Minimal residual disease: optimal methods, timing, and clinical relevance for an individual patient

Similar documents
Standard risk ALL (and its exceptions

First relapsed childhood ALL Role of chemotherapy

MRD in ALL: Correct interpretation in clinical practice. Deepak Bansal Prof., Pediatric Hematology-Oncology unit PGIMER, Chandigarh

Improving the Identification of High Risk Precursor B Acute Lymphoblastic Leukemia Patients with Earlier Quantification of Minimal Residual Disease

J Clin Oncol 26: by American Society of Clinical Oncology INTRODUCTION

Introduction LYMPHOID NEOPLASIA

Original article. Key words: Lymphoblastic leukemia, Acute, Childhood, Minimal residual disease, Flow cytometry

Risk Stratification in Childhood Leukemia

Multiparameter flow cytometry can be used to

Current Indications of Bone Marrow Transplantation (BMT) in Pediatric Malignant Conditions; a Review

Thiopurine Methyltransferase (TPMT) Genotype and Early Treatment Response to Mercaptopurine in Childhood Acute Lymphoblastic Leukemia

Published Ahead of Print on January 12, 2017, as doi: /haematol Copyright 2017 Ferrata Storti Foundation.

Summary. Olga Zając, Katarzyna Derwich, Katarzyna Stefankiewicz, Jacek Wachowiak. Rep Pract Oncol Radiother, 2007; 12(5):

Correlation of Sex and Remission of Acute Lymphoblastic Leukemia-L1 (ALL-L1) in Children

PAX5-JAK2 fusion in acute lymphoblastic leukaemia. Dr Andrew Baldi Royal Children s Hospital 24 February 2017 Melbourne

Case 1. Sa A.Wang, MD UT MD Anderson Cancer Center Houston, TX

Elisabeth Koller 3rd Medical Dept., Center for Hematology and Oncology, Hanusch Hospital, Vienna, Austria

Johann Hitzler, MD, FRCPC, FAAP Jacqueline Halton, MD, FRCPC Jason D. Pole, PhD

How Do You Measure Success in ALL?: Assessment of MRD

Acute Lymphoblastic Leukemia (ALL) Ryan Mattison, MD University of Wisconsin March 2, 2010

Received April 13, 2010

High-Risk Pediatric Acute Lymphoblastic Leukemia: To Transplant or Not to Transplant?

Improved outcome in childhood ALL with intensive consolidation and hematopoietic stem cell transplant

Philadelphia chromosome-positive acute lymphoblastic leukemia in childhood

Minimal residual disease (MRD) in AML; coming of age. Dr. Mehmet Yılmaz Gaziantep University Medical School Sahinbey Education and Research hospital

Acute Lymphoblastic and Myeloid Leukemia

MP BCR-ABL1 Testing in Chronic Myelogenous Leukemia and Acute Lymphoblastic Leukemia

Controversies in Hematology: Case-Based Discussion. Acute leukemia in Adolescents and Young adults October 2018, Chiang Mai Thailand

Can ALL be managed without chemotherapy/transplant? (Position: NO) D.Hoelzer J.W.Goethe University Frankfurt

Supplementary appendix

Mixed Phenotype Acute Leukemias

ADVANCES IN CHILDHOOD ACUTE LEUKEMIAS : GENERAL OVERVIEW

Corporate Medical Policy. Policy Effective February 23, 2018

IKZF1 deletion is an independent predictor of outcome in pediatric acute lymphoblastic leukemia treated according to the ALL-BFM 2000 protocol

AIEOP-BFM consensus guidelines 2016 flow cytometric immunophenotyping in pediatric acute leukemias

Clinical characteristics and prognosis of pediatric patients with B cell acute lymphoblastic leukemia relapse

The probability of curing children with acute. brief report

Identification of residual leukemic cells by flow cytometry in childhood

Prognostic significance of monitoring leukemia-associated immunophenotypes by eight-color flow cytometry in adult B-acute lymphoblastic leukemia

Management of Acute Lymphoblastic Leukemia

Abstract 861. Stein AS, Topp MS, Kantarjian H, Gökbuget N, Bargou R, Litzow M, Rambaldi A, Ribera J-M, Zhang A, Zimmerman Z, Forman SJ

Handout for lecture on lymphoblastic neoplasms presented by Rob McKenna

ETP - Acute Lymphoblastic Leukaemia

BCR ABL1 like ALL: molekuliniai mechanizmai ir klinikinė reikšmė. IKAROS delecija: molekulinė biologija, prognostinė reikšmė. ASH 2015 naujienos

Acute myeloid leukemia. M. Kaźmierczak 2016

2011: ALL Pre-HCT. Subsequent Transplant

Prophylactic Cranial Irradiation in Acute Lymphoblastic Leukemia: Is there still an indication? Celine Bicquart, MD Radiation Medicine May 5, 2010

Acute myeloid leukemia: prognosis and treatment. Dimitri A. Breems, MD, PhD Internist-Hematoloog Ziekenhuis Netwerk Antwerpen Campus Stuivenberg

Treatment of High-risk Acute Lymphoblastic Leukemia With Precision Medicine

TCF3 breakpoints of TCF3-PBX1 (patients 1a 5a) and TCF3-HLF (patients 6a 9a and11a) translocations.

TRANSPARENCY COMMITTEE OPINION. 14 February 2007

MRD detection in AML. Adriano Venditti Hematology Fondazione Policlinico Tor Vergata Rome

MANAGEMENT OF ACUTE LYMPHOBLASTIC LEUKEMIA. BY Dr SUBHASHINI 1 st yr PG DEPARTMENT OF PEDIATRICS

Childhood Acute Lymphoblastic Leukemia

Relapsed acute lymphoblastic leukemia. Lymphoma Tumor Board. July 21, 2017

Prognostic significance of leukopenia in childhood acute lymphoblastic leukemia

Outcome of acute leukemia patients with central nervous system (CNS) involvement treated with total body or CNS irradiation before transplantation

The Role of Prognostic Features in the Treatment of Childhood Acute Lymphoblastic Leukemia

ACUTE LYMPHOBLASTIC LEUKEMIA

Acute Lymphoblastic Leukaemia Guidelines

Characteristics and Outcome of Therapy-Related Acute Promyelocytic Leukemia After Different Front-line Therapies

Acute leukemia and myelodysplastic syndromes

BiTE in ALL and AML. Ibrahim Aldoss, MD Assistant Professor, City of Hope Hematology and Hematopoietic Cell Transplantation

Minimal residual disease is an important predictive factor of outcome in children with relapsed high-risk acute lymphoblastic leukemia

MUD SCT for Paediatric AML?

Predicting relapse risk in childhood acute lymphoblastic leukaemia

Pediatric Acute Leukemia: The Effect of Prognostic Factors on Clinical Outcomes at Phramongkutklao Hospital, Bangkok, Thailand

Molecular Markers in Acute Leukemia. Dr Muhd Zanapiah Zakaria Hospital Ampang

Myelodysplasia/Myeloproliferative Neoplasms (MDS/MPN) Post-HCT Data

High-risk childhood acute lymphoblastic leukemia in first remission treated with novel intensive chemotherapy and allogeneic transplantation

National Institute for Health and Care Excellence. Single Technology Appraisal (STA)

ΙL-8 and MCP-1 in children with acute lymphoblastic leukemia and potential correlation with neurotoxicity and thromboembolic phenomena

Pediatric Acute Lymphoblastic Leukemia. Highlights of ASH 2015

Jordi Esteve Hospital Clínic (Barcelona) Acute Leukemia Working Party. The European Group for Blood and Marrow Transplantation

ALL-RELAPSE GROUP Päivi Lähteenmäki on behalf of the group

Lymphoblastic Leukemia / Lymphoma

Ferrata Storti Foundation

J Clin Oncol 29: by American Society of Clinical Oncology INTRODUCTION

Guideline on the use of minimal residual disease as a clinical endpoint in multiple myeloma studies

Adult ALL: NILG experience

Leukemia. Andre C. Schuh. Princess Margaret Cancer Centre Toronto

Role of FISH in Hematological Cancers

HEMATOLOGIC MALIGNANCIES BIOLOGY

ONE STEP MULTIPLEX RT-PCR FOR BCRlABL GENE IN MALAY PATIENTS DIAGNOSED AS LEUKAEMIA

IN PHILADELPHIA CHROMOSOME positive (Ph )

Prognostic significance of minimal residual disease in infants with acute lymphoblastic leukemia treated within the Interfant-99 protocol

UNTHSC Scholarly Repository. University of North Texas Health Science Center

PDF of Trial CTRI Website URL -

Test Name Results Units Bio. Ref. Interval. Positive

Early Clearance of Peripheral Blood Blasts Predicts Response to Induction Chemotherapy in Acute Myeloid Leukemia

Case Report T-Cell Lymphoblastic Leukemia/Lymphoma: Relapse 16 Years after First Remission

Childhood Acute Lymphoblastic Leukemia: Progress Through Collaboration

Minimal Residual Disease in Hematologic Malignancies

Kerrie Clerici, Michael Swain, Dominic Fernandez, Julia Schulz, Matthew Archer, Janine Campbell

Timing and complications of allogeneic stem cell transplant in Ph + ALL

Philadelphia-Positive B-Acute Lymphoblastic Leukemia: Does it Differ from Philadelphia-Negative One?

NUP214-ABL1 Fusion: A Novel Discovery in Acute Myelomonocytic Leukemia

Extramedullary precursor T-lymphoblastic transformation of CML at presentation

Acute leukemia & agressive lymphoma in children

Molecular Advances in Hematopathology

Transcription:

DYNAMIC DISCOVERIES AND DIRECTIONS IN PEDIATRIC LEUKEMIAS Minimal residual disease: optimal methods, timing, and clinical relevance for an individual patient Martin Schrappe 1 1 Department of Pediatrics at the Christian-Albrechts-University of Kiel, University Medical Center Schleswig-Holstein, Kiel, Germany After approximately 20 years of development and after several prospective clinical trials, the detection of minimal residual disease (MRD) has emerged as part of state-of-the-art diagnostics to guide the majority of contemporary treatment programs both in pediatric and adult acute lymphoblastic leukemia (ALL). For ALL, several methods of MRD analysis are available, but 2 are widely applicable. One is based on the detection of aberrant expression of leukemia specific antigens by flow cytometry and the other one uses the specific rearrangements of the TCR or Ig genes, which can be detected by quantitative PCR in the DNA of leukemic cells. In some cases with known fusion genes such as BCR/ABL, RT-PCR can be used as a third method of identifying leukemic cells by analyzing RNA in patient samples. Clinical application of such sophisticated tools in the stratification and treatment of ALL requires reliable, reproducible, and quality-assured methods to ensure patient safety. Introduction Several pediatric and adult acute lymphoblastic leukemia (ALL) study groups have established informative checkpoints in their respective treatment protocols that are being used for risk stratification. Predefined levels of minimal residual disease (MRD) at these checkpoints predict the risk of relapse, but large differences in the response rates of various ALL subtypes have been found. MRD detection before and after stem cell transplantation (SCT) has also been established for individual treatment adaptation, and whether response assessment by MRD detection can be used as a surrogate clinical end point for the evaluation of single agents or combination therapy is now being investigated. In vivo sensitivity of ALL can be defined by the early blast cell reduction in the peripheral blood (PB) or BM after exposure to one or several antileukemic agents. Systematic response assessment is regarded as a necessary tool to risk-stratify patients with ALL. Lack of adequate response, in particular at the end of remission induction, indicates poor prognosis, but this may vary significantly according to individual patient characteristics. 1 Cytological response assessment early in treatment can distinguish prognostic subgroups very well if done by experienced investigators in one reference laboratory either in the PB (day 8) 2 or the BM (day 15). 3 Despite the clear separation of risk groups through BM cytology, two-thirds of the relapses are nevertheless comprised in patients with M1 or M2 BM on day 15 of induction. 3 Therefore, advanced and highly sensitive methods for response assessment were needed and developed to detect MRD. The choice of technique for the detection of MRD depends mainly on the aims of the clinical trial and on the availability of resources. 4-7 If MRD analysis is used to identify high-risk (HR) patients, it may be sufficient to use a faster but less sensitive method. 8 If the aim is also to identify super responders, it is necessary to seek the highest sensitivity because the lack of signal in the corresponding MRD investigation must be absolutely reliable, particularly if such patients shall be spared additional therapy or even be assigned to reduced therapy. The second most important prerequisite is the prospective analysis with the MRD method of choice to determine the prognostic significance of certain MRD levels on the background of a predefined uniform chemotherapy regimen. 4,9-15 It is now widely acknowledged that MRD detection is part of state-of-the-art diagnostics and is necessary in the management of ALL. 7,16-18 This short review describes the clinical utility of MRD application in the treatment of ALL, with a specific focus on pediatric ALL. The necessity to work with well-established definitions is emphasized. In addition, a few examples in which MRD was used for assessment of efficacy of novel treatment modalities, either alone or in combination with conventional end points, are briefly reviewed. Pros and cons of commonly used techniques for MRD assessment Table 1 illustrates some key characteristics of the methods used in ALL for MRD detection. These factors were recently reevaluated before being put into a consensus recommendation in Europe. 4 Although RT-PCR may provide highly sensitive MRD detection in ALL with specific fusion genes (eg, BCR/ABL or MLL rearrangements), the main limitation is the lack of such targets in the majority of ALL cases. It is highly relevant, however, for quantitative monitoring of BCR/ABL ALL when being treated with novel tyrosine kinase inhibitors. 4,19 Both PCR and flow cytometry (FCM) require minimum cell counts at diagnosis and during follow-up. This needs to be strictly observed because there is a risk of misinterpretation of quantitative readouts if cell counts are low. 5,7 For reproducible quantification, clear guidelines are needed, especially if several reference laboratories perform the initial and follow-up diagnostics in the same clinical trial. 20 MRD after 1 or 2 weeks of therapy: impact of early response assessment In many ALL protocols, days 8 and 15 of induction therapy are considered the first checkpoints to test the in vivo sensitivity of the leukemia in the individual patient. 3,21-24 The general message for the individual patient is simple: the fast reduction or even elimination of the leukemia (or its predominant clone) is highly predictive of Hematology 2012 137

Table 1. Characteristics of the techniques currently used for MRD detection in ALL 4 PCR analysis of Ig and TCR gene rearrangements PCR analysis of BCR-ABL transcripts Multiparameter FCM Sensitivity RQ-PCR: 10-4 -10-5 10-4 -10-6 3- to 4-color: 10-3 -10-4 6- to 9-color: 10-4 -10-5 Also depends on cell input Quantitative range RQ-PCR: 10-2 -10-4 Not yet defined Not yet defined Applicability pcb-all: 90%-95% T-ALL: 90%-95% Advantages Disadvantages High sensitivity High degree of standardization reached Well-established stratification tool in various clinical protocols Most published data for evidencebased treatment decisions Applicable for almost all ALL patients Stability of DNA (multicenter setting) Time-consuming marker characterization Pretreatment sample required to sequence the patient-specific diagnostic clone Potential instability of targets (clonal evolution phenomena) Extensive knowledge and experience needed Relatively expensive Ph indicates Philadelphia chromosome positive. Ph ALL (5%-8% of children with pcb-all, 30%-35% of adults with pcb-all) High sensitivity Stability of target during course of treatment Fast Relatively easy/cheap Applicable only in Ph patients Instability of RNA Differences in expression levels possible Standardization necessary Risk of false positivity due to contamination pcb-all: 80%-95% T-ALL: 90%-95% Depends also on number of colors Applicable for almost all ALL patients Rapid Quantitative Additional information on benign cells Additional information on malignant cells Growing standardization throughout Europe Immunophenotypic shifts Expanded pcb-cell compartment during regeneration Low cellularity during/after induction Relatively expensive (depends on number of markers/colors and ulterior cytometer utilizations) Limited sensitivity/applicability using 3- to 4-color FCM 6-color FCM: extensive knowledge and experience for sensitive and standardized analysis needed superior relapse-free survival. 9,25 Borowitz et al demonstrated that distinct levels of MRD (by FCM) in the PB at day 8 of induction therapy in a large set of precursor B-cell ALL (pcb ALL; n 2143) patients were associated with the probability of event-free survival (pefs). Patients with 0.01% or less MRD (called MRD ) had a 5-year pefs of 90% 2%, whereas the worst group, with MRD at 10%, had a pefs of only 54% 7%. Interestingly, among the patients who cleared MRD in the BM by the end of induction (day 29 in that regimen), the prognosis of patients with more than 1% MRD in the PB at day 8 (18% of all) was worse than that of patients who had responded more favorably at day 8 (79% 4% vs 90% 1%, respectively). 9 Another large study performed by Basso et al compared BM day 15 MRD results generated by FCM with cytomorphological response assessment and PCR analysis of MRD at the end of induction (day 33) and at the end of consolidation (day 78). Levels of MRD in the BM at day 15 were well correlated with risk of relapse: less than 0.1% of MRD conferred a cumulative incidence of relapse of 8% 1.7% in pcb-cell ALL and only 3.3% 3.3% in T-cell ALL. These fast responders comprised 43% of pcb-cell ALL patients and 34% of T-cell ALL patients, respectively. In both major subgroups of ALL, a distinct poor-risk group was identified by high levels of MRD ( 10%) at day 15, comprising 10% of pcb-all and 21% of T-ALL, respectively: the 5-year cumulative incidence of relapse was 45.5% 6.8% in pcb-all and 55.6% 11.7% in T-ALL. 15 Investigators from Prague reported on the specific prognostic information of MRD in PB on day 15 of a Berlin-Frankfurt-Münster (BFM) based regimen compared with MRD in the BM (both measured by PCR in PcB-ALL). 26 They were able to demonstrate that 35 of 78 patients were MRD (or low positive below 10 4 ) in the PB at day 15, with an excellent relapse-free survival of 100%. At day 8 of therapy, the same low level of MRD also identified a subset of patients without relapse, but that group was much smaller. Therefore, day 15 appears to be an informative MRD checkpoint, both by MRD in PB and in the BM if treatment follows the BFM regimen. This is somewhat surprising, because the first 2 weeks of this therapy comprised only a 7-day prophase with prednisone and 1 dose of intrathecal methotrexate, followed by vincristine and daunorubicin on day 8 and asparaginase on day 12. Usually, the day 15 PB and BM samples are taken before the next doses of vincristine, daunorubicin, and asparaginase are given on day 15. 21 Is early MRD response assessment by FCM sufficient to identify the patient at risk of relapse and the patient who can be spared therapy? To this end, the analysis by Basso et al reveals that 41% of the patients who are classified as low risk by FCM ( 0.1%) on day 15 of induction therapy according to protocol Italian Association of Pediatric Hematology and Oncology (AIEOP)-BFM ALL 2000 are not MRD by PCR at the end of induction (day 33). The rate of relapses was very low among FCM day 15 good responders ( 0.1%), but 12 of 19 recurrences within that group were observed in the subset of patients with any level of MRD positivity by PCR at day 33. 15 Another large study comparing MRD results obtained in the same protocol by several reference laboratories also revealed that concordance, sensitivity, and specificity of MRD analysis by FCM and PCR largely depend on the time in therapy and on the cell 138 American Society of Hematology

Figure 1. pcb- and T-ALL in AIEOP-BFM ALL 2000. Shown is the distribution of patients in the different MRD risk groups according to immunophenotype. 29,37 MRD-SR indicates no MRD detectable at days 33 and 78 from diagnosis, with a sensitivity for the 2 targets of at least 10 4 ; MRD-HR indicates that the MRD level at day 78 is 10 3 ; and MRD-IR indicates all other constellations of MRD. material used. 27 Both studies reflect the practical realities of large clinical study groups, and these comparisons reveal that both techniques, FCM on day 15 and PCR at end of induction and at additional later time points, may be complementary in optimizing the treatment stratification. Rapid clearance of blasts as measured by PCR-based MRD detection is also associated with superior prognosis in standard-risk (SR) adult ALL. Approximately 10% of the patients have undetectable or low positive ( 10 4 ) MRD at days 11 and 24 of induction. Their prognosis is excellent, with 3-year disease-free and overall survival of 100%. 28 Prognostic information based on MRD at end of induction In the largest study for de novo ALL published so far, the AIEOP and BFM study groups used MRD by PCR for risk stratification in a total of 3184 pcb-all and 464 T-ALL patients. All patients were treated by identical chemotherapy in the first 9 weeks of therapy. Large differences in pefs between MRD-defined subgroups were found. 29,30 MRD was analyzed in the BM at the end of induction (day 33) and at the end of induction consolidation (day 78) and revealed major differences in the kinetics of the MRD response between pcb-all and T-ALL (Figure 1) and large similarities in the pefs of all 3 MRD risk groups: In pcb-all, the pefs at 5 years was 92.3% 0.9% for MRD-SR, 77.6% 1.3% for MRD-intermediate risk (MRD-IR), and 50.1% 4.1% for MRD- HR. The corresponding results in T-ALL were 93.0% 3.0% (5-year pefs for MRD-SR), 80.6% 2.3% (MRD-IR), and 49.8% 5.1% (MRD-HR), respectively. Interestingly, the AIEOP- BFM trial showed that patients with the unfavorable subtype of early T-precursor ALL 31 were also more frequently slow MRD responders, which resulted in treatment in the most intensive treatment group. Within pcb-all, the prognostic impact of MRD was maintained, even in the 2 large subgroups of TEL/AML1 and hyperdiploid ALL. 29 Two major differences between pcb-all and T-ALL can be found: (1) MRD at the end of induction (day 33) is more informative in pcb-all, whereas MRD at the end of consolidation (day 78) is more informative in T-ALL, and (2) MRD levels in pcb-all are correlated with risk of systemic relapse, whereas MRD in T-ALL is predictive of both systemic and extramedullary relapse. The results obtained here, in combination with the results obtained by FCM 15 in the same trial, extend the current risk group definition for MRD-HR patients. Any patient who has 10% leukemic blasts by FCM on day 15 is enrolled into the MRD-HR group; any patient with pcb-all who has MRD 10 3 (0.1%) at day 33 and is still MRD at day 78 is stratified into the MRD-HR group; any pcb-all patient with MRD 0.1% at day 15 is eligible for being considered MRD-SR if the PCR results (with at least one highly sensitive molecular target, quantitative range up to 10 4 ) at days 33 and 78 are also negative. When the results of Conter et al 29 are compared with the large study by Borowitz et al in pcb-all, 9 one important difference can be found: MRD negativity at end of induction is strongly associated with highly favorable outcome independent of other risk factors. In the Children s Oncology Group (COG) trials, MRD negativity at end of induction was also associated with favorable outcome, but the group was much larger and half of the events still occurred within that group. This most likely reflects the different MRD techniques and sensitivities, but the different treatment intensity applied in the COG trials may also have contributed to this observation. Other groups have also demonstrated the prognostic significance of end of induction MRD despite differences in the composition of induction therapy. 4,32,33 Postremission MRD and subsequent MRD surveillance at later time points Additional postremission MRD assessment has been performed in several clinical trials and these were reviewed by Brüggemann et al. 4 In AIEOP-BFM ALL 2000, all patients with MRD at 10 3 at day 78, were stratified into the MRD-HR group and then monitored after each reconsolidation element. This strategy is now used to adjust further chemotherapy and to prepare for allogeneic SCT. Although postinduction MRD was also found to have a significant prognostic impact in relapsed ALL, 14 MRD monitoring of the pre-sct response demonstrated the necessity to optimize the quality of remission before the procedure to prevent post-sct relapse. 34,35 Another recent investigation of MRD in adult patients with HR-ALL demonstrated that high MRD at day 71 after induction was associated with only 32% 6% disease-free survival compared with 66% 8% in HR patients with molecular complete remission. Allogeneic SCT appeared to be beneficial in patients with molecular failure, because those patients who did not receive Hematology 2012 139

Table 2. Proposals for definitions of MRD terms in ALL 4 MRD term Proposed definition Remission assessment Complete MRD response Minimal technical requirements fulfilled and nondetectable MRD* MRD persistence Minimal technical requirements fulfilled and quantifiable MRD positivity for at least 2 time-points Postremission monitoring MRD reappearance Minimal technical requirements fulfilled and conversion after MRD negativity to quantifiable MRD positivity *Treatment modifications depending on these measurements are strongly recommended to be based on at least 2 analyzed samples. Time points should be specified within the respective protocol and at least one relevant treatment element should be administered in between. MRD reappearance can be equated to MRD relapse (which would then be considered event ) if a particular ALL protocol has provided unequivocal evidence that MRD reappearance is closely associated with hematological relapse. the procedure had a disease-free survival of only 6% 5%. In SR patients, similar but less pronounced observations were made. If MRD at week 16 was analyzed, this difference between transplanted and nontransplanted patients could also be seen. 36 Therefore, allogeneic SCT should be offered rather soon because relapse will occur early in such patients. MRD in relapse and clonal evolution MRD monitoring in relapsed patients carries some potential pitfalls, mainly due to clonal evolution. 38 Detailed analysis of all molecular markers at first diagnosis and at the time of relapse may reveal a different origin of the predominant clone. 39 This implies that regular monitoring of MRD by those markers defined at first diagnosis may fail if clonal evolution occurs after first relapse. 40 Genome-wide copy number profiling in pairs of primary/relapsed ALL patients demonstrated a large number of differential somatic copy number alterations. 41,42 Definitions of MRD status in the management of ALL Although there is great consensus about the definition of complete cytological remission in ALL, 1 it has become more difficult to achieve common definitions for certain MRD terms. These definitions are urgently needed for comparison of results in clinical trials, but more importantly, to provide safe guidelines for patient management. The consensus proposal summarized by Brüggemann et al seems useful in providing the terms for remission assessment and postremission monitoring (Table 2). MRD as an end point for clinical intervention After MRD analysis became a common diagnostic tool in most multiagent clinical trials for ALL, the question arose of whether the MRD response can also be used as a primary end point in phase 2 trials. Many scenarios can be described in which response is used for the first screening to assess the efficacy of a single agent or a drug combination. However, the MRD response is only the more sensitive further development of the cytological response. Certainly, the efficacy of a given intervention with regard to response does not need to translate into the relevant clinical end points such as event-free and overall survival. However, recent novel immunotherapeutic strategies (eg, with an anti-cd3, anti-cd19 bispecific mab) that show strong efficacy in the MRD response may contribute to long-term remission because they may provide the necessary quality of remission needed to perform subsequent allogeneic SCT successfully. 43,44 In contrast, a British study comparing reinduction therapy with mitoxantrone versus idarubicin in relapsed pediatric ALL demonstrated that the MRD response was not predictive of treatment efficacy. 45 This observation indicates that MRD response may be misleading in drug evaluation as long as activity is considered to be equal to efficacy. Molecular predictors of MRD response An obvious disadvantage of any response assessment is the fact that the information about inadequate response to a given drug combination, such as induction therapy in ALL, comes only after these (inefficient) drugs have already been applied. Therefore, the patient may have been exposed to a toxic regimen without any benefit or with only minor benefit. The search for markers predictive of nonresponse at time of diagnosis is thus highly relevant. 46-48 Quite recently, it was also found that new genetic markers for ALL may have additional prognostic power alone or in combination with MRD response assessment. 49,50 High expression of CRLF2 was shown to predict a high relapse rate, even though the MRD response of this subgroup was considered rather favorable. 51 In addition, certain germline genetic variants were demonstrated to be associated with MRD response. 42,52 In summary, MRD has evolved as one of the most powerful diagnostic tools in the clinical management of ALL. At this time, it cannot yet be replaced by upfront diagnostic markers to predict response and relapse with the same level of sensitivity and specificity. Clinical intervention based on MRD results must rely on robust quality assurance of any given MRD detection method, because phenomena such as regenerating immature (normal) lymphocytes or clonal evolution of the leukemia may interfere with the interpretation of MRD results. Disclosures Conflict-of-interest disclosure: The author has received research funding from EUSAPharma, Medac, and Novartis. Off-label drug use: None disclosed. Correspondence Martin Schrappe, MD, Department of Pediatrics, Christian- Albrechts-University of Kiel, University Medical Center Schleswig- Holstein, Schwanenweg 20, 24105 Kiel, Germany; Phone: 49-431- 5971621; Fax: 49-431-5973966; e-mail: m.schrappe@ pediatrics.uni-kiel.de. References 1. Schrappe M, Hunger SP, Pui CH, et al. Outcomes after induction failure in childhood acute lymphoblastic leukemia. N Engl J Med. 2012;366(15):1371-1381. 2. Riehm H, Reiter A, Schrappe M, et al. Corticosteroiddependent reduction of leukocyte count in blood as a prognostic factor in acute lymphoblastic leukemia in childhood (therapy study ALL-BFM 83) [article in German]. Klin Padiatr. 1987; 199:151-160. 3. Lauten M, Moricke A, Beier R, et al. Prediction of outcome by early bone marrow response in childhood acute lymphoblastic 140 American Society of Hematology

leukemia treated in the trial ALL-BFM 95. Differential effects in precursor B-cell and T-cell leukemia. Haematologica. 2012; 97(7):1048 1056. 4. 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;24(1):521-535. 5. van der Velden VH, 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;21(4):604-611. 6. Szczepánski T, Harrison CJ, van Dongen JJ. Genetic aberrations in paediatric acute leukaemias and implications for management of patients. Lancet Oncol. 2010;11(9):880-889. 7. 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;22(4):771-782. 8. Coustan-Smith E, Ribeiro RC, Stow P, et al. A simplified flow cytometric assay identifies children with acute lymphoblastic leukemia who have a superior clinical outcome. Blood. 2006; 108(1):97-102. 9. 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;111(12):5477-5485. 10. 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;339(9):591-598. 11. Coustan-Smith E, Behm FG, Sanchez J, et al. Immunological detection of minimal residual disease in children with acute lymphoblastic leukaemia. Lancet. 1998;351(9102):550-554. 12. 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;99(12):1952-1958. 13. van Dongen JJM, Seriu T, Panzer-Grümayer ER, et al. Prognostic value of minimal residual disease in childhood acute lymphoblastic leukemia: A prospective study of the International BFM Study Group. Lancet. 1998;352(9142):1731-1738. 14. Eckert C, Biondi A, Seeger K, et al. Prognostic value of minimal residual disease in relapsed childhood acute lymphoblastic leukaemia. Lancet. 2001;358(9289):1239-1241. 15. Basso G, Veltroni M, Valsecchi MG, et al. Risk of relapse of childhood acute lymphoblastic leukemia is predicted by flow cytometric measurement of residual disease on day 15 bone marrow. J Clin Oncol. 2009;27(31):5168-5174. 16. Biondi A, Valsecchi MG, Seriu T, et al. Molecular detection of minimal residual disease is a strong predictive factor of relapse in childhood B-lineage acute lymphoblastic leukemia with medium risk features. A case control study of the International BFM study group. Leukemia. 2000;14(11):1939-1943. 17. Schultz KR, Pullen DJ, Sather HN, et al. Risk- and responsebased classification of childhood B-precursor acute lymphoblastic leukemia: a combined analysis of prognostic markers from the Pediatric Oncology Group (POG) and Children s Cancer Group (CCG). Blood. 2007;109(3):926-935. 18. McGregor S, McNeer J, Gurbuxani S. Beyond the 2008 World Health Organization classification: the role of the hematopathology laboratory in the diagnosis and management of acute lymphoblastic leukemia. Semin Diagn Pathol. 2012;29(1):2-11. 19. 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;106(2):458-463. 20. 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;74(6):331-340. 21. Möricke A, Reiter A, Zimmermann M, et al. Risk-adjusted therapy of acute lymphoblastic leukemia can decrease treatment burden and improve survival: treatment results of 2169 unselected pediatric and adolescent patients enrolled in the trial ALL-BFM 95. Blood. 2008;111(9):4477-4489. 22. Möricke A, Zimmermann M, Reiter A, et al. Long-term results of five consecutive trials in childhood acute lymphoblastic leukemia performed by the ALL-BFM study group from 1981 to 2000. Leukemia. 2010;24(2):265-284. 23. Gaynon PS, Angiolillo AL, Carroll WL, et al. Long-term results of the children s cancer group studies for childhood acute lymphoblastic leukemia 1983-2002: a Children s Oncology Group Report. Leukemia. 2010;24(2):285-297. 24. Conter V, Arico M, Basso G, et al. Long-term results of the Italian Association of Pediatric Hematology and Oncology (AIEOP) Studies 82, 87, 88, 91 and 95 for childhood acute lymphoblastic leukemia. Leukemia. 2010;24(2):255-264. 25. Fronkova E, Mejstrikova E, Avigad S, et al. Minimal residual disease (MRD) analysis in the non-mrd-based ALL IC-BFM 2002 protocol for childhood ALL: is it possible to avoid MRD testing? Leukemia. 2008;22(5):989-997. 26. Volejnikova J, Mejstrikova E, Valova T, et al. Minimal residual disease in peripheral blood at day 15 identifies a subgroup of childhood B-cell precursor acute lymphoblastic leukemia with superior prognosis. Haematologica. 2011;96:1815-1821. 27. Gaipa G, Cazzaniga G, Valsecchi MG, et al. Time pointdependent concordance of flow cytometry and RQ-PCR in minimal residual disease detection in childhood acute lymphoblastic leukemia [published online ahead of print May 11, 2012]. Haematologica. doi:10.3324/haematol.2011.06042. 28. 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;107(3): 1116-1123. 29. 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;115(16):3206-3214. 30. Schrappe M, Valsecchi MG, Bartram CR, et al. Late MRD response determines relapse risk overall and in subsets of childhood T-cell ALL: results of the AIEOP-BFM-ALL 2000 study. Blood. 2011;118(8):2077-2084. 31. Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol. 2009;10(2):147-156. 32. Zhou J, Goldwasser MA, Li A, et al. Quantitative analysis of minimal residual disease predicts relapse in children with B-lineage acute lymphoblastic leukemia in DFCI ALL Consortium Protocol 95-01. Blood. 2007;110(5):1607-1611. 33. Nyvold C, Madsen HO, Ryder LP, et al. Precise quantification Hematology 2012 141

of minimal residual disease at day 29 allows identification of children with acute lymphoblastic leukemia and an excellent outcome. Blood. 2002;99(4):1253-1258. 34. Bader P, Kreyenberg H, Henze GH, et al. Prognostic value of minimal residual disease quantification before allogeneic stemcell transplantation in relapsed childhood acute lymphoblastic leukemia: the ALL-REZ BFM Study Group. J Clin Oncol. 2009;27(3):377-384. 35. Leung W, Pui CH, Coustan-Smith E, et al. Detectable minimal residual disease before hematopoietic cell transplantation is prognostic but does not preclude cure for children with very-high-risk leukemia. Blood. 2012;120(2):468-472. 36. Gokbuget N, Kneba M, Raff T, et al. Adults with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies. Blood. 2012;120(9):1868-1876. 37. Schrappe M, Valsecchi MV, Bartram CR, et al. Late MRD response determines relapse risk overall and in subsets of childhood T-cell ALL: results of the AIEOP-BFM-ALL 2000 study. Blood. 2011;118(8):2077-2084. 38. Eckert C, Flohr T, Koehler R, et al. Very early/early relapses of acute lymphoblastic leukemia show unexpected changes of clonal markers and high heterogeneity in response to initial and relapse treatment. Leukemia. 2011;25(8):1305-1313. 39. Szczepanski T, van der Velden VH, Waanders E, et al. Late recurrence of childhood T-cell acute lymphoblastic leukemia frequently represents a second leukemia rather than a relapse: first evidence for genetic predisposition. J Clin Oncol. 2011; 29(12):1643-1649. 40. Guggemos A, Eckert C, Szczepanski T, et al. Assessment of clonal stability of minimal residual disease targets between 1st and 2nd relapse of childhood precursor B-cell acute lymphoblastic leukemia. Haematologica. 2003;88(7):737-746. 41. Mullighan CG, Phillips LA, Su X, et al. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science. 2008;322(5906):1377-1380. 42. Yang JJ, Cheng C, Yang W, et al. Genome-wide interrogation of germline genetic variation associated with treatment response in childhood acute lymphoblastic leukemia. JAMA. 2009;301(4):393-403. 43. Handgretinger R, Zugmaier G, Henze G, Kreyenberg H, Lang P, von Stackelberg A. Complete remission after blinatumomabinduced donor T-cell activation in three pediatric patients with post-transplant relapsed acute lymphoblastic leukemia. Leukemia. 2011;25(1):181-184. 44. Topp MS, Kufer P, Gokbuget N, et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapyrefractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol. 2011;29(18): 2493-2498. 45. Parker C, Waters R, Leighton C, et al. Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial. Lancet. 2010;376(9757):2009-2017. 46. Cario G, Stanulla M, Fine BM, et al. Distinct gene expression profiles determine molecular treatment response in childhood acute lymphoblastic leukemia. Blood. 2005;105(2):821-826. 47. Flotho C, Coustan-Smith E, Pei D, et al. Genes contributing to minimal residual disease in childhood acute lymphoblastic leukemia: prognostic significance of CASP8AP2. Blood. 2006; 108(3):1050-1057. 48. Bhojwani D, Kang H, Menezes RX, et al. Gene expression signatures predictive of early response and outcome in high-risk childhood acute lymphoblastic leukemia: A Children s Oncology Group Study [corrected]. J Clin Oncol. 2008;26(27):4376-4384. 49. Mullighan CG, Su X, Zhang J, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med. 2009;360(5):470-480. 50. Chen IM, Harvey RC, Mullighan CG, et al. Outcome modeling with CRLF2, IKZF1, JAK, and minimal residual disease in pediatric acute lymphoblastic leukemia: a Children s Oncology Group study. Blood. 2012;119(15):3512-3522. 51. Cario G, Zimmermann M, Romey R, et al. Presence of the P2RY8-CRLF2 rearrangement is associated with a poor prognosis in non-high-risk precursor B-cell acute lymphoblastic leukemia in children treated according to the ALL-BFM 2000 protocol. Blood. 2010;115(26):5393-5397. 52. Davies SM, Borowitz MJ, Rosner GL, et al. Pharmacogenetics of minimal residual disease response in children with B- precursor acute lymphoblastic leukemia: a report from the Children s Oncology Group. Blood. 2008;111(6):2984-2990. 142 American Society of Hematology