Allogeneic hematopoietic stem cell transplantation (SCT) is well accepted as a curative treatment approach for younger patients with myelodysplastic syndrome (MDS) and has become one of the most frequent indications for allogeneic SCT as reported to the Center for International Blood and Marrow Transplant Research. However, MDS patients are usually elderly with a median age of approximately 75 years at diagnosis. Large register studies have confirmed the feasibility of the procedure in elderly MDS patients; and in the register of the European Group for Blood and Marrow Transplantation, one-third of the allogeneic transplant procedures for MDS were performed in 2010 in patients older than 60 years. Despite its curative potential, its role in the treatment of elderly MDS patients is less defined. Because of the inherent complications of the transplantation leading to treatment-related mortality and the risk of relapse, a careful calculation of the benefit for each patient is mandatory, taking into account comorbidities, disease status, donor selection, and effective nontransplant therapies. Prospective multicenter studies are needed to define optimal intensity of the conditioning regimen, timing of transplantation within a treatment algorithm, including drug-based therapies, and posttransplant strategies to reduce the risk of relapse.

Myelodysplastic syndrome (MDS) summarizes a heterogeneous group of hematologic diseases, which are characterized by a clonal abnormality of hematopoietic stem cells resulting in cytopenias, abnormal blasts, and risk of transformation into acute myeloid leukemia (AML). The clinical course of the disease varies from an indolent course over several years to a more rapid progression within months.1,2  MDS is predominantly a malignant disease of an elderly person, with a median age at diagnosis of approximately 75 years,3  and more than 80% are reported to be older than 60 years.4  Allogeneic stem cell transplantation (SCT) is considered to be a curative treatment option in MDS patients, and its role in treatment of “younger” patients with MDS is well established,5-12  even if the data relating outcomes are obtained mainly from retrospective studies. For many years, because of therapy-related morbidity and mortality, allogeneic SCT has been performed only in younger patients with MDS, whereas elderly patients who represent the majority of MDS patients have been excluded. It has to be pointed out that, in the transplant setting, patients older than 50 years are considered to be “elderly patients.” Furthermore, even when transplantation is performed on patients up to the age of 75 years, the majority of transplanted patients are still younger than the median age of MDS patients at diagnosis (∼ 75 years).

The introduction of toxicity-reduced, reduced-intensity conditioning (RIC) or nonmyeloablative conditioning regimens has resulted in a drastic reduction of transplant-related toxicity and mortality, leading to a rapidly growing number of transplantations in elderly patients with hematologic diseases.13-15  In the register of the European Group for Blood and Marrow Transplantation (EBMT), the numbers of allogeneic SCT for MDS/secondary acute myeloid leukemia have increased from 737 in 2001 to 1636 in 2010 (Figure 1). In parallel, the percentage of transplantation in MDS patients older than 50 years, older than 60 years, or older than 65 years increased from 47%, 10%, and 2%, respectively, in 2001 to 64%, 33%, and 14%, respectively, in 2010 (Figure 1). Despite the increase of literature reporting the feasibility of this approach in elderly MDS patients, prospective comparing trials are lacking; and despite the curative potential even in elderly patients, there is great need to define the population of elderly MDS patients who will benefit with a high probability of long-term freedom from disease versus the inherent complications of the transplantation procedure. It is also important to develop the timing of treatment sequence of allogeneic transplantation in the context of novel drugs, such as hypomethylating agents or immunomodulating drugs.

Figure 1

Development of allogeneic SCT for MDS/sAML in EBMT register according to age categories.

Figure 1

Development of allogeneic SCT for MDS/sAML in EBMT register according to age categories.

Close modal

The majority of the large retrospective trials consider a patient's age as a major prognostic factor for therapy-related mortality.7,8,16  In one of these studies, patient age is also associated with a high risk of relapse.16  These results were obtained mainly after standard myeloablative conditioning. Two recent large register trials address the specific issue of elderly MDS patients and allogeneic SCT (Table 1).

Table 1

Allogeneic SCT in elderly MDS patients

ReferenceNMedian age, y (range)ConditioningAge, yNRM, % (y)Relapse, % (y)DFS, % (y)OS, % (y)
Lim et al,17  EBMT (2010) 1333 56 (50-74) RIC: 833 (62%) — 36 (44) 36 (44) — 31 (4) 
  34% > 60 MA: 500 (38%) — 36 (50-60) 32 (50-60); P = .02 — 34 (50-60) 
  — — — 39 (> 60) 41 (> 60); P = .02 — 27 (> 60) 
McClune et al,18 * CIBMTR (2010) 1080 MDS: 40-78  40-54 25 (2) 33 (2) 42 (2) 44 (2) 
 MDS: n = 535 10% > 65 Only RIC or NMA (2 Gy TBI) 55-59 22 (2) 34 (2) 43 (2) 50 (2) 
 AML 1 CR: n = 545 AML: 40-79  60-64 32 (2) 37 (2) 31 (2) 34 (2) 
  12% > 65  ≥ 65 34 (2) 33 (2) 34 (2) 36 (2) 
ReferenceNMedian age, y (range)ConditioningAge, yNRM, % (y)Relapse, % (y)DFS, % (y)OS, % (y)
Lim et al,17  EBMT (2010) 1333 56 (50-74) RIC: 833 (62%) — 36 (44) 36 (44) — 31 (4) 
  34% > 60 MA: 500 (38%) — 36 (50-60) 32 (50-60); P = .02 — 34 (50-60) 
  — — — 39 (> 60) 41 (> 60); P = .02 — 27 (> 60) 
McClune et al,18 * CIBMTR (2010) 1080 MDS: 40-78  40-54 25 (2) 33 (2) 42 (2) 44 (2) 
 MDS: n = 535 10% > 65 Only RIC or NMA (2 Gy TBI) 55-59 22 (2) 34 (2) 43 (2) 50 (2) 
 AML 1 CR: n = 545 AML: 40-79  60-64 32 (2) 37 (2) 31 (2) 34 (2) 
  12% > 65  ≥ 65 34 (2) 33 (2) 34 (2) 36 (2) 

— indicates not applicable; MA, myeloablative; NMA, nonmyeloablative; and TBI, total body irradiation.

*

This study also includes AML patients.

The retrospective study of the of the EBMT reported on 1333 patients older than 50 years with MDS.17  Interestingly, despite advanced age, 38% of the patients received a standard myeloablative conditioning regimen. The patients were divided into age categories of 50 to 60 years and more than 60 years. The cumulative incidence of nonrelapse mortality (NRM) at 4 years was 36% for the 50- to 60-year group and 39% for the group 60 years or older (P = .39). The cumulative incidence of relapse at 4 years was 32% for the 50- to 60-year group and 41% for the group 60 years or older (P = .02), whereas overall survival (OS) at 4 years did not differ between the groups (34% vs 27%, P = .2). In a multivariate analysis for OS, only advanced stage of the disease at time of transplantation (hazard ratio [HR] = 1.55) was associated with inferior survival.

The Center for International Blood and Marrow Transplantation (CIBMTR) performed a similar study in 1080 elderly patients with MDS or AML in complete remission (CR) who received an RIC or nonmyeloablative conditioning regimen followed by allogeneic SCT. The patients were divided into 4 age categories: 40 to 54 years, 55 to 69 years, 60 to 65 years, and 65 years of age or older. There was a trend for a higher incidence of relapse and NRM in the group 60 to 65 years and 65 years of age or older compared with the 2 groups younger than 60 years (Table 1), but none of these differences reached statistical significance. However, a selection bias for the older transplant cohort has to be taken into account while interpreting the data. Disease-free survival (DFS) and OS did not differ between the age categories. In a multivariate analysis, age had no significant effect on OS, whereas low performance status, mismatched unrelated donors, and unfavorable cytogenetic were associated with inferior OS.18  It is controversial whether elderly MDS patients who undergo allogeneic SCT have a higher risk of GVHD. Whereas in some HLA-identical sibling transplantation studies recipient age was associated with an increased risk of graft versus host disease,19  in unrelated transplantation the National Marrow and Donor Program reported that only increased donor age was associated with a higher risk of severe acute GVHD.20  In sibling transplantation, donor age is closely linked to recipient age, and so it has been difficult to demonstrate an effect of age on GVHD incidence in matched sibling transplant. A more recent trial, including more than 1000 patients, did not show significant impact of age on occurrence of GVHD.18  However, if GVHD occurs, steroids are often less tolerated in older than in younger patients.

These 2 large retrospectives studies suggest that recipient age alone cannot be considered as contraindication for allogeneic SCT.

Because patient's age per se is not a major risk factor, other factors, such as comorbidities that by nature increase with increasing age, are taken into account. Comorbidity has a major impact on outcome of MDS patients in the nontransplant setting,21  and several scores have been developed for allogeneic SCT. The Hematopoietic Cell Transplantation Comorbidity Index (HCT-CI), developed by Sorror et al, has been demonstrated to be useful in patients with MDS or AML who underwent allogeneic SCT after RIC or standard conditioning regimen.22  If patients were stratified according to HCT-CI score and disease status, the risk of NRM increased with higher HCT-CI score and high-risk disease status. Patients with the highest HCT-CI score and also high-risk disease had the worst survival rate (OS at 2 years: 29%) independent of the intensity of the conditioning regimen because the lower NRM in the RIC-treated patients was offset by a higher risk of relapse.22  Other reports confirmed inferior survival in MDS patients with higher comorbidity scores after allogeneic SCT independent of the chronologic age of the patients.23,24 

Iron overload, which is not part of most comorbidity scores, may also influence toxicity and survival after transplantation and should be considered as comorbidity. Iron overload in MDS patients is caused by red blood cell transfusion and also by an increased gastrointestinal absorption of iron because of ineffective erythropoiesis.25  This iron is accumulated in macrophages in tissues, such as liver, spleen, and bone marrow.

Figure 2

SCT in elderly MDS patients.

Figure 2

SCT in elderly MDS patients.

Close modal

Once the capacity of plasma transferrin to bind iron is exhausted, nontransferrin-bound iron (NTBI) appears in blood and labile plasma iron as redox-active component of NTBI and is considered the most likely mediation of tissue damage and higher risk of infectious complications.26 

Iron overload is usually determined by serum ferritin in patients who undergo allogeneic SCT. An elevated serum ferritin of more than or equal to 1000 μg/L has been associated with a higher NRM and increased risk of infection after allogeneic SCT and myeloablative conditioning.27,28  The retrospective study by the Guppo Italiano Trapianto Midollo Osseo confirmed the impact of elevated serum ferritin on outcome of allogeneic transplantation but only after myeloablative conditioning and not after RIC.29  More appropriate methods for iron overload, such as liver or cardiac MRI, transfusion frequencies, and measurement of NTBI, and/or labile plasma iron, are needed to determine the impact of iron overload on outcome after SCT.30  Most of the transplant-related mortality resulting from elevated serum ferritin occurs within the first 3 months after SCT.27  It is very likely that the complications of iron overload, such as infections and organ toxicity, are the result of the release of NTBI and labile plasma iron induced by the cytotoxic conditioning regimen catalyzing reactive oxygen species. In our own study, we could show that, in patients who underwent allogeneic SCT after myeloablative conditioning regimen NTBI and transferring, saturation peaked early after starting conditioning therapy and remained elevated up to 2 weeks after initiation of the conditioning regimen.31  First attempts to use chelation, such as deferasirox, during conditioning regimen have shown dramatic reduction of labile plasma iron.32  Alternatively, administration of plasma apotransferrin has been shown to lower NTBI during SCT and restored the inhibitory effect on growth of Staphylococcus epidermidis in vitro.33 

The available data suggest a major impact of comorbidity on outcome after transplantation, but the most valuable comorbidity score needs to be determined and validated in larger cohorts of patients. Iron overload can be considered as comorbidity and is associated with a higher NRM. Prospective allogeneic SCT trials in MDS should include measurement of overload and determination of comorbidity indices. However, it is currently unclear how to treat iron overload in patients who undergo allogeneic SCT.

The exploration of RIC regimens resulted in less toxicity and has broadened the application of allogeneic SCT, especially in the cohort of elderly patients with hematologic malignancies.13,14  The rationale for RIC is to promote graft-versus-leukemia effect without excessive toxicity of the conditioning regimen, therefore resulting in less treatment-related mortality. The EBMT performed a retrospective comparing RIC with standard myeloablative conditioning in 836 patients with MDS who received stem cell grafts from HLA-identical siblings. The EBMT found a lower risk for treatment-related mortality (P = .015) but a significantly higher risk for relapse (P = .001) for the RIC transplanted patients, resulting in similar survival rates in both groups.34  The CIBMTR compared transplant outcome in more than 3000 patients with AML or MDS who received allogeneic SCT either after RIC or nonmyeloablative conditioning or after standard myeloablative conditioning. Those patients who received a RIC regimen had a higher risk of relapse and less probability of disease control compared with those who received a conventional ablative regimen.35  Other single-center retrospective studies have shown that a lower intensity of the conditioning regimen results in higher relapse rate and inferior outcome, especially in patients with more advanced disease.36,37  A more recent retrospective analysis from EBMT also suggests that the very low-intensity approach (2 Gy total body irradiation) has the highest risk of relapse in MDS and AML patients, and the treatment-related mortality after RIC transplantation has not reached a plateau after 1 year and differs only slightly at 5 years compared with myeloablative conditioning (Rodrigo Martino, oral presentation at EBMT meeting, Geneva, April 2012). All these studies have been performed retrospectively, and the inherent bias in selecting patients for a specific intensity of conditioning regimen should be considered because patient assignment to either RIC or myeloablative conditioning was based on physician or patient preference. An ongoing prospective randomized trial comparing RIC versus myeloablative conditioning from the MDS Subcommittee of the EBMT is addressing this issue (#NCT00682396).

However, in discussing intensity of conditioning regimen, it has to be pointed out that there is no clear cut-off between RIC and the myeloablative conditioning regimen. The major achievement of the new conditioning regimens is the reduced organ toxicity that broadens the application of allogeneic SCT to elderly patients. However, reducing organ toxicity does not exclude myeloablation or reduced antileukemic activity. For instance, targeting drug level or using intravenous formulation as part of the conditioning regimen has substantially reduced toxicity of busulfan; both have been shown to reduce transplant-related complications and improve outcome for MDS patients,38,39  allowing the use of this drug at myeloablative doses safely. Furthermore, cyclophosphamide as a toxic drug with only limited antileukemic activity can be substituted by fludarabine, inducing less organ toxicity without obvious loss of antileukemic activity.40  Other alkylating drugs, such as treosulfan, can also be used safely as part of a myeloablative conditioning regimen with low toxicity and NRM in MDS patients.41,42 

Even if results from prospective randomized trials are lacking, available literature suggests that patients up to 70 years of age can tolerate myeloablative conditioning regimen and that age per se should not be a criterion for selecting the intensity of the conditioning regimen rather than performance status/comorbidity and the status of the disease at time of transplantation. The current data also support that there is no “one-size-fits-all” conditioning regimen in elderly MDS patients, and the selection of regimen intensity has to become more individualized.

Cytogenetic abnormalities have an important impact on prognosis of patients undergoing allogeneic SCT for MDS. The established risk groups regarding cytogenetic abnormalities within the International Prognostic Scoring System (IPSS) have also major impact on outcome after allogeneic SCT.43,44 

The EBMT reported 692 patients with an OS of 47% for good-risk, 40% for intermediate-risk, and 31% for high-risk cytogenetic IPSS.44  The failure to transplant was mainly attributed to a higher risk of relapse, which was 34% for good-risk, 35% for intermediate-risk, and 57% for high-risk patients.44  Once patients with IPSS high-risk cytogenetics were analyzed in detail, more recent data from EBMT suggest that single abnormalities on chromosome 7 had significantly better survival than complex or monosomal karyotype.45  If outcome of transplantation was analyzed according to the new 5 cytogenetic risk groups proposed for MDS, a significant difference for the risk of relapse could be observed between the very-good-risk (8%), good-risk (17%), intermediate-risk (19%), poor-risk (26%), and the very-poor-risk cytogenetics (48%) groups.46  Improvement in outcome might be achieved using pretransplantation novel drugs, such as lenalidomide or hypomethylating agents, which have been shown to induce cytogenetic remission in patients with 5q− or monosomy 7 abnormalities.47,48 

Besides cytogenetic abnormalities, modern genomic technology, such as next-generation sequencing and mass spectrometry-based genotyping, allow the detection of molecular abnormalities, even in cytogenetically normal patients, such as mutations in TP53, EHZ2, ETV6, RUNX1, TET2, and ASXL1, which influence survival in a nontransplantation setting.49  Furthermore, newly discovered mutations of the splicing machinery have become the most frequent detectable mutations in MDS patients, which might also have an impact on outcome in a nontransplantation setting.50,51 

The EBMT study of allogeneic SCT in elderly MDS patients suggests that, in advanced stage, relapse is the most frequent cause of treatment failure.17  The number of blasts and not being in CR at time of transplantation are the most significant factors for inferior outcome, especially after RIC transplantation in MDS patients.7,16,34,37,52  Therefore, the issue of performing induction chemotherapy before transplantation has been a matter of debate since starting allogeneic transplantation in MDS patients. Unfortunately, the only randomized study from the EBMT had to be stopped because of slow recruitment, and as a result, no valid data are available. Smaller retrospective single-center studies showed no conclusive results,52-54  and it is probable that obtained results are biased by the selection of patients who received intensive chemotherapy before transplantation and those who did not achieve CR and never received a transplantation. Especially in the older populations, the intensive induction chemotherapy carries the risk of long-lasting myelosuppression and organ toxicities. To overcome the limitation of long-lasting myelosuppression but gain the antileukemic effect of pretransplantation induction chemotherapy, some centers have evaluated AML-like induction chemotherapies, such as anthracycline/cytosine, arabinoside/fludarabine, or clofarabine/cytosine arabinoside-based chemotherapy, followed by only 3 days' rest before performing a RIC and subsequent allogeneic SCT.55-57  With those conditioning regimens and matched related or unrelated donors, 2-year OS rates of 69% and 56% can be achieved.56,57 

Newer agents, such as 5-azacytidine or decitabine, which have been shown to be active in MDS,58,59  may also be used as pretransplantation cytoreductive therapy. However, the CR rate of approximately 10% is much lower than after conventional induction chemotherapy, and the reported trials confirmed the feasibility without significant survival benefit.60,61  Larger studies from EBMT and CIBMTR are currently under investigation regarding this issue.

The clinical course of MDS patients varies from several years to only a few months. Prognostic score systems, such as the IPSS.1  which includes number of blasts, cytopenias and cytogenetic, or the WHO prognostic scoring system, which includes also the transfusion dependency,62  may help to determine the prognosis of MDS patients at diagnosis or during the course of the disease.63  Although these scoring systems have been validated in a nontransplantation MDS cohort, both scoring systems have also been shown to be useful as a predictive model for outcome after allogeneic SCT.64,65  The arguments for early transplantation are based on low incidence of relapse. The Seattle group reported on outcome of unrelated SCT in MDS patients according to IPSS before transplantation. No relapse was seen in the IPSS low-risk group; whereas in the IPSS high-risk group, the relapse rate increases to 42%, resulting in an relapse-free survival of 80% in the low-risk group and only 29% among patients with an IPPS score higher than 2.16  A similar study was performed from EBMT in 374 patients with early disease defined as refractory anemia or refractory anemia with ring sideroblasts because IPSS scores were only available in half of the patients. The predictive factors for survival were recipient age, year of transplantation, and interval between diagnosis and transplantation. Earlier transplantation after diagnosis was associated with a 10% increase of survival rate at 4 years (57% vs 47%).66 

Whether these data support early transplantation in elderly MDS patients is questionable because no comparisons with nontransplantation approaches were made, and IPSS low-risk patients often do not require treatment. Furthermore, the risk of NRM should be carefully balanced; and even if NRM has decreased in recent years, the NRM is still considerably high and should be carefully balanced with a nontransplantation approach.67  Despite its curative potential, in the face of treatment-related mortality and the risk of relapse, one might ask whether elderly patients should be offered allogeneic SCT at all and whether conservative, less toxic approaches would be more suitable.

So far, no prospective trial has compared allogeneic SCT with nontransplantation approaches. In the past, the nontransplantation options were supportive without obvious prolongation of life. Hypomethylating agents, such as 5-azacytidine, have shown prolongation of survival in MDS patients compared with other nontransplantation therapies.58,59  If younger patients with MDS who received an HLA-identical sibling transplantation after standard myeloablative conditioning were compared with a nontransplantation cohort within a multistate model (Markov model), the obtained results suggest that IPSS high and intermediate II patients benefit from immediate transplantation, whereas intermediate I or low-risk patients benefit more from a delayed (in case of progression) transplantation.67 

The EBMT also recently used a multistate model to compare elderly (> 55 years) advanced MDS patients (RAEB or RAEB-t) who received allogeneic SCT and were reported to EBMT with a nontransplantation cohort from the Düsseldorf MDS registry. Here no benefit for allogeneic SCT could be found, but the analysis also showed the limitation of even sophisticated statistical analysis if patients from 2 different registries are compared and the selection of patients for transplant or nontransplantation is not clear.68  To address this important issue, the German MDS study group has started a prospective study in elderly (55-69 years) IPSS intermediate II or high-risk patients, comparing 5-azacytidine with a reduced-intensity allograft (#NCT01404741). Those studies should also include quality-of-life evaluation because any potential survival benefit should also be balanced with the gain or loss in quality of life.

From the clinical viewpoint, it is important to define optimal timing of allogeneic SCT in treatment algorithms, including hypomethylating and other effective drug-based agents, such as lenalidomide. For clinicians, it is reasonable to start with hypomethylating agents in elderly patients with intermediate II or high-risk IPSS while searching for a donor. But it remains unclear at which point of time the transplantation should be performed in responding patients: at time of best response or after progression? After failure to hypomethylating agents, the median survival is only 5.6 months. Long-term survival can then be achieved only by allogeneic SCT as salvage therapy (median survival, 19.5 months).69 

SCTs from unrelated donors have been increasingly used in recent years. In the EBMT registry, unrelated SCTs accounted for 59% of all allogeneic transplants for MDS in 2009. Although early studies on unrelated SCT in MDS reported a NRM of more than 40%,70  the incidence of NRM has been considerably and steadily improved in recent years and is comparable to HLA-identical sibling transplantation after adjusting for other risk factors (N.K., unpublished EBMT data, March 2011). In addition, the National Marrow Donor Program reported a relative risk for DFS of 1.43 (P = .03) for unrelated stem cell transplantations for MDS performed between 1988 and 1993 versus more recent transplantations.71 

In elderly patients with MDS, the available HLA-identical sibling is likely to have similar age as the recipient. Because in unrelated SCT increasing donor age is associated with inferior survival,20  the MDS subcommittee of EBMT investigated the influence of donor age in elderly MDS patients (> 50 years) who underwent allogeneic SCT. The question of the study was whether in elderly MDS patients a young unrelated donor would result in better outcome after allogeneic SCT than an HLA-identical sibling. A total of 719 patients with MDS and a median age of 58 years (range, 50-73 years) were included. The median age of the HLA-identical sibling donors was 56 years, and of the unrelated donors was 34 years. Although there was no influence of donor age in the HLA-identical sibling cohort, age of matched unrelated donor had significant influence on survival (HR = 1.03, P = .02), resulting in significantly improved survival in elderly MDS patients if the age of the unrelated donor was less than 30 years compared with HLA-identical sibling donors and matched unrelated donors older than 30 years: 40% versus 33% versus 24% (P = .04).

This retrospective study suggests that for elderly MDS patients, a young unrelated donor (< 30 years) is a more preferable donor than an HLA-identical sibling, but an HLA-identical donor should be preferred to an older matched unrelated donor.72 

Relapse has become the main reason for treatment failure after allogeneic SCT in MDS patients. Although the cumulative incidence of NRM has substantially decreased in recent years, no major advancement in reducing the risk of relapse has been made.73 

In the EBMT trial,17  the risk of relapse was significantly higher in patients 60 years of age or older compared with the age category 50 to 59 years (41% vs 32%, P = .02). This could be because those patients received more RIC regimen than the younger group but also because of biologic factors. Prevention of relapse after allogeneic SCT in MDS patients is therefore of even greater need in elderly patients. Because the treatment of relapse resulted in disappointing results, more effort has to be put into prevention of relapse. Limited experience of DLIs for relapse treatment or relapse prevention exists: in 2 studies, 14% to 22% achieved a CR, which lasted in 2 patients more than 5 years74,75  and prophylactic DLI improved relapse-free survival, but this has not been investigated in a randomized fashion and the results might be biased by patient selection.76 

5-azacytidine in combination with DLI has shown activity with 23% CR after allogenic SCT in relapsed AML/MDS patients.77  The MD Anderson Cancer Center investigated 5-azacytidine in a dose-finding study as maintenance therapy after transplantation in high-risk AML and MDS patients. They observed no severe side effects and no impact of azacytidine on GVHD and chimerism. The recommended dose is 24 mg/m2 5 times and has currently been tested in a randomized trial.78,79  Potential effects of hypomethylating agents after transplantation are the increase expression on cancer-testis antigen, which may become the target for donor cells to induce a tumor-specific immune response.80,81  However, other in vitro studies show immunosuppressive properties of 5-azacytidine, which, in contrast, may abrogate effective immune response by increasing T-regulatory cells.82 

To avoid overtreatment by prophylactic post-transplantation strategies, such as DLI or hypomethylating agents, a more optimized way would be therapeutic intervention based on detectable minimal molecular residual disease after transplantation, as it has been shown for CML, MPN, or AML.83 

Monitoring CD34+ lineage-specific chimerism might be able to detect residual disease or early relapse and might allow successful intervention with 5-azacitidine.84  The more recent discovery of mutation in MDS patients,49,85  by modern sequencing techniques, allows for screening patients before transplantation for specific mutations and can be used after transplantation as a marker for residual disease and as a guide for adoptive immunotherapies to prevent clinical relapse.

In conclusion, allogeneic SCT is well accepted as curative treatment approach in patients with MDS and has become the third most frequent indication for allogeneic SCT as reported to the CIBMTR.86  The majority of MDS patients are elderly, with a median age of 65 to 70 years at diagnosis. However, the role of SCT in elderly patients with MDS is not well defined. Large retrospective studies confirmed the feasibility of allogeneic SCT in elderly MDS patients as well as curative potential, but benefit in terms of survival has not been demonstrated yet in randomized trials. Chronologic age, which has been used for a long time as exclusion criteria for allogeneic SCT, is probably only a poor predictor for survival because of the lack of data on organ dysfunction and performance status. Besides chronologic age, physical function and organ comorbidities should be taken into account for selecting patients for transplantation. The intensity of the conditioning regimen is not well established, but retrospective studies suggest that there is no “one-size-fits-all” conditioning regimen for elderly MDS patients, and comorbidities and risk of relapse are the main determinants for selecting the intensity of the conditioning regimen in a more “individualized” fashion. Allogeneic SCT should early be included in the treatment plan for MDS patients, which includes novel agents, such as hypomethylating agents or immunomodulatory drugs to achieve cytogenetic response or/and reduction of the number of blasts prior transplantation. Molecular marker detectable with new sequencing methods should be investigated at the time of diagnosis and used as residual disease marker after transplantation to guide either drug-based or immunologic based therapies, such as donor lymphocyte infusion, alloreactive NK cells, or vaccination strategies to lower the risk of relapse and improve OS.

The author thanks the staff of the Department of Stem Cell Transplantation at the University Medical Center Hamburg for providing excellent care of the patients, Karen Klemt for editing the manuscript, and the members of the MDS subcommittee of the Chronic Leukemia Working Party of EBMT, especially Theo de Witte for fruitful discussion and study proposals to improve transplant results in MDS patients.

Contribution: N.K. wrote the manuscript.

Conflict-of-interest disclosure: The author declares no competing financial interests.

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

1
Greenberg
 
P
Cox
 
C
LeBeau
 
MM
, et al. 
International scoring system for evaluating prognosis in myelodysplastic syndromes.
Blood
1997
, vol. 
89
 
6
(pg. 
2079
-
2088
)
2
Nimer
 
S
Myelodysplastic syndromes.
Blood
2008
, vol. 
111
 
10
(pg. 
4841
-
4851
)
3
Swerdlow
 
SH
Campo
 
E
Harris
 
NL
, et al. 
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
2008
Lyon, France
IARC Press
4
Ma
 
X
Does
 
M
Raza
 
A
, et al. 
Myelodysplastic syndromes: incidence and survival in the United States.
Cancer
2007
, vol. 
109
 
8
(pg. 
1536
-
1542
)
5
Oliansky
 
DM
Antin
 
JH
Bennett
 
JM
, et al. 
The role of cytotoxic therapy with hematopoietic stem cell transplantation in the therapy of myelodysplastic syndromes: an evidence-based review.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
2
(pg. 
137
-
172
)
6
Appelbaum
 
FR
Storb
 
R
Ramberg
 
RE
, et al. 
Treatment of preleukemia syndromes with marrow transplantation.
Blood
1987
, vol. 
69
 
1
(pg. 
92
-
96
)
7
de Witte
 
T
Hermans
 
J
Vossen
 
J
, et al. 
Haematopoietic stem cell transplantation for patients with myelodysplastic syndromes and secondary acute myeloid leukemia: a report on behalf of the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT).
Br J Haematol
2000
, vol. 
110
 
3
(pg. 
620
-
630
)
8
Sierra
 
J
Pérez
 
WS
Rozman
 
C
, et al. 
Bone marrow transplantation from HLA-identical siblings as treatment for myelodysplasia.
Blood
2002
, vol. 
100
 
6
(pg. 
1997
-
2004
)
9
Sutton
 
L
Chastang
 
C
Ribaud
 
P
, et al. 
Factors influencing outcome in de-novo myelodysplastic syndromes treated by allogeneic bone marrow transplantation: a long term study of 71 patients. Societé Francaise de Greffe de Moelle.
Blood
1996
, vol. 
88
 
1
(pg. 
358
-
365
)
10
Anderson
 
J
Anasetti
 
C
Appelbaum
 
F
, et al. 
Unrelated donor bone marrow transplantation for myelodysplasia (MDS) and MDS-related acute myeloid leukemia.
Br J Haematol
1996
, vol. 
93
 
1
(pg. 
59
-
67
)
11
Oostervald
 
M
Suciu
 
S
Verhoef
 
G
, et al. 
The presence of an HLA-identical sibling donor has no impact on outcomes of patients with high risk MDS or secondary AML (sAML) treated with intensive chemotherapy followed by transplantation: results of a prospective study of the EORTC, EBMT, SAKK, and GIMEMA Leukemia groups (EORTC 06921).
Leukemia
2003
, vol. 
17
 
5
(pg. 
859
-
868
)
12
Deeg
 
HJ
Shulman
 
HM
Anderson
 
JE
, et al. 
Allogeneic and syngeneic marrow transplantation for myelodysplastic syndrome in patients 55 to 65 years of age.
Blood
2000
, vol. 
95
 
4
(pg. 
1188
-
1194
)
13
Slavin
 
S
Nagler
 
A
Naperstek
 
E
, et al. 
Non-myeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and non-malignant hematologic diseases.
Blood
1998
, vol. 
91
 
3
(pg. 
756
-
763
)
14
Giralt
 
S
Estey
 
E
Albitar
 
M
, et al. 
Engraftment of allogeneic hematopoietic progenitor cells with purine analog containing chemotherapy: harnessing graft-versus-leukemia without myeloablative therapy.
Blood
1997
, vol. 
89
 
12
(pg. 
4531
-
4536
)
15
Laport
 
GG
Sandmaier
 
BM
Storer
 
BE
, et al. 
Reduced-intensity conditioning followed by allogeneic hematopoietic cell transplantation for adult patients with myelodysplastic syndrome and myeloproliferative disorders.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
2
(pg. 
246
-
255
)
16
Deeg
 
HJ
Storer
 
B
Slattery
 
JT
, et al. 
Conditioning with targeted busulfan and cyclophosphamide for hemopoietic stem cell transplantation from related and unrelated donors in patients with myelodysplastic syndrome.
Blood
2002
, vol. 
100
 
4
(pg. 
1201
-
1207
)
17
Lim
 
Z
Brand
 
R
Martino
 
R
, et al. 
Allogeneic hematopoietic stem cell transplantation for patients 50 years or older with myelodysplastic syndromes or secondary acute myeloid leukemia.
J Clin Oncol
2010
, vol. 
28
 
3
(pg. 
405
-
411
)
18
McClune
 
BL
Weisdorf
 
DJ
Pedersen
 
TL
, et al. 
Effect of age on outcome of reduced-intensity hematopoietic cell transplantation for older patients with acute myeloid leukemia in first complete remission or with myelodysplastic syndrome.
J Clin Oncol
2010
, vol. 
28
 
11
(pg. 
1878
-
1887
)
19
Weisdorf
 
D
Hakke
 
R
Blazar
 
B
, et al. 
Risk factors for acute graft-versus-host disease in histocompatible donor bone marrow transplantation.
Transplantation
1991
, vol. 
51
 
6
(pg. 
1197
-
1203
)
20
Kollmann
 
C
Howe
 
CW
Anasetti
 
C
, et al. 
Donor characteristics as risk factors in recipients after transplantation of bone marrow from unrelated donors: the effect of donor age.
Blood
2001
, vol. 
98
 
7
(pg. 
2043
-
2051
)
21
Zipperer
 
E
Pelz
 
D
Nachtkamp
 
K
, et al. 
The hematopoietic stem cell transplantation comorbidity index is of prognostic relevance for patients with myelodysplastic syndrome.
Haematologica
2009
, vol. 
94
 
5
(pg. 
729
-
732
)
22
Sorror
 
ML
Sandmaier
 
BM
Storer
 
BE
, et al. 
Comorbidity and disease status based risk stratification of outcomes among patients with acute myeloid leukemia or myelodysplasia receiving allogeneic hematopoietic cell transplantation.
J Clin Oncol
2007
, vol. 
25
 
27
(pg. 
4246
-
4254
)
23
Lim
 
ZY
Ingram
 
W
Brand
 
R
, et al. 
Impact of pretransplant comorbidities on alemtuzumab-based reduced-intensity conditioning allogeneic hematopoietic SCT for patients with high-risk myelodysplastic syndrome and AML.
Bone Marrow Transplant
2010
, vol. 
45
 
4
(pg. 
633
-
639
)
24
Boehm
 
A
Sperr
 
WR
Leitner
 
G
, et al. 
Comorbidity predicts survival in myelodysplastic syndromes or secondary acute myeloid leukemia after allogeneic stem cell transplantation.
Eur J Clin Invest
2008
, vol. 
38
 
12
(pg. 
945
-
952
)
25
Cortelezzi
 
A
Cattaneo
 
C
Christiani
 
S
, et al. 
Non-transferrin-bound iron in myelodysplastic syndromes: a marker of ineffective erythropoiesis?
Hematol J
2000
, vol. 
1
 
3
(pg. 
153
-
158
)
26
Pullarkat
 
V
Objectives of iron chelation therapy in myelodysplastic syndromes: more than meets the eye?
Blood
2009
, vol. 
114
 
26
(pg. 
5251
-
5255
)
27
Armand
 
P
Kim
 
HT
Cutler
 
CS
, et al. 
Prognostic impact of elevated pretransplantation serum ferritinin patients undergoing myeloablative stem cell transplantation.
Blood
2007
, vol. 
109
 
10
(pg. 
4586
-
4588
)
28
Pullarkat
 
V
Blanchard
 
S
Tegtmeier
 
B
, et al. 
Iron overload adversely affects outcome of allogeneic hematopoietic cell transplantation.
Bone Marrow Transplant
2008
, vol. 
42
 
12
(pg. 
799
-
805
)
29
Alessandrino
 
EP
Della Porta
 
MG
Bacigalupo
 
A
, et al. 
Prognostic impact of pre-transplantation transfusion history and secondary iron overload in patients with myelodysplastic syndrome undergoing allogeneic stem cell transplantation: a GITMO study.
Haematologica
2010
, vol. 
95
 
3
(pg. 
476
-
484
)
30
Armand
 
P
Kim
 
HT
Rhodes
 
J
, et al. 
Iron overload in patients with acute leukemia or MDS undergoing myeloablative stem cell transplantation.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
6
(pg. 
852
-
860
)
31
Dürken
 
M
Nielsen
 
P
Knobel
 
S
, et al. 
Nontransferrin-bound iron in serum of patients receiving bone marrow transplants.
Free Radic Biol Med
1997
, vol. 
22
 
7
(pg. 
1159
-
1163
)
32
Fritsch
 
A
Langebrake
 
C
Nielsen
 
P
, et al. 
Deferasirox (Exjade) given during conditioning regimen (FLAMSA/Busulfan/ATG) reduces the appearance of labile plasma iron in patients undergoing allogeneic stem cell transplantation [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
 
Abstract 3023
33
Sahlstedt
 
L
von Bornsdorf
 
L
Ebeling
 
F
, et al. 
Effective binding of free iron by a single intravenous dose of human apotransferrin in haematological stem cell transplant patients.
Br J Haematol
2002
, vol. 
119
 
2
(pg. 
547
-
553
)
34
Martino
 
R
Iacobelli
 
S
Brand
 
R
, et al. 
Myelodysplastic Syndrome Subcommittee of the Chronic Leukemia Working Party of the European Blood and Marrow Transplantation Group: retrospective comparison of reduced-intensity conditioning and conventional high-dose conditioning for allogeneic hematopoietic stem cell transplantation using HLA-identical sibling donors in myelodysplastic syndromes.
Blood
2006
, vol. 
108
 
3
(pg. 
836
-
846
)
35
Luger
 
S
Ringden
 
O
Zhang
 
MJ
, et al. 
Similar outcomes using myeloablative vs reduced-intensity allogeneic transplant preparative regimens for AML or MDS.
Bone Marrow Transplant
2012
, vol. 
47
 
2
(pg. 
203
-
211
)
36
de Lima
 
M
Anagnostopoulos
 
A
Munsell
 
M
, et al. 
Nonablative versus reduced-intensity conditioning regimens in the treatment of acute myeloid leukemia and high-risk myelodysplastic syndrome: dose is relevant for long-term disease control after allogeneic hematopoietic stem cell transplantation.
Blood
2004
, vol. 
104
 
3
(pg. 
865
-
872
)
37
Shimoni
 
A
Hardan
 
I
Shem-Tov
 
N
, et al. 
Allogeneic hematopoietic stem cell transplantation in AML and MDS using myeloablative versus reduced-intensity conditioning: the role of dose intensity.
Leukemia
2006
, vol. 
20
 
2
(pg. 
322
-
328
)
38
Andersson
 
BS
Kashyap
 
A
Gian
 
V
, et al. 
Conditioning therapy with intravenous busulfan and cyclophosphamide (IV BuCy2) for hematologic malignancies prior to allogeneic stem cell transplantation: a phase II study.
Biol Blood Marrow Transplant
2002
, vol. 
8
 
3
(pg. 
145
-
154
)
39
Russell
 
JA
Tran
 
HAT
Quinlan
 
D
, et al. 
Once daily intravenous busulfan given with fludarabine as conditioning for allogeneic stem cell transplantation study of pharmacokinetics and early clinical outcomes.
Biol Blood Marrow Transplant
2002
, vol. 
8
 
9
(pg. 
468
-
476
)
40
de Lima
 
M
Couriel
 
D
Thall
 
PF
, et al. 
Once daily intravenous busulfan and fludarabine: clinical and pharmacokinetic results of a myeloablative reduced-toxicity conditioning regimen for allogeneic stem cell in AML and MDS.
Blood
2004
, vol. 
104
 
3
(pg. 
857
-
864
)
41
Ruutu
 
T
Volin
 
L
Beelen
 
DW
, et al. 
Reduced-toxicity conditioning with treosulfan and fludarabine in allogeneic hematopoietic stem cell transplantation for myelodysplastic syndromes: final results of an international prospective phase II trial.
Haematologica
2011
, vol. 
96
 
9
(pg. 
1344
-
1350
)
42
Kröger
 
N
Shimoni
 
A
Zabelina
 
T
, et al. 
Reduced-toxicity conditioning with treosulfan, fludarabine, and ATG as preparative regimen for allogeneic stem cell transplantation (alloSCT) in elderly patients with secondary acute myeloid leukemia (sAML) or myelodysplastic syndrome (MDS).
Bone Marrow Transplant
2006
, vol. 
37
 
4
(pg. 
339
-
344
)
43
Nevill
 
TJ
Shepherd
 
JD
Sutherland
 
HJ
, et al. 
IPSS poor-risk karyotype as a predictor of outcome for patients with myelodysplastic syndrome following myeloablative stem cell transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
2
(pg. 
205
-
213
)
44
Onida
 
F
Brand
 
R
van Biezen
 
A
, et al. 
Impact of cytogenetics on outcome of patients with MDS or secondary AML undergoing allogeneic HSCT from HLA-identical siblings: a retrospective analysis of the EBMT-CLWP [abstract].
Blood (ASH Annual Meeting Abstracts)
2006
, vol. 
108
  
Abstract 2653
45
Van Gelder
 
M
Schetelig
 
J
Volin
 
Liisa
, et al. 
Monosomal karyotype predicts poor outcome for MDS/sAML patients with chromosome 7 abnormalities after allogeneic stem cell transplantation for MDS/sAML: a study of the MDS subcommittee of the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT) [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
 
Abstract 293
46
Deeg
 
HJ
Scott
 
BL
Appelbaum
 
FR
Gooley
 
T
Impact of novel 5-group cytogenetic risk classification of MDS on outcome after allogeneic hematopoietic cell transplantation (HCT) [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
  
Abstract 666
47
Platzbecker
 
U
Mohr
 
B
von Bonin
 
M
, et al. 
Lenalidomide as induction therapy before allogenic stem cell transplantation in a patient with proliferative CMML-2 and del(5q) not involving ERG1 locus.
Leukemia
2007
, vol. 
21
 (pg. 
2384
-
2385
)
48
Rüter
 
B
Wijermans
 
P
Claus
 
R
, et al. 
Preferential cytogenetic response to continuous intravenous low-dose decitabine (DAC) administration in myelodysplastic syndrome with monosomy 7.
Blood
2007
, vol. 
110
 (pg. 
1080
-
1082
)
49
Bejar
 
R
Stevenson
 
K
Abdel-Wahab
 
O
, et al. 
Clinical effect of point mutations in myelodysplastic syndromes.
N Engl J Med
2011
, vol. 
364
 
26
(pg. 
2496
-
2506
)
50
Yoshida
 
K
Sanada
 
M
Shiraishi
 
Y
, et al. 
Frequent pathway mutations of splicing machinery in myelodysplasia.
Nature
2011
, vol. 
478
 (pg. 
64
-
69
)
51
Malcovati
 
L
Papaemmanuil
 
E
Bowen
 
DT
, et al. 
Clinical significance of SF3B mutations in myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms.
Blood
2011
, vol. 
118
 (pg. 
6239
-
6246
)
52
Nakai
 
K
Kanda
 
Y
Fukuhara
 
S
, et al. 
Value of chemotherapy before allogeneic hematopoietic stem cell transplantation from HLA-identical sibling donor for myelodysplastic syndrome.
Leukemia
2005
, vol. 
19
 
3
(pg. 
396
-
401
)
53
Scott
 
BL
Storer
 
B
Loken
 
MR
, et al. 
Pre-transplantation induction chemotherapy and post-transplantation relapse in patients with advanced myelodysplastic syndrome.
Biol Blood Marrow Transplant
2005
, vol. 
11
 
1
(pg. 
65
-
73
)
54
Oran
 
B
Giralt
 
S
Saliba
 
R
, et al. 
Allogeneic hematopoietic stem cell transplantation for the treatment of high-risk myelogenous leukemia and myelodysplastic syndrome using reduced-intensity conditioning with fludarabine and melphalan.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
4
(pg. 
454
-
462
)
55
Schmid
 
C
Schleuning
 
M
Ledderose
 
G
, et al. 
Sequential regimen of chemotherapy, reduced-intensity conditioning for allogeneic stem cell transplantation, and prophylactic donor lymphocyte transfusion in high-risk acute myeloid leukemia and myelodysplastic syndrome.
J Clin Oncol
2005
, vol. 
23
 
24
(pg. 
5675
-
5687
)
56
Kröger
 
N
Zabelina
 
T
Wolschke
 
C
, et al. 
Induction chemotherapy followed immediately by busulfan-based reduced conditioning and allografting in elderly patients with advanced MDS or sAML [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
  
Abstract 3387
57
Buchholz
 
S
Dammann
 
E
Stadler
 
M
, et al. 
Cytoreductive treatment with clofarabine/ara-C combined with reduced-intensity conditioning and allogeneic stem cell transplantation in patients with high-risk, relapsed, or refractory acute myeloid leukemia and advanced myelodysplastic syndrome.
Eur J Haematol
2012
, vol. 
88
 
1
(pg. 
52
-
60
)
58
Silverman
 
LR
Demakos
 
EP
Peterson
 
BL
, et al. 
Randomized controlled trial of azacitidine in patients with myelodysplastic syndrome: a study of the Cancer and Leukemia Group B.
J Clin Oncol
2002
, vol. 
20
 
10
(pg. 
2429
-
2440
)
59
Fenaux
 
P
Mufti
 
GJ
Hellstrom-Lindberg
 
E
, et al. 
Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomized, open-label, phase III study.
Lancet Oncol
2009
, vol. 
10
 
3
(pg. 
223
-
232
)
60
de Padua
 
SL
de Lima
 
M
Kantarjian
 
H
, et al. 
Feasibility of allo SCT after hypomethylating therapy with decitabine for myelodysplastic syndrome.
Bone Marrow Transplant
2009
, vol. 
43
 
11
(pg. 
839
-
843
)
61
Field
 
T
Perkins
 
J
Huang
 
Y
, et al. 
5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation.
Bone Marrow Transplant
2010
, vol. 
45
 
2
(pg. 
255
-
260
)
62
Malcovati
 
L
Porta
 
MG
Pascutto
 
C
, et al. 
Prognostic factors and life expectancy in myelodysplastic syndromes classified according to WHO criteria: a basis for clinical decision making.
J Clin Oncol
2005
, vol. 
23
 
30
(pg. 
7594
-
7603
)
63
Malcovati
 
L
Germing
 
U
Kuendgen
 
A
, et al. 
Time-dependent prognostic scoring system for predicting survival and leukemic evolution in myelodysplastic syndromes.
J Clin Oncol
2007
, vol. 
25
 
23
(pg. 
3503
-
3510
)
64
Alessandrino
 
EP
Della Porta
 
MG
Bacigalupo
 
A
, et al. 
WHO classification and WPSS predict posttransplantation outcome in patients with myelodysplastic syndrome: a study from the Gruppo Italiano Trapianto di Modollo Osseo (GITMO).
Blood
2008
, vol. 
112
 
3
(pg. 
895
-
902
)
65
Appelbaum
 
FR
Anderson
 
J
Allogeneic bone marrow transplantation for myelodysplastic syndromes: outcomes analysis according to IPSS scores.
Leukemia
1998
, vol. 
12
 
suppl 1
(pg. 
S25
-
S29
)
66
de Witte
 
T
Brand
 
R
van Biezen
 
A
, et al. 
Allogeneic stem cell transplantation for patients with refractory anaemia with matched related and unrelated donors: delay of the transplant is associated with inferior survival.
Br J Haematol
2009
, vol. 
146
 
6
(pg. 
627
-
636
)
67
Cutler
 
CS
Lee
 
SJ
Greenberg
 
P
, et al. 
A decision analysis of allogeneic bone marrow transplantation for the myelodysplastic syndromes: delayed transplantation for low-risk myelodysplasia is associated with improved outcome.
Blood
2004
, vol. 
104
 
2
(pg. 
579
-
585
)
68
Kröger
 
N
Putter
 
H
Brand
 
R
, et al. 
Comparison of allogeneic stem cell transplantation and best supportive care in elderly patients with advanced MDS [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
  
Abstract 3363
69
Prébet
 
T
Gore
 
SD
Esterni
 
B
, et al. 
Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure.
J Clin Oncol
2011
, vol. 
29
 
24
(pg. 
3322
-
3327
)
70
Arnold
 
R
de Witte
 
T
van Biezen
 
A
, et al. 
Unrelated bone marrow transplantation in patients with myelodysplastic syndromes and secondary acute myeloid leukemia: an EBMT survey.
Bone Marrow Transplant
1998
, vol. 
21
 
12
(pg. 
1213
-
1216
)
71
Castro-Malaspina
 
H
Harris
 
RE
Gajewski
 
J
, et al. 
Unrelated donor marrow transplantation for myelodysplastic syndromes: outcome analysis in 510 transplants facilitated by the National Marrow Donor Program.
Blood
2002
, vol. 
99
 
6
(pg. 
1943
-
1951
)
72
Kröger
 
N
Zabelina
 
T
van Biezen
 
A
, et al. 
Donor selection for allogeneic stem cell transplantation in elderly patients with advanced MDS: younger matched-unrelated donor or HLA-identical sibling [abstract]?
Blood (ASH Annual Meeting Abstracts)
2010
, vol. 
116
  
Abstract 912
73
Bishop
 
MR
Alyea
 
EP
Cairo
 
MS
, et al. 
National Cancer Institute's First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: summary and recommendations from the organizing committee.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
4
(pg. 
443
-
454
)
74
Campregher
 
PV
Gooley
 
T
Scott
 
BL
, et al. 
Results of donor lymphocyte infusions for relapsed myelodysplastic syndrome after hematopoietic cell transplantation.
Bone Marrow Transplant
2007
, vol. 
40
 
10
(pg. 
965
-
971
)
75
Depil
 
S
Deconinck
 
E
Milpied
 
N
, et al. 
Donor lymphocyte infusion to treat relapse after allogeneic bone marrow transplantation for myelodysplastic syndrome.
Bone Marrow Transplant
2004
, vol. 
33
 
5
(pg. 
531
-
534
)
76
Kolb
 
HJ
Schmid
 
C
Tischer
 
J
, et al. 
Allogeneic stem cell transplantation for MDS and sAML following reduced-intensity conditioning and preemptive donor lymphocyte transfusion [abstract].
Blood (ASH Annual Meeting Abstracts)
2006
, vol. 
108
  
Abstract 324
77
Schroeder
 
T
Czibere
 
A
Kröger
 
N
, et al. 
Phase II study of azacitidine (Vidaza, Aza) and donor lymphocyte infusions (DLI) as first salvage therapy in patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS) relapsing after allogeneic hematopoietic stem cell transplantation (allo-SCT): final results from AZARELA-trial (NCT-00795548) [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
 
Abstract 656
78
Jabbour
 
E
Giralt
 
S
Kantarjian
 
H
, et al. 
Low-dose azacitidine after allogeneic stem cell transplantation for acute leukemia.
Cancer
2009
, vol. 
115
 
9
(pg. 
1899
-
1905
)
79
de Lima
 
M
Giralt
 
S
Thall
 
PF
, et al. 
Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogeneous leukemia or myelodysplastic syndrome: a dose and schedule finding study.
Cancer
2010
, vol. 
116
 
23
(pg. 
5420
-
5431
)
80
Atanackovich
 
D
Luetkens
 
T
Kloth
 
B
, et al. 
Cancer-testis antigen expression and its epigenetic modulation in acute myeloid leukemia.
Am J Hematol
2011
, vol. 
86
 
11
(pg. 
918
-
922
)
81
Goodyear
 
O
Agathanggelou
 
A
Novitzky-Basso
 
I
, et al. 
Induction of a CD8+ T-cell response to MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia.
Blood
2010
, vol. 
116
 
11
(pg. 
1908
-
1918
)
82
Stübig
 
T
Badbaran
 
A
Wolschke
 
C
, et al. 
Immune-modulation of CD4+ and CD8+ cells by 5-azacitidine [abstract].
Haematologica
2010
, vol. 
95
 
suppl 2
pg. 
308
  
Abstract 0738
83
Kröger
 
N
Bacher
 
U
Bader
 
P
, et al. 
NCI first international workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: report from the committee on disease-specific methods and strategies for monitoring relapse following allogeneic stem cell transplantation: II. Chronic leukemias, myeloproliferative neoplasms, and lymphoid malignancies.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
10
(pg. 
1325
-
1346
)
84
Platzbecker
 
U
Wermke
 
M
Radke
 
J
, et al. 
Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial.
Leukemia
2012
, vol. 
26
 
3
(pg. 
381
-
389
)
85
Thol
 
F
Friesen
 
I
Damm
 
F
, et al. 
Prognostic significance of ASXL1 mutations with myelodysplastic syndromes.
J Clin Oncol
2011
, vol. 
29
 
18
(pg. 
2499
-
2506
)
86
Center for International Blood & Marrow Transplant Research
Report on state of the art in blood and marrow transplantation: 2009; Slide 9.
Accessed 2009 
Sign in via your Institution