The clinical results in 107 patients receiving a peripheral blood stem cell (PBSC) graft mobilized by granulocyte colony-stimulating factor (G-CSF) from HLA-A, -B, and -DR–compatible unrelated donors were compared to 107 matched controls receiving unrelated bone marrow (BM) transplants. Engraftment was achieved in 94% of the patients in both groups. The PBSC graft contained significantly more nucleated cells, CD34+, CD3+, and CD56+ cells (P < .001), and resulted in a significantly shorter time-to-neutrophil (15 versus 19 days) and platelet engraftment (20 versus 27 days), compared to the BM control group (P < .001). Probabilities of acute graft-versus-host disease (GVHD) grades II to IV were 35% and 32% (not significant [NS]) and of chronic GVHD 61% and 76% (NS) in the PBSC and BM groups, respectively. There was no difference between the 2 groups in bacteremia, cytomegalovirus reactivation or disease, and fungal infection. The 3-year transplant-related mortality (TRM) rates were 42% in the PBSC group and 31% in the BM controls (P = .7) and the survival rates were 46% and 51%, respectively. The probability of relapse was 25% and 31% in both groups (NS), resulting in disease-free survival rates of 43% in the PBSC group and 46% in the BM controls (NS). In the multivariate analysis, early disease, acute GVHD grade 0 to I, and presence of chronic GVHD were independent factors associated with a better disease-free survival in this study. PBSC from HLA-compatible unrelated donors can be used safely as an alternative to BM for stem cell transplantation.

The first report using peripheral blood as a source of stem cells for allogeneic hematopoietic stem cell transplantation (SCT) was published in 1989.1 Since then, the number of peripheral blood stem cell (PBSC) transplants has steadily increased.2-9 In 1995, about 500 PBSC transplants from HLA-identical siblings were reported to the European Group for Blood and Marrow Transplantation (EBMT); by 1996, the figure had increased to more than 1000.8 Until recent years most reports concerned PBSC grafts using HLA-A, -B, and -DR–identical sibling donors; data have been reported on only a few patients who received PBSC from unrelated donors.10-14 There have been several reasons for the reluctance to use PBSCs from unrelated donors. One concern has been the ethics of administering granulocyte colony-stimulating factor (G-CSF) to mobilize PBSCs from healthy volunteers. Another concern has been that the 10- to 15-fold higher donor T-cell content of PBSC would increase the risk of graft-versus-host disease (GVHD).13,15,16 Some reports have indicated a higher incidence of chronic GVHD after sibling PBSC transplantation.17-19 We report here the experience of PBSC grafts from unrelated donors at 3 European bone marrow transplant (BMT) units.

Patients

The study group consisted of 107 consecutive patients who underwent PBSC transplantation with an HLA-identical unrelated donor at Essen (n = 44), Huddinge Hospital (n = 32), or Idar-Oberstein (n = 31) between February 1993 and September 1999. The patients were eligible for transplantation with PBSCs if they had a disease for which BMT was indicated and after approved informed consent was obtained from the donor and recipient. Most of the patients had a hematologic malignancy. Before 1998, relatively more patients underwent transplantation while not in remission, compared to those having the procedure later. The characteristics of the patients and donors are given in Table 1.

Table 1.

Patient and donor characteristics

PBSC groupBM group
No. of patients 107 107  
Recipient age 35 (5-56)  37 (1-55)   
Donor age 33 (20-53) 36 (18-55)  
Recipient sex (M/F) 56/51 60/47  
Donor sex (M/F) 72/35 62/44 
Diagnoses   
 AML   
  CR1 12 11 
  CR2 11  
  Not in remission 16 13 
 ALL   
  CR1 7  
  CR2-3 
  Not in remission 
 CML   
  CP1 39 42  
  CP2 
  Acc phase 6  
  Blast crisis 
 Lymphoma 0  
 Myelodysplastic syndrome 
 Aspartylglycosaminurea 1  
Recipient CMV serology (−/+) 43/64 47/59  
Donor CMV serology (−/+) 61/46 58/48  
Follow-up, mo, median (range) 11.1 (1.9-49)* 20.8 (2.8-66) 
PBSC groupBM group
No. of patients 107 107  
Recipient age 35 (5-56)  37 (1-55)   
Donor age 33 (20-53) 36 (18-55)  
Recipient sex (M/F) 56/51 60/47  
Donor sex (M/F) 72/35 62/44 
Diagnoses   
 AML   
  CR1 12 11 
  CR2 11  
  Not in remission 16 13 
 ALL   
  CR1 7  
  CR2-3 
  Not in remission 
 CML   
  CP1 39 42  
  CP2 
  Acc phase 6  
  Blast crisis 
 Lymphoma 0  
 Myelodysplastic syndrome 
 Aspartylglycosaminurea 1  
Recipient CMV serology (−/+) 43/64 47/59  
Donor CMV serology (−/+) 61/46 58/48  
Follow-up, mo, median (range) 11.1 (1.9-49)* 20.8 (2.8-66) 

AML indicates acute myeloid leukemia; ALL, acute lymphocytic leukemia; Acc phase, accelerated phase.

*

P < .001.

BMT control group

For each unrelated patient undergoing PBSC transplantation we selected a control patient who had received an HLA-A, -B, and -DR–compatible unrelated bone marrow (BM) graft at almost the same time. Controls were matched for diagnosis, stage of disease, age (< 20 years or > 20 years), and GVHD prophylaxis. All controls, except one, were matched for transplant center. Eighty of 107 (75%) controls were matched for all 4 criteria, and 101 of 107 (94%) were matched for at least 3 criteria. Among the controls, 43 came from Essen, 33 from Huddinge, and 31 from Idar-Oberstein. Patient and donor characteristics are given in Table 1.

Donors

All donors were HLA-A, -B, and -DR compatible with the patients. Before 1997, class I HLA typing was serologic. Since then, polymerase chain reaction-sequence-specific primer (PCR-SSP) low-resolution typing for class I was used. For HLA class II, genomic high-resolution DNA-based typing (PCR-SSP) was used.20 All patients undergoing transplantation at Huddinge University Hospital have recently been retrospectively retyped using PCR-SSP high-resolution typing for both HLA class I and II antigens (submitted). Before apheresis, all donors of PBSCs were treated with G-CSF (Rhône-Poulenc Rorer, Lyon, France or Amgen-Roche, Thousand Oaks, CA) for 4 to 6 days. The dose of G-CSF ranged from 9 to 12.5 μg/kg per day, administered subcutaneously once daily.

Conditioning

As preparative treatment before transplantation, most patients received cyclophosphamid (Cy) 60 mg/kg for 2 consecutive days, combined with total body irradiation (TBI) in a dose ranging from 10 to 13.5 Gy (Table 2). TBI doses were mainly 10 Gy in Huddinge (single dose) and Essen (fractionated) and 12 to 13.5 Gy (fractionated) in Idar-Oberstein. Thirty-eight patients were given busulfan (Bu) 16 mg/kg followed by 120 mg/kg Cy.21 Other combinations of conditioning regimens were also used and are given in Table 2. A priority was given to TBI, but if not possible, Bu was used.21 Detailed descriptions of the myeloablative therapy, immunosuppression, and patient care at the different centers have been reported elsewhere.10,22,23 

Table 2.

Conditioning and immunosuppression

PBSC groupBM group
No. 107 107 
Conditioning   
 Cy/TLI 1  
 Cy/TBI 67 80 
 Cy/TBI + other drug 7  
 Holoxan/TBI 
 Bu/Cy 22 16  
 Bu/Cy + other drug 
 Bu/other 0  
 Cy/MEA 
 ATG/OKT-3 25/11 21/16  
GVHD prophylaxis   
 CsA or MTX 3  
 CsA + pred 3  
 CsA + MMF 0  
 CsA + MTX 71 69 
 CsA + MTX + pred 25 31  
 CsA + pred + MMF 
 CsA + MTX + pred + MMF 1  
Patients receiving G-CSF 53* 32  
Median d with G-CSF 10 (1-24) 11 (5-30) 
PBSC groupBM group
No. 107 107 
Conditioning   
 Cy/TLI 1  
 Cy/TBI 67 80 
 Cy/TBI + other drug 7  
 Holoxan/TBI 
 Bu/Cy 22 16  
 Bu/Cy + other drug 
 Bu/other 0  
 Cy/MEA 
 ATG/OKT-3 25/11 21/16  
GVHD prophylaxis   
 CsA or MTX 3  
 CsA + pred 3  
 CsA + MMF 0  
 CsA + MTX 71 69 
 CsA + MTX + pred 25 31  
 CsA + pred + MMF 
 CsA + MTX + pred + MMF 1  
Patients receiving G-CSF 53* 32  
Median d with G-CSF 10 (1-24) 11 (5-30) 

TLI indicates total lymphoid irradiation; MMF, mycomofetilphenolate; pred, prednisolone; MEA, mitoxantrone + etoposide + arabinoside-C.

*

P < .01.

As additional immunosuppressive treatment before transplant, one third of the patients (mainly at Huddinge) were given antithymocyte globulin (ATG-Fresenius, Fresenius AG, Bad Homburg, Germany, or Thymoglobulin, IMTIX-Sangstat, Lyon, France) or monoclonal anti–T-cell antibodies (Orthoclone OKT3, Ortho Biotech, Raritan, NJ) for 4 to 5 days.24 

GVHD prophylaxis

The most common immunosuppression agent was cyclosporin A (CsA) combined with a short course (4 doses) of methotrexate (MTX; Table 2). The combination of CsA, MTX, and prednisolone was used in about one fourth of the patients.

Engraftment

Neutrophil engraftment was defined as the first of 3 consecutive days with an absolute neutrophil count (ANC) of 0.5 × 109/L or above. Platelet engraftment was defined as the first of 7 days with platelet counts of 50 × 109/L or above without transfusion.

GVHD

Acute GVHD was graded 0 to IV, according to standard criteria.25 Chronic GVHD was assessed in patients surviving more than 90 days after transplantation and defined as limited or extensive.26 

Other definitions

Bacteremia was defined as the first positive blood culture related to a febrile episode during the first 30 days after transplantation. Cytomegalovirus (CMV) reactivation was determined weekly with PCR in leukocytes.27 Patients in Idar-Oberstein and Essen were followed weekly by the pp 65 antigen in blood leukocytes. CMV disease was defined as symptoms from the affected organ, combined with detection of CMV by histology, isolation, or immunohistochemistry, or typical retinitis changes.28Determinations of CD3+, CD34+, and CD56+ cells were performed on unseparated BM or PBSCs by flow cytometry, as previously described.12 G-CSF was given to 85 patients after transplantation for a median of 11 days (range, 1-30 days; Table 2).

Statistics

Analysis was performed on March 25, 2000, with a median follow-up time of 11.1 months (range, 1.9-49 months) in the PBSC group and 20.8 months (range, 2.8-66 months) in the BM group. Patients in the control group were matched according to (in ranking) center, diagnosis, disease stage, age (< 20 years or ≥ 20 years) and GVHD prophylaxis.

The probabilities of acute and chronic GVHD, transplant-related mortality (TRM), relapse, overall survival, and disease-free survival (DFS) were calculated with the Kaplan-Meier method,29comparing the groups with use of the log-rank test (Mantel-Haentszel).30 We used the Mann-WhitneyU test to compare the cell yield, time to engraftment, transfusions, and hospitalizations. In the univariate and multivariate analyses of risk factors for acute GVHD and prognostic factors for engraftment, the logistic regression model was used.31Patients dying before day 30 without GVHD were omitted from the GVHD analysis. When analyzing prognostic factors for chronic GVHD, TRM, survival, relapse, and DFS, the Cox regression model was used.32 

In analyzing risk factors for relapse and chronic GVHD, only patients surviving more than 90 days after hematopoietic stem cell transplantation (HSCT) or having the event (relapse or chronic GVHD) before day 90 were included in the analysis. Factors significant at the 10% level in the univariate analysis were included in the multivariate analysis. The following factors were analyzed: MTX, nucleated cell dose, CD34 dose, CD3 dose, CD56 dose, ATG, G-CSF after transplant, stem cell source, disease, disease stage, pretransplant CMV serology, conditioning, patient age (< 30 years or ≥ 30 years), donor age (< 35 years or ≥ 35 years), and sex. For TRM, relapse, and survival, GVHD was also included.

Graft content

The nucleated cell dose was 3 times higher in the PBSC group than in the BM control group (P < .001; Table3). The CD34+ cell dose was 2-fold and the CD3+ and the CD56+ cell doses more than 10-fold higher in the PBSC group (P < .001).

Table 3.

Cell yield in PBSC and BM grafts

PBSCBMP
Nucleated cell dose (× 108/kg) 10.0  (2.2-51.1) 3.1  (0.2-11.6) < .001 
CD34+ cell dose (× 106/kg) 6.1  (2.0-18.3) 3.1  (0.5-10.7) < .001  
CD3+ cell dose (× 106/kg) 336  (75-1527) 25.3  (3.5-57.1) < .001  
CD56+ cell dose (× 106/kg) 32.4  (8-379) 2.4  (0.9-14.7) < .001 
PBSCBMP
Nucleated cell dose (× 108/kg) 10.0  (2.2-51.1) 3.1  (0.2-11.6) < .001 
CD34+ cell dose (× 106/kg) 6.1  (2.0-18.3) 3.1  (0.5-10.7) < .001  
CD3+ cell dose (× 106/kg) 336  (75-1527) 25.3  (3.5-57.1) < .001  
CD56+ cell dose (× 106/kg) 32.4  (8-379) 2.4  (0.9-14.7) < .001 

Engraftment

Six patients in the PBSC group and 6 in the BM group died early, before engraftment (ANC > 0.5 × 109/L), giving an engraftment rate of 94% in both groups. Time-to-neutrophil engraftment was significantly shorter in the PBSC group (median, 15 days; range, 7-27 days) compared to the BM group (median, 19 days; range, 10-31 days) (P < .001). In the multivariate analysis, PBSC graft (odds ratio [OR] 3.74, CI 2.03-6.89, P < .001) and G-CSF after transplant (OR 2.87, CI 1.55-5.37,P = .001) were independent factors associated with faster neutrophil engraftment.

Platelet engraftment (> 50 × 109/L) was also significantly faster in the PBSC group (20 days; range, 8-395 days) but 27 days (18-210; P < .001) in the BM group. The CD34+ cell dose (> 5 × 106/L; OR 2.72, CI 1.38-5.37, P = .004) was the only factor associated with faster platelet engraftment in the multivariate analysis.

The median numbers of days from transplantation to discharge from hospital were similar in both groups, 37 days (range, 13-104 days) and 39 days (range, 14-150 days), respectively.

Transfusions

The numbers of platelet, erythrocyte, and granulocyte transfusions did not differ between the 2 groups, although there was a tendency to more platelet transfusions in the BM group, median 14 (range, 0-167) (P = .06) versus 10 (range, 0-144) in the PBSC group.

Infections

Bacteremia occurred in 45 (42%) of the patients in both groups. In the PBSC group, 33 patients had a coagulase-negative staphylococci (CNS) infection, 3 had a streptococcal infection, 5 had a pseudomonal infection, and 4 had other bacteria. In the BM group, 34 had a CNS infection, 4 a streptococcal infection, 3 a pseudomonal infection, and 4 other bacteria. CMV reactivation, as detected by PCR or pp 65 antigen, occurred in 33 and 34 patients in the PBSC and BM groups, respectively. CMV disease developed in 5 and 4 patients in the 2 groups, respectively. CMV disease consisted of interstitial pneumonitis in 4 patients and enteritis in one patient in the PBSC group. In the BM group, 2 patients had interstitial pneumonitis and one each had enteritis and retinitis. Invasive fungal infection was diagnosed in 8 patients in each group. Among the PBSC patients,Aspergillus was found in 3, Candida albicans in 3, and Candida glabrata in 2. In the BM group,Aspergillus was found in 5, C. albicans in 2, andPneumocystis carinii in 1.

GVHD

The cumulative probabilities of acute GVHD grades II to IV were similar in the 2 groups, 35% in the PBSC group and 32% in the BM group (NS). The probabilities of grades III to IV acute GVHD were 18% and 22% in the 2 groups, respectively. In the multivariate analysis, the use of OKT-3 was a significant risk factor for acute GVHD grades II to IV (OR 3.74, CI 1.62-8.67, P = .002).

The cumulative probabilities of chronic GVHD were 61% among patients receiving a PBSC graft and 76% in patients receiving a BM graft (NS; Figure 1). Seven patients in the PBSC group and 10 in the BM group developed extensive chronic GVHD (NS). In the multivariate analysis, acute GVHD of any grade (relative risk [RR] 3.03, CI 1.72-5.37, P < .001), no ATG (RR 2.83, CI 1.51-5.31, P = .001), and chronic myeloid leukemia (CML; RR 1.84, CI 1.20-2.83, P = .005) were correlated to the development of chronic GVHD.

Fig. 1.

Probability of chronic GVHD.

Time and cumulative probabilities of chronic GVHD after SCT with PBSC and BM, using unrelated donors

Fig. 1.

Probability of chronic GVHD.

Time and cumulative probabilities of chronic GVHD after SCT with PBSC and BM, using unrelated donors

Close modal

Survival and relapse

The TRM, all deaths except relapse, did not differ between the 2 groups (Figure 2). The TRM rate was 18% and 19% at 100 days and 32% and 31% at 12 months after transplantation in the 2 groups, respectively.

Fig. 2.

Probability of TRM.

Time and cumulative probabilities of TRM after SCT with PBSC and BM, using unrelated donors

Fig. 2.

Probability of TRM.

Time and cumulative probabilities of TRM after SCT with PBSC and BM, using unrelated donors

Close modal

In the multivariate analysis, acute GVHD grades II to IV (relative hazard [RH] 8.94, CI 4.76-16.78, P < .001) was the only independent risk factor for TRM. Higher patient age (≥ 30 years; RH 2.01, 95% CI 1.19-3.42, P = .011) and late disease (RH 1.99, CI 1.17-3.39, P = .011) were independent risk factors for TRM if acute GVHD was not added in the multivariate analysis.

The 3-year probabilities of survival were 46% in the PBSC group and 51% in the BM group (NS; Figure 3). Among patients with a malignant disease in first remission (CR) or first chronic phase (CP; early disease), the probabilities of survival at 2 years were 61% and 68% in the PBSC group and the BM group, respectively (NS). In patients beyond the first CR/CP (late disease), the 2-year survival rates were 37% and 29%, respectively (NS). In the multivariate analysis, late disease (RH 2.25, CI 1.46-3.46,P < .001) and acute GVHD grades II to IV (RH 3.03, CI 1.97-4.66, P < .001) were independent risk factors for death after HSCT in this material. If GVHD was not introduced into the analysis, pretransplant CMV seropositivity (RH 1.54, CI 1.01-2.34,P = .046) was also associated with death.

Fig. 3.

Probability of survival.

Time and cumulative probability of survival after SCT with PBSC and BM, using unrelated donors.

Fig. 3.

Probability of survival.

Time and cumulative probability of survival after SCT with PBSC and BM, using unrelated donors.

Close modal

Hematologic relapse was detected in 18 patients receiving PBSC and 20 receiving BM, giving a cumulative probability of 25% in both groups. Among patients having early disease, the 2-year probabilities of relapse were 2% in the PBSC group and 6% in the BM group (NS). Corresponding figures for patients in late disease were 49% and 52% (NS).

In the multivariate analysis, advanced disease (RH 10.91, CI 4.18-28.5,P < .001) and absence of chronic GVHD (RH 2.84, CI 1.42-5.64, P = .003) were independent risk factors for relapse. If GVHD was removed from the analysis, advanced disease and acute leukemia (AL; RH 2.51, CI 1.06-5.93, P = .04) were significantly associated with relapse.

The rates for DFS at 3 years were similar in the 2 groups, 43% in patients in the PBSC group and 46% in the BM group (NS; Figure4). Among patients with CML, the DFS rates at 2 years were 62% and 64% in the PBSC and the BM groups, respectively.

Fig. 4.

Probability of DFS.

Time and cumulative probabilities of DFS after SCT among patients in early and late diseases given PBSC and BM from unrelated donors.

Fig. 4.

Probability of DFS.

Time and cumulative probabilities of DFS after SCT among patients in early and late diseases given PBSC and BM from unrelated donors.

Close modal

Corresponding figures for patients with AL were 43% and 51% (NS). The probabilities of DFS and relapse in patients with early and late disease in the various diagnostic groups are given in Table4.

Table 4.

Probabilities of relapse and relapse-free survival among CML and AL patients, divided into early (CR1/CP1) and late (> CR1/CP1) disease

No.Relapse (2 y), %DFS (2 y), %
PBSC    
 CML early disease 39 64  
 CML late disease 12 21 45  
 AL early disease 17 68 
 AL late disease 37 57 25  
Bone marrow    
 CML early disease 42 70  
 CML late disease 10 38 40  
 AL early disease 18 21 64 
 AL late disease 36 57 26 
No.Relapse (2 y), %DFS (2 y), %
PBSC    
 CML early disease 39 64  
 CML late disease 12 21 45  
 AL early disease 17 68 
 AL late disease 37 57 25  
Bone marrow    
 CML early disease 42 70  
 CML late disease 10 38 40  
 AL early disease 18 21 64 
 AL late disease 36 57 26 

DFS indicates disease-free survival.

Among patients not in remission at transplant, DFS rates at 2 years were 25% for patients receiving a PBSC graft and 11% in the BM group (P = .11; Figure 5).

Fig. 5.

Probability of DFS.

Time and cumulative probabilities of DFS after SCT among patients not in remission/chronic phase given PBSC and BM from unrelated donors.

Fig. 5.

Probability of DFS.

Time and cumulative probabilities of DFS after SCT among patients not in remission/chronic phase given PBSC and BM from unrelated donors.

Close modal

In the multivariate analysis, early disease (RH 2.97, CI 1.97-4.48,P < .001), acute GVHD grades 0 to I (RH 2.31, CI 1.54-3.49, P < .001), and the presence of chronic GVHD (RH 1.98, CI 1.28-3.04, P = .002) were independent prognostic factors for better DFS in this material. If GVHD was not added to the analysis, early disease was the only significant factor for a better DFS.

The PBSC grafts contained a significantly higher cell dose than the BM grafts and this was correlated to faster neutrophil and platelet engraftment. This is in agreement with most other reports using sibling donors.5-8,12,14 The higher nucleated cell dose and CD34+ cell content seem to be the main factors for faster engraftment in the PBSC group.12,13,33 In the present study, more patients in the PBSC group received posttransplant G-CSF treatment, which promotes engraftment. In the multivariate analysis, G-CSF and PBSCs were independent prognostic factors for faster neutrophil engraftment. For platelet engraftment, the CD34+cell dose was the main factor for a faster engraftment. This agrees with other studies.12,33 Despite a faster engraftment being achieved, no difference in the number of days to discharge from hospital was seen. There may be several explanations for this. Even after engraftment, problems such as malnutrition, fever, GVHD, and infections remain before the patients can be discharged. The incidence of infections was the same as in the BM group despite the faster engraftment in the PBSC group.

In contrast, in a double-blinded randomized study, Powles and coworkers found that patients receiving PBSCs from HLA-identical siblings were discharged significantly earlier than those given BM.9This difference compared with our results could be explained by the fact that recipients of stem cells from siblings have fewer transplant-related complications than recipients of unrelated grafts.22 

We found no difference in the incidence of moderate-to-severe acute GVHD between the 2 groups. This is important because it has been feared that the higher T-cell content in the PBSC grafts might cause more GVHD when using unrelated donors. This does not agree with the update of the EBMT randomized study in leukemic patients receiving grafts from HLA-identical siblings, where those receiving PBSC grafts had significantly more acute GVHD (N. Schmitz, oral communication, August 2000). One bias in the present study may have been that more patients in the PBSC group received G-CSF after transplant.

Some studies have indicated that G-CSF may affect the incidence and severity of acute GVHD, although most have not.34 But in both univariate and multivariate analysis, G-CSF after transplant did not affect acute GVHD in this study.

Roughly one third of the patients in both groups in our study developed moderate-to-severe acute GVHD, which is less than that reported by others using unrelated marrow.35,36 The use of ATG as part of pretransplant conditioning in some patients may have helped to reduce the incidence of acute GVHD.37 Polyclonal ATG was correlated to less acute GVHD in the univariate but not in the multivariate analysis. As we have previously reported, the use of OKT-3 increases the risk for acute GVHD.24 This is probably caused by a “cytokine release syndrome” on administration of OKT-3. However, the number of patients receiving ATG or OKT-3 were similar in the 2 groups.

No difference in the probability of chronic GVHD was seen in this study. Using HLA-identical sibling donors, some studies have reported a higher incidence of chronic GVHD when using PBSC.17-19The reason for absence of a difference in the present study may be the higher incidence of chronic GVHD using unrelated donors, compared to HLA-identical siblings.35,36,38,39 In this study, the incidences of chronic GVHD were 61% and 76% in the PBSC and BM groups, respectively. As previously reported, diagnosis of CML and acute GVHD of any grade were correlated to chronic GVHD.40 

We found that patients not receiving polyclonal ATG had more chronic GVHD. The reason for this is that the absence of ATG is correlated to more acute GVHD and acute GVHD triggers chronic GVHD.40But, once again, the use of ATG was similar in the 2 groups. The incidences of bacteremia and CMV and fungal infections were almost identical in the 2 groups. Theoretically, the risk of CMV infection may be higher when using PBSC because the higher cell dose contains more viruses, but we found no such difference in this study. However, we analyzed only the number of patients with a CMV infection and not the occurrence of repeated infections.

With regard to survival, relapse, and DFS, the outcome was the same in patients receiving PBSC or BM. This agrees with most reports comparing PBSC with BM from HLA-identical sibling donors.7,8 Two studies, one from the International Bone Marrow Transplant Registry (IBMTR) and another from the M. D. Anderson Hospital, reported better DFS rates among patients with advanced leukemia using PBSCs rather than BM, using HLA-identical sibling donors.41,42 A randomized trial from Seattle also showed that leukemic patients with more advanced disease had fewer relapses and better DFS if they received PBSCs rather than BM.43 In our material, we found no difference in DFS among patients with CML or AL with early or late disease (Table 4 and Figure 4). However, when analyzing only patients not in remission at transplant, we found, in accordance with the studies from Seattle, IBMTR, and the M. D. Anderson Hospital, Houston, TX, in HLA-identical siblings, a better DFS for patients receiving PBSCs (Figure 5). But in our study the difference was not statistically significant (P = .11). This may be due to the low number of patients in this cohort and conclusions cannot be drawn based on this material. Further studies with PBSC using unrelated donors including more patients in advanced disease are needed.

There was a longer follow-up with BM in this study, because with more experience with PBSCs more patients and donors selected PBSCs compared to BM. In the PBSC group, 32% of the patients received transplants before 1998, compared to 53% in the BM group. However, indications for SCT using unrelated donors were the same during the period 1993-1999, when the patients were included for this study. Furthermore, any changes in treatment protocols that may have influenced the outcome variables have not been introduced in any of the 3 centers during this time period. Different GVHD prophylaxis and conditioning protocols were used in this study, but no statistical difference between the 2 groups existed. Furthermore, in the multivariate analysis significant and almost significant factors were included. Therefore, the finding of a similar survival using BM or PBSC seems sound and is not due to selection bias or differences in treatment or follow-up between the 2 groups.

When using unrelated donors, the incidence of chronic GVHD is higher than in HLA-identical siblings, which also carries a graft-versus-leukemia effect,44,45 thus reducing the risk of relapse. The shorter follow-up in the PBSC group may also have affected the results in the present study. However, this seems unlikely because the same protocol has been used throughout the study. HLA typing for class I has been genomic at one center during the last 3 years. This has not affected the outcome because genomic or serologic class I mismatched donor-recipient pairs were not included in this material.

Factors associated with a better DFS in our study (early disease, grades 0-I acute GVHD, and chronic GVHD) agree with other studies.46,47 Patients with chronic GVHD may run a slightly higher risk of late death not related to relapse, but this will probably be balanced by a lower risk of relapse. A long-term follow-up is warranted to find the answer.

To conclude, this study has shown that the use of PBSCs from unrelated donors is a safe and well-tolerated procedure, resulting in faster engraftment than BM, a good survival rate, and DFS.

We thank all nurses at the departments for excellent and skillful care and all volunteer donors for their generous gift to the patients.

Supported by grants from Swedish Cancer Society (0070-B97-11XBC), the Children's Cancer Foundation (1997/073, 1998/002), the Swedish Medical Research Council (K98-06X-05 971-18B), the FRF Foundation, and the Tobias Foundation (B11/98).

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
Kessinger
A
Smith
D
Strandjord
S
Allogeneic transplantation of blood-derived, T cell-depleted hematopoietic stem cells after myeloablative treatment in a patient with acute lymphoblastic leukemia.
Bone Marrow Transplant.
4
1989
643
646
2
Dreger
P
Suttorp
M
Haferlach
T
Löffler
H
Schmitz
N
Schroyes
W
Allogeneic granulocyte colony-stimulating factor mobilized peripheral blood progenitor cells for treatment of engraftment failure after bone marrow transplantation.
Blood.
81
1993
1404
1407
3
Russell
NH
Hunter
AE
Peripheral blood stem cells for allogeneic transplantation [editorial].
Bone Marrow Transplant.
13
1994
353
355
4
Bensinger
WI
Weaver
CH
Appelbaum
FR
et al
Transplantation of allogeneic peripheral blood stem cells mobilized by recombinant human granulocyte colony-stimulating factor.
Blood.
85
1995
1655
1658
5
Korbling
M
Przepiorka
D
Huh
YO
et al
Allogeneic blood stem cell transplantation for refractory leukemia and lymphoma: potential advantage of blood over marrow allografts.
Blood.
85
1995
1659
1665
6
Schmitz
N
Dreger
P
Suttorp
M
et al
Primary transplantation of allogeneic peripheral blood progenitor cells mobilized by filgrastim (granulocyte colony-stimulating factor).
Blood.
85
1995
1666
1672
7
Bensinger
WI
Clift
R
Martin
P
et al
Allogeneic peripheral blood stem cell transplantation in patients with advanced hematologic malignancies: a retrospective comparison with marrow transplantation.
Blood.
88
1996
2794
2800
8
Schmitz
N
Bacigalupo
A
Hasenclever
D
et al
Allogeneic bone marrow transplantation vs filgrastim-mobilised peripheral blood progenitor cell transplantation in patients with early leukaemia: first results of a randomised multicentre trial of the European Group for Blood and Marrow Transplantation [see comments].
Bone Marrow Transplant.
21
1998
995
1003
9
Powles
R
Mehta
J
Kulkarni
S
et al
Allogeneic blood and bone-marrow stem-cell transplantation in haematological malignant diseases: a randomised trial.
Lancet.
355
2000
1231
1237
10
Blau
IW
Basara
N
Lentini
G
et al
Feasibility and safety of peripheral blood stem cell transplantation from unrelated donors: results of a single-center study.
Bone Marrow Transplant.
27
2001
27
33
11
Fauser
AA
Basara
N
Blau
IW
Kiehl
MG
A comparative study of peripheral blood stem cell vs bone marrow transplantation from unrelated donors (MUD): a single-centre study.
Bone Marrow Transplant.
25(suppl 2)
2000
S27
S31
12
Hägglund
H
Ringdén
O
Remberger
M
et al
Faster neutrophil and platelet engraftment, but no difference in acute GVHD or survival, using peripheral blood stem cells from related and unrelated donors, compared to bone marrow.
Bone Marrow Transplant.
22
1998
131
136
13
Ringdén
O
Remberger
M
Runde
V
et al
Peripheral blood stem cell transplantation from unrelated donors: a comparison with marrow transplantation.
Blood.
94
1999
455
464
14
Ringdén
O
Hägglund
H
Runde
V
et al
Faster engraftment of peripheral blood progenitor cells compared to bone marrow from unrelated donors.
Bone Marrow Transplant.
21(suppl 3)
1998
S81
S84
15
Singhal
S
Powles
R
Kulkarni
S
et al
Comparison of marrow and blood cell yields from the same donors in a double-blind, randomized study of allogeneic marrow vs blood stem cell transplantation.
Bone Marrow Transplant.
25
2000
501
505
16
Ringdén
O
Lönnqvist
B
Hägglund
H
et al
Transplantation with peripheral blood stem cells from unrelated donors without serious graft-versus-host disease [letter].
Bone Marrow Transplant.
16
1995
856
857
17
Storek
J
Gooley
T
Siadak
M
et al
Allogeneic peripheral blood stem cell transplantation may be associated with a high risk of chronic graft-versus-host disease.
Blood.
90
1997
4705
4709
18
Scott
MA
Gandhi
MK
Jestice
HK
Mahendra
P
Bass
G
Marcus
RE
A trend towards an increased incidence of chronic graft-versus-host disease following allogeneic peripheral blood progenitor cell transplantation: a case-controlled study.
Bone Marrow Transplant.
22
1998
273
276
19
Blaise
D
Kuentz
M
Fortanier
C
et al
Randomized trial of bone marrow versus lenograstim-primed blood cell allogeneic transplantation in patients with early-stage leukemia: a report from the Societe Francaise de Greffe de Moelle.
J Clin Oncol.
18
2000
537
546
20
Olerup
O
Zetterqvist
H
HLA-DR typing by PCR amplification with sequence-specific primers (PCR-SSP) in 2 hours: an alternative to serological DR typing in clinical practice including donor-recipient matching in cadaveric transplantation.
Tissue Antigens.
39
1992
225
235
21
Ringdén
O
Ruutu
T
Remberger
M
et al
A randomized trial comparing busulphan with total body irradiation as conditioning in allogenic marrow transplant recipients with leukemia: a report from the Nordic Bone Marrow Transplantation Group.
Blood.
83
1994
2723
2730
22
Ringdén
O
Remberger
M
Persson
U
et al
Similar incidence of graft-versus-host disease using HLA-A, -B and -DR identical unrelated bone marrow donors as with HLA-identical siblings.
Bone Marrow Transplantation.
15
1995
619
625
23
Beelen
DW
Ottinger
HD
Elmaagacli
A
et al
Transplantation of filgrastim-mobilized peripheral blood stem cells from HLA-identical sibling or alternative family donors in patients with hematologic malignancies: a prospective comparison on clinical outcome, immune reconstitution, and hematopoietic chimerism.
Blood.
90
1997
4725
4735
24
Remberger
M
Svahn
B-M
Hentschke
P
Löfgren
C
Ringdén
O
Effect on cytokine release and graft-versus-host disease of different anti-T-cell antibodies during conditioning for unrelated haematopoietic stem cell transplantation.
Bone Marrow Transplant.
24
1999
823
830
25
Glucksberg
H
Storb
R
Fefer
A
et al
Clinical manifestations of graft-versus-host disease in human recipients of marrow from HLA-matched sibling donors.
Transplantation.
18
1974
295304
26
Shulman
H
Sullivan
K
Weiden
P
et al
Chronic graft-versus-host syndrome in man: a clinicopathological study of 20 long-term Seattle patients.
Am J Med.
69
1980
204
217
27
Ljungman
P
Lore
K
Aschan
J
et al
Use of a semi-quantitative PCR for cytomegalovirus DNA as a basis for pre-emptive antiviral therapy in allogeneic bone marrow transplant patients.
Bone Marrow Transplant.
17
1996
583
587
28
Ljungman
P
Plotkin
S
Definition, clinical severity scores and new syndromes.
Scand J Infect Dis Suppl.
99
1995
87
88
29
Kaplan
E
Meier
P
Nonparametric estimation from incomplete observations.
J Am Stat Assoc.
53
1958
457
481
30
Peto
R
Pike
MC
Armitage
P
et al
Design and analysis of randomized clinical trials requiring prolonged observation of each patient, II: analysis and examples.
Br J Cancer.
35
1977
1
39
31
Breslow
N
Day
N
Davis
W
Statistical Methods in Cancer Research.
1. The Analysis of Case-Control Studies
1980
International Agency for Research on Cancer
Lyon, France
32
Cox
D
Regression models and life-tables.
J R Stat Soc (Series B).
34
1972
187
220
33
Mavroudis
D
Read
E
Cottler-Fox
M
et al
CD34+ cell dose predicts survival, posttransplant morbidity, and rate of hematologic recovery after allogeneic marrow transplants for hematologic malignancies.
Blood.
88
1996
3223
3228
34
Bishop
MR
Tarantolo
SR
Geller
RB
et al
A randomised, double-blind trial of filgrastim (granulocyte colony-stimulating factor) versus placebo following allogeneic blood stem cell transplantation.
Blood.
96
2000
80
85
35
Beatty
P
Hansen
J
Longton
G
et al
Marrow transplantation from HLA-matched unrelated donors for treatment of hematologic malignancies.
Transplantation.
51
1991
443
447
36
Kernan
NA
Bartsch
G
Ash
RC
et al
Analysis of 462 transplantations from unrelated donors facilitated by the National Marrow Donor Program [see comments].
N Engl J Med.
328
1993
593
602
37
Ringdén
O
Remberger
M
Carlens
S
et al
Low incidence of acute graft-versus-host disease, using unrelated HLA-A, -B and -DR compatible donors and conditioning, including anti-T-cell antibodies.
Transplantation.
66
1998
620
625
38
Storb
R
Deeg
H
Whitehead
J
et al
Methotrexate and cyclosporine compared with cyclosporine alone for prophylaxis of acute graft-versus-host disease after marrow transplantation for leukemia.
N Engl J Med.
314
1986
729
735
39
Ringdén
O
Paulin
T
Lönnqvist
B
Nilsson
B
An analysis of factors predisposing for chronic graft-versus-host disease.
Exp Hematol.
13
1985
1062
1067
40
Carlens
S
Ringdén
O
Remberger
M
et al
Risk factors for chronic graft-versus-host disease after bone marrow transplantation: a retrospective single centre analysis.
Bone Marrow Transplant.
22
1998
755
761
41
Champlin
RE
Schmitz
N
Horowitz
MM
et al
Blood stem cells compared with bone marrow as a source of hematopoietic cells for allogeneic transplantation. IBMTR Histocompatibility and Stem Cell Sources Working Committee and the European Group for Blood and Marrow Transplantation (EBMT).
Blood.
95
2000
3702
3709
42
Przepiorka
D
Anderlini
P
Ippoliti
C
et al
Allogeneic blood stem cell transplantation in advanced hematologic cancers.
Bone Marrow Transplant.
19
1997
455
460
43
Bensinger
W
Martin
P
Clift
R
et al
A prospective, randomised trial of peripheral blood stem cells (PBSC) or marrow (BM) for patients undergoing allogeneic transplantation for hematologic malignancies.
Blood.
94(suppl 1)
1999
368a
44
Ringdén
O
Labopin
M
Gluckman
E
et al
Graft-versus-leukemia effect in allogeneic marrow transplant recipients with acute leukemia is maintained using cyclosporin A combined with methotrexate as prophylaxis. Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation.
Bone Marrow Transplant.
18
1996
921
929
45
Horowitz
M
Gale
R
Sondel
P
et al
Graft-versus-leukemia reactions following bone marrow transplantation in humans.
Blood.
75
1989
555
562
46
Carlens
S
Ringdén
O
Aschan
J
et al
Risk factors in bone marrow transplant recipients with leukaemia: increased relapse risk in patients treated with ciprofloxacin for gut decontamination.
Clin Transplant.
12
1989
84
92
47
Gratwohl
A
Hermans
J
Apperly
J
et al
Acute graft-versus-host disease: grade and outcome in patients with chronic myelogenous leukemia. Working Party Chronic Leukemia of the European Group for Blood and Marrow Transplantation.
Blood.
86
1995
813
818

Author notes

Mats Remberger, Department of Clinical Immunology, F79, Huddinge University Hospital, SE-141 86 Huddinge, Sweden; e-mail:mats.remberger@impi.ki.se.

Sign in via your Institution