Significant improvements have been made in the treatment of acute lymphoblastic leukemia (ALL) during the past 2 decades, and measurement of submicroscopic (minimal) levels of residual disease (MRD) is increasingly used to monitor treatment efficacy. For a better comparability of MRD data, there are ongoing efforts to standardize MRD quantification using real-time quantitative PCR of clonal immunoglobulin and T-cell receptor gene rearrangements, real-time quantitative-based detection of fusion gene transcripts or breakpoints, and multiparameter flow cytometric immunophenotyping. Several studies have demonstrated that MRD assessment in childhood and adult ALL significantly correlates with clinical outcome. MRD detection is particularly useful for evaluation of treatment response, but also for early assessment of an impending relapse. Therefore, MRD has gained a prominent position in many ALL treatment studies as a tool for tailoring therapy with growing evidence that MRD supersedes most conventional stratification criteria at least for Ph-negative ALL. Most study protocols on adult ALL follow a 2-step approach with a first classic pretherapeutic and a second MRD-based risk stratification. Here we discuss whether and how MRD is ready to be used as main decisive marker and whether pretherapeutic factors and MRD are really competing or complementary tools to individualize treatment.

Treatment outcome in acute lymphoblastic leukemia (ALL) patients depends on a combination of multiple factors, such as properties of the leukemic cells (eg, proliferative capacity, susceptibility to drugs, and other escape mechanisms), host factors (eg, general fitness and concomitant diseases, treatment compliance, host pharmacodynamics and pharmacogenetics), and treatment given to eradicate the disease. Many of intrinsic leukemia cell and host factors have already been elucidated with immunologic and molecular methods and are ongoing translated into providing prognostic information (Table 1). Based on retrospective analyses of large cohorts of patients, conventional pretherapeutic risk criteria, including age, elevated white blood cell count at diagnosis, adverse immunophenotypic features, and cytogenetic as well as molecular aberrations provide the basis for upfront risk stratification in current treatment protocols. It has to be acknowledged, however, that these advances in our understanding of ALL biology in the past have only to a limited extent been accompanied by improved survival of adult ALL patients with relapse still being the main clinical problem. The source of these relapses is the persistence of minimal residual disease (MRD) that is undetectable by standard diagnostic techniques. Several studies have shown that detection of MRD in childhood and adult ALL is an independent risk parameter of high clinical relevance, both in de novo and relapsed ALL as well as in ALL patients undergoing stem cell transplantation (SCT).1-14  Consequently, an increasing number of treatment protocols use MRD as a tool for treatment stratification. In addition, postremission MRD monitoring is also used to predict an impending relapse and to start preemptive salvage treatment in time. Therefore, MRD is not only a prognostic factor but challenges the traditional concept of defining remission and relapse. Prerequisite for application of MRD for treatment tailoring is an adequate, sensitive, and standardized MRD methodology. The focus of this Perspective is therefore to highlight pros and cons of the different MRD techniques, to present the published experience of MRD analysis and MRD-guided treatment in adult ALL, and to discuss the value of MRD in different clinical settings compared with other prognostic factors.

Table 1

Pretherapeutic factors associated with outcome in adult ALL81-83 

FactorCategoryPrognostic impactPotential impact on targeted therapy
Age — Worse outcome with advancing age84,85  — 
White blood cell count at diagnosis B: > 30 × 109/L (B) High WBC associated with poor prognosis84,85  — 
 T: > 100 × 109/L (T)   
Immunophenotype CD20 expression Conflicting data concerning prognosis74,75  Monoclonal antibodies 
 T versus B Independent prognostic significance (T-ALL with better prognosis) mainly in early studies84,85  Monoclonal antibodies Bispecific T-cell engager nelarabine 
Cytogenetics t(9;22)/BCR-ABL Poor prognosis86,87  TKI 
 t(4;11)/MLL-AF4 Poor prognosis86,87  — 
 t(8;14)   
 Hypodiploidy*   
 Near triploidy   
 Complex karyotype   
 t(1;19) Conflicting data concerning prognosis88,89  — 
 High hyperdiploidy Better prognosis86  — 
 del(9p)   
Specific molecular alterations JAK mutations Emerging significance of poor prognosis90  JAK inhibitors 
 IKZF Deletions/sequence mutations Emerging significance of poor prognosis91,92   
 CRLF2 overexpression Emerging significance (mainly childhood ALL) of poor prognosis92  CRLF antibodies 
 ERG/BAALC expression Conflicting data concerning prognosis93,94   
 NOTCH1 mutations Conflicting data concerning prognosis95,96  NOTCH1 targeting 
FactorCategoryPrognostic impactPotential impact on targeted therapy
Age — Worse outcome with advancing age84,85  — 
White blood cell count at diagnosis B: > 30 × 109/L (B) High WBC associated with poor prognosis84,85  — 
 T: > 100 × 109/L (T)   
Immunophenotype CD20 expression Conflicting data concerning prognosis74,75  Monoclonal antibodies 
 T versus B Independent prognostic significance (T-ALL with better prognosis) mainly in early studies84,85  Monoclonal antibodies Bispecific T-cell engager nelarabine 
Cytogenetics t(9;22)/BCR-ABL Poor prognosis86,87  TKI 
 t(4;11)/MLL-AF4 Poor prognosis86,87  — 
 t(8;14)   
 Hypodiploidy*   
 Near triploidy   
 Complex karyotype   
 t(1;19) Conflicting data concerning prognosis88,89  — 
 High hyperdiploidy Better prognosis86  — 
 del(9p)   
Specific molecular alterations JAK mutations Emerging significance of poor prognosis90  JAK inhibitors 
 IKZF Deletions/sequence mutations Emerging significance of poor prognosis91,92   
 CRLF2 overexpression Emerging significance (mainly childhood ALL) of poor prognosis92  CRLF antibodies 
 ERG/BAALC expression Conflicting data concerning prognosis93,94   
 NOTCH1 mutations Conflicting data concerning prognosis95,96  NOTCH1 targeting 

— indicates not applicable; CRLF2, cytokine receptor-like factor 2; ERG, v-ets erythroblastosis virus E26 oncogene homolog (avian); BAALC, brain and acute leukemia, cytoplasmic; IKZF, IKAROS family zinc finger; JAK, Janus kinase; TKI, tyrosine kinase inhibitors; and WBC, white blood cell count.

*

< 44 chromosomes/leukemic cell.

> 5 abnormalities.

> 50 chromosomes/leukemic cell.

MRD assessment relies on the identification of specific molecular or immunophenotypic markers on the leukemia cells. Flow cytometry (FCM) is applied to detect combinations of cell markers that are present on the leukemic but not on normal bone marrow cells. PCR is used to detect leukemia-specific fusion transcripts (eg, BCR-ABL) or clone specific immunoglobulin (Ig) or T-cell receptor (TCR) genes.

Each of these techniques has its own strengths and limitations, which are summarized in Table 2 and are described in more detail within the next 3 sections.

Table 2

Features of techniques currently employed for MRD detection in ALL

General statementsProsCons
Flow cytometry Aberrant antigene expression Applicable for almost all ALL patients Immunophenotypic shifts of leukemic cells 
 Sensitivity depends on technology (number of colors) and on cell input Availability of methodology in many laboratories Expanded and altered precursor-B-cell compartment during regeneration 
  Rapid Low cellularity during/after induction 
  Quantitative Relatively high costs (depends on cell input, number of markers/colors and ulterior cytometer use) 
  Additional information on benign cells Limited sensitivity/applicability using 3- to 4-color flow cytometry 
  Additional information on malignant cells ≥ 6-color flow cytometry: extensive knowledge and experience for sensitive and standardized analysis needed 
  Identification and monitoring of treatment targets possible No sample asservation and retrospective analysis possible (on-site availability of expert operators necessary) 
  Growing standardization (mainly throughout Europe)  
DNA-based RQ-PCR Targets mainly Ig and TCR gene rearrangements but also MLL gene rearrangements or SIL-TAL-deletions Applicable for almost all ALL patients Time-consuming marker characterization 
  High sensitivity Potential instability of targets (clonal evolution phenomena, therefore need for preferably 2 targets/patient) 
  High degree of standardization Extensive knowledge and experience needed 
  Accepted and uniformly used definition of quantitative range and sensitivity Relatively expensive 
  Well-established stratification tool in various clinical protocols  
  Most published data for evidence-based treatment decisions  
  Stability of DNA (multicenter setting, shipment time)  
  Possibility of sample storage and retrospective/batch analysis  
Quantitative RT-PCR of fusion transcripts Targets mainly BCR-ABL transcripts (∼ 35% of adult B-cell precursor ALL) High sensitivity Useful only in a minority of patients 
  Unequivocal link with leukemic/preleukemic clone Instability of RNA 
  Stability of target during course of treatment Uncertain quantitation because of unknown number of RNA transcripts/cell (potential differences during course of treatment) 
  Fast False positivity resulting from cross-contamination 
  Relatively cheap Standardization necessary 
General statementsProsCons
Flow cytometry Aberrant antigene expression Applicable for almost all ALL patients Immunophenotypic shifts of leukemic cells 
 Sensitivity depends on technology (number of colors) and on cell input Availability of methodology in many laboratories Expanded and altered precursor-B-cell compartment during regeneration 
  Rapid Low cellularity during/after induction 
  Quantitative Relatively high costs (depends on cell input, number of markers/colors and ulterior cytometer use) 
  Additional information on benign cells Limited sensitivity/applicability using 3- to 4-color flow cytometry 
  Additional information on malignant cells ≥ 6-color flow cytometry: extensive knowledge and experience for sensitive and standardized analysis needed 
  Identification and monitoring of treatment targets possible No sample asservation and retrospective analysis possible (on-site availability of expert operators necessary) 
  Growing standardization (mainly throughout Europe)  
DNA-based RQ-PCR Targets mainly Ig and TCR gene rearrangements but also MLL gene rearrangements or SIL-TAL-deletions Applicable for almost all ALL patients Time-consuming marker characterization 
  High sensitivity Potential instability of targets (clonal evolution phenomena, therefore need for preferably 2 targets/patient) 
  High degree of standardization Extensive knowledge and experience needed 
  Accepted and uniformly used definition of quantitative range and sensitivity Relatively expensive 
  Well-established stratification tool in various clinical protocols  
  Most published data for evidence-based treatment decisions  
  Stability of DNA (multicenter setting, shipment time)  
  Possibility of sample storage and retrospective/batch analysis  
Quantitative RT-PCR of fusion transcripts Targets mainly BCR-ABL transcripts (∼ 35% of adult B-cell precursor ALL) High sensitivity Useful only in a minority of patients 
  Unequivocal link with leukemic/preleukemic clone Instability of RNA 
  Stability of target during course of treatment Uncertain quantitation because of unknown number of RNA transcripts/cell (potential differences during course of treatment) 
  Fast False positivity resulting from cross-contamination 
  Relatively cheap Standardization necessary 

Multiparameter flow cytometry

MRD measurement by FCM is based on the detection of leukemia-associated immunophenotypes that can be used to distinguish them from normal hematopoietic cells. Leukemia-associated immunophenotypes usually describe a subpopulation of cells of a given lineage at a particular differentiation stage with aberrant molecular expression patterns, asynchrony, and/or profound overexpression or underexpression of molecules.15  Therefore, unlike molecular methods, FCM needs the identification of a cluster of events for definition of MRD positivity. Using 4-color flow cytometry, leukemia-associated immunophenotypes can be identified in ∼ 90% of B-precursor and > 95% of all T-ALL patients reaching detection limits of 10−3-10−4.5,16-22 

The major advantage of FCM is its rapidity, which allows reporting of quantitative results within 1 day. This is specifically useful when MRD results are needed quickly to guide therapy. In addition, FCM allows the simultaneous assessment of cell qualities requisite for emerging targeted therapies in ALL.23,24  A potential pitfall of the method results from similarities between leukemic lymphoblasts and nonmalignant lymphoid precursors in various phases of regeneration or chemotherapy-induced alterations that may lead to false positivity.18,25  In addition, phenotypic shifts frequently occur in leukemic cells during induction among others because of steroid-induced gene expression modulation.23-28  In times of targeted therapies (eg, anti-CD19, anti-CD20, anti-CD22, or anti-CD33 treatment), also completely unknown marker shifts may occur potentially influencing detectability of residual leukemic cells. Therefore, interpretation of MRD FCM data requires a deep understanding of the expression patterns of benign hematogones and leukemic cells within different treatment phases and should be restricted to reference laboratories, even if FCM is broadly available in many hematolgoy centers.16 

In addition, careful standardization of experimental setup and interpretation is needed to obtain comparable MRD results in multicenter studies, which is one topic of international study groups.29-33  Additional efforts focus on improving the method and its sensitivity through usage of more colors (≥ 8), inclusion of new markers, possibly more specific for leukemic cells, and development of new software for fast and easy automated data analysis.

PCR analysis of Ig and TCR gene rearrangements

Ig and TCR gene rearrangements represent fingerprint-like DNA regions of individual lymphoid cells and its descendants, making them attractive targets for clone specific PCR. Prerequisite is the molecular characterization of leukemia-specific Ig/TCR gene rearrangements for each individual patient, which is possible for the majority (> 95%) of B- and T-lineage ALL. Quantification of these target genes is nowadays performed with quantitative real-time quantitative (RQ)–PCR that allows for a higher degree of automation and sample throughput without the need of post-PCR manipulation of samples.34-37  Similarly to rearranged immune genes, also some leukemia specific translocations (in particular MLL rearrangements) can be used as targets for DNA-based quantitative RQ-PCR analysis after the rearrangement break point at the DNA level has been characterized.38,39  Sensitivity is determined exactly for each assay and generally reaches 1:104-1:105, which is ∼ 0.5-1.0 log higher than for published FCM-based MRD assays.19,21,40 

Right from the start, DNA-based MRD diagnostics has been highly standardized through the efforts of the EuroMRD Group. Guidelines are published for determination of linearity, sensitivity, specificity, and reproducibility for each clone-specific assay.36  Usage of DNA as analytical sample allows long shipping times, sample storage, and retrospective cumulative PCR analyses of sample batches. Nevertheless, there are some caveats, including clonal evolution of Ig/TCR genes, which may lead to false negativity. It is therefore recommended to use at least 2 independent targets. On the other hand, false-positive MRD results cannot be completely excluded as massive regeneration of normal lymphoid progenitors might lead to low levels of nonspecific amplification.41,42  In addition, the method needs the time-consuming initial characterization of the leukemic Ig/TCR rearrangements with a panel of different PCR and sequencing reactions. However, recent advances in sequencing technology (next-generation sequencing) will probably accelerate this process in the near future and even add a new molecular technology for MRD assessment.43 

PCR analysis of fusion transcripts

Leukemia-specific gene fusions represent ideal targets for MRD detection because they are linked to the oncogenic process and are therefore highly specific. Only few translocations (mainly MLL rearrangements38  and SIL-TAL1 translocations) are analyzed on DNA level, the broadest routine clinical use is the quantification of aberrant BCR-ABL mRNA transcripts in Philadelphia (Ph)–positive ALL.

Advantages of quantitative reverse transcriptase-PCR (RT-PCR) to evaluate fusion transcripts in Ph-positive ALL include the high sensitivity of up to 10−6 because of presence of multiple specific mRNA-transcripts per cell. Furthermore, these transcripts allow the usage of the same primer/probe-combinations for quantitative RT-PCR of many patients, thus keeping the costs for the individual analyses low. In addition, there is a broad experience in BCR-ABL PCR assays because it is widely used for MRD monitoring in CML. However, methodologies are not fully comparable for both entities (dominance of minor break-point transcript in Ph-positive ALL compared with major breakpoint cluster region in CML, differences in relevant MRD thresholds with higher sensitivities being required in Ph-positive ALL).

A crucial point for BCR-ABL PCR is the sample quality, which is adversely influenced by long transport times resulting in degradation of RNA. In addition, lack of standardization of RNA extraction, cDNA synthesis, selection of housekeeping genes, and variation in the way quantitative RT-PCR analyses are carried out lead to substantial differences in results and carry the risk of false negativity. In addition, false positivity cannot be ruled out (eg, because of cross-contamination) and is observed in external quality controls even among experienced laboratories. There are ongoing efforts to standardize methodology and interpretation to allow a better comparability of PCR results within different clinical trials.44 

Proposal for a uniform description of MRD results

An important step toward a better comparability of MRD data are the standardized and uniform description of MRD results. Irrespective of the methodology used a testing result may be (1) negative, (2) positive without being quantifiable because of the rarity of the event, and (3) positive at a quantifiable level (Figure 1).

Figure 1

Proposals for definition of MRD terms in ALL. Data from Brüggemann et al16 with permission.

Figure 1

Proposals for definition of MRD terms in ALL. Data from Brüggemann et al16 with permission.

Close modal

Concerning Ig/TCR-PCR, these results are defined by the EuroMRD Group based on the general performance, linearity, reproducibility, and background of the individual assay.36  In addition, for BCR-ABL and FCM MRD, there are ongoing international efforts to define generally accepted guidelines for standardized analysis and interpretation of results.30,33,44 

Based on these efforts, also a uniform MRD terminology referring to terms that are already established for cytomorphology with definitions of complete MRD response, MRD persistence, and MRD relapse seems feasible16  (Figure 1).

The 2 major applications of MRD in adult ALL are (1) the assessment of response to initial therapy for definition of MRD-based risk groups and (2) the subsequent monitoring of remission patients to detect MRD reoccurrence and allow a preemptive salvage treatment.

MRD for initial remission assessment

The most significant application of MRD for de novo ALL is the sensitive assessment of treatment efficacy in patients reaching a complete morphological remission, thereby refining initial risk stratification. Compared with childhood ALL with reports on several thousand patients,2,45  MRD in adult ALL has been studied less extensively. Nevertheless, also in adult ALL large studies have shown that initial MRD kinetics is highly predictive for outcome (Table 3).

Table 3

Selected MRD studies for assessment of treatment response in adult ALL

StudyNo. and risk group of patientsTreatmentMRD assessment/prognostic significance of MRD/conclusionsRetaining prognostic significance of classic risk factors/comments
Ph-negative ALL*     
    Vidriales22  (2003) 102 patients According to clinical risk factors: FCM (3 colors) during (d+14) and after (d+35) induction: MRD most relevant independent prognostic factor 
 B- and T-lineage     Chemo (n = 65) Prognostic influence of MRD at both time points Additional independent prognostic factors: age, WBC, Ph positivity 
 CR after induction     Plus auto-SCT (n = 14) Poor prognosis in patients with MRD persistence ≥ 5 × 10−3 d+14 and/or ≥ 5 × 10−4 d+35
Excellent prognosis in small subgroup of patients with early MRD clearance (< 3 × 10−4, d+14) 
      Plus allo-SCT (n = 23)  
    Brüggemann3  (2006) 196 patients
B- and T-lineage
SR ALL 
Chemo RQ-PCR (Ig/TCR) at 9 time points (d+11, d+24, d+44, d+71, wk+16, wk+22, wk+30, wk+41, wk+52) Study population homogeneous concerning other prognostic factors
MRD only variable with significant impact on outcome
No prognostic impact of remaining variables (age, WBC, T- vs B-lineage) 
   Prognostic influence of MRD at all time points
Excellent prognosis in small subgroup of patients with early MRD clearance (MRD negativity at d+14)
Poor prognosis in patients with MRD persistence (MRD > 1 × 10−4 from d+71 onwards) 
    Holowiecki48  (2008) 116 patients
B- and T-lineage
SR (n = 34) and HR (n = 82) ALL 
Chemo (n = 54)
Auto-SCT (n = 27)
Allo-SCT (n = 35) 
FCM (3 colors) after induction and Cons
Poor prognosis in SR and HR patients with MRD > 10−3 after induction and being treated with Chemo alone
Higher proportion of patients with MRD > 10−3 in HR patients than in SR patients
No prognostic impact of MRD on T-ALL
No significant impact of MRD after Cons 
MRD after induction most relevant independent prognostic factor
Additional independent prognostic factors: age and WBC 
    Bassan1  (2009) 112 patients
B- and T-lineage
All risk groups 
According to MRD and clinical risk factors
Chemo (n = 51)
Allo-SCT (n = 25)
Auto-SCT (n = 21)
No additional treatment (relapse, toxicity: (n = 15) 
RQ-PCR (Ig/TCR and/or fusion genes) at 3 time points (wk+10, wk+16, wk+22) Combined MRD information at wk+16 and wk+22 most relevant independent prognostic factor 
   Good prognosis in patients with MRD < 10−4 (wk+16) and any MRD negativity (wk+22) irrespective of classic risk group
Poor outcome in patients with MRD persistence without differences related to original clinical risk class
Early MRD (wk+10) predicted late MRD (wk+16/wk+22) 
Additional independent prognostic factor: WBC
Allo-SCT not needed in HR patients with MRD LR profile 
    Patel47  (2010)§ 159 patients
B-lineage ALL 
Irrespective of risk group
Chemotherapy (n = 94)
Auto-SCT (n = 25)
Allo-SCT (n = 40) 
DNA fingerprinting/RQ-PCR (Ig/TCR) at 3(-5) time points (wk+5, wk+10, wk+17, for patients without SCT: also wk+28, wk+39):
Prognostic influence of MRD in patients receiving Chemo or auto-SCT as postremission treatment
Most discriminative time points: wk+10, wk+17
No significant impact of MRD in patients with allo-SCT 
No multivariate analysis performed 
    Gökbuget46  (2012) 580 patients
B- and T-lineage ALL
SR (n = 434) and HR (n = 146) patients) 
According to MRD (only SR ALL) and clinical risk factors: Chemo (n = 425)
Allo-SCT (n = 155) 
RQ-PCR (Ig/TCR) at 2 time points (d+71, wk+16)
Prognostic influence of MRD at d+71 and wk+16
Complete MRD response rate higher in SR vs HR patients (d+71: 77% vs 51%), clinical benefit of complete response comparable for SR and HR patients 
Multivariate analysis: MRD only parameter with significant prognostic impact 
   MRD-based allo-SCT in MRD-HR patients  
       SCT performed in half of the patients  
       Improved DFS and OS for patients with SCT  
Ph-positive ALL     
    Pane10  (2005) 45 Ph+ patients Chemo followed by allo-SCT (in case of available donor, n = 20) RT-PCR (BCR-ABL) at 2 time points (end of induction, end of first Cons) Age and WBC did not differ between both MRD groups 
  No TKI Heterogeneous sensitivity to treatment  
   Better DFS and OS in good molecular responders ( > 2 log reduction after induction + > 3 log reduction after Cons 1) compared with poor molecular responders  
    Ottmann97  (2007) 49 Ph+ patients Randomized induction: Chemo vs TKI RT- and qualitative nested PCR (BCR-ABL) after end of induction, each Cons/reinduction cycle Lower WBC at diagnosis in MRD negative patients 
 Elderly patients (54-79 y) Cons: Chemo + TKI Heterogeneous sensitivity to treatment  
   MRD response in patients with TKI induction compared to Chemo:  
           Better initial MRD response in TKI group  
           No difference in median MRD level after Cons  
   MRD negativity at any time associated with more favorable outcome  
    Yanada53  (2008) 100 Ph+ patients‖ Induction Chemo + TKI RT-PCR (BCR-ABL) at d+28, d+63 (end of induction),  
  Donor: allo-SCT in CR1 (n = 60) end of Cons 1 (+later time points)  
  No donor: Cons Chemo + TKI (n = 37) No impact of rapid MRD clearance (MRD negativity at d+28, d+62 or after Cons 1) on relapse-free survival  
    Lee51  (2009) 52 Ph+ patients Treatment sequence: Induction Chemo
4 wk TKI
Cons Chemo
4 wk TKI
Allo-SCT in CR1 (n = 48) or non-CR (n = 4) 
RT-PCR (BCR-ABL) after induction Chemo, first TKI, after Cons Chemo, after second course TKI
No prognostic impact of MRD after induction Chemo before TKI
Better DFS and lower relapse rate in patients with ≥ 3 log reduction after first imatinib course 
MRD after TKI most significant independent prognostic factor
Chronic GVHD only additional variable with an independent prognostic impact 
StudyNo. and risk group of patientsTreatmentMRD assessment/prognostic significance of MRD/conclusionsRetaining prognostic significance of classic risk factors/comments
Ph-negative ALL*     
    Vidriales22  (2003) 102 patients According to clinical risk factors: FCM (3 colors) during (d+14) and after (d+35) induction: MRD most relevant independent prognostic factor 
 B- and T-lineage     Chemo (n = 65) Prognostic influence of MRD at both time points Additional independent prognostic factors: age, WBC, Ph positivity 
 CR after induction     Plus auto-SCT (n = 14) Poor prognosis in patients with MRD persistence ≥ 5 × 10−3 d+14 and/or ≥ 5 × 10−4 d+35
Excellent prognosis in small subgroup of patients with early MRD clearance (< 3 × 10−4, d+14) 
      Plus allo-SCT (n = 23)  
    Brüggemann3  (2006) 196 patients
B- and T-lineage
SR ALL 
Chemo RQ-PCR (Ig/TCR) at 9 time points (d+11, d+24, d+44, d+71, wk+16, wk+22, wk+30, wk+41, wk+52) Study population homogeneous concerning other prognostic factors
MRD only variable with significant impact on outcome
No prognostic impact of remaining variables (age, WBC, T- vs B-lineage) 
   Prognostic influence of MRD at all time points
Excellent prognosis in small subgroup of patients with early MRD clearance (MRD negativity at d+14)
Poor prognosis in patients with MRD persistence (MRD > 1 × 10−4 from d+71 onwards) 
    Holowiecki48  (2008) 116 patients
B- and T-lineage
SR (n = 34) and HR (n = 82) ALL 
Chemo (n = 54)
Auto-SCT (n = 27)
Allo-SCT (n = 35) 
FCM (3 colors) after induction and Cons
Poor prognosis in SR and HR patients with MRD > 10−3 after induction and being treated with Chemo alone
Higher proportion of patients with MRD > 10−3 in HR patients than in SR patients
No prognostic impact of MRD on T-ALL
No significant impact of MRD after Cons 
MRD after induction most relevant independent prognostic factor
Additional independent prognostic factors: age and WBC 
    Bassan1  (2009) 112 patients
B- and T-lineage
All risk groups 
According to MRD and clinical risk factors
Chemo (n = 51)
Allo-SCT (n = 25)
Auto-SCT (n = 21)
No additional treatment (relapse, toxicity: (n = 15) 
RQ-PCR (Ig/TCR and/or fusion genes) at 3 time points (wk+10, wk+16, wk+22) Combined MRD information at wk+16 and wk+22 most relevant independent prognostic factor 
   Good prognosis in patients with MRD < 10−4 (wk+16) and any MRD negativity (wk+22) irrespective of classic risk group
Poor outcome in patients with MRD persistence without differences related to original clinical risk class
Early MRD (wk+10) predicted late MRD (wk+16/wk+22) 
Additional independent prognostic factor: WBC
Allo-SCT not needed in HR patients with MRD LR profile 
    Patel47  (2010)§ 159 patients
B-lineage ALL 
Irrespective of risk group
Chemotherapy (n = 94)
Auto-SCT (n = 25)
Allo-SCT (n = 40) 
DNA fingerprinting/RQ-PCR (Ig/TCR) at 3(-5) time points (wk+5, wk+10, wk+17, for patients without SCT: also wk+28, wk+39):
Prognostic influence of MRD in patients receiving Chemo or auto-SCT as postremission treatment
Most discriminative time points: wk+10, wk+17
No significant impact of MRD in patients with allo-SCT 
No multivariate analysis performed 
    Gökbuget46  (2012) 580 patients
B- and T-lineage ALL
SR (n = 434) and HR (n = 146) patients) 
According to MRD (only SR ALL) and clinical risk factors: Chemo (n = 425)
Allo-SCT (n = 155) 
RQ-PCR (Ig/TCR) at 2 time points (d+71, wk+16)
Prognostic influence of MRD at d+71 and wk+16
Complete MRD response rate higher in SR vs HR patients (d+71: 77% vs 51%), clinical benefit of complete response comparable for SR and HR patients 
Multivariate analysis: MRD only parameter with significant prognostic impact 
   MRD-based allo-SCT in MRD-HR patients  
       SCT performed in half of the patients  
       Improved DFS and OS for patients with SCT  
Ph-positive ALL     
    Pane10  (2005) 45 Ph+ patients Chemo followed by allo-SCT (in case of available donor, n = 20) RT-PCR (BCR-ABL) at 2 time points (end of induction, end of first Cons) Age and WBC did not differ between both MRD groups 
  No TKI Heterogeneous sensitivity to treatment  
   Better DFS and OS in good molecular responders ( > 2 log reduction after induction + > 3 log reduction after Cons 1) compared with poor molecular responders  
    Ottmann97  (2007) 49 Ph+ patients Randomized induction: Chemo vs TKI RT- and qualitative nested PCR (BCR-ABL) after end of induction, each Cons/reinduction cycle Lower WBC at diagnosis in MRD negative patients 
 Elderly patients (54-79 y) Cons: Chemo + TKI Heterogeneous sensitivity to treatment  
   MRD response in patients with TKI induction compared to Chemo:  
           Better initial MRD response in TKI group  
           No difference in median MRD level after Cons  
   MRD negativity at any time associated with more favorable outcome  
    Yanada53  (2008) 100 Ph+ patients‖ Induction Chemo + TKI RT-PCR (BCR-ABL) at d+28, d+63 (end of induction),  
  Donor: allo-SCT in CR1 (n = 60) end of Cons 1 (+later time points)  
  No donor: Cons Chemo + TKI (n = 37) No impact of rapid MRD clearance (MRD negativity at d+28, d+62 or after Cons 1) on relapse-free survival  
    Lee51  (2009) 52 Ph+ patients Treatment sequence: Induction Chemo
4 wk TKI
Cons Chemo
4 wk TKI
Allo-SCT in CR1 (n = 48) or non-CR (n = 4) 
RT-PCR (BCR-ABL) after induction Chemo, first TKI, after Cons Chemo, after second course TKI
No prognostic impact of MRD after induction Chemo before TKI
Better DFS and lower relapse rate in patients with ≥ 3 log reduction after first imatinib course 
MRD after TKI most significant independent prognostic factor
Chronic GVHD only additional variable with an independent prognostic impact 

Only MRD studies being published as peer reviewed papers are listed.

Chemo indicates chemotherapy; Cons, consolidation; CR, complete remission; D, day; DFS, disease free survival; FCM, flow cytometry; HR, high risk; Ind, induction; LR, low risk; OS, overall survival; Ph, Philadelphia chromosome; RQ-PCR, real-time quantitative PCR; RT-PCR, reverse transcriptase-PCR; SCT, stem cell transplantation; SR, standard risk; TKI, tyrosine kinase inhibitors; TP(s), time-point(s); Wk, week; and WBC, white blood cell count.

*

Only study of Bassan et al includes Ph-positive patients.1 

Eleven patients with SCT censored at time of SCT.

Clinical risk factors for assignment to allo-SCT independent of MRD: presence of t(9;22) or t(4;11).

§

Update from an interim report of Mortuza et al.9 

Ph-negative ALL.

Several studies reported on the independent prognostic value of MRD in adult Ph-negative ALL performing either Ig/TCR-quantitative RQ-PCR1,3,9,46,47  or FCM12,22,48  (Table 3). Concordantly, different European study groups demonstrated that MRD persistence measured at different time points 1-6 months after initial diagnosis is associated with a poor prognosis.1,3,9,22,46-48  Brüggemann et al3  and Vidriales et al22  additionally identified a small subset of patients with a very rapid tumor clearance (low-level/undetectable MRD after 2 weeks of therapy) and an excellent prognosis. The presently largest MRD study on adult ALL was recently published by Gökbuget et al and analyzed MRD in Ph-negative patients with standard risk (SR, n = 434) and high risk (HR, n = 146) features.46  A complete MRD response after induction 2 and/or consolidation 1 was associated with a comparable clinical benefit irrespective of pretherapeutic risk factors. MRD was the only parameter with significant prognostic impact in multivariate analysis. The biologic differences between SR and HR patients were reflected by the significant different proportion of patients reaching a complete MRD response between SR and HR patients with ∼ 20% point lower rates of MRD negativity in HR patients.

Whether or not rare factors, such as t(4;11), may retain their prognostic significance is hard to state as these patients are diluted within the whole population. Cimino et al analyzed a limited number of adult MLL-AF4–positive patients demonstrating the prognostic impact of MRD also for this rare biologic entity.49 

Ph-positive ALL.

In Ph-positive ALL, most published studies focus on detection and quantification of BCR-ABL transcripts. In the pre-tyrosine kinase inhibitors (TKI) era, the level of MRD after induction and/or consolidation treatment turned out to be a powerful indicator of prognosis, although data differed concerning the discriminative value of different time points for MRD assessment10,50  (Table 3). Introduction of TKI substantially changed treatment outcome and MRD kinetics. In a study by Lee et al, MRD assessment after induction chemotherapy did not retain its prognostic significance when followed by imatinib treatment.51  Conversely, a reduction of BCR-ABL transcript levels of at least 3 log after the first 4-week imatinib therapy was identified as the most powerful predictor of a better disease free survival (DFS) and overall survival (OS) rate after SCT (4-year DFS 82.1% vs 41.7%, P = .009; 4-year OS 82.3 vs 48.6, P = .007). These findings indicate that poor response to chemotherapy may be compensated by subsequent administration of imatinib. Leguay et al presented data of the GRAAL AFR03 study that imatinib combined with high-dose chemotherapy improved molecular remission rate before transplantation and led to an improved outcome.52  In contrast, Yanada et al failed to establish an association between an early MRD response to imatinib combined chemotherapy and outcome and concluded that relapse risk may depend on factors unrelated to initial treatment response53  (eg, outgrowth of preexisting subclones with resistance mutations54 ).

Postremission MRD monitoring

A second important application of MRD is postremission monitoring of patients reaching complete MRD response for early detection of an impending relapse.

Ph-negative ALL.

Raff at al55  were the first to demonstrate within the German Multicenter ALL Study Group (GMALL) trials that molecular relapse defined as reconversion to quantifiable molecular MRD positivity was followed by a clinical relapse after a median time of 4.1 month in Ph-negative ALL. Remarkably, low-level, nonquantifiable MRD was not necessarily related to a subsequent relapse confirming this MRD value as a sort of gray area.16  A recent update of the Raff et al study55  by Gökbuget et al46  confirmed the close correlation between conversion to quantifiable MRD positivity and subsequent relapse: in 34 patients with conversion to quantifiable MRD positivity (“MRD relapse,” n = 13 during first year of treatment, n = 21 subsequently), the probability of continuous complete remission (CCR) was 21% ± 9% at 5 years. If patients with MRD-based SCT in first CR were excluded the probability of CCR was only 0% (5% ± 5% at 3 years).

Ph-positive ALL.

In Ph-positive ALL, several studies published already in the 1990s showed a significant relationship between conversion to MRD positivity and subsequent relapse in the pre-TKI era.10,56-59  Yanada et al confirmed these findings also for Ph-positive patients being treated with imatinib-combined chemotherapy.53  In a prospective study on 100 adult patients with Ph-positive ALL, 29 showed an MRD elevation in CR. Of these, 12 of 13 who had not undergone allogeneic (allo) SCT experienced a relapse, whereas only 3 of 16 patients who underwent allo-SCT relapsed. The authors concluded that an increase in MRD is predictive of a subsequent relapse, but such patients can be successfully treated with allo-SCT.

Treatment stratification according to initial MRD response

The objective of measuring MRD response to initial therapy is to adjust treatment with the ultimate goal to improve outcome of MRD-HR patients and to reduce toxicity in MRD-LR patients without worsening their prognosis.

Several clinical trials on adult ALL implemented MRD into their treatment stratification16 : The PETHEMA ALL-AR-03 trial12,60  focuses on HR Ph-negative ALL and passes on SCT in first complete remission in case of standard cytologic response (< 10% blasts in BM on day 14) and MRD < 5 × 10−4 after early consolidation. In contrast, patients with a slow cytologic response and/or MRD > 5 × 10−4 after early consolidation receive allo-SCT further on. Preliminary results indicate that the prognosis of HR patients with adequate response to induction and adequate clearance of MRD is not worsened by avoiding allo-SCT. The combined MRD level after induction and consolidation therapy was the main prognostic factor for CR, DFS, and OS, although a part of MRD-LR patients received a MRD-based de-escalated therapy.

Within the GMALL 07/03 trial, patients with persistent MRD > 10−4 after induction (day 71) and/or first consolidation (week 16) were allocated to the MRD-HR group61  and qualified for allo-SCT. Gökbuget et al recently showed the first results of MRD-based treatment intensification in these patients: 120 of 504 evaluated patients (24%) were allocated to the MRD-HR group (89 SR and 31 HR patients defined by conventional criteria).46  In 47% of these patients, SCT was realized in first CR, with the SCT rate being significantly higher in HR compared with SR patients (71% vs 39%, P < .002). The probability of CCR after 5 years was significantly higher for patients receiving an MRD-directed SCT in first CR compared with those without SCT in first CR (66% ± 7% vs 12% ± 5%, P < .0001). This also translated into a better OS at 5 years (54% ± 8% vs 33% ± 7%, P = .06).

Bassan et al described an MRD-oriented therapy for all t(4;11)/t(9,22) ALL patients within the Northern Italy Leukemia Group.1  MRD-positive patients (defined as MRD > 10−4 before induction-consolidation cycle 6 and MRD positivity before cycle 8) were allocated to allogeneic or autologous SCT, whereas MRD-negative patients received standard maintenance regardless of classic risk factors. Four-year DFS was 76% in MRD-LR patients versus only 24% in the MRD-HR group despite of treatment intensification.

The first phase 2 clinical study prospectively analyzing an MRD-based targeted therapy administered blinatumomab monotherapy in patients with MRD failure or MRD reappearance (defined as quantifiable MRD > 1 × 10−4 after end of first consolidation).62  Sixteen of 20 evaluable patients (15 patients with MRD failure, 5 patients with MRD relapse) became MRD negative after 1 cycle of blinatumomab treatment. Twelve of the 16 MRD responders had never achieved MRD negativity before. MRD negativity translated into an ongoing hematologic remission in all 16 MRD-negative patients within a median observation time of 405 days (1-year relapse free survival probability 78%). One patient was censored because of withdrawal of informed consent during second treatment cycle, 8 of these patients consecutively underwent allo-SCT without any treatment related mortality, and 7 patients did not receive any further consolidation treatment, indicating the possibility of an improved outcome with this MRD-guided treatment, even if data can only be compared with historical controls.

Preemptive treatment in case of MRD recurrence

MRD assessment is also used for preemptive treatment intervention in case of MRD recurrence. In the GMALL 07/03 trial, salvage treatment was intended to be started at time of reoccurrence of quantifiable MRD. In a recent publication,46  10 of 34 patients with MRD relapse underwent MRD triggered SCT in ongoing first CR. Three-year probability of CCR was 80% ± 18% compared with only 5% ± 5% in patients without transplantation. Other study groups also perform postremission MRD monitoring for selected patient subgroups, with some of them drawing clinical consequences in case of high-level MRD.16  Besides SCT, also targeted therapies are investigated to improve outcome in patients with an MRD relapse.

In Ph-positive ALL, postremission MRD monitoring is mainly used to tailor treatment after SCT. Wassmann et al investigated the effect of imatinib to decrease the relapse probability in case of reconversion to MRD positivity after SCT.63  BCR-ABL transcripts became undetectable by both quantitative and nested RT-PCR in 15 of 29 (52%) patients. This was associated with a sustained remission, whereas MRD persistence 6-10 weeks after start of imatinib treatment correlated with an almost certain relapse.

Optimal sampling time points and sampling frequency have to be defined according to the individual protocol depending on the treatment protocol and the stratification aim. In de novo Ph-negative ALL, postinduction MRD assessment (after 2-4 months of treatment) is considered to have the most important role for evaluation of initial treatment response and MRD-based risk-stratification. An MRD assessment during induction (after ∼ 2 weeks of treatment) additionally identifies patients with a rapid tumor clearance and a particular good outcome. Concerning postremission monitoring of MRD for early relapse identification, the GMALL proposes 3-monthly intervals for a total of 3 years as the majority of clinical relapses occur within this time64  and reconversion to MRD-positivity precedes a clinical relapse with a median time of 4.1 months between first quantifiable MRD positivity and relapse.

In Ph-positive ALL, the value of MRD for initial remission assessment is more limited in the era of TKI, whereas MRD assessment is frequently used for postremission monitoring. However, compared with Ph-negative ALL, relapse kinetics seem to be more rapid with median time between MRD elevation and relapse of only 2-3 months with53  or without59  application of TKI. Therefore, sampling frequency is recommended to be higher than in Ph-negative ALL.

Comparisons of MRD results obtained by different methodologies show that different techniques cannot be considered fully interchangeable. The main difference between FCM and Ig/TCR-PCR seems to be sensitivity,19,21,40  with both methods quantifying single signals/leukemic cell. In contrast, BCR-ABL-PCR measures multiple transcripts/cell with copy numbers potentially varying during the course of treatment. As there is evidence of multilineage involvement of Ph-positive cells, target cells also may not be fully concordant to other MRD techniques. Therefore, choice of the MRD method in a particular protocol should be guided by the question to be answered, the experience gained in former MRD trials, the available technical expertise, the logistics, and whether or not treatment intervention is planned according to MRD results.

The topic of this Perspective is to debate whether MRD superseded other risk factors. To answer this question, it is essential to illuminate the value of MRD within different clinical settings but also to discuss pretherapeutic factors that partly changed their meaning from prognostic to predictive classifiers.

Classic risk factors versus MRD: the conventional concept of risk-oriented therapy

During the past 3 decades, treatment of adult ALL patients composed an induction/consolidation chemotherapy followed either by additional consolidation cycles and maintenance treatment (partly supported by auto SCT) or allo-SCT. Although there was also a discussion on optimization of chemotherapy elements, the main therapeutic decision was whether or not to apply allo-SCT, which on the one hand leads to a reduced relapse rate but on the other hand is accompanied by severe side effects and a considerable treatment-related mortality. Data exist for both treatment paradigms: Collaboration from the United Kingdom Medical Research Council and the Eastern Cooperative Oncology Group demonstrated the superiority of allo-SCT at least in younger patients.65  On the other hand, pediatric-inspired chemotherapy regimens show favorable outcomes in adolescents restraining indication of allo-SCT.66-68  Most protocols struck a balance between both approaches generally recommending SCT for patients with clinical risk factors and a high risk of postchemotherapy relapse. Risk assignment is conventionally based on indirect measures of leukemic burden and chemosensitivity, such as chromosome, molecular, and immunophenotypic analyses at diagnosis (Table 1; Figure 2A). Compared with all these pretherapeutic factors, MRD directly measures chemosensitivity for individual patients and integrates different host-, leukemia-, and treatment-related components of treatment outcome.

Figure 2

Prospective therapeutic shifts according to conventional pretreatment stratification criteria and MRD. (A) Potential treatment decisions based on pretreatment factors. (B) Potential treatment decisions based on MRD. Cons indicates consolidation; DLI, donor lymphocyte infusion; and TD, treatment decision.

Figure 2

Prospective therapeutic shifts according to conventional pretreatment stratification criteria and MRD. (A) Potential treatment decisions based on pretreatment factors. (B) Potential treatment decisions based on MRD. Cons indicates consolidation; DLI, donor lymphocyte infusion; and TD, treatment decision.

Close modal

Indeed, most published data recognize MRD as most important independent prognostic factor in adult ALL that supersedes all other risk factors, at least for Ph-negative ALL. Prospective studies on MRD-oriented therapy also indicate that MRD-based treatment is safely possible, allo-SCT may be avoided in MRD-LR Ph-negative patients and seems to be particularly active in MRD-HR patients. In addition, MRD measurement helps to prevent the problem of stratifying patients with newly identified genetic risk factors that are too rare to allow a reasonable assignment to a special risk group.

However, available MRD data presented here are not fully consistent. Optimal sampling time points and MRD thresholds seem to differ between ALL subgroups, in particular in Ph-positive compared with Ph-negative ALL. In addition, multivariate analyses in published studies partially retain prognostic factors other than MRD as independent variables (interestingly, the “old-fashioned” factor elevated white blood cell count at diagnosis emerges repeatedly). As an additional restriction, as true also for all other prognostic factors, MRD assessment is not realizable in all patients because of technical limitations (eg, lack of identification of leukemia specific markers, missing follow-up samples). Depending on the minimum technical requirements, the MRD methodology, and the adherence to the protocol, MRD-based stratification using stringent criteria seems to be feasible in ∼ 80%-90% of patients.2,69 

It also must be admitted that no published study on adult ALL performed a randomized comparison between action and no action on MRD but only compared the results of MRD-based treatment to historical outcome data or to groups of patients that did not receive an MRD guided therapy for whatever reasons. Therefore, as already started in childhood ALL, also in adult ALL randomized trials have to be done to confirm these encouraging data. MRD excellently qualifies for such an approach because it not only serves as marker for initial treatment stratification but also allows for an ongoing monitoring (Figure 2B). Thereby MRD can serve as “safety net” enabling early reintensification in case of MRD-based treatment de-escalation.

Targeted therapies: prognostic factors become predictive

Increased understanding of molecular mechanisms of cancer and availability of drugs targeting them is changing the meaning of particular leukemic markers from solely prognostic toward predictive factors (Table 1). In adult ALL, BCR-ABL positivity can serve as a model for this transformation: Formerly, the detection of the Ph chromosome during diagnostic workup of adult ALL prognosticated an extremely poor outcome with remission rates being at least 10% lower than in Ph-negative ALL and with a median survival of only 8 months.70  The sole curative option was an allo-SCT. but even in this setting Ph positivity formed the risk group with the poorest outcome, making Ph positivity a “poorly tractable therapeutic problem.”71  However, things changed with the implementation of TKIs as a targeted therapy: CR rates improved and also evidence of a survival benefit emerges. Now BCR-ABL is not only a prognostic marker but also predicts response to TKI. In addition, this new therapeutic approach changes MRD kinetics in Ph-positive ALL. Whereas in the pre-TKI era studies suggested a good correlation between MRD and outcome, MRD data in the TKI setting are more conflicting, and optimal time-points and thresholds are still a matter of debate. A potential reason for the discrepancy between initial MRD response and outcome is the existence of low-level BCR-ABL kinase domain mutations before treatment.54,72,73  Whereas the leukemic bulk may well respond to treatment the small-sized resistant subclone may be the origin of relapse potentially necessitating MRD analysis of subclones in the future.

Even though many of new potential predictive markers are not ready to be used as treatment targets in clinical routine because of issues related to reproducibility, statistical significance, and practical applications, the possibilities of targeted therapy are intriguing. However, when implementing new elements into treatment protocols, the sensitivity of leukemic cells to a particular drug is not known in advance. Traditional risk groups may well respond differently to targeted therapies than they do to classic chemotherapy, potentially leading to unpredictable treatment responses in different subgroups of ALL. For instance, CD20 expression in B-lineage ALL, which is under debate to be an adverse prognostic factor,74,75  is increasingly targeted by protocols including rituximab. Data from the MD Anderson Cancer Center indicate that addition of rituximab to polychemotherapy alters MRD kinetics and significantly improves MRD response.76  However, the effect of the same rituximab dose seems to be different in SR compared with HR patients.77  Therefore, it is particularly important to evaluate treatment success separately within the different subgroups to identify sensitive subpopulations. Ignoring baseline risk factors means obscuring a possibly relevant treatment effect in distinct biologic subsets by diluting in the whole population. Therefore, assessment of pretherapeutic markers cannot be skipped in diagnostic workup of ALL, even if MRD is closely monitored as pretherapeutic and MRD markers increasingly become complementary tools to tailor therapy.

In times of targeted and sequential therapies, definition of an adequate and discriminatory primary variable for judging treatment success and planning subsequent treatment steps becomes more and more important. OS, the classic gold standard for a primary end point, as reliable, objective, and easily determined parameter gets problematic in times of multimodal and sequential therapies, including SCT. The other endpoint, the CR rate, is less objective in particular in B-cell precursor ALL where the distinction between leukemic cells and hematogones is exceedingly difficult in bone marrow during recovery after chemotherapy or SCT. The value of CR assessment is also limited by the fact that current treatment protocols lead to CR rate of ∼ 90% in adult ALL. In contrast, MRD offers itself as an endpoint in this setting as it is a clinical parameter that integrates different leukemia, host, and treatment aspects into one highly sensitive parameter. As a prerequisite for a usage as primary endpoint, there has to be (1) a plausible biologic relationship between reduction of MRD and response of the disease to therapy, (2) the prognostic value of the surrogate for the clinical outcome has to be validated, and (3) the evidence from clinical trials has to exist that treatment effects on the surrogate correspond to effects on the clinical outcome.78  A necessary technical precondition is the availability of a standardized technology for MRD measurement regarding both standardized analysis and interpretation. This is currently primarily fulfilled for MRD assessment using DNA-based quantitative RQ-PCR analysis according to EuroMRD standards36  and related consensus definitions obtained at the second international symposium on MRD assessment.16  Ten years of international quality controls with > 40 participating laboratories demonstrated the robustness of the system with intra- and inter-assay variability of less than half a log. In addition, FCM-based MRD quantification and quantitative RT-PCR of BCR-ABL transcripts are increasingly standardized within different international collaborations. These analyses have to be performed in accredited specialized laboratories as tests used in decision-making in clinical trials generally have to conform to the Organisation for Economic Co-Operation and Development (OECD) Guidelines on Good Laboratory Practice79  or the Clinical Laboratory Improvement Amendments.80  As shown in large-scale pediatric trials, this does not hamper a broad application to all eligible ALL patients. Although not yet generally accepted as a primary endpoint of clinical trials by the European Medicines Agency and the Food and Drug Administration, MRD measurement in combination with adaptive trial designs may overcome part of the current difficulties in evaluating the efficiency of new agents in ALL.

In conclusion, published data on MRD assessment in adult ALL have shown a strong correlation between MRD response and outcome as well as the prognostic value of MRD reappearance for hematologic relapse. In times of individualized targeted therapies, MRD will not substitute baseline risk factors but evaluate their impact within different treatment regimens and will help to optimize treatment sequence. In this context, MRD has also to be considered as quantitative and objective extension of established endpoints of hematologic remission and relapse more than a substitute of pretherapeutic risk factors.

The authors thank Nicola Gökbuget, Dieter Hoelzer, and the participants of the German Multicenter Study Group for adult ALL for their close collaboration in the MRD studies.

This work was supported in part by the Wilhelm Sander Stiftung (2001-074.1 and 2001-074.2) and the Deutsche Krebshilfe (702657Ho2).

Contribution: M.B. wrote the first draft of the manuscript; T.R. contributed to the writing and prepared figures; and M.B., T.R., and M.K. finalized the manuscript.

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

Correspondence: Monika Brüggemann, Department of Hematology, University Hospital Schleswig-Holstein, Campus Kiel, Chemnitzstrasse 33, D-24116 Kiel, Germany; e-mail: m.brueggemann@med2.uni-kiel.de.

1
Bassan
 
R
Spinelli
 
O
Oldani
 
E
, et al. 
Improved risk classification for risk-specific therapy based on the molecular study of minimal residual disease (MRD) in adult acute lymphoblastic leukemia (ALL).
Blood
2009
, vol. 
113
 
18
(pg. 
4153
-
4162
)
2
Borowitz
 
MJ
Devidas
 
M
Hunger
 
SP
, et al. 
Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: a Children's Oncology Group study.
Blood
2008
, vol. 
111
 
12
(pg. 
5477
-
5485
)
3
Brüggemann
 
M
Raff
 
T
Flohr
 
T
, et al. 
Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia.
Blood
2006
, vol. 
107
 
3
(pg. 
1116
-
1123
)
4
Cavé
 
H
van der Werff ten Bosch
 
J
Suciu
 
S
, et al. 
Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia: European Organization for Research and Treatment of Cancer–Childhood Leukemia Cooperative Group.
N Engl J Med
1998
, vol. 
339
 
9
(pg. 
591
-
598
)
5
Coustan-Smith
 
E
Sancho
 
J
Hancock
 
ML
, et al. 
Clinical importance of minimal residual disease in childhood acute lymphoblastic leukemia.
Blood
2000
, vol. 
96
 
8
(pg. 
2691
-
2696
)
6
Eckert
 
C
Biondi
 
A
Seeger
 
K
, et al. 
Prognostic value of minimal residual disease in relapsed childhood acute lymphoblastic leukaemia.
Lancet
2001
, vol. 
358
 
9289
(pg. 
1239
-
1241
)
7
Lee
 
S
Kim
 
DW
Cho
 
B
, et al. 
Risk factors for adults with Philadelphia-chromosome-positive acute lymphoblastic leukaemia in remission treated with allogeneic bone marrow transplantation: the potential of real-time quantitative reverse-transcription polymerase chain reaction.
Br J Haematol
2003
, vol. 
120
 
1
(pg. 
145
-
153
)
8
Lee
 
S
Kim
 
DW
Kim
 
YJ
, et al. 
Minimal residual disease-based role of imatinib as a first-line interim therapy prior to allogeneic stem cell transplantation in Philadelphia chromosome-positive acute lymphoblastic leukemia.
Blood
2003
, vol. 
102
 
8
(pg. 
3068
-
3070
)
9
Mortuza
 
FY
Papaioannou
 
M
Moreira
 
IM
, et al. 
Minimal residual disease tests provide an independent predictor of clinical outcome in adult acute lymphoblastic leukemia.
J Clin Oncol
2002
, vol. 
20
 
4
(pg. 
1094
-
1104
)
10
Pane
 
F
Cimino
 
G
Izzo
 
B
, et al. 
Significant reduction of the hybrid BCR/ABL transcripts after induction and consolidation therapy is a powerful predictor of treatment response in adult Philadelphia-positive acute lymphoblastic leukemia.
Leukemia
2005
, vol. 
19
 
4
(pg. 
628
-
635
)
11
Raghavachar
 
A
Thiel
 
E
Bartram
 
CR
Analyses of phenotype and genotype in acute lymphoblastic leukemias at first presentation and in relapse.
Blood
1987
, vol. 
70
 
4
(pg. 
1079
-
1083
)
12
Ribera
 
JM
Oriol
 
A
Morgades
 
M
, et al. 
Treatment of high-risk (HR) Philadelphia chromosome-negative (Ph-) adult acute lymphoblastic leukemia (ALL) according to baseline risk factors and minimal residual disease (MRD): results of the PETHEMA ALL-AR-03 Trial including the use of Propensity Score (PS) method to reduce assignment bias [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
 
Abstract 322
13
Spinelli
 
O
Peruta
 
B
Tosi
 
M
, et al. 
Clearance of minimal residual disease after allogeneic stem cell transplantation and the prediction of the clinical outcome of adult patients with high-risk acute lymphoblastic leukemia.
Haematologica
2007
, vol. 
92
 
5
(pg. 
612
-
618
)
14
van Dongen
 
JJ
Seriu
 
T
Panzer-Grumayer
 
ER
, et al. 
Prognostic value of minimal residual disease in acute lymphoblastic leukaemia in childhood.
Lancet
1998
, vol. 
352
 
9142
(pg. 
1731
-
1738
)
15
Mejstríková
 
E
Fronkova
 
E
Kalina
 
T
, et al. 
Detection of residual B precursor lymphoblastic leukemia by uniform gating flow cytometry.
Pediatr Blood Cancer
2010
, vol. 
54
 
1
(pg. 
62
-
70
)
16
Brüggemann
 
M
Schrauder
 
A
Raff
 
T
, et al. 
Standardized MRD quantification in European ALL trials: proceedings of the Second International Symposium on MRD assessment in Kiel, Germany, 18-20 September 2008.
Leukemia
2010
, vol. 
24
 
3
(pg. 
521
-
535
)
17
Coustan-Smith
 
E
Campana
 
D
Immunologic minimal residual disease detection in acute lymphoblastic leukemia: a comparative approach to molecular testing.
Best Pract Res Clin Haematol
2010
, vol. 
23
 
3
(pg. 
347
-
358
)
18
Dworzak
 
MN
Froschl
 
G
Printz
 
D
, et al. 
Prognostic significance and modalities of flow cytometric minimal residual disease detection in childhood acute lymphoblastic leukemia.
Blood
2002
, vol. 
99
 
6
(pg. 
1952
-
1958
)
19
Gaipa
 
G
Cazzaniga
 
G
Valsecchi
 
MG
, et al. 
Time point-dependent concordance of flow cytometry and RQ-PCR in minimal residual disease detection in childhood acute lymphoblastic leukemia.
Haematologica
2012
, vol. 
97
 
10
(pg. 
1582
-
1593
)
20
Kerst
 
G
Kreyenberg
 
H
Roth
 
C
, et al. 
Concurrent detection of minimal residual disease (MRD) in childhood acute lymphoblastic leukaemia by flow cytometry and real-time PCR.
Br J Haematol
2005
, vol. 
128
 
6
(pg. 
774
-
782
)
21
Malec
 
M
van der Velden
 
VHJ
Bjorklund
 
E
, et al. 
Analysis of minimal residual disease in childhood acute lymphoblastic leukemia: comparison between RQ-PCR analysis of Ig/TcR gene rearrangements and multicolor flow cytometric immunophenotyping.
Leukemia
2004
, vol. 
18
 
10
(pg. 
1630
-
1636
)
22
Vidriales
 
MB
Perez
 
JJ
Lopez-Berges
 
MC
, et al. 
Minimal residual disease in adolescent (older than 14 years) and adult acute lymphoblastic leukemias: early immunophenotypic evaluation has high clinical value.
Blood
2003
, vol. 
101
 
12
(pg. 
4695
-
4700
)
23
Dworzak
 
MN
Schumich
 
A
Printz
 
D
, et al. 
CD20 up-regulation in pediatric B-cell precursor acute lymphoblastic leukemia during induction treatment: setting the stage for anti-CD20 directed immunotherapy.
Blood
2008
, vol. 
112
 
10
(pg. 
3982
-
3988
)
24
Gaipa
 
G
Basso
 
G
Maglia
 
O
, et al. 
Drug-induced immunophenotypic modulation in childhood ALL: implications for minimal residual disease detection.
Leukemia
2005
, vol. 
19
 
1
(pg. 
49
-
56
)
25
van der Sluijs-Gelling
 
AJ
Van der Velden
 
VHJ
Roeffen
 
ET
Veerman
 
AJ
van Wering
 
ER
Immunophenotypic modulation in childhood precursor-B-ALL can be mimicked in vitro and is related to the induction of cell death.
Leukemia
2005
, vol. 
19
 
10
(pg. 
1845
-
1847
)
26
Stams
 
WA
Den Boer
 
ML
Beverloo
 
HB
, et al. 
Effect of the histone deacetylase inhibitor depsipeptide on B-cell differentiation in both TEL-AML1-positive and negative childhood acute lymphoblastic leukemia.
Haematologica
2005
, vol. 
90
 
12
(pg. 
1697
-
1699
)
27
van Wering
 
ER
Beishuizen
 
A
Roeffen
 
ET
, et al. 
Immunophenotypic changes between diagnosis and relapse in childhood acute lymphoblastic leukemia.
Leukemia
1995
, vol. 
9
 
9
(pg. 
1523
-
1533
)
28
Dworzak
 
MN
Gaipa
 
G
Schumich
 
A
, et al. 
Modulation of antigen expression in B-cell precursor acute lymphoblastic leukemia during induction therapy is partly transient: evidence for a drug-induced regulatory phenomenon. Results of the AIEOP-BFM-ALL-FLOW-MRD-Study Group.
Cytometry B Clin Cytom
2010
, vol. 
78
 
3
(pg. 
147
-
153
)
29
da Costa
 
ES
Peres
 
RT
Almeida
 
J
, et al. 
Harmonization of light scatter and fluorescence flow cytometry profiles obtained after staining peripheral blood leucocytes for cell surface-only versus intracellular antigens with the Fix & Perm reagent.
Cytometry B Clin Cytom
2010
, vol. 
20;78
 
1
(pg. 
11
-
20
)
30
Dworzak
 
MN
Gaipa
 
G
Ratei
 
R
, et al. 
Standardization of flow cytometric minimal residual disease evaluation in acute lymphoblastic leukemia: multicentric assessment is feasible.
Cytometry B Clin Cytom
2008
, vol. 
74
 
6
(pg. 
331
-
340
)
31
Pedreira
 
CE
Costa
 
ES
Almeida
 
J
, et al. 
A probabilistic approach for the evaluation of minimal residual disease by multiparameter flow cytometry in leukemic B-cell chronic lymphoproliferative disorders.
Cytometry A
2008
, vol. 
73A
 
12
(pg. 
1141
-
1150
)
32
Pedreira
 
CE
Costa
 
ES
Barrena
 
S
, et al. 
Generation of flow cytometry data files with a potentially infinite number of dimensions.
Cytometry A
2008
, vol. 
73
 
9
(pg. 
834
-
846
)
33
van Dongen
 
JJM
Lhermitte
 
L
Böttcher
 
S
, et al. 
EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes.
Leukemia
2012
, vol. 
26
 
9
(pg. 
1908
-
1975
)
34
Brüggemann
 
M
Droese
 
J
Bolz
 
I
, et al. 
Improved assessment of minimal residual disease in B cell malignancies using fluorogenic consensus probes for real-time quantitative PCR.
Leukemia
2000
, vol. 
14
 
8
(pg. 
1419
-
1425
)
35
Brüggemann
 
M
van der Velden
 
VHJ
Raff
 
T
, et al. 
Rearranged T-cell receptor beta genes represent powerful targets for quantification of minimal residual disease in childhood and adult T-cell acute lymphoblastic leukemia.
Leukemia
2004
, vol. 
18
 
4
(pg. 
709
-
719
)
36
Van der Velden
 
VHJ
Cazzaniga
 
G
Schrauder
 
A
, et al. 
Analysis of minimal residual disease by Ig/TCR gene rearrangements: guidelines for interpretation of real-time quantitative PCR data.
Leukemia
2007
, vol. 
21
 
4
(pg. 
604
-
611
)
37
Van der Velden
 
VHJ
van Dongen
 
JJM
MRD detection in acute lymphoblastic leukemia patients using Ig/TCR gene rearrangements as targets for real-time quantitative PCR.
Methods Mol Biol
2009
, vol. 
538
 (pg. 
115
-
150
)
38
Burmeister
 
T
Marschalek
 
R
Schneider
 
B
, et al. 
Monitoring minimal residual disease by quantification of genomic chromosomal breakpoint sequences in acute leukemias with MLL aberrations.
Leukemia
2006
, vol. 
20
 
3
(pg. 
451
-
457
)
39
Van der Velden
 
VHJ
Corral
 
L
Valsecchi
 
MG
, et al. 
Prognostic significance of minimal residual disease in infants with acute lymphoblastic leukemia treated within the Interfant-99 protocol.
Leukemia
2009
, vol. 
23
 
6
(pg. 
1073
-
1079
)
40
Neale
 
GA
Coustan-Smith
 
E
Stow
 
P
, et al. 
Comparative analysis of flow cytometry and polymerase chain reaction for the detection of minimal residual disease in childhood acute lymphoblastic leukemia.
Leukemia
2004
, vol. 
18
 
5
(pg. 
934
-
938
)
41
Fronkova
 
E
Muzikova
 
K
Mejstrikova
 
E
, et al. 
B-cell reconstitution after allogeneic SCT impairs minimal residual disease monitoring in children with ALL.
Bone Marrow Transplant
2008
, vol. 
42
 
3
(pg. 
187
-
196
)
42
van der Velden
 
VHJ
Wijkhuijs
 
JM
van Dongen
 
JJM
Non-specific amplification of patient-specific Ig//TCR gene rearrangements depends on the time point during therapy: implications for minimal residual disease monitoring.
Leukemia
2008
, vol. 
22
 
3
(pg. 
641
-
644
)
43
Faham
 
M
Zheng
 
J
Moorhead
 
M
, et al. 
Deep sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia [published online ahead of print October 16, 2012].
Blood
 
44
Pfeifer
 
H
Cazzaniga
 
G
Spinelli
 
O
, et al. 
International standardization of minimal residual disease assessment in Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ ALL) expressing m-BCR-ABL transcripts: updated results of quality control procedures by the EWALL and ESG-MRD-ALL Consortia [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
 
Abstract 2535
45
Conter
 
V
Bartram
 
CR
Valsecchi
 
MG
, et al. 
Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: results in 3184 patients of the AIEOP-BFM ALL 2000 study.
Blood
2010
, vol. 
115
 
16
(pg. 
3206
-
3214
)
46
Gökbuget
 
N
Kneba
 
M
Raff
 
T
, et al. 
Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies.
Blood
2012
, vol. 
120
 
9
(pg. 
1868
-
1876
)
47
Patel
 
B
Rai
 
L
Buck
 
G
, et al. 
Minimal residual disease is a significant predictor of treatment failure in non T-lineage adult acute lymphoblastic leukaemia: final results of the international trial UKALL XII/ECOG2993.
Br J Haematol
2010
, vol. 
148
 
1
(pg. 
80
-
89
)
48
Holowiecki
 
J
Krawczyk-Kulis
 
M
Giebel
 
S
, et al. 
Status of minimal residual disease after induction predicts outcome in both standard and high-risk Ph-negative adult acute lymphoblastic leukaemia: the Polish Adult Leukemia Group ALL 4-2002 MRD Study.
Br J Haematol
2008
, vol. 
142
 
2
(pg. 
227
-
237
)
49
Cimino
 
G
Elia
 
L
Rapanotti
 
MC
, et al. 
A prospective study of residual-disease monitoring of the ALL1/AF4 transcript in patients with t(4;11) acute lymphoblastic leukemia.
Blood
2000
, vol. 
95
 
1
(pg. 
96
-
101
)
50
Dombret
 
H
Gabert
 
J
Boiron
 
JM
, et al. 
Outcome of treatment in adults with Philadelphia chromosome-positive acute lymphoblastic leukemia: results of the prospective multicenter LALA-94 trial.
Blood
2002
, vol. 
100
 
7
(pg. 
2357
-
2366
)
51
Lee
 
S
Kim
 
YJ
Chung
 
NG
, et al. 
The extent of minimal residual disease reduction after the first 4-week imatinib therapy determines outcome of allogeneic stem cell transplantation in adults with Philadelphia chromosome-positive acute lymphoblastic leukemia.
Cancer
2009
, vol. 
115
 
3
(pg. 
561
-
570
)
52
Leguay
 
T
Witz
 
F
De Botton
 
S
, et al. 
Post-remission therapy with imatinib and HAM improve MRD before transplant for patients with Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL): results of the GRAALL AFR03 Study [abstract].
Blood (ASH Annual Meeting Abstracts)
2006
, vol. 
108
 
11
 
Abstract 1877
53
Yanada
 
M
Sugiura
 
I
Takeuchi
 
J
, et al. 
Prospective monitoring of BCR-ABL1 transcript levels in patients with Philadelphia chromosome-positive acute lymphoblastic leukaemia undergoing imatinib-combined chemotherapy.
Br J Haematol
2008
, vol. 
143
 
4
(pg. 
503
-
510
)
54
Pfeifer
 
H
Lange
 
T
Wystub
 
S
, et al. 
Prevalence and dynamics of bcr-abl kinase domain mutations during imatinib treatment differ in patients with newly diagnosed and recurrent bcr-abl positive acute lymphoblastic leukemia.
Leukemia
2012
, vol. 
26
 
7
(pg. 
1475
-
1481
)
55
Raff
 
T
Gökbuget
 
N
Lüschen
 
S
, et al. 
Molecular relapse in adult standard-risk ALL patients detected by prospective MRD monitoring during and after maintenance treatment: data from the GMALL 06/99 and 07/03 trials.
Blood
2007
, vol. 
109
 
3
(pg. 
910
-
915
)
56
Mitterbauer
 
G
Nemeth
 
P
Wacha
 
S
, et al. 
Quantification of minimal residual disease in patients with BCR-ABL-positive acute lymphoblastic leukaemia using quantitative competitive polymerase chain reaction.
Br J Haematol
1999
, vol. 
106
 
3
(pg. 
634
-
643
)
57
Miyamura
 
K
Tanimoto
 
M
Morishima
 
Y
, et al. 
Detection of Philadelphia chromosome-positive acute lymphoblastic leukemia by polymerase chain reaction: possible eradication of minimal residual disease by marrow transplantation.
Blood
1992
, vol. 
79
 
5
(pg. 
1366
-
1370
)
58
Preudhomme
 
C
Henic
 
N
Cazin
 
B
, et al. 
Good correlation between RT-PCR analysis and relapse in Philadelphia (Ph1)-positive acute lymphoblastic leukemia (ALL).
Leukemia
1997
, vol. 
11
 
2
(pg. 
294
-
298
)
59
Radich
 
J
Gehly
 
G
Lee
 
A
, et al. 
Detection of bcr-abl transcripts in Philadelphia chromosome-positive acute lymphoblastic leukemia after marrow transplantation.
Blood
1997
, vol. 
89
 
7
(pg. 
2602
-
2609
)
60
Ribera
 
J
Oriol
 
A
Morgades
 
M
, et al. 
Treatment of high-risk (HR) Philadelphia chromosome-negative (Ph−) adult acute lymphoblastic leukemia (ALL) according to classical risk factors and minimal residual disease (MRD): interim results of the PETHEMA ALL-AR-03 Trial [abstract].
Blood (ASH Annual Meeting Abstracts)
2006
, vol. 
108
 
11
 
Abstract 1872
61
Gökbuget
 
N
Kneba
 
M
Raff
 
T
, et al. 
Risk-adapted treatment according to minimal residual disease in adult ALL.
Best Pract Res Clin Haematol
2002
, vol. 
15
 
4
(pg. 
639
-
652
)
62
Topp
 
MS
Kufer
 
P
Gokbuget
 
N
, et al. 
Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival.
J Clin Oncol
2011
, vol. 
29
 
18
(pg. 
2493
-
2498
)
63
Wassmann
 
B
Pfeifer
 
H
Stadler
 
M
, et al. 
Early molecular response to posttransplantation imatinib determines outcome in MRD+ Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL).
Blood
2005
, vol. 
106
 
2
(pg. 
458
-
463
)
64
Huguet
 
F
Leguay
 
T
Raffoux
 
E
, et al. 
Pediatric-inspired therapy in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: the GRAALL-2003 study.
J Clin Oncol
2009
, vol. 
27
 
6
(pg. 
911
-
918
)
65
Goldstone
 
AH
Richards
 
SM
Lazarus
 
HM
, et al. 
In adults with standard-risk acute lymphoblastic leukemia, the greatest benefit is achieved from a matched sibling allogeneic transplantation in first complete remission, and an autologous transplantation is less effective than conventional consolidation/maintenance chemotherapy in all patients: final results of the International ALL Trial (MRC UKALL XII/ECOG E2993).
Blood
2008
, vol. 
111
 
4
(pg. 
1827
-
1833
)
66
Boissel
 
N
Auclerc
 
MF
Lheritier
 
V
, et al. 
Should adolescents with acute lymphoblastic leukemia be treated as old children or young adults? Comparison of the French FRALLE-93 and LALA-94 trials.
J Clin Oncol
2003
, vol. 
21
 
5
(pg. 
774
-
780
)
67
Hallböök
 
H
Gustafsson
 
G
Smedmyr
 
B
Soderhall
 
S
Heyman
 
M
Treatment outcome in young adults and children > 10 years of age with acute lymphoblastic leukemia in Sweden: a comparison between a pediatric protocol and an adult protocol.
Cancer
2006
, vol. 
107
 
7
(pg. 
1551
-
1561
)
68
Stock
 
W
La
 
M
Sanford
 
B
, et al. 
What determines the outcomes for adolescents and young adults with acute lymphoblastic leukemia treated on cooperative group protocols? A comparison of Children's Cancer Group and Cancer and Leukemia Group B studies.
Blood
2008
, vol. 
112
 
5
(pg. 
1646
-
1654
)
69
Flohr
 
T
Schrauder
 
A
Cazzaniga
 
G
, et al. 
Minimal residual disease-directed risk stratification using real-time quantitative PCR analysis of immunoglobulin and T-cell receptor gene rearrangements in the international multicenter trial AIEOP-BFM ALL 2000 for childhood acute lymphoblastic leukemia.
Leukemia
2008
, vol. 
22
 
4
(pg. 
771
-
782
)
70
Secker-Walker
 
LM
Craig
 
JM
Hawkins
 
JM
Hoffbrand
 
AV
Philadelphia positive acute lymphoblastic leukemia in adults: age distribution, BCR breakpoint and prognostic significance.
Leukemia
1991
, vol. 
5
 
3
(pg. 
196
-
199
)
71
Fielding
 
AK
Current treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia.
Hematology Am Soc Hematol Educ Program
2011
, vol. 
2011
 (pg. 
231
-
237
)
72
Pfeifer
 
H
Wassmann
 
B
Pavlova
 
A
, et al. 
Kinase domain mutations of BCR-ABL frequently precede imatinib-based therapy and give rise to relapse in patients with de novo Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL).
Blood
2007
, vol. 
110
 
2
(pg. 
727
-
734
)
73
Soverini
 
S
Vitale
 
A
Poerio
 
A
, et al. 
Philadelphia-positive acute lymphoblastic leukemia patients already harbor BCR-ABL kinase domain mutations at low levels at the time of diagnosis.
Haematologica
2011
, vol. 
96
 
4
(pg. 
552
-
557
)
74
Mannelli
 
F
Gianfaldoni
 
G
Intermesoli
 
T
, et al. 
CD20 expression has no prognostic role in Philadelphia-negative B-precursor acute lymphoblastic leukemia: new insights from the molecular study of minimal residual disease.
Haematologica
2012
, vol. 
97
 
4
(pg. 
568
-
571
)
75
Thomas
 
DA
O'Brien
 
S
Jorgensen
 
JL
, et al. 
Prognostic significance of CD20 expression in adults with de novo precursor B-lineage acute lymphoblastic leukemia.
Blood
2009
, vol. 
113
 
25
(pg. 
6330
-
6337
)
76
Thomas
 
DA
O'Brien
 
S
Faderl
 
S
, et al. 
Chemoimmunotherapy with a modified hyper-CVAD and rituximab regimen improves outcome in de novo Philadelphia chromosome-negative precursor B-lineage acute lymphoblastic leukemia.
J Clin Oncol
2010
, vol. 
28
 
24
(pg. 
3880
-
3889
)
77
Hoelzer
 
D
Huettmann
 
A
Kaul
 
F
, et al. 
Immunochemotherapy with rituximab improves molecular CR rate and outcome in CD20+ B-lineage standard and high risk patients: results of 263 CD20+ patients studied prospectively in GMALL Study 07/2003 [abstract].
Blood (ASH Annual Meeting Abstracts)
2010
, vol. 
116
 
21
 
Abstract 170
78
Committee for Proprietary Medicinal Products
Note for Guidance on Statistical Principles for Clinical Trials (CPMP/ICH/363/96): European Medicine Agency; 1998.
Accessed June 9, 2012 
79
Organisation for Economic Co-Operation and Development (OECD)
Good Laboratory Practice: OECD Principles and Guidance for Compliance Monitoring.
Paris, France
OECD Publishing 2006
80
United States Department of Health and Human Services
Clinical Laboratory Improvement Amendments; 2011.
Accessed June 9, 2012 
81
Fielding
 
AK
Current therapeutic strategies in adult acute lymphoblastic leukemia.
Hematol Oncol Clin North Am
2011
, vol. 
25
 
6
(pg. 
1255
-
1279
)
82
Mullighan
 
CG
New strategies in acute lymphoblastic leukemia: translating advances in genomics into clinical practice.
Clin Cancer Res
2011
, vol. 
17
 
3
(pg. 
396
-
400
)
83
Rowe
 
JM
Prognostic factors in adult acute lymphoblastic leukaemia.
Br J Haematol
2010
, vol. 
150
 
4
(pg. 
389
-
405
)
84
Hoelzer
 
D
Thiel
 
E
Loffler
 
H
, et al. 
Prognostic factors in a multicenter study for treatment of acute lymphoblastic leukemia in adults.
Blood
1988
, vol. 
71
 
1
(pg. 
123
-
131
)
85
Rowe
 
JM
Buck
 
G
Burnett
 
AK
, et al. 
Induction therapy for adults with acute lymphoblastic leukemia. Results of more than 1500 patients from the international ALL trial: MRC UKALL XII/ECOG E2993.
Blood
2005
, vol. 
106
 
12
(pg. 
3760
-
3767
)
86
Moorman
 
AV
Harrison
 
CJ
Buck
 
GA
, et al. 
Karyotype is an independent prognostic factor in adult acute lymphoblastic leukemia (ALL): analysis of cytogenetic data from patients treated on the Medical Research Council (MRC) UKALLXII/Eastern Cooperative Oncology Group (ECOG) 2993 trial.
Blood
2007
, vol. 
109
 
8
(pg. 
3189
-
3197
)
87
Pullarkat
 
V
Slovak
 
ML
Kopecky
 
KJ
Forman
 
SJ
Appelbaum
 
FR
Impact of cytogenetics on the outcome of adult acute lymphoblastic leukemia: results of Southwest Oncology Group 9400 study.
Blood
2008
, vol. 
111
 
5
(pg. 
2563
-
2572
)
88
Burmeister
 
T
Gökbuget
 
N
Schwartz
 
S
, et al. 
Clinical features and prognostic implications of TCF3-PBX1 and ETV6-RUNX1 in adult acute lymphoblastic leukemia.
Haematologica
2010
, vol. 
95
 
2
(pg. 
241
-
246
)
89
Piccaluga
 
PP
Malagola
 
M
Rondoni
 
M
, et al. 
Poor outcome of adult acute lymphoblastic leukemia patients carrying the (1;19)(q23;p13) translocation.
Leuk Lymphoma
2006
, vol. 
47
 
3
(pg. 
469
-
472
)
90
Flex
 
E
Petrangeli
 
V
Stella
 
L
, et al. 
Somatically acquired JAK1 mutations in adult acute lymphoblastic leukemia.
J Exp Med
2008
, vol. 
205
 
4
(pg. 
751
-
758
)
91
Martinelli
 
G
Iacobucci
 
I
Storlazzi
 
CT
, et al. 
IKZF1 (Ikaros) deletions in BCR-ABL1-positive acute lymphoblastic leukemia are associated with short disease-free survival and high rate of cumulative incidence of relapse: a GIMEMA AL WP report.
J Clin Oncol
2009
, vol. 
27
 
31
(pg. 
5202
-
5207
)
92
Mi
 
JQ
Wang
 
X
Yao
 
Y
, et al. 
Newly diagnosed acute lymphoblastic leukemia in China (II): prognosis related to genetic abnormalities in a series of 1091 cases.
Leukemia
2012
, vol. 
26
 
7
(pg. 
1507
-
1516
)
93
Baldus
 
CD
Martus
 
P
Burmeister
 
T
, et al. 
Low ERG and BAALC expression identifies a new subgroup of adult acute T-lymphoblastic leukemia with a highly favorable outcome.
J Clin Oncol
2007
, vol. 
25
 
24
(pg. 
3739
-
3745
)
94
Ben
 
AR
Asnafi
 
V
Leguay
 
T
, et al. 
Pediatric-inspired intensified therapy of adult T-ALL reveals the favorable outcome of NOTCH1/FBXW7 mutations, but not of low ERG/BAALC expression: a GRAALL study.
Blood
2011
, vol. 
118
 
19
(pg. 
5099
-
5107
)
95
Asnafi
 
V
Buzyn
 
A
Le
 
NS
, et al. 
NOTCH1/FBXW7 mutation identifies a large subgroup with favorable outcome in adult T-cell acute lymphoblastic leukemia (T-ALL): a Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) study.
Blood
2009
, vol. 
113
 
17
(pg. 
3918
-
3924
)
96
Mansour
 
MR
Sulis
 
ML
Duke
 
V
, et al. 
Prognostic implications of NOTCH1 and FBXW7 mutations in adults with T-cell acute lymphoblastic leukemia treated on the MRC UKALLXII/ECOG E2993 protocol.
J Clin Oncol
2009
, vol. 
27
 
26
(pg. 
4352
-
4356
)
97
Ottmann
 
OG
Wassmann
 
B
Pfeifer
 
H
, et al. 
Imatinib compared with chemotherapy as front-line treatment of elderly patients with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL).
Cancer
2007
, vol. 
109
 
10
(pg. 
2068
-
2076
)
Sign in via your Institution