone marrow transplantation.
Ann Hematol
74
1997
1
52
Hale
G
Waldmann
H
for CAMPATH users
Risks of developing EBV-related lymphoproliferative disorders following T cell depleted marrow transplants.
Blood
91
1998
1
53
Willemze
R
Richel
DJ
Falkenberg
JHF
Hale
G
Waldmann
H
Zwaan
FE
Fibbe
WE
In vivo use of CAMPATH-1G to prevent graft-versus-host disease and graft rejection after bone marrow transplantation.
Bone Marrow Transplant
9
1992
255
54
Hamblyn
M
Marsh
JCW
Lawler
M
McCann
SR
Dunlop
L
Ball
S
Davies
G
Hale
G
Waldmann
H
Gordon-Smith
EC
CAMPATH-1G in vivo confers a low incidence of graft-versus-host disease associated with a high incidence of mixed chimerism after BMT for severe aplastic anaemia using HLA sibling donors.
Bone Marrow Transplant
17
1996
819
55
Jacobs
P
Wood
L
Fullard
L
Waldmann
H
Hale
G
T-cell depletion by exposure to CAMPATH-1G in vitro prevents graft-versus-host disease.
Bone Marrow Transplant
13
1993
763
Sign in via your Institution