The choice of either induction or postremission therapy for adults with acute myeloid leukemia is still largely based on the “one size fits all” principle. Moreover, pretreatment prognostic parameters, especially chromosome and gene abnormalities, may fail in predicting individual patient outcome. Measurement of minimal residual disease (MRD) is nowadays recognized as a potential critical tool to assess the quality of response after chemotherapy and to plan postremission strategies that are, therefore, driven by the individual risk of relapse. PCR and multiparametric flow cytometry have become the most popular methods to investigate MRD because they have been established as sensitive and specific enough to allow MRD to be studied serially. In the present review, we examine the evidence supporting the appropriateness of incorporating MRD detection into the AML risk assessment process. A comprehensive prognostic algorithm, generated by combining pretreatment cytogenetics/genetics and posttreatment MRD determination, should promote advances in development of personalized therapeutic approaches.

In adult patients with acute myeloid leukemia (AML), intensive chemotherapy achieves complete remission (CR) rates ranging from 50% to 80%. Despite these encouraging results, the majority of responding patients will eventually relapse, with only 30% to 40% of young and less than 20% of elderly patients being long-term survivors.1,,,5  Advances in biologic characterization are expected to provide a more proper risk stratification of AML, allowing delivery of treatments proportional to the real aggressiveness of disease. In this view, cytogenetic abnormalities represent the most reliable prognosticator in adult AML.6,,,10  Identification of specific gene abnormalities (eg, FLT3, NPM1, CEBPA, and DMNT3A) has further improved prognostic allocation of patients with AML, especially within homogeneous karyotypic groups (ie, intermediate karyotypes or favorable karyotypes), where the possible concomitant mutations of KIT, occurring in the context of core binding factor (CBF) translocations, confer a negative prognosis.11,,,15  A proper assignment to low- or high-risk category is a critical step in the therapeutic decision-making process of patients with AML. Indeed, patients belonging to the low-risk category would not benefit from routine use of allogeneic stem cell transplantation (ASCT) in first CR because any advantages in terms of reduced relapse incidence will be counterbalanced by procedure-related morbidity and mortality.16  On the other hand, missing early identification of high-risk patients might hamper or delay the timely use of intensive treatments, such as ASCT.17  However, it is also well recognized that cytogenetic/genetic signature cannot always reliably predict outcome in individual patients; indeed, approximately 40% to 50% of those with favorable karyotype will eventually experience a relapse. In this view, measure of minimal residual disease (MRD) promises to be an efficient tool to establish on an individual basis the patient's leukemia's susceptibility to treatment, enhancing delivery of risk-adapted therapies.18,19  Despite these premises, the systematic applicability of MRD analysis in AML is not yet accomplished. For cases with a genetic signature that accounts for 60% to 70% of AML, the molecular approach still suffers from lack of standardized assays and cut-offs. Similarly, although flow cytometry can potentially investigate more than 85% of AML, its widespread applicability is complicated by lack of standardized procedure. The present review will focus on the technical issues and clinical relevance of MRD determination in non-M3 AML. In particular, we will try to outline how MRD assessment might improve risk stratification.

Clonal chromosome alterations are detected in AML in more than 50% of adults and are universally considered the strongest predictor of duration of response and overall survival (OS). Based on cytogenetics, patients are generally stratified into 3 risk groups: favorable, intermediate, and unfavorable.6,,,10  Accordingly, patients falling into the category of favorable risk karyotype (F-RK) can expect an approximate 65% to 70% likelihood of cure, those who have an intermediate risk karyotype (I-RK) an approximate 40% chance of long-term disease-free survival (DFS), whereas patients belonging to the unfavorable risk karyotype (U-RK) category have a very dismal outcome, with less than 5% to 10% becoming long-term survivors.6,,,10  However, it has become clear that cytogenetic risk allocation may help guide therapeutic decision, particularly for those who are at the extremes (F-RK or U-RK), whereas, because of the heterogeneity of this category, for patients within I-RK the therapeutic decision may be problematic. Indeed, 40% to 50% of patients categorized in this group lack any clonal abnormalities on standard cytogenetic analysis. Furthermore, together with cytogenetically normal AML (CN-AML), the I-RK also includes a miscellany of different structural and numerical changes that are too infrequent to be reliably assigned a prognostic significance and are not encompassed in the 2 other risk groups. In recent years, various molecular markers have been identified, allowing to dissect further cytogenetically defined subsets.20  This is critical to risk assessment of patients with CN-AML (Table 1). Although the expectations of using genetics to guide therapy were enormous, the delivery to the clinics has made slow progress and the therapeutic strategy for most patients still remains controversial. On one side, risk assessment based on pretreatment factors composes a large group of patients with broad differences in terms of risk of relapse. On the other side, fitting the increasing number of new gene abnormalities into a practical, prospective, decision-making algorithm is a very difficult task. Indeed, the excess of dissection in homogeneous genetic subsets may lead to a plethora of subgroups failing to provide physicians with an adequate statistical power (Figure 1). A reasonable way to improve outcome prediction in AML might be attained by combining pretreatment and posttreatment parameters into a common prognostic algorithm. Such a hypothesis found acceptance in a seminal manuscript published by Wheatley et al who generated a prognostic scoring system by integrating pretreatment cytogenetics and achievement of CR after 1 or 2 cycles of induction therapy.21  Patients with U-RK who entered CR after 2 induction courses had the worst outcome. The design of the analysis represented a first attempt to put together into a risk stratification algorithm 2 different classes of prognostic parameters: pretreatment factors and indicators of the quality of response after a chemotherapy-induced morphologic CR. Nowadays, such a working hypothesis can be developed further, thanks to the availability of techniques, such as PCR and multiparametric flow cytometry (MPFC). These techniques can reliably detect, at high sensitivity, leukemic cells at submicroscopic level, thus offering the opportunity to investigate MRD.18,19  Assessment of MRD promises to be a powerful and accurate tool to refine patients' risk category assignment as initially established on the basis of the sole cytogenetic/genetic findings.17,22,23 

Table 1

Gene mutations occurring in cytogenetically normal acute myeloid leukemia

GeneIncidenceBiologic featuresImpact on clinical outcome and targeted therapy opportunities
FLT3    
ITD Found in 28%-34% of CN-AML Member of the class III receptor tyrosine kinase family Consistently associated with inferior outcome in terms of OS, RFS, and EFS33,34  
  In-frame mutations, mostly in exons 14 and 15 of the juxtamembrane domain Constitutively active FLT3 molecules are targets for specific molecular inhibitors (phase 2 or 3 trials) 
  Level of mutant allele probably of importance  
  Homozygous FLT3 mutations as a result of mitotic recombination leading to partial UPD  
TKD Found in 11%-14% of CN-AML In-frame point mutations of the TKD codon 835 and 836 Meta-analysis suggesting a negative prognostic impact32  
  Level of mutant allele may be of importance More recently associated with better OS31  
   High-level mutants associated with improved OS30  
NPM1 Found in 25%-35% of AML(45%-62% of CN-AML) The mutational event modifies specific nucleoli binding and nuclear export signal motifs coded by the exon 12 and determines an abnormal cytoplasmic localization of nucleophosmin NPM1mut/FLT3-ITD genotype associated with a favorable OS, RFS, and CR rate38,3940  
  Associated with presenting clinical and laboratory features, such as female sex, higher BM blast counts, and LDH levels, as well as high CD33 but low or absent CD34 levels Patients with NPM1mut/FLT3-ITD genotype may not benefit from MRD allogeneic transplantation in first CR11  
  Associated with FLT3-ITD and TKD mutations (40% of cases) Pharmacologic modulation of the RA-signaling pathway has been suggested as a therapeutic option11  
  NPM1 acts as a corepressor in RA-associated transcriptional regulation41   
CEBPA Found in 15%-20% of CN-AML Transcription factor mediating lineage specification and differentiation of multipotent myeloid progenitors into mature neutrophils Associated with higher CR rate and better RFS and OS45,46  
  Concurrent mutations (ie, FLT3-ITD and NPM1) were virtually not present in cases with CEBPA–biallelic mutation compared with CEBPA–single allelic mutation44  The presence of CEBPA biallelic (bm) but not single allelic mutation (sm) is an independent factor for a favorable outcome44  
MLL PTD found in 5%-11% of CN-AML Usually involves exons 5-11 or, less frequently, exons 5-12 Associated with shorter CR duration or inferior RFS and EFS60  
  MLL wild-type allele is silenced, probably as a result of differential DNA methylation and histone modifications Allogeneic transplantation may improve outcome11  
  Rationale for the use of DNA methyltransferase and/or histone deacetylase inhibitors based on in vitro data  
IDH1/2 Found in 15% of AML Correlated with accumulation of the cancer-associated metabolite 2-hydroxyglutarate IDH1 mutations identify patients at higher risk of relapse within the favorable risk category of NPM1 mutated AML50,,53  
  IDH2 gene encodes a mitochondrial protein homologous to IDH1 that also catalyzes isocytratecarboxylation IDH2 mutations get along with specific pretreatment features (low WBC, lack of other mutations, and primary resistance to chemotherapy)50,,53  
  IDH1 and IDH2 mutations are mutually exclusive  
DNMT3A Found in 22% of AML. Somatic mutation in DNMT3A, encoding a DNA methyltransferase Independent determinant of dismal prognosis both in the overall study population and, significantly, within high-risk patients (FLT3-ITD, age > 60 years)54  
  Highly enriched in the group of patients with an intermediate-risk cytogenetic profile (56/66, 33.7%) Only ASCT provided a significant benefit54  
  Absent in the subgroup of 79 patients carrying a favorable-risk cytogenetic  
  A perturbation in the methylation of specific DNA sequences has been postulated  
BAALC overexpression Rare Expressed in neuroectoderm-derived tissues and hematopoietic precursors Significantly worse CR rates and shorter DFS, EFS, and OS than patients with low expression55  
  Detected in a subset of karyotypically normal adults < 60 years with de novo AML SCT in first CR might overcome the adverse prognostic effect of high BAALC expression55  
ERG overexpression Rare Identified in CN-AML patients and patients with prognostically unfavorable complex karyotypes with cryptic amplification of chromosome Significantly shorter OS and higher CIR56,57  
RAS Found in 9%-13% of CN-AML Constitutively activated by point mutations either in the GTP-binding domain (codon 12/13) or in the GTPase domain (codon 61) A clear prognostic role could not be elucidated yet42  
  Possible susceptibility blasts to FTIs  
WT1 Found in ∼ 10% of CN-AML The majority of mutations in both studies were frameshift mutations of exon 7, whereas only one was identified in exon 9 Initial studies on small patient cohorts suggest association with induction failure47,49  
GeneIncidenceBiologic featuresImpact on clinical outcome and targeted therapy opportunities
FLT3    
ITD Found in 28%-34% of CN-AML Member of the class III receptor tyrosine kinase family Consistently associated with inferior outcome in terms of OS, RFS, and EFS33,34  
  In-frame mutations, mostly in exons 14 and 15 of the juxtamembrane domain Constitutively active FLT3 molecules are targets for specific molecular inhibitors (phase 2 or 3 trials) 
  Level of mutant allele probably of importance  
  Homozygous FLT3 mutations as a result of mitotic recombination leading to partial UPD  
TKD Found in 11%-14% of CN-AML In-frame point mutations of the TKD codon 835 and 836 Meta-analysis suggesting a negative prognostic impact32  
  Level of mutant allele may be of importance More recently associated with better OS31  
   High-level mutants associated with improved OS30  
NPM1 Found in 25%-35% of AML(45%-62% of CN-AML) The mutational event modifies specific nucleoli binding and nuclear export signal motifs coded by the exon 12 and determines an abnormal cytoplasmic localization of nucleophosmin NPM1mut/FLT3-ITD genotype associated with a favorable OS, RFS, and CR rate38,3940  
  Associated with presenting clinical and laboratory features, such as female sex, higher BM blast counts, and LDH levels, as well as high CD33 but low or absent CD34 levels Patients with NPM1mut/FLT3-ITD genotype may not benefit from MRD allogeneic transplantation in first CR11  
  Associated with FLT3-ITD and TKD mutations (40% of cases) Pharmacologic modulation of the RA-signaling pathway has been suggested as a therapeutic option11  
  NPM1 acts as a corepressor in RA-associated transcriptional regulation41   
CEBPA Found in 15%-20% of CN-AML Transcription factor mediating lineage specification and differentiation of multipotent myeloid progenitors into mature neutrophils Associated with higher CR rate and better RFS and OS45,46  
  Concurrent mutations (ie, FLT3-ITD and NPM1) were virtually not present in cases with CEBPA–biallelic mutation compared with CEBPA–single allelic mutation44  The presence of CEBPA biallelic (bm) but not single allelic mutation (sm) is an independent factor for a favorable outcome44  
MLL PTD found in 5%-11% of CN-AML Usually involves exons 5-11 or, less frequently, exons 5-12 Associated with shorter CR duration or inferior RFS and EFS60  
  MLL wild-type allele is silenced, probably as a result of differential DNA methylation and histone modifications Allogeneic transplantation may improve outcome11  
  Rationale for the use of DNA methyltransferase and/or histone deacetylase inhibitors based on in vitro data  
IDH1/2 Found in 15% of AML Correlated with accumulation of the cancer-associated metabolite 2-hydroxyglutarate IDH1 mutations identify patients at higher risk of relapse within the favorable risk category of NPM1 mutated AML50,,53  
  IDH2 gene encodes a mitochondrial protein homologous to IDH1 that also catalyzes isocytratecarboxylation IDH2 mutations get along with specific pretreatment features (low WBC, lack of other mutations, and primary resistance to chemotherapy)50,,53  
  IDH1 and IDH2 mutations are mutually exclusive  
DNMT3A Found in 22% of AML. Somatic mutation in DNMT3A, encoding a DNA methyltransferase Independent determinant of dismal prognosis both in the overall study population and, significantly, within high-risk patients (FLT3-ITD, age > 60 years)54  
  Highly enriched in the group of patients with an intermediate-risk cytogenetic profile (56/66, 33.7%) Only ASCT provided a significant benefit54  
  Absent in the subgroup of 79 patients carrying a favorable-risk cytogenetic  
  A perturbation in the methylation of specific DNA sequences has been postulated  
BAALC overexpression Rare Expressed in neuroectoderm-derived tissues and hematopoietic precursors Significantly worse CR rates and shorter DFS, EFS, and OS than patients with low expression55  
  Detected in a subset of karyotypically normal adults < 60 years with de novo AML SCT in first CR might overcome the adverse prognostic effect of high BAALC expression55  
ERG overexpression Rare Identified in CN-AML patients and patients with prognostically unfavorable complex karyotypes with cryptic amplification of chromosome Significantly shorter OS and higher CIR56,57  
RAS Found in 9%-13% of CN-AML Constitutively activated by point mutations either in the GTP-binding domain (codon 12/13) or in the GTPase domain (codon 61) A clear prognostic role could not be elucidated yet42  
  Possible susceptibility blasts to FTIs  
WT1 Found in ∼ 10% of CN-AML The majority of mutations in both studies were frameshift mutations of exon 7, whereas only one was identified in exon 9 Initial studies on small patient cohorts suggest association with induction failure47,49  

LDH indicates lactate dehydrogenase; CN, cytogenetically normal; TKD, tyrosine kinase domain; MRD, matched related donor; JM, juxtamembrane domain; UPD, uniparental disomy; EFS, event-free survival; PTD, partial tandem duplication; NPM1, nucleophosmin (nucleolar phosphoprotein B23, numatrin); FLT3-ITD, internal tandem duplication of the fms-related tyrosine kinase 3 (FLT3) gene; RA, retinoic acid; DFS, disease-free survival; CEBPA, CCAAT/enhancer binding protein (C/EBP)–α; CRD, CR duration; MLL-PTD, partial tandem duplication of the myeloid/lymphoid or mixed-lineage leukemia (MLL) gene; IDH1/2, isocitrate dehydrogenase 1 and 2; DNMT3A, DNA methyltransferase 3A gene; BAALC, brain and acute leukemia gene, cytoplasmic; ERG, v-erythroblastosis virus E26 oncogene-like (avian); GTP, guanosine triphosphate; and FTIs, farnesyltransferase inhibitors.

Figure 1

Pie chart depicting the molecular heterogeneity of CN-AML. The analysis is based on mutations in the NPM1, CEBPA, MLL, and FLT3 (ITD and tyrosine kinase domain [TKD] mutations at codons D835 and I836), NRAS, and WT1 genes. Data are derived from mutational analysis of 485 younger adult patients with CN-AML from the German AML Study Group.90 

Figure 1

Pie chart depicting the molecular heterogeneity of CN-AML. The analysis is based on mutations in the NPM1, CEBPA, MLL, and FLT3 (ITD and tyrosine kinase domain [TKD] mutations at codons D835 and I836), NRAS, and WT1 genes. Data are derived from mutational analysis of 485 younger adult patients with CN-AML from the German AML Study Group.90 

Close modal

PCR and MPFC have proven sensitive and specific enough to allow MRD to be investigated and represent today the gold standard for MRD monitoring in AML.

MRD detection by PCR

Leukemic fusion genes.

The power of PCR relies on cloning of breakpoints of the chromosomal rearrangements and allows their detection in the postremission phase in at least 30% of the patients, using RT-PCR or real-time quantitative PCR. Common targets for PCR-based MRD detection are fusion transcripts of CBF-positive AM (eg, RUNX1-RUNX1T1, CBFB-MYH11, and MLL-gene fusions). RT-PCR in CBF-positive AML has a limited clinical applicability as persistent PCR positivity has been observed in long survivors even after ASCT.24  Real-time quantitative PCR has proven potentially more valuable because of its capability to anticipate impending relapse during follow-up monitoring.25  In a recent report, Corbacioglu et al established clinically relevant MRD cut-points at which persistence of CBFB-MYH11 transcript positivity singled out patients with significantly increased risk of relapse.26  The authors conclude that monitoring of CBFB-MYH11 transcript levels should be incorporated into future clinical trials to guide therapeutic decisions.26 

Mutations.

The discovery of gene mutations in fusion gene-negative AML has potentially increased to 60% to 70% the percentage of cases suitable for PCR-based MRD monitoring (Table 1).27,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,59  The key question about the use of these genes as candidates for MRD monitoring regards their stability over the course of disease. The receptor tyrosine kinase FLT3 is mutated in a relevant proportion of CN-AML. Mutations resulting in the constitutive activation of FLT3 have been identified in 2 functional domains of the receptor: the juxtamembrane domain and the split tyrosine kinase domain.28,29  The juxtamembrane domain is disrupted by internal tandem duplications (ITDs) of various sizes in 28% to 34% of CN-AML, whereas point mutations of the tyrosine kinase domain codon 835 or 836 have been reported in 11% to 14% of CN-AML.30,32  The role of FLT3-ITD in MRD monitoring is controversial: lack of stability of FLT3 mutations in paired samples from diagnosis and relapse raises concerns about the clinical usefulness of this marker.29,36  However, some authors claim that this lack of longitudinal stability might relate to lack of sensitivity. Using high-sensitivity RT-PCR, it was observed that, in all 25 cases under study, FLT3-ITD was detected at diagnosis and relapse. In 4 of them, relapse was predictable based on serially documented reappearance of FLT3-ITD. These results suggest that there is still room for revisiting the role of FLT3-ITD as a potential marker for MRD monitoring.60  Although there are only initial experiences dealing with the use of CEBPA and MLL-PTD in MRD monitoring,61,62  increasing knowledge is being accumulated on the role of NPM1 mutations. NPM1 mutations can be identified in 45% to 60% of patients with CN-AML, accounting for the most frequent genetic change in this subset. The mutational event modifies specific nucleoli binding and nuclear export signal motifs coded by exon 12 and determines an abnormal cytoplasmic localization of NPM1.37,38  Approximately 40% of patients with NPM1 mutations also carry FLT3-ITD. Some studies indicate that NPM1 mutations are very stable at relapse, and thus that they might have a role in MRD assessment.63  Schnittger et al developed a highly sensitive real-time quantitative PCR assay able to prime 17 different mutations of NPM1.64  In 252 NPM1-mutated AML, high levels of NPM1mut, quantified at 4 different time points, were significantly correlated with outcome at each tested time point. Multivariate analysis, including age, FLT3-ITD status, and the level of NPM1, demonstrated that the latter was the most relevant prognostic factor affecting event-free survival during first-line treatment, also in the subgroup of patients who underwent ASCT. Furthermore, the authors compared the kinetics of NPM1 and FLT3-ITD fluctuations and found that FLT3-ITD levels suffer from an unpredictable instability, making it unreliable for MRD purposes.29,36  In a further refinement of such an approach, Krönke et al demonstrated that the level of NPM1mut, measured after double induction and consolidation therapy, impacted on OS and cumulative incidence of relapse (CIR; P < .001 for all comparisons).65 

Gene overexpression.

Techniques of real-time quantitative PCR intended to quantify genes overexpression theoretically provide the opportunity to study the whole population of AML. Various candidates have been proposed, with WT1 being the most reliable. Retrospective clinical data indicate that WT1 assessment after induction therapy is a predictor of outcome, with lower levels being associated with long-term remission.66 WT1 overexpression can be exploited as a marker to establish the presence, persistence, or reappearance of leukemic hematopoiesis. Cilloni et al have measured the levels of WT1 after chemotherapy-induced morphologic CR, in peripheral blood (PB), and BM of patients with AML.67  In multivariate analysis, they found that a more than or equal to 2-log MRD reduction in PB and/or BM was associated with a significantly lower CIR (P = .004). Although the manuscript is of outstanding relevance because of the provision of a common standardized protocol on the behalf of the LeukemiaNet, only in 46% and 13% of PB and BM samples, respectively, were the levels of WT1 sufficiently overexpressed, compared with normal samples, to allow a prognostic stratification. Based on this, concerns still remain about the confounding role of the physiologic background of WT1 in normal PB and BM.

MRD detection by flow cytometry

MRD monitoring by MPFC relies on the expression of “leukemia-associated immunophenotypes” (LAIPs) defined as the presence of a combination of antigens and/or flow cytometric physical abnormalities that are absent or very infrequent in normal BM. The growing interest surrounding this technique is because of its wide applicability (> 85% of AML), quickness, specificity, and ability to distinguish viable cells from BM debris and dead cells. Furthermore, the diffusion of devices equipped with multiple lasers allowed implementation of multiple color assays (> 4 or 5 monoclonal antibody combinations), thus favoring increment of sensitivity that, now, can be reasonably placed in between 10−4 and 10−5. An additional advantage of multiple color assays consists of a significant attenuation of concerns of phenotypic shifts that can be observed on recurrence.68  There is evidence that the use of an expanded 6-9 color polychromatic assay not only increases sensitivity69  but also results in improved qualitative information on the leukemic clone because of the superior definition of its phenotypic composition. In this context, MRD monitoring might not be compromised, especially if serial determinations are performed. The results from the literature demonstrate that detection of MRD by MPFC is technically sound and represents a powerful tool to segregate patients with AML into categories of risk (Table 2). Most studies do not deal with the MRD issue as a simple matter of a positive or negative finding; rather, they set a threshold below or above which the outcome can be significantly different. A common approach is to set up empirically the most significant level of MRD by choosing a logarithmic scale (eg, 101, 102, 103) or a quartile segregation that correlates with survival estimates and CIR. An alternative approach relies on the evaluation of the prognostic role of MRD in a continuous variable model. In the experience of Kern et al, such an analysis demonstrated a strong correlation with outcome when assessed on day 16 from induction and after consolidation.70,71  However, when it came to stratifying the patients in low- or high-risk categories, significant thresholds were identified. At least 4 manuscripts tried to address the issue of MRD “prognostically significant levels” applying specific statistical methods that would help selecting thresholds more appropriately.72,,75  In the paper by Al-Mawali et al, to determine the optimal cut-off yielding the best segregation of AML patients into categories of risk, a receiver operating characteristic analysis was carried out.75  The threshold was established at the value of 0.15% residual leukemic cells; therefore, patients with MRD more than 0.15% qualified as MRD-positive, whereas those with MRD less than or equal to 0.15% as MRD-negative. We evaluated the trend of standardized log-rank statistics using relapse-free survival (RFS) and OS as dependent variables and the values of residual leukemic cells determined after induction and consolidation as independent variables (maximally selected log-rank statistics).76  Based on the results of this test, we currently use the value of 0.035% BM residual leukemic cells to discriminate MRD-negative from MRD-positive cases, both after induction and consolidation. Although the use of a dedicated statistical approach to establish the threshold for MRD negativity/positivity is recommended, the reproducibility of that specific threshold throughout different laboratories still remains an issue. Should we select a particular cut-off that different laboratories can refer to or should any laboratory set up its own? A universal cut-off would be desirable, but it might represent a very troublesome mission to accomplish because standardizing thresholds is affected by a number of technical (equipment, fluorochromes, sensitivity of LAIPs, procedures of acquisition and analysis) and clinical factors. Among clinical factors, the different intensity of chemotherapeutic regimens delivered may play a critical role. Indeed, the clinically relevant threshold of MRD may depend on the therapy used and might change with changes in therapeutic schedule. Divergence between our experience72,73  and that of San Miguel et al is an example of such a situation.77,78  In the study of San Miguel et al, the induction, consolidation, and intensification therapy consisted of the combination of an anthracycline and cytosine arabinoside.77,78  In our study, the patients were treated on 3 drug-based regimens, associating an anthracycline with cytosine arabinoside and etoposide.72,73  In addition, cytosine arabinoside administration was prolonged through 10 days instead of the conventional 7 during the induction phase. Thus, one may assume that, in the study of San Miguel et al, the less intensive therapy delivered was associated with a milder debulking effect that in turn may account for the higher level of MRD at which a significant influence on disease outcome was found.77,78  Based on this, we can also assume that each new protocol will most likely define a new threshold.

Table 2

MRD by MPFC studies including more than 50 patients

ReferenceNo. of patientsTime pointThreshold postinduction, %Threshold postconsolidation, %MethodMultivariate analysisSurvival parameter
San Miguel77  53 I, C 0.5 0.2 NA I,C RFS 
Venditti87  56 I, C 0.045 0.035 empirical OS, RFS 
San Miguel78  126 < 0.01 0.01-0.1, 0.1-1, > 1 NA NA RFS 
Kern70  62 I, C Continuous analysis log-difference Continuous analysis log-difference 75th percentiles of log difference RFS 
Kern71  106 Day 16 after start of induction Continuous analysis log-difference NA Median of log difference Day 16 after start of induction EFS, RFS 
Buccisano72  100 I, C 0.035 0.035 Maximally selected log-rank statistic OS, RFS 
Maurillo73  142 I, C 0.035 0.035 Maximally selected log-rank statistic OS, RFS 
Al Mawali75  54 0.15 0.15 ROC analysis RFS, OS 
ReferenceNo. of patientsTime pointThreshold postinduction, %Threshold postconsolidation, %MethodMultivariate analysisSurvival parameter
San Miguel77  53 I, C 0.5 0.2 NA I,C RFS 
Venditti87  56 I, C 0.045 0.035 empirical OS, RFS 
San Miguel78  126 < 0.01 0.01-0.1, 0.1-1, > 1 NA NA RFS 
Kern70  62 I, C Continuous analysis log-difference Continuous analysis log-difference 75th percentiles of log difference RFS 
Kern71  106 Day 16 after start of induction Continuous analysis log-difference NA Median of log difference Day 16 after start of induction EFS, RFS 
Buccisano72  100 I, C 0.035 0.035 Maximally selected log-rank statistic OS, RFS 
Maurillo73  142 I, C 0.035 0.035 Maximally selected log-rank statistic OS, RFS 
Al Mawali75  54 0.15 0.15 ROC analysis RFS, OS 

I indicates induction; C, consolidation; NA, not available; EFS, event-free survival; and ROC, receiver operating characteristic.

PB may represent an alternative source of cells for the purpose of MRD studies. This is based on the assumption that the presence of circulating blasts at the time of CR might reflect the persistence of malignant cells in the BM. Studies using real-time quantitative PCR to monitor MRD in CBF-positive AML showed that the transcripts were detectable in PB and BM with a comparable sensitivity.79,80  The levels of WT1 transcript measured after consolidation in PB and BM were found to be equally associated with the risk of relapse, and the sensitivity of PB WT1 analysis resulted to be equivalent if not better than that of BM.66,67  We have demonstrated the feasibility of MRD detection in PB of 50 adult AML patients using MPFC.74  The levels of MRD after induction and consolidation in PB significantly reproduced those observed in BM (r = 0.86, P < .0001; and r = 0.82, P < .0001; respectively). A level of MRD more than 0.015% in PB after consolidation was associated with a significant likelihood of subsequent relapse and a shorter RFS. Our data suggest that PB may be a complementary source of cells for MRD studies in patients with AML. In addition, combined measurement of MRD in BM and PB might improve the risk stratification process. The clinical impact of MRD contamination of PB apheresis product is a further subject of research because it has been associated with a shorter RFS and OS.81  Further studies are warranted to clarify these issues.

Timing of PCR assessment

As soon as experience was accumulated, it became clear that selection of the most appropriate time point to determine MRD was a key issue to have a risk-adapted approach transposed into the clinical reality. Ideally, such a time point should be the one havingthe power to provide the most informative prognostic indication, so that the choice of postremission therapy is driven by the actual risk of relapse. The paradigm of such a situation is represented by acute promyelocytic leukemia, where the persistence of PML-RARA fusion transcript at the end of consolidation therapy or subsequent recurrence of PCR positivity in patients previously in molecular remission was demonstrated to precede overt relapse.82,83  CBF-positive cases have been extensively investigated by real-time quantitative PCR for MRD persistence at specific time points. Perea et al found that MRD positivity was associated with an increased risk of relapse at any time of assessment but only MRD persistence at the end of treatment and during subsequent follow-up significantly anticipated relapse.24  Similar results have been reported in CBFB-MYH11 AML by Corbacioglu et al who identified in the postconsolidation and early follow-up phase (≤ 3 months) the critical time points allowing patients at higher risk of relapse to be recognized.26  Krönke et al observed that detection of high NPM1mut transcript levels after double induction and consolidation correlated significantly with an increased CIR.65  In accordance with what we have learned by assessing MRD in acute promyelocytic leukemia, experience with CBF-positive and NPM1mut AML appears to indicate that MRD levels as measured at delayed rather early time points are of superior prognostic relevance. At variance with this assumption, Cilloni et al suggest that risk stratification is indeed improved by early assessment.67  Indeed, they reported an increased risk of relapse when the level of WT1 failed to reduce more than or equal to 2-log after induction; the magnitude of WT1 reduction remained independently significant, even after adjusting for competitive covariates, such as age, white blood cell count, and cytogenetics. A final issue pertains to the hypothesis that the optimal sampling interval varies with molecular subgroups. Ommen et al have demonstrated that the kinetics of molecular relapse are remarkably different among NPM1, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 AML.84  The authors derived a mathematical model to investigate the molecular relapse and the time from molecular relapse to hematologic relapse. They found that CBFB-MYH11 AML displayed a slower clone regrowth than AML with other molecular signature. These results will potentially optimize MRD monitoring, allowing the identification of suitable sampling intervals for other molecular signatures.

Timing of MPFC assessment

With regard to the MPFC approach, the issue of the best time point is even more controversial. The German AML Cooperative group demonstrated that assessment of MRD persistence on day 16 from induction and the log-difference between MRD-positive cells on day 1 and 16 from induction represented an independent prognostic factor affecting CR, event-free survival, OS, and RFS.71  However, it is important to note that this analysis is something different from MRD assessment, for it pertains to the concept of “speed of blast clearance.” “Speed of blast clearance” is supposed to reflect the chemosensitivity of the leukemic clone, but it does not necessarily relate to the quality of response as determined, later on, on full hematopoietic reconstitution. To the best of our knowledge, no formal demonstration has been published on the correlation between “fast blast clearance” and achievement of MRD negativity.85,86  San Miguel et al demonstrated that a stratification according to levels of MRD less than 0.01%, 0.01% to 0.1%, 0.1% to 1%, and more than 1% after induction therapy, resulted in significant differences in OS.77,78  Al-Mawali et al found that a threshold of 0.15% residual leukemic cells discriminated MRD-negative from MRD-positive cases with optimal sensitivity and specificity, allowing impending relapse to be predicted at postinduction and postconsolidation time points.75  Multivariate analysis showed that the postinduction MRD level affected independently RFS and OS. However, also diverging opinions have been published supporting the hypothesis that delayed time points may be even more informative compared with earlier ones. We demonstrated that levels of MRD more than or equal to 0.035%, as measured after consolidation therapy, predicted a high frequency of relapse and a short duration of OS and RFS; the prognostic role of MRD positivity after consolidation was confirmed in multivariate analysis.72,73,87  In line with our experience, Kern et al reported that the 75th percentile of the MRD log-difference between the first day of induction and postconsolidation time point was the sole variable dividing the patients in 2 groups with significantly different OS.70  Selecting an early or delayed time point for MRD determination might entail the choice of different therapeutic options: the early time point option may prove useful to identify as soon as possible high-risk patients for whom a fast allocation to very intensive treatments is required. For these patients, approaches, such as dose-dense schedule88  and/or ASCT, could be incorporated into the upfront treatment strategy.89  On the other hand, opponents of this hypothesis raise concerns about situations of overtreatment for patients showing a slow blast clearance. In our experience, approximately 30% of patients who are MRD-positive after induction become negative at the end of consolidation and the clinical outcome of these “slow responders” was not significantly different from that of patients who tested MRD-negative soon after induction.72,73  Finally, we think that special consideration should be given to specific situations where a serial sampling may be recommended. In our experience, sequential MRD monitoring is required in the phase after autologous stem cell transplantation to enhance detection of impending relapse in patients who are MRD-negative after consolidation.90 

In AML, cytogenetic and molecular findings at diagnosis are critical determinants of outcome and allow stratification of approximately 40% of patients into “good risk” (based on the presence of mutated NPM1 without FLT3-ITD or F-RK) or “poor risk” (FLT3-ITD mutations or U-RK) categories.91  Good risk patients achieve high OS and DFS rates with standard treatments, whereas poor risk do unsuccessfully without intensified therapy with ASCT. On the other hand, there are no accepted criteria to direct the decision-making process after induction/consolidation for patients included in the I-RK (∼ 60%). For these patients, evaluation of MRD status appears appropriate to extrapolate those at high (MRD-positive) or low (MRD-negative) risk of relapse, for whom differentiated treatments may be adopted. Based on this assumption, we have tried to optimize risk assessment of patients with AML by integrating evaluation of pretreatment cytogenetics/genetics and MRD status at the postconsolidation time point.92  Of 143 adult patients, those with F-RK and I-RK who were MRD-negative had 4-year RFS of 70% and 63%, and OS of 84% and 67%, respectively. Patients with F-RK and I-RK who were MRD-positive had 4-year RFS of 15% and 17%, and OS of 38% and 23%, respectively (P < .001 for all comparisons; Figure 2A-B). Likewise, FLT3 wild-type patients achieving a MRD-negative status (Figure 2C-D) had a better outcome than those who remained MRD-positive after consolidation (4-year RFS 54% vs 17% P < .0001, OS 60% vs 23% P = .002). Therefore, patients with F-RK, I-RK, or FLT3 wild-type had a very different outcome depending on MRD status at the end of consolidation. The Children's Oncology Group has recently carried out the same analysis, based on which children with standard risk AML and MRD positivity after the first induction were reallocated to the high-risk category, whereas those MRD-negative were added to the favorable-risk cohort.93  This risk stratification system generated 2 new categories with a 3-year DFS of 20% and 68%, respectively (P < .001). The authors conclude that cytogenetics, molecular genotyping, and postinduction MPFC analysis provide a robust means of stratifying pediatric AML into 2 risk groups with significantly different outcomes.

Figure 2

Subgroup analysis of RFS and CIR of 143 AML patients stratified according to pretreatment karyotype or FLT3 status and levels of MRD after consolidation. (A-B) Those with a level of residual leukemic cells < 0.035% are referred to as intermediate karyotype-MRD, favorable karyotype-MRD, or FLT-wt MRD, whereas those with levels ≥ 0.035% are categorized as intermediate karyotype-MRD+, favorable karyotype-MRD+, or FLT-wt MRD+. Survival outcomes of these subsets and of U-RK and FLT3-ITD category are shown (P < .001 for all comparisons). (C-D) FLT-wt patients achieving a MRD-negative status show a better outcome than those who remained MRD-positive after consolidation (P < .001).

Figure 2

Subgroup analysis of RFS and CIR of 143 AML patients stratified according to pretreatment karyotype or FLT3 status and levels of MRD after consolidation. (A-B) Those with a level of residual leukemic cells < 0.035% are referred to as intermediate karyotype-MRD, favorable karyotype-MRD, or FLT-wt MRD, whereas those with levels ≥ 0.035% are categorized as intermediate karyotype-MRD+, favorable karyotype-MRD+, or FLT-wt MRD+. Survival outcomes of these subsets and of U-RK and FLT3-ITD category are shown (P < .001 for all comparisons). (C-D) FLT-wt patients achieving a MRD-negative status show a better outcome than those who remained MRD-positive after consolidation (P < .001).

Close modal

The role of the leukemic stem cell (LSC) in AML recurrence is now more than a simple hypothesis. Relapses derive from re-expansion of residual leukemic cells escaping the cytotoxic effect of chemotherapy; this leukemic cell population is thought to reside within the stem cell CD34+CD38 compartment. Experimental data indicate that LSC are more resistant to chemotherapy than the more mature CD34+CD38+ progeny and can be distinguished from their normal counterpart because of the expression of LSC-specific immunophenotype.94  Therefore, the concept of LAIP expression is something that applies, even to LSC on which aberrant overexpression of CD47,95  CD123, and CD44 has been described.94  Among markers of LCS aberrancy, C-type lectin-like molecule-1 (CLL-1) promises to be one of the most appealing because its expression is lacking on normal stem cells.96,97  Therefore, CLL-1 has the double role of LSC specific antigen and potential target for future LSC directed-therapy. Using a monoclonal antibody targeting CLL-1, Terwijn et al demonstrated that a LSC frequency more than 1 × 10−3 after induction and more than 2 × 10−4 after the second induction and consolidation predicted a short RFS (P = .00003 and .004, respectively).98  By combining the residual LSC and the “whole MRD” fractions, they came up with 4 different categories whose outcome was the best for the group with a negative residual LSC status.98  The persistence of residual LSC may explain why a certain proportion of MRD-negative patients experience disease recurrence: in our experience, such a proportion accounts for 20% to 25% of MRD-negative patients. Therefore, monitoring of LSC-LAIPs represents an additional tool capable to refine the MPFC MRD assessment, and the combination of LSC-LAIPs and “whole MRD blast” frequencies might prove useful to guide future therapeutic intervention.

The choice of postremission therapy is too often driven by a “genetic randomization” resulting in a given patient being allocated to ASCT once the availability of an HLA-compatible sibling donor is documented. Meta-analysis of large prospective studies indicates that the beneficial effect of ASCT takes place as soon as the risk of relapse exceeds 35% to 40%99 ; when probabilities of relapse are below those percentages the risk of treatment-related mortality will attenuate the survival advantage of this procedure. Furthermore, the extensive use of ASCT is hampered by the paucity of candidates (25%-30%) with a fully matched family donor. For the remaining, even if a timely search of either international registries or cord blood banks is started, the probability of identifying a suitable donor is 46% and 73% at 3 and 6 months, respectively (W. Arcese, personal oral communication, June 30, 2011). In the meantime, almost 40% of candidates for transplant die of relapse or are relegated to less effective approaches, such as chemotherapy or autologous stem cell transplantation. We have demonstrated that an adjusted risk stratification based on pretreatment genetics/cytogenetics and MRD status at the end of consolidation refines the upfront genetic/cytogenetic risk classification.92  In this view, MRD assessment will greatly enhance selection procedures so that patients are assigned to ASCT not on the basis of donor availability but of the real risk of relapse.17  Applying this adjusted risk stratification, we distinguished 2 categories of patients: (1) low-risk: F-RK and I-RK, which were MRD-negative after consolidation; and (2) high-risk: U-RK, FLT3-ITD mutated cases, F-RK and I-RK, which were MRD-positive after consolidation (Figure 3). After these observations, we started a program for high-risk AML based on a prospective assignment to ASCT that should be delivered in the form of matched sibling donor, matched unrelated donor, umbilical cord blood, or haploidentical related donor transplant. We analyzed preliminarily a cohort of 21 high-risk patients who, according to the aforementioned policy, were assigned to ASCT (8 matched sibling donor, 7 matched unrelated donor/umbilical cord blood, and 6 haploidentical related donor). For comparative purposes, a matched historical cohort of 36 high-risk patients was analyzed: 12 were given matched sibling donor-ASCT and 24, lacking a matched sibling donor, received autologous stem cell transplantation. Survival estimates were significantly better for the prospective cohort compared with the control group (DFS 70% vs 20%, P = .000 47; OS 69% vs 24%, P = .046; Figure 4). The prospective cohort also showed a lower relapse rate (20% vs 52%, P = .003).

Figure 3

Risk assessment combining pretreatment and post-treatment prognosticators. Applying an adjusted risk stratification, combining genetics/cytogenetics and level of MRD at the end of consolidation therapy, we distinguished 2 categories of patients: (1) low-risk, including F-RK and I-RK that were MRD-negative; and (2) high-risk, including U-RK, FLT3-ITD cases, and F-RK/I-RK that were MRD-positive. The first group stands for significantly longer OS (73% vs 17%), RFS (58% vs 22%), and cumulative incidence of relapse (17% vs 77%; P < .001 for all comparisons).

Figure 3

Risk assessment combining pretreatment and post-treatment prognosticators. Applying an adjusted risk stratification, combining genetics/cytogenetics and level of MRD at the end of consolidation therapy, we distinguished 2 categories of patients: (1) low-risk, including F-RK and I-RK that were MRD-negative; and (2) high-risk, including U-RK, FLT3-ITD cases, and F-RK/I-RK that were MRD-positive. The first group stands for significantly longer OS (73% vs 17%), RFS (58% vs 22%), and cumulative incidence of relapse (17% vs 77%; P < .001 for all comparisons).

Close modal
Figure 4

Comparison of clinical outcome in the “prospective” and “historical” cohort. The prospective cohort included 21 high-risk patients (4 MRD-positive favorable-karyotype, 9 MRD-positive intermediate-karyotype, 4 unfavorable-karyotype, and 4 FLT3-ITD) who underwent ASCT (8 matched sibling donor, 6 haploidentical related, and 7 matched unrelated/umbilical cord blood donor). The historical cohort accounted for 36 high-risk patients (6 MRD-positive favorable-karyotype, 23 MRD-positive intermediate-karyotype, 1 unfavorable-karyotype, and 6 FLT3-ITD). ASCT was offered to 12 patients with an available matched sibling donor, whereas those lacking this option were given autologous stem cell transplantation (n = 24). With a median follow-up of 18 months, survival estimates were significantly superior for the prospective cohort compared with the historical control (DFS 70% vs 20%, P = .000 47; OS 69% vs 24%, P = .046).

Figure 4

Comparison of clinical outcome in the “prospective” and “historical” cohort. The prospective cohort included 21 high-risk patients (4 MRD-positive favorable-karyotype, 9 MRD-positive intermediate-karyotype, 4 unfavorable-karyotype, and 4 FLT3-ITD) who underwent ASCT (8 matched sibling donor, 6 haploidentical related, and 7 matched unrelated/umbilical cord blood donor). The historical cohort accounted for 36 high-risk patients (6 MRD-positive favorable-karyotype, 23 MRD-positive intermediate-karyotype, 1 unfavorable-karyotype, and 6 FLT3-ITD). ASCT was offered to 12 patients with an available matched sibling donor, whereas those lacking this option were given autologous stem cell transplantation (n = 24). With a median follow-up of 18 months, survival estimates were significantly superior for the prospective cohort compared with the historical control (DFS 70% vs 20%, P = .000 47; OS 69% vs 24%, P = .046).

Close modal

In conclusion, the paradigm of treatment for adult AML has largely been based on the “one size fits all” approach: in the short term, this has led to satisfactory rates of CR; but in the long term, survival estimates for young and elderly patients are disappointing. In acute promyelocytic leukemia and acute lymphoblastic leukemia, MRD positivity has consistently been shown to increase probabilities of relapse; therefore, attainment of MRD-negative remission represents a “gold standard,” leading to prolonged duration of CR.100  More recently, a trial of prospective MRD-driven therapy has also been initiated in childhood non-M3 AML, demonstrating an improvement of outcome in high-risk patients.101  Altogether, these observations suggest that, whatever the method, even in non-M3 AML measurement of MRD will potentially contribute refining risk assessment. In this view, a comprehensive risk stratification, generated by integrating the prognostic weight of pretreatment and posttreatment parameters (MRD),92  might help to allocate the majority of patients to a more realistic category of risk, thus favoring selection of more appropriate postremission strategies. In the context of such an integrated approach, pretreatment cytogenetic/genetic profile will dictate intensity and type of induction therapy, whereas posttreatment MRD status helps modulating intensity of postremission strategies, allowing a treatment proportional to the individual risk of relapse to be delivered. The final purpose is that patients will be assigned to ASCT not on the basis of donor availability (donor vs no donor approach) but on the basis of their adjusted (cytogenetic/genetic plus MRD) risk of relapse (transplant vs no transplant approach). This will also comply with the primary goal of saving additional lives, even without any major advances in chemotherapy or transplant technologies.17 

Contribution: All authors wrote the manuscript.

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

Correspondence: Adriano Venditti, Ematologia, Fondazione Policlinico Tor Vergata, Viale Oxford 81-00133, Rome, Italy; e-mail: adriano.venditti@uniroma2.it.

1
Löwenberg
G
Strategies in the treatment of acute myeloid leukemia.
Haematologica
2004
89
9
1029
1032
2
Burnett
AK
Goldstone
AH
Stevens
RM
et al
Randomised comparison of addition of autologous bone marrow transplantation to intensive chemotherapy for acute myeloid leukemia in first remission: results of MRC AML10 trial. UK Medical Research Council Adult and Children's Leukemia Working Parties.
Lancet
1998
351
9104
700
708
3
Cassileth
PA
Harrington
DP
Appelbaum
FR
et al
Chemotherapy compared with autologous or allogeneic bone marrow transplantation in the management of acute myeloid leukemia in first remission.
N Engl J Med
1998
339
23
1649
1656
4
Harousseau
JL
Cahn
JY
Pignon
B
et al
Comparison of autologous bone marrow transplantation and intensive chemotherapy as post-remission therapy in adult acute myeloid leukemia: the Groupe Ouest Est Leucemies Aigues Myeloblastiques (GOELAM).
Blood
1997
90
8
2978
2986
5
Suciu
S
Mandelli
F
de Witte
T
et al
Allogeneic compared with autologous stem cell transplantation in the treatment of patients younger than 46 years with acute myeloid leukemia (AML) in first complete remission (CR1): an intention-to-treat analysis of the EORTC/GIMEMAAML-10 trial.
Blood
2003
102
4
1232
1240
6
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
116
3
354
365
7
Grimwade
D
Walker
H
Oliver
F
et al
The importance of diagnostic cytogenetics on outcome in AML: analysis of 1,612 patients entered into the MRC AML 10 trial.
Blood
1998
92
7
2322
2333
8
Slovak
ML
Kopecky
KJ
Cassileth
PA
et al
Karyotypic analysis predicts outcome of pre-remission and post-remission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group study.
Blood
2000
96
13
4075
4083
9
Mrózek
K
Heerema
NA
Bloomfield
CD
Cytogenetics in acute leukemia.
Blood Rev
2004
18
12
115
136
10
Byrd
JC
Mrózek
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
100
13
4325
4336
11
Mrózek
K
Marcucci
G
Paschka
P
et al
Advances in molecular genetics and treatment of core-binding factor acute myeloid leukemia.
Curr Opin Oncol
2008
20
6
711
718
12
Schlenk
RF
Dohner
K
Krauter
J
et al
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia.
N Engl J Med
2008
358
18
1909
1918
13
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
107
9
3463
3468
14
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
24
24
3904
3911
15
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
107
5
1791
1799
16
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
301
22
2349
2361
17
Appelbaum
FR
Incorporating hematopoietic cell transplantation (HCT) into the management of adults aged under 60 years with acute myeloid leukemia (AML).
Best Pract Res Clin Haematol
2008
21
4
85
92
18
Campana
D
Pui
C-H
Detection of minimal residual disease in acute leukemia: methodological advances and clinical significance.
Blood
1995
85
6
1416
1434
19
Campana
D
determination of minimal residual disease in leukemia patients.
Br J Haematol
2003
121
6
823
838
20
Mrózek
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
109
2
431
448
21
Wheatley
K
Burnett
AK
Goldstone
AH
et al
A simple, robust, validated and highly predictive index for the determination of risk-directed therapy in acute myeloid leukemia derived from the MRC AML 10 trial.
Br J Haematol
1999
107
1
69
79
22
Freeman
S
Jovanovic
J
Grimwade
D
Development of minimal residual disease: directed therapy in acute myeloid leukemia.
Semin Oncol
2008
35
4
388
400
23
Yin
A
Grimwade
D
Minimal residual disease evaluation in acute myeloid leukaemia.
Lancet
2002
360
9327
160
162
24
Perea
G
Lasa
A
Aventín
A
et al
Prognostic value of minimal residual disease (MRD) in acute myeloid leukemia (AML) with favorable cytogenetics [t(8;21) and inv(16)].
Leukemia
2006
20
1
87
94
25
Martinelli
G
Rondoni
M
Buonamici
S
et al
Molecular monitoring to identify a threshold of CBFbeta/MYH11 transcript below which continuous complete remission of acute myeloid leukemia inv16 is likely.
Haematologica
2004
89
4
495
497
26
Corbacioglu
A
Scholl
C
Schlenk
RF
et al
Prognostic impact of minimal residual disease in CBFB-MYH11-positive acute myeloid leukemia.
J Clin Oncol
2010
28
23
3724
3729
27
Mrózek
K
Bloomfield
CD
Chromosome aberrations, gene mutations and expression changes, and prognosis in adult acute myeloid leukemia.
Hematology Am Soc Hematol Educ Program
2006
2006
169
77
28
Nakao
M
Yokota
S
Iwai
T
et al
Internal tandem duplication of the FLT3 gene found in acute myeloid leukemia.
Leukemia
1996
10
12
1911
1918
29
Yamamoto
Y
Kiyoi
H
Nakano
Y
et al
Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies.
Blood
2001
97
8
2434
2439
30
Frohling
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
100
13
4372
4380
31
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
99
12
4326
4335
32
Mead
AJ
Linch
DC
Hills
RK
et al
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
110
4
1262
1270
33
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
19
8
1345
1349
34
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
98
6
1752
1759
35
Whitman
SP
Archer
KJ
Feng
L
et al
Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a Cancer and Leukemia Group B study.
Cancer Res
2001
61
19
7233
7239
36
Cloos
J
Goemans
BF
Hess
CJ
et al
Stability and prognostic influence of FLT3 mutations in paired initial and relapsed AML samples.
Leukemia
2006
20
7
1217
1220
37
Falini
B
Mecucci
C
Tiacci
E
et al
Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype [erratum in: N Engl J Med. 2005;352(7):740].
N Engl J Med
2005
352
3
254
265
38
Döhner
H
Implication of the molecular characterization of acute myeloid leukemia.
Hematology Am Soc Hematol Educ Program
2007
2007
412
419
39
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
106
12
3733
3739
40
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
106
12
3740
3746
41
Verhaak
RG
Goudswaard
CS
van Putten
W
et al
Mutations in nucleophosmin (NPM1) in acute myeloid leukemia (AML): association with other gene abnormalities and previously established gene expression signatures and their favorable prognostic significance.
Blood
2005
106
12
3747
3754
42
Liu
H
Tan
BC
Tseng
KH
et al
Nucleophosmin acts as a novel AP2alpha-binding transcriptional corepressor during cell differentiation.
EMBO Rep
2007
8
4
394
400
43
Mrózek
K
Döhner
H
Bloomfield
CD
Influence of new molecular prognostic markers in patients with karyotypically normal acute myeloid leukemia: recent advances.
Curr Opin Hematol
2007
14
2
106
114
44
Estey
E
Döhner
H
Acute myeloid leukaemia.
Lancet
2006
368
9550
1894
1907
45
Taskesen
E
Bullinger
L
Corbacioglu
A
et al
Prognostic impact, concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity.
Blood
2011
117
8
2469
2475
46
Whitman
SP
Liu
S
Vukosavljevic
T
et al
The MLL partial tandem duplication: evidence for recessive gain-of-function in acute myeloid leukemia identifies a novel patient subgroup for molecular-targeted therapy.
Blood
2005
106
1
345
352
47
Döhner
K
Tobis
K
Ulrich
R
et al
Prognostic significance of partial tandem duplications of the MLL gene in adult patients 16 to 60 years old with acute myeloid leukemia and normal cytogenetics: a study of the Acute Myeloid Leukemia Study Group Ulm.
J Clin Oncol
2002
20
15
3254
3261
48
Summers
K
Stevens
J
Kakkas
I
et al
Wilms' tumour 1 mutations are associated with FLT3-ITD and failure of standard induction chemotherapy in patients with normal karyotype AML.
Leukemia
2007
21
3
550
551
49
Virappane
P
Gale
R
Hills
R
et al
Mutation of the Wilms' tumor 1 gene is a poor prognostic factor associated with chemotherapy resistance in normal karyotype acute myeloid leukemia: the United Kingdom Medical Research Council Adult Leukaemia Working Party.
J Clin Oncol
2008
26
33
5429
5435
50
Paschka
P
Marcucci
G
Ruppert
AS
et al
Wilms' tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
J Clin Oncol
2008
26
28
4595
4602
51
Mardis
ER
Ding
L
Dooling
DJ
et al
Recurring mutations found by sequencing an acute myeloid leukemia genome.
N Engl J Med
2009
361
11
1058
1066
52
Thol
F
Damm
F
Wagner
K
et al
Prognostic impact of IDH2 mutations in cytogenetically normal acute myeloid leukemia.
Blood
2010
116
4
614
616
53
Marcucci
G
Maharry
K
Wu
YZ
et al
IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
J Clin Oncol
2010
28
14
2348
2355
54
Schnittger
S
Haferlach
C
Ulke
M
et al
IDH1 mutations are detected in 6.6% of 1414 AML patients and are associated with intermediate risk karyotype and unfavorable prognosis in adults younger than 60 years and unmutated NPM1 status.
Blood
2010
116
25
5486
5496
55
Ley
TJ
Ding
L
Walter
MJ
et al
DNMT3A mutations in acute myeloid leukemia.
N Engl J Med
2010
363
25
2424
2433
56
Baldus
CD
Tanner
SM
Ruppert
AS
et al
BAALC expression predicts clinical outcome of de novo acute myeloid leukemia patients with normal cytogenetics: a Cancer and Leukemia Group B Study.
Blood
2003
102
5
1613
1618
57
Baldus
CD
Liyanarachchi
S
Mrozek
K
et al
Acute myeloid leukemia with complex karyotypes and abnormal chromosome 21: amplification discloses overexpression of APP, ETS2, and ERG genes.
Proc Natl Acad Sci U S A
2004
101
11
3915
3920
58
Marcucci
G
Maharry
K
Whitman
SP
et al
High expression levels of the ETS-related gene, ERG, predict adverse outcome and improve molecular risk-based classification of cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B Study.
J Clin Oncol
2007
25
22
3337
3343
59
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
121
5
775
777
60
Schnittger
S
Schoch
C
Dugas
M
et al
Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease.
Blood
2002
100
1
59
66
61
Smith
LL
Pearce
D
Smith
ML
Development of a quantitative real-time polymerase chain reaction method for monitoring CEBPA mutations in normal karyotype acute myeloid leukaemia.
Br J Haematol
2006
133
1
103
105
62
Weisser
M
Kern
W
Schoch
C
et al
Risk assessment by monitoring expression levels of partial tandem duplications in the MLL gene in acute myeloid leukemia during therapy.
Haematologica
2005
90
7
881
889
63
Gorello
P
Cazzaniga
G
Alberti
F
et al
Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations.
Leukemia
2006
20
6
1103
1108
64
Schnittger
S
Kern
W
Tschulik
C
et al
Minimal residual disease levels assessed by NPM1 mutation specific real-time quantitative PCR provide important prognostic information in AML.
Blood
2009
114
11
2220
2231
65
Krönke
J
Schlenk
R
Jensen
KO
et al
Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia: a study from the German-Austrian Acute Myeloid Leukemia Study Group.
J Clin Oncol
2011
29
19
2709
2716
66
Cilloni
D
Messa
F
Arruga
F
et al
Early prediction of treatment outcome in acute myeloid leukemia by measurement of WT1 transcript levels in peripheral blood samples collected after chemotherapy.
Haematologica
2008
93
6
921
924
67
Cilloni
D
Renneville
A
Hermitte
F
et al
Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: a European LeukemiaNet study.
J Clin Oncol
2009
27
31
5195
5201
68
Voskova
D
Schoch
C
Schnittger
S
Hiddemann
W
Haferlach
T
Kern
W
Stability of leukemia associated aberrant immunophenotypes in patients with acute leukemia between diagnosis and relapse: comparison with cytomorphologic, cytogenetic and molecular genetic findings.
Cytometry B Clin Cytom
2004
62
1
25
38
69
Olaru
D
Campos
L
Flandrin
P
et al
Multiparametric analysis of normal and postchemotherapy bone marrow: implication for the detection of leukemia-associated immunophenotypes.
Cytometry B Clin Cytom
2008
74
1
17
24
70
Kern
W
Voskova
D
Schoch
C
Hiddemann
W
Schnittger
S
Haferlach
T
Determination of relapse risk based on assessment of minimal residual disease during complete remission by multiparameter flow cytometry in unselected patients with acute leukemia.
Blood
2004
104
10
3078
3085
71
Kern
W
Haferlach
T
Schoch
C
et al
Early blast clearance by remission induction therapy is a major independent prognostic factor for both achievement of complete remission and long-term outcome in acute myeloid leukemia: data from the German AML Cooperative Group (AMLCG) 1992 Trial.
Blood
2003
101
1
64
70
72
Buccisano
F
Maurillo
L
Gattei
V
et al
The kinetics of reduction of minimal residual disease impacts on duration of response and survival of patients with acute myeloid leukemia.
Leukemia
2006
20
10
1783
1789
73
Maurillo
L
Buccisano
F
Del Principe
MI
et al
Toward optimization of postremission therapy for residual disease-positive patients with acute myeloid leukemia.
J Clin Oncol
2008
26
30
4944
4951
74
Maurillo
L
Buccisano
F
Spagnoli
A
et al
Monitoring of minimal residual disease in adult acute myeloid leukemia using peripheral blood as an alternative source to bone marrow.
Haematologica
2007
92
5
605
611
75
Al-Mawali
A
Gillis
D
Lewis
I
The use of receiver operating characteristic analysis for detection of minimal residual disease using five-color multiparameter flow cytometry in acute myeloid leukemia identifies patients with high risk of relapse.
Cytometry B Clin Cytom
2009
76
2
91
101
76
Hothorn
T
Lausen
B
On the exact distribution of maximally selected rank statistics.
Comput Statist Data Analysis
2003
43
121
137
77
San Miguel
JF
Martinez
A
Macedo
A
et al
Immunophenotyping investigation of minimal residual disease is a useful approach for predicting relapse in acute myeloid leukemia patients.
Blood
1997
90
6
2465
2470
78
San Miguel
JF
Vidriales
MB
Lopez Berges
C
et al
Early immunophenotypical evaluation of minimal residual disease in acute myeloid leukemia identifies different patient risk groups and may contribute to post-induction treatment stratification.
Blood
2001
98
6
1746
1751
79
Leroy
H
de Botton
S
Grardel-Duflos
N
et al
Prognostic value of realtime quantitative PCR (real-time quantitative PCR) in AML with t(8;21).
Leukemia
2005
19
3
367
372
80
Stentoft
J
Hokland
P
Ostergaard
M
et al
Minimal residual core binding factor AMLs by real time quantitative PCR-initial response to chemotherapy predicts event free survival and close monitoring of peripheral blood unravels the kinetics of relapse.
Leuk Res
2006
30
4
389
395
81
Feller
N
van der Pol
MA
van Stijn
A
et al
MRD parameters using immunophenotypic detection methods are highly reliable in predicting survival in acute myeloid leukaemia.
Leukemia
2004
18
8
1380
1390
82
Sanz
MA
Lo-Coco
F
Modern approaches to treating acute promyelocytic leukemia.
J Clin Oncol
2011
29
5
495
503
83
Diverio
D
Rossi
V
Avvisati
G
et al
Early detection of relapse by prospective reverse transcriptase-polymerase chain reaction analysis of the PML/RARalpha fusion gene in patients with acute promyelocytic leukemia enrolled in the GIMEMA-AIEOP multicenter “AIDA” trial: GIMEMA-AIEOP Multicenter “AIDA” Trial.
Blood
1998
92
3
784
789
84
Ommen
HB
Schnittger
S
Jovanovic
JV
et al
Strikingly different molecular relapse kinetics in acute myeloid leukemias.
Blood
2010
115
2
198
205
85
Gianfaldoni
G
Mannelli
F
Bencini
S
Leoni
F
Baldini
S
Bosi
A
Peripheral blood blast clearance during induction therapy in acute myeloid leukemia.
Blood
2008
111
3
1746
1747
86
Elliott
MA
Litzow
MR
Letendre
LL
et al
Early peripheral blood blast clearance during induction chemotherapy for acute myeloid leukemia predicts superior relapse-free survival.
Blood
2007
110
13
4172
4174
87
Venditti
A
Buccisano
F
Del Poeta
G
et al
Level of minimal residual disease after consolidation therapy predicts outcome in acute myeloid leukemia.
Blood
2000
96
12
3948
3952
88
Braess
J
Spiekermann
K
Staib
P
et al
Dose-dense induction with sequential high-dose cytarabine and mitoxantone (S-HAM) and pegfilgrastim results in a high efficacy and a short duration of critical neutropenia in de novo acute myeloid leukemia: a pilot study of the AMLCG.
Blood
2009
113
17
3903
3910
89
Schaich
M
Illmer
T
Aulitzky
WE
et al
Upfront allogeneic stem cell transplantation for remission induction in high-risk acute myeloid leukemia patients within the randomized multi-center trial AML2003.
Blood
2008
112
11
978a
90
Venditti
A
Maurillo
L
Buccisano
F
et al
Pretransplant minimal residual disease level predicts clinical outcome in patients with acute myeloid leukemia receiving high-dose chemotherapy and autologous stem cell transplantation.
Leukemia
2003
17
11
2178
2182
91
Döhner
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
115
3
453
474
92
Buccisano
F
Maurillo
L
Spagnoli
A
et al
Cytogenetic and molecular diagnostic characterization combined to post-consolidation minimal residual disease assessment by flow-cytometry improves risk stratification in adult acute myeloid leukemia.
Blood
2010
116
13
2295
2303
93
Alonzo
TA
Ho
PA
Gerbing
RB
et al
Conventional cytogenetics, molecular profiling, and flow cytometric response data allow the creation of a two-tiered risk-group system for risk-based therapy allocation in childhood AML: a Report From the Children's Oncology Group.
Blood
2010
116
21
761a
94
van Rhenen
A
Moshaver
B
Kelder
A
et al
Aberrant marker expression patterns on the CD34+CD38- stem cell compartment in acute myeloid leukemia allows to distinguish the malignant from the normal stem cell compartment both at diagnosis and in remission.
Leukemia
2007
21
8
1700
1707
95
Majeti
R
Chao
MP
Alizadeh
AA
et al
CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells.
Cell
2009
138
2
286
299
96
van Rhenen
A
Feller
N
Kelder
A
et al
High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival.
Clin Cancer Res
2005
11
18
6520
6527
97
Bakker
AB
van den Oudenrijn
S
Bakker
AQ
et al
C-type lectin-like molecule-1: a novel myeloid cell surface marker associated with acute myeloid leukemia.
Cancer Res
2004
64
22
8443
8450
98
Terwijn
M
Kelder
A
Rutten
AP
et al
Leukemic stem cell assessment in remission bone marrow of acute myeloid leukemia patients is a new prognostic parameter.
Blood
2009
114
22
399a
99
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
109
9
3658
3666
100
Cheson
BD
Bennett
JM
Kopecky
KJ
et al
International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia: revised recommendations of the International Working Group for diagnosis, standardizations of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia.
J Clin Oncol
2003
21
24
4642
4649
101
Rubnitz
JE
Inaba
H
Dahl
G
et al
Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: results of the AML02 multicentre trial.
Lancet Oncol
2010
11
6
543
552
Sign in via your Institution