T cell–depleted haploidentical hematopoietic stem cell transplantation (haploHSCT) is an option to treat children with very high-risk acute lymphoblastic leukemia (ALL) lacking an HLA-identical donor. We analyzed 127 children with ALL who underwent haploHSCT in first (n = 22), second (n = 48), or third (n = 32), complete remission or in relapse (n = 25). The 5-year leukemia-free survival (LFS) was 30%, 34%, 22%, and 0%, respectively. A risk-factor analysis was performed for patients who underwent transplantation in remission (n = 102). Five-year nonrelapse mortality (NRM), relapse incidence (RI), and LFS were 37%, 36%, and 27%, respectively. A trend of improved LFS rate and decreased RI was observed for children given a graft with higher number of CD34+ cells (adjusted P = .09 and P = .07, respectively). In a multivariate analysis, haploHSCT performed in larger centers (performing ≥ 231 allotransplantations in the studied period) was associated with improved LFS rate and decreased RI (adjusted P = .01 and P = .04, respectively), adjusting for different patient-, disease-, and transplant-related factors such as number of previous autotransplantations, cytomegalovirus serology status, type of T-cell depletion, and use of total body irradiation and antithymocyte globulin. In conclusion, higher CD34+ cell dose and better patient selection may improve outcomes of children with ALL who undergo a haploHSCT. Transplant centers initiating programs on haploHSCT for children may collaborate with more experienced centers.

Allogeneic hematopoietic stem cell transplantation (HSCT) is being increasingly used for pediatric patients with acute lymphoblastic leukemia (ALL) in second or subsequent complete remission (CR) after marrow relapse, as well as in patients in first CR but with high-risk characteristics.1-7  However, HLA-identical sibling donors are not available for approximately 75% of the patients, and unrelated donors, matched at the allelic level, cannot be found in time for all patients who need an allograft. Therefore, transplantations using alternative donor sources such as unrelated cord blood8-12  or haploidentical stem cells are increasingly used.13-15  The Perugia Group has first shown, in adult patients with hematologic malignancies who received a transplant from an HLA-disparate relative, that the infusion of a large number of extensively T cell–depleted CD34+ cells ensures sustained engraftment of donor hematopoiesis and minimizes the risk of both acute and chronic graft-versus-host disease (GVHD).15,16  The feasibility of haploHSCT was reproduced also in children, in particular in patients with ALL, transplanting high numbers of positively selected stem cells.17  However, data on the role of haploHSCT in childhood ALL are still limited and the reports analyzing outcomes and risk factors that are available in the literature include a limited number of children.

With the goal to evaluate the role of haploHSCT in childhood ALL, we have studied in a retrospective, multicenter, registry-based analysis the outcomes of 127 children given a T cell–depleted allograft from an HLA-disparate relative and risk factors of 102 patients who underwent transplantation in remission in European transplant centers and reported to the Pediatric diseases and the Acute Leukemia working parties European Group for Blood and Marrow Transplantation (EBMT) registry.

Selection criteria

The study used data reported to the EBMT registry and selected the patients according to the following criteria: (1) children with a diagnosis of ALL aged 16 years or younger; (2) T cell–depleted bone marrow or peripheral blood allografts from a relative; (3) 2 or 3 antigen HLA differences at loci A, B, or DRB1 in the donor-recipient pair; (4) transplantations performed in EBMT centers from 1995 to 2004. A total of 127 patients who underwent transplantation in 36 EBMT centers met these eligibility criteria. For these patients, the missing information was retrieved by the statistical office of the EBMT from each center providing patients for analysis.

Definitions

High-resolution typing of recipient and donor HLA-C, and low-resolution typing of HLA-B and HLA-A alleles of recipient and donor was used to segregate patients and donors in the following killer immunoglobulin-like receptor (KIR) ligand groups according to the list of HLA-C, group 1 and group 2, HLA-Bw4, and HLA-Bw6 alleles by Ruggeri et al.18  A3/A11 were not included because their role in vitro is controversial. We were unable to study the influence of noninherited maternal antigens and noninherited paternal antigens with outcomes because most of the donors were parents, and there were very few siblings according to definition published by van Rood et al.19 

Center size was defined based on the total number of allogeneic HSCTs performed in children (< 16 years old) for all diseases, during the same time period, by the EBMT transplant centers that have included consecutive haploHSCT. In the EBMT megafile, we have found that the median number of allotransplantations performed by the same transplant centers was 231. The median number of transplantations was the best cutoff number that was statistically associated with leukemia-free survival (LFS) and it was used to discriminate large and small centers. Based on this definition, 8 transplant centers performing a median of 8 haploHSCTs and more than 231 allotransplantations were classified as larger centers, whereas 28 transplant centers performing a median of 1 haploHSCT and fewer than 231 allotransplantations were classified as smaller centers.

End points

Myeloid engraftment was defined as the first of 3 consecutive days when the absolute neutrophil count was 0.5 × 109/L or higher with evidence of donor hematopoiesis. Engraftment that occurred after day 60 was considered as nonengraftment. Acute GVHD was diagnosed and graded according to published criteria.20  Chronic GVHD was diagnosed according to standard criteria.21  Criteria for classification of GVHD proposed by National Institutes of Health consensus were not used in this retrospective analysis.22  Children with evidence of donor engraftment who survived more than 90 days from transplantation were evaluated for occurrence of chronic GVHD. Nonrelapse mortality (NRM) was defined as any death without evidence of relapse. Relapse was defined on the basis of morphologic evidence of leukemia in bone marrow or other sites. Survival was calculated from transplantation to death due to any cause, and leukemia-free survival (LFS) was defined as the time from transplantation to either first relapse or death in complete remission, whichever occurred first.

Statistical analysis

The duration of follow-up was the time to the last assessment for survivors. Nonrelapse mortality (NRM) and relapse incidence (RI) were expressed as cumulative incidence curves, to adjust the analysis for competing risks.23  Probabilities of LFS were calculated using the Kaplan-Meier estimate. Univariate analyses were done using log-rank test for LFS, whereas the Gray test was applied for RI and NRM. A risk-factor analysis was restricted to patients who underwent transplantation in remission because all patients who underwent transplantation in relapse died. Patients receiving a second transplant for graft failure were kept in the analysis, and their follow-up was considered after second transplantation until relapse or death. The variables considered in risk-factor analyses for NRM, RI, and LFS were as follows: recipient age, sex, and cytomegalovirus (CMV) serology; disease characteristics (cytogenetic, disease status at transplantation [CR1, CR2, CR3]), donor characteristics (age, sex, female donor to male recipient, CMV serology); transplant characteristics (number of antigen incompatibilities [2 vs > 2 of 6], previous autologous transplantation, dose of CD34+ and CD3+ cells infused, conditioning regimen including total body irradiation [TBI] or not, use of antithymocyte/antilymphocyte globulin [ATG/ALG], year of transplantation, and centers performing ≥ 231 or < 231 alloHSCTs during the period studied). KIR matching was analyzed only for patients with available information. For prognostic analyses, continuous variables were categorized as follows: each variable was first divided into 5 categories at approximately the 20th, 40th, 60th, and 80th percentiles. In the absence of a clear pattern among the 5 categories, the median was used as a cutoff point. This approach allows distinguishing 2 types of centers according to the number of allotransplantations performed during the period studied. Multivariate analyses were performed using Cox proportional-hazard regression model, including all factors associated with a P value less than .2 by univariate analysis and all factors statistically different among the 2 types of centers (P < .10). Then a stepwise backward procedure was used with a cutoff significance level of .05 for removal of factors from the model. P values are 2-sided, with a type I error rate fixed at .05. Statistical analyses were performed with SPSS 17.0 and R 2.9.0 software.

Patient, donor, and disease characteristics

Table 1 lists the characteristics of the 102 children who underwent transplantation in remission. Median age of children at time of transplantation was 8.7 years (range, 0.6-16 years); the median number of white blood cell count at diagnosis was 27 × 109/L (range, 2-517 × 109/L). Twenty-five (29%) of the 86 patients with cytogenetic data available had high-risk ALL defined by the presence of t(9;21) or t(4;11). At transplantation, 22 (22%) patients were in first complete remission (CR1); 48 (47%), in CR2; and 32 (31%), in CR3. In fact, of 22 children who underwent transplantation in CR1, 16 patients had poor cytogenetics (t(9;22) in 10 patients, t(4;11) in 5 patients, and monosomy 7 in 1 patient). Three patients had white blood cell count at diagnosis higher than 190 × 109/L (1 had hypodiploidy), and 3 patients achieved CR after 80 days of induction. For 48 patients who underwent transplantation in CR2, 35% of the patients relapsed before 18 months after achieving first CR; 41%, between 18 and 30 months; and 24% had later first relapse but poor cytogenetics, including more frequently t(9;22) or t(4;11). All patients who obtained CR3 were offered a haploHSCT, independent of the ALL diagnosis characteristics or time to relapse. Among them, 6 patients had been previously autografted.

Table 1

Patient characteristics: patients in remission (n = 102)

CharacteristicNo.
Patient-related factors  
    Age at transplantation, y (range) 8.7 (0.64-16) 
    Sex, male/female 70/32 
    CMV serology  
        Negative 40 
        Positive 59 
        Not known 
Donor-related factors  
    Age at transplantation, y (range) 36.6 (18-53.8) 
    Sex, male/female 55/47 
    Female donor to male recipient  
        No 71 
        Yes 31 
    CMV serology  
        Negative 32 
        Positive 61 
        Not known 
Disease-related factors  
    WBC at diagnosis × 109/L 27.2 (1.9-517) 
    Immunophenotype  
        B 84 
        T 11 
        Biphenotypic 
        Not known 
    Cytogenetic  
        No Ph+ 63 
        Ph+ or t(4;11) 24 
        Not performed/not available 15 
    Status at transplantation  
        CR1 22 (22%) 
        CR2 48 (47%) 
        CR3 32 (31%) 
    Median time from first diagnosis to transplantation, mo  
        CR1 6 (3-16) 
        CR2 32 (6-80) 
        CR3 57 (11-141) 
    Previous autologous transplantation  
        No 94 
        Yes 
Transplant-related factors  
    Year of transplantation  
        1995 
        1996 
        1997 
        1998 14 
        1999 11 
        2000 17 
        2001 10 
        2002 10 
        2003 14 
        2004 11 
        Median, y 2000 
    Donor type  
        Mother 44 
        Father 51 
        Brother 
        Sister 
    No. of antigen mismatches in HLA-A, -B, -DR  
        2 34 
        3 68 
    KIR mismatches between donor and recipients  
        Yes 25 
        No 41 
        Not known 36 
    Cell source  
        BM 
        PB 96 
    In vitro T-cell depletion, device used  
        CliniMacs 79 
        Baxter 12 
        Cellpro 
        Negative T depletion 
    Serotherapy with ATG or ALG  
        No 22 
        Yes 76 
        Not known 
    Use of monoclonal antibodies  
        No 57 
        Yes 44 
        Not known 
    Total body irradiation  
        No 24 
        Yes 78 
    No. of CD34+ × 106/kg body weight  
        All 12.33 (1.36-95.2) 
        PB only 12.4 (1.9-67) 
    No. of CD3+ × 106/kg body weight*  
        All 0.05 (0-129) 
        PB only 0.045 (0-128.7) 
    Donor lymphocyte infusion performed  
        No 86 
        Yes 16 
CharacteristicNo.
Patient-related factors  
    Age at transplantation, y (range) 8.7 (0.64-16) 
    Sex, male/female 70/32 
    CMV serology  
        Negative 40 
        Positive 59 
        Not known 
Donor-related factors  
    Age at transplantation, y (range) 36.6 (18-53.8) 
    Sex, male/female 55/47 
    Female donor to male recipient  
        No 71 
        Yes 31 
    CMV serology  
        Negative 32 
        Positive 61 
        Not known 
Disease-related factors  
    WBC at diagnosis × 109/L 27.2 (1.9-517) 
    Immunophenotype  
        B 84 
        T 11 
        Biphenotypic 
        Not known 
    Cytogenetic  
        No Ph+ 63 
        Ph+ or t(4;11) 24 
        Not performed/not available 15 
    Status at transplantation  
        CR1 22 (22%) 
        CR2 48 (47%) 
        CR3 32 (31%) 
    Median time from first diagnosis to transplantation, mo  
        CR1 6 (3-16) 
        CR2 32 (6-80) 
        CR3 57 (11-141) 
    Previous autologous transplantation  
        No 94 
        Yes 
Transplant-related factors  
    Year of transplantation  
        1995 
        1996 
        1997 
        1998 14 
        1999 11 
        2000 17 
        2001 10 
        2002 10 
        2003 14 
        2004 11 
        Median, y 2000 
    Donor type  
        Mother 44 
        Father 51 
        Brother 
        Sister 
    No. of antigen mismatches in HLA-A, -B, -DR  
        2 34 
        3 68 
    KIR mismatches between donor and recipients  
        Yes 25 
        No 41 
        Not known 36 
    Cell source  
        BM 
        PB 96 
    In vitro T-cell depletion, device used  
        CliniMacs 79 
        Baxter 12 
        Cellpro 
        Negative T depletion 
    Serotherapy with ATG or ALG  
        No 22 
        Yes 76 
        Not known 
    Use of monoclonal antibodies  
        No 57 
        Yes 44 
        Not known 
    Total body irradiation  
        No 24 
        Yes 78 
    No. of CD34+ × 106/kg body weight  
        All 12.33 (1.36-95.2) 
        PB only 12.4 (1.9-67) 
    No. of CD3+ × 106/kg body weight*  
        All 0.05 (0-129) 
        PB only 0.045 (0-128.7) 
    Donor lymphocyte infusion performed  
        No 86 
        Yes 16 

CMV indicates cytomegalovirus; WBC, white blood cell count; CR, complete remission; Ph+, Philadelphia chromosome positive; ATG, antithymocyte globulin; ALG, antilymphocyte globulin; PB, peripheral blood; and BM, bone marrow.

*

n = 10 not known.

Sixty-eight patients received a graft with 3 HLA antigen disparities (Table 1). In 66 patients, sufficient information on HLA-C, -Bw4, and -Bw6 typing was available to classify patients into KIR ligand matching between donors and recipients. KIR ligand was matched with the donor in the GVHD direction in 41 patients and mismatched in 25 patients.

The donors were mothers in 44 cases, fathers in 51 cases, brothers in 5 cases, and sisters in 2 cases.

Transplantation and graft cell selection

Hematopoietic stem cells were collected either by the transplant center or at the local transfusion service and processed by cellular therapy laboratories. All patients but 6, who received a bone marrow cell transplant, were given T cell–depleted peripheral blood (PB) progenitor cells, which were mobilized in the donor through the use of granulocyte colony-stimulating factor and collected by a cell separator. In most cases, CD34+ cells were positively selected using the CliniMacs one-step procedure (Miltenyi Biotec). The median number of CD34+ and CD3+ cells infused using PB was 12.3 × 106/kg (range, 1.36-95 × 104/kg) and 5.0 × 104/kg (range, 0-12.9 × 104/kg). In vivo T-cell depletion by ATG or ALG was used in 78% of reported transplantations. In vivo use of monoclonal antibodies was added in 44 children. All patients received a myeloablative conditioning regimen; total body irradiation was used in 76% of patients (Table 1).

A total of 127 children with ALL underwent haploHSCT from 1995 to 2004 in 36 EBMT centers. Twenty-two received a transplant in first complete remission (CR1); 48, in second CR (CR2); and 32, in third CR (CR3); and 25 were not in remission. According to disease phase, the 5-year probability of LFS after haploHSCT was 30% (± 10%), 34% (± 7%), 22% (± 7%), and 0%, respectively. Because all 25 children with active disease who underwent transplantation died, description of the population and risk factor analyses were restricted to patients in remission.

Outcomes

Neutrophil recovery.

Engraftment was observed in 92 patients (91%). Median time to reach a neutrophil count higher than 500/mm3 was 15 days (range, 8-55 days). Chimerism data were not obtained from 9 patients who died early or were in aplasia, but data were available for 87 of 93 patients; 9% of the patients experienced autologous reconstitution (n = 8); 10%, mixed chimerism (n = 9); and 80%, full donor chimerism (n = 70) in the 3 months after transplantation. Primary graft failure was observed in 9 and secondary graft failure in 4 children. A second transplantation was performed in 12 patients, and second engraftment was successful in 10.

Acute and chronic graft-versus-host disease.

Acute GVHD was absent or grade 1 in 80 children, grade 2 in 13, grade 3 in 6, and grade 4 in 3 cases, and chronic GVHD was observed in 14 of the 84 patients at risk. Preemptive DLI was given for only 1 child who has not developed GVHD.

Relapse incidence.

The cumulative incidence of hematologic relapse at 5 years was 36% (± 4%). Table 2 lists the univariate analysis for RI. None of the factors studied in univariate analysis were statistically associated with RI. KIR incompatibilities were not associated with RI (Table 2). Relapse cumulative incidence at 5 years was 32% (± 8%) for patients with no incompatibility and 36% (± 10%) for those with KIR incompatibilities in the GVHD direction (P = .55). Of note, RI was 41% (± 7%) for those children who underwent transplantation in smaller centers and 31% (± 6%) for those who underwent transplantation in larger centers (P = .26, Figure 1B). However, after adjusting for other prognostic factors (Table 2) and the differences between smaller and larger centers (Table 3), RI was decreased in larger centers (hazard ratio [HR]: 0.49, 95% confidence interval [95% CI]: 0.24-0.98; P = .04). In addition, in a multivariate model, there was a trend of decreased RI for children with higher number of CD34+ cells who underwent transplantation (HR: 0.52, 95% CI: 0.25-1.06; P = .07).

Table 2

Univariate analysis for outcomes after haploHSCT for childhood ALL in remission (n = 102): 5-year results

No.LFS, %RI, %NRM, %
Overall 102 27 ± 5 36 ± 4 37 ± 4 
Patient-related factors     
    Age, y     
        Younger than 8.7 51 30 ± 7 30 ± 6 39 ± 6 
        8.7 or older 51 24 ± 6 42 ± 6 34 ± 6 
        P  .43 .37 .93 
    Sex     
        Male 70 28 ± 6 35 ± 5 37 ± 5 
        Female 32 25 ± 8 40 ± 10 35 ± 9 
        P  .96 .59 .69 
    CMV serology     
        Negative 40 25 ± 8 47 ± 8 28 ± 8 
        Positive 59 28 ± 6 30 ± 6 42 ± 6 
        P  .34 .15 .053 
Donor-related factors     
    Age, y     
        Younger than median 45 31 ± 7 34 ± 7 35 ± 7 
        Older than median 45 23 ± 7 40 ± 7 37 ± 7 
        P  .48 .67 .7 
    Sex     
        Male 55 34 ± 7 28 ± 6 38 ± 7 
        Female 47 19 ± 6 45 ± 7 36 ± 8 
        P  .26 .08 .67 
    CMV serology     
        Negative 32 24 ± 8 42 ± 8 35 ± 9 
        Positive 61 30 ± 6 31 ± 6 39 ± 6 
        P  .8 .31 .35 
Disease-related factors     
    Status at transplantation     
        CR1 22 15 ± 12 33 ± 10 52 ± 18 
        CR2 48 34 ± 7 36 ± 6 30 ± 7 
        CR3 32 22 ± 7 37 ± 9 42 ± 9 
        P  .26 .99 .26 
    Cytogenetics     
        No Ph+ 63 29 ± 6 38 ± 7 33 ± 7 
        Ph+ and t(4;11) 24 32 ± 10 30 ± 10 39 ± 9 
        P  .83 .71 .63 
    Previous autologous transplantation     
        No 94 29 ± 5 38 ± 5 34 ± 5 
        Yes 0 (1vv) 75 ± 15 
        P  .12 .18 .002* 
Transplant-related factors     
    No. of HLA antigen mismatches in -A, -B, -DR     
        2 34 18 ± 9 44 ± 9 39 ± 12 
        3 68 30 ± 6 32 ± 6 38 ± 6 
        P  .3 .18 .73 
    KIR (C and Bw4) mismatch     
        No 41 32 ± 8 32 ± 8 36 ± 8 
        Yes 25 36 ± 10 36 ± 10 28 ± 9 
        P  .82 .55 .59 
    Female donor to male recipient     
        No 71 31 ± 6 31 ± 5 38 ± 6 
        Yes 31 18 ± 8 47 ± 9 35 ± 10 
        P  .58 .2 .62 
    Year of transplantation     
        2000 or earlier 57 26 ± 6 36 ± 6 34 ± 6 
        Later than 2000 45 25 ± 7 38 ± 8 37 ± 7 
        P  .73 .98 .71 
    Use of ATG/ALG     
        No 22 32 ± 10 32 ± 11 36 ± 11 
        Yes 76 24 ± 6 38 ± 5 38 ± 6 
        P  .67 .61 .99 
    Conditioning with TBI     
        No 24 20 ± 9 35 ± 11 44 ± 11 
        Yes 78 30 ± 5 36 ± 5 34 ± 5 
        P  .29 .99 .36 
    CD34+ cell dose × 106/kg body weight     
        Lower than 12.4 48 17 ± 6 45 ± 8 38 ± 7 
        12.4 or higher 48 35 ± 7 27 ± 7 37 ± 7 
        P  .06 .13 .74 
    CD3+ cell dose × 106/kg body weight     
        Lower than 0.05 48 26 ± 7 30 ± 7 41 ± 8 
        0.05 or higher 42 28 ± 7 38 ± 8 31 ± 8 
        P  .98 .35 .35 
Center-related factor     
    No. of alloHSCTs performed     
        Fewer than 231 45 15 ± 6 41 ± 7 44 ± 7 
        231 or more 57 39 ± 7 31 ± 6 30 ± 6 
        P  .005* .26 .17 
No.LFS, %RI, %NRM, %
Overall 102 27 ± 5 36 ± 4 37 ± 4 
Patient-related factors     
    Age, y     
        Younger than 8.7 51 30 ± 7 30 ± 6 39 ± 6 
        8.7 or older 51 24 ± 6 42 ± 6 34 ± 6 
        P  .43 .37 .93 
    Sex     
        Male 70 28 ± 6 35 ± 5 37 ± 5 
        Female 32 25 ± 8 40 ± 10 35 ± 9 
        P  .96 .59 .69 
    CMV serology     
        Negative 40 25 ± 8 47 ± 8 28 ± 8 
        Positive 59 28 ± 6 30 ± 6 42 ± 6 
        P  .34 .15 .053 
Donor-related factors     
    Age, y     
        Younger than median 45 31 ± 7 34 ± 7 35 ± 7 
        Older than median 45 23 ± 7 40 ± 7 37 ± 7 
        P  .48 .67 .7 
    Sex     
        Male 55 34 ± 7 28 ± 6 38 ± 7 
        Female 47 19 ± 6 45 ± 7 36 ± 8 
        P  .26 .08 .67 
    CMV serology     
        Negative 32 24 ± 8 42 ± 8 35 ± 9 
        Positive 61 30 ± 6 31 ± 6 39 ± 6 
        P  .8 .31 .35 
Disease-related factors     
    Status at transplantation     
        CR1 22 15 ± 12 33 ± 10 52 ± 18 
        CR2 48 34 ± 7 36 ± 6 30 ± 7 
        CR3 32 22 ± 7 37 ± 9 42 ± 9 
        P  .26 .99 .26 
    Cytogenetics     
        No Ph+ 63 29 ± 6 38 ± 7 33 ± 7 
        Ph+ and t(4;11) 24 32 ± 10 30 ± 10 39 ± 9 
        P  .83 .71 .63 
    Previous autologous transplantation     
        No 94 29 ± 5 38 ± 5 34 ± 5 
        Yes 0 (1vv) 75 ± 15 
        P  .12 .18 .002* 
Transplant-related factors     
    No. of HLA antigen mismatches in -A, -B, -DR     
        2 34 18 ± 9 44 ± 9 39 ± 12 
        3 68 30 ± 6 32 ± 6 38 ± 6 
        P  .3 .18 .73 
    KIR (C and Bw4) mismatch     
        No 41 32 ± 8 32 ± 8 36 ± 8 
        Yes 25 36 ± 10 36 ± 10 28 ± 9 
        P  .82 .55 .59 
    Female donor to male recipient     
        No 71 31 ± 6 31 ± 5 38 ± 6 
        Yes 31 18 ± 8 47 ± 9 35 ± 10 
        P  .58 .2 .62 
    Year of transplantation     
        2000 or earlier 57 26 ± 6 36 ± 6 34 ± 6 
        Later than 2000 45 25 ± 7 38 ± 8 37 ± 7 
        P  .73 .98 .71 
    Use of ATG/ALG     
        No 22 32 ± 10 32 ± 11 36 ± 11 
        Yes 76 24 ± 6 38 ± 5 38 ± 6 
        P  .67 .61 .99 
    Conditioning with TBI     
        No 24 20 ± 9 35 ± 11 44 ± 11 
        Yes 78 30 ± 5 36 ± 5 34 ± 5 
        P  .29 .99 .36 
    CD34+ cell dose × 106/kg body weight     
        Lower than 12.4 48 17 ± 6 45 ± 8 38 ± 7 
        12.4 or higher 48 35 ± 7 27 ± 7 37 ± 7 
        P  .06 .13 .74 
    CD3+ cell dose × 106/kg body weight     
        Lower than 0.05 48 26 ± 7 30 ± 7 41 ± 8 
        0.05 or higher 42 28 ± 7 38 ± 8 31 ± 8 
        P  .98 .35 .35 
Center-related factor     
    No. of alloHSCTs performed     
        Fewer than 231 45 15 ± 6 41 ± 7 44 ± 7 
        231 or more 57 39 ± 7 31 ± 6 30 ± 6 
        P  .005* .26 .17 

LFS indicates leukemia-free survival; RI, relapse incidence; NRM, nonrelapse mortality; CMV, cytomegalovirus; WBC, white blood cell count; CR, complete remission; Ph+, Philadelphia chromosome–positive; ATG, antithymocyte globulin; ALG, antilymphocyte globulin; and TBI, total body irradiation.

*

Statistically significant.

Figure 1

(A) Leukemia-free survival after haploHSCT in children with ALL according to number of alloHSCTs performed in participating transplant centers. (B) Cumulative incidence of relapse after haploHSCT in children with ALL according to number of alloHSCTs performed in participating transplant centers. (C) Cumulative incidence of nonrelapse mortality in patients with ALL in remission only, according to number of haploHSCTs.

Figure 1

(A) Leukemia-free survival after haploHSCT in children with ALL according to number of alloHSCTs performed in participating transplant centers. (B) Cumulative incidence of relapse after haploHSCT in children with ALL according to number of alloHSCTs performed in participating transplant centers. (C) Cumulative incidence of nonrelapse mortality in patients with ALL in remission only, according to number of haploHSCTs.

Close modal
Table 3

Patient-, donor-, disease-, and transplant-related factors in 102 children who underwent haploHSCT for ALL in remission, according to number of transplantations performed during the study period

Fewer than 231 alloHSCTs performed231 or more alloHSCTs performedP
Patient-related factors    
    Age, y 8.8 (0.96-16) 8.7 (0.64-15.7) .64 
    Sex    
        Male 32 38 .63 
        Female (%) 13 (29) 19 (33)  
    CMV serology    
        Negative 32 < .001* 
        Positive (%) 35 (81) 24 (43)  
Donor-related factors    
    Age, y 36 (18.6-49.5) 36.9 (18-53.8) .8 
    Sex    
        Male 20 35 .09 
        Female (%) 25 (56) 22 (39)  
    CMV serology    
        Negative 24 .02* 
        Positive (%) 30 (79) 31 (56)  
Disease-related factors    
    WBC at diagnosis 21.25 (2.8-517) 27.9 (1.9-272.4) .67 
    Cytogenetics    
        No Ph+ 27 36 .25 
        Ph+ or t(4;11) (%) 7 (21) 17 (32)  
    Previous autologous transplantation    
        No 39 55 .07 
        Yes (%) 6 (13) 2 (4)  
    Status at transplantation (%)    
        CR1 6 (13) 16 (28) .2 
        CR2 23 (51) 25 (44)  
        CR3 16 (36) 16 (28)  
Transplant-related factors    
    Year of transplantation 2000 (1995-2004) 2000 (1995-2004) .8 
    No. of antigen mismatch in HLA-A, -B, -DR    
        2 17 17 .31 
        3 (%) 28 (62) 40 (70)  
    Female donor to male recipient    
        No 28 43 .15 
        Yes (%) 17 (38) 14 (25)  
    Cell source    
        BM .77 
        PB (%) 42 (93) 54 (95)  
    Type of T-cell depletion (%)    
        CliniMacs 30 (68) 48 (84) .04* 
        Isolex 9 (21) 3 (5)  
        Cellpro 4 (9) 2 (4)  
        Negative T-depletion 1 (2) 4 (7)  
    No. of CD34+ cells infused ×106/kg body weight 12.7 (1.36-95.2) 12.1 (3-33) .68 
    TBI in conditioning    
        No 16 .01* 
        Yes (%) 29 (64) 49 (86)  
    ATG/ALG in conditioning    
        No 18 .01* 
        Yes (%) 38 (90) 38 (68)  
Fewer than 231 alloHSCTs performed231 or more alloHSCTs performedP
Patient-related factors    
    Age, y 8.8 (0.96-16) 8.7 (0.64-15.7) .64 
    Sex    
        Male 32 38 .63 
        Female (%) 13 (29) 19 (33)  
    CMV serology    
        Negative 32 < .001* 
        Positive (%) 35 (81) 24 (43)  
Donor-related factors    
    Age, y 36 (18.6-49.5) 36.9 (18-53.8) .8 
    Sex    
        Male 20 35 .09 
        Female (%) 25 (56) 22 (39)  
    CMV serology    
        Negative 24 .02* 
        Positive (%) 30 (79) 31 (56)  
Disease-related factors    
    WBC at diagnosis 21.25 (2.8-517) 27.9 (1.9-272.4) .67 
    Cytogenetics    
        No Ph+ 27 36 .25 
        Ph+ or t(4;11) (%) 7 (21) 17 (32)  
    Previous autologous transplantation    
        No 39 55 .07 
        Yes (%) 6 (13) 2 (4)  
    Status at transplantation (%)    
        CR1 6 (13) 16 (28) .2 
        CR2 23 (51) 25 (44)  
        CR3 16 (36) 16 (28)  
Transplant-related factors    
    Year of transplantation 2000 (1995-2004) 2000 (1995-2004) .8 
    No. of antigen mismatch in HLA-A, -B, -DR    
        2 17 17 .31 
        3 (%) 28 (62) 40 (70)  
    Female donor to male recipient    
        No 28 43 .15 
        Yes (%) 17 (38) 14 (25)  
    Cell source    
        BM .77 
        PB (%) 42 (93) 54 (95)  
    Type of T-cell depletion (%)    
        CliniMacs 30 (68) 48 (84) .04* 
        Isolex 9 (21) 3 (5)  
        Cellpro 4 (9) 2 (4)  
        Negative T-depletion 1 (2) 4 (7)  
    No. of CD34+ cells infused ×106/kg body weight 12.7 (1.36-95.2) 12.1 (3-33) .68 
    TBI in conditioning    
        No 16 .01* 
        Yes (%) 29 (64) 49 (86)  
    ATG/ALG in conditioning    
        No 18 .01* 
        Yes (%) 38 (90) 38 (68)  

CMV indicates cytomegalovirus; WBC, white blood cell count; Ph+, Philadelphia chromosome–positive; CR, complete remission; BM, bone marrow; PB, peripheral blood; TBI, total body irradiation; ATG, antithymocyte globulin; and ALG, antilymphocyte globulin.

*

Statistically significant.

Nonrelapse mortality.

The cumulative incidence of NRM at 5 years was 37% (± 4%). Table 2 lists the univariate analysis of factors potentially influencing NRM. A higher cumulative incidence of NRM (75% ± 15%) was observed in the 8 children who had previously received an autologous transplant compared with 34% (± 5%) for the remaining patients (P = .002). In univariate analysis, recipient's positive CMV serology and previous autologous transplantation were associated with higher NRM (Table 2), however in a multivariate analysis, only previous autograft was associated with increased NRM (HR: 3.01, 95% CI: 1.16-7.84; P = .024). No other patient-, disease-, donor-, or transplantation-related variable or size of transplant center was statistically associated with NRM (Figure 1C).

Leukemia-free survival.

The estimate probability of LFS at 5 years of patients who underwent transplantation in CR was 27% (± 5%). Factors such as KIR matching and donor type were not associated with LFS, as shown in Table 2. In univariate (Table 2 and Figure 1A) and in multivariate analyses, size of the center was significantly associated with LFS (HR: 1.9, 95% CI: 1.15-3.11; P = .01). There was also a trend of improved LFS rate in univariate (P = .06, Figure 2) and multivariate analyses in children given a higher CD34+ cell dose than 12.4 × 106/kg (HR: 1.53, 95% CI: 0.93-2.52; P = .09).

Figure 2

Leukemia-free survival of patients with ALL in remission according to CD34+ cell dose in the graft.

Figure 2

Leukemia-free survival of patients with ALL in remission according to CD34+ cell dose in the graft.

Close modal

Survival and causes of death.

The estimated probability of overall survival (OS) at 5 years was 29% (± 5%) for the whole cohort, 28% (± 10%) for patients who underwent transplantation in CR1, 39% (± 7%) for those in CR2, and 32% (± 8%) for those in CR3. Five-year OS was 38% (± 6%) in larger centers (n = 57, deaths = 34) and 18% (± 6%) in smaller centers (n = 45, deaths = 35; P = .008). Thirty-one patients died of recurrence of the disease, 16 in larger centers and 15 in smaller centers, and 22 died of infection (11 and 11 according to the center size, respectively). Other causes of death were GVHD in 9 cases and interstitial pneumonitis in 5 patients.

Impact of center size on outcomes

The differences in terms of patient-, donor-, and transplantation-related factors between large and small centers are shown in Table 3. In smaller centers, children were more likely to have CMV-positive serology (P < .001), to have previously undergone an autologous HSCT (P = .07), to have received ATG/ALG in the preparative regimen (P = .01), and to have undergone transplantation without TBI (P = .01). In addition, the technique of T-cell depletion was different because larger centers more frequently used CliniMacs device (P = .04). All of these differences were taken into consideration in the multivariate model for outcomes.

Center size, defined as the total number of allotransplantations performed during the studied period in transplant center, was associated in univariate analysis with LFS (Table 2). After statistical adjustments of the differences between larger and smaller centers RI was decreased (HR: 0.49, 95% CI: 0.24-0.98; P = .04) and LFS improved (HR: 1.9, 95% CI: 1.15-3.11; P = .01) in larger transplant centers.

Most reports in the literature regarding haploidentical transplantations include a limited number of patients and those reports are heterogeneous because they include patients with malignant and nonmalignant diseases, adolescents and young adults, as well as patients who received a transplant from one antigen–disparate related donor, whose outcome is known to be similar to matched donor transplantations.14,15,17,24-31  All of these factors preclude the possibility of drawing a reliable conclusion on the role of haploHSCT in specific pediatric hematologic disorders. We analyzed a homogenous cohort of children (≤ 16 years) who underwent transplantation over a 10-year period in European Group for Blood and Marrow Transplantation (EBMT) centers using related donors with at least 2 HLA antigen disparities and after a relatively homogenous procedure of T-cell depletion, namely positive selection of CD34+ cells.

The first message of our study is that haploHSCT should not be considered an option if complete remission is not achieved before transplantation. In fact, although the 5-year probability of LFS after haploHSCT was 27% for patients who underwent transplantation in remission, it was 0% for children with active/resistant disease who underwent transplantation.

LFS after other strategies, such as unrelated donors or unrelated cord blood transplantation, for children with ALL lacking a matched sibling donors varies between 30% and 58% in various series.2,4,8  Whether outcomes after haploHSCT are comparable with those strategies is not known and should be further investigated. It must be emphasized that those comparisons are possible only after adjustments for differences among these strategies, such as the waiting time for locating a suitable donor. Importantly, retrospective studies cannot take into consideration the previous selection of patients given that the main advantage of haploHSCT over unrelated donor transplantation is the easy, rapid access to donor and the possibility of using cells for immunotherapy.

The second aim of our study was to analyze risk factors affecting the outcomes of patients who undergo haploHSCT in remission. Interestingly, in our study, LFS did not differ among patients who underwent transplantation in CR1, CR2, or CR3, suggesting that even patients who undergo transplantation in early disease phase represent a very high-risk population. The quite low survival rate of 30% in patients who received a transplant in first CR is a concern. One could argue that the reason for this poor LFS is related to a very high-risk group of patients with a high frequency of poor-risk cytogenetics (16/22 patients had poor cytogenetics [t(9;22) in 10 patients, t(4;11) in 5 patients, and monosomy 7 in 1 patient], 3 patients had white blood cell count at diagnosis higher than 190 × 109/L [1 hypodiploidy], and 3 patients achieved CR after 80 days of induction). Another speculative explanation for the poor result is the lack of graft-versus-leukemia effect needed in this specific group. It is difficult to draw definitive conclusions of the place of haploHSCT in CR1 patients based on this low number of patients, but indication of haploidentical HSCT in CR1 patients should be properly evaluated.

Increased NRM was observed in children who had received a previous autologous transplant. This is not unexpected because patients who already underwent myeloablative therapy have a higher burden of toxicity, eventually resulting in death. In univariate analysis, a recipient's positive CMV serology was associated with higher NRM, but his/her factor was not isolated in the final multivariate model. Of note, none of the graft-related factors, such as type and number of HLA differences in the haplotype, transplant from a female donor to a male recipient, year of transplantation, use of serotherapy, the type of conditioning, and the number of CD3+ cells, had influence on the cumulative incidence of NRM.

Importantly, in the multivariate analysis, we have found that there was a trend of improved LFS rate and decreased RI in patients receiving higher number of CD34+ cells in the graft. This is an important finding, because the number of hematopoietic progenitors is, to some degree, under the control of clinicians. However, the experimental data at the basis of the “megadose” concept and identifying the magnitude of CD34+ cell dose as critical for successfully overcoming the HLA barrier, and in establishing tolerance, provide support to the conclusion that, whenever possible, the maximum number of CD34+ cells should be infused.32,33  Whether a higher CD34+ cell dose infused is associated with a higher graft-versus-leukemia effect has to be further investigated. In accordance with this hypothesis, the Tübingen group has demonstrated that a CD34+ cell dose greater than 20 × 106/kg is associated with a faster immunologic recovery.34 

In contrast to other investigators35-40  who did describe a benefit for patients with KIR ligand mismatch and alloreactive natural killer cells in the haploidentical setting, we did not find any impact of KIR mismatch between donor and recipient in our study for ALL. However this finding should be taken with caution because the number of patients with available information on KIR ligand was small. Moreover, some other factors such as use of ATG/ALG could negatively impact the KIR ligand effect. Another donor-related factor that we would expect to be associated with outcomes was type of related donor. Recently, a mother donor was associated with better survival in 118 patients with leukemia.35  Those patients underwent transplantation in 2 centers and were more frequently adults with acute myeloid leukemia. This finding was not observed in our study: LFS was 19% (± 6%) when the donor was a female, and it was 34% (± 7%) when the donor was a male (P = not significant).

An important finding of our study was the association of outcomes, LFS and RI, with the center size. The “center-effect” issue is known in transplantation medicine,41,42  and it has been associated with outcomes after HSCT in adults43 ; however in pediatric HSCT, it has not been appropriately studied. Indeed, the outcome of bone marrow transplantation for acute myeloid leukemia in CR1 was demonstrated to be influenced by the center at which the procedure was performed, even after adjustment for known prognostic risk factors.43  In a first analysis, we have found that patients who underwent transplantation in centers performing more than 9 haploHSCTs during the studied period had improved LFS compared with transplant centers performing fewer (data not shown). To better define center size, we addressed the question of whether those transplant centers performing more allogeneic HSCTs during the period study would change the analysis. Once more, we found that center size (defined as more than or fewer than 231 allotransplantations performed during the period study) was associated with outcomes of haploHSCT.

We found that several variables were differently distributed between larger and smaller centers (Table 3). These differences may have all contributed to the better results obtained in the centers with larger activity; however, the center size differences cannot be explained only by a confounding factor related to patient selection or a specific therapy administrated at the transplant center. In fact, use of TBI, use of ATG, as well as patient's CMV serology or previous autologous transplantation were not associated with outcome in univariate or multivariate analyses, and cannot explain the findings related to center size. This observation indicates that many factors related to the center cannot be measured, such as personal experience of physicians and nurses that lead to good choice of patients, timing and transplantation procedure, infrastructure of laboratories, and infection surveillance. We also think that in future studies of haploHSCT, it is important that center effect should be further considered and used to adjust for prognostic factors. Finally, center size or experience effect is found to be the most important factor associated with outcomes in this setting, independent of any other prognostic factor analyzed. We strongly believe in the importance of this message in helping to improve patient outcomes and, importantly, if transplant centers want to initiate a haploHSCT program, they need training and sharing of knowledge from experienced centers.

Despite the number of patients analyzed, our findings should be confirmed by other groups because of the retrospective and multicenter design of our study; in addition, the time period (1995-2004) may be too long considering the rapid development of transplantation medicine.44  We cannot conclude that factors not reaching statistical significance as prognostic factors had no impact on outcome because this may be due to a lack of power. Their impact should be tested in larger series of patients. Moreover, some more recent data have described the impact of minimal residual disease and use of tyrosine kinase inhibitors before and after transplantation,45-47  but unfortunately it was not possible to evaluate in our retrospective analysis.

In conclusion, haploHSCT is an alternative option to treat children with very high-risk ALL lacking an HLA-identical donor and should be considered whenever there is an indication to allogeneic transplantation,17,30  and not as the last option in relapse of the disease. The final outcome of patients undergoing a haploHSCT may be improved by increasing the number of CD34+ cells infused and performing transplantation in experienced centers with programs dedicated to this type of allograft. Innovative procedures of progenitor cell selection, such as those based on the negative depletion of CD3/CD19 cells,48,49  use of natural killer alloreactive donors,18  as well as strategies of adoptive cell therapy with T-cell lines or clones aimed at enhancing the graft-versus-leukemia and pathogen-specific immune reconstitution may also contribute to ameliorate the outcome of children undergoing a haploHSCT.3,50  Exciting perspectives for haploHSCT may also result from cotransplantation of mesenchymal stem cells.51 

An Inside Blood analysis of this article appears at the front of this issue.

The online version of this article contains a data supplement.

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 USC section 1734.

Contribution: T.K. wrote the paper, worked on study design, and contributed patients; J.C. worked on study design, participated in the writing committee, and contributed patients; M.L. designed and performed statistical analysis; F.L., P.D., R.H., A.B., J.O.-L., F.F., R.O., C.P., F.A., and G.D. contributed clinical data and worked on the writing committee; E.P. collected and checked clinical data; and V.R. wrote the paper, worked on study design, verified and confirmed clinical data, and helped M.L. perform statistical analysis.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

For a complete list of participants from the Acute Leukaemia and Paediatric Diseases Working Parties of European Blood and Marrow Transplant Group Registry, see the supplemental Appendix (available on the Blood Web site; see the Supplemental Materials link at the top of the online article).

Correspondence: Thomas Klingebiel, Theodor Stern Kai 7, Klinik für Kinder-und Jugendmedizin III, Johann Wolfgang Goethe Universität, 60590 Frankfurt, Germany; e-mail: thomas.klingebiel@kgu.de.

1
Dopfer
 
R
Henze
 
G
Bender-Gotze
 
C
, et al. 
Allogeneic bone marrow transplantation for childhood acute lymphoblastic leukemia in second remission after intensive primary and relapse therapy according to the BFM- and CoALL-protocols: results of the German Cooperative Study.
Blood
1991
, vol. 
78
 
10
(pg. 
2780
-
2784
)
2
Borgmann
 
A
Von Stackelberg
 
A
Hartmann
 
R
, et al. 
Unrelated donor stem cell transplantation compared with chemotherapy for children with acute lymphoblastic leukemia in a second remission: a matched-pair analysis.
Blood
2003
, vol. 
101
 
10
(pg. 
3835
-
3839
)
3
Bader
 
P
Kreyenberg
 
H
Hoelle
 
W
, et al. 
Increasing mixed chimerism is an important prognostic factor for unfavorable outcome in children with acute lymphoblastic leukemia after allogeneic stem-cell transplantation: possible role for pre-emptive immunotherapy?
J Clin Oncol
2004
, vol. 
22
 
9
(pg. 
1696
-
1705
)
4
Balduzzi
 
A
Valsecchi
 
MG
Uderzo
 
C
, et al. 
Chemotherapy versus allogeneic transplantation for very-high-risk childhood acute lymphoblastic leukaemia in first complete remission: comparison by genetic randomisation in an international prospective study.
Lancet
2005
, vol. 
366
 
9486
(pg. 
635
-
642
)
5
Lang
 
P
Handgretinger
 
R
Niethammer
 
D
, et al. 
Transplantation of highly purified CD34+ progenitor cells from unrelated donors in pediatric leukemia.
Blood
2003
, vol. 
101
 
4
(pg. 
1630
-
1636
)
6
Peters
 
C
Schrauder
 
A
Schrappe
 
M
, et al. 
Allogeneic haematopoietic stem cell transplantation in children with acute lymphoblastic leukaemia: the BFM/IBFM/EBMT concepts.
Bone Marrow Transplant
2005
, vol. 
35
 
suppl 1
(pg. 
S9
-
S11
)
7
Schrauder
 
A
Reiter
 
A
Gadner
 
H
, et al. 
Superiority of allogeneic hematopoietic stem-cell transplantation compared with chemotherapy alone in high-risk childhood T-cell acute lymphoblastic leukemia: results from ALL-BFM 90 and 95.
J Clin Oncol
2006
, vol. 
24
 
36
(pg. 
5742
-
5749
)
8
Eapen
 
M
Raetz
 
E
Zhang
 
MJ
, et al. 
Outcomes after HLA-matched sibling transplantation or chemotherapy in children with B-precursor acute lymphoblastic leukemia in a second remission: a collaborative study of the Children's Oncology Group and the Center for International Blood and Marrow Transplant Research.
Blood
2006
, vol. 
107
 
12
(pg. 
4961
-
4967
)
9
Eapen
 
M
Rubinstein
 
P
Zhang
 
MJ
, et al. 
Comparable long-term survival after unrelated and HLA-matched sibling donor hematopoietic stem cell transplantations for acute leukemia in children younger than 18 months.
J Clin Oncol
2006
, vol. 
24
 
1
(pg. 
145
-
151
)
10
Rocha
 
V
Garnier
 
F
Ionescu
 
I
Gluckman
 
E
Hematopoietic stem-cell transplantation using umbilical-cord blood cells.
Rev Invest Clin
2005
, vol. 
57
 
2
(pg. 
314
-
323
)
11
Rocha
 
V
Gluckman
 
E
Clinical use of umbilical cord blood hematopoietic stem cells.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
1 suppl 1
(pg. 
34
-
41
)
12
Rocha
 
V
Cornish
 
J
Sievers
 
EL
, et al. 
Comparison of outcomes of unrelated bone marrow and umbilical cord blood transplants in children with acute leukemia.
Blood
2001
, vol. 
97
 
10
(pg. 
2962
-
2971
)
13
Klingebiel
 
T
Lang
 
P
Schumm
 
M
, et al. 
Experiences with haploidentical stem cell transplantation in children with acute lymphoblastic leukemia.
Pathol Biol (Paris)
2005
, vol. 
53
 
3
(pg. 
159
-
161
)
14
Handgretinger
 
R
Klingebiel
 
T
Lang
 
P
, et al. 
Megadose transplantation of purified peripheral blood CD34(+) progenitor cells from HLA-mismatched parental donors in children.
Bone Marrow Transplant
2001
, vol. 
27
 
8
(pg. 
777
-
783
)
15
Aversa
 
F
Tabilio
 
A
Velardi
 
A
, et al. 
Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype.
N Engl J Med
1998
, vol. 
339
 
17
(pg. 
1186
-
1193
)
16
Aversa
 
F
Terenzi
 
A
Carotti
 
A
, et al. 
Improved outcome with T-cell-depleted bone marrow transplantation for acute leukemia.
J Clin Oncol
1999
, vol. 
17
 
5
(pg. 
1545
-
1550
)
17
Klingebiel
 
T
Handgretinger
 
R
Lang
 
P
Bader
 
P
Niethammer
 
D
Haploidentical transplantation for acute lymphoblastic leukemia in childhood.
Blood Rev
2004
, vol. 
18
 
3
(pg. 
181
-
192
)
18
Ruggeri
 
L
Mancusi
 
A
Capanni
 
M
, et al. 
Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: challenging its predictive value.
Blood
2007
, vol. 
110
 
1
(pg. 
433
-
440
)
19
van Rood
 
JJ
Loberiza
 
FR
Zhang
 
MJ
, et al. 
Effect of tolerance to noninherited maternal antigens on the occurrence of graft-versus-host disease after bone marrow transplantation from a parent or an HLA-haploidentical sibling.
Blood
2002
, vol. 
99
 
5
(pg. 
1572
-
1577
)
20
Glucksberg
 
H
Storb
 
R
Fefer
 
A
, et al. 
Clinical manifestations of graft-versus-host disease in human recipients of marrow from HL-A-matched sibling donors.
Transplantation
1974
, vol. 
18
 
4
(pg. 
295
-
304
)
21
Shulman
 
HM
Sullivan
 
KM
Weiden
 
PL
, et al. 
Chronic graft-versus-host syndrome in man: a long-term clinicopathologic study of 20 Seattle patients.
Am J Med
1980
, vol. 
69
 
2
(pg. 
204
-
217
)
22
Filipovich
 
AH
Weisdorf
 
D
Pavletic
 
S
, et al. 
National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I, diagnosis and staging working group report.
Biol Blood Marrow Transplant
2005
, vol. 
11
 
12
(pg. 
945
-
956
)
23
Gooley
 
TA
Leisenring
 
W
Crowley
 
J
Storer
 
BE
Estimation of failure probabilities in the presence of competing risks: new representations of old estimators.
Stat Med
1999
, vol. 
18
 
6
(pg. 
695
-
706
)
24
Henslee-Downey
 
PJ
Abhyankar
 
SH
Parrish
 
RS
, et al. 
Use of partially mismatched related donors extends access to allogeneic marrow transplant.
Blood
1997
, vol. 
89
 
10
(pg. 
3864
-
3872
)
25
Aversa
 
F
Terenzi
 
A
Felicini
 
R
, et al. 
Mismatched T cell-depleted hematopoietic stem cell transplantation for children with high-risk acute leukemia.
Bone Marrow Transplant
1998
, vol. 
22
 
suppl 5
(pg. 
S29
-
S32
)
26
Peters
 
C
Matthes-Martin
 
S
Fritsch
 
G
, et al. 
Transplantation of highly purified peripheral blood CD34+ cells from HLA-mismatched parental donors in 14 children: evaluation of early monitoring of engraftment.
Leukemia
1999
, vol. 
13
 
12
(pg. 
2070
-
2078
)
27
Sedlácek
 
P
Stary
 
J
Vodvakova
 
S
, et al. 
Hematopoietic stem cell transplantation in children with hematological malignancies across HLA barriers: reasonable alternative?
Neoplasma
2001
, vol. 
48
 
4
(pg. 
302
-
306
)
28
Ortín
 
M
Raj
 
R
Kinning
 
E
Williams
 
M
Darbyshire
 
PJ
Partially matched related donor peripheral blood progenitor cell transplantation in paediatric patients adding fludarabine and anti-lymphocyte gamma-globulin.
Bone Marrow Transplant
2002
, vol. 
30
 
6
(pg. 
359
-
366
)
29
Lang
 
P
Greil
 
J
Bader
 
P
, et al. 
Long-term outcome after haploidentical stem cell transplantation in children.
Blood Cells Mol Dis
2004
, vol. 
33
 
3
(pg. 
281
-
287
)
30
Marks
 
DI
Khattry
 
N
Cummins
 
M
, et al. 
Haploidentical stem cell transplantation for children with acute leukaemia.
Br J Haematol
2006
, vol. 
134
 
2
(pg. 
196
-
201
)
31
Szydlo
 
R
Goldman
 
JM
Klein
 
JP
, et al. 
Results of allogeneic bone marrow transplants for leukemia using donors other than HLA-identical siblings.
J Clin Oncol
1997
, vol. 
15
 
5
(pg. 
1767
-
1777
)
32
Reisner
 
Y
Bachar-Lustig
 
E
Li
 
HW
Aversa
 
F
Velardi
 
A
Martelli
 
MF
The role of megadose CD34+ progenitor cells in the treatment of leukemia patients without a matched donor and in tolerance induction for organ transplantation.
Ann N Y Acad Sci
1999
, vol. 
872
 (pg. 
336
-
348
discussion 348–350
33
Reisner
 
Y
Stem cell transplantation across major genetic barriers.
Ann N Y Acad Sci
2001
, vol. 
938
 (pg. 
322
-
326
discussion 326–327
34
Handgretinger
 
R
Schumm
 
M
Lang
 
P
, et al. 
Transplantation of megadoses of purified haploidentical stem cells.
Ann N Y Acad Sci
1999
, vol. 
872
 (pg. 
351
-
361
discussion 361–362
35
Stern
 
M
Ruggeri
 
L
Mancusi
 
A
, et al. 
Survival after T cell-depleted haploidentical stem cell transplantation is improved using the mother as donor.
Blood
2008
, vol. 
112
 
7
(pg. 
2990
-
2995
)
36
Ruggeri
 
L
Capanni
 
M
Mancusi
 
A
, et al. 
Natural killer cell alloreactivity in haploidentical hematopoietic stem cell transplantation.
Int J Hematol
2005
, vol. 
81
 
1
(pg. 
13
-
17
)
37
Ruggeri
 
L
Capanni
 
M
Mancusi
 
A
, et al. 
Alloreactive natural killer cells in mismatched hematopoietic stem cell transplantation.
Blood Cells Mol Dis
2004
, vol. 
33
 
3
(pg. 
216
-
221
)
38
Ruggeri
 
L
Capanni
 
M
Tosti
 
A
, et al. 
Innate immunity against hematological malignancies.
Cytotherapy
2002
, vol. 
4
 
4
(pg. 
343
-
346
)
39
Ruggeri
 
L
Capanni
 
M
Urbani
 
E
, et al. 
Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants.
Science
2002
, vol. 
295
 
5562
(pg. 
2097
-
2100
)
40
Locatelli
 
F
Pende
 
D
Maccario
 
R
Mingari
 
MC
Moretta
 
A
Moretta
 
L
Haploidentical hemopoietic stem cell transplantation for the treatment of high-risk leukemias: how NK cells make the difference.
Clin Immunol
2009
, vol. 
133
 
2
(pg. 
171
-
178
)
41
Salvalaggio
 
PR
Modanlou
 
KA
Edwards
 
EB
Harper
 
AM
Abecassis
 
MM
Hepatic artery thrombosis after adult living donor liver transplantation: the effect of center volume.
Transplantation
2007
, vol. 
84
 
7
(pg. 
926
-
928
)
42
Gjertson
 
DW
Revisiting the center effect.
Clin Transpl
2005
(pg. 
333
-
341
)
43
Frassoni
 
F
Labopin
 
M
Powles
 
R
, et al. 
Effect of centre on outcome of bone-marrow transplantation for acute myeloid leukaemia: acute Leukaemia Working Party of the European Group for Blood and Marrow Transplantation.
Lancet
2000
, vol. 
355
 
9213
(pg. 
1393
-
1398
)
44
Locatelli
 
F
Zecca
 
M
Messina
 
C
, et al. 
Improvement over time in outcome for children with acute lymphoblastic leukemia in second remission given hematopoietic stem cell transplantation from unrelated donors.
Leukemia
2002
, vol. 
16
 
11
(pg. 
2228
-
2237
)
45
Bader
 
P
Hancock
 
J
Kreyenberg
 
H
, et al. 
Minimal residual disease (MRD) status prior to allogeneic stem cell transplantation is a powerful predictor for post-transplant outcome in children with ALL.
Leukemia
2002
, vol. 
16
 
9
(pg. 
1668
-
1672
)
46
Krejci
 
O
van der Velden
 
VH
Bader
 
P
, et al. 
Level of minimal residual disease prior to haematopoietic stem cell transplantation predicts prognosis in paediatric patients with acute lymphoblastic leukaemia: a report of the Pre-BMT MRD Study Group.
Bone Marrow Transplant
2003
, vol. 
32
 
8
(pg. 
849
-
851
)
47
Wassmann
 
B
Pfeifer
 
H
Stadler
 
M
, et al. 
Early molecular response to posttransplantation imatinib determines outcome in MRD+ Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL).
Blood
2005
, vol. 
106
 
2
(pg. 
458
-
463
)
48
Lang
 
P
Pfeiffer
 
M
Muller
 
I
, et al. 
Haploidentical stem cell transplantation in patients with pediatric solid tumors: preliminary results of a pilot study and analysis of graft versus tumor effects.
Klin Padiatr
2006
, vol. 
218
 
6
(pg. 
321
-
326
)
49
Bethge
 
WA
Haegele
 
M
Faul
 
C
, et al. 
Haploidentical allogeneic hematopoietic cell transplantation in adults with reduced-intensity conditioning and CD3/CD19 depletion: fast engraftment and low toxicity.
Exp Hematol
2006
, vol. 
34
 
12
(pg. 
1746
-
1752
)
50
Schleuning
 
M
Adoptive allogeneic immunotherapy: history and future perspectives.
Transfus Sci
2000
, vol. 
23
 
2
(pg. 
133
-
150
)
51
Ball
 
LM
Bernardo
 
ME
Roelofs
 
H
, et al. 
Cotransplantation of ex vivo expanded mesenchymal stem cells accelerates lymphocyte recovery and may reduce the risk of graft failure in haploidentical hematopoietic stem-cell transplantation.
Blood
2007
, vol. 
110
 
7
(pg. 
2764
-
2767
)
Sign in via your Institution