Progress in the last decade has improved the understanding of leukemia biology. Molecular markers in combinations with cytogenetics have improved the risk stratification of acute myeloid leukemia (AML) and informed decision-making. In parallel, several important advances in the transplant field, such as better supportive care, improved transplant technology, increased availability of alternative donors, and reduced-intensity conditioning have improved the safety as well as access of allogeneic hematopoietic cell transplantation (HCT) for a larger number of patients. In this review, the positioning of HCT in the management of patients with AML is evaluated in view of changing risk/benefit ratios associated with both conventional treatments and transplantation, and some of the controversies are addressed in light of emerging data. Increasing data demonstrate outcomes of alternative donor transplantation approaching HLA-identical sibling donors in high-risk AML supporting the inclusion of alternative donors in trials of prospective studies evaluating post remission strategies for high-risk AML. The use of reduced-intensity conditioning has expanded the eligibility of HCT to older patients with AML, and outcome data are encouraging. Continued study of HCT versus alternative therapies is required to optimize patients' outcomes in AML.

Allogeneic hematopoietic cell transplantation (HCT) is a curative treatment option for patients with acute myeloid leukemia (AML). The curative effect of HCT in patients with AML is contributed both by the chemotherapy and/or radiation in the preparative regimen and more importantly by the immunologic graft-versus-leukemia (GVL) effect.1  Currently, AML is the most common indication for HCT. Data from Center for International Blood and Marrow Transplant Research (CIBMTR) indicate a sustained increase in the number of AML allogeneic transplants in the last decade (Table 1). Although the number of transplants from related donors has remained stable during the last decade, transplants from unrelated donors (URDs) are growing (Table 1). The growth of transplant activity is mainly in adults and is attributable to increased use of URDs, especially in first remission (CR1; Table 1, Figure 1). CIBMTR data demonstrate that 47% of CR1 allografts (all ages) in AML in 2008 were performed with the use of URDs. Among URDs, umbilical cord blood (UCB) is becoming an important graft source in adults and contributed to 11% of adult transplants from URDs in 2008 (Table 1). Despite these trends, there remains wide variation in the application of HCT in AML patients, especially during CR1. The guidelines of various major organizations in Europe and the United States on the use of HCT for AML patients are not consistent.2 

Table 1

Trends in AML allogeneic transplant activity: cases registered with the CIBMTR

19981999200020012002200320042005200620072008
No of centers reporting to CIBMTR 162 171 189 208 220 208 222 210 212 197 184 
AML patients undergoing allo-HCT, all ages            
    All patients 1778 1826 2045 2119 2324 2326 2621 2735 2697 2581 2557 
    RD 1286 1288 1359 1319 1462 1382 1444 1485 1436 1284 1223 
    vURD 443 474 627 697 744 822 1046 1091 1053 1085 1114 
    UCB 49 64 59 103 118 122 131 159 208 212 220 
Adult*AML allo-HCT patients using vURD            
    CR1 72 88 153 186 211 273 353 422 441 467 518 
    ≥ CR2 99 104 148 162 173 161 245 258 212 239 221 
    Not in remission 126 118 149 155 179 193 242 216 225 173 278 
Adult*AML allo-HCT patients using UCB            
    CR1 14 14 16 29 43 39 47 
    ≥ CR2 12 17 27 30 39 45 
    Not in remission 16 14 11 21 24 38 
Graft source on all adult AML patients, %            
    Related donor            
        PB 43 54 65 74 76 83 83 87 87 88 88 
        BM 57 46 34 26 24 17 17% 13 13 12 12 
        UCB 0.5 0.3 0.6 0% 0.1 0.2 
    Unrelated donor            
        PB 13 29 39 46 60 68 69 70 71 75 
        BM 91 83 68 55 50 34 26 23 19 18 14 
        UCB 11 11 11 
19981999200020012002200320042005200620072008
No of centers reporting to CIBMTR 162 171 189 208 220 208 222 210 212 197 184 
AML patients undergoing allo-HCT, all ages            
    All patients 1778 1826 2045 2119 2324 2326 2621 2735 2697 2581 2557 
    RD 1286 1288 1359 1319 1462 1382 1444 1485 1436 1284 1223 
    vURD 443 474 627 697 744 822 1046 1091 1053 1085 1114 
    UCB 49 64 59 103 118 122 131 159 208 212 220 
Adult*AML allo-HCT patients using vURD            
    CR1 72 88 153 186 211 273 353 422 441 467 518 
    ≥ CR2 99 104 148 162 173 161 245 258 212 239 221 
    Not in remission 126 118 149 155 179 193 242 216 225 173 278 
Adult*AML allo-HCT patients using UCB            
    CR1 14 14 16 29 43 39 47 
    ≥ CR2 12 17 27 30 39 45 
    Not in remission 16 14 11 21 24 38 
Graft source on all adult AML patients, %            
    Related donor            
        PB 43 54 65 74 76 83 83 87 87 88 88 
        BM 57 46 34 26 24 17 17% 13 13 12 12 
        UCB 0.5 0.3 0.6 0% 0.1 0.2 
    Unrelated donor            
        PB 13 29 39 46 60 68 69 70 71 75 
        BM 91 83 68 55 50 34 26 23 19 18 14 
        UCB 11 11 11 

allo-HCT indicates allogeneic hematopoietic cell transplantation; AML, acute myeloid leukemia; BM, bone marrow, CIBMTR, Center for International Blood and Marrow Transplant Research; CR, complete remission; CR1, first complete remission; CR2, second complete remission; PB, peripheral blood; RD, related donor; UCB, umbilical cord blood; and vURD, voluntary unrelated donor.

*

Adult defined as ≥ 18 years.

Figure 1

Trends in allogeneic HCT activity in adult AML patients (≥ 18 years) by the use of unrelated donors according to disease status at HCT: cases registered with CIBMTR from 1998 to 2008. (A) Adult AML patients (≥18 years), CR1, CR2, CR3, and not in remission, undergoing unrelated HCT. (B) Adult AML patients (≥ 18 years) undergoing unrelated HCT in CR1.

Figure 1

Trends in allogeneic HCT activity in adult AML patients (≥ 18 years) by the use of unrelated donors according to disease status at HCT: cases registered with CIBMTR from 1998 to 2008. (A) Adult AML patients (≥18 years), CR1, CR2, CR3, and not in remission, undergoing unrelated HCT. (B) Adult AML patients (≥ 18 years) undergoing unrelated HCT in CR1.

Close modal

Progress in the last 2 decades in understanding the disease biology of AML may guide further practice changes. Cytogenetics is the most robust prognostic marker for risk stratification of AML at the time of diagnosis as well as in selection of postremission treatments.3-5  On the basis of specific structural and numerical cytogenetic abnormalities, patients with AML are divided into favorable, intermediate-, or adverse-risk groups. More recently, new molecular markers such as FMS-related tyrosine kinase 3–internal tandem duplication (FLT3-ITD), CCAAT/enhancer-binding protein-α (CEBPA), and the nucleophosmin-1 (NPM1) mutation have further refined the risk stratification, particularly in patients with cytogenetically normal AML (CN-AML).6  These cytogenetic and molecular markers have helped identify patients at different risk of relapse after achieving CR. In parallel, major advances have developed in the transplant field. Better supportive care, and selection of URD with the use of high-resolution allele-level HLA-typing have improved the safety of transplantation. Increasing use of UCB, and application of reduced-intensity conditioning (RIC) regimens have increased the candidacy for HCT to include a wider group of patients, particularly older or ethnic minority patients who were not well served by earlier HCT and donor search options. In this review, we evaluate the positioning of HCT in the management of patients with AML in the light of changing risk/benefit ratios associated with both conventional chemotherapy and HCT and address controversies highlighted by emerging data.

Several groups have investigated the role of HLA-matched sibling donor (MSD) transplantation in patients with AML in CR1. The designs of these studies involve genetic assignment substituting for randomization on the basis of the availability of a MSD. These studies were generally not powered to evaluate the outcome of HCT according to cytogenetic risks. Subgroup analyses of these studies on the basis of cytogenetics included small sample sizes. In a recent systematic review and meta-analysis of prospective biologic assignment studies, researchers analyzed 3638 patients with AML in CR1 by cytogenetic risk and showed a significant survival advantage of HCT in comparison with nonallogeneic treatments for AML patients with intermediate and unfavorable cytogenetics, although not for good risk cytogenetics.7  Recent progress in the understanding of leukemia biology has further refined prognostication of intermediate-risk group patients, especially CN-AML, and has contributed to the identification of subgroups among those with favorable risk cytogenetics at greater risk of relapse. Current guidelines and new evidence for AML patients in CR1 on the basis of leukemia biology raise new questions: (1) Should all patients with CN-AML with a suitable donor be referred for HCT in CR1? (2) Is there a role of HCT for a subgroup of patients with good-risk cytogenetics in CR1? (3) If a MSD is not available, is alternative donor HCT indicated in CR1?

CN-AML is a heterogeneous group comprising approximately 40%-50% of AML patients < 60 years, less common in patients > 60, and currently categorized as intermediate risk.3-5  Recently, several molecular alternations have helped researchers to improve the individual risk assessment of CN-AML patients.8  Several authors have shown the prognostic significance for mutations in the NPM1, CEBPA, and FLT3 genes alone or in combination in both younger and older adults with AML. The presence of the FLT3-ITD mutation has been identified as a powerful indicator predicting more frequent and early relapse.9-12  The adverse impact in FLT3-ITD–positive patients may be greater in those with a high level of the mutated allele.10,13  The prognostic significance of FLT3-TKD mutations remains controversial in view of conflicting data.14-16 NPM1 mutation (mNPM1) in the absence of FLT3-ITD mutation in CN-AML has been associated with lower cumulative incidence of relapse, resulting in better leukemia-free survival (LFS) and overall survival (OS).17-20  The outcomes of AML with genotype “mNPM1 without FLT3-ITD” treated with conventional chemotherapy appear favorable and similar to patients with t(8;21) or inv(16).11,19  CN-AML with mutations in CEBPA (mCEBPA) are associated with a favorable prognosis.11,21-24  The role of molecular genetic alterations in AML has been recognized in the 2008 revised World Health Organization classification, and AML with mNPM1 or mCEBPA have been incorporated in the World Health Organization classification as provisional entities.25 

In a study from Germany, researchers analyzed the role of mutational status of NPM1, FLT3, CEBPA, along with MLL and NRAS in guiding postremission therapy for CN-AML in CR1.11  They the analyzed data from CN-AML patients treated in 4 prospective clinical trials. Treatment intention was similar in all 4 trials, and patients with a HLA-matched related donor (MRD) were assigned to undergo HCT in CR1; those without a donor were randomly assigned to receive high-dose cytarabine (HiDAC) consolidation or autologous transplant. HCT was performed in 82% of assigned patients. Autologous transplant or consolidation therapy resulted in similar outcomes. An intention-to-treat analysis on the basis of donor availability demonstrated significantly longer relapse-free survival (RFS) in the donor group (P = .009). Data were further analyzed on the basis of mutation status, and patients were further subdivided into 2 groups: patients with “mNPM1 without FLT3-ITD,” and patients with all other genotypes. Because of small numbers, patients with mCEBPA were excluded. No differences in RFS was seen with or without a donor in the favorable genotype patients with mNPM1 without FLT3-ITD (P = .71). Among the patients with CN-AML with genotypes other than “mNPM1 without FLT3-ITD,” superior RFS was observed in those with a donor. At present, there are no comparative data available on outcomes of HCT versus nonallogeneic treatments in CN-AML patients with “mCEBPA.” The outcome of “mCEBPA” patients is similar to “mNPM without FLT3-ITD.”11  Increasing data show that the better outcome of CEBPA mutations is restricted to patients with double mutation only.24,26,27  Therefore among the CN-AML, HCT in CR1 could be deferred in favorable genotype of “mCEBPA or mNPM1 without FLT3-ITD.” HCT should be considered in all other patients with CN-AML in CR1, at least in those up to age 60 years.

The definition of favorable genotype may be further refined with the identification of new mutations, although the favorable subtypes may remain uncommon. Recently identified isocitrate dehydrogenase enzyme isoform 1 mutations (mIDH1) and isoform 2 mutation (mIDH2) have been associated with a poor prognosis in patients with AML.28,29  Of particular note, mIDH1 was seen among 21 of 86 patients with mNPM1. None of the 22 patients with mCEBPA had mIDH1.28  None of the patients with mNPM1 or mCEBPA had mIDH2. The authors suggested that favorable genotype among CN-AML patients should be modified to “mNPM1 or mCEBPA with neither FLT3-ITD nor mIDH1.” Other recently described mutations in the DNA methyltransferase gene DNMT3A are frequent in patients with intermediate-risk cytogenetics and associated with increased relapse risk and poor outcome.30  Of note, a significant benefit of HCT was observed in this small series. Findings from these recent but small studies need further validation before routine application. Evolving technologies such as microarray profiling31-33 and microRNA signatures34  have shown promise in improving the prognostic classification of AML; however, these methodologies are not yet available for routine clinical use.

Patients with core binding factor (CBF) leukemia (ie, t[8;21] or inv[16] or t[16;16]) or acute promyelocytic leukemia (APL) with t(15;17) are considered at relatively lower risk of relapse and thus are considered to have favorable risk cytogenetics.3-5  The meta-analysis of prospective genetic randomization studies showed no benefit of HCT in patients with favorable risk cytogenetics in CR1.7  With all-trans retinoic acid or arsenic trioxide–based treatments, the outcome of APL with t(15;17) has significantly improved. These treatment strategies lead to cure in most patients, and the expected relapse rate is usually 10%-25%.35  However, in CBF-AML, only 50% of patients were alive at 5 years.36  Therefore, some of these patients may still have a high risk of relapse. The role of KIT mutations (mKIT) in CBF-AML has been investigated to identify a high-risk subset in the otherwise-favorable CBF group.37-40  Two commonly identified KIT mutations (exon17 [mKIT 17] and exon 8 [mKIT 8]) in CBF-AML appear to have prognostic relevance; however, the data from these studies are not consistent. Discrepant results may be related to small number of patients and differences in the treatments.

Repetitive cycles of HiDAC as postremission therapy are associated with favorable outcomes in patients with CBF-AML.41,42  In a Cancer and Leukemia Group B (CALGB) study, authors investigated the role of mKIT in 61 adult patients with inv(16) and 49 patients with t(8;21) assigned to postremission therapy with HiDAC.40  In patients with inv(16), the 5-year relapse-risk was significantly greater in patients with mKIT (56% vs 29%, P = .05) and in particular mKIT17 (80% vs 29%, P = .002) in comparison with wtKIT. Similarly, in patients with t(8;21), the 5-year relapse-risk was significantly greater in those mKIT (70% vs 36%, P = .017). This relapse pattern is similar to that seen with patients with adverse cytogenetics. In view of these data, we may suggest that patients with “CBF-AML with mKIT” be considered for HCT in CR1. It is noteworthy that at present, there are no available data to show the benefit of HCT in “CBF-AML with mKIT” although resistance to HiDAC or other consolidation strategies may not imply resistance to the allogeneic GVL effect. The high-risk of relapse in these patients merit the investigation of alternative treatment strategies including HCT or possibly molecular targeted therapies with tyrosine kinase inhibitors in future studies.

Most AML patients with adverse cytogenetics relapse within a year, and likelihood of subsequent CR is very low.43,44  Meta-analysis of prospective biologic assignment studies comparing the role of HCT to non-HCT treatments for patients with adverse cytogenetics in CR1 have shown a strong survival advantage for HCT during CR1.7,45  Most patients in these studies received HCT via the use of MSD, and comparisons of URD HCT to nonallogeneic treatments are limited.46,47  In a recent study from CIBMTR, researchers compared the outcomes of URD and MSD transplants in patients with AML in CR1 with unfavorable cytogenetics.48  This study showed similar LFS and OS of HLA-well matched URD (no known disparity at HLA-A, B, C, or DRB1) in comparison with MSD, whereas outcomes were not as good for human leukocyte antigen (HLA)-partially matched URD (disparity at one locus). Prospective comparisons of MSD and URD for high-risk AML patients in CR1 on a limited number of patients show equivalent outcomes.46,47  Therefore, if a MSD is not available, an HLA-well matched URD is appropriate for patients with AML with adverse cytogenetics in CR1.

There are only scanty data on comparisons of MRD and URD transplantation in intermediate-risk AML in CR1. In a study from Seattle, researchers compared 85 patients with URD versus 135 patients undergoing MRD transplantation in CR1.49  In the intermediate-risk group, 58 and 83 patients underwent HCT via the use of URD and MRD, respectively. Most patients in intermediate-risk had a normal karyotype. Although these sample sizes are modest, the outcomes of URD and MRD appear similar. Among the patients with normal karyotype, those with FLT3-ITD mutations are at a greater risk of relapse. At present, no comparative data are available on outcomes of MSD and URD in AML patients with FLT3-ITD mutations. The high risk of relapse in these patients merits the use of HCT with either related or URD HCT and further investigation of novel molecular targeted therapies. A high risk of early relapse and lengthy time for volunteer URD searches confound the use of URD HCT in CR1 in high-risk AML patients. Therefore, donor searches for such patients should be initiated early, that is, during the initial induction.

An important but as yet incompletely addressed question is the utility of UCB-HCT for patients with high-risk AML in CR1. Previous registry studies have compared the outcomes of unrelated UCB with unrelated bone marrow (URD-BM) grafts in adults with acute leukemia.50,51  A study from the European Group of Blood and Marrow Transplantation (EBMT) showed similar outcomes among the 2 cohorts,51  whereas the study from CIBMTR50  showed lower treatment-related mortality (TRM), treatment failure, and overall mortality among the recipients of HLA-matched BM grafts; similar outcomes were found for patients receiving mismatched UCB transplants and mismatched URD-BM grafts.50  In a recent study from CIBMTR, the authors compared the outcomes of UCB (n = 165) with unrelated peripheral blood (URD-PB; n = 888), URD-BM (n = 472) in patients with acute leukemia.52  The majority of patients receiving UCB graft were 4/6 antigen match (70%). The LFS in patients after 4-6/6 matched UCB-HCT were comparable with that after 8/8 or 7/8 allele-matched URD-PB or URD-BM recipients; however, the TRM was greater after UCB-HCT. A recently published study from Minnesota and Seattle demonstrated similar outcomes in double unit UCB and matched URD compared with MSD after myeloablative HCT in hematologic malignancies.53  Because these studies analyzed impact of graft source in patients with mixture of different disease status in acute leukemia50-52  or hematologic malignancies,53  the results are hard to interpret for AML in CR1.

In a recent study from the Japan, researchers evaluated the disease-specific comparison of unrelated UCB recipients and HLA-allele–matched URD-BM recipients in 484 adult patients with AML (173, CB; and 311, BM).54  In this study, 180 AML patients underwent transplant in CR1 (50, CB; and 130, BM). Multivariate analysis showed inferior survival of patients in CR1 receiving UCB-HCT versus URD-BM (relative risk 2.92 (95% confidence interval 1.38-6.18), P = .005). The inferior survival with UCB-HCT in these patients was associated with greater TRM in the UCB group. Contrary to the commonly held belief that GVL effect after URD or mismatched donor is more potent in comparison with MSD, the studies in which the authors compared different donor sources show that the relapse rate was not decreased after well-matched URD48,55  or mismatched URD/UCB-HCT.48,52,54 

Should haplo-identical transplantation (haplo-HCT) be a suitable option for high-risk AML patients in CR1? Very limited data are available on use of haplo-HCT in patients with AML in CR1. In a recent study from the EBMT, authors analyzed the outcomes of haplo-HCT in patients with acute leukemia in remission.56  This study included 86 patients with AML in remission (25 in CR1). The 2-year cumulative incidence of TRM, relapse, and probability of LFS were 36%, 16%, and 48%, respectively in CR1 patients. The effect of donor graft sources in AML from various studies are summarized in Table 2.46,48,49,54,57-59  These data are intriguing but insufficient to guide decisions about URD, UCB, or haplo-HCT, particularly in centers with little experience with either UCB or haplo-HCT where complications of graft failure or slower immune recovery need extra attention, even if graft-versus-host-disease is less common. However, their rapid availability offers an important clinical advantage for patients in whom donor search was delayed or cohorts of greater risk where remission duration may be brief. Current data suggest that these options be used only in the absence of a timely available URD. No data exist on the comparison of UCB and haplo-HCT to nonallogeneic treatments in AML in CR1 at present; emerging data support the inclusion of UCB, haplo-HCT in addition to volunteer URD in prospective studies in which the authors evaluate postremission strategies for high-risk AML in CR1.

Table 2

Comparative outcomes of various donor sources for HCT in AML

StudySample size and patient populationDisease statusMain comparisonLFS and OSAdditional comments
Hegenbert et al, 2006,57  Multicenter, Seattle Consortium 122, age 17-74 y, using NMA conditioning only CR1 (n = 51), CR2 (n = 39), advanced (n = 32) MRD (n = 58) vs MUD (n = 64) No difference between MRD and MUD Disease status at HCT and cytogenetics most important factors for LFS and OS 
Moore et al, 2007,58  Multicenter, Australasian Registry 210, age 16-59 y, using MAC only CR1 (n = 36), > CR1/others (n = 174) MSD (n = 105) vs URD (n = 105) No difference between MSD and URD Matched case-controlled study 
Schetelig et al, 2008,59  Multicenter, Germany 368, age 50-73 y, both MAC and RIC CR1 (n = 136), advanced, (n = 228), others (n = 4) MSD (n = 168) vs M/MRD (n = 12) vs MUD (n = 51) vs possibly MUD (n = 68) vs partially MUD (n = 45) vs poorly MUD (n = 24) No difference between different donor types Advanced disease at HCT, secondary AML, and high risk cytogenetics associated with poor outcomes 
Atsuta et al, 2009,54  Multicenter, JMDP/JCBBN 484, age 16-69 y, using MAC only CR1 (n = 180), Others (n = 304) MUD (n = 311) vs UCB-HCT (n = 173) Inferior outcomes in UCB-HCT attributable to increased TRM No difference in risk of relapse 
Walter et al, 2010,49  single center, Seattle 220, age 18-69 y, using MAC only CR1, Intermediate cytogenetics (n = 141), high risk cytogenetics (n = 60), others (n = 19) MRD (n = 135) vs 10/10 MUD (n = 62) vs 9/10 URD (n = 23) No difference between MRD vs 10/10 MUD vs 9/10 URD Unfavorable cytogenetics and high HCT-CI score associated with worse outcomes 
Gupta et al, 2010,48  Multicenter, CIBMTR 584, age <1-74 y, Both MAC and RIC AML in CR1 with adverse cytogenetics MSD (n = 226) vs well-matched URD (n = 254) vs Partially matched URD (n = 104) Similar between MSD and well-matched URD, inferior for partially matched URD Lower risk of relapse in patients with chronic GVHD 
Schlenk et al, 2010,46  Multicenter, Germany and Austria 162, age 19-61 y, Both MAC and RIC High-risk AML in CR1, patients refractory to induction therapy MRD (n = 62) vs MUD (n = 89) vs CB/HaploHCT (n = 11) Similar between MSD and MUD Prospective study, also compared with patients who could not get transplant, benefit of HCT seen in comparison with non-HCT patients in landmark analysis 
StudySample size and patient populationDisease statusMain comparisonLFS and OSAdditional comments
Hegenbert et al, 2006,57  Multicenter, Seattle Consortium 122, age 17-74 y, using NMA conditioning only CR1 (n = 51), CR2 (n = 39), advanced (n = 32) MRD (n = 58) vs MUD (n = 64) No difference between MRD and MUD Disease status at HCT and cytogenetics most important factors for LFS and OS 
Moore et al, 2007,58  Multicenter, Australasian Registry 210, age 16-59 y, using MAC only CR1 (n = 36), > CR1/others (n = 174) MSD (n = 105) vs URD (n = 105) No difference between MSD and URD Matched case-controlled study 
Schetelig et al, 2008,59  Multicenter, Germany 368, age 50-73 y, both MAC and RIC CR1 (n = 136), advanced, (n = 228), others (n = 4) MSD (n = 168) vs M/MRD (n = 12) vs MUD (n = 51) vs possibly MUD (n = 68) vs partially MUD (n = 45) vs poorly MUD (n = 24) No difference between different donor types Advanced disease at HCT, secondary AML, and high risk cytogenetics associated with poor outcomes 
Atsuta et al, 2009,54  Multicenter, JMDP/JCBBN 484, age 16-69 y, using MAC only CR1 (n = 180), Others (n = 304) MUD (n = 311) vs UCB-HCT (n = 173) Inferior outcomes in UCB-HCT attributable to increased TRM No difference in risk of relapse 
Walter et al, 2010,49  single center, Seattle 220, age 18-69 y, using MAC only CR1, Intermediate cytogenetics (n = 141), high risk cytogenetics (n = 60), others (n = 19) MRD (n = 135) vs 10/10 MUD (n = 62) vs 9/10 URD (n = 23) No difference between MRD vs 10/10 MUD vs 9/10 URD Unfavorable cytogenetics and high HCT-CI score associated with worse outcomes 
Gupta et al, 2010,48  Multicenter, CIBMTR 584, age <1-74 y, Both MAC and RIC AML in CR1 with adverse cytogenetics MSD (n = 226) vs well-matched URD (n = 254) vs Partially matched URD (n = 104) Similar between MSD and well-matched URD, inferior for partially matched URD Lower risk of relapse in patients with chronic GVHD 
Schlenk et al, 2010,46  Multicenter, Germany and Austria 162, age 19-61 y, Both MAC and RIC High-risk AML in CR1, patients refractory to induction therapy MRD (n = 62) vs MUD (n = 89) vs CB/HaploHCT (n = 11) Similar between MSD and MUD Prospective study, also compared with patients who could not get transplant, benefit of HCT seen in comparison with non-HCT patients in landmark analysis 

CIBMTR indicates Center for International Blood and Marrow Transplant Research; CR1, first complete remission; CR2, second complete remission; GVHD, graft versus host disease; haploHCT, haplo-identical cell transplantation; HCT, hematopoietic cell transplantation; HCT-CI, hematopoietic cell transplantation comorbidity index; JCCBN, Japanese cord blood bank network; JMDP, Japanese Marrow Donor Program; LFS, leukemia-free survival; MAC, myeloablative conditioning; NMA, nonmyeloablative; OS, overall survival; M/MRD, mismatched related donor; MRD, matched related donor; MSD, matched sibling donor; MUD, matched unrelated donor; RIC, reduced-intensity conditioning; TRM, treatment-related mortality; UCB, umbilical cord blood; and URD, unrelated donor.

The treatment of AML in first relapse is associated with unsatisfactory results and survival is usually poor. There are no prospective studies in which the authors evaluate the outcome of HCT in comparison to conventional chemotherapy in patients with relapsed AML. All reported data are retrospective in nature and have the limitations of treatment heterogeneity and selection bias. However, a prospective study in this area is logistically difficult and unlikely to be performed. HCT is often used in this setting, despite only limited evidence about its outcome.

One study from Europe evaluated the outcomes of 667 AML patients in first relapse among 1540 newly diagnosed non-M3 AML patients (age 15-60 years) entered into 3 consecutive cooperative group trials.60  The authors identified 4 prognostic factors in multivariate analysis: relapse-free interval from CR1, cytogenetics at diagnosis, age at first relapse, and autologous or HCT before first relapse. On the basis of these factors, a weighted prognostic score was proposed to identify 3 risk groups: favorable, intermediate, and poor risk. For patients able to achieve CR2, comparison of chemotherapy versus HCT among these 3 groups showed superior 5-year survival in patients undergoing HCT (favorable, 88% vs 33%; intermediate, 48% vs 31%; poor, 26% vs 6%). Achievement of CR2 and application of salvage HCT are crucial for improving the prognosis of these patients.60,61  Survival of patients in first relapse undergoing salvage HCT was significantly better for those who achieved CR2 compared with those not in remission at HCT (3-year survival 59% vs 21%).61  Few data on comparison of UCB-HCT/ haplo-HCT versus chemotherapy in this setting are reported. Given the poor prognosis associated with chemotherapy alone, UCB or haplo-HCT for patients in CR2 may be valuable, particularly in light of their rapid availability.

The utility of HCT for patients with AML with active disease remains controversial. Several authors62-68  have reported the outcomes of HCT in patients with acute leukemia not in remission at the time of transplant with variable outcomes. Small sample sizes; a mixture of patients with AML, ALL, and CML blast crisis; and publication bias confound interpretation of data.

In a recent CIBMTR study, researchers evaluated the outcome of 1673 patients with AML not in remission at the time of HCT.69  Survival at 3 and 5 years was 19% and 17%, respectively. In the multivariate analysis, 5 adverse patient-, disease- and transplant- related factors were identified: first CR duration < 6 months, circulating blasts, donor other than MSD, performance score < 90%, and adverse risk cytogenetics. The 3-year survival of patients with none of these risk factors was 42%, and those with 1, 2, or ≥ 3 risk factors was 28%, 15%, and 6%, respectively. These results provide important guidance in identifying groups where the transplant procedure has reasonable chances of success, as well as identifying patients for whom transplant procedure is likely to be futile. This study included only patients treated with myeloablative conditioning. The role of RIC in patients with active acute leukemia is unclear. Some investigators have suggested that RIC may be an option,67,70,71  whereas others have reported futility of this procedure in such patients.72 

For patients with refractory AML (primary induction failure, ie, no CR after 2 cycles of therapy), relapse after a CR1 < 6 months, second or greater relapse, or resistant elapsed disease, success in achieving CR with any further salvage therapy is very low (10%-15% at best). Further chemotherapy extends the risks of opportunistic, often fungal infection and organ toxicities, any of which can increase TRM of future HCT. A sequential approach of use of salvage chemotherapy for reduction of leukemia burden followed by RIC was reported from Germany.71  This approach led to encouraging 3-year LFS and OS of 30% and 32% in patients with refractory AML, yet still reflects substantial patient selection.

Relapse is the major cause of failure in AML patients not in remission at HCT, and novel strategies to improve the efficacy of conditioning such as addition of targeted radiation,73  intensity modulated radiation therapy,74  or post-HCT strategies such as azacytidine, prophylactic donor lymphocyte infusions (DLIs), adoptive transfer of natural killer cells, leukemia-specific T cells or leukemia vaccines aimed at promoting a more potent or durable GvL effect require study.71,75-77 

Guidelines for the indications of HCT in AML from various donor sources are summarized in Table 3. With in the framework of these guidelines, each patient should be carefully evaluated for the risk posed by disease itself versus risk from the transplant procedure taking in considerations factors such as age, performance status, comorbidities, and donor-recipient matching. Decisions may further be guided by the likely ability to achieve CR2 in case of relapse.

Table 3

Guidelines for indications for HCT in adult patients with AML

MSDMUDUCBHaplo-identical
First remission     
    Favorable cytogenetics     
        APL No No No No 
        CBF-AML* No No No No 
        With mKIT Uncertain Uncertain Uncertain Uncertain 
        Without mKIT No No No No 
    CN-AML* Yes Uncertain Uncertain Uncertain 
        “mNPM1 without FLT3ITD” No No No No 
        “mCEBPA No No No No 
        Others than above Yes Uncertain Uncertain Uncertain 
    Intermediate risk with abnormal cytogenetics Yes Uncertain Uncertain Uncertain 
    Adverse Yes Yes Yes Yes 
Second remission Yes Yes Yes Yes 
Not in remission§ Yes Yes Uncertain Uncertain 
MSDMUDUCBHaplo-identical
First remission     
    Favorable cytogenetics     
        APL No No No No 
        CBF-AML* No No No No 
        With mKIT Uncertain Uncertain Uncertain Uncertain 
        Without mKIT No No No No 
    CN-AML* Yes Uncertain Uncertain Uncertain 
        “mNPM1 without FLT3ITD” No No No No 
        “mCEBPA No No No No 
        Others than above Yes Uncertain Uncertain Uncertain 
    Intermediate risk with abnormal cytogenetics Yes Uncertain Uncertain Uncertain 
    Adverse Yes Yes Yes Yes 
Second remission Yes Yes Yes Yes 
Not in remission§ Yes Yes Uncertain Uncertain 

Uncertain implies insufficient published data for a recommendation.

APL indicates acute promyelocytic leukemia; CBF, core binding factor [t(8;21) or Inv(16)]; CN-AML, cytogenetically normal AML; FLT3ITD, FMS-related tyrosine kinase 3–internal tandem duplication; HCT, hematopoietic cell transplantation; mCEBPA, mutated CEBPA; mKIT, KIT mutations; MSD, matched sibling donor; mNPM1, mutated NPM1; MUD, matched unrelated donor; and UCB, umbilical cord blood.

*

If the data on molecular markers are not available.

Increasing data show that the beneficial effect may be restricted only to patients with double mutations.

Only at experienced centers and in the absence of a timely available MUD.

§

Carefully selected patients with good performance status and low disease burden; CIBMTR risk score may aid the patient selection.

With the expanding pool of cancer survivors, therapy-related AML (t-AML) is increasingly encountered and constitutes approximately 10%-20% of newly diagnosed patients with AML.78  Conventional chemotherapy is not curative, and HCT is a potential treatment for patients with t-AML. However, the best t-AML candidates for HCT are not well defined. There are no prospective data defining the value of HCT in patients with t-AML to guide treatment decisions.

Outcomes of HCT in patients with t-AML are inferior to published data on de novo AML.79-81  This finding is attributable in part to a high number of t-AML patients with poor leukemia biology, including unfavorable cytogenetics, previous therapy-related myelodysplastic syndrome (t-MDS), and active disease at the time of HCT. However in some reports, when adjusted for disease status and cytogenetics, there was no difference in outcomes of t-AML and de novo AML.82,83  The results of t-MDS/AML from 3 major registry studies from the French Society of Bone Marrow Transplantation,81  EBMT,80  and CIBMTR79  are summarized in Table 4. Several common conclusions can be drawn from these studies. HCT yields encouraging outcomes in younger patients in remission with MSD, well-matched URD (8/8), or partially matched URD (7/8). Poor-risk cytogenetics has a significant adverse impact on relapse. On the basis of 4 prognostic factors for survival (age > 35 years, poor risk cytogenetics, AML not in remission or advanced MDS, and donors other than MSD, well-matched or partially matched URD), the CIBMTR study proposed a prognostic scoring system for t-AML undergoing HCT.79  Five-year survival of patients with score 0, 1, 2, 3, and 4 was 50%, 26%, 21%, 10%, and 4%, respectively. This scoring system may be particularly useful to guide selection of subset of patients likely to benefit from HCT and suggest investigational or palliative approaches for those lacking the favorable features.

Cytogenetic classification is an independent prognostic parameter in patients with t-AML.84  However, when comparable cytogenetic groups were evaluated, the survival of t-AML with favorable cytogenetics was inferior to those with de novo AML. Among the CBF-AML, the outcome of t(8;21) in t-AML appears inferior compared with de novo AML,84,85  whereas the outcomes appear similar for the rare patients with inv(16).84  The outcome of t-APL does not appear different from de novo-APL when treated with an all-trans retinoic acid–containing regimen.86  For t-AML, should patients with rare CBF leukemia or CN-AML with favorable molecular profile be referred for HCT in CR1? Currently, there are no data to determine the prognostic impact of “mNPM1 without FLT3-ITD” or “mCEBPA” in the small subset of t-AML patients in comparison to de novo AML patients.

Given the poor prognosis of t-AML with conventional chemotherapy, HCT should be considered during CR1 for all patients in the transplant age group with suitable donors. HCT can be deferred for t-APL, and possibly inv(16) in CR1, but even these data are limited.

Similar to t-AML, AML evolving from preceding myelodysplastic syndrome or myeloproliferative disorder is recognized as high risk and as such, is not curable with conventional chemotherapy. HCT has promise, but no prospective data directly addresses this topic.

Conventional chemotherapy options are not curative in a majority of AML patients ≥ 60 years of age.87  High peritransplantation mortality with myeloablative transplant was assumed to be a major barrier resulting in only limited application of HCT in older patients.88  The introduction of RIC has enabled to overcome the barrier of early TRM, and several authors89-91  have shown the feasibility and reasonable outcomes with RIC in older patients with AML ≥ 60 years using related and URDs. It appears that the likelihood of older patients being referred for HCT is very low in comparison to younger patients.92  Hesitancy of treating physicians, uncertainty regarding outcomes of HCT, lack of comparative data on the outcomes of HCT versus nontransplant treatments in older patients, and insurance coverage are some of the barriers contributing to underuse of HCT in older patients.

In a recent study from CIBMTR, authors evaluated the impact of age in 545 patients with AML in CR1 with age ≥ 40 years undergoing RIC (40-54 years, 201; 55-59 years, 149; 60-64 years, 132; ≥ 65 years, 63).93  In this study, 2-year LFS and OS in all age groups were similar, and no impact of age was observed. Two single-center studies showed no impact of age on posttransplant outcomes in patients ≥ 60 years treated with RIC.94,95  In another multicenter study from the Seattle consortium, the outcomes of 274 patients with AML treated with a nonmyeloablative conditioning included 135 patients with AML ≥ 60 years and no impact of age was observed.96  An important observation was that MSD and well-matched URD transplants led to similar survival.93-96  In view of the aforementioned data, HCT can be considered in patients up to the age 70 years. Very few HCT patients older than 70 years of age have been reported. Therefore, suitably fit patients between the age 60-70 years should be informed about the option of HCT, and donor searches should be initiated promptly after diagnosis to allow this option if a CR is achieved.

Most of the comparisons of allogeneic versus non-allogeneic treatments in AML are reported in younger patients up to the age 60 years; at present, there are no prospective comparisons of allogeneic versus conventional treatments in older patients. Preliminary results of a retrospective case-controlled study from the CIBMTR and CALGB comparing the outcomes of RIC transplantation (n = 100) with conventional chemotherapy (n = 96) in patients ≥ 60 years with AML were recently reported.97  To avoid selection bias for the HCT arm, only patients remaining in CR1 for at least 4 months treated on CALGB trials were included in the chemotherapy arm. The 3-year LFS from CR1 for HCT patients was 32% compared with 15% for chemotherapy-treated patients (P = .006). Although relapse-risk in HCT arm was significantly lower (32% vs 81%, P < .001); TRM was significantly greater (22% vs 3%, P < .001) resulting in marginal difference in OS (HCT, 37% vs chemotherapy, 25%;P = .08). These data indicate the importance of ongoing efforts to improve the results of HCT with a focus to decrease TRM as well as novel strategies to decrease the relapse risk in non-allogeneic treatments. Continued efforts should be made to recruit these patients in to prospective studies comparing HCT versus nonallogeneic treatments.

The preferred intensity of conditioning therapy for AML patients remains a subject of debate. In the last decade, a plethora of RIC regimens have been developed. The spectrum of intensity of RIC regimens vary from minimal to moderately intense. To facilitate comparisons of intensity of conditioning regimens, members of the CIBMTR has developed guidelines to define these regimens, and on the basis of the expected duration of cytopenia and on the requirement for stem cell support, these regimens are defined as(1) myeloablative conditioning (MAC),(2) RIC, and (3) nonmyeloablative (NMA) conditioning.98 

Various studies in which authors compared the outcomes of RIC/NMA conditioning with MAC in AML patients are summarized in Table 5.70,72,79-81,99-105  Results of these retrospective studies are limited by significant differences in patient populations and the analysis may be influenced by selection bias. Patients expected to have a high risk of relapse may be selected for MAC; and patients with advanced age and/or comorbidities may more often be selected for RIC/NMA regimens. Missing data on comorbidities and details of the decision-making process for RIC/NMA are other major limitations. Importantly, all studies have reported similar LFS and OS in patients undergoing MAC and RIC, respectively. Some have reported decreased TRM with RIC in comparison with MAC,70,101,104,106  whereas others have reported similar TRM.99,102,103,105  A comparison of NMA versus RIC versus MAC regimens showed similar outcomes of RIC and MAC, whereas outcomes were inferior for NMA as the result of increased relapse.105  A comparison of RIC versus NMA in patients with AML/MDS suggested better outcomes with RIC.107  Relapse rates between MAC and RIC appear similar in patients with leukemia in CR at the time of HCT72,99,105 ; dose intensity appears particularly important for patients with leukemia not in remission at the time of HCT.70,72  Of importance, none of the studies so far has shown superiority of RIC/NMA to MAC in AML. Therefore, RIC/NMA should only be offered to the patients considered ineligible for MAC. At present, the indications of RIC/NMA are not consistent. Tools such as HCT-specific comorbidity index (HCT-CI),108  pretransplant assessment of mortality score,109  and understanding of interaction of comorbidities, performance status, with age may aid in developing objective criteria for candidacy for RIC/NMA. The best regimen for RIC/NMA is not known, but available data support the use of RIC over NMA regimens for AML.

Table 4

Multicenter studies in which the authors evaluated prognostic risk factors in patients with t-MDS/AML undergoing HCT

StudySample sizeAdverse prognostic factors for survival
Other comments
AgeCytogeneticsDisease statusDonor typeOther factors
Yakoub-Agha et al, JCO, 200081  (The French Society of BMT) 70 t-MDS = 31 t-AML = 39 > 37 y Not evaluated Not in CR at HCT Not evaluated, only few URD Male Recipient CMV Positive Intensive MAC All received MAC 
Kroger et al, Haematologica, 200980  (EBMT) 461 t-AML = 293 t-MDS = 168 > 40 y Abnormal ncytogenetics Not in CR at HCT MSD vs URD not significant – No difference in MAC vs RIC, based on prognostic factors identified 3 groups with different survival at 3 y: 62%, 33%, and 24% 
Litzow et al, Blood, 201079  (CIBMTR) 868 t-AML = 545 t-MDS = 323 > 35 y Poor risk cytogenetics* AML not in remission or advanced MDS Similar outcomes with MSD, well-matched URD (8/8) or partially matched URD (7/8); worse outcomes with mismatched RD or URD  No difference in MAC vs RIC, based on 4 risk factors generated a risk score. Patients with risk score of 0, 1, 2, 3, and 4 had a 5-year survival of 50%, 26%, 21%, 10%, and 4%, respectively 
StudySample sizeAdverse prognostic factors for survival
Other comments
AgeCytogeneticsDisease statusDonor typeOther factors
Yakoub-Agha et al, JCO, 200081  (The French Society of BMT) 70 t-MDS = 31 t-AML = 39 > 37 y Not evaluated Not in CR at HCT Not evaluated, only few URD Male Recipient CMV Positive Intensive MAC All received MAC 
Kroger et al, Haematologica, 200980  (EBMT) 461 t-AML = 293 t-MDS = 168 > 40 y Abnormal ncytogenetics Not in CR at HCT MSD vs URD not significant – No difference in MAC vs RIC, based on prognostic factors identified 3 groups with different survival at 3 y: 62%, 33%, and 24% 
Litzow et al, Blood, 201079  (CIBMTR) 868 t-AML = 545 t-MDS = 323 > 35 y Poor risk cytogenetics* AML not in remission or advanced MDS Similar outcomes with MSD, well-matched URD (8/8) or partially matched URD (7/8); worse outcomes with mismatched RD or URD  No difference in MAC vs RIC, based on 4 risk factors generated a risk score. Patients with risk score of 0, 1, 2, 3, and 4 had a 5-year survival of 50%, 26%, 21%, 10%, and 4%, respectively 

CIBMTR, Center for International Blood and Marrow Transplant Research; CMV, CR, complete remission; EBMT, European Group of Blood and Marrow Transplantation; HCT, allogeneic hematopoietic cell transplantation; MAC, myeloablative conditioning; MDS, myelodysplastic syndrome; MSD, matched sibling donor; RIC, reduced intensity conditioning; t-MDS/AML, therapy-related MDS/AML; and URD, unrelated donor.

*

Defined according to Southwest Oncology Group/Eastern Cooperative Oncology Group classification for AML patients and International Prognostic Scoring System classification for MDS patients;

Table 5

Comparisons of myeloablative and reduced intensity or nonmyeloablative conditioning in adult patients with AML

StudyPatient populationSample sizeLFSOSOther comments
Aoudjhane et al, 2005104 ; Multicenter, EBMT AML, > 50 y, MSD only 722, RIC = 315, MAC = 407 Similar Similar Decreased acute GVHD, chronic GVHD, and TRM, but increased relapse with RIC 
Alyea et al, 200670 ; single center, Boston AML/MDS, 21-70 y, MRD and URD donors 136, (AML, 82) RIC = 39, MAC = 97 Similar Similar Decreased TRM, and increased relapse with RIC 
Scott et al, 2006102 ; single center, Seattle MDS/sAML with previous MDS, 40-72 y, MRD and URD 150, (AML, 55) NMA = 38, MAC = 112 Similar Similar No difference in relapse/TRM 
Shimoni et al, 200672 ; single center, Tel-Hashomer AML/MDS, 17-70 y, MRD and URD 112, (AML, 56) RIC = 67, MAC = 45 Similar Similar Similar outcomes for patients in remission at HCT, inferior outcomes of patients with active disease treated with RIC 
Flynn et al, 2007103 ; single center, Minnesota AML/MDS, 19-69 y, MRD and URD (included UCB grafts) 219, (AML, 160) RIC = 32, MAC = 187 Similar Similar Similar TRM, but increase in relapse with RIC 
Ringden et al, 2009101 ; EBMT multicenter AML, 16-76 y, URD transplants only 1555, RIC = 401, MAC = 1154 Similar Not reported Reduced NRM in ≥50 y, and increased relapse in patients <50 y with RIC. 
Lim et al, 2010106 ; EBMT, multicenter MDS/sAML with previous MDS, ≥ 50 y, MRD and URD 1333, (AML, 334), RIC = 833, MAC = 500 Not reported Similar Increased relapse, and decreased TRM with RIC 
Khabori et al, 201099 ; single center, Toronto AML/MDS, 40-60 y, MRD and URD transplants 101, (AML, 77), RIC = 39, MAC = 62 Similar Similar Poor outcome in patients with high-risk disease biology attributable to higher relapse rate 
Luger et al, CIBMTR105 , multicenter AML/MDS, 18-70 y, MRD and URD 5179, RIC/NMA = 1448, MAC = 3731 Similar between MAC vs RIC, inferior for NMA Similar between MAC vs RIC, More relapse with NMA Late TRM negated any advantage offered by RIC or NMA 
StudyPatient populationSample sizeLFSOSOther comments
Aoudjhane et al, 2005104 ; Multicenter, EBMT AML, > 50 y, MSD only 722, RIC = 315, MAC = 407 Similar Similar Decreased acute GVHD, chronic GVHD, and TRM, but increased relapse with RIC 
Alyea et al, 200670 ; single center, Boston AML/MDS, 21-70 y, MRD and URD donors 136, (AML, 82) RIC = 39, MAC = 97 Similar Similar Decreased TRM, and increased relapse with RIC 
Scott et al, 2006102 ; single center, Seattle MDS/sAML with previous MDS, 40-72 y, MRD and URD 150, (AML, 55) NMA = 38, MAC = 112 Similar Similar No difference in relapse/TRM 
Shimoni et al, 200672 ; single center, Tel-Hashomer AML/MDS, 17-70 y, MRD and URD 112, (AML, 56) RIC = 67, MAC = 45 Similar Similar Similar outcomes for patients in remission at HCT, inferior outcomes of patients with active disease treated with RIC 
Flynn et al, 2007103 ; single center, Minnesota AML/MDS, 19-69 y, MRD and URD (included UCB grafts) 219, (AML, 160) RIC = 32, MAC = 187 Similar Similar Similar TRM, but increase in relapse with RIC 
Ringden et al, 2009101 ; EBMT multicenter AML, 16-76 y, URD transplants only 1555, RIC = 401, MAC = 1154 Similar Not reported Reduced NRM in ≥50 y, and increased relapse in patients <50 y with RIC. 
Lim et al, 2010106 ; EBMT, multicenter MDS/sAML with previous MDS, ≥ 50 y, MRD and URD 1333, (AML, 334), RIC = 833, MAC = 500 Not reported Similar Increased relapse, and decreased TRM with RIC 
Khabori et al, 201099 ; single center, Toronto AML/MDS, 40-60 y, MRD and URD transplants 101, (AML, 77), RIC = 39, MAC = 62 Similar Similar Poor outcome in patients with high-risk disease biology attributable to higher relapse rate 
Luger et al, CIBMTR105 , multicenter AML/MDS, 18-70 y, MRD and URD 5179, RIC/NMA = 1448, MAC = 3731 Similar between MAC vs RIC, inferior for NMA Similar between MAC vs RIC, More relapse with NMA Late TRM negated any advantage offered by RIC or NMA 

CIBMTR indicates Center for International Blood and Marrow Transplant Research; EBMT, European Group for Blood and Marrow Transplantation; GVHD, graft versus host disease; HCT, hematopoietic cell transplantation; LFS, leukemia-free survival; MAC, myeloablative conditioning; MRD, matched related donor; MUD, matched unrelated donor; NMA, nonmyeloablative, NRM, nonrelapse mortality; OS, overall survival; RIC, reduced-intensity conditioning; sAML, secondary AML; TRM, treatment-related mortality; UCB, umbilical cord blood; and URD, unrelated donor.

Current data on application of RIC/NMA regimens in AML lack prospective comparison of allogeneic versus nonallogeneic treatments. The authors of a retrospective study evaluated RIC HCT in comparison with chemotherapy in 95 patients with high-risk AML in CR1.110  When an intention-to-treat approach was used, a “donor” versus “no donor” comparison showed better LFS in the donor group (54% vs 30%, P = .01). These encouraging results further need prospective validation including from studies presently in progress.

Relapse is one of most common cause of failure of HCT for AML and is associated with very poor prognosis.111  The preferred management of relapse after HCT is not known. Conventional treatment options include supportive care, chemotherapy, second HCT using the same or alternative donor, DLI, and cytokines. Substantial selection bias confounds interpretation of the reported literature.

Second HCT after failure of first HCT has limited efficacy in patients with AML.112-116  Available literature suggests that duration of remission after first HCT is the most important prognostic factor.114-116  A reasonable outcome can be expected in selected patients, whose duration of remission after first HCT is ≥ 1 year, and who achieve CR before a second HCT.114-116  DLI has limited efficacy in patients with AML. In 2 prospective studies authors have evaluated the strategy of reduction of disease burden with chemotherapy followed by granulocyte colony-stimulating factor-primed DLI in patients with advanced myeloid malignancies.117,118  This strategy was beneficial to those who were able to achieve CR. Patients with remission lasting > 6 months had a greater likelihood of response. The efficacy of DLI was retrospectively studied in a EBMT study, including 399 patients in first relapse after HCT for AML who received DLI (n = 171), or not (n = 228).119  Clinical benefit of DLI was seen only in a minority particularly with those with a lower tumor burden at relapse (< 35% BM blasts), female sex, favorable cytogenetics, and remission before DLI.

Therefore, a second allogeneic treatment can be considered in selected patients, whose duration of remission after first HCT is > 6 months. Decision-making and the selection of second HCT versus DLI should be individualized on the basis of donor availability and achievement of remission before DLI or second HCT. Recently, low-dose azacytidine has been reported to be of benefit in patients relapsing after HCT in small series, and this strategy may be useful tool for disease debulking as an alternative to chemotherapy before DLI/second HCT.75  More research is needed to understand the biology of relapse to develop novel interventions for this difficult problem.

The trend of growth of numbers of HCT in adult patients with AML can be expected to continue based on acceptance and availability of URD and UCB donor. The highly specialized nature and resource intensity of HCT will lead to major challenges for manpower and resources. Few data are available for the reliable estimates of the number of transplants for AML and the total number of AML patients for whom transplantation is appropriate. With the expansion of donor registries, and availability of UCB grafts a suitable donor can potentially be found for nearly all patients with AML.46  However, there appears a great discrepancy in the number of newly diagnosed AML patients in the transplant age group and the number of cases reported to the CIBMTR. Volunteer URD searches limit timely availability of graft as patients with acute leukemia may relapse while waiting for transplant.120  In addition, although race and ethnicity may limit URD availability, significant barriers such as age, sex, socioeconomic status, donor registry and center funding plus insurance restrictions, and other unknown barriers for access to HCT still exist.121  Addressing these challenges will be required to harness the full potential of HCT for patients with AML.

We acknowledge assistance from the CIBMTR for the data on trends in AML HCT.

Contribution: V.G. prepared the initial draft of manuscript; M.ST. and D.J.W. provided further knowledge, insights, and helped in critical review; and all authors approved the final version of the manuscript.

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

Correspondence: Dr Vikas Gupta, MD, FRCP, FRCPath, Blood and Marrow Transplant Program, Princess Margaret Hospital, Suite 5-217, 610 University Ave, Toronto, M5G 2M9, ON, Canada; e-mail: vikas.gupta@uhn.on.ca.

1
Horowitz
 
MM
Gale
 
RP
Sondel
 
PM
, et al. 
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
1990
, vol. 
75
 
3
(pg. 
555
-
562
)
2
Hubel
 
K
Weingart
 
O
Naumann
 
F
Bohlius
 
J
Wheatley
 
K
Engert
 
A
Allogeneic stem cell transplantation in adult patients with acute myeloid leukemia: to transplant or not to transplant? A Systematic review of international guidelines [abstract].
Blood
2009
, vol. 
114
 
22
(pg. 
577
-
78
Abstract 1419
3
Byrd
 
JC
Mrozek
 
K
Dodge
 
RK
, et al. 
Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461).
Blood
2002
, vol. 
100
 
13
(pg. 
4325
-
4336
)
4
Grimwade
 
D
Hills
 
RK
Moorman
 
AV
, et al. 
Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials.
Blood
2010
, vol. 
116
 
3
(pg. 
354
-
365
)
5
Slovak
 
ML
Kopecky
 
KJ
Cassileth
 
PA
, et al. 
Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study.
Blood
2000
, vol. 
96
 
13
(pg. 
4075
-
4083
)
6
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
)
7
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
)
8
Mrozek
 
K
Marcucci
 
G
Paschka
 
P
Whitman
 
SP
Bloomfield
 
CD
Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a prognostically prioritized molecular classification?
Blood
2007
, vol. 
109
 
2
(pg. 
431
-
448
)
9
Kottaridis
 
PD
Gale
 
RE
Frew
 
ME
, et al. 
The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials.
Blood
2001
, vol. 
98
 
6
(pg. 
1752
-
1759
)
10
Thiede
 
C
Steudel
 
C
Mohr
 
B
, et al. 
Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis.
Blood
2002
, vol. 
99
 
12
(pg. 
4326
-
4335
)
11
Schlenk
 
RF
Dohner
 
K
Krauter
 
J
, et al. 
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia.
N Engl J Med
2008
, vol. 
358
 
18
(pg. 
1909
-
1918
)
12
Fröhling
 
S
Schlenk
 
RF
Breitruck
 
J
, et al. 
Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm.
Blood
2002
, vol. 
100
 
13
(pg. 
4372
-
4380
)
13
Gale
 
RE
Green
 
C
Allen
 
C
, et al. 
The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia.
Blood
2008
, vol. 
111
 
5
(pg. 
2776
-
2784
)
14
Yanada
 
M
Matsuo
 
K
Suzuki
 
T
Kiyoi
 
H
Naoe
 
T
Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a meta-analysis.
Leukemia
2005
, vol. 
19
 
8
(pg. 
1345
-
1349
)
15
Mead
 
AJ
Linch
 
DC
Hills
 
RK
Wheatley
 
K
Burnett
 
AK
Gale
 
RE
FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia.
Blood
2007
, vol. 
110
 
4
(pg. 
1262
-
1270
)
16
Whitman
 
SP
Ruppert
 
AS
Radmacher
 
MD
, et al. 
FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications.
Blood
2008
, vol. 
111
 
3
(pg. 
1552
-
1559
)
17
Döhner
 
K
Schlenk
 
RF
Habdank
 
M
, et al. 
Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations.
Blood
2005
, vol. 
106
 
12
(pg. 
3740
-
3746
)
18
Schnittger
 
S
Schoch
 
C
Kern
 
W
, et al. 
Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype.
Blood
2005
, vol. 
106
 
12
(pg. 
3733
-
3739
)
19
Thiede
 
C
Koch
 
S
Creutzig
 
E
, et al. 
Prevalence and prognostic impact of NPM1 mutations in 1485 adult patients with acute myeloid leukemia (AML).
Blood
2006
, vol. 
107
 
10
(pg. 
4011
-
4020
)
20
Rollig
 
C
Thiede
 
C
Gramatzki
 
M
, et al. 
A novel prognostic model in elderly patients with acute myeloid leukemia: results of 909 patients entered into the prospective AML96 trial.
Blood
, vol. 
116
 
6
(pg. 
971
-
978
)
21
Preudhomme
 
C
Sagot
 
C
Boissel
 
N
, et al. 
Favorable prognostic significance of CEBPA mutations in patients with de novo acute myeloid leukemia: a study from the Acute Leukemia French Association (ALFA).
Blood
2002
, vol. 
100
 
8
(pg. 
2717
-
2723
)
22
Fröhling
 
S
Schlenk
 
RF
Stolze
 
I
, et al. 
CEBPA mutations in younger adults with acute myeloid leukemia and normal cytogenetics: prognostic relevance and analysis of cooperating mutations.
J Clin Oncol
2004
, vol. 
22
 
4
(pg. 
624
-
633
)
23
Marcucci
 
G
Maharry
 
K
Radmacher
 
MD
, et al. 
Prognostic significance of, and gene and microRNA expression signatures associated with, CEBPA mutations in cytogenetically normal acute myeloid leukemia with high-risk molecular features: a Cancer and Leukemia Group B Study.
J Clin Oncol
2008
, vol. 
26
 
31
(pg. 
5078
-
5087
)
24
Green
 
CL
Koo
 
KK
Hills
 
RK
Burnett
 
AK
Linch
 
DC
Gale
 
RE
Prognostic significance of CEBPA mutations in a large cohort of younger adult patients with acute myeloid leukemia: impact of double CEBPA mutations and the interaction with FLT3 and NPM1 mutations.
J Clin Oncol
2010
, vol. 
28
 
16
(pg. 
2739
-
2747
)
25
Vardiman
 
JW
Thiele
 
J
Arber
 
DA
, et al. 
The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes.
Blood
2009
, vol. 
114
 
5
(pg. 
937
-
951
)
26
Wouters
 
BJ
Lowenberg
 
B
Erpelinck-Verschueren
 
CA
van Putten
 
WL
Valk
 
PJ
Delwel
 
R
Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome.
Blood
2009
, vol. 
113
 
13
(pg. 
3088
-
3091
)
27
Pabst
 
T
Eyholzer
 
M
Fos
 
J
Mueller
 
BU
Heterogeneity within AML with CEBPA mutations; only CEBPA double mutations, but not single CEBPA mutations are associated with favourable prognosis.
Br J Cancer
2009
, vol. 
100
 
8
(pg. 
1343
-
1346
)
28
Boissel
 
N
Nibourel
 
O
Renneville
 
A
, et al. 
Prognostic impact of isocitrate dehydrogenase enzyme isoforms 1 and 2 mutations in acute myeloid leukemia: a study by the acute leukemia French association group.
J Clin Oncol
2010
, vol. 
28
 
23
(pg. 
3717
-
3723
)
29
Paschka
 
P
Schlenk
 
RF
Gaidzik
 
VI
, et al. 
IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication.
J Clin Oncol
2010
, vol. 
28
 
22
(pg. 
3636
-
3643
)
30
Ley
 
TJ
Ding
 
L
Walter
 
MJ
, et al. 
DNMT3A Mutations in Acute Myeloid Leukemia.
N Engl J Med
2010
, vol. 
363
 
25
(pg. 
2424
-
2433
)
31
Bullinger
 
L
Dohner
 
K
Bair
 
E
, et al. 
Use of gene-expression profiling to identify prognostic subclasses in adult acute myeloid leukemia.
N Engl J Med
2004
, vol. 
350
 
16
(pg. 
1605
-
1616
)
32
Valk
 
PJ
Verhaak
 
RG
Beijen
 
MA
, et al. 
Prognostically useful gene-expression profiles in acute myeloid leukemia.
N Engl J Med
2004
, vol. 
350
 
16
(pg. 
1617
-
1628
)
33
Radmacher
 
MD
Marcucci
 
G
Ruppert
 
AS
, et al. 
Independent confirmation of a prognostic gene-expression signature in adult acute myeloid leukemia with a normal karyotype: a Cancer and Leukemia Group B study.
Blood
2006
, vol. 
108
 
5
(pg. 
1677
-
1683
)
34
Garzon
 
R
Volinia
 
S
Liu
 
CG
, et al. 
MicroRNA signatures associated with cytogenetics and prognosis in acute myeloid leukemia.
Blood
2008
, vol. 
111
 
6
(pg. 
3183
-
3189
)
35
Tallman
 
MS
Altman
 
JK
How I treat acute promyelocytic leukemia.
Blood
2009
, vol. 
114
 
25
(pg. 
5126
-
5135
)
36
Marcucci
 
G
Mrozek
 
K
Ruppert
 
AS
, et al. 
Prognostic factors and outcome of core binding factor acute myeloid leukemia patients with t(8;21) differ from those of patients with inv(16): a Cancer and Leukemia Group B study.
J Clin Oncol
2005
, vol. 
23
 
24
(pg. 
5705
-
5717
)
37
Cairoli
 
R
Beghini
 
A
Grillo
 
G
, et al. 
Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study.
Blood
2006
, vol. 
107
 
9
(pg. 
3463
-
3468
)
38
Schnittger
 
S
Kohl
 
TM
Haferlach
 
T
, et al. 
KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival.
Blood
2006
, vol. 
107
 
5
(pg. 
1791
-
1799
)
39
Care
 
RS
Valk
 
PJ
Goodeve
 
AC
, et al. 
Incidence and prognosis of c-KIT and FLT3 mutations in core binding factor (CBF) acute myeloid leukaemias.
Br J Haematol
2003
, vol. 
121
 
5
(pg. 
775
-
777
)
40
Paschka
 
P
Marcucci
 
G
Ruppert
 
AS
, et al. 
Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B Study.
J Clin Oncol
2006
, vol. 
24
 
24
(pg. 
3904
-
3911
)
41
Byrd
 
JC
Dodge
 
RK
Carroll
 
A
, et al. 
Patients with t(8;21)(q22;q22) and acute myeloid leukemia have superior failure-free and overall survival when repetitive cycles of high-dose cytarabine are administered.
J Clin Oncol
1999
, vol. 
17
 
12
(pg. 
3767
-
3775
)
42
Byrd
 
JC
Ruppert
 
AS
Mrozek
 
K
, et al. 
Repetitive cycles of high-dose cytarabine benefit patients with acute myeloid leukemia and inv(16)(p13q22) or t(16;16)(p13;q22): results from CALGB 8461.
J Clin Oncol
2004
, vol. 
22
 
6
(pg. 
1087
-
1094
)
43
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
)
44
Rowe
 
JM
Optimal induction and post-remission therapy for AML in first remission.
Hematology Am Soc Hematol Educ Program
2009
(pg. 
396
-
405
)
45
Yanada
 
M
Matsuo
 
K
Emi
 
N
Naoe
 
T
Efficacy of allogeneic hematopoietic stem cell transplantation depends on cytogenetic risk for acute myeloid leukemia in first disease remission: a metaanalysis.
Cancer
2005
, vol. 
103
 
8
(pg. 
1652
-
1658
)
46
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
)
47
Basara
 
N
Schulze
 
A
Wedding
 
U
, et al. 
Early related or unrelated haematopoietic cell transplantation results in higher overall survival and leukaemia-free survival compared with conventional chemotherapy in high-risk acute myeloid leukaemia patients in first complete remission.
Leukemia
2009
, vol. 
23
 
4
(pg. 
635
-
640
)
48
Gupta
 
V
Tallman
 
MS
He
 
W
, et al. 
Comparable survival after HLA-well-matched unrelated or matched sibling donor transplantation for acute myeloid leukemia in first remission with unfavorable cytogenetics at diagnosis.
Blood
2010
, vol. 
116
 
11
(pg. 
1839
-
1848
)
49
Walter
 
RB
Pagel
 
JM
Gooley
 
TA
, et al. 
Comparison of matched unrelated and matched related donor myeloablative hematopoietic cell transplantation for adults with acute myeloid leukemia in first remission.
Leukemia
2010
, vol. 
24
 
7
(pg. 
1276
-
1282
)
50
Laughlin
 
MJ
Eapen
 
M
Rubinstein
 
P
, et al. 
Outcomes after transplantation of cord blood or bone marrow from unrelated donors in adults with leukemia.
N Engl J Med
2004
, vol. 
351
 
22
(pg. 
2265
-
2275
)
51
Rocha
 
V
Labopin
 
M
Sanz
 
G
, et al. 
Transplants of umbilical-cord blood or bone marrow from unrelated donors in adults with acute leukemia.
N Engl J Med
2004
, vol. 
351
 
22
(pg. 
2276
-
2285
)
52
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
)
53
Brunstein
 
CG
Gutman
 
JA
Weisdorf
 
DJ
, et al. 
Allogeneic hematopoietic cell transplantation for hematological malignancy: relative risks and benefits of double umbilical cord blood.
Blood
2010
, vol. 
116
 
22
(pg. 
4693
-
4699
)
54
Atsuta
 
Y
Suzuki
 
R
Nagamura-Inoue
 
T
, et al. 
Disease-specific analyses of unrelated cord blood transplantation compared with unrelated bone marrow transplantation in adult patients with acute leukemia.
Blood
2009
, vol. 
113
 
8
(pg. 
1631
-
1638
)
55
Ringden
 
O
Pavletic
 
SZ
Anasetti
 
C
, et al. 
The graft-versus-leukemia effect using matched unrelated donors is not superior to HLA-identical siblings for hematopoietic stem cell transplantation.
Blood
2009
, vol. 
113
 
13
(pg. 
3110
-
3118
)
56
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
)
57
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
)
58
Moore
 
J
Nivison-Smith
 
I
Goh
 
K
, et al. 
Equivalent survival for sibling and unrelated donor allogeneic stem cell transplantation for acute myelogenous leukemia.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
5
(pg. 
601
-
607
)
59
Schetelig
 
J
Bornhauser
 
M
Schmid
 
C
, et al. 
Matched unrelated or matched sibling donors result in comparable survival after allogeneic stem-cell transplantation in elderly patients with acute myeloid leukemia: a report from the cooperative German Transplant Study Group.
J Clin Oncol
2008
, vol. 
26
 
32
(pg. 
5183
-
5191
)
60
Breems
 
DA
Van Putten
 
WL
Huijgens
 
PC
, et al. 
Prognostic index for adult patients with acute myeloid leukemia in first relapse.
J Clin Oncol
2005
, vol. 
23
 
9
(pg. 
1969
-
1978
)
61
Kurosawa
 
S
Yamaguchi
 
T
Miyawaki
 
S
, et al. 
Prognostic factors and outcomes of adult patients with acute myeloid leukemia after first relapse.
Haematologica
2010
, vol. 
95
 
11
(pg. 
1857
-
1864
)
62
Biggs
 
JC
Horowitz
 
MM
Gale
 
RP
, et al. 
Bone marrow transplants may cure patients with acute leukemia never achieving remission with chemotherapy.
Blood
1992
, vol. 
80
 
4
(pg. 
1090
-
1093
)
63
Brown
 
RA
Wolff
 
SN
Fay
 
JW
, et al. 
High-dose etoposide, cyclophosphamide, and total body irradiation with allogeneic bone marrow transplantation for patients with acute myeloid leukemia in untreated first relapse: a study by the North American Marrow Transplant Group.
Blood
1995
, vol. 
85
 
5
(pg. 
1391
-
1395
)
64
Sierra
 
J
Storer
 
B
Hansen
 
JA
, et al. 
Transplantation of marrow cells from unrelated donors for treatment of high-risk acute leukemia: the effect of leukemic burden, donor HLA-matching, and marrow cell dose.
Blood
1997
, vol. 
89
 
11
(pg. 
4226
-
4235
)
65
Michallet
 
M
Thomas
 
X
Vernant
 
JP
, et al. 
Long-term outcome after allogeneic hematopoietic stem cell transplantation for advanced stage acute myeloblastic leukemia: a retrospective study of 379 patients reported to the Societe Francaise de Greffe de Moelle (SFGM).
Bone Marrow Transplant
2000
, vol. 
26
 
11
(pg. 
1157
-
1163
)
66
Greinix
 
HT
Reiter
 
E
Keil
 
F
, et al. 
Leukemia-free survival and mortality in patients with refractory or relapsed acute leukemia given marrow transplants from sibling and unrelated donors.
Bone Marrow Transplant
1998
, vol. 
21
 
7
(pg. 
673
-
678
)
67
Wong
 
R
Shahjahan
 
M
Wang
 
X
, et al. 
Prognostic factors for outcomes of patients with refractory or relapsed acute myelogenous leukemia or myelodysplastic syndromes undergoing allogeneic progenitor cell transplantation.
Biol Blood Marrow Transplant
2005
, vol. 
11
 
2
(pg. 
108
-
114
)
68
Oyekunle
 
AA
Kroger
 
N
Zabelina
 
T
, et al. 
Allogeneic stem-cell transplantation in patients with refractory acute leukemia: a long-term follow-up.
Bone Marrow Transplant
2006
, vol. 
37
 
1
(pg. 
45
-
50
)
69
Duval
 
M
Klein
 
JP
He
 
W
, et al. 
Hematopoietic stem-cell transplantation for acute leukemia in relapse or primary induction failure.
J Clin Oncol
2010
, vol. 
28
 
23
(pg. 
3730
-
3738
)
70
Alyea
 
EP
Kim
 
HT
Ho
 
V
, et al. 
Impact of conditioning regimen intensity on outcome of allogeneic hematopoietic cell transplantation for advanced acute myelogenous leukemia and myelodysplastic syndrome.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
10
(pg. 
1047
-
1055
)
71
Schmid
 
C
Schleuning
 
M
Schwerdtfeger
 
R
, et al. 
Long-term survival in refractory acute myeloid leukemia after sequential treatment with chemotherapy and reduced-intensity conditioning for allogeneic stem cell transplantation.
Blood
2006
, vol. 
108
 
3
(pg. 
1092
-
1099
)
72
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
)
73
Pagel
 
JM
Gooley
 
TA
Rajendran
 
J
, et al. 
Allogeneic hematopoietic cell transplantation after conditioning with 131I-anti-CD45 antibody plus fludarabine and low-dose total body irradiation for elderly patients with advanced acute myeloid leukemia or high-risk myelodysplastic syndrome.
Blood
2009
, vol. 
114
 
27
(pg. 
5444
-
5453
)
74
Rosenthal
 
J
Wong
 
J
Stein
 
A
, et al. 
Phase I/II trial of total marrow and lymph node irradiation to augment reduced intensity transplant for advanced hematological malignancies.
Blood
2011
, vol. 
117
 
1
(pg. 
309
-
315
)
75
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
)
76
Kolb
 
HJ
Graft-versus-leukemia effects of transplantation and donor lymphocytes.
Blood
2008
, vol. 
112
 
12
(pg. 
4371
-
4383
)
77
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
2011
, vol. 
116
 
23
(pg. 
5420
-
5431
)
78
Leone
 
G
Mele
 
L
Pulsoni
 
A
Equitani
 
F
Pagano
 
L
The incidence of secondary leukemias.
Haematologica
1999
, vol. 
84
 
10
(pg. 
937
-
945
)
79
Litzow
 
MR
Tarima
 
S
Perez
 
WS
, et al. 
Allogeneic transplantation for therapy-related myelodysplastic syndrome and acute myeloid leukemia.
Blood
2010
, vol. 
115
 
9
(pg. 
1850
-
1857
)
80
Kroger
 
N
Brand
 
R
van Biezen
 
A
, et al. 
Risk factors for therapy-related myelodysplastic syndrome and acute myeloid leukemia treated with allogeneic stem cell transplantation.
Haematologica
2009
, vol. 
94
 
4
(pg. 
542
-
549
)
81
Yakoub-Agha
 
I
de La Salmoniere
 
P
Ribaud
 
P
, et al. 
Allogeneic bone marrow transplantation for therapy-related myelodysplastic syndrome and acute myeloid leukemia: a long-term study of 70 patients-report of the French society of bone marrow transplantation.
J Clin Oncol
2000
, vol. 
18
 
5
(pg. 
963
-
971
)
82
Chang
 
C
Storer
 
BE
Scott
 
BL
, et al. 
Hematopoietic cell transplantation in patients with myelodysplastic syndrome or acute myeloid leukemia arising from myelodysplastic syndrome: similar outcomes in patients with de novo disease and disease following prior therapy or antecedent hematologic disorders.
Blood
2007
, vol. 
110
 
4
(pg. 
1379
-
1387
)
83
Armand
 
P
Kim
 
HT
DeAngelo
 
DJ
, et al. 
Impact of cytogenetics on outcome of de novo and therapy-related AML and MDS after allogeneic transplantation.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
6
(pg. 
655
-
664
)
84
Schoch
 
C
Kern
 
W
Schnittger
 
S
Hiddemann
 
W
Haferlach
 
T
Karyotype is an independent prognostic parameter in therapy-related acute myeloid leukemia (t-AML): an analysis of 93 patients with t-AML in comparison to 1091 patients with de novo AML.
Leukemia
2004
, vol. 
18
 
1
(pg. 
120
-
125
)
85
Gustafson
 
SA
Lin
 
P
Chen
 
SS
, et al. 
Therapy-related acute myeloid leukemia with t(8;21) (q22;q22) shares many features with de novo acute myeloid leukemia with t(8;21)(q22;q22) but does not have a favorable outcome.
Am J Clin Pathol
2009
, vol. 
131
 
5
(pg. 
647
-
655
)
86
Beaumont
 
M
Sanz
 
M
Carli
 
PM
, et al. 
Therapy-related acute promyelocytic leukemia.
J Clin Oncol
2003
, vol. 
21
 
11
(pg. 
2123
-
2137
)
87
Kantarjian
 
H
Ravandi
 
F
O'Brien
 
S
, et al. 
Intensive chemotherapy does not benefit most older patients (age 70 years or older) with acute myeloid leukemia.
Blood
2010
, vol. 
116
 
22
(pg. 
4422
-
4429
)
88
Wallen
 
H
Gooley
 
TA
Deeg
 
HJ
, et al. 
Ablative allogeneic hematopoietic cell transplantation in adults 60 years of age and older.
J Clin Oncol
2005
, vol. 
23
 
15
(pg. 
3439
-
3446
)
89
Gupta
 
V
Daly
 
A
Lipton
 
JH
, et al. 
Nonmyeloablative stem cell transplantation for myelodysplastic syndrome or acute myeloid leukemia in patients 60 years or older.
Biol Blood Marrow Transplant
2005
, vol. 
11
 
10
(pg. 
764
-
772
)
90
Spyridonidis
 
A
Bertz
 
H
Ihorst
 
G
Grullich
 
C
Finke
 
J
Hematopoietic cell transplantation from unrelated donors as an effective therapy for older patients (≥60 years) with active myeloid malignancies.
Blood
2005
, vol. 
105
 
10
(pg. 
4147
-
4148
)
91
Bertz
 
H
Potthoff
 
K
Finke
 
J
Allogeneic stem-cell transplantation from related and unrelated donors in older patients with myeloid leukemia.
J Clin Oncol
2003
, vol. 
21
 
8
(pg. 
1480
-
1484
)
92
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
)
93
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
)
94
Deschler
 
B
Binek
 
K
Ihorst
 
G
, et al. 
Prognostic factor and quality of life analysis in 160 patients aged > or =60 years with hematologic neoplasias treated with allogeneic hematopoietic cell transplantation.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
7
(pg. 
967
-
975
)
95
Koreth
 
J
Aldridge
 
J
Kim
 
HT
, et al. 
Reduced-intensity conditioning hematopoietic stem cell transplantation in patients over 60 years: hematologic malignancy outcomes are not impaired in advanced age.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
6
(pg. 
792
-
800
)
96
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
)
97
Farag
 
S
Maharry
 
K
Perez
 
WS
, et al. 
Allogeneic hematopoietic stem cell transplantation (HCT) compared with chemotherapy only in acute myeloid leukemia (AML) patients 60 years and older: a Center for International Blood and Marrow Transplantation Research (CIBMTR)/Cancer and Leukemia Group B (CALGB) study [abstract].
Blood
2009
, vol. 
114
 
22
pg. 
274
  
Abstract 657
98
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
)
99
Khabori
 
MA
El-Emary
 
M
Xu
 
W
, et al. 
Impact of intensity of conditioning therapy in patients aged 40-60 years with AML/myelodysplastic syndrome undergoing allogeneic transplantation [published online ahead of print July 12, 2010].
Bone Marrow Transplant
 
100
Massenkeil
 
G
Nagy
 
M
Neuburger
 
S
, et al. 
Survival after reduced-intensity conditioning is not inferior to standard high-dose conditioning before allogeneic haematopoietic cell transplantation in acute leukaemias.
Bone Marrow Transplant
2005
, vol. 
36
 
8
(pg. 
683
-
689
)
101
Ringden
 
O
Labopin
 
M
Ehninger
 
G
, et al. 
Reduced intensity conditioning compared with myeloablative conditioning using unrelated donor transplants in patients with acute myeloid leukemia.
J Clin Oncol
2009
, vol. 
27
 
27
(pg. 
4570
-
4577
)
102
Scott
 
BL
Sandmaier
 
BM
Storer
 
B
, et al. 
Myeloablative vs nonmyeloablative allogeneic transplantation for patients with myelodysplastic syndrome or acute myelogenous leukemia with multilineage dysplasia: a retrospective analysis.
Leukemia
2006
, vol. 
20
 
1
(pg. 
128
-
135
)
103
Flynn
 
CM
Hirsch
 
B
Defor
 
T
, et al. 
Reduced intensity compared with high dose conditioning for allotransplantation in acute myeloid leukemia and myelodysplastic syndrome: a comparative clinical analysis.
Am J Hematol
2007
, vol. 
82
 
10
(pg. 
867
-
872
)
104
Aoudjhane
 
M
Labopin
 
M
Gorin
 
NC
, et al. 
Comparative outcome of reduced intensity and myeloablative conditioning regimen in HLA identical sibling allogeneic haematopoietic stem cell transplantation for patients older than 50 years of age with acute myeloblastic leukaemia: a retrospective survey from the Acute Leukemia Working Party (ALWP) of the European group for Blood and Marrow Transplantation (EBMT).
Leukemia
2005
, vol. 
19
 
12
(pg. 
2304
-
2312
)
105
Luger
 
S
Ringden
 
O
Perez
 
WS
, et al. 
Similar outcomes using myeloablative versus reduced intensity and non-myeloablative allogeneic transplant preparative regimens for AML or MDS: from the Center for International Blood and Marrow Transplant Research [abstract].
Blood
2008
, vol. 
112
 
11
pg. 
136
  
Abstract 348
106
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
)
107
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
)
108
Sorror
 
ML
Giralt
 
S
Sandmaier
 
BM
, et al. 
Hematopoietic cell transplantation specific comorbidity index as an outcome predictor for patients with acute myeloid leukemia in first remission: combined FHCRC and MDACC experiences.
Blood
2007
, vol. 
110
 
13
(pg. 
4606
-
4613
)
109
Parimon
 
T
Au
 
DH
Martin
 
PJ
Chien
 
JW
A risk score for mortality after allogeneic hematopoietic cell transplantation.
Ann Intern Med
2006
, vol. 
144
 
6
(pg. 
407
-
414
)
110
Mohty
 
M
de Lavallade
 
H
Ladaique
 
P
, et al. 
The role of reduced intensity conditioning allogeneic stem cell transplantation in patients with acute myeloid leukemia: a donor vs no donor comparison.
Leukemia
2005
, vol. 
19
 
6
(pg. 
916
-
920
)
111
Mortimer
 
J
Blinder
 
MA
Schulman
 
S
, et al. 
Relapse of acute leukemia after marrow transplantation: natural history and results of subsequent therapy.
J Clin Oncol
1989
, vol. 
7
 
1
(pg. 
50
-
57
)
112
Radich
 
JP
Sanders
 
JE
Buckner
 
CD
, et al. 
Second allogeneic marrow transplantation for patients with recurrent leukemia after initial transplant with total-body irradiation-containing regimens.
J Clin Oncol
1993
, vol. 
11
 
2
(pg. 
304
-
313
)
113
Hosing
 
C
Saliba
 
RM
Shahjahan
 
M
, et al. 
Disease burden may identify patients more likely to benefit from second allogeneic hematopoietic stem cell transplantation to treat relapsed acute myelogenous leukemia.
Bone Marrow Transplant
2005
, vol. 
36
 
2
(pg. 
157
-
162
)
114
Bosi
 
A
Laszlo
 
D
Labopin
 
M
, et al. 
Second allogeneic bone marrow transplantation in acute leukemia: results of a survey by the European Cooperative Group for Blood and Marrow Transplantation.
J Clin Oncol
2001
, vol. 
19
 
16
(pg. 
3675
-
3684
)
115
Michallet
 
M
Tanguy
 
ML
Socie
 
G
, et al. 
Second allogeneic haematopoietic stem cell transplantation in relapsed acute and chronic leukaemias for patients who underwent a first allogeneic bone marrow transplantation: a survey of the Societe Francaise de Greffe de moelle (SFGM).
Br J Haematol
2000
, vol. 
108
 
2
(pg. 
400
-
407
)
116
Eapen
 
M
Giralt
 
SA
Horowitz
 
MM
, et al. 
Second transplant for acute and chronic leukemia relapsing after first HLA-identical sibling transplant.
Bone Marrow Transplant
2004
, vol. 
34
 
8
(pg. 
721
-
727
)
117
Levine
 
JE
Braun
 
T
Penza
 
SL
, et al. 
Prospective trial of chemotherapy and donor leukocyte infusions for relapse of advanced myeloid malignancies after allogeneic stem-cell transplantation.
J Clin Oncol
2002
, vol. 
20
 
2
(pg. 
405
-
412
)
118
Choi
 
SJ
Lee
 
JH
Kim
 
S
, et al. 
Treatment of relapsed acute myeloid leukemia after allogeneic bone marrow transplantation with chemotherapy followed by G-CSF-primed donor leukocyte infusion: a high incidence of isolated extramedullary relapse.
Leukemia
2004
, vol. 
18
 
11
(pg. 
1789
-
1797
)
119
Schmid
 
C
Labopin
 
M
Nagler
 
A
, et al. 
Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party.
J Clin Oncol
2007
, vol. 
25
 
31
(pg. 
4938
-
4945
)
120
Kollman
 
C
Weis
 
T
Switzer
 
GE
, et al. 
Non-HLA barriers to unrelated donor stem cell transplantation.
Bone Marrow Transplant
2001
, vol. 
27
 
6
(pg. 
581
-
587
)
121
Majhail
 
NS
Omondi
 
NA
Denzen
 
E
Murphy
 
EA
Rizzo
 
JD
Access to hematopoietic cell transplantation in the United States.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
8
(pg. 
1070
-
1075
)
Sign in via your Institution