Case 1: A 35-year-old man with a normal white blood cell (WBC) count (9.3 × 109/L) was diagnosed with acute myeloid leukemia (AML) with a t(8;21)(q22;q22) translocation in 25/25 metaphases. The RUNX1-RUNX1T1 fusion gene was detected by real-time quantitative polymerase chain reaction (PCR), whereas studies for mutations involving KIT and FLT3 were negative. After 1 cycle of induction therapy with cytarabine/idarubicin according to the “7+3” schema, he achieved a morphologic complete remission (CR) with a 2-log reduction of RUNX1-RUNX1T1 transcript levels. The patient has an excellent performance status and no comorbidities. Should you recommend allogeneic hematopoietic cell transplantation (HCT)?

Case 2: A 43-year-old woman was diagnosed with cytogenetically normal AML; molecular studies for gene mutations involving NPM1, CEBPA, and FLT3 were negative. After standard induction chemotherapy, she achieved a morphologic CR and then underwent 1 cycle of consolidation therapy with high-dose cytarabine. During the pre-HCT work-up in anticipation of matched related donor transplant, she is found to have evidence of minimal residual disease (MRD) by multiparameter flow cytometry (MFC); no prior MFC studies are available. She has no comorbidities other than arterial hypertension, and her performance status is excellent. Are you recommending additional cycle(s) of chemotherapy to attempt MRD eradication before HCT?

In recent years, several methods have been developed to detect submicroscopic MRD in AML patients in morphologic remission.1-4  The existence of small numbers of leukemic cells among normal hematopoietic cells can be identified based on numeric or structural chromosomal changes, gene mutations, antigen receptor rearrangements, abnormal gene expression, altered cell growth, and immunophenotypic abnormalities. Thus far, most exploited for MRD detection and quantification in AML are MFC- and PCR-based approaches, which can achieve sensitivities up to 10−5 to 10−6.1-5  MFC has gained popularity for the detection of MRD in AML because it can be applied to the vast majority of AML patients, although the identification of immunophenotypic abnormalities can be challenging, especially if a diagnostic specimen is not available or the disease has evolved over time. PCR-based approaches are typically limited to specific patient subsets, but recent methodologic advances (eg, based on next generation sequencing or digital PCR) allow leukemia-associated mutations to be tracked more comprehensively, thereby broadening the scope of molecular MRD detection (defined in this study as the detection of chimeric fusion genes, somatic mutations, or aberrant gene expression).

For several reasons, including variations in health care provision and laboratory infrastructures between countries and, perhaps, the fluidity with which MRD detection methodologies are evolving, implementation of standardized MRD assessments into clinical practice has remained a major challenge. Nevertheless, increasing evidence indicates that the presence of MRD, measured either molecularly (as in case 1) or by MFC (as in case 2), identifies patients at particularly high risk of relapse and provides powerful prognostic information beyond pretreatment characteristics, such as cytogenetic or molecular abnormalities.4  This observation has sparked interest in risk-adapted treatment strategies that are based on the MRD status to improve patient outcomes. Herein, we examined the evidence supporting such an approach.

A systematic literature search, restricted to humans and the English language, was conducted using MEDLINE (October 24, 2014), Embase (October 31, 2014), and CENTRAL (Cochrane Register of Controlled Trials; November 10, 2014) (see supplemental Tables A1 and A2, available on the Blood Web site). Three authors reviewed all abstracts. Studies were included if they provided useful extractable data for AML (other than acute promyelocytic leukemia [APL]) for which MRD parameters were used to direct therapy. Potential unpublished articles were also sought by searching Web of Science, the Web sites for the conference proceedings from the American Society of Hematology (2012-2014) and the American Society of Clinical Oncology (2012-2014), as well as the Clinical Trial Registry (www.clinicaltrials.gov; November 10, 2014). Recommendations were developed based on the Grading of Recommendations Assessment Development and Evaluation system (Table 1).6 

Table 1

Summary of GRADE recommendations on rating the strength of recommendations and quality of evidence

Strength of recommendationQuality of evidence
1 (“Strong”) Desirable effects of an intervention clearly outweigh (or clearly do not outweigh) the undesirable effects A (“High”) Further research is very unlikely to change our confidence in the estimate of effect 
2 (“Weak”) Trade-offs between desirable and undesirable effects are less certain (eg, because of low-quality evidence or evidence suggesting closely balanced effects) B (“Moderate”) Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate 
  C (“Low”) Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate 
  D (“Very low”) Any estimate of effect is very uncertain 
Strength of recommendationQuality of evidence
1 (“Strong”) Desirable effects of an intervention clearly outweigh (or clearly do not outweigh) the undesirable effects A (“High”) Further research is very unlikely to change our confidence in the estimate of effect 
2 (“Weak”) Trade-offs between desirable and undesirable effects are less certain (eg, because of low-quality evidence or evidence suggesting closely balanced effects) B (“Moderate”) Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate 
  C (“Low”) Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate 
  D (“Very low”) Any estimate of effect is very uncertain 

GRADE, Grading of Recommendations Assessment Development and Evaluation.

Each recommendation consists of a numerical score denoting the strength of the recommendation and a letter denoting the quality of the evidence.6 

Our systematic literature search resulted in 603 records after duplicates had been removed (MEDLINE, n = 236; Embase, n = 402; and CENTRAL, n = 16). Of these, 28 were reviewed in full. No randomized, controlled study was found to address MRD-directed therapy for non-APL AML.

Case 1: MRD-directed therapy of favorable-risk AML

Patients with t(8;21)(q22;q22) AML generally have a relatively favorable prognosis. With intensive induction chemotherapy, nearly all individuals who do not die of treatment-related toxicities will achieve a morphologic CR, and with repeated courses of intensive consolidation therapy, the relapse risk may not exceed 20% to 35% in 3 to 5 years.7-11  Consequently, these patients have, on average, no survival advantage with allogeneic HCT while in first remission because the transplant-related mortality is greater than the decrease in relapse rates afforded by the transplant.12,13  However, significant heterogeneity within t(8;21)(q22;q22) leukemias is widely appreciated. Several variables associated with worse outcome have been recognized in at least some studies, including a high WBC count and the presence of KIT or FLT3 mutations at diagnosis.7,14-19  Recent studies have highlighted this disease heterogeneity by identifying subsets of patients with distinct risks of disease recurrence based on the degree of reduction in RUNX1-RUNX1T1 transcripts.9-11,20-23  Specifically, in the largest study conducted to date on 163 patients, a >3 log reduction in transcript burden after the first course of induction therapy and a >4 log reduction after the first course of postremission therapy were associated with cumulative incidences of relapse (CIR) of only 4% and 13%, respectively; in this study, the clinicians were blinded to the MRD results, which thus did not influence patient management.9  Other studies came to qualitatively similar conclusions.10,22,23  By comparison, other series, including one on 116 patients, have suggested that MRD levels after induction have no prognostic relevance while they are informative after consolidation therapy.11,20  Direct comparison of these studies is hindered by the fact that different real-time quantitative PCR methodologies and data normalizations were employed, the timing of MRD assessment varied, definitions for the quality of MRD response differed, and variable chemotherapy regimens were used. Moreover, because of their retrospective nature without independent prospective confirmation, estimates from these studies may be subject to significant bias. Nonetheless, the available evidence suggests that optimal outcomes are achieved when patients with t(8;21)(q22;q22) AML obtain either a molecular remission or very significant reductions in RUNX1-RUNX1T1 transcripts with induction and postremission therapy; higher-intensity regimens may lead to deeper log reductions after the first course of chemotherapy.9,24  Emerging evidence from a study by Jourdan et al also suggests that information from posttreatment RUNX1-RUNX1T1 transcript levels may be preferable over high WBC or KIT/FLT3 mutational status to identify patients with high-risk t(8;21)(q22;q22) AML, as only MRD but not the other factors was of significant prognostic impact in multivariate analyses.25 

No randomized trial has so far tested whether patients with t(8;21)(q22;q22) AML who are in morphologic CR with suboptimal reduction in RUNX1-RUNX1T1 transcript levels would benefit from allogeneic HCT. However, a recent multicenter study suggests the potential value of such an approach.11  This study examined 116 patients aged 15 to 60 years with t(8;21)(q22;q22) AML who achieved morphologic remission with 1 to 2 courses of induction therapy according to the “7+3” schema, and then completed 2 cycles of consolidation therapy consisting of intermediate-dose cytarabine (1 to 2 g/m2 every 12 hours for 3 days) with or without an anthracycline. The lack of a major molecular response (defined as a >3 log reduction in RUNX1-RUNX1T1 transcript levels from baseline) after the second course of consolidation, or loss of major molecular response within 6 months, was used to categorize patients into high- and low-risk. High-risk patients were recommended to proceed to myeloablative allogeneic HCT, whereas low-risk patients were advised to undergo 6 additional cycles of chemotherapy (intermediate-dose cytarabine for cycles 3 and 4, then cytarabine 100 mg/m2 for 7 days in combination with an anthracycline [cycle 5], homoharringtonine [cycle 6], mitoxantrone [cycle 7], or aclamycin [cycle 8]); autologous HCT was permitted after 4 courses of consolidation. Sixty-nine of the 116 patients (59%) were compliant with this risk-adapted approach (with 40/69 high-risk patients undergoing allogeneic HCT and 29/47 low-risk patients receiving chemotherapy); the remaining patients served as non-risk adapted controls. Overall, the risk-adapted treatment approach resulted in survival outcomes similar to what has been reported earlier. In additional “as-treated” landmark analyses, allogeneic HCT was associated with a lower relapse rate and better survival, as compared with chemotherapy in high-risk patients (5-year CIR: 22.1% vs 78.9%, P < .0001; 5-year disease-free survival [DFS]: 61.7% vs 19.6%, P = .001; 5-year overall survival [OS]: 71.6% vs 26.7%, P = .007). Conversely, low-risk patients did not significantly benefit from allografting with regard to CIR (14.7% vs 5.3%, P = .33) and even had inferior DFS relative to those treated with chemotherapy/autologous HCT (70.3% vs 94.7%, P = .024).11  However, because of the possibility of significant bias in the above analyses, the benefit of allogeneic HCT for high-risk patients with t(8;21)(q22;q22) AML needs to be confirmed in further, better controlled studies; if large enough, such studies could also assess which role different transplant conditionings and donor sources might play for high-risk t(8;21)(q22;q22) AML. Although some patients with insufficiently reduced transcript levels can remain relapse-free even without allogeneic HCT, others will experience disease recurrence even when transplanted, therefore a more aggressive therapy (like allogeneic HCT) is not always associated with a better outcome in these patients.11  Future investigations will also need to carefully revisit the impact on OS, given that some studies with careful long-term follow-up data indicate that many patients with favorable-risk AML can be salvaged after first disease recurrence.7,26 

Recommendation.

For an adult with t(8;21)(q22;q22) AML who has achieved morphologic CR with persistence of RUNX1-RUNX1T1 transcripts with 1 course of induction therapy, no data exists to advocate the immediate use of allogeneic HCT. We suggest monitoring RUNX1-RUNX1T1 transcript levels and considering allogeneic HCT if a >3 log reduction in RUNX1-RUNX1T1 transcript levels from baseline is not reached after the 2nd course of consolidation or lost within 6 months (grade 2C). The optimal definition of what should be considered insufficient reduction in RUNX1-RUNX1T1 transcripts levels and the best timing of this assessment might change based on future data.

Case 2: MRD-directed therapy of intermediate-risk AML

Unlike favorable-risk patients, those with intermediate-risk disease based on revised Medical Research Council/National Cancer Research Institute (NCRI) or European LeukemiaNet criteria have generally been considered appropriate candidates for allogeneic HCT in first morphologic CR, particularly if comorbidity scores are low and an HLA-matched donor is available.12,13  This recommendation has recently been challenged by analyses from the Medical Research Council/NCRI suggesting that equivalent OS in this risk group may be achievable by delaying transplantation until after the first relapse.26  Even if an allogeneic HCT is performed in first morphologic CR, posttransplant relapse remains a substantial risk. Several retrospective studies have suggested that, on average, standard cytarabine-based consolidation chemotherapy before allogeneic HCT for AML patients of all risk groups in first morphologic CR does not improve posttransplant outcomes.27-30  Unfortunately, in all these trials, information on MRD was not available, and it is unknown whether additional postremission therapy could benefit a subset of patients with MRD. Numerous studies have convincingly demonstrated that MRD before allogeneic HCT is independently associated with a significantly increased risk of subsequent relapse and inferior survival.1,5  This relationship would justify risk-stratified treatment allocation, including the use of additional pretransplant chemotherapy, under the assumption that a further reduction of tumor burden would optimize the benefit conferred by allogeneic HCT. However, so far, no well-controlled studies (eg, investigating immediate vs delayed transplantation in MRD-positive patients with available donors) have been conducted to rigorously test this hypothesis. Because MRD is fundamentally also an indicator for the reduced sensitivity of leukemia cells to prior therapies, the presence of residual disease could thus simply mark those patients who are unlikely to be cured with subsequent similar-type therapies, even if disease levels are brought temporarily below the level of detection. Moreover, additional therapy to reduce the tumor burden in patients with MRD is associated with a risk of complications, such as organ toxicities or infections, that could delay or prevent transplantation, increase transplant-related mortality, and offset any potential benefit of improved disease control.

Recommendation.

There is currently no evidence to support or refute a benefit of additional chemotherapy for patients with intermediate-risk AML in first morphologic CR planned to undergo allogeneic HCT. The presence of MRD is not a contraindication to allogeneic HCT. Although MRD is associated with a several fold increased risk of post-HCT relapse even after adjustment for other predictive factors, up to 20% to 30% of patients with MRD at the time of transplantation experience prolonged DFS (ie, some MRD-positive patients will be salvaged with either myeloablative or nonmyeloablative conditioning allogeneic HCT).5,31,32  Outside of a clinical trial, we suggest transplantation without additional chemotherapy in this situation. We acknowledge the controversy regarding the value of allogeneic HCT in first CR for intermediate-risk AML, for which chemotherapy-based postremission therapy followed by close observation and transplantation in second CR, if obtained, may be a reasonable alternative.33 

In APL, molecular assessment of disease response has become standard practice, and MRD-directed therapy quite plausibly improves outcome, particularly in patients with high-risk disease.34,35  In the other forms of AML, attempts to measure MRD are complicated by the genetic and molecular complexity/diversity at initial presentation and disease evolution over the course of the illness with the possibility that minor subclones can emerge at the time of recurrence.36,37  Not all abnormalities are therefore equally suited as MRD parameters. Nonetheless, MRD is now established as an independent marker of increased relapse risk in non-APL AML and may be able to replace morphologic examinations as the gold standard for the assessment of treatment responses.4  On the other hand, conclusive data on the value of MRD-based, risk-stratified therapy is currently not available. A widely cited study in pediatric AML has used a combination of MRD measurement and genetic disease features to direct decisions on the second induction course and subsequent therapy, and based on a comparison with previous treatment cohorts suggested that this approach could improve outcomes.38  However, because of patient heterogeneity and improving supportive care measures, comparisons with “historic” control groups can be problematic,39  and better controlled and ideally randomized studies will ultimately be required to make a compelling argument for MRD-directed interventions in non-APL AML.

APL exemplifies that periodic MRD monitoring for patients with acute leukemia can be adopted as “standard” once its value is demonstrated. The impact of MRD monitoring in non-APL AML on survival, quality of life, and resource utilization is currently being explored in the United Kingdom’s NCRI AML17 trial. In this trial, patients with leukemias that have informative molecular markers are randomized to a “MRD monitoring vs no MRD monitoring” strategy, with the question of therapeutic intervention being left to the primary hematologist/oncologist. So far, however, the use of MRD monitoring as a routine tool in non-APL AML is hampered by interlaboratory differences in the assays and preferred analytical methods, varying approaches to defining MRD positivity/negativity with need to identify the cutoff values that are most informative at a given time point, differences in source of material (bone marrow vs peripheral blood) and correction for hemodilution if marrow is examined, and variation in the exact timing and frequency of MRD sampling. The remaining key issues with respect to MRD detection in AML are summarized in Table 2. Inconsistencies in MRD assays limit and complicate their interpretation and transferability of results and, likely, curb the enthusiasm of regulatory authorities to use MRD as an end point in the drug approval process. As an important step toward optimized use of MRD assays, it will be critical to address these current shortcomings through the adoption of standardized methodologic approaches with frequent external quality control, and validation and clarification of regulatory considerations. Although their need is well recognized, efforts in that direction have only begun.4,40-44  In light of these limitations, we would highly encourage the research community to work toward standardized methods for the detection and monitoring of MRD levels and use them as soon as they become available, and to conduct well controlled, ideally randomized trials for evaluating the value of MRD-directed treatment escalation or de-escalation in AML. Recent studies in acute lymphoblastic leukemia demonstrate that such trials are feasible and can provide definitive evidence that modification of postremission treatment intensity based on MRD status can optimize treatment outcomes.45,46 

Table 2

MRD detection in AML: remaining key issues

Detection methodIssue
All Definition of MRD positivity/negativity 
 Differences in source of material (bone marrow vs peripheral blood) 
 For bone marrow: hemodilution 
 Variation in timing/frequency of MRD sampling 
 Regulatory approval/validation of assay 
 Insufficient assay sensitivity 
 Disease evolution with change in targets suitable for MRD detection 
 Requirement of expertise for data interpretation 
 Sample degradation 
Molecular Choice of target(s), target specificity 
 Quality of cDNA synthesis 
 Efficacy of PCR amplification 
 Insufficient primer specificity 
 Sensitivity of target gene overexpression limited by normal tissue expression 
 Target stability 
 Data normalization; choice of housekeeping gene 
 Contamination 
Flow cytometry Choice of antigens and antibody panels 
 Lack of immunophenotypic abnormalities 
 Lack of diagnostic specimen to determine immunophenotypic abnormalities sufficient for MRD detection 
 Choice of analysis strategy for MRD detection (diagnostic leukemia-associated immunophenotypes vs “different-from-normal” analysis) 
 Lack of automatic analysis algorithms 
Detection methodIssue
All Definition of MRD positivity/negativity 
 Differences in source of material (bone marrow vs peripheral blood) 
 For bone marrow: hemodilution 
 Variation in timing/frequency of MRD sampling 
 Regulatory approval/validation of assay 
 Insufficient assay sensitivity 
 Disease evolution with change in targets suitable for MRD detection 
 Requirement of expertise for data interpretation 
 Sample degradation 
Molecular Choice of target(s), target specificity 
 Quality of cDNA synthesis 
 Efficacy of PCR amplification 
 Insufficient primer specificity 
 Sensitivity of target gene overexpression limited by normal tissue expression 
 Target stability 
 Data normalization; choice of housekeeping gene 
 Contamination 
Flow cytometry Choice of antigens and antibody panels 
 Lack of immunophenotypic abnormalities 
 Lack of diagnostic specimen to determine immunophenotypic abnormalities sufficient for MRD detection 
 Choice of analysis strategy for MRD detection (diagnostic leukemia-associated immunophenotypes vs “different-from-normal” analysis) 
 Lack of automatic analysis algorithms 

cDNA, complementary DNA.

Key issues for MRD detection in AML have been previously highlighted by several investigators.1-4,36,40,41,43,44,47-51 

The online version of this article contains a data supplement.

The authors thank Drs Elihu H. Estey and Brent L. Wood for critical reading of the manuscript.

This study was supported by grants from the National Institute for Health Research under its Programme Grants for Applied Research (RP- PG-0108-10093) (D.G.), the Leukaemia & Lymphoma Research of Great Britain, the Guy’s and St. Thomas’ Charity, and the MRD Workpackage (WP12) of the European LeukemiaNet (D.G.), the Else Kröner-Fresenius-Stiftung (P80/08/A65/08) (R.F.S.), the German Bundesministerium für Bildung und Forschung (01GI9981, 01KG0605, and 01KG1004) (R.F.S.), and the Deutsche José Carreras Leukämie-Stiftung (DJCLS H 09/22) (R.F.S.). The views expressed are those of the authors and not necessarily those of the National Health Service, the National Institute for Health Research, or the Department of Health. R.B.W. is a Leukemia & Lymphoma Society scholar in Clinical Research.

Contribution: S.K. and R.B.W. were responsible for the concept of this review, contributed to the literature search data collection/quality assessment, analyzed and interpreted data, and wrote the manuscript; R.F.S. and D.G. analyzed and interpreted data, and critically revised the manuscript; and V.E.D.Y. designed the literature search, performed the data extraction and quality assessment, analyzed and interpreted data, and critically revised the manuscript.

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

Correspondence: Sabine Kayser, Department of Internal Medicine V, University Hospital of Heidelberg, Im Neuenheimer Feld 410; 69120 Heidelberg, Germany; e-mail: sabine.kayser@med.uni-heidelberg.de.

1
Buccisano
 
F
Maurillo
 
L
Del Principe
 
MI
, et al. 
Prognostic and therapeutic implications of minimal residual disease detection in acute myeloid leukemia.
Blood
2012
, vol. 
119
 
2
(pg. 
332
-
341
)
2
Paietta
 
E
Minimal residual disease in acute myeloid leukemia: coming of age.
Hematology Am Soc Hematol Educ Program
2012
, vol. 
2012
 (pg. 
35
-
42
)
3
Hourigan
 
CS
Karp
 
JE
Minimal residual disease in acute myeloid leukaemia.
Nat Rev Clin Oncol
2013
, vol. 
10
 
8
(pg. 
460
-
471
)
4
Grimwade
 
D
Freeman
 
SD
Defining minimal residual disease in acute myeloid leukemia: which platforms are ready for “prime time”?
Blood
2014
, vol. 
124
 
23
(pg. 
3345
-
3355
)
5
Buckley
 
SA
Appelbaum
 
FR
Walter
 
RB
Prognostic and therapeutic implications of minimal residual disease at the time of transplantation in acute leukemia.
Bone Marrow Transplant
2013
, vol. 
48
 
5
(pg. 
630
-
641
)
6
Guyatt
 
GH
Oxman
 
AD
Vist
 
GE
, et al. 
GRADE Working Group
GRADE: an emerging consensus on rating quality of evidence and strength of recommendations.
BMJ
2008
, vol. 
336
 
7650
(pg. 
924
-
926
)
7
Schlenk
 
RF
Benner
 
A
Krauter
 
J
, et al. 
Individual patient data-based meta-analysis of patients aged 16 to 60 years with core binding factor acute myeloid leukemia: a survey of the German Acute Myeloid Leukemia Intergroup.
J Clin Oncol
2004
, vol. 
22
 
18
(pg. 
3741
-
3750
)
8
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
)
9
Yin
 
JA
O’Brien
 
MA
Hills
 
RK
Daly
 
SB
Wheatley
 
K
Burnett
 
AK
Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: results of the United Kingdom MRC AML-15 trial.
Blood
2012
, vol. 
120
 
14
(pg. 
2826
-
2835
)
10
Hoyos
 
M
Nomdedeu
 
JF
Esteve
 
J
, et al. 
Core binding factor acute myeloid leukemia: the impact of age, leukocyte count, molecular findings, and minimal residual disease.
Eur J Haematol
2013
, vol. 
91
 
3
(pg. 
209
-
218
)
11
Zhu
 
HH
Zhang
 
XH
Qin
 
YZ
, et al. 
MRD-directed risk stratification treatment may improve outcomes of t(8;21) AML in the first complete remission: results from the AML05 multicenter trial.
Blood
2013
, vol. 
121
 
20
(pg. 
4056
-
4062
)
12
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
)
13
Cornelissen
 
JJ
Gratwohl
 
A
Schlenk
 
RF
, et al. 
The European LeukemiaNet AML Working Party consensus statement on allogeneic HSCT for patients with AML in remission: an integrated-risk adapted approach.
Nat Rev Clin Oncol
2012
, vol. 
9
 
10
(pg. 
579
-
590
)
14
Nguyen
 
S
Leblanc
 
T
Fenaux
 
P
, et al. 
A white blood cell index as the main prognostic factor in t(8;21) acute myeloid leukemia (AML): a survey of 161 cases from the French AML Intergroup.
Blood
2002
, vol. 
99
 
10
(pg. 
3517
-
3523
)
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
, vol. 
107
 
5
(pg. 
1791
-
1799
)
16
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
)
17
Boissel
 
N
Leroy
 
H
Brethon
 
B
, et al. 
Acute Leukemia French Association (ALFA); Leucémies Aiguës Myéloblastiques de l’Enfant (LAME) Cooperative Groups
Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML).
Leukemia
2006
, vol. 
20
 
6
(pg. 
965
-
970
)
18
Paschka
 
P
Marcucci
 
G
Ruppert
 
AS
, et al. 
Cancer and Leukemia Group B
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
)
19
Allen
 
C
Hills
 
RK
Lamb
 
K
, et al. 
The importance of relative mutant level for evaluating impact on outcome of KIT, FLT3 and CBL mutations in core-binding factor acute myeloid leukemia.
Leukemia
2013
, vol. 
27
 
9
(pg. 
1891
-
1901
)
20
Schnittger
 
S
Weisser
 
M
Schoch
 
C
Hiddemann
 
W
Haferlach
 
T
Kern
 
W
New score predicting for prognosis in PML-RARA+, AML1-ETO+, or CBFBMYH11+ acute myeloid leukemia based on quantification of fusion transcripts.
Blood
2003
, vol. 
102
 
8
(pg. 
2746
-
2755
)
21
Krauter
 
J
Görlich
 
K
Ottmann
 
O
, et al. 
Prognostic value of minimal residual disease quantification by real-time reverse transcriptase polymerase chain reaction in patients with core binding factor leukemias.
J Clin Oncol
2003
, vol. 
21
 
23
(pg. 
4413
-
4422
)
22
Leroy
 
H
de Botton
 
S
Grardel-Duflos
 
N
, et al. 
Prognostic value of real-time quantitative PCR (RQ-PCR) in AML with t(8;21).
Leukemia
2005
, vol. 
19
 
3
(pg. 
367
-
372
)
23
Weisser
 
M
Haferlach
 
C
Hiddemann
 
W
Schnittger
 
S
The quality of molecular response to chemotherapy is predictive for the outcome of AML1-ETO-positive AML and is independent of pretreatment risk factors.
Leukemia
2007
, vol. 
21
 
6
(pg. 
1177
-
1182
)
24
Prebet
 
T
Bertoli
 
S
Delabesse
 
E
, et al. 
Impact of anthracycline dose intensification on minimal residual disease and outcome of core binding factor acute myeloid leukemias. [abstract]
Blood
2013
, vol. 
122
 
21
pg. 
2681
 
25
Jourdan
 
E
Boissel
 
N
Chevret
 
S
, et al. 
French AML Intergroup
Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia.
Blood
2013
, vol. 
121
 
12
(pg. 
2213
-
2223
)
26
Burnett
 
AK
Goldstone
 
A
Hills
 
RK
, et al. 
Curability of patients with acute myeloid leukemia who did not undergo transplantation in first remission.
J Clin Oncol
2013
, vol. 
31
 
10
(pg. 
1293
-
1301
)
27
Tallman
 
MS
Rowlings
 
PA
Milone
 
G
, et al. 
Effect of postremission chemotherapy before human leukocyte antigen-identical sibling transplantation for acute myelogenous leukemia in first complete remission.
Blood
2000
, vol. 
96
 
4
(pg. 
1254
-
1258
)
28
Cahn
 
JY
Labopin
 
M
Sierra
 
J
, et al. 
Acute Leukaemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT)
No impact of high-dose cytarabine on the outcome of patients transplanted for acute myeloblastic leukaemia in first remission.
Br J Haematol
2000
, vol. 
110
 
2
(pg. 
308
-
314
)
29
Warlick
 
ED
Paulson
 
K
Brazauskas
 
R
, et al. 
Effect of postremission therapy before reduced-intensity conditioning allogeneic transplantation for acute myeloid leukemia in first complete remission.
Biol Blood Marrow Transplant
2014
, vol. 
20
 
2
(pg. 
202
-
208
)
30
Yeshurun
 
M
Labopin
 
M
Blaise
 
D
, et al. 
Impact of postremission consolidation chemotherapy on outcome after reduced-intensity conditioning allogeneic stem cell transplantation for patients with acute myeloid leukemia in first complete remission: a report from the Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation.
Cancer
2014
, vol. 
120
 
6
(pg. 
855
-
863
)
31
Walter
 
RB
Buckley
 
SA
Pagel
 
JM
, et al. 
Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission.
Blood
2013
, vol. 
122
 
10
(pg. 
1813
-
1821
)
32
Walter
 
RB
Gyurkocza
 
B
Storer
 
BE
, et al. 
Comparison of minimal residual disease as outcome predictor for AML patients in first complete remission undergoing myeloablative or nonmyeloablative allogeneic hematopoietic cell transplantation.
Leukemia
2015
, vol. 
29
 
1
(pg. 
137
-
144
)
33
Stone
 
RM
Acute myeloid leukemia in first remission: to choose transplantation or not?
J Clin Oncol
2013
, vol. 
31
 
10
(pg. 
1262
-
1266
)
34
Grimwade
 
D
Jovanovic
 
JV
Hills
 
RK
Can we say farewell to monitoring minimal residual disease in acute promyelocytic leukaemia?
Best Pract Res Clin Haematol
2014
, vol. 
27
 
1
(pg. 
53
-
61
)
35
National Comprehensive Cancer Network
 
NCCN clinical practice guidelines in oncology: acute myeloid leukemia. Version 1. Available at: http://www.nccn.org. Accessed January 11, 2015
36
Zeijlemaker
 
W
Gratama
 
JW
Schuurhuis
 
GJ
Tumor heterogeneity makes AML a “moving target” for detection of residual disease.
Cytometry B Clin Cytom
2014
, vol. 
86
 
1
(pg. 
3
-
14
)
37
Graubert
 
TA
Brunner
 
AM
Fathi
 
AT
New molecular abnormalities and clonal architecture in AML: from reciprocal translocations to whole-genome sequencing.
Am Soc Clin Oncol Educ Book
2014
(pg. 
e334
-
340
)
38
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
, vol. 
11
 
6
(pg. 
543
-
552
)
39
Walter
 
RB
Appelbaum
 
FR
Tallman
 
MS
Weiss
 
NS
Larson
 
RA
Estey
 
EH
Shortcomings in the clinical evaluation of new drugs: acute myeloid leukemia as paradigm.
Blood
2010
, vol. 
116
 
14
(pg. 
2420
-
2428
)
40
Beillard
 
E
Pallisgaard
 
N
van der Velden
 
VH
, et al. 
Evaluation of candidate control genes for diagnosis and residual disease detection in leukemic patients using “real-time” quantitative reverse-transcriptase polymerase chain reaction (RQ-PCR) —a Europe Against Cancer Program.
Leukemia
2003
, vol. 
17
 
12
(pg. 
2474
-
2486
)
41
Gabert
 
J
Beillard
 
E
van der Velden
 
VH
, et al. 
Standardization and quality control studies of “real-time” quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia—a Europe Against Cancer Program.
Leukemia
2003
, vol. 
17
 
12
(pg. 
2318
-
2357
)
42
Kröger
 
N
Bacher
 
U
Bader
 
P
, et al. 
NCI First International Workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: report from the Committee on Disease-Specific Methods and Strategies for Monitoring Relapse following Allogeneic Stem Cell Transplantation. Part I: methods, acute leukemias, and myelodysplastic syndromes. [published correction appears in Biol Blood Marrow Transplant. 2010;16(12):1752].
Biol Blood Marrow Transplant
2010
, vol. 
16
 
9
(pg. 
1187
-
1211
)
43
Østergaard
 
M
Nyvold
 
CG
Jovanovic
 
JV
, et al. 
Development of standardized approaches to reporting of minimal residual disease data using a reporting software package designed within the European LeukemiaNet.
Leukemia
2011
, vol. 
25
 
7
(pg. 
1168
-
1173
)
44
Feller
 
N
van der Velden
 
VH
Brooimans
 
RA
, et al. 
Defining consensus leukemia-associated immunophenotypes for detection of minimal residual disease in acute myeloid leukemia in a multicenter setting.
Blood Cancer J
2013
, vol. 
3
 pg. 
e129
 
45
Vora
 
A
Goulden
 
N
Wade
 
R
, et al. 
Treatment reduction for children and young adults with low-risk acute lymphoblastic leukaemia defined by minimal residual disease (UKALL 2003): a randomised controlled trial.
Lancet Oncol
2013
, vol. 
14
 
3
(pg. 
199
-
209
)
46
Vora
 
A
Goulden
 
N
Mitchell
 
C
, et al. 
Augmented post-remission therapy for a minimal residual disease-defined high-risk subgroup of children and young people with clinical standard-risk and intermediate-risk acute lymphoblastic leukaemia (UKALL 2003): a randomised controlled trial.
Lancet Oncol
2014
, vol. 
15
 
8
(pg. 
809
-
818
)
47
van der Velden
 
VH
Boeckx
 
N
Gonzalez
 
M
, et al. 
Europe Against Cancer Program
Differential stability of control gene and fusion gene transcripts over time may hamper accurate quantification of minimal residual disease—a study within the Europe Against Cancer Program.
Leukemia
2004
, vol. 
18
 
4
(pg. 
884
-
886
)
48
Kern
 
W
Bacher
 
U
Haferlach
 
C
Schnittger
 
S
Haferlach
 
T
The role of multiparameter flow cytometry for disease monitoring in AML.
Best Pract Res Clin Haematol
2010
, vol. 
23
 
3
(pg. 
379
-
390
)
49
Hokland
 
P
Ommen
 
HB
Nyvold
 
CG
Roug
 
AS
Sensitivity of minimal residual disease in acute myeloid leukaemia in first remission—methodologies in relation to their clinical situation.
Br J Haematol
2012
, vol. 
158
 
5
(pg. 
569
-
580
)
50
Ravandi
 
F
Jorgensen
 
JL
Monitoring minimal residual disease in acute myeloid leukemia: ready for prime time?
J Natl Compr Canc Netw
2012
, vol. 
10
 
8
(pg. 
1029
-
1036
)
51
Schrappe
 
M
Minimal residual disease: optimal methods, timing, and clinical relevance for an individual patient.
Hematology Am Soc Hematol Educ Program
2012
, vol. 
2012
 (pg. 
137
-
142
)

Supplemental data

Sign in via your Institution