Allogeneic hematopoietic stem cell transplantation (HSCT) is the only proven curative therapy for juvenile myelomonocytic leukemia (JMML). We, the European Working Group on Childhood MDS (EWOG-MDS) and the European Blood and Marrow Transplantation (EBMT) Group, report the outcome of 100 children (67 boys and 33 girls) with JMML given unmanipulated HSCT after a preparative regimen including busulfan, cyclophosphamide, and melphalan. Forty-eight and 52 children received transplants from an HLA-identical relative or an unrelated donor (UD), respectively. The source of hematopoietic stem cells was bone marrow, peripheral blood, and cord blood in 79, 14, and 7 children, respectively. Splenectomy had been performed before HSCT in 24 children. The 5-year cumulative incidence of transplantation-related mortality and leukemia recurrence was 13% and 35%, respectively. Age older than 4 years predicted an increased risk of disease recurrence. The 5-year probability of event-free survival for children given HSCT from either a relative or a UD was 55% and 49%, respectively (P = NS), with median observation time of patients alive being 40 months (range, 6 to 144). In multivariate analysis, age older than 4 years and female sex predicted poorer outcome. Results of this study compare favorably with previously published reports. Disease recurrence remains the major cause of treatment failure. Outcome of UD-HSCT recipients is comparable to that of children receiving transplants from an HLA-identical sibling. (Blood. 2005;105:410-419)

Juvenile myelomonocytic leukemia (JMML) is a rare hematopoietic malignancy of early childhood, representing 2% to 3% of all pediatric leukemias.1-3  JMML usually runs an aggressive clinical course, with median duration of survival for children left untreated being less than 12 months from diagnosis.1-5  Some young children with JMML (mainly those diagnosed before 2 years of age) may experience a longer course, sometimes characterized by temporary clinical improvement in the absence of therapy.2,6  Death usually occurs as the result of tumor cell infiltration of organs, leading to organ dysfunction, infection, and bleeding.

Neither intensive nor moderate chemotherapy approaches have been demonstrated to consistently improve the outcome of children with JMML,4-9  and allogeneic hematopoietic stem cell transplantation (HSCT) is presently the only curative treatment for this disorder. Different studies have reported that a significant proportion of children with JMML are cured by an allograft.10-15  Leukemia recurrence represents the main cause of treatment failure in children with JMML given HSCT, with the relapse rate being as high as 50%.13,14  Relapse occurs early, at a median of 4 months from transplantation and generally within the first year after the allograft.10,11,15 

All studies published so far on the results of HSCT in children with JMML enrolled a limited number of patients treated with heterogeneous approaches. This fact significantly precluded the possibility of identifying patient-, disease-, and transplantation-related variables predicting the clinical course of a child with JMML treated with HSCT.

In this paper we analyze the outcome of 100 children with JMML given unmanipulated HSCT after an homogeneous preparative regimen consisting of 3 alkylating agents: busulfan, cyclophosphamide, and melphalan.

Approval for the study was obtained from the institutional review board of each single institution. Written informed consent was provided by the patients' parents, according to the Declaration of Helsinki. The patients included in this study were diagnosed as affected by JMML according to previously published criteria.16,17  A total of 100 children who underwent transplantation from January 1993 through December 2002 in 29 centers from 7 countries and reported to the European Working Group on Myelodysplastic Syndrome in Childhood (EWOG-MDS) were evaluated. Data concerning patient and disease characteristics and transplantation outcome were collected by standardized questionnaires for each child enrolled into this study. Submitted data were reviewed by 1 physician (C.M.N.), and computerized error checks were performed to ensure data quality.

Patient characteristics, preparative regimen, GVHD prophylaxis, and supportive therapy

The characteristics of the 100 children (67 boys and 33 girls) and of the transplantation procedure are detailed in Tables 1 and 2, respectively. Fourteen children (7 boys and 7 girls) had clinical evidence of neurofibromatosis type 1 (NF1) in association with JMML.

Table 1.

Patient characteristics


No. of patients enrolled  

100  
Median patient sex, M/F   67/33  
Median patient age at diagnosis, y (range)   1.4 (0.1-14)  
Median patient age at HSCT, y (range)   2.5 (0.3-15)  
Median interval between diagnosis and HSCT, mo (range)   6 (0.3-49)  
Median WBC count at diagnosis,* × 109/L (range)   34 (3-240)  
Median monocyte count at diagnosis,† × 109/L (range)   5.5 (1-50)  
Median platelet count at diagnosis,‡ × 109/L (range)   65 (9-530)  
Percentage of HbF at diagnosis§ (range)   9 (0-80)  
Karyotype  
Normal karyotype   66  
Monosomy 7   20  
Trisomy 8   9  
Other abnormalities   4  
Missing/unknown   1  
No. of patients with clinical evidence of NF1∥   14  
Median WBC count at HSCT,¶ × 109/L (range)   9.6 (0.7-320)  
Percentage of marrow blasts at HSCT** (range)   6 (0-85)  
Splenectomy before HSCT   24  
Median spleen size at HSCT,†† cm (range)   5 (0-16)  
Patient HCMV serology  
Negative   57  
Positive   39  
Unknown
 
4
 

No. of patients enrolled  

100  
Median patient sex, M/F   67/33  
Median patient age at diagnosis, y (range)   1.4 (0.1-14)  
Median patient age at HSCT, y (range)   2.5 (0.3-15)  
Median interval between diagnosis and HSCT, mo (range)   6 (0.3-49)  
Median WBC count at diagnosis,* × 109/L (range)   34 (3-240)  
Median monocyte count at diagnosis,† × 109/L (range)   5.5 (1-50)  
Median platelet count at diagnosis,‡ × 109/L (range)   65 (9-530)  
Percentage of HbF at diagnosis§ (range)   9 (0-80)  
Karyotype  
Normal karyotype   66  
Monosomy 7   20  
Trisomy 8   9  
Other abnormalities   4  
Missing/unknown   1  
No. of patients with clinical evidence of NF1∥   14  
Median WBC count at HSCT,¶ × 109/L (range)   9.6 (0.7-320)  
Percentage of marrow blasts at HSCT** (range)   6 (0-85)  
Splenectomy before HSCT   24  
Median spleen size at HSCT,†† cm (range)   5 (0-16)  
Patient HCMV serology  
Negative   57  
Positive   39  
Unknown
 
4
 

WBC indicates white blood cell.

*Three unknown; †6 unknown; ‡3 unknown; §23 unknown; ∥8 unknown; ¶8 unknown; **13 unknown; ††6 unknown.

Table 2.

Transplantation procedure


 

 
Donor  
HLA-identical family donor   48  
Matched unrelated donor   52  
Donor sex, M/F*  50/46  
Median donor age, y (range)   19 (1-54)  
Donor HCMV serology  
Negative   55  
Positive   38  
Unknown   7  
Stem cell source  
Bone marrow   79  
Peripheral blood   14  
Cord blood   7  
Median mononuclear cell dose infused  
Bone marrow, × 108/kg (range)   6.1 (0.6-18.9)  
Peripheral blood, CD34+ × 106/kg (range)   20 (10-30)  
Cord blood, × 107/kg (range)   5 (4-14)  
GVHD prophylaxis: matched family donor; matched unrelated donor  
None   1; 0  
Cs-A   32; 0  
MTX   2; 0  
Cs-A + MTX   7; 3  
Cs-A + ALG or MoAbs   3; 3  
Cs-A + MTX + ALG or MoAbs   1; 41  
Cs-A ± PDN ± MTX ± MoAbs
 
2; 5
 

 

 
Donor  
HLA-identical family donor   48  
Matched unrelated donor   52  
Donor sex, M/F*  50/46  
Median donor age, y (range)   19 (1-54)  
Donor HCMV serology  
Negative   55  
Positive   38  
Unknown   7  
Stem cell source  
Bone marrow   79  
Peripheral blood   14  
Cord blood   7  
Median mononuclear cell dose infused  
Bone marrow, × 108/kg (range)   6.1 (0.6-18.9)  
Peripheral blood, CD34+ × 106/kg (range)   20 (10-30)  
Cord blood, × 107/kg (range)   5 (4-14)  
GVHD prophylaxis: matched family donor; matched unrelated donor  
None   1; 0  
Cs-A   32; 0  
MTX   2; 0  
Cs-A + MTX   7; 3  
Cs-A + ALG or MoAbs   3; 3  
Cs-A + MTX + ALG or MoAbs   1; 41  
Cs-A ± PDN ± MTX ± MoAbs
 
2; 5
 

Cl-A indicates cyclosporin-A; MTX, short-term methotrexate; ALG, antilymphocyte globulin; MoAbs, monoclonal antibodies.

*Four unknown; †21 unknown.

The median age at presentation was 1.4 years, with a range between 0.1 and 14 years. Forty-eight children received transplants from an HLA-identical relative (sibling in 46 cases and phenotypically identical aunt or mother in 1 case each); the remaining 52 children were given the allograft from an unrelated donor (UD).

Successful cytogenetic analysis of malignant cells was available in all cases but one. For 81 patients, karyotype taken within 6 weeks prior to HSCT was available; for the remaining 18 children the karyotype obtained at time of diagnosis was used. Monosomy of chromosome 7 was the most frequent cytogenetic abnormality, being observed in 20 of the 33 patients with an abnormal karyotype.

Splenectomy before transplantation had been performed in 24 children.

To evaluate the impact of therapy before transplantation on posttransplantation outcome, patients were subdivided into 2 groups according to the different kind of therapy received before transplantation: Group 1 comprised 84 children given either no treatment (43 patients, 12 of whom were splenectomized), differentiative therapy (ie, cis-retinoic acid; 1 patient who subsequently received splenectomy), or low-intensity chemotherapy (ie, 6-thioguanine, 6-mercaptopurine, low-dose cytarabine, others; 40 patients, 9 of whom were splenectomized); group 2 consisted of 16 children (2 of whom were splenectomized) receiving schemes of chemotherapy similar to those adopted for children with acute myelogenous leukemia (AML).

The preparative regimen was based on the use of busulfan (16 to 20 mg/kg given orally over 4 consecutive days), cyclophosphamide (60 mg/kg/d for 2 consecutive days), and melphalan (140 mg/m2 in single dose).18  In 25 children who underwent transplantation in centers in Italy, a pharmacokinetic (PK) study of busulfan was performed. In detail, the PK study was performed on the first administration of busulfan and the dosage adjusted starting from the fifth administration to maintain plasmatic levels between 500 and 800 ng/mL.19 

Most children (35 of 48; 73%) given the allograft from an HLA-identical relative received cyclosporine-A (Cs-A) (1-3 mg/kg/d) alone to prevent occurrence of graft-versus-host disease (GVHD). The combination of Cs-A, short-term methotrexate (MTX) (15 mg/m2 on day +1 and 10 mg/m2 on days +3, +6, and +11), and either antithymocyte globulin (ATG) or, in a few cases, the monoclonal antibody Campath1-G was used in most patients receiving transplants from an unrelated volunteer.

Supportive therapy as well as prophylaxis and treatment of infections were similar among centers participating in this study. Human cytomegalovirus (HCMV) serologic status was studied before transplantation in donor-recipient pairs (see Tables 1 and 2). In all patients, HCMV infection was monitored and treated according to previously reported strategies.20 

HLA typing

HLA-A and -B antigen serologic typing and a low-resolution generic DRB1 oligotyping were available for all donor-recipient pairs. Unrelated donors were located through networks of national and international bone marrow and cord blood donor registries. In all unrelated donor-recipient pairs, as well as when the donor was a relative other than an HLA-identical sibling, histocompatibility was determined by serology for HLA-A and -B antigens and by high-resolution DNA typing for DRB1 antigen. All children receiving transplants from an unrelated volunteer were either identical or had one antigenic/allelic disparity with their donor.

Definitions

Acute GVHD was diagnosed and graded by investigators at each transplantation center according to previously reported criteria.21  All patients surviving more than 10 days after transplantation were considered at risk for developing acute GVHD. Children alive at day +100 after transplantation with sustained donor engraftment were considered to be evaluable for chronic GVHD, which was classified according to previously reported criteria.22  Tissue biopsy samples were obtained to confirm diagnosis of GVHD whenever clinically indicated and feasible. Treatment of both acute and chronic GVHD was administered according to the protocols in use at each single institution.

Myeloid engraftment was defined as the first of 3 consecutive days when the neutrophil count was higher than 0.5 × 109/L and platelet engraftment as the first of 7 consecutive days with an unsupported platelet count higher than 50 × 109/L. Patients who did not engraft, as well as those with transient engraftment of donor cells, were considered to have graft failure.

Statistical analysis

Analysis used July 1, 2003, as the reference date (ie, the day at which all centers locked data on patient outcomes).

Overall survival (OS) was defined as the probability of survival, irrespective of disease state, at any point in time; patients alive at their last follow-up were censored, while only death was considered as an event. Event-free survival (EFS) was defined as the probability of being alive, disease-free, and with complete donor chimerism at any time point; death, relapse, rejection, and graft failure were considered as events, while patients alive and disease free with donor engraftment at their last follow-up were censored. Both these probabilities were analyzed by the Kaplan-Meier method, and comparisons between probabilities in different patient groups were performed using the log-rank test.23 

Relapse incidence (RI) was defined as the probability of having a relapse before time t; death without experiencing a relapse was considered a competing event. On the contrary, transplantation-related mortality (TRM) was defined as the probability of dying without previous occurrence of a relapse, which was the competing event. Both these probabilities were estimated as cumulative incidence curves, as previously described.24-26 

Also, the probabilities of acute and chronic GVHD and those of neutrophil and platelet engraftment were estimated as cumulative incidence. For acute GVHD analysis, relapse, death, and either rejection or graft failure were treated as competing events, while patients alive and relapse free at day +100 without having experienced acute GVHD were censored. For chronic GVHD, only patients surviving in remission and with donor engraftment for at least 100 days were evaluated. Also in this case, relapse, death, and either rejection or graft failure were considered competing events, data being censored at time of last follow-up for patients who did not experience chronic GVHD.

Finally, for neutrophil and platelet engraftment, competing events were relapse, death, or rejection before engraftment.

All results were expressed as 5-year probability or 5-year cumulative incidence (%) and 95% confidence interval (95% CI).

A univariate analysis of EFS, RI, and TRM was performed for the whole study population considering the following variables: patient and donor age; sex mismatch; interval between diagnosis and HSCT; leukocyte, monocyte, and platelet count at diagnosis; hemoglobin F (HbF) percentage corrected for patient age at diagnosis; karyotype, leukocyte count, and bone marrow blast percentage at HSCT; NF1; spleen size or splenectomy prior to HSCT; pretransplantation treatment; type of donor; stem cell source; infused cell dose; recipient and donor HCMV serology; type of GVHD prophylaxis; busulfan PK study; development of grades II-IV acute GVHD; and development of chronic GVHD.

For this purpose, continuous variables were categorized as follows: Each variable was first divided into 4 categories at approximately the 25th, 50th, and 75th percentiles. If the relative event rates (ratio of the observed number of events to the expected number of events in a category, assuming no variation across categories) in 2 or more adjacent categories (and the mean times to event) were not substantially different, these categories were grouped. If no clear pattern was observed for the primary outcome, the median was taken as cut point.27 

For multivariate analyses, the Cox proportional hazard regression model was used, including in the models all the variables with P less than .1 in univariate analysis.28,29 

The χ2 test was used to compare differences in percentages.

All P values were 2-sided, and values less than .05 were considered statistically significant. P values above .1 were reported as nonsignificant (NS), whereas those between .05 and .1 were reported in detail.

The SAS package (SAS Institute, Cary, NC) and NCSS 2001 (Number Cruncher Statistical Systems, Kaysville, UT) were used for the analysis of the data.

Engraftment and GVHD occurrence

Information on kinetics of myeloid recovery was available for all children included in this study. Three patients failed to engraft, 2 of whom had received a UD HSCT. Two more patients, both receiving transplants from a UD, presented a secondary marrow failure 27 and 39 days after HSCT. No other factor was associated with the occurrence of either primary or secondary graft failure. In children with sustained engraftment of donor cells, the median time to achieve neutrophil recovery was 18 days (range, 8 to 44). In the Cox analysis on the whole population, the use of cord blood as stem cell source and the absence of grades II-IV acute GVHD were factors associated with a delayed neutrophil engraftment (P = .0015, relative risk [RR] = 0.16, 95% CI: 0.051-0.49; and P = .017, RR = 0.58, 95% CI: 0.38-0.91; respectively).

The median time to obtain a self-sustained platelet count higher than 50 × 109/L was 30 days (range, 11 to 148). From the Cox model, we found that the most adverse factors for platelet recovery in the overall population were a platelet count at diagnosis less than 100 × 109/L and the use of cord blood as stem cell source (P = .02, RR = 0.54, 95% CI: 0.32-0.91; and P = .0065, RR = 0.21, 95% CI: 0.07-0.65; respectively).

Grades II-IV acute GVHD developed in 40 patients. The cumulative incidence at day 100 of grades II-IV acute GVHD was 40% (31% to 51%) whereas that of grades III-IV acute GVHD was 17% (11% to 26%) (Figure 1). Children given HSCT from an HLA-compatible family donor had a cumulative incidence of grades II-IV acute GVHD comparable to that of patients receiving transplants from an unrelated volunteer (46% versus 35%, respectively; P = NS). No patient- or transplantation-related variable was significantly associated with the development of grades II-IV acute GVHD in a multivariate model.

Figure 1.

Cumulative incidence of grades II-IV and grades III-IV acute GVHD in the overall cohort of patients studied.

Figure 1.

Cumulative incidence of grades II-IV and grades III-IV acute GVHD in the overall cohort of patients studied.

Close modal

Thirteen (15%) of the 86 patients at risk developed chronic GVHD, which was limited in 6 cases and extensive in the remaining 7 patients. The overall cumulative incidence of chronic GVHD was 17% (10% to 28%). Children given HSCT from an HLA-compatible family donor had a cumulative incidence of chronic GVHD similar to that of patients receiving transplants from an unrelated volunteer (17% versus 16%, respectively; P = NS). From the multivariate analysis performed using the Cox model, we found that previous grades II-IV acute GVHD was the only statistically significant risk factor for the occurrence of chronic GVHD (P = .016, RR = 4.96, 95% CI: 1.35-18.2).

Transplantation-related mortality

Thirteen patients died of transplantation-related causes, with the 5-year cumulative incidence of TRM being 13% (8% to 22%) (Figure 2). The 5-year cumulative incidence of TRM for patients receiving transplants from either an HLA-identical sibling or an unrelated volunteer was 10% (5% to 24%) and 16% (8% to 30%), respectively (P = NS; Figure 3). The median time to treatment-related death was 2.7 months (range, 1 to 16). Table 3 lists probabilities of TRM, RI, and EFS not adjusted for differences in factors that influence transplantation outcome. In univariate analysis, we found that patients receiving transplants from a female donor had a statistically higher probability of dying of transplantation-related causes; all other variables did not have any impact on the probability of death due to transplantation complications, possibly because of the limited number of events. None of the variables considered influenced TRM in multivariate analysis (Table 4).

Figure 2.

Kaplan-Meier estimate of overall survival (surv) and event-free survival (EFS) and cumulative incidence of relapse (RI) and transplantation-related mortality (TRM) in the overall population.

Figure 2.

Kaplan-Meier estimate of overall survival (surv) and event-free survival (EFS) and cumulative incidence of relapse (RI) and transplantation-related mortality (TRM) in the overall population.

Close modal
Figure 3.

Kaplan-Meier estimate of event-free survival (EFS) and transplantation-related mortality (TRM) according to the type of donor used.

Figure 3.

Kaplan-Meier estimate of event-free survival (EFS) and transplantation-related mortality (TRM) according to the type of donor used.

Close modal
Table 3.

Univariate analysis of 5-year event-free survival probability (EFS), cumulative incidence of relapse (RI), and cumulative incidence of transplantation-related mortality (TRM)




EFS

RI

TRM

No. of patients
Probability
95% CI
Cumulative incidence
95% CI
Cumulative incidence
95% CI
Overall probability or incidence   100   52%   42-62   35%   27-46   13   8-22  
Patient sex        
Male   67   61%   49-73   30%   20-44   9%   4-19  
Female   33   33%   17-49   45%   31-66   21%   11-41  
p  —   .0021   —   .012   —   .065   —  
Donor sex        
Male   50   53%   38-67   41%   30-59   6%   2-18  
Female   46   53%   39-68   25%   15-42   22%   13-38  
Missing   4   —   —   —   —   —   —  
p  —   NS   —   NS   —   .035   —  
Age at diagnosis        
Less than 1 y   38   65%   46-78   17%   8-35   21%   12-40  
1-2 y   18   67%   45-88   22%   9-53   11%   3-41  
2-3 y   9   47%   10-84   53%   27-100   0%   —  
3-4 y   18   50%   27-73   44%   27-75   6%   1-37  
4 y or more   17   16%   0-34   73%   53-98   12%   3-43  
p  —   .013   —   .0003   —   NS   —  
Less than 2 y   56   64%   51-76   18%   10-32   18%   10-32  
2-4 y   27   50%   31-70   46%   30-70   4%   1-25  
4 y   17   16%   0-34   73%   53-98   12%   3-43  
p  —   .0020   —   .0001   —   NS   —  
Interval between diagnosis and HSCT        
Less than 3 mo   24   56%   35-77   31%   17-59   13%   4-36  
3-6 mo   29   54%   35-73   39%   24-62   7%   2-28  
6-9 mo   25   47%   27-67   37%   22-62   16%   7-40  
9 mo or more   22   50%   29-71   32%   17-59   18%   7-44  
p  —   NS   —   NS   —   NS   —  
Age at HSCT        
Less than 1 y   17   64%   41-87   12%   3-45   24%   10-55  
1-2 y   27   66%   48-84   19%   9-42   15%   6-37  
2-3 y   13   62%   35-88   23%   9-62   15%   4-55  
3-4 y   18   55%   32-78   45%   27-75   0%   —  
4 y or more   25   23%   6-40   65%   48-87   12%   4-35  
p  —   .014   —   .0008   —   NS   —  
Less than 2 y   44   65%   51-79   16%   8-32   18%   10-34  
2-4 y   31   57%   39-75   37%   23-59   6%   2-25  
4 y or more   25   23%   6-40   65%   48-87   12%   4-35  
p  —   .0022   —   .0008   —   NS   —  
Donor age        
Less than 10 y   31   60%   42-78   34%   20-56   6%   2-25  
10-20 y   9   33%   3-64   33%   13-84   33%   13-84  
20 y or more   39   47%   31-63   37%   25-57   16%   8-33  
Missing   21   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Leukocyte count at diagnosis, × 109/L        
Less than 20   25   51%   31-71   45%   29-70   4%   1-27  
20-39   35   49%   32-67   30%   18-51   20%   10-39  
40-59   17   40%   17-64   36%   19-69   24%   10-55  
60-79   11   64%   35-92   36%   17-79   0%   —  
80 or more   9   63%   30-97   37%   15-91   0%   —  
Missing   3   —   —   —   —   —   —  
p  —   NS   —   NS   —   .09   —  
Monocyte count at diagnosis, × 109/L        
1-2   13   54%   27-80   31%   14-70   15%   4-55  
2-3   15   51%   25-78   42%   23-78   7%   1-44  
3-5   16   56%   32-81   31%   15-65   13%   3-46  
5-10   25   38%   19-58   37%   22-62   25%   12-50  
10 or more   25   62%   43-82   34%   19-59   4%   1-27  
Missing   6   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Platelet count at diagnosis, × 109/L        
Less than 50   38   44%   28-60   42%   29-62   13%   6-30  
50-100   27   48%   28-69   37%   22-63   15%   6-37  
100 or more   32   62%   45-79   29%   17-50   9%   3-28  
Missing   3   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Karyotype        
Normal   66   49%   36-61   37%   27-52   14%   8-25  
Monosomy 7   20   68%   46-89   22%   9-54   10%   3-37  
Other   13   46%   19-73   46%   26-83   8%   1-51  
Missing   1   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
HbF %        
Less than 5%   26   61%   42-80   20%   9-43   20%   9-43  
5%-9%   14   43%   17-69   57%   36-90   0%   —  
10%-39%   23   51%   29-72   32%   17-59   17%   7-42  
40% or more   14   19%   0-41   81%   62-100   0%   —  
Missing   23   —   —   —   —   —   —  
p  —   NS   —   .0083   —   NS   —  
Less than 40%   63   53%   41-66   33%   23-47   14   8-26  
40% or more   14   19%   0-41   81%   62-100   0   —  
Missing   23   —   —   —   —   —   —  
p  —   .07   —   .004   —   NS   —  
Clinical evidence of NF1        
No   78   55%   43-66   34%   24-47   12%   6-22  
Yes   14   36%   11-61   50%   30-84   14%   4-52  
Missing   8   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Leukocyte count at HSCT, × 109/L        
Less than 10   46   53%   38-68   32%   20-49   15%   8-30  
10-19   20   47%   25-70   37%   20-67   16%   6-45  
20-39   17   45%   20-69   43%   25-77   12%   3-43  
40 or more   9   44%   12-77   56%   31-100   0%   —  
Missing   8   —   —   —   —   —   —  
p  —   NS   —   NS   —   —   —  
Bone marrow blast percentage at HSCT        
Less than 5%   30   63%   46-81   20%   10-41   17%   7-37  
5%-19%   47   52%   37-67   35%   24-52   13%   6-27  
20% or more   10   0%   —   90%   73-100   10%   2-64  
Missing   13   —   —   —   —   —   —  
p  —   .10   —   .017   —   NS   —  
Spleen size at HSCT        
Less than 5 cm   34   61%   44-78   24%   13-44   15%   7-33  
5 cm or more   36   44%   26-62   45%   30-67   11%   4-28  
Splenectomized   24   48%   28-69   39%   23-65   13%   4-36  
Missing   6   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Pre-HSCT treatment        
None or low dose   84   52%   41-63   35%   26-47   13%   8-23  
AML-like   16   50%   26-75   38%   20-71   13%   3-46  
p  —   NS   —   NS   —   NS   —  
Donor        
Matched family donor   48   55%   41-70   35%   23-52   10%   5-24  
Unrelated donor   52   49%   35-63   36%   24-52   16%   8-30  
p  —   NS   —   NS   —   NS   —  
Stem cell source        
Bone marrow   79   51%   40-62   35%   26-47   14%   8-24  
Peripheral blood   14   55%   28-82   36%   18-72   9%   1-59  
Cord blood   7   54%   14-93   32%   10-100   14%   2-88  
p  —   NS   —   NS   —   NS   —  
Stem cell dose, BM only        
Less than 5 × 108/kg   22   58%   37-79   33%   18-61   9%   2-34  
5 × 108/kg or more   46   54%   39-68   31%   20-48   15%   8-30  
Missing   11   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
HCMV serology        
Negative/Negative   41   49%   33-65   39%   26-58   13%   6-29  
Other   53   54%   40-67   33%   22-49   13%   7-26  
Missing   6   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Busulfan PK study        
Yes   25   60%   40-79   32%   18-57   8%   2-30  
No   75   49%   37-61   36%   27-59   15%   9-269  
p  —   NS   —   NS   —   NS   —  
GVHD prophylaxis        
All transplantations         
Monotherapy   35   59%   42-75   33%   20-54   9%   3-25  
Combination treatment   14   36%   11-61   36%   18-72   29%   12-65  
Serotherapy   51   52%   37-66   36%   25-53   12%   6-25  
p  —   NS   —   NS   —   NS   —  
Sibling donor transplantations         
Monotherapy   35   59%   42-75   33%   20-54   9%   3-25  
Combination treatment   8   38%   4-71   38%   11-92   25%   8-83  
Serotherapy   5   60%   17-100   40%   14-100   0%   —  
p  —   NS   —   NS   —   —   —  
Unrelated donor transplantations         
Monotherapy   0   —   —   —   —   —   —  
Combination treatment   6   33%   0-71   33%   11-100   33%   11-100  
Serotherapy   46   51%   36-66   36%   24-53   13%   6-28  
p  —   NS   —   NS   —   0.08   —  
Acute GVHD occurrence        
Grades 0-I   60   48%   35-61   37%   26-52   15%   8-28  
Grades II-IV   40   57%   42-73   33%   21-51   10%   4-25  
p  —   NS   —   NS   —   .083   —  
Chronic GVHD        
Absent   73   59%   48-71   35%   26-48   6%   2-15  
Present   13   67%   41-94   25%   9-67   8%   1-51  
Not evaluable   14   —   —   —   —   —   —  
p
 

 
NS
 

 
NS
 

 
NS
 

 



EFS

RI

TRM

No. of patients
Probability
95% CI
Cumulative incidence
95% CI
Cumulative incidence
95% CI
Overall probability or incidence   100   52%   42-62   35%   27-46   13   8-22  
Patient sex        
Male   67   61%   49-73   30%   20-44   9%   4-19  
Female   33   33%   17-49   45%   31-66   21%   11-41  
p  —   .0021   —   .012   —   .065   —  
Donor sex        
Male   50   53%   38-67   41%   30-59   6%   2-18  
Female   46   53%   39-68   25%   15-42   22%   13-38  
Missing   4   —   —   —   —   —   —  
p  —   NS   —   NS   —   .035   —  
Age at diagnosis        
Less than 1 y   38   65%   46-78   17%   8-35   21%   12-40  
1-2 y   18   67%   45-88   22%   9-53   11%   3-41  
2-3 y   9   47%   10-84   53%   27-100   0%   —  
3-4 y   18   50%   27-73   44%   27-75   6%   1-37  
4 y or more   17   16%   0-34   73%   53-98   12%   3-43  
p  —   .013   —   .0003   —   NS   —  
Less than 2 y   56   64%   51-76   18%   10-32   18%   10-32  
2-4 y   27   50%   31-70   46%   30-70   4%   1-25  
4 y   17   16%   0-34   73%   53-98   12%   3-43  
p  —   .0020   —   .0001   —   NS   —  
Interval between diagnosis and HSCT        
Less than 3 mo   24   56%   35-77   31%   17-59   13%   4-36  
3-6 mo   29   54%   35-73   39%   24-62   7%   2-28  
6-9 mo   25   47%   27-67   37%   22-62   16%   7-40  
9 mo or more   22   50%   29-71   32%   17-59   18%   7-44  
p  —   NS   —   NS   —   NS   —  
Age at HSCT        
Less than 1 y   17   64%   41-87   12%   3-45   24%   10-55  
1-2 y   27   66%   48-84   19%   9-42   15%   6-37  
2-3 y   13   62%   35-88   23%   9-62   15%   4-55  
3-4 y   18   55%   32-78   45%   27-75   0%   —  
4 y or more   25   23%   6-40   65%   48-87   12%   4-35  
p  —   .014   —   .0008   —   NS   —  
Less than 2 y   44   65%   51-79   16%   8-32   18%   10-34  
2-4 y   31   57%   39-75   37%   23-59   6%   2-25  
4 y or more   25   23%   6-40   65%   48-87   12%   4-35  
p  —   .0022   —   .0008   —   NS   —  
Donor age        
Less than 10 y   31   60%   42-78   34%   20-56   6%   2-25  
10-20 y   9   33%   3-64   33%   13-84   33%   13-84  
20 y or more   39   47%   31-63   37%   25-57   16%   8-33  
Missing   21   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Leukocyte count at diagnosis, × 109/L        
Less than 20   25   51%   31-71   45%   29-70   4%   1-27  
20-39   35   49%   32-67   30%   18-51   20%   10-39  
40-59   17   40%   17-64   36%   19-69   24%   10-55  
60-79   11   64%   35-92   36%   17-79   0%   —  
80 or more   9   63%   30-97   37%   15-91   0%   —  
Missing   3   —   —   —   —   —   —  
p  —   NS   —   NS   —   .09   —  
Monocyte count at diagnosis, × 109/L        
1-2   13   54%   27-80   31%   14-70   15%   4-55  
2-3   15   51%   25-78   42%   23-78   7%   1-44  
3-5   16   56%   32-81   31%   15-65   13%   3-46  
5-10   25   38%   19-58   37%   22-62   25%   12-50  
10 or more   25   62%   43-82   34%   19-59   4%   1-27  
Missing   6   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Platelet count at diagnosis, × 109/L        
Less than 50   38   44%   28-60   42%   29-62   13%   6-30  
50-100   27   48%   28-69   37%   22-63   15%   6-37  
100 or more   32   62%   45-79   29%   17-50   9%   3-28  
Missing   3   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Karyotype        
Normal   66   49%   36-61   37%   27-52   14%   8-25  
Monosomy 7   20   68%   46-89   22%   9-54   10%   3-37  
Other   13   46%   19-73   46%   26-83   8%   1-51  
Missing   1   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
HbF %        
Less than 5%   26   61%   42-80   20%   9-43   20%   9-43  
5%-9%   14   43%   17-69   57%   36-90   0%   —  
10%-39%   23   51%   29-72   32%   17-59   17%   7-42  
40% or more   14   19%   0-41   81%   62-100   0%   —  
Missing   23   —   —   —   —   —   —  
p  —   NS   —   .0083   —   NS   —  
Less than 40%   63   53%   41-66   33%   23-47   14   8-26  
40% or more   14   19%   0-41   81%   62-100   0   —  
Missing   23   —   —   —   —   —   —  
p  —   .07   —   .004   —   NS   —  
Clinical evidence of NF1        
No   78   55%   43-66   34%   24-47   12%   6-22  
Yes   14   36%   11-61   50%   30-84   14%   4-52  
Missing   8   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Leukocyte count at HSCT, × 109/L        
Less than 10   46   53%   38-68   32%   20-49   15%   8-30  
10-19   20   47%   25-70   37%   20-67   16%   6-45  
20-39   17   45%   20-69   43%   25-77   12%   3-43  
40 or more   9   44%   12-77   56%   31-100   0%   —  
Missing   8   —   —   —   —   —   —  
p  —   NS   —   NS   —   —   —  
Bone marrow blast percentage at HSCT        
Less than 5%   30   63%   46-81   20%   10-41   17%   7-37  
5%-19%   47   52%   37-67   35%   24-52   13%   6-27  
20% or more   10   0%   —   90%   73-100   10%   2-64  
Missing   13   —   —   —   —   —   —  
p  —   .10   —   .017   —   NS   —  
Spleen size at HSCT        
Less than 5 cm   34   61%   44-78   24%   13-44   15%   7-33  
5 cm or more   36   44%   26-62   45%   30-67   11%   4-28  
Splenectomized   24   48%   28-69   39%   23-65   13%   4-36  
Missing   6   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Pre-HSCT treatment        
None or low dose   84   52%   41-63   35%   26-47   13%   8-23  
AML-like   16   50%   26-75   38%   20-71   13%   3-46  
p  —   NS   —   NS   —   NS   —  
Donor        
Matched family donor   48   55%   41-70   35%   23-52   10%   5-24  
Unrelated donor   52   49%   35-63   36%   24-52   16%   8-30  
p  —   NS   —   NS   —   NS   —  
Stem cell source        
Bone marrow   79   51%   40-62   35%   26-47   14%   8-24  
Peripheral blood   14   55%   28-82   36%   18-72   9%   1-59  
Cord blood   7   54%   14-93   32%   10-100   14%   2-88  
p  —   NS   —   NS   —   NS   —  
Stem cell dose, BM only        
Less than 5 × 108/kg   22   58%   37-79   33%   18-61   9%   2-34  
5 × 108/kg or more   46   54%   39-68   31%   20-48   15%   8-30  
Missing   11   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
HCMV serology        
Negative/Negative   41   49%   33-65   39%   26-58   13%   6-29  
Other   53   54%   40-67   33%   22-49   13%   7-26  
Missing   6   —   —   —   —   —   —  
p  —   NS   —   NS   —   NS   —  
Busulfan PK study        
Yes   25   60%   40-79   32%   18-57   8%   2-30  
No   75   49%   37-61   36%   27-59   15%   9-269  
p  —   NS   —   NS   —   NS   —  
GVHD prophylaxis        
All transplantations         
Monotherapy   35   59%   42-75   33%   20-54   9%   3-25  
Combination treatment   14   36%   11-61   36%   18-72   29%   12-65  
Serotherapy   51   52%   37-66   36%   25-53   12%   6-25  
p  —   NS   —   NS   —   NS   —  
Sibling donor transplantations         
Monotherapy   35   59%   42-75   33%   20-54   9%   3-25  
Combination treatment   8   38%   4-71   38%   11-92   25%   8-83  
Serotherapy   5   60%   17-100   40%   14-100   0%   —  
p  —   NS   —   NS   —   —   —  
Unrelated donor transplantations         
Monotherapy   0   —   —   —   —   —   —  
Combination treatment   6   33%   0-71   33%   11-100   33%   11-100  
Serotherapy   46   51%   36-66   36%   24-53   13%   6-28  
p  —   NS   —   NS   —   0.08   —  
Acute GVHD occurrence        
Grades 0-I   60   48%   35-61   37%   26-52   15%   8-28  
Grades II-IV   40   57%   42-73   33%   21-51   10%   4-25  
p  —   NS   —   NS   —   .083   —  
Chronic GVHD        
Absent   73   59%   48-71   35%   26-48   6%   2-15  
Present   13   67%   41-94   25%   9-67   8%   1-51  
Not evaluable   14   —   —   —   —   —   —  
p
 

 
NS
 

 
NS
 

 
NS
 

 

GVHD prophylaxis was defined as follows: monotherapy, a single drug used (cyclosporin-A or methotrexate; the single patient who did not receive any GVHD prophylaxis was included in this group); combination treatment: more than one drug used (cyclosporin-A + methotrexate or steroids) without the addition of antilymphocyte globulin or monoclonal antibodies; serotherapy: any drug combination plus the addition of antilymphocyte globulin or monoclonal antibodies (Campath-1G).

—indicates that no value is available.

Table 4.

Multivariate analysis of variables influencing the probability of event-free survival (EFS), relapse incidence (RI), and transplantation-related mortality (TRM)




Relative risk

95% CI

P
EFS    
Patient age at HSCT, 4 y or more vs less than 4 y   2.24   1.07-4.69   .032  
Patient sex, female vs male   2.22   1.09-4.50   .028  
HbF %, 40% or more vs less than 40%   1.20   0.52-2.72   NS  
Bone marrow blast % at HSCT     
5%-19% vs less than 5%   1.70   0.76-3.79   NS  
20% or more vs less than 5%   1.82   0.64-5.15   NS  
RI    
Patient age at HSCT, 4 y or more vs less than 4 y   2.96   1.26-6.92   .012  
Patient sex, female vs male   1.80   0.77-4.20   NS  
HbF %, 40% or more vs less than 40%   1.90   0.79-4.54   NS  
Bone marrow blast % at HSCT     
5%-19% vs less than 5%   2.08   0.78-5.55   NS  
20% or more vs less than 5%   2.06   0.60-7.06   NS  
TRM    
Patient sex, female vs male   2.18   0.66-7.22   NS  
Donor sex, female vs male   2.87   0.76-10.87   NS  
WBC count at diagnosis, × 109/L; 30 or more vs less than 30   3.16   0.68-14.69   NS  
GVHD prophylaxis     
Cs-A + MTX vs monotherapy   3.95   0.77-20.35   NS  
Cs-A + MTX + ALG vs monotherapy
 
1.43
 
0.35-5.92
 
NS
 



Relative risk

95% CI

P
EFS    
Patient age at HSCT, 4 y or more vs less than 4 y   2.24   1.07-4.69   .032  
Patient sex, female vs male   2.22   1.09-4.50   .028  
HbF %, 40% or more vs less than 40%   1.20   0.52-2.72   NS  
Bone marrow blast % at HSCT     
5%-19% vs less than 5%   1.70   0.76-3.79   NS  
20% or more vs less than 5%   1.82   0.64-5.15   NS  
RI    
Patient age at HSCT, 4 y or more vs less than 4 y   2.96   1.26-6.92   .012  
Patient sex, female vs male   1.80   0.77-4.20   NS  
HbF %, 40% or more vs less than 40%   1.90   0.79-4.54   NS  
Bone marrow blast % at HSCT     
5%-19% vs less than 5%   2.08   0.78-5.55   NS  
20% or more vs less than 5%   2.06   0.60-7.06   NS  
TRM    
Patient sex, female vs male   2.18   0.66-7.22   NS  
Donor sex, female vs male   2.87   0.76-10.87   NS  
WBC count at diagnosis, × 109/L; 30 or more vs less than 30   3.16   0.68-14.69   NS  
GVHD prophylaxis     
Cs-A + MTX vs monotherapy   3.95   0.77-20.35   NS  
Cs-A + MTX + ALG vs monotherapy
 
1.43
 
0.35-5.92
 
NS
 

All variables with a P ≤ .1 in univariate analysis were considered as covariates and included in the Cox proportional hazard regression model.

Hepatic venoocclusive disease occurred in 11 patients (5 of whom received transplants from an unrelated volunteer) but, fortunately, it was not fatal in any of them. Three of these 11 patients relapsed, with the remaining 8 still being alive and in complete remission.

Relapse incidence

Thirty-four patients had hematologic relapse after transplantation, at a median time of 6 months (range, 2 to 36) after the allograft. Twenty-one children died due to disease progression at a median of 11 months after transplantation (range, 2 to 65). The 5-year cumulative incidence of relapse was 35% (Figure 2), with no significant difference between patients receiving transplants from either a relative or an unrelated donor (Table 3).

In univariable analysis (Table 3), the following features were associated with increased RI: female sex, age at diagnosis older than 4 years, high percentage of HbF, and blast percentage in the bone marrow at time of transplantation above 20%. From the Cox model, we found that only age at diagnosis older than 4 years remained a predictive variable for an increased risk of relapse (Table 4 and Figure 4A).

Figure 4.

Cumulative incidence of relapse (RI) and Kaplan-Meier estimate of event-free survival (EFS) according to age at diagnosis.

Figure 4.

Cumulative incidence of relapse (RI) and Kaplan-Meier estimate of event-free survival (EFS) according to age at diagnosis.

Close modal

Survival and leukemia-free survival

Overall, 66 children remain alive after HSCT, the 5-year Kaplan-Meier estimate of survival being 64% (54% to 74%) (Figure 2).

Fifty-three patients are alive in first complete remission after HSCT, with a median observation time of 40 months (range, 6 to 144). The 5-year cumulative probability of EFS after the first allograft is 52% (42% to 62%) (Figure 2) for the whole cohort of patients studied, being 55% (41% to 70%) and 49% (35% to 63%) for patients given HSCT from either a relative or an unrelated donor, respectively (P = NS; Figure 3). Six patients are alive with disease, and 7 patients are alive in hematologic remission after a second allograft, which was performed in a total of 15 cases. Five of these 15 patients given a second transplantation died because of further disease recurrence, and 3 died due to transplantation-related complications. In 10 of the 15 children who received a second transplantation, the same donor used in the first HSCT was used and total body irradiation was added as part of the preparative regimen in 8 of these 15 patients. Moreover, less intensive GVHD prophylaxis was adopted to exploit a graft-versus-leukemia (GVL) effect; this choice resulted in the occurrence of grades II-IV acute GVHD in 8 of the 15 patients given a second allograft. The median follow-up of the 7 patients who are alive and disease free after the second HSCT is 2.3 years (range, 0.4 to 5.4).

Univariate analysis of factors related to the patient, disease, and transplantation influencing EFS showed that male sex and both age at diagnosis and age at transplantation younger than 4 years were associated with a better outcome. Both age at diagnosis and patient sex remained significant in multivariate analysis (Table 4 and Figures 4B and 5). All other variables did not have any impact on the probability of EFS. In particular, no significant differences in terms of EFS, RI, and TRM were observed between children given AML-type chemotherapy or less intensive treatment (Table 3). Also, neither splenectomy prior to HSCT nor spleen size at time of transplantation influenced the outcome (Figure 6). Of particular interest are the data on cytogenetics: Patients with monosomy 7 had an outcome comparable to that of children with either a normal karyotype or other cytogenetics abnormalities (Table 3 and Figure 7).

Figure 5.

Kaplan-Meier estimate of event-free survival (EFS) according to the patient's sex.

Figure 5.

Kaplan-Meier estimate of event-free survival (EFS) according to the patient's sex.

Close modal
Figure 6.

Kaplan-Meier estimate of event-free survival (EFS) in patients splenectomized before transplantation and according to spleen size at time of allograft.

Figure 6.

Kaplan-Meier estimate of event-free survival (EFS) in patients splenectomized before transplantation and according to spleen size at time of allograft.

Close modal
Figure 7.

Kaplan-Meier estimate of event-free survival (EFS) according to the patient's karyotype.

Figure 7.

Kaplan-Meier estimate of event-free survival (EFS) according to the patient's karyotype.

Close modal

Finally, the probability of EFS of the 14 patients with NF1 was lower, although not statistically significantly, than that of children who did not have NF1 (Table 3).

This is the largest study reported so far on children with JMML treated in the context of a prospective clinical trial with the same preparative regimen. With the follow-up now available, our data support the conclusion that allogeneic HSCT may cure approximately 50% of patients with JMML, with disease recurrence being the major cause of treatment failure. The probability of children enrolled in this study being alive and disease free compares favorably with that of many previously published reports on HSCT in children with JMML.10-14  In particular, the overall probability of EFS of the 48 children receiving transplants from an HLA-identical relative is 55%, a value better than that (38%) reported in the retrospective analysis published by our EWOG-MDS group on 24 children given the allograft from a family donor.11  Likewise, the EFS probability of 49% at 5 years after the allograft we have obtained in children receiving transplants from an unrelated volunteer is higher than that reported by the recent retrospective analysis of the National Marrow Donor Program in 46 children (24% at 2 years after transplantation).14 

The choice of adopting a preparative regimen consisting of 3 alkylating agents was based on a preliminary study demonstrating the safety of this therapy18  and on the fact that a retrospective analysis of the EWOG-MDS group showed that a myeloablative therapy including busulfan was associated with a better EFS and a lower relapse incidence in comparison with regimens employing total body irradiation.11  Furthermore, we reasoned that avoiding radiotherapy could have the advantage of reducing the risk of severe radiation-induced growth retardation,30  endocrine and neuropsychological sequelae,31-33  and secondary malignancies.34,35  The results of this study confirm that the preparative regimen is safe, as the cumulative incidence of TRM was only 13%, with no significant difference between recipients of either HLA-identical sibling or UD transplants.

Also, in terms of ultimate outcome, our results seem to indicate that using UDs offers minimal or possibly no significant disadvantage as compared with employing an HLA-identical sibling. These data are in agreement with previously published studies in children with acute leukemia, where the probability of EFS in recipients of sibling HSCT was reported to be comparable to that of children given a UD allograft in the most recent years, thus suggesting the possibility of applying the same indications for HSCT independently of the type of donor available (ie, an HLA-identical sibling or an HLA-matched UD).36,37  Several factors may have contributed to the favorable outcome of our children receiving transplants from a UD. The possibility of selecting the donor using high-resolution molecular typing of HLA loci has been suggested to be potentially able to decrease the risk of graft failure, GVHD, and TRM.38,39  A learning and experience effect in handling recipients of UD HSCT, as well as optimization of the strategies of both prevention and treatment of GVHD, are also variables that could have contributed to the improved outcome.

Our results confirm the conclusion, reported in previously published studies,11-15  that relapse is the major cause of treatment failure in patients with JMML undergoing allogeneic HSCT. Relapse occurred in one third of our patients after a relatively short time from the allograft, with the median time from HSCT being 6 months with only 2 patients having relapsed later than 18 months after transplantation. Disease progression was also the most frequent cause of death. Previously published studies have found that older age,11,15  increased HbF,14  and abnormal karyotype15  are patient-specific risk factors for relapse after transplantation and that occurrence of chronic GVHD protects from the risk of disease relapse.14  Age above 2 years at diagnosis and a high percentage of HbF at diagnosis have been found to predict short survival also in studies analyzing the natural history of the disease in patients with JMML not given HSCT.2,5,6  In univariate analysis, we found that older age, female sex, increased percentage of HbF, and blast percentage in the bone marrow above 20% predicted the occurrence of leukemia relapse. However, only the former of these 4 variables remained significant in multivariate analysis.

Despite the usually aggressive reemergence of the malignant clone and the short time interval between first and second HSCT, a substantial number of our children (7 of the 15 who were given a second allograft) have achieved a second sustained hematologic remission thanks to a second transplantation. It is reasonable to hypothesize that less intensive GVHD prophylaxis could have contributed to the sustained remission after the second allograft in these patients by better preserving GVL effect. This finding is in agreement with previously published reports40,41  and indicates that leukemia relapse does not necessarily means a desperate prognosis and that a second transplantation should be considered as an option to be offered to every patient in good clinical condition.

Despite the delayed hematologic recovery, the ultimate outcome of cord blood transplant recipients was comparable to that of children given HSCT using either bone marrow or peripheral blood progenitors, thus providing further support to previously published studies that reported similar probabilities of EFS in children with malignancies undergoing transplantation with either placental blood or bone marrow-derived hematopoietic stem cells.42,43  The advantages of using cord blood are mainly represented by the prompt availability of this source of hematopoietic progenitors, which shortens the time needed to locate a suitable donor, and by the possibility of performing transplantations in the presence of 1 or 2 HLA disparities in the donor-recipient pairs.43,44  Both these factors can be relevant for treating children with JMML, whose disease, often running an aggressive clinical course, might not allow an extended time for finding a suitable unrelated bone marrow donor.

Two thirds of our patients had a normal karyotype, monosomy 7 being the most frequent cytogenetic anomaly. In contrast with a previous report documenting a negative impact of abnormal karyotype on the probability of OS after HSCT,15  we found that neither monosomy 7 nor other cytogenetic abnormalities confer a worse prognosis. This finding supports a recently published study from a United Kingdom study group on childhood MDS, which reported that in children with JMML monosomy 7 was associated with an outcome comparable to or even better than that of patients with normal karytotype.45 

Splenectomy before HSCT, as well as spleen size at time of the allograft, did not appear to have an impact on posttransplantation outcome. One could argue that patients given splenectomy before transplantation were those with the largest spleen and, thus, with the greatest tumor burden, this possibly being associated with a higher risk of treatment failure. However, the fact that spleen size at time of transplantation influenced neither the risk of relapse nor the probability of survival in patients who did not undergo splenectomy does not support this hypothesis. The results of this study, as well those of previously published reports,11,14,46  are not in favor of an indiscriminate use of splenectomy before transplantation, the potential advantages having to be weighed against the risks related to the procedure or to postsplenectomy infections. The indication of performing splenectomy has to be carefully evaluated for each single child, the presence of massive splenomegaly with evidence of hypersplenism and/or refractoriness to platelet transfusions being an argument for considering this procedure in order to promote engraftment, to hasten hematologic recovery, and to lower the risk of hemorrhagic complications.

Clinical remissions and long-term survival after AML-type combination therapy have been reported in small series of children with JMML.8,47,48  Other investigators, however, pointed out that intensive chemotherapy is notably unsuccessful, especially in patients with aggressive disease.6,9,49  Neither EFS was improved; nor was relapse incidence reduced in our patients who had received intensive chemotherapy before the allograft. Thus, in view of these results, intensive chemotherapy prior to allogeneic HSCT cannot be recommended.

The worse outcome of female patients, also confirmed in multivariate analysis, is a finding never reported in previously published studies on children with JMML given HSCT.10-15  There is no immediate explanation for this finding, although it is noteworthy that among females there was a relatively higher percentage of patients with both NF1 and a low platelet count at time of diagnosis.

In conclusion, this study indicates that HSCT, after a preparative regimen consisting of busulfan, cyclophosphamide, and melphalan, may cure approximately 50% of patients with JMML and that nowadays results achievable using UD are comparable to those obtained employing an HLA-compatible related donor. Identification of factors influencing relapse rate and EFS can be of help in counseling patients. Disease recurrence remains the major cause of treatment failure, and novel strategies to lower the risk of relapse are warranted. In this regard, a reduction in both intensity and duration of GVHD prophylaxis might favor the emergence of a GVL effect displayed by donor lymphocytes, thus contributing to better leukemia control.

TBL5 

The following transplantation teams enrolled patients in the present study.


Transplant center

Country

Principal investigator
Berlin   Germany   Wolfram Ebell  
Bologna   Italy   Andrea Pession  
Cagliari   Italy   Franca Argiolu  
Copenhagen   Denmark   Carsten Heilmann  
Dublin   Ireland   Angus O'Marcaigh  
Düsseldorf   Germany   Dagmar Dilloo  
Erlangen   Germany   Wolfgang Holter  
Essen   Germany   Bernhard Kremens  
Frankfurt   Germany   Thomas Klingebiel  
Freiburg   Germany   Charlotte Niemeyer  
Geneva   Switzerland   Pierre Wacker  
Genova   Italy   Giorgio Dini  
Giessen   Germany   Alfred Reiter  
Greifswald   Germany   James Beck  
Hamburg   Germany   Hartmut Kabisch  
Hannover   Germany   Karl Sykora  
Jena   Germany   Felix Zintl  
Kiel   Germany   Alexander Claviez  
Leiden   The Netherlands   Elisabeth Korthof  
Lund   Sweden   Albert Bekassy  
Monza   Italy   Cornelio Uderzo  
München   Germany   Monika Führer  
Padova   Italy   Chiara Messina  
Paraná   Brazil   Carmen S. Bonfim  
Pavia   Italy   Franco Locatelli  
Pisa   Italy   Claudio Favre  
Prague   Czech Republic   Jan Stary  
Tübingen   Germany   Peter Bader  
Uppsala   Sweden   Johan Arvidson  
Utrecht   The Netherlands   Tom Révész  
Wien
 
Austria
 
Christina Peters
 

Transplant center

Country

Principal investigator
Berlin   Germany   Wolfram Ebell  
Bologna   Italy   Andrea Pession  
Cagliari   Italy   Franca Argiolu  
Copenhagen   Denmark   Carsten Heilmann  
Dublin   Ireland   Angus O'Marcaigh  
Düsseldorf   Germany   Dagmar Dilloo  
Erlangen   Germany   Wolfgang Holter  
Essen   Germany   Bernhard Kremens  
Frankfurt   Germany   Thomas Klingebiel  
Freiburg   Germany   Charlotte Niemeyer  
Geneva   Switzerland   Pierre Wacker  
Genova   Italy   Giorgio Dini  
Giessen   Germany   Alfred Reiter  
Greifswald   Germany   James Beck  
Hamburg   Germany   Hartmut Kabisch  
Hannover   Germany   Karl Sykora  
Jena   Germany   Felix Zintl  
Kiel   Germany   Alexander Claviez  
Leiden   The Netherlands   Elisabeth Korthof  
Lund   Sweden   Albert Bekassy  
Monza   Italy   Cornelio Uderzo  
München   Germany   Monika Führer  
Padova   Italy   Chiara Messina  
Paraná   Brazil   Carmen S. Bonfim  
Pavia   Italy   Franco Locatelli  
Pisa   Italy   Claudio Favre  
Prague   Czech Republic   Jan Stary  
Tübingen   Germany   Peter Bader  
Uppsala   Sweden   Johan Arvidson  
Utrecht   The Netherlands   Tom Révész  
Wien
 
Austria
 
Christina Peters
 

Prepublished online as Blood First Edition Paper, September 7, 2004; DOI 10.1182/blood-2004-05-1944.

A complete list of the transplantation teams that enrolled patients in the present study appears in the “Appendix.”

Partly supported by grants from AIRC (Associazione Italiana Ricerca sul Cancro), CNR (Consiglio Nazionale delle Ricerche), 6th Framework Programme European Community “ALLOSTEM,” and IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Policlinico S. Matteo to F.L.

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
Freedman MH, Estrov Z, Chan HS. Juvenile chronic myelogenous leukemia.
Am J Pediatr Hematol Oncol.
1988
;
10
:
261
-267.
2
Niemeyer CM, Aricò M, Basso G, et al. Chronic myelomonocytic leukemia in childhood: a report of 110 cases.
Blood.
1997
;
89
:
3534
-3543.
3
Emanuel PD, Shannon KM, Castleberry RP. Juvenile myelomonocytic leukemia: molecular understanding and prospects for therapy.
Mol Med Today.
1996
;
2
:
468
-475.
4
Luna-Fineman S, Shannon KM, Atwater SK, et al. Myelodysplastic and myeloproliferative disorders of childhood: a study of 167 patients.
Blood.
1999
;
93
:
459
-466.
5
Passmore SJ, Hann IM, Stiller CA, et al. Pediatric myelodysplasia: a study of 68 children and a new prognostic scoring system.
Blood.
1995
;
85
:
1742
-1750.
6
Castro-Malaspina H, Schaison G, Passe S, et al. Subacute and chronic myelomonocytic leukemia in children (Juvenile CML).
Cancer.
1984
;
54
:
675
-686.
7
Chan HSL, Estrov Z, Weitzman SS, et al. The value of intensive combination chemotherapy for juvenile chronic myelogenous leukemia.
J Clin Oncol.
1987
;
5
:
1960
-1967.
8
Woods WG, Barnard DR, Alonzo TA, et al. Prospective study of 90 children requiring treatment for juvenile myelomonocytic leukemia or myelodysplastic syndrome: a report from the Children's Cancer Group.
J Clin Oncol.
2002
;
20
:
434
-440.
9
Hasle H, Kerndrup G, Yssing M, et al. Intensive chemotherapy in childhood myelodysplastic syndrome. A comparison with results in acute myeloid leukemia.
Leukemia.
1996
;
10
:
1269
-1273.
10
Sanders JE, Buckner CD, Thomas ED, et al. Allogeneic marrow transplantation for children with juvenile chronic myelogenous leukemia.
Blood.
1988
;
71
:
1144
-1146.
11
Locatelli F, Niemeyer C, Angelucci E, et al. Allogenic bone marrow transplantation for chronic myelomonocytic leukemia in childhood: a report from the European Working Group on Myelodysplastic Syndrome in Childhood.
J Clin Oncol.
1997
;
15
:
566
-573.
12
MacMillan ML, Davies SM, Orchard PJ, et al. Haemopoietic cell transplantation in children with juvenile myelomonocytic leukaemia.
Br J Haematol.
1998
;
103
:
552
-558.
13
Matthes-Martin S, Mann G, Peters C, et al. Allogeneic bone marrow transplantation for juvenile myelomonocytic leukaemia: a single centre experience and review of the literature.
Bone Marrow Transplant.
2000
;
26
:
377
-382.
14
Smith FO, King R, Nelson G, et al. Unrelated donor bone marrow transplantation for children with juvenile myelomonocytic leukaemia.
Br J Haematol.
2002
;
116
:
716
-724.
15
Manabe A, Okamura J, Yumura-Yagi K, et al. Allogeneic hematopoietic stem cell transplantation for 27 children with juvenile myelomonocytic leukemia diagnosed based on the criteria of the International JMML Working Group.
Leukemia.
2002
;
16
:
645
-649.
16
Niemeyer CM, Fenu S, Hasle H, et al. Differentiating juvenile myelomonocytic leukemia from infectious disease.
Blood.
1998
;
91
:
365
-367.
17
Hasle H, Niemeyer CM, Chessells JM, et al. A pediatric approach to the WHO classification of myelodysplastic and myeloproliferative diseases.
Leukemia.
2003
;
17
:
277
-282.
18
Locatelli F, Pession A, Comoli P, et al. Role of allogeneic bone marrow transplantation from HLA-identical sibling or matched unrelated donor in the treatment of children with juvenile chronic myeloid leukaemia.
Br J Haematol.
1996
;
92
:
49
-54.
19
Regazzi M, Locatelli F, Buggia I, Bonetti F, Zecca M, Pregnolato M. Disposition of high dose busulfan in pediatric patients undergoing bone marrow transplantation.
Clin Pharmacol Ther.
1993
;
53
:
45
-52.
20
Locatelli F, Percivalle E, Comoli P, et al. Human cytomegalovirus infection in pediatric patients given allogeneic bone marrow transplantation: role of early treatment of antigenemia on patients' outcome.
Br J Haematol.
1994
;
88
:
64
-71.
21
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.
1974
;
18
:
295
-304.
22
Storb R, Prentice RL, Sullivan KM, et al. Predictive factors in chronic graft-versus-host disease in patients with aplastic anemia treated by bone marrow transplantation from HLA-identical siblings.
Ann Intern Med.
1983
;
98
:
461
-466.
23
Kaplan EL, Meier P. Nonparametric estimation from incomplete observations.
J Am Stat Assoc.
1958
;
53
:
457
-481.
24
Pepe MS, Longton G, Pettinger M, Mori M, Fisher LD, Storb R. Summarizing data on survival, relapse, and chronic graft-versus-host disease after bone marrow transplantation: motivation for and description of new methods.
Br J Haematol.
1993
;
83
:
602
-607.
25
Gooley TA, Leisenring W, Crowley J, Storer BE. Estimation of failure probabilities in the presence of competing risks: new representation of old estimators.
Stat Med.
1999
;
18
:
695
-706.
26
Klein JP, Rizzo JD, Zhang M-J, Keiding N. Statistical methods for the analysis and presentation of the results of bone marrow transplants. Part 1: unadjusted analysis.
Bone Marrow Transplant.
2001
;
10
:
909
-915.
27
Byar DP. Identification of prognostic factors. In: Buyse ME, Staquet MJ, Sylvester RJ, eds.
Cancer Clinical Trials: Methods and Practice
. Oxford, United Kingdom: Oxford Medical Publications;
1988
:
423
-443.
28
Cox DR. Regression models and life tables (with discussion).
J R Stat Soc B.
1972
;
34
:
187
-220.
29
Klein JP, Rizzo JD, Zhang M-J, Keiding N. Statistical methods for the analysis and presentation of the results of bone marrow transplants. Part 2: regression modeling.
Bone Marrow Transplant.
2001
;
11
:
1001
-1011.
30
Giorgiani G, Bozzola M, Locatelli F, et al. Role of busulfan and total body irradiation on growth of pre-pubertal children receiving bone marrow transplantation and results of treatment with recombinant human growth hormone.
Blood.
1995
;
86
:
825
-831.
31
Michel G, Gluckman E, Esperou-Bourdeau H, et al. Allogeneic bone marrow transplantation for children with acute myeloblastic leukemia in first complete remission: impact of conditioning regimen without total-body irradiation—a report from the Societe Francaise de Greffe de Moelle.
J Clin Oncol.
1994
;
12
:
1217
-1222.
32
Parth P, Kennedy RS, Lane NE, Dunlap WP, Ordy JM. Motor and cognitive testing of bone marrow transplant patients after chemo-radiotherapy.
Percept Mot Skills.
1989
;
68
:
1227
-1241.
33
Locatelli F, Labopin M, Ortega J, et al. Factors influencing outcome and incidence of long-term complications in children who underwent autologous stem cell transplantation for acute myeloid leukemia in first complete remission.
Blood.
2003
;
101
:
1611
-1619.
34
Socie G, Curtis RE, Deeg HJ, et al. New malignant diseases after allogeneic marrow transplantation for childhood acute leukemia.
J Clin Oncol.
2000
;
18
:
348
-357.
35
Curtis R, Rowlings PA, Deeg HJ, et al. Solid cancers after bone marrow transplantation.
N Engl J Med.
1997
;
336
:
897
-904.
36
Saarinen-Pihkala UM, Gustafsson G, Ringden O, et al. No disadvantages in outcome of using matched unrelated donors as compared with matched sibling donors for bone marrow transplantation in children with acute lymphoblastic leukemia in second remission.
J Clin Oncol.
2001
;
19
:
3406
-3414.
37
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
;
16
:
2228
-2237.
38
Sasazuki T, Juji T, Morishima Y, et al. Effect of matching of class I HLA alleles on clinical outcome after transplantation of hematopoietic stem cells from an unrelated donor.
N Engl J Med.
1998
;
339
:
1177
-1193.
39
Petersdorf EW, Gooley TA, Anasetti C, et al. Optimizing outcome after unrelated marrow transplantation by comprehensive matching of HLA class I and II alleles in the donor and recipient.
Blood.
1998
;
92
:
3515
-3520.
40
Pamphilon DH, Cornish JM, Goodman S, et al. Successful second unrelated donor BMT in a child with juvenile chronic myeloid leukaemia: documentation of chimaerism using the polymerase chain reaction.
Bone Marrow Transplant.
1993
;
11
:
81
-84.
41
Mahfuzah M, Flotho C, Arvidson J, et al. Second allogeneic stem cell transplantation (SCT) for relapse after first SCT for juvenile myelomonocytic leukaemia (JMML).
Bone Marrow Transplant
. In press.
42
Rocha V, Wagner J, Sobocinski K A, et al. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling. Eurocord and International Bone Marrow Transplant Registry Working Committee on Alternative Donor and Stem Cell Sources.
N Engl J Med.
2000
;
342
:
1846
-1854.
43
Rocha V, Cornish J, Sievers EL, et al. Comparisons of outcomes of unrelated bone marrow and umbilical cord blood transplants in children with acute leukaemia.
Blood.
2001
;
97
:
2962
-2971.
44
Gluckman E, Locatelli F. Umbilical cord blood transplants.
Curr Opin Haematol.
2000
;
7
:
353
-357.
45
Passmore SJ, Chessells JM, Kempski H, Hann IM, Brownbill PA, Stiller CA. Paediatric MDS and JMML in the UK: a population-based study of incidence and survival.
Br J Haematol.
2003
;
121
;
758
-767.
46
Maguire AM, Vowels MR, Russell S, et al. Allogeneic bone marrow transplant improves outcome for juvenile myelomonocytic leukaemia.
J Paediatr Child Health.
2002
;
38
:
166
-169.
47
Chan HS, Estrov Z, Weitzman SS, et al. The value of intensive combination chemotherapy for juvenile chronic myelogenous leukemia.
J Clin Oncol.
1987
;
5
:
1960
-1967.
48
Diaz de Heredia C, Ortega JJ, Coll MT, et al. Results of intensive chemotherapy in children with juvenile chronic myelomonocytic leukemia: a pilot study.
Med Pediatr Oncol.
1998
;
31
:
516
-520.
49
Lutz P, Zix-Kieffer I, Souillet G, et al. Juvenile myelomonocytic leukemia: analyses of treatment results in the EORTC children leukemia cooperative group (CLCG).
Bone Marrow Transplant.
1996
;
18
:
1111
-1116.
Sign in via your Institution