Postremission therapy in patients with acute myeloid leukemia (AML) may consist of continuing chemotherapy or transplantation using either autologous or allogeneic stem cells. Patients with favorable subtypes of AML generally receive chemotherapeutic consolidation, although recent studies have also suggested favorable outcome after hematopoietic stem cell transplantation (HSCT). Although allogeneic HSCT (alloHSCT) is considered the preferred type of postremission therapy in poor- and very-poor-risk AML, the place of alloHSCT in intermediate-risk AML is being debated, and autologous HSCT is considered a valuable alternative that may be preferred in patients without minimal residual disease after induction chemotherapy. Here, we review postremission transplantation strategies using either autologous or allogeneic stem cells. Recent developments in the field of alternative donors, including cord blood and haploidentical donors, are highlighted, and we discuss reduced-intensity alloHSCT in older AML recipients who represent the predominant category of patients with AML who have a high risk of relapse in first remission.

Although the majority of patients with acute myeloid leukemia (AML) enter remission upon induction chemotherapy, the risk of relapse is considerable. That risk varies greatly according to age and genetic subtype as major denominators.1-3  Taking an increasingly detailed risk evaluation into account, the intensity and type of postremission therapy is generally tailored according to risk profile, whereby chemotherapeutic consolidation is favored in good-risk AML and allogeneic hematopoietic stem cell transplantation (alloHSCT) is favored in poor-risk AML.1,4,5  These approaches are currently weighed and debated in intermediate-risk AML, and interest in autologous HSCT (autoHSCT) has recently been revived.6 

The last decade has witnessed an enormous increase in knowledge of the pathogenesis of AML. Genetic characterization has yielded important prognostic information, which has necessitated taking the most important mutational aberrations into account for therapeutic decision making.1,7  Furthermore, translational research has now established AML as a multiclonal disease that develops stepwise according a Darwinian model of leukemogenesis.8  The latter insight has important consequences for therapy, because it became unlikely that targeted monotherapies aimed at a single AML oncogene could match the efficacy of the tyrosine kinase inhibitors in monoclonal malignancies (eg, chronic myeloid leukemia). Indeed, current therapeutic developments aim to incorporate targeted therapy into established intensive chemotherapeutic schedules.9,10  In addition, the advent of new treatment modalities such as chimeric antigen receptor T cells and various monoclonal antibodies has renewed interest in immunotherapeutic approaches for treatment of multiclonal leukemias such as AML and acute lymphoblastic leukemia.11,12  A particularly potent immunotherapeutic approach is the allogeneic graft-versus-leukemia (GVL) effect, which has been shown to exert its influence across all genomic subsets with similar relative potency.13  However, its inherent toxicity is increasingly being debated, which makes using transplantation protocols associated with an acceptable limited toxicity profile a greater necessity. Conversely, the urge to preserve and exploit the GVL effect is an important issue, especially in the poor-risk genetic subtypes of AML. Here, we address 3 important issues in the field of postremission therapy by alloHSCT or autoHSCT in first complete response (CR1) in patients with AML: What is the current place of reduced-intensity conditioning (RIC) and nonmyeloablative (NMA) alloHSCT in both older and younger patients with AML in CR1? Which type of alternative donor is currently preferred and what is the current status of haploidentical donor transplantation in AML? Is autoHSCT a valuable alternative for alloHSCT in intermediate-risk AML?

Following the observation that both organ toxicity and severe graft-versus-host disease (GVHD) are related to the intensity of the conditioning regimen,14  it was hypothesized that reducing the intensity may limit these complications, while still allowing a sufficient GVL effect.15,16  Nearly 20 years later, it is clear that the hypothesis has been proved by showing reduced non-relapse mortality (NRM) and persistent GVL effects.17  GVL effects after RIC appear to depend on several factors, including a stronger reduction of relapse in patients experiencing chronic GVHD (cGVHD) and higher relapse risk after more stringent T-cell depletion or T-cell inhibition.18-20  As the first and major consequence, the development of RIC alloHSCT has enabled the use of alloHSCT in older and medically infirm patients. It represents a major change because this population is at greater risk for complications and has the poorest prognosis.3,21  That poor prognosis is mainly related to the underlying mutational profile22  and ineligibility to qualify for and benefit from intensive chemotherapy.23  Early attempts have confirmed the feasibility of RIC alloHSCT in older recipients with AML,24,25  which has resulted in a worldwide increase in alloHSCT activity.26  Although recent reviews27-29  have highlighted the development of RIC alloHSCTs and weighed their efficacy, several questions remain.

Is there an optimal conditioning dose intensity that allows for a well-balanced antileukemic effect and limited toxicity?

By definition, myeloablative conditioning (MAC) regimens do not allow for autologous recovery and require stem cell support whereas NMA regimens do not require stem cell support. Regimens that do not fit these criteria can then be classified as RIC, whereby the dose of total body irradiation or the alkylating agent is usually reduced by at least 30% compared with an ablative regimen.30  The NMA conditioning regimen developed by Storb et al consists of low-dose total body irradiation and fludarabine, which effectively allows for engraftment and appears to be associated with limited early mortality.16  However, antileukemic activity may be compromised, notably in advanced or high-risk diseases in which higher rates of relapse have been observed.31  In a prospective trial not limited to AML, it was reported that such an NMA regimen is indeed associated with low NRM but at the expense of a somewhat higher relapse rate, eventually resulting in the same outcome as that in patients treated with more intensive conditioning.32  Other centers developed RIC regimens based on the combination of alkylating agents with a purine analog28,33-35  followed by attempts to increase antileukemic activity by increasing the dose of the alkylating drug.36  Overall, such RIC regimens were suggested to be associated with lower relapse rates compared with NMA regimens, but prospective randomized studies are lacking. Most retrospective comparisons are hampered by patient selection and also by incomplete genetic characterization of the underlying AML as the most important prognostic parameter. To address the question of whether an increased dose of busulfan as part of RIC may be associated with better leukemia-free survival, a French cooperative group has set out to prospectively compare 3 different dosages of busulfan when combined with fludarabine prior to HLA-identical sibling alloHSCT (NCT01985061). Meanwhile, the experience of the local team and the number of transplantations being performed annually also have an impact on results.37  Thus, it is very difficult to favor one regimen over many others at this time, although an NMA regimen may not be preferred in more advanced AML.

Do anti-T-cell antibodies in the conditioning regimen allow for more effective GVHD prophylaxis without compromising GVL?

To effectively prevent GVHD and especially cGVHD in the elderly, some teams have proposed adding additional immunosuppression that incorporates antithymocyte immunoglobulins (ATGs) or alemtuzumab into the conditioning regimen. Finke et al38  reported a lower incidence of acute GVHD (aGVHD) and cGVHD using ATG before myeloablative unrelated donor (UD) alloHSCT. However, a survival benefit could not be demonstrated in that study or in a meta-analysis of several prospective studies.39  No prospective comparative study evaluated these agents in RIC alloHSCT, and registry data (Center for International Blood and Marrow Transplant Research [CIBMTR]; European Society for Blood and Marrow Transplantation [EBMT]) have not precisely defined the place for ATG.40,41  However, in the context of RIC using an alkylating agent (busulfan) and fludarabine, an intermediate ATG dose (5 mg/kg total dose) was suggested to be associated with effective prevention of cGVHD while preserving GVL effects.42,43  In contrast, it was suggested that a higher dose of ATG blunted the GVL effect and resulted in a high rate of opportunistic infections as a result of more stringent T-cell depletion.44  Whether such a strategy applies to both matched unrelated donors and sibling donors is currently unknown. In a recent phase 2 study that included patients older than age 55 years conditioned with fludarabine, intravenous busulfan, and 2 days of ATG before sibling alloHSCT, an acceptable NRM of 9% and a favorable quality of life was reported, whereas relapse appeared to be limited.45  More stringent in vivo T-cell depletion may be achieved by using alemtuzumab, which has been used extensively in the United Kingdom. A recent study of AML patients age 35 to 60 years suggested a low NRM and very effective prevention of GVHD in those who received matched sibling transplants, but results in recipients of UD grafts were less favorable, which might be explained by a higher incidence of opportunistic infections.46  Altogether these studies suggested that GVHD may be controlled in a more effective way by incorporating ATG or alemtuzumab, but the comparative value of these approaches with conventional prevention of GVHD is still unclear because randomized studies are lacking. Finally, the strategy of posttransplant immunosuppression may be profoundly changed by the introduction of posttransplant high-dose cyclophosphamide (PT-HDCy), which was developed in haploidentical alloHSCT but is currently being explored after matched sibling alloHSCT.47 

Age and comorbidities

Considering the significant impact of comorbidities, one may wonder whether age itself or whether age-associated comorbidities have a significant impact on risk for NRM. Several retrospective studies addressed that question, but an independent impact of age was often absent.45,48-51  Those studies suggested no definite age limit, but they noted that the assessment of comorbidities was an essential part of the workup for every patient for whom alloHSCT was considered.52  In addition, the use of a geriatric evaluation in patients older than age 60 to 65 years is also important. Recently, Muffly et al53  have shown that the combined evaluation of geriatric and comorbidity status has a discriminative impact on outcome. Younger patients (younger than age 60 years) would benefit from conditioning regimen with a more acceptable toxicity profile. Several meta-analyses of prospective trials that used a donor vs no-donor comparison have established that donor availability may be associated with a strong reduction of relapse, which is generally compromised by NRM in patients older than age 40 years.54,55  All patients who have received transplants in the latter studies received standard MAC, combining either full-dose total body irradiation and an alkylating agent or 2 alkylating agents at a myeloablative dosage. Because of increased NRM in patients older than age 40 years and the favorable experience in older patients with RIC alloHSCT, several cooperative groups introduced RIC alloHSCT for patients in the 40 to 60 years age category. Two recent retrospective studies suggested better outcome following transplantation in patients older than age 40 years compared with chemotherapy because of the reduced NRM and preserved GVL effects.46,56  These studies suggest that in the context of intensified and optimized induction chemotherapy, a RIC regimen prior to alloHSCT might offer an attractive approach in AML patients in CR1.

Recommendations for RIC alloHSCT in AML patients in CR1

Given the paucity of prospective randomized studies, one needs to be cautious about making firm recommendations. But it seems reasonable to state that RIC alloHSCT is an established type of postremission therapy in elderly patients with AML and may offer a strong antileukemic effect.1,2,57-59  However, the use of alloHSCT would need to be tailored according to risk for NRM and morbidity,51,60  whereby patients at higher risk for NRM might qualify for NMA conditioning or, alternatively, an autograft61  or chemotherapy.2  Weighing risk factors might be done as earlier suggested by the European Leukemia Net5  and as presented in Table 1. Risk factors and their individual or composite characteristics will evolve, but the need to discuss their impact on relapse and NRM continues. Although studies that evaluate alloHSCT by donor availability or by using transplantation as a time-dependent covariate provide the main basis for decision making,59,62  the opposite effects of reduced relapse and increased NRM limit the precise applicability of those studies for individual patients. Therefore, we recommend weighing these effects by taking into account their projected, personalized risks for an individual patient, according to the European Leukemia Net model presented in Table 1. The place of RIC alloHSCT in patients younger than age 60 years is less clear. A recent meta-analysis comparing RIC and MAC regimens did not reveal any significant differences in terms of overall survival (OS) or progression-free survival, although RIC recipients were older and had more advanced disease.63  The prospective randomized study by Bornhäuser et al64  yielded a similar picture, although the RIC regimen came close to myeloablation, and the study was prematurely closed. Meanwhile, results of the prospective randomized study comparing RIC and MAC that was performed in the United States (BMT CTN 0901) are eagerly awaited.

Table 1

Recommendation for alloHSCT in AML CR1 based on integrated risk profiles

AML risk group‡AML risk assessment criteria at diagnosisMRD after cycle 2Risk of relapse following consolidation approachPrognostic scores for NRM that indicate alloHSCT as preferred consolidation
Chemotherapy or autoHSCT (%)AlloHSCT (%)EBMT score52 HCT-CI score53 NRM risk (%)
Good –t(8;21) or AML1-ETO, WBC <20 Positive or negative 35-40 15-20 NA (≤1) NA (<1) 10-15 
–inv16/t(16;16) or CBFB-MYH11 
–CEBPA-biallelic mutant-positive 
–FLT3-ITD-negative/NMP1-positive 
Intermediate –CN –X –Y, WBC <100, CRe Negative 50-55 20-25 ≤2 ≤2 <20-25 
–t(8;21) or AML1-ETO plus WBC >20 
or mutant KIT 
Poor –CN –X –Y, WBC <100, CRe Positive 70-80 30-40 ≤3-4 ≤3-4 <30 
–t(8;21) or AML1-ETO, WBC >20 Positive 
and/or mutant KIT  
–CN –X –Y, WBC <100, no CRe Negative 
–CN –X –Y, WBC >100 Negative 
–CA, but non-CBF, MK-negative, no abn3q26  
Very poor –CN –X –Y, WBC >100 Positive >90 40-50 ≤5 ≤5 <40 
–CA, but non-CBF, MK-negative, no abn3q26, EVI1-negative Positive 
–MK-positive Positive or negative 
–abn3q26  
–Non-CBF, EVI1-positive  
–Non-CBF with mutant p53, or –mutant RUNX1, or mutant ASXL1 –or biallelic FLT3-ITD with –FLT3-ITD:FLT3 WT ratio of >0.6  
AML risk group‡AML risk assessment criteria at diagnosisMRD after cycle 2Risk of relapse following consolidation approachPrognostic scores for NRM that indicate alloHSCT as preferred consolidation
Chemotherapy or autoHSCT (%)AlloHSCT (%)EBMT score52 HCT-CI score53 NRM risk (%)
Good –t(8;21) or AML1-ETO, WBC <20 Positive or negative 35-40 15-20 NA (≤1) NA (<1) 10-15 
–inv16/t(16;16) or CBFB-MYH11 
–CEBPA-biallelic mutant-positive 
–FLT3-ITD-negative/NMP1-positive 
Intermediate –CN –X –Y, WBC <100, CRe Negative 50-55 20-25 ≤2 ≤2 <20-25 
–t(8;21) or AML1-ETO plus WBC >20 
or mutant KIT 
Poor –CN –X –Y, WBC <100, CRe Positive 70-80 30-40 ≤3-4 ≤3-4 <30 
–t(8;21) or AML1-ETO, WBC >20 Positive 
and/or mutant KIT  
–CN –X –Y, WBC <100, no CRe Negative 
–CN –X –Y, WBC >100 Negative 
–CA, but non-CBF, MK-negative, no abn3q26  
Very poor –CN –X –Y, WBC >100 Positive >90 40-50 ≤5 ≤5 <40 
–CA, but non-CBF, MK-negative, no abn3q26, EVI1-negative Positive 
–MK-positive Positive or negative 
–abn3q26  
–Non-CBF, EVI1-positive  
–Non-CBF with mutant p53, or –mutant RUNX1, or mutant ASXL1 –or biallelic FLT3-ITD with –FLT3-ITD:FLT3 WT ratio of >0.6  

Adapted from the European Leukemia Net recommendation by adding new molecular markers and MRD.5  The proposed patient-specific use of alloHSCT in AML CR1 integrates the individual risks for relapse and NRM and aims for a disease-free survival benefit of at least 10% for the individual patient compared with consolidation by a non-alloHSCT approach. The categorization of AML is based on cytogenetic, molecular, and clinical parameters (including white blood cell count [WBC]); subcategories are now designated good, intermediate, poor, and very poor, as currently used by the HOVON-SAKK Cooperative Consortium.

CA, cytogentic abnormalities; CBF, core binding factor; CN, cytogenetically normal; CRe, early complete remission; HCT-CI, hematopoietic cell transplantation comorbidity index; ITD, internal tandem duplication; MK, monosomal karyotype; NA, not applicable; –X –Y, deleted X or Y chromosome.

It seems fair to state that patients with poor-risk AML primarily qualify for alloHSCT, whereas patients at higher risk for NRM might be offered a RIC regimen before transplantation. The option of alloHSCT in patients with intermediate-risk AML would need to be weighed, with autoHSCT as an alternative option (discussed in “Autologous transplantation”). The subset of patients with very-poor-risk AML needs special consideration. Although the immunologic GVL effect is operational in patients with very-poor-risk AMLs,13  outcomes with alloHSCT are poor, and experimental approaches that explore ways to exploit the GVL effect in those patients are urgently needed. Such approaches include timing of transplantation, new combinations of conditioning after preceding chemotherapy, and development of new immunotherapeutic and chemotherapeutic approaches after transplantation.65,66  In that respect, it is especially encouraging to note that new immunotherapeutic approaches are being developed to be used either pre- or posttransplant.11,12,67  Overall, recommendations apply to transplants performed with matched sibling and well-matched UDs, because these donor types have been demonstrated to result in fairly similar outcomes.68  However, the last decade has witnessed an enormous development in alternative stem cell donors or stem cell sources, which are also currently used in RIC and NMA alloHSCT and therefore need to be discussed separately.

Adult UDs and umbilical cord blood

The lack of potential stem cell donors has long been a major limitation for using alloHSCT. Fortunately, recent advances have greatly expanded the pools of potential adult volunteer donors and stem cell donors. First, international registries of adult stem cell donors have expanded to approximately 25 million donors. In addition, the use of high-resolution typing has allowed selection of UDs who are matched for HLA-A, -B, -C, -DRB1, and -DQB1 at the molecular level for an increasing percentage of patients.69  Results with respect to OS and disease-free survival with well-matched donors currently compare well with those obtained with sibling donors.70-74  Few clinical trials have prospectively addressed this question, but nearly 30 000 patients have been compared in different studies that overall show similar results.73  Second, after initial experience in pediatric patients, umbilical cord blood (UCB) transplantation was extended to adult patients. Although both MAC and RIC regimens are used,75  the RIC regimen developed by the transplantation team of the University of Minnesota combines strong immunosuppression, antileukemic activity, and an acceptable NRM.76,77  The incidence of graft failure, however, may still be higher in pediatric patients, and cell dose was identified as a pivotal parameter.78  These results initiated attempts to increase the cell dose, including double UCB transplantation79  and ex vivo expansion.80  Overall, when a minimal cell dose is used, survival appears to be similar to that in HSCT using adult UDs. Indeed, in a large retrospective study from the CIBMTR and EBMT with more than 1500 patients, UCB transplantation was associated with higher NRM but lower relapse and GVHD rates, resulting in similar leukemia-free survival.81  The results of that study and others established UCB transplantation as an alternative stem cell source for patients lacking an HLA-matched sibling donor. Results with double UCB transplantation suggested that the use of two cord blood (CB) donors may be associated with less graft failure and also a somewhat stronger GVL effect.82  With that background, a randomized trial was performed that compared one-unit vs two-unit UCB transplantation in young patients, including those with myeloid malignancies.83  All patients were to receive a minimum of 2.5 nucleated cells per kilogram. Survival rates were similar after one-unit and two-unit UCB transplantations. However, a one-unit CB transplantation appeared to be associated with a lower risk of GVHD, suggesting that a single unit may be preferred if the unit contains sufficient nucleated cells. Mature studies addressing the value of expanded UCB in AML patients are currently lacking.

Haploidentical family donors

More recently, T-cell repleted haploidentical transplantation has become a third player in the field. Interest in haploidentical HSCT started in the mid 1990s with the transplantation team of the University of Perugia.84  However, their sophisticated approach using a highly myeloablative regimen and extensive in vitro and ex vivo T-cell depletion appeared to be associated with impaired immune reconstitution, compromising both the GVL effect and anti-infectious immunity.85  More recently, T-cell repleted haploidentical HSCT was developed based on different immunosuppressive approaches varying from pretransplant ATG to PT-HDCy. Initial reports of T-cell repleted haploidentical HSCT showed high rates of engraftment, acceptable rates of severe aGVHD and NRM, and low incidences of severe cGVHD.86-88  Subsequently, many reports have confirmed these initial findings, although development varied worldwide.89-94  Collectively, the experience successfully challenged the earlier notion that haploidentical HSCT would generally be associated with a high NRM. Some cooperative groups or registries76,95-97  have already addressed the question of which alternative donor is to be preferred.98-100  Overall, these reports suggest similar results between alternative donor or stem cell sources (Table 2), but prospective studies are scarce. The US clinical trial network is conducting a prospective study comparing PT-HDCy haploidentical HSCT and CB transplantation. Because the preferred alternative donor is currently unknown, it is of utmost importance to include as many patients as possible in prospective studies. Finally, it should be noted that the number of studies specifically reporting on patients with AML is very limited (Table 2). However, Wang et al101  recently reported results from a prospective multicenter trial that compared matched sibling alloHSCT (n = 219) with haploidentical HSCT (n = 231) (Table 2). The results suggested that outcome after haploidentical alloHSCT may be comparable to survival after alloHSCT using a matched sibling donor for patients with AML in CR. However, more studies with long-term follow-up and molecular characterization of the underlying AML are needed to better define the place of each type of alternative donor and to develop more detailed guidelines. An increase in transplantation activity may be expected in the years to come, because alternative donors have now become available for nearly every individual patient in need of alloHSCT. However, a number of questions remain, precluding the precise formulation of guidelines, donor hierarchy, and preferred conditioning regimen.

Table 2

Recent studies of alloHSCT using haploidentical donors in AML

First author, year of study, study group, type of studyAML patients characteristicsOutcome, by donor typeStudy conclusions
Age at diagnosis (y) (range)AML in CR1 (%)Median follow-up, moConditioningStem cell sourceGVHD preventionDonor type and No.NRMRelapseLFS
Ciceri, 2008, retrospective, multicentric96  36 (16-66) 14 47 Myeloablative, 100% PBSC Ex vivo T-cell depletion, 100%; ATG, 89% Haploidentical, 86 0.36 0.16 0.48 The choice between haploidentical and UCB transplantation may be based on center expertise, policy, costs of the procedures, and the availability of clinical trials. 
    
    
Ruggeri, 2015 EBMT, retrospective, multicentric95  45 (18-72) 34 24 Myeloablative, 61% BM; PBSC PT-HDCy, 32% UCB, 558 0.30 0.32 0.38 Cumulative incidence of relapse was not different between the 2 groups; adjusted LFS and OS were comparable. 
Haploidentical, 360 0.27 0.41 0.32 
    
Wang, 2015, prospective, multicentric101  28 (15-57) 100 32 Myeloablative, 100% BM + PBSC ATG, 100% MSD, 219 0.08 0.15 0.78 This comparison suggests that outcome after haploidentical HSCT (with ATG) is comparable to matched sibling alloHSCT. 
Haploidentical, 231 0.13 0.15 0.74 
Ciurea, 2015, IBMTR, retrospective, multicentric97  57 (21-70) 47 30-39 Myeloablative, 54% BM PT-HDCy, 100% MAC MUD, 1245; 0.20 0.39 0.42 These data suggest that OS after haploidentical HSCT with PT-HDCy is comparable to MUD alloHSCT. 
Haploidentical, 104 0.14 0.44 0.41 
RIC MUD, 737 0.23 0.42 0.37 
Haploidentical, 88 0.09 0.58 0.35 
First author, year of study, study group, type of studyAML patients characteristicsOutcome, by donor typeStudy conclusions
Age at diagnosis (y) (range)AML in CR1 (%)Median follow-up, moConditioningStem cell sourceGVHD preventionDonor type and No.NRMRelapseLFS
Ciceri, 2008, retrospective, multicentric96  36 (16-66) 14 47 Myeloablative, 100% PBSC Ex vivo T-cell depletion, 100%; ATG, 89% Haploidentical, 86 0.36 0.16 0.48 The choice between haploidentical and UCB transplantation may be based on center expertise, policy, costs of the procedures, and the availability of clinical trials. 
    
    
Ruggeri, 2015 EBMT, retrospective, multicentric95  45 (18-72) 34 24 Myeloablative, 61% BM; PBSC PT-HDCy, 32% UCB, 558 0.30 0.32 0.38 Cumulative incidence of relapse was not different between the 2 groups; adjusted LFS and OS were comparable. 
Haploidentical, 360 0.27 0.41 0.32 
    
Wang, 2015, prospective, multicentric101  28 (15-57) 100 32 Myeloablative, 100% BM + PBSC ATG, 100% MSD, 219 0.08 0.15 0.78 This comparison suggests that outcome after haploidentical HSCT (with ATG) is comparable to matched sibling alloHSCT. 
Haploidentical, 231 0.13 0.15 0.74 
Ciurea, 2015, IBMTR, retrospective, multicentric97  57 (21-70) 47 30-39 Myeloablative, 54% BM PT-HDCy, 100% MAC MUD, 1245; 0.20 0.39 0.42 These data suggest that OS after haploidentical HSCT with PT-HDCy is comparable to MUD alloHSCT. 
Haploidentical, 104 0.14 0.44 0.41 
RIC MUD, 737 0.23 0.42 0.37 
Haploidentical, 88 0.09 0.58 0.35 

BM, bone marrow; LFS, leukemia-free survival; MSD, matched sibling donor; MUD, matched unrelated donor; UCB, unrelated cord blood.

Although alloHSCT is the preferred type of postremission therapy in poor- and very-poor-risk AML, the place of alloHSCT in intermediate-risk AML is being debated, and chemotherapy or autoHSCT may also offer long-term survival, although the risk of relapse may be higher than that after alloHSCT. Chemotherapeutic postremission therapy seems to be favored in the United States, but several European collaborative groups continue to use autoHSCT, especially since the introduction of peripheral blood stem cells (PBSCs), which have been shown to improve hematologic recovery and thereby treatment-related morbidity and mortality.

Results with PBSCs

Many phase 2 and retrospective studies have been reported, but few prospective randomized studies are available. The study performed by the Dutch-Belgian Hemato-Oncology Cooperative Group/Swiss Group for Clinical Cancer Research (HOVON-SAKK) Cooperative Consortium included 517 patients who were randomly assigned between 1995 and 2006.102  With a mature follow-up of more than 5 years, the actual rates of relapse after chemotherapy vs after autoHSCT were 70% vs 58%, respectively (P = .02). The study showed a nonsignificant difference in relapse-free survival (RFS) of 29% vs 38% (P = .065). OS did not differ between the 2 groups and was estimated to be 41% vs 44%, respectively, at 5 years from randomization. Salvage treatment by alloHSCT was more frequently performed in relapsing chemotherapy recipients compared with autograft recipients (25% vs 17%). Of note, those percentages seem rather low given the current availability of alternative donors and the increased use of RIC. Therefore, the HOVON-SAKK Cooperative Consortium more recently re-analyzed the use of alloHSCT in relapsing autograft patients and showed that the rate of salvage alloHSCT had increased to approximately 30%.56  As a result, a retrospective comparison of patients age 40 to 60 years suggested better OS after autoHSCT in intermediate-risk patients as a result of improved salvage possibilities.56  Furthermore, outcome after alloHSCT or autoHSCT no longer differed in terms of OS, whereas alloHSCT after either MAC or RIC still appeared to be associated with better RFS (Figure 1). These results compare well to results from a retrospective study by the CIBMTR that suggested similar outcome for younger AML patients in CR1 receiving either alloHSCT from an HLA-identical sibling or an autograft using PBSCs.103  Although recipients of alloHSCT exhibited more high-risk features, had longer follow-up, and experienced a lower risk of treatment failure, no significant difference in OS was noted. The EBMT noted similar observations in older AML patients. Herr et al61  reported that RIC alloHSCT was similar to postremission therapy with autologous PBSC transplantation in terms of OS and RFS if it was used in CR1. Of note, a lower relapse rate of 36% vs 50% was counterbalanced by increased NRM in alloHSCT recipients. Moreover, autoHSCT may be used in patients up to age 70 years with an acceptable NRM of approximately 8%, which compares favorably to 17% as was observed after RIC alloHSCT.61  The conditioning regimen for autoHSCT was based on busulfan in 85% of patients. Although both oral and intravenous busulfan were used in that study,61  it has become clear that the intravenous administration of busulfan should be preferred because of fewer complications and higher reproducibility of levels that were aimed for.35  Given the higher NRM and nonrelapse toxicity (primarily GVHD and its consequences) associated with RIC alloHSCT, these results compel us to redefine the place of RIC alloHSCT and autoHSCT, especially in intermediate-risk AML patients in CR1 who harbor a risk of relapse that may vary between 40% and 50%.

Figure 1

Kaplan-Meier estimates of overall survival of AML intermediate-risk patients in CR1, age 40 to 60 years, by type of postremission therapy (updated results from Cornelissen et al53). HSCT recipients showed significantly better OS than patients receiving chemotherapeutic postremission therapy (P = .001). AlloMAB, myeloablative alloHSCT; AlloRIC, reduced intensity conditioning alloHSCT; Auto, autologous HSCT; CT, chemotherapy; ELN, European Leukemia Net. F, female; LR, logistic regression.

Figure 1

Kaplan-Meier estimates of overall survival of AML intermediate-risk patients in CR1, age 40 to 60 years, by type of postremission therapy (updated results from Cornelissen et al53). HSCT recipients showed significantly better OS than patients receiving chemotherapeutic postremission therapy (P = .001). AlloMAB, myeloablative alloHSCT; AlloRIC, reduced intensity conditioning alloHSCT; Auto, autologous HSCT; CT, chemotherapy; ELN, European Leukemia Net. F, female; LR, logistic regression.

Close modal

Prognostic parameters

Several studies have suggested using dedicated risk scores for the application of autoHSCT. For example Pfirrmann et al104  developed a composite risk score based on age, CD34+ blast count, Flt3 mutant:wild-type ratio, cytogenetic risk, and secondary origin of AML. Three groups could be discriminated. AutoHSCT appeared to be associated with better survival compared with alloHSCT in their newly defined intermediate risk group. Although the principle of using a risk score in intermediate-risk patients is attractive, disadvantages include the poor reproducibility of parameters such as CD34 blast count and Flt3 mutant:wild-type ratio. In addition, the recent study by the HOVON-SAKK Cooperative Consortium could not reproduce the predictive power of the Pffirmann score in AML patients age 40 to 60 years in CR1 for whom all parameters were available.56  Alternatively, decision-making might benefit from taking minimal residual disease (MRD) into account. Following a 3- to 4-decade-long development, it is now well established that the assessment of MRD after induction and/or consolidation therapy in AML by either multiparameter flow cytometry or by quantitative polymerase chain reaction for specific molecular markers significantly predicts subsequent outcome.105-109  Moreover, MRD has been shown to predict outcome after different modes of postremission therapy such as continued chemotherapy, autoHSCT, or alloHSCT. As a consequence, several collaborative groups such as the Italian Gruppo Italiano Malattie Ematologiche dell’ Adulto (GIMEMA) and the HOVON-SAKK Cooperative Consortium have now incorporated MRD as a decisive parameter for choice of postremission therapy type in intermediate-risk AML patients upon achieving CR1 (Table 1; www.HOVON.nl), whereby MRD-negative patients may be consolidated by autoHSCT and MRD-positive patients may proceed to alloHSCT. The preferred type of postremission therapy in intermediate-risk AML is not definitely settled and continues to evolve. Currently, the choice may depend on multiple parameters, including the risk score of the underlying leukemia, the response to chemotherapy as assessed by morphology and MRD markers, and the projected risks associated with type of transplantation. However, alloHSCT is increasingly used selectively in poor- and very-poor-risk patients, whereas autoHSCT is generating new interest, especially in intermediate-risk patients who became MRD negative upon induction chemotherapy. Good-risk AML patients qualify for chemotherapeutic consolidation, but a recent report by Schlenk et al110  suggested favorable outcome for good-risk patients with autoHSCT, which provides a possible option in that category of patients. Importantly, new developments in the field of immunotherapy11,12,111  that will be evaluated in the context of either autoHSCT or alloHSCT are currently being pursued. Hopefully, they will yield better prospects for older patients with AML and help us to redefine the preferred type of postremission therapy and possibly also type of immunotherapeutic maintenance after transplantation.

Contribution: J.J.C. and D.B. contributed equally to outlining this review, formulating questions, searching for and selecting literature, and writing and final approval of the manuscript.

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

Correspondence: Jan J. Cornelissen, Erasmus MC Cancer Institute/Erasmus University Medical Center, Groene Hilledijk 301, Rotterdam, 3075 EA, The Netherlands; e-mail: j.cornelissen@erasmusmc.nl.

1
Estey
 
EH
Acute myeloid leukemia: 2014 update on risk-stratification and management.
Am J Hematol
2014
, vol. 
89
 
11
(pg. 
1063
-
1081
)
2
Ossenkoppele
 
G
Lowenberg
 
B
How I treat the older patient with acute myeloid leukemia.
Blood
2015
, vol. 
125
 
5
(pg. 
767
-
774
)
3
Burnett
 
A
Wetzler
 
M
Lowenberg
 
B
Therapeutic advances in acute myeloid leukemia.
J Clin Oncol
2011
, vol. 
29
 
5
(pg. 
487
-
494
)
4
Dohner
 
H
Estey
 
EH
Amadori
 
S
, et al. 
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet.
Blood
2010
, vol. 
115
 
3
(pg. 
453
-
474
)
5
Cornelissen
 
JJ
Gratwohl
 
A
Schlenk
 
RF
, et al. 
The European LeukemiaNet AML Working Party consensus statement on allogeneic HSCT for patients with AML in remission: an integrated-risk adapted approach.
Nat Rev Clin Oncol
2012
, vol. 
9
 
10
(pg. 
579
-
590
)
6
Ferrara
 
F
Renaissance of autologous stem cell transplantation for AML?
Lancet Oncol
2012
, vol. 
13
 
2
(pg. 
121
-
123
)
7
Sanders
 
MA
Valk
 
PJ
The evolving molecular genetic landscape in acute myeloid leukaemia.
Curr Opin Hematol
2013
, vol. 
20
 
2
(pg. 
79
-
85
)
8
Grove
 
CS
Vassiliou
 
GS
Acute myeloid leukaemia: a paradigm for the clonal evolution of cancer?
Dis Model Mech
2014
, vol. 
7
 
8
(pg. 
941
-
951
)
9
Engen
 
CB
Wergeland
 
L
Skavland
 
J
Gjertsen
 
BT
Targeted Therapy of FLT3 in Treatment of AML-Current Status and Future Directions.
J Clin Med
2014
, vol. 
3
 
4
(pg. 
1466
-
1489
)
10
Fielding
 
AK
Treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia in adults: a broader range of options, improved outcomes, and more therapeutic dilemmas.
Am Soc Clin Oncol Educ Book
2015
, vol. 
35
 (pg. 
e352
-
e359
)
11
Pegram
 
HJ
Smith
 
EL
Rafiq
 
S
Brentjens
 
RJ
CAR therapy for hematological cancers: can success seen in the treatment of B-cell acute lymphoblastic leukemia be applied to other hematological malignancies?
Immunotherapy
2015
, vol. 
7
 
5
(pg. 
545
-
561
)
12
Topp
 
MS
Gokbuget
 
N
Stein
 
AS
, et al. 
Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study.
Lancet Oncol
2015
, vol. 
16
 
1
(pg. 
57
-
66
)
13
Cornelissen
 
JJ
Breems
 
D
van Putten
 
WL
, et al. 
Comparative analysis of the value of allogeneic hematopoietic stem-cell transplantation in acute myeloid leukemia with monosomal karyotype versus other cytogenetic risk categories.
J Clin Oncol
2012
, vol. 
30
 
17
(pg. 
2140
-
2146
)
14
Holler
 
E
Kolb
 
HJ
Moller
 
A
, et al. 
Increased serum levels of tumor necrosis factor alpha precede major complications of bone marrow transplantation.
Blood
1990
, vol. 
75
 
4
(pg. 
1011
-
1016
)
15
Nagler
 
A
Slavin
 
S
Varadi
 
G
Naparstek
 
E
Samuel
 
S
Or
 
R
Allogeneic peripheral blood stem cell transplantation using a fludarabine-based low intensity conditioning regimen for malignant lymphoma.
Bone Marrow Transplant
2000
, vol. 
25
 
10
(pg. 
1021
-
1028
)
16
McSweeney
 
PA
Niederwieser
 
D
Shizuru
 
JA
, et al. 
Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects.
Blood
2001
, vol. 
97
 
11
(pg. 
3390
-
3400
)
17
Blaise
 
D
Castagna
 
L
 
Do different conditioning regimens really make a difference? Hematology Am Soc Hematol Educ Program. 2012;2012:237-245
18
Weisdorf
 
D
Zhang
 
MJ
Arora
 
M
Horowitz
 
MM
Rizzo
 
JD
Eapen
 
M
Graft-versus-host disease induced graft-versus-leukemia effect: greater impact on relapse and disease-free survival after reduced intensity conditioning.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
11
(pg. 
1727
-
1733
)
19
Storb
 
R
Gyurkocza
 
B
Storer
 
BE
, et al. 
Graft-versus-host disease and graft-versus-tumor effects after allogeneic hematopoietic cell transplantation.
J Clin Oncol
2013
, vol. 
31
 
12
(pg. 
1530
-
1538
)
20
Craddock
 
C
Nagra
 
S
Peniket
 
A
, et al. 
Factors predicting long-term survival after T-cell depleted reduced intensity allogeneic stem cell transplantation for acute myeloid leukemia.
Haematologica
2010
, vol. 
95
 
6
(pg. 
989
-
995
)
21
Hayat
 
MJ
Howlader
 
N
Reichman
 
ME
Edwards
 
BK
Cancer statistics, trends, and multiple primary cancer analyses from the Surveillance, Epidemiology, and End Results (SEER) Program.
Oncologist
2007
, vol. 
12
 
1
(pg. 
20
-
37
)
22
Kantarjian
 
H
O’Brien
 
S
Cortes
 
J
, et al. 
Results of intensive chemotherapy in 998 patients age 65 years or older with acute myeloid leukemia or high-risk myelodysplastic syndrome: predictive prognostic models for outcome.
Cancer
2006
, vol. 
106
 
5
(pg. 
1090
-
1098
)
23
Menzin
 
J
Lang
 
K
Earle
 
CC
Kerney
 
D
Mallick
 
R
The outcomes and costs of acute myeloid leukemia among the elderly.
Arch Intern Med
2002
, vol. 
162
 
14
(pg. 
1597
-
1603
)
24
Estey
 
E
de Lima
 
M
Tibes
 
R
, et al. 
Prospective feasibility analysis of reduced-intensity conditioning (RIC) regimens for hematopoietic stem cell transplantation (HSCT) in elderly patients with acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (MDS).
Blood
2007
, vol. 
109
 
4
(pg. 
1395
-
1400
)
25
Hegenbart
 
U
Niederwieser
 
D
Sandmaier
 
BM
, et al. 
Treatment for acute myelogenous leukemia by low-dose, total-body, irradiation-based conditioning and hematopoietic cell transplantation from related and unrelated donors.
J Clin Oncol
2006
, vol. 
24
 
3
(pg. 
444
-
453
)
26
Passweg
 
JR
Baldomero
 
H
Bader
 
P
, et al. 
Hematopoietic SCT in Europe 2013: recent trends in the use of alternative donors showing more haploidentical donors but fewer cord blood transplants.
Bone Marrow Transplant
2015
, vol. 
50
 
4
(pg. 
476
-
482
)
27
Reshef
 
R
Porter
 
DL
Reduced-intensity conditioned allogeneic SCT in adults with AML.
Bone Marrow Transplant
2015
, vol. 
50
 
6
(pg. 
759
-
769
)
28
Gyurkocza
 
B
Sandmaier
 
BM
Conditioning regimens for hematopoietic cell transplantation: one size does not fit all.
Blood
2014
, vol. 
124
 
3
(pg. 
344
-
353
)
29
Sengsayadeth
 
S
Savani
 
BN
Blaise
 
D
Malard
 
F
Nagler
 
A
Mohty
 
M
Reduced intensity conditioning allogeneic hematopoietic cell transplantation for adult acute myeloid leukemia in complete remission - a review from the Acute Leukemia Working Party of the EBMT.
Haematologica
2015
, vol. 
100
 
7
(pg. 
859
-
869
)
30
Bacigalupo
 
A
Ballen
 
K
Rizzo
 
D
, et al. 
Defining the intensity of conditioning regimens: working definitions.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
12
(pg. 
1628
-
1633
)
31
Gyurkocza
 
B
Storb
 
R
Storer
 
BE
, et al. 
Nonmyeloablative allogeneic hematopoietic cell transplantation in patients with acute myeloid leukemia.
J Clin Oncol
2010
, vol. 
28
 
17
(pg. 
2859
-
2867
)
32
Blaise
 
D
Tabrizi
 
R
Boher
 
JM
, et al. 
Randomized study of 2 reduced-intensity conditioning strategies for human leukocyte antigen-matched, related allogeneic peripheral blood stem cell transplantation: prospective clinical and socioeconomic evaluation.
Cancer
2013
, vol. 
119
 
3
(pg. 
602
-
611
)
33
Shimoni
 
A
Hardan
 
I
Shem-Tov
 
N
, et al. 
Comparison between two fludarabine-based reduced-intensity conditioning regimens before allogeneic hematopoietic stem-cell transplantation: fludarabine/melphalan is associated with higher incidence of acute graft-versus-host disease and non-relapse mortality and lower incidence of relapse than fludarabine/busulfan.
Leukemia
2007
, vol. 
21
 
10
(pg. 
2109
-
2116
)
34
Danylesko
 
I
Shimoni
 
A
Nagler
 
A
Treosulfan-based conditioning before hematopoietic SCT: more than a BU look-alike.
Bone Marrow Transplant
2012
, vol. 
47
 
1
(pg. 
5
-
14
)
35
Ciurea
 
SO
Andersson
 
BS
Busulfan in hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
5
(pg. 
523
-
536
)
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
Gratwohl
 
A
Baldomero
 
H
Schwendener
 
A
, et al. 
The EBMT activity survey 2008: impact of team size, team density and new trends.
Bone Marrow Transplant
2011
, vol. 
46
 
2
(pg. 
174
-
191
)
38
Finke
 
J
Bethge
 
WA
Schmoor
 
C
, et al. 
Standard graft-versus-host disease prophylaxis with or without anti-T-cell globulin in haematopoietic cell transplantation from matched unrelated donors: a randomised, open-label, multicentre phase 3 trial.
Lancet Oncol
2009
, vol. 
10
 
9
(pg. 
855
-
864
)
39
Kumar
 
A
Mhaskar
 
AR
Reljic
 
T
, et al. 
Antithymocyte globulin for acute-graft-versus-host-disease prophylaxis in patients undergoing allogeneic hematopoietic cell transplantation: a systematic review.
Leukemia
2012
, vol. 
26
 
4
(pg. 
582
-
588
)
40
Soiffer
 
RJ
Lerademacher
 
J
Ho
 
V
, et al. 
Impact of immune modulation with anti-T-cell antibodies on the outcome of reduced-intensity allogeneic hematopoietic stem cell transplantation for hematologic malignancies.
Blood
2011
, vol. 
117
 
25
(pg. 
6963
-
6970
)
41
Baron
 
F
Labopin
 
M
Blaise
 
D
, et al. 
Impact of in vivo T-cell depletion on outcome of AML patients in first CR given peripheral blood stem cells and reduced-intensity conditioning allo-SCT from a HLA-identical sibling donor: a report from the Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation.
Bone Marrow Transplant
2014
, vol. 
49
 
3
(pg. 
389
-
396
)
42
Devillier
 
R
Furst
 
S
El-Cheikh
 
J
, et al. 
Antithymocyte globulin in reduced-intensity conditioning regimen allows a high disease-free survival exempt of long-term chronic graft-versus-host disease.
Biol Blood Marrow Transplant
2014
, vol. 
20
 
3
(pg. 
370
-
374
)
43
Mohty
 
M
Bay
 
JO
Faucher
 
C
, et al. 
Graft-versus-host disease following allogeneic transplantation from HLA-identical sibling with antithymocyte globulin-based reduced-intensity preparative regimen.
Blood
2003
, vol. 
102
 
2
(pg. 
470
-
476
)
44
Mohty
 
M
Jacot
 
W
Faucher
 
C
, et al. 
Infectious complications following allogeneic HLA-identical sibling transplantation with antithymocyte globulin-based reduced intensity preparative regimen.
Leukemia
2003
, vol. 
17
 
11
(pg. 
2168
-
2177
)
45
Blaise
 
D
Devillier
 
R
Lecoroller-Sorriano
 
AG
, et al. 
Low non-relapse mortality and long-term preserved quality of life in older patients undergoing matched related donor allogeneic stem cell transplantation: a prospective multicenter phase II trial.
Haematologica
2015
, vol. 
100
 
2
(pg. 
269
-
274
)
46
Russell
 
NH
Kjeldsen
 
L
Craddock
 
C
, et al. 
UK NCRI Adult AML Working Party. A comparative assessment of the curative potential of reduced intensity allografts in acute myeloid leukaemia.
Leukemia
2014
 
29(7):1478-1484
47
Kanakry
 
CG
O’Donnell
 
PV
Furlong
 
T
, et al. 
Multi-institutional study of post-transplantation cyclophosphamide as single-agent graft-versus-host disease prophylaxis after allogeneic bone marrow transplantation using myeloablative busulfan and fludarabine conditioning.
J Clin Oncol
2014
, vol. 
32
 
31
(pg. 
3497
-
3505
)
48
Sorror
 
ML
Sandmaier
 
BM
Storer
 
BE
, et al. 
Long-term outcomes among older patients following nonmyeloablative conditioning and allogeneic hematopoietic cell transplantation for advanced hematologic malignancies.
JAMA
2011
, vol. 
306
 
17
(pg. 
1874
-
1883
)
49
Brunner
 
AM
Kim
 
HT
Coughlin
 
E
, et al. 
Outcomes in patients age 70 or older undergoing allogeneic hematopoietic stem cell transplantation for hematologic malignancies.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
9
(pg. 
1374
-
1380
)
50
Federmann
 
B
Faul
 
C
Meisner
 
C
Vogel
 
W
Kanz
 
L
Bethge
 
WA
Influence of age on outcome after allogeneic hematopoietic cell transplantation: a single center study in patients aged 60.
Bone Marrow Transplant
2015
, vol. 
50
 
3
(pg. 
427
-
431
)
51
Versluis
 
J
Labopin
 
M
Niederwieser
 
D
, et al. 
Prediction of non-relapse mortality in recipients of reduced intensity conditioning allogeneic stem cell transplantation with AML in first complete remission.
Leukemia
2015
, vol. 
29
 
1
(pg. 
51
-
57
)
52
Sorror
 
ML
Storb
 
RF
Sandmaier
 
BM
, et al. 
Comorbidity-age index: a clinical measure of biologic age before allogeneic hematopoietic cell transplantation.
J Clin Oncol
2014
, vol. 
32
 
29
(pg. 
3249
-
3256
)
53
Muffly
 
LS
Kocherginsky
 
M
Stock
 
W
, et al. 
Geriatric assessment to predict survival in older allogeneic hematopoietic cell transplantation recipients.
Haematologica
2014
, vol. 
99
 
8
(pg. 
1373
-
1379
)
54
Cornelissen
 
JJ
van Putten
 
WL
Verdonck
 
LF
, et al. 
Results of a HOVON/SAKK donor versus no-donor analysis of myeloablative HLA-identical sibling stem cell transplantation in first remission acute myeloid leukemia in young and middle-aged adults: benefits for whom?
Blood
2007
, vol. 
109
 
9
(pg. 
3658
-
3666
)
55
Koreth
 
J
Schlenk
 
R
Kopecky
 
KJ
, et al. 
Allogeneic stem cell transplantation for acute myeloid leukemia in first complete remission: systematic review and meta-analysis of prospective clinical trials.
JAMA
2009
, vol. 
301
 
22
(pg. 
2349
-
2361
)
56
Cornelissen
 
JJ
Versluis
 
J
Passweg
 
JR
, et al. 
Comparative therapeutic value of post-remission approaches in patients with acute myeloid leukemia aged 40-60 years.
Leukemia
2015
, vol. 
29
 
5
(pg. 
1041
-
1050
)
57
Champlin
 
R
Reduced intensity allogeneic hematopoietic transplantation is an established standard of care for treatment of older patients with acute myeloid leukemia.
Best Pract Res Clin Haematol
2013
, vol. 
26
 
3
(pg. 
297
-
300
)
58
Ustun
 
C
Lazarus
 
HM
Weisdorf
 
D
To transplant or not: a dilemma for treatment of elderly AML patients in the twenty-first century.
Bone Marrow Transplant
2013
, vol. 
48
 
12
(pg. 
1497
-
1505
)
59
Versluis
 
J
Hazenberg
 
C
Passweg
 
JR
, et al. 
Post-remission treatment with allogeneic stem cell transplantation in patients aged 60 years and older with acute myeloid leukaemia: a time-dependent analysis.
Lancet Haematol
 
In press
60
Sorror
 
ML
Appelbaum
 
FR
Risk assessment before allogeneic hematopoietic cell transplantation for older adults with acute myeloid leukemia.
Expert Rev Hematol
2013
, vol. 
6
 
5
(pg. 
547
-
562
)
61
Herr
 
AL
Labopin
 
M
Blaise
 
D
, et al. 
HLA-identical sibling allogeneic peripheral blood stem cell transplantation with reduced intensity conditioning compared to autologous peripheral blood stem cell transplantation for elderly patients with de novo acute myeloid leukemia.
Leukemia
2007
, vol. 
21
 
1
(pg. 
129
-
135
)
62
Mohty
 
M
de Lavallade
 
H
El-Cheikh
 
J
, et al. 
Reduced intensity conditioning allogeneic stem cell transplantation for patients with acute myeloid leukemia: long term results of a ‘donor’ versus ‘no donor’ comparison.
Leukemia
2009
, vol. 
23
 
1
(pg. 
194
-
196
)
63
Abdul Wahid
 
SF
Ismail
 
NA
Mohd-Idris
 
MR
, et al. 
Comparison of reduced-intensity and myeloablative conditioning regimens for allogeneic hematopoietic stem cell transplantation in patients with acute myeloid leukemia and acute lymphoblastic leukemia: a meta-analysis.
Stem Cells Dev
2014
, vol. 
23
 
21
(pg. 
2535
-
2552
)
64
Bornhäuser
 
M
Kienast
 
J
Trenschel
 
R
, et al. 
Reduced-intensity conditioning versus standard conditioning before allogeneic haemopoietic cell transplantation in patients with acute myeloid leukaemia in first complete remission: a prospective, open-label randomised phase 3 trial.
Lancet Oncol
2012
, vol. 
13
 
10
(pg. 
1035
-
1044
)
65
Schmid
 
C
Schleuning
 
M
Tischer
 
J
, et al. 
Early allo-SCT for AML with a complex aberrant karyotype–results from a prospective pilot study.
Bone Marrow Transplant
2012
, vol. 
47
 
1
(pg. 
46
-
53
)
66
de Lima
 
M
Giralt
 
S
Thall
 
PF
, et al. 
Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogenous leukemia or myelodysplastic syndrome: a dose and schedule finding study.
Cancer
2010
, vol. 
116
 
23
(pg. 
5420
-
5431
)
67
Kochenderfer
 
JN
Dudley
 
ME
Carpenter
 
RO
, et al. 
Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation.
Blood
2013
, vol. 
122
 
25
(pg. 
4129
-
4139
)
68
Appelbaum
 
FR
Alternative donor transplantation for adults with acute leukemia.
Best Pract Res Clin Haematol
2014
, vol. 
27
 
3-4
(pg. 
272
-
277
)
69
Gragert
 
L
Eapen
 
M
Williams
 
E
, et al. 
HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. registry.
N Engl J Med
2014
, vol. 
371
 
4
(pg. 
339
-
348
)
70
Yakoub-Agha
 
I
Mesnil
 
F
Kuentz
 
M
, et al. 
Allogeneic marrow stem-cell transplantation from human leukocyte antigen-identical siblings versus human leukocyte antigen-allelic-matched unrelated donors (10/10) in patients with standard-risk hematologic malignancy: a prospective study from the French Society of Bone Marrow Transplantation and Cell Therapy.
J Clin Oncol
2006
, vol. 
24
 
36
(pg. 
5695
-
5702
)
71
Saber
 
W
Opie
 
S
Rizzo
 
JD
Zhang
 
MJ
Horowitz
 
MM
Schriber
 
J
Outcomes after matched unrelated donor versus identical sibling hematopoietic cell transplantation in adults with acute myelogenous leukemia.
Blood
2012
, vol. 
119
 
17
(pg. 
3908
-
3916
)
72
Kröger
 
N
Zabelina
 
T
de Wreede
 
L
, et al. 
Allogeneic stem cell transplantation for older advanced MDS patients: improved survival with young unrelated donor in comparison with HLA-identical siblings.
Leukemia
2013
, vol. 
27
 
3
(pg. 
604
-
609
)
73
Bacigalupo
 
A
 
Matched and mismatched unrelated donor transplantation: is the outcome the same as for matched sibling donor transplantation? Hematology Am Soc Hematol Educ Program. 2012;2012:223-229
74
Schlenk
 
RF
Dohner
 
K
Mack
 
S
, et al. 
Prospective evaluation of allogeneic hematopoietic stem-cell transplantation from matched related and matched unrelated donors in younger adults with high-risk acute myeloid leukemia: German-Austrian trial AMLHD98A.
J Clin Oncol
2010
, vol. 
28
 
30
(pg. 
4642
-
4648
)
75
Oran
 
B
Wagner
 
JE
DeFor
 
TE
Weisdorf
 
DJ
Brunstein
 
CG
Effect of conditioning regimen intensity on acute myeloid leukemia outcomes after umbilical cord blood transplantation.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
9
(pg. 
1327
-
1334
)
76
Brunstein
 
CG
Fuchs
 
EJ
Carter
 
SL
, et al. 
Alternative donor transplantation after reduced intensity conditioning: results of parallel phase 2 trials using partially HLA-mismatched related bone marrow or unrelated double umbilical cord blood grafts.
Blood
2011
, vol. 
118
 
2
(pg. 
282
-
288
)
77
Brunstein
 
CG
Eapen
 
M
Ahn
 
KW
, et al. 
Reduced-intensity conditioning transplantation in acute leukemia: the effect of source of unrelated donor stem cells on outcomes.
Blood
2012
, vol. 
119
 
23
(pg. 
5591
-
5598
)
78
Laughlin
 
MJ
Barker
 
J
Bambach
 
B
, et al. 
Hematopoietic engraftment and survival in adult recipients of umbilical-cord blood from unrelated donors.
N Engl J Med
2001
, vol. 
344
 
24
(pg. 
1815
-
1822
)
79
Barker
 
JN
Weisdorf
 
DJ
DeFor
 
TE
, et al. 
Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy.
Blood
2005
, vol. 
105
 
3
(pg. 
1343
-
1347
)
80
Kelly
 
SS
Sola
 
CB
de Lima
 
M
Shpall
 
E
Ex vivo expansion of cord blood.
Bone Marrow Transplant
2009
, vol. 
44
 
10
(pg. 
673
-
681
)
81
Eapen
 
M
Rocha
 
V
Sanz
 
G
, et al. 
Effect of graft source on unrelated donor haemopoietic stem-cell transplantation in adults with acute leukaemia: a retrospective analysis.
Lancet Oncol
2010
, vol. 
11
 
7
(pg. 
653
-
660
)
82
Stanevsky
 
A
Shimoni
 
A
Yerushalmi
 
R
Nagler
 
A
Double umbilical cord blood transplant: more than a cell dose?
Leuk Lymphoma
2010
, vol. 
51
 
6
(pg. 
975
-
982
)
83
Wagner
 
JE
Eapen
 
M
Carter
 
S
, et al. 
One-unit versus two-unit cord-blood transplantation for hematologic cancers.
N Engl J Med
2014
, vol. 
371
 
18
(pg. 
1685
-
1694
)
84
Aversa
 
F
Tabilio
 
A
Velardi
 
A
, et al. 
Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype.
N Engl J Med
1998
, vol. 
339
 
17
(pg. 
1186
-
1193
)
85
Aversa
 
F
Terenzi
 
A
Tabilio
 
A
, et al. 
Full haplotype-mismatched hematopoietic stem-cell transplantation: a phase II study in patients with acute leukemia at high risk of relapse.
J Clin Oncol
2005
, vol. 
23
 
15
(pg. 
3447
-
3454
)
86
Huang
 
XJ
Liu
 
DH
Liu
 
KY
, et al. 
Haploidentical hematopoietic stem cell transplantation without in vitro T-cell depletion for the treatment of hematological malignancies.
Bone Marrow Transplant
2006
, vol. 
38
 
4
(pg. 
291
-
297
)
87
Luznik
 
L
O’Donnell
 
PV
Symons
 
HJ
, et al. 
HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
6
(pg. 
641
-
650
)
88
Lee
 
KH
Lee
 
JH
Lee
 
JH
, et al. 
Hematopoietic cell transplantation from an HLA-mismatched familial donor is feasible without ex vivo-T cell depletion after reduced-intensity conditioning with busulfan, fludarabine, and antithymocyte globulin.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
1
(pg. 
61
-
72
)
89
Lee
 
KH
Lee
 
JH
Lee
 
JH
, et al. 
Reduced-intensity conditioning therapy with busulfan, fludarabine, and antithymocyte globulin for HLA-haploidentical hematopoietic cell transplantation in acute leukemia and myelodysplastic syndrome.
Blood
2011
, vol. 
118
 
9
(pg. 
2609
-
2617
)
90
Di Bartolomeo
 
P
Santarone
 
S
De Angelis
 
G
, et al. 
Haploidentical, unmanipulated, G-CSF-primed bone marrow transplantation for patients with high-risk hematologic malignancies.
Blood
2013
, vol. 
121
 
5
(pg. 
849
-
857
)
91
Wang
 
Y
Chang
 
YJ
Xu
 
LP
, et al. 
Who is the best donor for a related HLA haplotype-mismatched transplant?
Blood
2014
, vol. 
124
 
6
(pg. 
843
-
850
)
92
Bashey
 
A
Zhang
 
X
Sizemore
 
CA
, et al. 
T-cell-replete HLA-haploidentical hematopoietic transplantation for hematologic malignancies using post-transplantation cyclophosphamide results in outcomes equivalent to those of contemporaneous HLA-matched related and unrelated donor transplantation.
J Clin Oncol
2013
, vol. 
31
 
10
(pg. 
1310
-
1316
)
93
Raiola
 
AM
Dominietto
 
A
Ghiso
 
A
, et al. 
Unmanipulated haploidentical bone marrow transplantation and posttransplantation cyclophosphamide for hematologic malignancies after myeloablative conditioning.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
1
(pg. 
117
-
122
)
94
Castagna
 
L
Bramanti
 
S
Furst
 
S
, et al. 
Nonmyeloablative conditioning, unmanipulated haploidentical SCT and post-infusion CY for advanced lymphomas.
Bone Marrow Transplant
2014
, vol. 
49
 
12
(pg. 
1475
-
1480
)
95
Ruggeri
 
A
Labopin
 
M
Sanz
 
G
, et al. 
Comparison of outcomes after unrelated cord blood and unmanipulated haploidentical stem cell transplantation in adults with acute leukemia.
Leukemia
2015
 
29(9):1891-1900
96
Ciceri
 
F
Labopin
 
M
Aversa
 
F
, et al. 
A survey of fully haploidentical hematopoietic stem cell transplantation in adults with high-risk acute leukemia: a risk factor analysis of outcomes for patients in remission at transplantation.
Blood
2008
, vol. 
112
 
9
(pg. 
3574
-
3581
)
97
Ciurea
 
SO
Zhang
 
MJ
Bacigalupo
 
AA
, et al. 
Haploidentical transplant with posttransplant cyclophosphamide vs matched unrelated donor transplant for acute myeloid leukemia.
Blood
2015
 
126(8):1033-1040
98
Raiola
 
AM
Dominietto
 
A
di Grazia
 
C
, et al. 
Unmanipulated haploidentical transplants compared with other alternative donors and matched sibling grafts.
Biol Blood Marrow Transplant
2014
, vol. 
20
 
10
(pg. 
1573
-
1579
)
99
Di Stasi
 
A
Milton
 
DR
Poon
 
LM
, et al. 
Similar transplantation outcomes for acute myeloid leukemia and myelodysplastic syndrome patients with haploidentical versus 10/10 human leukocyte antigen-matched unrelated and related donors.
Biol Blood Marrow Transplant
2014
, vol. 
20
 
12
(pg. 
1975
-
1981
)
100
El-Cheikh
 
J
Crocchiolo
 
R
Furst
 
S
, et al. 
Unrelated cord blood compared with haploidentical grafts in patients with hematological malignancies.
Cancer
2015
, vol. 
121
 
11
(pg. 
1809
-
1816
)
101
Wang
 
Y
Liu
 
QF
Xu
 
LP
, et al. 
Haploidentical vs identical-sibling transplant for AML in remission: a multicenter, prospective study.
Blood
2015
, vol. 
125
 
25
(pg. 
3956
-
3962
)
102
Vellenga
 
E
van Putten
 
W
Ossenkoppele
 
GJ
, et al. 
Autologous peripheral blood stem cell transplantation for acute myeloid leukemia.
Blood
2011
, vol. 
118
 
23
(pg. 
6037
-
6042
)
103
Keating
 
A
DaSilva
 
G
Perez
 
WS
, et al. 
Autologous blood cell transplantation versus HLA-identical sibling transplantation for acute myeloid leukemia in first complete remission: a registry study from the Center for International Blood and Marrow Transplantation Research.
Haematologica
2013
, vol. 
98
 
2
(pg. 
185
-
192
)
104
Pfirrmann
 
M
Ehninger
 
G
Thiede
 
C
, et al. 
Prediction of post-remission survival in acute myeloid leukaemia: a post-hoc analysis of the AML96 trial.
Lancet Oncol
2012
, vol. 
13
 
2
(pg. 
207
-
214
)
105
Burnett
 
AK
Goldstone
 
A
Hills
 
RK
, et al. 
Curability of patients with acute myeloid leukemia who did not undergo transplantation in first remission.
J Clin Oncol
2013
, vol. 
31
 
10
(pg. 
1293
-
1301
)
106
Terwijn
 
M
van Putten
 
WL
Kelder
 
A
, et al. 
High prognostic impact of flow cytometric minimal residual disease detection in acute myeloid leukemia: data from the HOVON/SAKK AML 42A study.
J Clin Oncol
2013
, vol. 
31
 
31
(pg. 
3889
-
3897
)
107
Walter
 
RB
Gooley
 
TA
Wood
 
BL
, et al. 
Impact of pretransplantation minimal residual disease, as detected by multiparametric flow cytometry, on outcome of myeloablative hematopoietic cell transplantation for acute myeloid leukemia.
J Clin Oncol
2011
, vol. 
29
 
9
(pg. 
1190
-
1197
)
108
Schnittger
 
S
Kern
 
W
Tschulik
 
C
, et al. 
Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML.
Blood
2009
, vol. 
114
 
11
(pg. 
2220
-
2231
)
109
Jourdan
 
E
Boissel
 
N
Chevret
 
S
, et al. 
Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia.
Blood
2013
, vol. 
121
 
12
(pg. 
2213
-
2223
)
110
Schlenk
 
RF
Taskesen
 
E
van Norden
 
Y
, et al. 
The value of allogeneic and autologous hematopoietic stem cell transplantation in prognostically favorable acute myeloid leukemia with double mutant CEBPA.
Blood
2013
, vol. 
122
 
9
(pg. 
1576
-
1582
)
111
Lichtenegger
 
FS
Krupka
 
C
Kohnke
 
T
Subklewe
 
M
Immunotherapy for Acute Myeloid Leukemia.
Semin Hematol
2015
, vol. 
52
 
3
(pg. 
207
-
214
)
Sign in via your Institution