Although nonmyeloablative conditioning regimen transplantations (NMTs) induce engraftment of allogeneic stem cells with a low spectrum of toxicity, graft-versus-host disease (GVHD) remains a significant cause of morbidity and mortality. In vivo T-cell depletion, using alemtuzumab, has been shown to reduce the incidence of GVHD. However, this type of maneuver, although reducing GVHD, may have an adverse impact on disease response, because NMTs exhibit their antitumor activity by relying on a graft-versus-malignancy effect. To explore the efficacy of alemtuzumab compared with methotrexate (MTX) for GVHD prophylaxis, we have compared the results in 129 recipients of a sibling NMT enrolled in 2 prospective studies for chronic lymphoproliferative disorders. Both NMTs were based on the same combination of fludarabine and melphalan, but the United Kingdom regimen (group A) used cyclosporin A plus alemtuzumab, whereas the Spanish regimen (group B) used cyclosporin A plus MTX for GVHD prophylaxis. Patients receiving alemtuzumab had a higher incidence of cytomegalovirus (CMV) reactivation (85% versus 24%,P < .001) and a significantly lower incidence of acute GVHD (21.7% versus 45.1%, P = .006) and chronic GVHD (5% versus 66.7%, P < .001). Twenty-one percent of patients in group A and 67.5% in group B had complete or partial responses 3 months after transplantation (P < .001). Eighteen patients in group A received donor lymphocyte infusions (DLIs) to achieve disease control. At last follow-up there was no difference in disease status between the groups with 71% versus 67.5% (P = .43) of patients showing complete or partial responses in groups A and B, respectively. No significant differences were observed in event-free or overall survival between the 2 groups. In conclusion, alemtuzumab significantly reduced GVHD but its use was associated with a higher incidence of CMV reactivation. Patients receiving alemtuzumab often required DLIs to achieve similar tumor control but the incidence of GVHD was not significantly increased after DLI.

Nonmyeloablative transplants (NMTs) are currently being evaluated in patients with hematologic malignancies who are considered poor candidates for conventional allogeneic transplantation because of their advanced age or other concurrent medical conditions. The rationale for NMTs relies on previous observations that adoptive transfer of alloreactive donor lymphocytes may eradicate refractory or recurrent disease.1-5Therefore, in an effort to reduce the toxicity associated with high-dose chemotherapy,6-11 several groups have designed less toxic regimens that might exhibit their antitumor effect relying on a graft-versus-malignancy effect rather than on myeloablative chemotherapy.12-16 Although these regimens have been shown to permit engraftment of allogeneic hemopoietic stem cells with a low spectrum of toxicity, graft-versus-host disease (GVHD) remains a significant cause of increased morbidity and mortality.4-20 The ideal approach for GVHD prophylaxis remains uncertain and several agents have been introduced to reduce incidence and severity. The United Kingdom collaborative group has incorporated alemtuzumab (Campath-1H) as part of a fludarabine-based protocol and a low incidence of GVHD has been reported in both related and unrelated donor transplantation.21 However, this type of intervention to reduce the incidence and severity of GVHD may have an adverse impact on disease response, because several studies have suggested a positive correlation between GVHD and graft-versus-leukemia (GVL) effect,6,15,16,19,22-25 both in myeloid and in lymphoid malignancies18,20,25,26 and higher relapse rates have been reported after syngeneic or T-cell–depleted transplantation.27,28 Thus, the ideal approach is to establish the optimum balance between the risk and benefit of decreasing GVHD versus an increase in the risk of relapse.

To explore the efficacy of alemtuzumab compared with methotrexate (MTX; both in combination with cyclosporine) for GVHD prophylaxis and its impact on the outcome of patients receiving transplants for lymphoproliferative disorders, we have retrospectively compared the results of 2 prospective studies carried out in the United Kingdom21 and Spain.29 Both NMT schedules were based on the same combination of fludarabine and melphalan, but the United Kingdom regimen (group A) used cyclosporin A (CsA) plus alemtuzumab, whereas the Spanish regimen (group B) used CsA plus MTX. Patients included in this study underwent transplantation from February 1998 to January 2001 (group A) and from February 1999 to May 2001 (group B).

Patient characteristics

We have analyzed data on 129 patients with chronic lymphoproliferative disorders who were included in both registries and received a peripheral blood stem cell transplant from HLA-matched siblings. Both studies included patients considered candidates to receive a transplant from a related donor but who had a high risk of transplantation-related mortality (TRM) due to advanced age (≥ 45 years old, n = 77) or other concurrent medical condition (relapse after a previous autologous transplantation, n = 30; severe organ disfunction, active fungal infection, or heavily pretreated patients, n = 22). Patients gave written informed consent for inclusion in the protocols, which were approved by all local ethical review boards as well as the Spanish (protocol 99-0151) and the United Kingdom (study no. 974/0127) drug agencies.

No significant differences were observed between the 2 groups in median age, diagnosis, or previous transplantation. Disease status at transplantation was categorized as low risk (patients in CR1 and CR2), high risk (patients with refractory or progressive disease and more than first partial response [PR]) and intermediate risk (the rest of cases, ie, more than second complete response [CR]); no differences were observed in terms of disease status. In contrast, there was a sex mismatch in 56% of the alemtuzumab group (A) compared with 35% in MTX group (B; P = .031). In addition, in 37% of cases in group A both donor and recipient were cytomegalovirus (CMV) negative compared with only 9% in group B (P = .009). Other patient characteristics are specified in Table1.

Table 1.

Patient and disease characteristics

Fludarabine/melphalan/alemtuzumab (n = 78)Fludarabine/melphalan/MTX (n = 51)P
Age, y (median, SD) 44  (10,7) 47  (11,3) .94 
Previous transplantation 35  (45%) 26  (51%) .49 
Sex mismatched 34  (55.7%) 18  (35.3%) .031 
Patient CMV+ 37  (53%) 27  (81.8%)  
Patient CMV donor CMV+ 7  (10%) 3  (9.1%) .009  
Both CMV 26  (37%) 3  (9.1%)  
Disease status at transplantation categorized*    
 Low risk 7  (9.1%) 7  (13.7%) .25 
  CR1  6  2  
  CR2  1  5  
 Intermediate risk 53  (67.5%) 27  (52.9%)  
  More than CR2  7  2  
  1st PR 44 18  
  CR1/2 after 3rd line  2   
  Untreated relapse/SD treatment   7  
 High risk 18  (23.4%) 17  (33.3%)  
  Refractory/progressive 15 13  
  More than 1st PR  3  4  
Disease    
 HD 15 10 .52 
 CLL  8  7  
 MM 19 16  
 MCL  6    
 Low-grade NHL 11  7  
 High-grade NHL 12  4  
 T-NHL  2  4  
 OCLPDs  7  4  
Follow-up (median, SD) 400  (273) 253  (200) .05 
Fludarabine/melphalan/alemtuzumab (n = 78)Fludarabine/melphalan/MTX (n = 51)P
Age, y (median, SD) 44  (10,7) 47  (11,3) .94 
Previous transplantation 35  (45%) 26  (51%) .49 
Sex mismatched 34  (55.7%) 18  (35.3%) .031 
Patient CMV+ 37  (53%) 27  (81.8%)  
Patient CMV donor CMV+ 7  (10%) 3  (9.1%) .009  
Both CMV 26  (37%) 3  (9.1%)  
Disease status at transplantation categorized*    
 Low risk 7  (9.1%) 7  (13.7%) .25 
  CR1  6  2  
  CR2  1  5  
 Intermediate risk 53  (67.5%) 27  (52.9%)  
  More than CR2  7  2  
  1st PR 44 18  
  CR1/2 after 3rd line  2   
  Untreated relapse/SD treatment   7  
 High risk 18  (23.4%) 17  (33.3%)  
  Refractory/progressive 15 13  
  More than 1st PR  3  4  
Disease    
 HD 15 10 .52 
 CLL  8  7  
 MM 19 16  
 MCL  6    
 Low-grade NHL 11  7  
 High-grade NHL 12  4  
 T-NHL  2  4  
 OCLPDs  7  4  
Follow-up (median, SD) 400  (273) 253  (200) .05 
*

Low risk: CR1, CR2; high risk: refractory, progressive, more than 1st PR.

CR1 indicates 1st complete remission; CR2, 2nd complete remission; 1st PR, first partial response; SD, stable disease; HD, Hodgkin disease; CLL, chronic lymphoid leukemia; MM, multiple myeloma; MCL, mantle cell lymphoma; and OCLPDs, other chronic lymphoproliferative disorders, including Waldenstrom (n = 1/1), T-prolymphocytic leukemia (n = 1), low-grade transformed to high-grade HNL (n = 5/1), Richter (n = 0/2).

Mobilization regimens and peripheral blood stem cell collection procedures were similar in both trials.21,29 

Conditioning regimen and GVHD prophylaxis

In both trials the conditioning regimen consisted of fludarabine 30 mg/m2 intravenously for 5 days followed by melphalan 140 mg/m2 intravenously in 1 dose or divided in 2 doses: fludarabine 30 mg/m2 days −7 to −3 plus melphalan 140 mg/m2 on day −2 in group A and fludarabine 30 mg/m2 days −8 to −4 plus melphalan 140 mg/m2divided in 2 doses on days −3 and −2 in group B.21,29 

Acute and chronic GVHD were similarly graded by established criteria in both trials.30,31 In group A, GVHD prophylaxis consisted of CsA 3 mg/kg intravenously starting on day −1 and switched to an oral dose as soon as the patient could tolerate oral medications. The median duration of administration was 4 months for the first 40 patients; however, the observed low incidence of GVHD led physicians to taper the dose by the end of the second month for the rest of the patients. In patients without evidence of GVHD, CsA was discontinued by the end of the third month. alemtuzumab was administered at a dose of 20 mg/d by intravenous infusion over 8 hours on days −8 to −4.

In group B, CsA was started on day −7. In the absence of grade II or greater acute GVHD, CsA was tapered 10% weekly starting on day +90 and discontinued by day +150. In both groups CsA levels were monitored starting on day +1 and were maintained in the therapeutic range (150-300 ng/mL) until tapering. MTX in group B was given at a dose of 10 mg/m2 intravenously on days +1, +3, and +6, followed by folinic acid rescue (10 mg intravenously every 6 hours for 4 doses starting 24 hours after each dose of MTX).

Patients with relapsed/progressive disease after transplantation or those not evolving to 100% donor chimerism after discontinuing immunosuppression were considered eligible for donor lymphocyte infusion (DLI) in both studies.

An escalated dose regimen, starting at 1 × 106CD3+ T cells/kg with dose escalation at intervals of 3 months, was used in group A. The median interval from transplantation to the first DLI was 230 days (range, 182-781 days). Overall, 18 patients received DLI in group A. Fifteen patients received DLI for persistent disease after transplantation and for mixed chimerism. In group B, also an escalated dose regimen at intervals of 3 months was used; however, the starting dose was 1 × 107 CD3+ T cells/kg. In this group, 4 patients received DLI due to persistent disease (n = 3) or progression (n = 1). The median interval to start DLI after transplantation was +210 days (range, 120-243 days). Data on chimerism analysis and DLI (S.M., manuscript in preparation) have already been reported.21,29 Protocols for the prevention and treatment of transplantation complications and supportive care were similar in both groups.21,29 

Finally, patients were monitored for CMV reactivation by weekly CMV antigenemia (group B) or polymerase chain reaction (PCR) assay (group A) and preemptive therapy with ganciclovir32,33 was started after one positive antigenemia (group A) or after 2 consecutive positive PCR assays (group B).

Statistical methods

Comparison of relapse rate and GVHD between groups was performed using the χ2 and Fisher exact test. Events analyzed were calculated from the time of transplantation using Kaplan-Meier product-limit estimates. For TRM, patients were censored at last follow-up. Disease-free survival (DFS) was calculated from transplantation until disease progression for those patients reaching CR after transplantation and also for those reaching CR after DLI. Patients who suffered TRM and those who were still alive without progression at the time of reporting were censored at death and last follow-up, respectively. Event-free survival (EFS) was calculated from transplantation until disease progression or death and those patients who did not reach disease response (CR or PR) any time after transplantation were considered events. Overall survival (OS) was calculated from transplantation until death from any cause, and surviving patients were censored at last follow-up. The incidence and time to onset of chronic GVHD were calculated in patients followed for at least 90 days.

Engraftment

Both regimens induced an early and sustained engraftment.21,28 As shown in Table2, granulocyte recovery was faster in group A patients; the median time for more than 500 granulocytes/mm3 was 12.8 versus 14.7 days (P < .001) and for more than 1000 granulocytes/mm3 13.7 versus 17.6 days (P < .001) for groups A and B, respectively. In contrast, the use of alemtuzumab was associated with a significant delay in platelet recovery: median of 14.4 versus 11.2 days to reach more than 20 × 109/L platelets (P = .009) and 21.7 versus 14.1 days to reach more than 50 × 109/L platelets (P = .001) in groups A and B, respectively.

Table 2.

Transplantation-related morbidity and mortality

Fludarabine/melphalan/alemtuzumab (n = 78)Fludarabine/melphalan/MTX (n = 51)P
Days to reach    
 More than 500 granulocytes 12.88  (2.84) 14.79  (2.14) < .001  
 More than 1 000 granulocytes 13.7  (3.89) 17.68  (3.09) < .001 
 More than 20 000 platelets 14.49  (6.49) 11.25  (5.34)  .009 
 More than 50 000 platelets  21.73  (14.1) 14.14  (5.38)  .001  
Acute GVHD   Yes vs no .001  
 No 64  (82.1%) 28  (54.9%) 0-I vs II-IV 
 Grade I 9  (11.5%) 1  (2%) < .001  
 Grade II 5  (6.4%) 15  (29.4%) 0-II vs III-IV  
 Grade III-IV 2  (2.6%) 7  (13.7%)  .015  
Overall acute GVHD (before + after DLI)   Yes vs no .006  
 No 60  (76.9%) 28  (54.9%) 0-I vs II-IV  
 Grade I 10  (12.8%) 1  (2%) < .001  
 Grade II 3  (3.8%) 15  (29.4%) 0-II vs III-IV  
 Grade III-IV 4  (5.1%) 7  (13.7%)  .092  
Chronic GVHD    
 Limited  9 < .001 
 Extensive 13  
CMV reactivation 27  (46.6%) 10  (22.7%)  .018 
 CMV+ patient or donor 26  (86.7%) 7  (25%) < .001  
 CMV patient and donor  0  
TRM 8  (10.3%) 10  (20%)  .12 
Fludarabine/melphalan/alemtuzumab (n = 78)Fludarabine/melphalan/MTX (n = 51)P
Days to reach    
 More than 500 granulocytes 12.88  (2.84) 14.79  (2.14) < .001  
 More than 1 000 granulocytes 13.7  (3.89) 17.68  (3.09) < .001 
 More than 20 000 platelets 14.49  (6.49) 11.25  (5.34)  .009 
 More than 50 000 platelets  21.73  (14.1) 14.14  (5.38)  .001  
Acute GVHD   Yes vs no .001  
 No 64  (82.1%) 28  (54.9%) 0-I vs II-IV 
 Grade I 9  (11.5%) 1  (2%) < .001  
 Grade II 5  (6.4%) 15  (29.4%) 0-II vs III-IV  
 Grade III-IV 2  (2.6%) 7  (13.7%)  .015  
Overall acute GVHD (before + after DLI)   Yes vs no .006  
 No 60  (76.9%) 28  (54.9%) 0-I vs II-IV  
 Grade I 10  (12.8%) 1  (2%) < .001  
 Grade II 3  (3.8%) 15  (29.4%) 0-II vs III-IV  
 Grade III-IV 4  (5.1%) 7  (13.7%)  .092  
Chronic GVHD    
 Limited  9 < .001 
 Extensive 13  
CMV reactivation 27  (46.6%) 10  (22.7%)  .018 
 CMV+ patient or donor 26  (86.7%) 7  (25%) < .001  
 CMV patient and donor  0  
TRM 8  (10.3%) 10  (20%)  .12 

GVHD and infectious complications

Patients receiving alemtuzumab had a significantly lower incidence of acute GVHD (20.5%) when compared with patients receiving MTX (45.1%; P = .001). In addition, in patients receiving alemtuzumab the incidence of grades II to IV and III to IV acute GVHD was significantly lower compared with patients receiving MTX: 9% versus 43.1% grades II to IV acute GVHD in groups A and B, respectively (P < .001) and 2.6% versus 13.7% grades III to IV acute GVHD in groups A and B, respectively (P = .015). Interestingly, 18 patients received DLI in group A due to relapse/persistence of disease or mixed chimerism and only 3 of these patients developed GVHD (1 each grade I, grade IV, and limited chronic GVHD), so that a significantly lower incidence of acute GVHD persisted in patients receiving alemtuzumab even after DLI. However, there was no significant difference in terms of severe acute GVHD (grades III and IV) after DLI (P = .09; Table2).

There were also differences in the incidence of chronic GVHD between the groups. Only 2 patients receiving alemtuzumab developed chronic GVHD (1 limited, 1 extensive) in contrast to 22 (66.7% of patients at risk) patients receiving MTX (13 cases of extensive chronic GVHD;P < .001).

Patients were monitored for CMV reactivation by weekly CMV antigenemia (group B) or PCR assay (group A). Statistically significant differences were observed in the incidence between both groups. Although a lower percentage of patients in group B were CMV, only 22.7% developed CMV reactivation as compared with 47% in group A (P = .018). Even more strikingly, among CMV+patients, 87% had CMV reactivation in group A versus 25% in group B (P < .001). Despite the high incidence of reactivation only one patient died of CMV disease in group A and none in group B.

TRM

No significant differences were observed in terms of TRM between the groups, although a trend toward a higher mortality rate was observed in group B (10.2% versus 20% for groups A and B, respectively; P = .12; log-rank, P = .06; Figure 1).

Fig. 1.

Transplantation-related mortality.

Graph measures days from transplantation to death.

Fig. 1.

Transplantation-related mortality.

Graph measures days from transplantation to death.

Close modal

Overall 25.6% of patients in group A died compared with 29.4% in group B (P = .6); 33.4% of deaths in group B were related to GVHD compared with 5% in group A. By contrast, the most frequent cause of death in patients receiving alemtuzumab was disease relapse/progression (60% of deaths versus 20%). Data concerning morbidity and mortality are specified in Table 2.

Disease response

Disease status was evaluated 3 months after transplantation (Table3). After 3 months, 21.1% of patients in group A were in CR/PR in contrast to 67.5% in group B (P < .001). Interestingly, disease status at transplantation significantly influenced response to transplantation in patients receiving alemtuzumab but not in those receiving MTX. The most important differences in response were observed in patients categorized as high risk according to disease status before transplantation with no patient reaching CR or PR in group A versus 52.9% in group B (P < .001). In the intermediate-risk group of patients, 18.8% reached CR or PR in group A compared with 55.5% in group B (P < .001). In contrast, no significant differences were observed in the low-risk group, although the number of patients in this subgroup was too small to reach any conclusion. As previously stated, 18 patients received DLI in group A because of relapse/persistence of disease or mixed chimerism versus 4 patients in group B. Disease status at last follow-up (after CsA taper and DLI) is summarized in Table 3. Disease status at last follow-up was not significantly different between both groups for the overall series of patients. In addition, no significant differences were observed according to pretransplantation disease status, because in the high-risk group 53.8% of patients receiving alemtuzumab reached CR/PR versus 50% in the group of patients receiving MTX (P = .45). Also in the intermediate-risk group 55.8% of patients receiving alemtuzumab reached CR/PR compared with 56.5% of patients receiving MTX (P = .76).

Table 3.

Disease response and transplantation outcome

Fludarabine/
melphalan/
alemtuzumab (n = 78)
Fludarabine/
melphalan/MTX
(n = 51)
P
Disease response at 3 mo
after transplantation 
   
 CR 15  (21.1%) 19  (47.5%) < .001 
 PR 8  (20%)  
 Stable/progression free 48  (67.6%) 9  (22.5%)  
 Progression 8  (11.3%) 4  (10%)  
Disease status at last follow-up3-150    
 CR 28  (47.4%) 19  (47.5%) .43  
 PR 14  (23.7%) 8  (20%)  
 Stable/progression free 16  (27.1%) 9  (22.5%)  
 Progression 1  (1.8%) 4  (10%)  
Fludarabine/
melphalan/
alemtuzumab (n = 78)
Fludarabine/
melphalan/MTX
(n = 51)
P
Disease response at 3 mo
after transplantation 
   
 CR 15  (21.1%) 19  (47.5%) < .001 
 PR 8  (20%)  
 Stable/progression free 48  (67.6%) 9  (22.5%)  
 Progression 8  (11.3%) 4  (10%)  
Disease status at last follow-up3-150    
 CR 28  (47.4%) 19  (47.5%) .43  
 PR 14  (23.7%) 8  (20%)  
 Stable/progression free 16  (27.1%) 9  (22.5%)  
 Progression 1  (1.8%) 4  (10%)  
F3-150

Disease status after CsA taper and DLI.

On analyzing all patients (Table 4), we found an association between development of acute GVHD and disease response (55.2% of patients developing acute GVHD reached CR/PR compared with 29.5% for those who did not develop acute GVHD,P = .025) but this association disappeared after DLI. The same occurred when we analyzed patients developing grades II to IV acute GVHD. To evaluate whether these results could be attributed to any of the GVHD prophylaxis regimens both groups were separately analyzed. No clear relationship between acute GVHD and response to transplant or disease status after DLI was observed in either group A or B, although this might be related to the lower number of cases included, when both series were separately analyzed. In addition, 74% of patients developing chronic GVHD achieved CR/PR compared with 29% of patients who did not develop it (P < .001). In this case, the impact of DLI could not be evaluated because only 2 patients in group A developed chronic GHVD.

Table 4.

Response to transplantation according to GVHD development

YesNoP
Acute GVHD 
 Response to transplantation    
  CR/PR 16  (55.2%) 26  (29.5%) .025 
  Stable/progression 13  (44.8%) 56  (70.5%)  
 Disease status at last follow-up4-150    
  CR/PR 21  (75%) 47  (67.1%) .61  
  Stable/progression 7  (25%) 23  (32.9%)  
Chronic GVHD    
 Response to transplantation    
  CR/PR 17  (74%) 23  (29%) .016  
  Stable/progression 6  (23%) 57  (71%)  
YesNoP
Acute GVHD 
 Response to transplantation    
  CR/PR 16  (55.2%) 26  (29.5%) .025 
  Stable/progression 13  (44.8%) 56  (70.5%)  
 Disease status at last follow-up4-150    
  CR/PR 21  (75%) 47  (67.1%) .61  
  Stable/progression 7  (25%) 23  (32.9%)  
Chronic GVHD    
 Response to transplantation    
  CR/PR 17  (74%) 23  (29%) .016  
  Stable/progression 6  (23%) 57  (71%)  
F4-150

Disease status after CsA taper and DLI.

Finally, results from both registries were analyzed according to the type of underlying disease: in multiple myeloma both response to transplant and disease status after DLI were improved in patients receiving MTX (52.6% of patients in group A reached CR/PR at last follow-up compared with 68.8% in group B, P = .029). In patients with multiple myeloma there was no clear relation between acute GVHD and disease control; however, 50% of those patients who developed acute GVHD reached CR/PR compared with 25% for those who did not develop GVHD (P = .25).

In patients with Hodgkin disease, the initial response was better in group B; however, disease status after DLI was similar in both groups (53.3% of patients reached CR/PR in group A compared with 50% in group B, P = .21). Finally, within patients diagnosed with high-grade non-Hodgkin lymphoma (NHL; n = 21), only 2 were categorized as low-risk disease at transplantation, whereas the rest were categorized as intermediate (n = 9) and high risk (n = 10). Response to transplantation was significantly better for patients receiving MTX but, after examining disease status after DLI, no significant differences were observed between the 2 groups, although we observed a trend toward better disease control for patients in group B (26.7% of patients receiving alemtuzumab in CR/PR versus 50% for those receiving MTX). In view of the low number of patients no other disease categories were analyzed.

OS and DFS

Disease-free survival was first calculated identifying only those patients reaching a CR after transplantation. Median DFS was not reached for patients receiving alemtuzumab and not computed for those receiving MTX because no relapses were observed among patients reaching CR. No significant differences were found. When we also considered patients achieving CR after DLI, DFS at 2 years was 76% for patients in group A versus 100% in group B (P = .19). When we compared both groups according to pretransplantation disease status, no significant differences were found for any of the risk categories when we considered disease status after DLI.

Median EFS among patients receiving alemtuzumab was 438 days versus 267 days for patients receiving MTX. EFS at 2 years was 34% in group A versus 39% in group B (P = .14; Figure2). When both groups were examined according to pretransplantation disease status, no significant differences were found for any of the risk categories when we considered disease status after DLI. When we analyzed only those patients achieving any response (CR, PR, or stable disease), median EFS was not reached for any of the 2 groups. For these patients EFS at 2 years was 72% and 76% for groups A and B, respectively (P = .6; Figure 3).

Fig. 2.

Event-free survival.

Graph measures days from transplantation to death or relapse. Patients who did not reach CR or PR were considered events.

Fig. 2.

Event-free survival.

Graph measures days from transplantation to death or relapse. Patients who did not reach CR or PR were considered events.

Close modal
Fig. 3.

Event-free survival.

Graph measures days from transplantation to death or to relapse for patients who reached CR, PR, or SD after transplantation.

Fig. 3.

Event-free survival.

Graph measures days from transplantation to death or to relapse for patients who reached CR, PR, or SD after transplantation.

Close modal

With regard to OS, at the present time the median survival has not been reached for either of the 2 groups. No significant differences were observed between both groups (72% OS at 2 years in group A versus 66% in group B, P = .22) even when we considered the different risk categories (Figure 4).

Fig. 4.

Overall survival.

Graph measures days from transplantation to death.

Fig. 4.

Overall survival.

Graph measures days from transplantation to death.

Close modal

It has been well established that NMTs may offer a reduced risk of procedure-associated mortality,6,14-16,21,29 compared with conventional transplantation.8-10 However, despite the considerably lower toxicity, GVHD still remains a major limitation for this strategy.6,14-16,29 To explore the impact of alemtuzumab on the prevention of GVHD and the efficacy of NMT, we have performed the first comparative study between 2 prospective national studies in patients diagnosed with lymphoproliferative disorders.

Both conditioning regimens21,29 were based on the same doses of fludarabine and melphalan, with the only difference being the use of alemtuzumab (group A) or MTX (group B) in addition to CsA to prevent GVHD. Characteristics of patients at transplantation were similar, except for a higher incidence of sex-mismatched pairs in the group of patients receiving alemtuzumab and a lower percentage of CMV seronegativity in the group of patients receiving MTX.

This comparative study has confirmed that alemtuzumab significantly reduces the overall incidence of acute GVHD when compared with MTX. In addition, this reduction can be maintained even after DLI. Because the efficacy of this type of transplantation relies primarily on the graft-versus-tumor effect,6-10,24 any attempt to decrease GVHD may be offset by a higher risk of relapse and the results should be cautiously interpreted. Disease response at 3 months was significantly better in the group of patients receiving MTX. In addition, patients who developed GVHD obtained a better disease response after transplantation. Nevertheless, 18 patients in group A received DLI to generate a tumor response and our analysis has shown that disease status at last follow-up did not differ between both groups. Interestingly, disease response after DLI was not associated with a significant increase in GVHD because differences in GVHD incidence between both groups remained even after DLI. In addition, correlation between GVHD and response was not significant when we considered disease status after DLI. These results are in accordance with previous reports suggesting that the approach of using delayed DLI can induce disease response and may be associated with less GVHD compared with early infusion34 and also support previous reports showing that the use of DLI can contribute to overcome the negative effect on GVL effect of in vitro T-cell depletion using alemtuzumab in myeloid as well as in lymphoid malignancies.35 

This study also confirmed that lymphomas, chronic lymphoid leukemia, and multiple myeloma are susceptible to graft-versus-tumor effect.18,20,25,26,36-40 This effect is not equally potent across tumor subtypes and disorders with low proliferative capacity may respond better when compared to more aggressive forms of disease. We were also able to demonstrate a correlation between disease response and severity of GVHD and, not surprisingly, in our series of myeloma patients, response to transplantation was significantly better in group B. Interestingly, the improved outcome in group B remained even after DLI in group A, suggesting that an early onset of GVHD might be more beneficial in multiple myeloma.

Although the use of T-cell depletion combined with cyclosporine may have a deleterious effect on transplantation outcome,41recent reports have unexpectedly shown that Campath and cyclosporin can reduce the risk of death due to infections 6 months after the transplantation.42 Therefore, further studies will be needed to explore the mechanism of the cyclosporin effect and the potential benefit in combination with T-cell depletion.

As previously reported, the use of less intensive regimens may have a beneficial effect on infections.43 In the immediate posttransplantation period, the short duration of neutropenia may result in reduced incidence of bacterial infections. In the present study, alemtuzumab recipients experienced increased incidence of CMV reactivation compared with MTX, although only one patient in the former group died because of CMV disease. This difference in terms of CMV reactivation cannot be attributed to the different techniques used in both trials because several studies have demonstrated that antigenemia and PCR assay have similar predictive values and show a high linear correlation.44,45 According to these findings, the Centers for Disease Control and Prevention (CDC) have recently recommended that any of both techniques should be selected to determine the need for preemptive treatment.46 On the other hand, we were able to show that the use of MTX, as expected, was associated with delayed neutrophil engraftment, although unexpectedly, platelet engraftment was delayed in patients receiving alemtuzumab. These differences were not clinically important. The delay in platelet engraftment in group A might be related to the in vivo T-cell depletion induced by alemtuzumab, because T lymphocytes may be an important element for platelet engraftment,47 or in addition, alemtuzumab might have direct toxicity on megakaryocytic progenitors.

Although a major concern about the use of anti-T monoclonal antibodies is the increased incidence of posttransplantation lymphoproliferative disorders (PTLDs), in this study, none of the patients developed PTLDs. This is in contrast to previous reports on other anti-T monoclonal antibodies48,49 and is probably related to the anti–B-cell effect induced by alemtuzumab.

In conclusion, both nonmyeloablative regimens offer good results in patients considered poor candidates to undergo a conventional allogeneic transplantation, but a different pattern of complications was observed. Alemtuzumab significantly reduced GVHD but it was related to a higher incidence of CMV reactivation. These findings have prompted us to initiate a study to establish an optimum dose of alemtuzumab to maintain its efficacy for GVHD prophylaxis without increasing CMV reactivation.50 However, patients receiving alemtuzumab required DLI to achieve a similar response rate compared with those receiving MTX, but the incidence of GVHD did not significantly increase after DLI, thus suggesting the possibility of separating GVHD from GVL effect. Finally, better responses were observed early after transplantation in patients with multiple myeloma or with active disease before transplantation, indicating that these patients may benefit from MTX-containing regimens. Considering these results, a randomized trial comparing both protocols of GVHD prophylaxis is warranted.

The authors wish to thank Jose Manuel Garcı́a de Cecilia, Statistician of the SEK University (Segovia), for his support in the statistical analysis.

Prepublished online as Blood First Edition Paper, July 12, 2002; DOI 10.1182/blood-2002-03-0701.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Porter
DL
Roth
MS
McGarigle
C
Ferrara
JL
Antin
JH
Induction of graft versus host disease as immunotherapy for relapsed chronic myeloid leukemia.
N Engl J Med.
330
1994
100
106
2
Collins
RH
Jr
Shpilberg
O
Drobyski
WR
et al
Donor leukocyte infusions in 140 patients relapsed with malignancy after allogeneic bone marrow transplantation.
J Clin Oncol.
15
1997
433
444
3
Kolb
HJ
Schattenberg
A
Goldman
JM
et al
Graft versus leukemia effect of donor lymphocyte transfusions in marrow grafted patients.
Blood.
86
1995
2041
2050
4
Lockhorst
HM
Schattenberg
A
Cornelissen
JJ
Thomas
LLM
Verdonk
LF
Donor leukocyte infusions are effective in relapsed multiple myeloma after allogeneic bone marrow transplantation.
Blood.
90
1997
4206
4211
5
Kolb
HJ
Schattenberg
A
Goldman
JM
et al
Graft versus myeloma effect of donor lymphocyte transfusions in marrow grafted patients. European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia.
Blood.
86
1995
2041
2050
6
Pérez-Simón
JA
Caballero
D
Lopez-Pérez
R
et al
Chimerism and minimal residual disease monitoring after reduced intensity conditioning (RIC) allogeneic transplantation.
Leukemia.
16
2002
1423
1431
7
Bortin
MM
Horowitz
MM
Gale
RP
et al
Changing trends in allogeneic bone marrow transplantation for leukemia in the 1980s.
JAMA.
268
1992
607
612
8
Armitage
JO
Bone marrow transplantation.
N Engl J Med.
330
1994
827
838
9
Klingemann
HG
Storb
R
Fefer
A
et al
Bone marrow transplantation in patients aged 45 years and older.
Blood.
65
1986
770
776
10
Ringden
O
Horwitz
MH
Gale
RP
et al
Outcome after allogeneic bone marrow transplantation for leukemia in older patients.
JAMA.
270
1993
57
60
11
Blumme
KG
Forman
SJ
Nademanee
AP
et al
Bone marrow transplantation for hematologic malignancies in patients aged 30 years or older.
J Clin Oncol.
4
1986
1489
1492
12
Slavin
S
Weiss
L
Morecki
S
Weigensberg
M
Eradication of murine leukemia with histoincompatible marrow grafts in mice conditioned with total lymphoid irradiation.
Cancer Immunother.
11
1981
155
158
13
Storb
R
Yu
C
Wagner
JL
et al
Stable mixed hematopoietic chimerism in DLA-identical littermate dogs given sublethal total body irradiation before and pharmacological immunosuppression after marrow transplantation.
Blood.
89
1997
3048
3054
14
Slavin
S
Nagler
A
Naparstek
E
et al
Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases.
Blood.
91
1998
756
763
15
Giralt
S
Estey
E
Albitar
M
et al
Engraftment of allogeneic hematopoietic progenitor cells with purine analog-containing chemotherapy: harnessing graft versus leukemia without myeloablative therapy.
Blood.
89
1997
4531
4536
16
McSweeney
P
Niederwieser
D
Shizuru
J
et al
Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high dose cytotoxic therapy with graft versus tumor effect.
Blood.
97
2001
3390
3400
17
Giralt
S
Thall
P
Khouri
I
et al
Melphalan and purine analog-containing preparative regimens: reduced-intensity conditioning for patients with hematologic malignancies undergoing allogeneic progenitor cell transplantation.
Blood.
97
2001
631
637
18
Khouri
IF
Keating
M
Körbling
M
et al
Transplant-lite induction of graft-versus-malignancy using fludarabine based nonablative chemotherapy and allogeneic blood progenitor cell transplantation as treatment for lymphoid malignancies.
J Clin Oncol.
16
1998
2817
2824
19
Childs
R
Clave
E
Contentin
N
et al
Engraftment kinetics after nonmyeloablative allogeneic peripheral blood stem cell transplantation: full donor T-cell chimerism precedes alloimmune responses.
Blood.
94
1999
3234
3241
20
Badros
A
Barlogie
B
Morris
C
et al
High response rate in refractory and poor risk multiple myeloma after allotransplantation using a nonmyeloablative conditioning regimen and donor lymphocyte infusions.
Blood.
97
2001
2574
2579
21
Kottaridis
PD
Milligan
D
Chopra
R
et al
In vivo alemtuzumab prevents graft-versus-host disease following non myeloablative stem cell transplantation.
Blood.
96
2000
2419
2425
22
Weiden
PL
Sullivan
KM
Flournoy
N
Storb
R
Thomas
ED
Antileukemic effect of chronic graft versus host disease: contribution to improved survival after allogeneic marrow transplantation.
N Engl J Med.
304
1981
1529
1533
23
Horowitz
MM
Gale
RP
Sondel
PM
et al
Graft versus leukemia reactions after bone marrow transplantation.
Blood.
75
1990
555
562
24
Childs
RW
Chernoff
A
Contentin
N
et al
Regression of metastatic renal cell carcinoma after nonmyeloablative allogeneic peripheral blood stem cell transplantation.
N Engl J Med.
343
2000
750
758
25
Bertz
H
Burger
JA
Kunzmann
R
Mertelsmann
R
Finke
J
Adoptive immunotherapy for relapsed multiple myeloma after allogeneic bone marrow transplantation (BMT): evidence for a graft versus myeloma effect.
Leukemia.
11
1997
281
283
26
Nagler
A
Slavin
S
Varadi
G
Naparstek
E
Samuel
S
Or
R
Allogeneic peripheral blood stem cell transplantation using a fludarabine based low intensity conditioning regimen for malignant lymphomas.
Bone Marrow Transplant.
25
2000
1021
1028
27
Roux
E
Helg
C
Chapuis
B
Mixed chimerism after bone marrow transplantation and the risk of relapse.
Blood.
83
1994
4385
4386
28
Mackinnon
S
Barnett
L
Bourhis
JH
Black
P
Heller
G
O'Reilly
RJ
Myeloid and lymphoid chimaerism after T-cell depleted bone marrow transplantation: evaluation of conditioning regimens using the polymerase chain reaction to amplify human minisatellite regions of genomic DNA.
Blood.
80
1992
3235
3241
29
Martino
R
Caballero
D
Canals
C
et al
Allogeneic peripheral blood stem cell transplantation with reduced-intensity conditioning: results of a prospective multicenter study.
Br J Hematol.
115
2001
653
659
30
Przepiorka
D
Weisdorf
D
Martin
P
et al
Consensus Conference on Acute GVHD Grading.
Bone Marrow Transplant.
15
1995
825
830
31
Sullivan
KM
Shulman
HM
Storb
R
et al
Chronic graft-versus-host disease in 52 patients: adverse natural course and successful treatment with combination immunosuppression.
Blood.
57
1981
267
276
32
Boeckh
M
Bowden
RA
Gooley
T
Myerson
D
Corey
L
Successful modification of a pp65 antigenemia-based early treatment strategy for prevention of cytomegalovirus disease in allogeneic marrow transplant recipients.
Blood.
93
1999
1781
1787
33
Boeckh
M
Gooley
TA
Myerson
D
Cunningham
T
Schoch
G
Bowden
RA
Cytomegalovirus pp65 antigenemia-guided early treatment with ganciclovir versus ganciclovir at engraftment after allogeneic marrow transplantation: a randomized double-blind study.
Blood.
88
1996
4063
4071
34
Mackinnon
S
Papadopoulos
EP
Carabasi
MH
et al
Adoptive immunotherapy evaluating escalating doses of donor leucocytes for relapse of chronic myeloic leukemia following bone marrow transplantation: separation of graft versus leukemia responses from graft versus host disease.
Blood.
86
1995
1261
1268
35
Naparstek
E
Or
R
Nagler
A
et al
T-cell-depleted allogeneic bone marrow transplantation for acute leukaemia using Campath-1 antibodies and posttransplant administration of donor's peripheral blood lymphocytes for prevention of relapse.
Br J Haematol.
89
1995
506
515
36
Van Besien
K
Sobocinski
KA
Rowlings
PA
et al
Allogeneic bone marrow transplantation for low-grade lymphoma.
Blood.
92
1998
1832
1836
37
Rondon
G
Giralt
S
Huh
Y
et al
Graft-versus-leukemia effect after allogeneic bone marrow transplantation for chronic lymphocytic leukemia.
Bone Marrow Transplant.
18
1996
669
672
38
Khouri
IF
Przepiorka
D
van Besien
K
et al
Allogeneic blood or marrow transplantation for chronic lymphocytic leukaemia: timing of transplantation and potential effect of fludarabine on acute graft-versus-host disease.
Br J Haematol.
97
1997
466
473
39
Tricot
G
Vesole
DH
Jagannath
S
Hilton
J
Munshi
N
Barlogie
B
Graft-versus-myeloma effect: proof of principle.
Blood.
87
1996
1196
1198
40
Mackinnon
S
Who may benefit from donor leucocyte infusions after allogeneic stem cell transplantation?
Br J Haematol.
110
2000
12
17
41
Apperley
JF
Mauro
FR
Goldman
JM
et al
Bone marrow transplantation for chronic myeloid leukemia in first chronic phase: importance of a graft versus leukemia effect.
Br J Haematol.
69
1998
239
245
42
Hale
G
Cobbold
S
Novitzky
N
et al
on behalf of CAMPATH users. CAMPATH-1 antibodies in stem cell transplantation.
Cytotherapy.
3
2001
145
164
43
Martino
R
Caballero
D
Canals
C
et al
Reduced-intensity conditioning reduces the risk of severe infections after allogeneic peripheral Blood stem cell transplantation.
Bone Marrow Transplant.
28
2001
341
347
44
Camargo
LF
Uip
DE
Simpson
AA
et al
Comparison between antigenemia and a quantitative-competitive polymerase chain reaction for the diagnosis of cytomegalovirus infection after heart transplantation.
Transplantation.
71
2001
412
417
45
Guiver
M
Fox
AJ
Mutton
K
Mogulkoc
N
Egan
J
Evaluation of CMV viral load using TaqMan CMV quantitative PCR and comparison with CMV antigenemia in heart and lung transplantation.
Transplantation.
71
2001
1609
1615
46
Dykewicz
CA
Summary of the guidelines for preventing opportunistic infections among hematopoietic stem cell transplant recipients.
Clin Infect Dis.
33
2001
139
144
47
Martin
PJ
Influence of alloreactive T cells on initial hematopoietic reconstitution after marrow transplantation.
Exp Hematol.
23
1995
174
179
48
Curtis
RE
Travis
LB
Rowlings
PA
et al
Risk of lymphoproliferative disorders after bone marrow transplantation: a multi-institutional study.
Blood.
94
1999
2208
2216
49
Gross
TG
Steinbuch
M
DeFor
T
et al
B cell lymphoproliferative disorders following hematopoietic stem cell transplantation: risk factors, treatment and outcome.
Bone Marrow Transplant.
23
1999
251
258
50
Chakrabarti
S
Mackinnon
S
Chopra
R
et al
High incidence of cytomegalovirus infection after nonmyeloablative stem cell transplantation: potential role of CAMPATH-1H in delaying immune reconstitution.
Blood.
99
2002
4357
4363

Author notes

José A. Pérez-Simón, Servicio de Hematologı́a, Hospital Universitario de Salamanca, Paseo de San Vicente s/n, 37007, Spain; e-mail: pesimo@usal.es.

Sign in via your Institution