From 2002 to 2007, 103 patients with primary myelofibrosis or postessential thrombocythemia and polycythemia vera myelofibrosis and a median age of 55 years (range, 32-68 years) were included in a prospective multicenter phase 2 trial to determine efficacy of a busulfan (10 mg/kg)/fludarabine (180 mg/m2)–based reduced-intensity conditioning regimen followed by allogeneic stem cell transplantation from related (n = 33) or unrelated donors (n = 70). All but 2 patients (2%) showed leukocyte and platelet engraftment after a median of 18 and 22 days, respectively. Acute graft-versus-host disease grade 2 to 4 occurred in 27% and chronic graft-versus-host disease in 43% of the patients. Cumulative incidence of nonrelapse mortality at 1 year was 16% (95% confidence interval, 9%-23%) and significantly lower for patients with a completely matched donor (12% vs 38%; P = .003). The cumulative incidence of relapse at 3 years was 22% (95% confidence interval, 13%-31%) and was influenced by Lille risk profile (low, 14%; intermediate, 22%; and high, 34%; P = .02). The estimated 5-year event-free and overall survival was 51% and 67%, respectively. In a multivariate analysis, age older than 55 years (hazard ratio = 2.70; P = .02) and human leukocyte antigen–mismatched donor (hazard ratio = 3.04; P = .006) remained significant factors for survival. The study was registered at www.clinicaltrials.gov as #NCT 00599547.

The BCR-ABL–negative myeloproliferative disorder of primary myelofibrosis (PMF)1  and the advanced forms of essential thrombocythemia and polycythemia vera (ie, post-ET/PV myelofibrosis) are chronic hematologic malignancies characterized by splenomegaly, leuko-erythroblastosis, extramedullary hematopoiesis, and circulating CD34+ progenitor cells. Patients with symptomatic forms of PMF have a median survival of less than 5 years.2  The median age at diagnosis is in the seventh decade of life (median, 67 years).3  Curative therapy in PMF is currently possible only with allogeneic hematopoietic stem cell transplantation (AHSCT). Studies on conventional AHSCT included mainly younger patients with a median age between 38 and 54 years. The 5-year survival rate in 2 large studies was 47% and 61%, respectively.4,5  However, the nonrelapse mortality (NRM) at 1 year ranged from 20% to 48%, and one of the most significant prognostic factors for impaired survival was the increasing age of the patients.6  The introduction of reduced-intensity conditioning regimens is based on the concept of eradicating tumor cells to the immunologically mediated graft-versus-tumor effect rather than by high-dose chemotherapy. The potential advantages are less treatment-related morbidity and mortality and a broader applicability in elderly patients. Smaller studies showed a NRM rate of less than 20% and overall survival rates between 84% and 100% at 3 years.7-9  Evidence for an immunologically mediated graft-versus-myelofibrosis effect comes from reports in relapsed patients after AHSCT that show a remarkable reduction of bone marrow fibrosis after donor lymphocyte infusion.10-13 

We here report the outcome of a prospective multicenter phase 2 study in patients with advanced PMF evaluating a dose-reduced conditioning regimen followed by stem cell transplantation from related or unrelated donors.

Aim of the study

The primary endpoint was NRM by 1 year after transplantation, and secondary endpoints were incidence of acute and chronic graft-versus-host disease (GVHD), overall and disease-free survival, and incidence of relapse after transplantation.

Major inclusion criteria were histologically proven myelofibrosis with either an intermediate-risk or high-risk score according to the Lille scoring system14  or a low-risk Lille score but with constitutional symptoms, or a high-risk Cervantes score,15  and age between 18 and 70 years. Patients should have a human leukocyte antigen (HLA)–compatible related or unrelated donor. One HLA allele or antigen mismatch was allowed.

Between 2002 and 2007, 103 patients with PMF or post-ET/PV myelofibrosis and a median age of 55 years (range, 32-68 years) were enrolled. Seventeen transplantation centers from 3 nations included 62 male and 41 female patients with primary (n = 63) or post-PV/ET myelofibrosis (n = 40). Risk profile according the Lille score was low risk with constitutional symptoms (17%), intermediate risk (n = 53%), and high risk (n = 30%). All but 4 patients received peripheral blood stem cells as graft source, from either related (n = 33) or unrelated donors (n = 70). The median number of transplanted CD34+ cells per kilogram of body weight (kg BW) was 6.8 × 106 (range, 0.7-21.7 × 106). JAK2 V617 mutation status was available in 82 patients (Table 1). All patients gave written informed consent in accordance with the Declaration of Helsinki, and the study was approved by the ethic committees of all participating institutions and the national health authorities.

Table 1

Patient characteristics and results (n = 103 patients)

CharacteristicValue
Patient median age, y (range) 55 (32-68) 
Donor median age, y (range) 42 (17-69) 
Sex of patient, M/F 62/41 
Sex of donor, M/F 65/38 
PMF  
    Primary 63 
    Post-PV/ET 40 
Median time for diagnosis to SCT, mo (range) 19 (3-276) 
Risk profile according to Lille score  
    Low 17 
    Intermediate 55 
    High 31 
Risk profile according to Cervantes score  
    Low 23 
    High 76 
    Unknown 
Spleen size before stem cell transplantation  
    Enlarged 80 
    Not enlarged 
    Splenectomy 14 
    Unknown 
JAK2 V617F mutation status  
    Positive 59 
    Negative 23 
    Unknown 21 
Cytogenetics  
    Abnormal 29 
    Normal 37 
    Unknown 37 
HLA matching (10/10 allele)  
    Matched 82 
    Mismatched 21 
HLA-identical sibling 33 
Matched unrelated donor 70 
Stem cell source  
    Bone marrow 
    Peripheral blood stem cells 99 
    Peripheral blood stem cells plus bone marrow 
Median no. of CD34+ infused stem cells/kg BW (range) 6.8 × 106 (0.7-21.7) 
CMV serostatus of recipients  
    Positive 47 
    Negative 55 
    Unknown 
Fibrosis grading (n = 73)  
    Grade 0 
    Grade 1 
    Grade 2 28 
    Grade 3 37 
Primary graft failure 2 (2%) 
Median leukocyte engraftment (NC > 1.0 × 109/L), days (range) 18 (10-84) 
Median platelet engraftment (platelet > 20 × 109/L), days (range) 22 (8-145) 
Toxicity grade 3 or 4  
    Liver 10% 
Acute GVHD  
    Grade 0 to 1 74% 
    Grade 2 to 4 26% 
    Grade 3 or 4 11% 
Chronic GVHD  
    Limited 24% 
    Extensive 24% 
Veno-occlusive disease (n = 93)  
    None 80% 
    Mild 7% 
    Moderate 11% 
    Severe 2% 
Response  
    Complete histo-hematologic 44 
    Partial histo-hematologic 
    Treatment failure 11 
    Missing or unclear 45 
CharacteristicValue
Patient median age, y (range) 55 (32-68) 
Donor median age, y (range) 42 (17-69) 
Sex of patient, M/F 62/41 
Sex of donor, M/F 65/38 
PMF  
    Primary 63 
    Post-PV/ET 40 
Median time for diagnosis to SCT, mo (range) 19 (3-276) 
Risk profile according to Lille score  
    Low 17 
    Intermediate 55 
    High 31 
Risk profile according to Cervantes score  
    Low 23 
    High 76 
    Unknown 
Spleen size before stem cell transplantation  
    Enlarged 80 
    Not enlarged 
    Splenectomy 14 
    Unknown 
JAK2 V617F mutation status  
    Positive 59 
    Negative 23 
    Unknown 21 
Cytogenetics  
    Abnormal 29 
    Normal 37 
    Unknown 37 
HLA matching (10/10 allele)  
    Matched 82 
    Mismatched 21 
HLA-identical sibling 33 
Matched unrelated donor 70 
Stem cell source  
    Bone marrow 
    Peripheral blood stem cells 99 
    Peripheral blood stem cells plus bone marrow 
Median no. of CD34+ infused stem cells/kg BW (range) 6.8 × 106 (0.7-21.7) 
CMV serostatus of recipients  
    Positive 47 
    Negative 55 
    Unknown 
Fibrosis grading (n = 73)  
    Grade 0 
    Grade 1 
    Grade 2 28 
    Grade 3 37 
Primary graft failure 2 (2%) 
Median leukocyte engraftment (NC > 1.0 × 109/L), days (range) 18 (10-84) 
Median platelet engraftment (platelet > 20 × 109/L), days (range) 22 (8-145) 
Toxicity grade 3 or 4  
    Liver 10% 
Acute GVHD  
    Grade 0 to 1 74% 
    Grade 2 to 4 26% 
    Grade 3 or 4 11% 
Chronic GVHD  
    Limited 24% 
    Extensive 24% 
Veno-occlusive disease (n = 93)  
    None 80% 
    Mild 7% 
    Moderate 11% 
    Severe 2% 
Response  
    Complete histo-hematologic 44 
    Partial histo-hematologic 
    Treatment failure 11 
    Missing or unclear 45 

SCT indicates stem cell transplantation; CMV, cytomegalovirus; and NC, neutrophil count.

Treatment plan

The reduced-intensity conditioning regimen consisted of busulfan, 10 mg/kg BW orally (or busulfan, intravenously, in equivalent doses) given in 10 doses (1 mg/kg BW) over 3 days; fludarabine, 180 mg/m2, given as 30 mg/m2 over 6 days; and antilymphocyte-globulin (Fresenius), 3 × 10 mg/kg BW (for related transplantation) or 3 × 20 mg (for unrelated transplantation), followed by allogeneic stem cell transplantation.

HLA typing

HLA typing for inclusion required serologically testing for class I (HLA-A and HLA-B) and with sequence-specific oligonucleotide probes for class II (HLA-DRB1 and DQB1). For the analysis results, sequence-specific oligonucleotide probes for HLA-A, -B, -C, -DRB1, and -DQB1 alleles were used. Overall, according to high-resolution HLA typing, 82 patients had a completely HLA-matched donor; whereas in 21 patients, donors had at least one allele or antigen mismatch: A locus, n = 3; B locus, n = 1; C locus, n = 6; DRB1 locus, n = 2; DQB1 locus, n = 6; locus A plus C, n = 2; and locus A plus DQB1, n = 1.

GVHD prophylaxis and scoring

The GVHD prophylaxis consisted of cyclosporine A at a dose of 3 mg/kg BW as continuous infusion starting at day 1. The cyclosporine A dose should be adjusted according to the serum level of 200 ng/mL. If no GVHD occurs, cyclosporine A should be tapered and discontinued after day 180. Further GVHD prophylaxis consisted of methotrexate (10 mg/m2) at days 1, 3, and 6. The scoring of acute GVHD was performed according to Glucksberg et al16  and chronic GVHD according to the criteria of Shulman et al.17 

Response criteria

The following criteria were used for evaluating response to treatment. Complete hematologic remission (CHR) was defined as disappearance of all clinical signs of myelofibrosis, and peripheral blood and cytogenetic abnormalities attributable to the disease. Complete histo-hematologic remission was defined as a combination of a CHR with disappearance of myelofibrosis. Partial histo-hematologic remission was defined as CHR with partial regression of myelofibrosis. Treatment failure and relapse was defined as disease recurrence or an observed persistence of disease in patients who survived more than 30 days after transplantation.

Statistical analysis

Comparisons of subgroups with respect to nominal variables were made using the χ2 test and Fisher exact test. Survival curves were estimated using the Kaplan-Meier method (if no competing risk were involved) and then the significance of the log-rank test was used, or they were computed in a competing risk framework (to estimate the cumulative incidence of a specific event in case of competing risks). For multivariate analyses, the Cox model was used to estimate hazard ratios (HRs) and cause-specific hazards; but in case of competing risks, the curve estimates of the Cox model were not used because they are inappropriate in that case. For survival, the event was “death from any cause,” and the corresponding time interval was taken from transplantation to the date of this event (uncensored) or the last follow-up date (censored), whichever came first. For disease-free survival, the event was defined as “relapse or progression or death from any cause” and the corresponding time interval again from transplantation onward. For NRM, the event was defined as “death related to transplantation”; the corresponding time interval was again calculated from transplantation until this event (uncensored) or the last follow-up of the patient (censored), which implies that patients who died from other causes were censored at the time of death. The cumulative incidence method was used to estimate the incidence of both NRM and relapse to account for competing events. Calculations were performed in SPSS, Version 12/15 (SPSS). The competing risk analyses were done with the ACCorD (V. Gebski, National Health and Medical Research Council, Clinical Trial Center, University of Sydney). The following factors were included in the univariate analysis: age, HLA match, Lille score, Cervantes score, number of monocytes, cytogenetics, related versus unrelated donor, splenectomy, spleen size, bone marrow fibrosis grade, JAK2 V617F mutation, primary versus post-ET/PV MF, and time from diagnosis to transplantation. Factors that had P value of less than .1 univariately were included in multivariate analysis using the HRs estimated in Cox models.

Engraftment

All but 2 patients experienced engraftment of leukocytes. Two patients (2%) transplanted from a fully matched unrelated donor experienced primary graft failure. One patient received additional bone marrow cells from the same donor but did not show engraftment and died of aspergillus infection. The other patient recovered with autologous cells but died of sepsis 5 months after transplantation. The median time to leukocyte engraftment (absolute neutrophil count > 1.0 × 109/L) was 18 days (range, 10-84 days), and the median time to platelet engraftment (> 20 × 109/L) was 22 days (range, 8-145 days). Patients with splenectomy had a trend for faster leukocyte engraftment (16 vs 18 days). Eleven patients (11%) received an additional stem cell boost after transplantation resulting from poor graft function with persistent cytopenia (n = 8) or decreasing donor chimerism (n = 3).

GVHD

Overall, 73% of the study population did not experience any or more than grade 1 acute GVHD The incidence of acute GVHD grades 2 to 4 was 27%, and of severe GVHD grades 3 to 4, 11%. There was no difference in acute GVHD between transplantation from sibling or unrelated donor. The overall rate of chronic GVHD was 49%, which was limited disease in 24% and extensive disease in 24%. In a landmark analysis, including only patients who survived 1 year after transplantation, the estimated 3-year probability of posttransplantation survival was higher in patients with than without chronic GVHD (97% vs 89%; P = .04) supporting a graft-versus-myelofibrosis effect.

Mortality

During follow-up, 27 patients died. The main reasons for death were relapse or progression (n = 8), followed by GVHD (n = 7), infectious complications (n = 5), multiorgan failure (n = 2), liver toxicity (n = 1), posttransplantation lymphoma (n = 1), suicide (n = 1), accidental bleeding after central venous access (n = 1), and amyotrophic lateral sclerosis (n = 1). The cumulative incidence of NRM at 1 year was 16% (95% confidence interval [CI], 9%-23%). NRM at 1 year did not differ significantly between the HLA-identical sibling and the 10/10-matched unrelated group (10% vs 13%, P = .50) but was significantly higher after mismatched than after completely matched stem cell transplantation (38% vs 12%, P = .003; Table 2), which was the only significant factor in the univariate analysis.

Table 2

Univariate analysis of risk factors (only with P ≤ .1) after reduced-intensity conditioning and allogeneic stem cell transplantation for myelofibrosis

Variable5-year overall survivalP3-year disease-free survivalPNRM at 1 yearPRelapse at 3 yearsP
Lille score  .07  .01    .02 
    Low 94 (82-100)  80 (60-100)  —  14 (0-32)  
    Intermediate 63 (46-80)  61 (47-75)  —  22 (9-35)  
    High 64 (47-81)  41 (21-61)  —  34 (16-52)  
HLA  .003  .01  .003   
    Matched 74 (62-86)  63 (52-74)  12 (5-19)  —  
    Mismatched 38 (5-71)  39 (16-62)  38 (15-61)  —  
Age  .03       
    Less than or equal to 55 y 82 (71-93)  —  —  —  
    More than 55 y 48 (26-70)  —  —  —  
Splenectomy    .06    .005 
    Yes —  39 (12-66)  —  51 (20-82)  
    No —  61 (47-75)  —  20 (10-30)  
Variable5-year overall survivalP3-year disease-free survivalPNRM at 1 yearPRelapse at 3 yearsP
Lille score  .07  .01    .02 
    Low 94 (82-100)  80 (60-100)  —  14 (0-32)  
    Intermediate 63 (46-80)  61 (47-75)  —  22 (9-35)  
    High 64 (47-81)  41 (21-61)  —  34 (16-52)  
HLA  .003  .01  .003   
    Matched 74 (62-86)  63 (52-74)  12 (5-19)  —  
    Mismatched 38 (5-71)  39 (16-62)  38 (15-61)  —  
Age  .03       
    Less than or equal to 55 y 82 (71-93)  —  —  —  
    More than 55 y 48 (26-70)  —  —  —  
Splenectomy    .06    .005 
    Yes —  39 (12-66)  —  51 (20-82)  
    No —  61 (47-75)  —  20 (10-30)  

Values are shown as percentages (95% CI).

— indicates not applicable.

Response

Best hematologic and histopathologic response 1 year after transplantation was available in 58 patients. The percentage for complete histo-hematologic response was 74%, and for partial histo-hematologic response 5%, whereas 19% had treatment failure.

Relapse

The cumulative incidence of relapse and treatment failure at 3 and 5 years was 22% (95% CI, 13%-31%) and 29% (95% CI, 16%-42%), respectively. There was no significant difference in the incidence of relapse between sibling and unrelated donor transplantation (32% vs 20%; P = .6) and between the HLA-matched and mismatched stem cell donors (23% vs 30%; P = .35). In univariate analysis, significant factors for higher relapse rate at 3 years were risk profile according to Lille scores: low risk of 14% versus intermediate of 22% versus high risk of 34% (P = .02); and splenectomy of 51% versus no splenectomy of 20% (P = .005; Table 2). In the multivariate analysis, significant factors for a higher incidence of relapse at 3 years were splenectomy (HR = 3.58; 95% CI, 1.44-8.86; P = .006) and Lille high-risk score (HR = 5.23; 95% CI, 1.14-24.01; P = .003; Table 3).

Table 3

Multivariate analysis of risk factors after reduced-intensity conditioning and allogeneic stem cell transplantation for patients with myelofibrosis

Overall survival, HR (95% CI)PDisease-free survival, HR (95% CI)PRelapse, HR (95% CI)P
Age       
    Less than or equal to 55 y 1.0 .02 —  —  
    More than 55 y 2.70 (1.21-6.03)  —  —  
HLA       
    Match 1.0 .006 1.0 .01 —  
    Mismatched 3.04 (1.38-6.65)  2.39 (1.22-4.69)  —  
Lille score       
    Low —  1.0 .02 1.0 .03 
    Intermediate —  3.10 (0.91-10.58) .07 1.94 (0.42-9.02) .40 
    High —  5.37 (1.58-18.24) .007 5.23 (1.14-24.01) .03 
Splenectomy       
    No —  —  1.0 .006 
    Yes —  —  3.58 (1.44-8.86)  
Overall survival, HR (95% CI)PDisease-free survival, HR (95% CI)PRelapse, HR (95% CI)P
Age       
    Less than or equal to 55 y 1.0 .02 —  —  
    More than 55 y 2.70 (1.21-6.03)  —  —  
HLA       
    Match 1.0 .006 1.0 .01 —  
    Mismatched 3.04 (1.38-6.65)  2.39 (1.22-4.69)  —  
Lille score       
    Low —  1.0 .02 1.0 .03 
    Intermediate —  3.10 (0.91-10.58) .07 1.94 (0.42-9.02) .40 
    High —  5.37 (1.58-18.24) .007 5.23 (1.14-24.01) .03 
Splenectomy       
    No —  —  1.0 .006 
    Yes —  —  3.58 (1.44-8.86)  

— indicates not applicable.

Survival

After a median follow-up of 33 months (range, 12-76 months), 5-year estimated disease-free and overall survival was 51% (95% CI, 38%-64%) and 67% (95% CI, 55%-79%) (Figure 1). In the univariate analysis, significant factors for improved disease-free survival at 3 years were HLA-matched versus mismatched donor (59% vs 40% at 3 years, P = .01) and Lille score low versus intermediate versus high (80% vs 61% vs 41%; P = .01). For overall survival, younger age (< 55 years, 82% vs 48%, P = .003), HLA-matched versus mismatched donor (74% vs 38%, P = .003) had an improved survival rate (Table 2). Furthermore, JAK2+ patients had a better disease-free and overall survival rate than JAK2 patients (56% vs 34%, P = .04; and 76% vs 42%, P = .01) but were not included in the multivariate analysis because of incomplete dataset. In multivariate analysis, HLA-mismatched donor (HR = 2.39; 95% CI, 1.22-4.69; P = .01) and advanced Lille score (intermediate, HR = 3.10; P = .07; and high, HR = 5.37; P = .007) remained independent risk factors for disease-free and age older than 55 years (HR = 2.70; 95% CI, 1.20-6.03; P = .02) and HLA-mismatched transplantation (HR = 3.04; 95% CI, 1.38-6.65; P = .006) for overall survival (Table 3). If in multivariate analysis JAK2 mutation status is included, JAK2 positivity remained an independent factor for disease-free survival and for overall survival (HR = 2.00; 95% CI, 1.02-3.93; P = .04; and HR = 2.72; 95% CI, 1.18-6.28; P = .02, respectively).

Figure 1

Survival of patients with myelofibrosis after reduced-intensity allogeneic stem cell transplantation. According to age (A), donor (B), and Lille risk profile (C).

Figure 1

Survival of patients with myelofibrosis after reduced-intensity allogeneic stem cell transplantation. According to age (A), donor (B), and Lille risk profile (C).

Close modal

Fibrosis regression and JAK2 clearance

In a subset of patients (n = 16) treated in one center (Hamburg), a sequential analysis of bone marrow fibrosis regression was performed after engraftment (between day 30 and day 50), on day 100 and day 365 after transplantation by 2 hemato-pathologists (J.T., H.M.K.). Before transplantation, all patients had advanced grade 2 (n = 5) or grade 3 (n = 11) bone marrow fibrosis. Shortly after engraftment, 21% had a regression from grade 3 to 2 and 36% had near or complete resolution (grade 1 or 0) of bone marrow fibrosis. A near or complete resolution (grade 1 and 0) at day 100 and day 365 was observed in 69% and 93%, respectively (Figure 2).

Figure 2

Representative fibrosis regression after dose-reduced allograft. Bone marrow fibrosis grade 3 before transplantation (A), grade 1 after engraftment on day 30 (B), and grade 0 on day 100 after allogeneic stem cell transplantation (C). Image acquisition specifications: Zeiss Axioplan microscope; magnification ×180, oil; Plan Apochromat 20×/0.60 imaging medium/solution; Axiocam HRC camera; Adobe Photoshop software.

Figure 2

Representative fibrosis regression after dose-reduced allograft. Bone marrow fibrosis grade 3 before transplantation (A), grade 1 after engraftment on day 30 (B), and grade 0 on day 100 after allogeneic stem cell transplantation (C). Image acquisition specifications: Zeiss Axioplan microscope; magnification ×180, oil; Plan Apochromat 20×/0.60 imaging medium/solution; Axiocam HRC camera; Adobe Photoshop software.

Close modal

In 29 patients with JAK2V617F positivity, mutation could be followed after allogeneic stem cell transplantation with highly sensitive quantitative polymerase chain reaction technique.18  Twenty-one patients (72%) achieved JAK2 negativity after a median of 106 days (range, 20-437 days) after transplantation. Of the remaining 8 JAK2V617F+ patients, 1 died of therapy-related complications and 7 developed clinical relapse. Six of 7 patients achieved remission with JAK2 negativity after donor lymphocyte infusion (n = 2) or second allogeneic stem cell transplantation (n = 4).

Current medical therapeutic options for patients with PMF or myelofibrosis after PV or ET have not demonstrated an impact on the disease course, which exceeds the palliation of disease-related extramedullary hematopoiesis and alleviation of cytopenias. As in other hematologic malignancies, allogeneic stem cell transplantation can lead to “cure” but is limited because of the patient's age or comorbidities. Conventional myeloablative allogeneic stem cell transplantation was performed in younger patients, which does not reflect the median age of approximately 67 years for patients with myelofibrosis at time of diagnosis. However, this large prospective multicenter trial shows that a reduced-intensity conditioning regimen followed by allogeneic stem cell transplantation from related or unrelated histo-compatible donors is a reasonable and potential curative treatment option, even for elderly patients with PM or post-PV/ET myelofibrosis. The NRM was 16% and considerably lower than reported after standard myeloablative conditioning regimens. HLA-mismatched transplantation remains a major risk factor for therapy-related mortality and demonstrated the need for a careful donor selection. If patients without a fully matched HLA-compatible donor (n = 21) are excluded, the cumulative incidence of NRM at 1 year is only 12% and does not differ between HLA-identical siblings and fully matched unrelated donors (10% vs 13%). HLA-mismatched transplantation was also an independent risk factor for overall survival. If those patients are excluded, the 5-year estimated disease-free and overall survival rates increased from 51% to 59% and from 67% to 74%, respectively. Another important risk factor for outcome was age. Patients younger than 55 years had an estimated 5-year overall survival of 82%, whereas transplantation of patients older than 55 years of age resulted in an estimated 5-year survival of 48%. This is the result of a nonsignificant higher relapse and higher NRM for elderly patients. A third important risk factor is the stage of the disease determined by the Lille score. Independently, the stage of the disease influenced mostly the incidence of relapse, which was lowest for low-risk (14%) and highest for high-risk disease (34%), resulting in lower disease-free survival for patients with intermediate- and high-risk disease. Therefore, the timing of stem cell transplantation seems to be crucial and should be performed before the disease has developed to a very advanced stage. Whether a more intensified conditioning regimen might lower the risk of relapse in Lille high-risk patients should be investigated in a further trial. Results of standard myeloablative conditioning are similar, but the included patient population after myeloablative conditioning were younger with a median age of 42 years4  or 49 years,5  respectively. More recently, the Gruppo Italiano Trapianto di Midollo Osseo did not find a difference between reduced-intensity and standard myeloablative conditioning for patients with myelofibrosis, but the treatment-related mortality at 3 years with 43% was rather high.19  However, in the same study, a clear improvement in TRM in the more recent years could be observed. An unexpected finding of this study was a more than 3-fold increase of relapse for splenectomized patients who underwent allogeneic stem cell transplantation. The number of splenectomized patients in this trial was rather low (n = 14) to draw definite conclusions; but given the high surgery-related morbidity and mortality of splenectomy in myelofibrosis patients, splenectomy before stem cell transplantation should not be recommended, even if leukocyte engraftment tended to be faster in those patients as shown in our and other trials.4,5,20  It is also of note that those patients with nonenlarged or only modestly enlarged spleens (n = 8) also had an increased risk of relapse; but because of the low number, this factor was not included in the multivariate analysis. Other factors, such as cytogenetics, time between diagnosis and transplantation, primary MF versus MF post-PV/ET, and related versus unrelated donors, did not influence outcome. Interestingly, patients with the JAK2 V617 mutation had improved disease-free and overall survival rates; but because of an incomplete dataset, this factor was not included in the multivariate analysis. The place of allogeneic stem cell transplantation for patients with myelofibrosis has to be defined,21  especially in the era of molecular target therapy with the new JAK2 inhibitors. Recent data suggest that JAK2 inhibitors will not cure myelofibrosis patients but can relieve constitutional symptoms and splenomegaly.22  Therefore, the new JAK2-tyrosine-kinase inhibitors22  will not replace stem cell transplantation but could be used as remission-inducing drugs to make nontransplantation candidates fit to an allogeneic stem cell transplantation, which should be the subject of further clinical trials. The JAK2 V617 mutation level can be used as a marker for residual disease after transplantation and might be used to guide adoptive immunotherapy with donor lymphocyte infusion.12,21,23 

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.

The authors thank the staff of the clinics for providing excellent care to our patients, the medical technicians for their excellent work in the laboratories, and Karen Klemt for proof editing.

Contribution: N.K. designed the study, analyzed data, and wrote the manuscript; E.H., G.K., M.B., R.S., H.B., A.N., W.B., M.S., L.U., H.W., A.B., P.C., J.S., M.K., P.D., G.G.W., H.E., A.R.Z., and D.N. analyzed the data and approved the manuscript; T.Z. and R.B. performed statistical analysis; T.M.d.W. designed the study and analyzed the data; and H.M.K. and J.T. performed histology examinations.

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

Correspondence: Nicolaus Kröger, Center for Stem Cell Transplantation, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany; e-mail: nkroeger@uke.uni-hamburg.de.

1
Mesa
 
RA
Verstovsek
 
S
Cervantes
 
F
, et al. 
Primary myelofibrosis (PMF), post polycythemia vera myelofibrosis (post-PV MF), post essential thrombocythemia myelofibrosis (post-ET MF), blast phase PMF (PMF-BP): consensus on terminology by the international working group for myelofibrosis research and treatment (IWG-MRT).
Leuk Res
2007
, vol. 
31
 
6
(pg. 
737
-
740
)
2
Barosi
 
G
Hoffman
 
R
Idiopathic myelofibrosis.
Semin Hematol
2005
, vol. 
42
 
4
(pg. 
248
-
258
)
3
Mesa
 
RA
Silverstein
 
MN
Jacobsen
 
SJ
Wollan
 
PC
Tefferi
 
A
Population-based incidence and survival figures in essential thrombocythemia and agnogenic myeloid metaplasia: an Olmsted County Study, 1976-1995.
Am J Hematol
1999
, vol. 
61
 
1
(pg. 
10
-
15
)
4
Guardiola
 
P
Anderson
 
JE
Bandini
 
G
, et al. 
Allogeneic stem cell transplantation for agnogenic myeloid metaplasia: a European Group for Blood and Marrow Transplantation, Societe Francaise de Greffe de Moelle, Gruppo Italiano per il Trapianto del Midollo Osseo, and Fred Hutchinson Cancer Research Center Collaborative Study.
Blood
1999
, vol. 
93
 
9
(pg. 
2831
-
2838
)
5
Kerbauy
 
DM
Gooley
 
TA
Sale
 
GE
, et al. 
Hematopoietic cell transplantation as curative therapy for idiopathic myelofibrosis, advanced polycythemia vera, and essential thrombocythemia.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
3
(pg. 
355
-
365
)
6
Guardiola
 
P
Anderson
 
JE
Gluckman
 
E
Myelofibrosis with myeloid metaplasia [letter].
N Engl J Med
2000
, vol. 
343
 
9
pg. 
659
 
7
Rondelli
 
D
Barosi
 
G
Bacigalupo
 
A
, et al. 
Myeloproliferative Diseases-Research Consortium. Allogeneic hematopoietic stem-cell transplantation with reduced-intensity conditioning in intermediate- or high-risk patients with myelofibrosis with myeloid metaplasia.
Blood
2005
, vol. 
105
 
10
(pg. 
4115
-
4119
)
8
Kröger
 
N
Zabelina
 
T
Schieder
 
H
, et al. 
Pilot study of reduced-intensity conditioning followed by allogeneic stem cell transplantation from related and unrelated donors in patients with myelofibrosis.
Br J Haematol
2005
, vol. 
128
 
5
(pg. 
690
-
697
)
9
Kröger
 
N
Thiele
 
J
Kobbe
 
G
, et al. 
Rapid regression of bone marrow fibrosis after dose-reduced allogeneic stem cell transplantation.
Exp Hematol
2007
, vol. 
35
 
11
(pg. 
1719
-
1722
)
10
Byrne
 
JL
Beshti
 
H
Clark
 
D
, et al. 
Induction of remission after donor leucocyte infusion for the treatment of relapsed chronic idiopathic myelofibrosis following allogeneic transplantation: evidence for a ‘graft vs myelofibrosis’ effect.
Br J Haematol
2000
, vol. 
108
 
2
(pg. 
430
-
433
)
11
Cervantes
 
F
Rovira
 
M
Urbano-Ispizua
 
A
Rozman
 
M
Carreras
 
E
Montserrat
 
E
Complete remission of idiopathic myelofibrosis following donor lymphocyte infusion after failure of allogeneic transplantation: demonstration of a graft-versus-myelofibrosis effect.
Bone Marrow Transplant
2000
, vol. 
26
 
6
(pg. 
697
-
699
)
12
Kröger
 
N
Alchalby
 
H
Klyuchnikov
 
E
, et al. 
JAK2-V617F-triggered preemptive and salvage adoptive immunotherapy with donor-lymphocyte infusion in patients with myelofibrosis after allogeneic stem cell transplantation.
Blood
2009
, vol. 
113
 
8
(pg. 
1866
-
1868
)
13
Benjamini
 
O
Koren-Michowitz
 
M
Mariglio
 
N
Kroger
 
N
Nagler
 
A
Shimoni
 
A
Relapse of postpolycythemia myelofibrosis after allogeneic stem cell transplantation in a polycythemic phase: successful treatment with donor lymphocyte infusion directed by quantitative PCR test for V617F-JAK2 mutation.
Leukemia
2008
, vol. 
22
 
10
(pg. 
1961
-
1963
)
14
Dupriez
 
B
Morel
 
P
Demory
 
JL
, et al. 
Prognostic factors in agnogenic myeloid metaplasia: a report on 195 cases with a new scoring system.
Blood
1996
, vol. 
88
 (pg. 
1013
-
1018
)
15
Cervantes
 
F
Barosi
 
G
Demory
 
JL
, et al. 
Myelofibrosis with myeloid metaplasia in young individuals: disease characteristics, prognostic factors and identification of risk groups.
Br J Haematol
1998
, vol. 
102
 (pg. 
684
-
690
)
16
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
, vol. 
18
 (pg. 
295
-
304
)
17
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
)
18
Kröger
 
N
Badbaran
 
A
Holler
 
E
, et al. 
Monitoring of the JAK2-V617F mutation by highly sensitive quantitative real-time PCR after allogeneic stem cell transplantation in patients with myelofibrosis.
Blood
2007
, vol. 
109
 
3
(pg. 
1316
-
1321
)
19
Patriarca
 
F
Bacigalupo
 
A
Sperotto
 
A
, et al. 
Allogeneic hematopoietic stem cell transplantation in myelofibrosis: the 20 year experience of the Gruppo Italiano Trapianto di Midollo Osseo (GITMO).
Haematologica
2008
, vol. 
93
 
10
(pg. 
1514
-
1522
)
20
Tefferi
 
A
Mesa
 
RA
Nagorney
 
DM
Schroeder
 
G
Silverstein
 
MN
Splenectomy in myelofibrosis with myeloid metaplasia: a single-institution experience with 223 patients.
Blood
2000
, vol. 
95
 
7
(pg. 
2226
-
2233
)
21
Kröger
 
N
Mesa
 
RA
Choosing between stem cell therapy and drugs in myelofibrosis.
Leukemia
2008
, vol. 
22
 
3
(pg. 
474
-
486
)
22
Pardanini
 
A
JAK2 inhibitor therapy in myeloproliferative disorders: rationale, preclinical studies and ongoing clinical trials.
Leukemia
2008
, vol. 
22
 
1
(pg. 
23
-
30
)
23
Koren-Michowitz
 
M
Shimoni
 
A
Vivante
 
A
, et al. 
A new MALDI-TOF-based assay for monitoring JAK2 V617F mutation level in patients undergoing allogeneic stem cell transplantation (all SCT) for classic myeloproliferative disorders (MPD).
Leuk Res
2008
, vol. 
32
 
3
(pg. 
421
-
427
)
Sign in via your Institution