Anda di halaman 1dari 12

Cytometry Part B (Clinical Cytometry) 84B:359369 (2013)

Review Article

Detection of Minimal Residual Disease in


Pediatric Acute Lymphoblastic Leukemia
Giuseppe Gaipa,1* Giuseppe Basso,2 Andrea Biondi,1 and Dario Campana3
1

M. Tettamanti Research Center, Pediatric Clinic University of Milano Bicocca, Monza, Italy
Laboratorio di Oncoematologia Pediatrica, Department of Pediatrics, University of Padova, Padova, Italy
3
Department of Pediatrics, National University of Singapore, Singapore

Minimal residual disease (MRD) is a powerful predictor of the overall response to treatment in childhood
acute lymphoblastic leukemia (ALL). The most reliable and validated methods to assess MRD in ALL are
flow cytometric (FCM) analysis of leukemia-associated immunophenotypes and polymerase chain reaction (PCR) amplification of antigenreceptor gene rearrangements. Results of studies correlating MRD
with clinical outcome and technical improvements in FCM technology support the implementation of
MRD studies by this method in the clinic. Gene expression profiling of leukemic and normal cells has
identified new MRD markers, which can be incorporated to improve the applicability and sensitivity of
FCM-based MRD monitoring. The combined use of MRD and emerging information on genetic lesions of
C 2013 International Clinical
ALL offers the possibility of further refining risk-assignment approaches. V
Cytometry Society
Key words: childhood; acute lymphoblastic leukemia; minimal residual disease; flow cytometry; RQ-PCR

How to cite this article: Gaipa G, Basso G, Biondi A, Campana D. Detection of Minimal Residual Disease in
Pediatric Acute Lymphoblastic Leukemia. Cytometry Part B 2013; 84B: 359369.

INTRODUCTION
Most children with acute lymphoblastic leukemia
(ALL) achieve complete remission with current treatment regimens but leukemia relapse, the main cause of
treatment failure, still occurs in a significant proportion
of patients (1). Periodic assessment of treatment
response provides an indication of the sensitivity of leukemic cells to chemotherapy and of the overall treatment effectiveness. Historically, treatment response has
been monitored by morphological examination of bone
marrow aspirates, a task which is a fundamental component of the clinical care of patients with ALL (2).
Because this approach has a limited sensitivity and specificity, it can result in failure to detect residual leukemic
cells, potentially leading to under-treatment and an
increase risk of relapse. Conversely, misclassification of
normal cells as ALL blasts may result in over-treatment
and an increase in the risk of treatment-related
morbidity.
Over the last 23 decades, much effort has been
made to develop accurate and sensitive methods that
could determine response to treatment more precisely
than morphology, and to detect residual leukemia persisting in patients considered to be in morphologic

C 2013 International Clinical Cytometry Society


V

remission, i.e. minimal residual disease (MRD) (3,4).


Many studies have demonstrated that MRD is a powerful
predictor of clinical outcome in childhood ALL (512),
supporting the use of a more stringent definition of
hematological remission based on MRD assays rather
than morphology (13).
METHODOLOGIES FOR MRD DETECTION
To be informative, MRD assays for ALL should allow
to the detection of one leukemic cell among 10,000 normal cells or more. They should also reliably discriminate
leukemic and normal cells, and allow a timely output of
Correspondence to: Giuseppe Gaipa, M. Tettamanti Research Center, University of Milano Bicocca, Via Pergolesi, 33 Monza 20052,
Italy. E-mail g.gaipa@hsgerardo.org
Grant sponsor: Comitato M.L.Verga and Fondazione Tettamanti, Fondazione Citt
a della Speranza, Fondazione Cariparo, Associazione Gian
Franco Lupo, Associazione Italiana per la Ricerca sul Cancro, Fondazione Cariplo, and Ministero dellIstruzione, Universita e Ricerca
(MIUR).
Received 31 October 2012; Revised 2 April 2013; Accepted 23
March 2013
Published online 26 June 2013 in Wiley Online Library (wileyonline
library. com).
DOI: 10.1002/cyto.b.21101

360

GAIPA ET AL.

Table 1
Main Features and Applicability of the Methods Widely Used for Study MRD in Pediatric ALL
Method

Target

Applicability

Sensitivity
24

Flow cytometry

Leukemia-associated
immunophenotypes

95% or more

10

Real-time
quantitative
(RQ) PCR

Fusion gene
transcripts

40%

1024

Ig/TCR gene
rearrangements

9095%

1025

the results. Finally, they should be robust enough to support consistent MRD estimates when applied in different
laboratories (1418). Currently, the most reliable methods to study MRD in ALL are flow cytometric (FCM)
analysis of leukemia-associated immunophenotypes (i.e.,
aberrant phenotypes expressed in leukemic cells but
not in normal bone marrow or peripheral blood cells),
and polymerase chain reaction (PCR) amplification of
antigen receptor gene rearrangements (2,1423); gene
fusion transcripts can also be used as PCR targets in
some cases (24). The main features of these techniques
are outlined in Table 1.
A BRIEF HISTORY OF THE DEVELOPMENT OF MULTICOLOR
FLOW CYTOMETRY IMMUNOPHENOTYPING
Antigen expression is assessed by quantification of the
signal emitted by fluorochrome-conjugated-specific monoclonal antibodies (MoAb). Fluorescein excited by an argon
laser was initially used in the Herzenberg laboratory, followed by two-color fluorescence detection (25).
Then, additional antibody-conjugated fluorochromes
were developed to increase the number of colors
(26). From the 1980s, the capacity of several molecules
to absorb and transfer energy to other fluorescent molecules was exploited to produce tandem dyes such as PETexas Red, PE-Cy5, PE-Cy5.5, PE Cy7, and Alexa (27,28),
thus many laboratories were soon routinely performing
four-color experiments. Further advances rapidly led to
the possibility of studying a great number of functionally
distinct lymphocyte populations in human peripheral
blood by the simultaneous detection of six to eight different cell surface antigens (29). The introduction of violet lasers (405 nm) and of nanocrystals (called quantum
dots) or organic polymers capable of conducting electrons (30) led to the current polychromatic flow cytometry (PFC) capable of analyzing up to 18 colors in a single cell (31).
The development of PFC required a parallel advancement in hardware, software, and reagents. Engineering
advances in optics and signal processing (digital electronics) are areas of active development (26,32). Hardware improvements are supported by software development aiming at rapidly processing the bulk of raw data,

Advantages

Disadvantages

Widely applicable; rapid;


accurate and direct
MRD quantification
Rapid; reflects the
leukemic clone size
Widely applicable; highly
sensitive; accurate
MRD quantitation

Operator-dependent; need for


further standardization
Uncertain quantitation of MRD;
not completely standardized;
potential false-positive results
due to RNA degradation.
Laborious and costly;need for
experienced personnel and
standardization; oligoclonality
and clonal evolution may
produce false-negative results

to correct fluorescence spill-over between dyes (i.e.,


compensation), as well as to display and analyze
n-parameters and store them. Different software packages are now available, including FACSDivaTM [Becton
Dickinson], KaluzaTM [Beckman Coulter], FlowJoTM
[www.treestar.com], CytobankTM [www.cytobank.org],
SPICETM [http://exon.niaid.nih.gov/spice/], SamSPECTRALTM [R-package], FLAMETM, SPADETM [www.cytospade.org], FlowTypeTM [R-package], FlowCAPTM [flowcap.flowsite.org], GemStoneTM [www.vsh.com], InfinicyteTM
[www.infinicyt.com].
The increased number of fluorochromes did impact
on the quality of PFC analysis. For example, Bj
orklund
and collaborators used eight-color PFC to better define
the CD341 subsets in comparison with the previous
three-color method and proposed this approach to study
MRD in acute myeloid leukemia (AML) (33). Rare circulating progenitor cells of hematopoietic and/or endothelial origin could be detected with high sensitivity in
human peripheral blood or cord blood by using PFC
(34,35).The ability to measure more markers simultaneously can allow full antibody panels to be fitted into one
to two tubes, and the collection of meaningful information even from small samples. To this end, a 10-color
panel has been reported to detect leukemia and lymphoma cells in several types of small cell samples, a
development that could significantly improve diagnostic
procedures (36). Finally, the simultaneous analysis of
several parameters can also help to determine with
greater accuracy the presence of functionally important
cell subsets, such as those with stem cell features
(32). These advantages notwithstanding, several studies
demonstrated that MRD monitored by four-color FCM
can still be clinically informative (11,37,38).
PFC may introduce the potential for technical problems. For example, setting up multicolor FCM for MRD
analysis is not simply achieved by adding new reagents
to existing panels but requires careful testing, validation,
and quality control (26,39,40). In this process, antibody
clones must be carefully selected, as several clones recognizing the same marker may differ significantly in
their reactivity (e.g., CD34, a key marker for MRD detection) (41).

Cytometry Part B: Clinical Cytometry

MRD IN PEDIATRIC ALL

SOME CONSIDERATIONS REGARDING FLUOROCHROMES


Desirable fluorochromes for leukemia immunophenotyping are as follows: (i) biologically inert, (ii) have high
cell-associated fluorescence intensities, (iii) exhibit little
spectral overlap amongst each other, and (iv) can be easily conjugated to antibodies (26).
PE-conjugates are generally brighter than FITCconjugates, while PE-Cy5 and APC reagents show an
intermediate level of brightness. Other available fluorochromes and tandems include APC-Cy5, PerCP-Cy5,
APC-Cy7, and PerCP-Cy7. In general, the brightest fluorochrome should be used for detection of the weakest
marker. A surface molecule whose expression is
unknown is initially best studied with an antibody conjugate that uses one of the brightest fluorochrome (e.g.,
PE, PE-Cy5, or APC). The choice of fluorochrome for the
other markers depends on instrument characteristics. In
general, PerCP-conjugated MoAbs have some limitations
related to their low sensitivity and the restricted number
of commercially available MoAbs conjugated to this fluorochrome. PE-Cy5 tandem conjugates are widely available but have some nonspecific binding, especially to
monocytic cells (42). Another issue to be considered is
the possible spectral overlap between the various dyes
in the staining combinations. Although hardware and
software compensation can partially solve overlapping
problems, a careful choice of reagents can minimize the
requirement for such procedures. APC reagents are
quite sensitive; the combined use of APC with PE-Cy7
allows limited fluorescence compensation and reduces
nonspecific binding. However, the introduction of new
generation fluorochromes may also implicate compensation complications and attention should be drawn to
obtain optimal instrument setting; for example DiGiuseppe and collaborators calculated compensation values
for AmCyan and other fluorochromes using the beadsbased set provided by the manufacturers protocol, and
using unstained and singly stained lymphocytes as compensation controls. Improved compensation of the
violet-excited dye (i.e. AmCyan) was obtained when
cells rather than the commercial beads were used as
compensation controls (43).
IMMUNOLOGICAL MARKERS OF MRD IN ALL: EVOLUTION
AND DISCOVERY
It has been known for more than three decades that
ALL cells have immunophenotypic features that distinguish them from normal bone marrow and peripheral
blood cells. Initial studies were performed by two-color
immunofluorescence microscopy (2). With the advent of
FCM, it soon became clear that the features of this technology, such as the potential for detecting multiple
parameters in a large number of cells while quantifying
antigen expression, were particularly well suited to
MRD studies.
Two conceptually separate approaches are applied to
the selection of markers for MRD studies in T-lineage
and B-lineage ALL. In T-lineage ALL, the main task

Cytometry Part B: Clinical Cytometry

361

consists of identifying cells with immunophenotypic features of immature T cells. Normal thymocytes may
express identical cell marker profiles but they are normally confined to the thymic microenvironment. Hence,
the detection of cells with immunophenotypic features
of immature T cells (e.g., simultaneous expression of terminal deoxynucleotidase transferase and CD3) in the
blood or marrow of patients with T-lineage ALL indicates
MRD. In B-lineage ALL, simple identification of immature
B cells is generally not sufficient (see below for an
exception to this rule) because normal immature B cells
are a constituent of the normal bone marrow cell population and can be particularly abundant in bone marrow
samples recovering from chemotherapy (44,45). Therefore, B-lineage ALL cells must be distinguished from normal B-cell progenitors by virtue of cell markers that
become aberrantly expressed during leukemogenesis
(4649). However, if the follow-up bone marrow specimen does not contain lymphoid progenitors (a scenario
that presents itself during the first 23 weeks of
remission-induction therapy) then the mere presence of
cells with features of immature B cells can be an indication of residual disease. Indeed, in bone marrow samples collected from 11 patients with T-lineage ALL after
2 weeks of remission induction therapy, CD191 cells
expressing CD10 and/or CD34 were either undetectable
or represented <0.01% of bone marrow mononuclear
cells in all patients (50). Moreover, MRD levels measured
by these three markers correlated well with those measured by more complex FCM analysis or PCR amplification of antigen-receptor genes (50). This exception notwithstanding, all other MRD studies in B-lineage ALL
require the identification of the aberrant immunophenotypes expressed by each leukemia, which is typically
undertaken at diagnosis. Some investigators studied
MRD in follow-up samples without prior knowledge of
the immunophenotype of the leukemic cells, a process
that involves searching for aberrant profiles by using a
very large panel of antibody combinations (51). This
approach may be the only option when patients are
referred from another center for continuation therapy or
transplant, but a negative MRD result would be clearly
difficult to interpret.
Aberrant immunophenotypes can be found in the
majority of patients. For example, in the St Jude Total
XV study, 482 of 492 (98%) enrolled patients with newly
diagnosed ALL had leukemic cells at diagnosis that
expressed leukemia-associated immunophenotypes that
would allow the detection of MRD at the 0.01% level at
any time point during therapy, including at times of vigorous bone marrow cell recovery (52). However, the differences between ALL and normal cells are often subtle
and discerning them takes much experience. Logically,
the greater the immunophenotypic difference between
the two cell types the easier it should be to recognize
ALL cells. In this context, the identification of new
markers to be incorporated into MRD panels should be
beneficial by expanding the multidimensional approach
to MRD data analysis and taking advantage to the

362

GAIPA ET AL.

FIG. 1. Markers that can be used to monitor MRD in B-lineage ALL.


Black dots correspond to markers that denote immature B-lymphoid
cells but cannot distinguish between normal and leukemic cells. The
remaining dots indicate markers that can be used to define leukemiaassociated immunophenotypes: markers identified in early studies
(1st generation) are shown in the gray dots; those identified more
recently through gene expression array comparisons (62) are in white.
Examples of antibody combinations and their fluorochromes which can
be used to study MRD are shown in the table.

capabilities of contemporary instruments which have at


least doubled the number of parameters that could be
traditionally measured.
The task of identifying new markers of ALL was traditionally based on observations made by directly comparing the immunophenotype of normal and leukemic cells
(48,53). This approach was also used in more recent
studies which identified CD81 (54), CD49f (55), and
CD11b (56) as potentially useful markers. Another strategy is to compare the gene expression profiles of normal and leukemic CD191 CD101 cells and search for
differentially expressed genes. In an early study, this
approach led to the identification of CD58 as a useful
marker (57), a finding which was confirmed by several
other studies (5861). More recently, a study compared
genome-wide gene expression of leukemic cells from
270 patients with B-lineage ALL to that of purified normal CD191CD101 B cell progenitors from four healthy
donors (62). Among the differentially expressed genes,
30 were selected for further studies by FCM in 200 Blineage ALL and 61 non-leukemic bone marrow samples
(62). Twenty-two of the 30 markers (CD44, BCL2,
HSPB1, CD73, CD24, CD123, CD72, CD86, CD200,
CD79b, CD164, CD304, CD97, CD102, CD99, CD300a,
CD130, PBX1, CTNNA1, ITGB7, CD69, CD49f) were differentially expressed in about 80% of ALL cases (62).
Notably, some of the markers, such as CD304 (63) and
CD123 (64), were independently found to be differentially expressed in ALL by other investigators. Studies
using paired diagnosis-relapse samples demonstrated the
stability of the new markers, which have allowed MRD
monitoring by FCM in more than 150 consecutive
patients with a potential sensitivity of 1 in 100,000 (62).
Figure 1 summarizes first and second generation markers
for MRD studies in ALL; examples of the immunophenotypic differences between normal and leukemic cells
detected by some of the new markers are shown in
Figure 2.

STANDARDIZATION OF FLOW CYTOMETRIC METHODS FOR


MULTICENTRIC MRD STUDIES
Early efforts in standardization of flow cytometric protocols for MRD detection in ALL were initially carried
out in the context of BIOMED-1 Concerted Action by
six different European laboratories (47). In that study,
characterization of normal bone marrow-derived precursor-B cell subpopulations by three-color immunophenotyping provided templates as a frame of reference for
discriminating normal from malignant precursor-B cells.
An extensive standardization of four-color FCM MRD
protocol for multicentric applications was performed by
four laboratories within the AIEOP-BFM-ALL 2000 study
(23). This addressed several technical issues including
methodological harmonization regarding sample preparation, reagent selection and instrument set-up, intra and
inter-laboratory quality monitoring, as well as specific
personnel training focused on FCM data interpretation.
Despite differences in cytometers and software usage,
blinded inter-laboratory tests of list-mode data interpretation gave highly concordant results (intraclass correlation coefficient [ICC], 0.979). The reproducibility in
inter-laboratory sample exchange was also assessed:
among 63 follow-up samples, exchanged between two
centers, the positive/negative concordance as well as
the quantitative reproducibility of MRD-values, were
very high (23). Concordance between all four centers in
the analysis of artificial mixtures of normal and leukemic
cells was also high (ICC: 0.98): of 164 MRD-values available, there were five false values each among 113 positive and 51 negative values, resulting in a sensitivity of
95.6% and a specificity of 90.2% (23). The application of
such a standardized multicentric FCM protocol to study
MRD has yielded significant correlations with treatment
outcome in pediatric ALL. Thus, Ratei et al. (65) showed
that flow cytometric blast reduction rates on day 8 and
day 15 had a predictive impact on the remission status
after induction therapy in the AIEOP-BFM ALL 2000 protocol, and Basso et al. demonstrated feasibility and

FIG. 2. Immunophenotypic differences between normal and leukemic


cells. Flow cytometric dot plots show the immunophenotype of gated
CD191 CD101 cells from normal bone marrow (BM) samples (top
row) and from diagnostic samples of B-lineage ALL (bottom row) as
defined by some of the recently identified markers. Dashed lines
enclose areas were most normal CD191 CD101 cells are located.

Cytometry Part B: Clinical Cytometry

363

MRD IN PEDIATRIC ALL

robustness of this approach in refining MRD-based stratification with measurements on day 15 (37). The feasibility of standardized multicentric MRD assessment has
also been shown by others groups (66,67). Six-eightcolor-FCM MRD techniques are currently employed for
leukemia immunophenotyping. Advances in algorithm
and methods for analyzing complex data should further
facilitate MRD data analysis and foster standardized analytical approaches (68). The recently reported efforts of
the EuroFlow group should contribute to standardize
leukemia immunophenotyping in a major way, and also
have an impact on MRD protocols (69).
MOLECULAR METHODS FOR MRD DETECTION IN ALL
AntigenReceptor Gene Rearrangements
Somatic rearrangement of Ig and TcR gene loci occurs
during early differentiation of any B and T cell, by joining the germline variable (V), diversity (D), and joining
(J) gene segments (3). By this process, each lymphocyte
gets a specific combination of V-(D-) J segments that
encodes the variable domains of Ig or TcR molecules.
The uniqueness of each rearrangement further depends
on random insertion and deletion of nucleotides at the
junction sites of V, (D), and J gene segments, making
the junctional regions of Ig and TcR genes as
fingerprint-like sequences. This combined sequence
constitutes a specific signature of each lymphocyte.
Because of the clonal origin of the neoplasm, each
malignant lymphoid disease will represent the expansion
of a clonal population with a specific Ig/TcR signature
(70).
Virtually all B-lineage ALL patients have rearranged
Immunoglobulin heavy chain (IgH) genes (70). In addition, rearrangements of the Ig Kappa deleting element
(Kde) occur at a relatively high frequency (approximately 60%) (71). Cross-lineage incomplete TCRD rearrangements occur in more than 40% of all patients (40%
Vd2-Dd3, 19% Dd2-Dd3 and 13% showed both) (72).
Complete TCRD rearrangements (Vd-Jd) are very rare in
B-lineage ALL. Detection of TCRG rearrangements
occurs in more than 50% of the patients with B-lineage
ALL (72). Cross-lineage TCRB recombinations occur in a
small percentage (1520%) of patients with B-lineage
ALL. The success rate of detecting appropriate MRDPCR targets is lower in T-ALL compared to patients with
B-lineage ALL. The addition of TCRB gene rearrangements to MRD-PCR target identification increases the
availability of targets in T-ALL. Moreover, the junctional
regions of (complete) TCRD rearrangements, similar to
IGH in B-lineage ALL, frequently include extensive Nnucleotide insertions, thus enabling the design of highly
specific ASO primers. In contrast, the junctions of
TCRG and incomplete IGH rearrangements are commonly smaller resulting in a significantly lower ratio of
sensitive targets (about 75%).
For MRD studies, the specific rearrangements must be
sequenced in each case upfront. Ig/TcR gene rearrangements can be used as universal PCR-MRD targets. In

Cytometry Part B: Clinical Cytometry

combination with fluorescent probes, allele-specific oligonucleotide (ASO) primers can be designed complementary to the patient- and tumor-specific junctional
region sequence of each target. A large standardization,
quality control and guidelines on RQ-PCR analysis and
interpretation have been assessed within the EuroMRD group (previous European Study Group for MRD
detection in ALL, ESG-MRD ALL) (16). More recently,
the possibility to use high-throughput sequencing (HTS)
for disease-specific IGH sequence quantification was
proposed and it is likely that new and fast technologies
for rapid sequencing will broaden the availability of
MRD quantification B and T cell malignancies (73).
However, the predictive value of MRD was also demonstrated in childhood T-lineage ALL (74,75), infant ALL
(76), in mature-B ALL (77), as well as in patients with
intra chromosomal amplification of chromosome 21
(78). It is likely that in a near future PCR studies in ALL
MRD will benefit of more rapid and sensitive
approaches such as next-generation sequencing technologies (73,79).
Gene Fusions
Somatic chromosome aberrations can be used as PCR
targets for MRD detection because they are tumorspecific and stable during the disease course. The most
widely used for MRD detection are gene fusions, such as
BCR-ABL1, MLL-AFF1, TCF3-PBX1, and ETV6-RUNX1,
resulting in the expression of aberrant mRNA transcripts. Recurrent abnormalities suitable for routine
MRD studies in clinical samples are present in approximately 40% of children with ALL. Among more recently
identified abnormalities that could, in principle, be used
for monitoring MRD, are gene fusions involving CRLF2
(e.g., IGH@-CRLF2 and P2RY8-CRLF2). A consensus on
the reagents and minimal requirements for MRD monitoring by RQ-PCR of fusion transcripts has been reached
by several European laboratories collaborating on a
Europe Against Cancer program (80). Currently the use
of RQ-PCR of fusion genes is limited to the Ph1 ALL
(81).
CLINICAL SIGNIFICANCE OF MRD IN PEDIATRIC ALL
Many studies have demonstrated the prognostic significance of MRD levels in pediatric ALL. Such studies have
focused on newly diagnosed patients (58,8289),
patients with first relapse leukemia (9,9092), and
patients undergoing allogeneic hematopoietic stem cell
transplant (HSCT) (9396). They unanimously showed
that MRD is a powerful predictor of outcome, leading to
the adoption of MRD as a critical risk-assignment criteria
(12,52).
Recent studies have provided strong evidence that
MRD can also be informative among patients with specific subtypes of ALL. Thus, Conter et al. (12) found that
MRD was predictive of outcome among patients with
the TEL-AML1, high hyperdiploidy, or BCR-ABL1 molecular abnormalities. Schrappe et al. (75) found that MRD
measurements on days 33 and 78 of treatment allowed

364

GAIPA ET AL.

the delineation of distinct prognostic subgroups among


patients with T-ALL. Bowman et al. (97) reported that
MRD measurements in peripheral blood on day 8 and in
the bone marrow on day 29 predicted outcome in children with B-lineage ALL receiving augmented therapy
because of high-risk presenting clinical features. Finally,
van der Velden et al. (76) found that MRD levels at the
end of induction and consolidation could predict outcome in infants with ALL.
One of the main clinical uses of MRD monitoring is to
measure the kinetics of initial leukemia cytoreduction,
which, in turn, is a predictor of response overall. To this
end, it is becoming clear that studies of MRD at early
time points during therapy can be extremely powerful.
Early studies demonstrated that MRD testing after 23
weeks of remission induction therapy could predict outcome (7,98). More recently, Sutton et al. (85) measured
MRD during treatment in 108 patients and found that
measurements on day 15 were strong predictors of
relapse risk. A study of the Associazione Italiana Ematologia Oncologia Pediatrica assessed MRD in bone marrow samples from 830 children with ALL collected on
day 15 of treatment (37). MRD levels 10%, seen in
11% of patients, were associated with a 5-year relapse
rate of 47.2%, 0.1% to < 10% levels (47%) a rate of
17.5% and <0.1% levels (42%) with a rate of 7.5%. In a
multivariate analysis, MRD was the strongest prognostic
factor when compared to presenting features, and
retained prognostic significance when MRD detected by
PCR at later time points was added to the model. MRD
studies at these early time points are technically less
challenging that those are later time points because of
the paucity of normal lymphoid progenitors present in
bone marrow (these cells are eliminated by remission
induction chemotherapy) (50). Hence, the demonstration of their clinical utility is of particular practical
importance.
FLOW CYTOMETRY VERSUS PCR: CONCORDANCE,
CORRELATIONS, AND COMPLEMENTARY USE
Several studies have compared MRD estimates
obtained by FCM and PCR (99104). Most of these
reports considered a cut-off of 0.01% for positivity, and
showed a degree of concordance of >90%. Most of discrepancies were due to FCM-negative/PCR positive
results occurring at the lowest level of MRD. Discrepancies (either qualitative or quantitative) may be explained
by different factors affecting either PCR or FCM, such as
quality of clonal PCR-markers (22), nonspecific amplification of normal DNA (105), oligoclonality and/or clonal
evolution (106111), age-related (47) or therapy-related
bone marrow B-cell precursor regeneration status
(112,113), immunophenotypic changes between diagnosis and relapse (114,115) or drug-induced immunophenotypic modulation (59). This latter phenomenon has
been described during early phases of treatment in BFMoriented protocols, when steroids components are
largely predominant, in particular down modulation of
CD10 and CD34, and up modulation of CD19, CD20,

CD45RA, CD11a was observed, whereas CD58 was stable (59). Despite this, the accuracy of MRD detection
was not negatively affected, as residual modulated blasts
were located outside regions of residual normal B cells
from the same follow-up sample. Finally, Dworzak et al.
demonstrated that drug-induced up modulation of CD10
and CD34 were reversible after stop of steroid containing chemotherapy (116); therefore, the leukemiaassociated immunophenotypes seen at diagnosis remain
an important frame of reference even though transient
changes may occur during the early phase of treatment.
However, prevalence and inter-individual variations of B
cell precursor subpopulations have been described so
far in pediatric bone marrow during and after chemotherapy (112,113).
Table 2 summarizes the results obtained in some of
the studies comparing FCM and PCR in the simultaneous detection of MRD. Variations in sample collection
may affect the concordance between the two methods.
Both FCM and PCR samples should be collected from
the same bone marrow aspirate; MRD levels in a second
bone marrow aspiration through the same needle could
be lower because of hemodilution (117). The use of
mononucleated cells from the same density centrifugation separation should increase the concordance
between the two methods. However, several studies
have compared PCR and FCM in MRD detection by
using mononuclear cells and total nucleated cells,
respectively (see Table 2). Among these, Th
orn et al.
(118) adopted a virtual Ficoll gate when they analyzed
FCM data files in order to mimic the procedure of density gradient centrifugation. The calculated fraction of
mononuclear cells was determined by applying a mathematical formula and then used to extrapolate the MRD
values that would be found in Ficoll-separated cells. One
study found that the impact of using either monucleated
or total nucleated cells on the concordance of FCM
with PCR-results was minimal, and that concordance
rates may be influenced by the time point at which
MRD is measured (104).
In the final analysis, the choice of the MRD method to
be used is primarily dependent on the expertise and
resources available, and on clinical trial design. To this
end, for risk assignment strategies relying on early evaluations of treatment response, i.e. day 15, FCM should be
preferable as current PCR methods to detect antigen
receptor genes require a longer (typically 3 weeks)
preparation time.
CONCLUSIONS
MRD studies have now been incorporated into clinical
protocols where they are used to assess risk of relapse
and rationalize treatment decisions. MRD studies at early
time points can identify patients with good treatment
response, who may be curable with less intensity therapy. Current clinical trials are testing this possibility
(50). Another attractive application of MRD is in the
context of trials of novel agents or treatment regimens,
where MRD measurements can provide a rapid

Cytometry Part B: Clinical Cytometry

365

97.1%
93.3%
90.0%
80.0%
0.01%
0.01%
0.1%
0.01%
4
3
3/4
4
10 210
1024
1023
1024

10
1024
1023
1024

89.0%
0.01%
3/4

25
23

102321025
1024

1
10
4
3

Cytometry Part B: Clinical Cytometry

Kerst et al. (102)


Ryan et al.(103)
Th
orn et al. (118)
Gaipa et al. (104)

22
Malec et al. (101)

MNC, mononucleated cells; TNC, total nucleated cells.

MNC
MNC
FCM: TNC, PCR: MNC
FCM: TNC, PCR: MNC
105
151
726
2,701
30
29
228
1115

2
FCM: TNC, PCR: MNC
71

1
3
FCM: TNC, PCR: MNC
MNC
69
227

AIEOP-BFM-2000
St.Jude Total XIII;
XIV; XV.
NOPHO 92 and
NOPHO 2000
Not reported
UK-ALL 97/99
NOPHO 2000
AIEOP-BFM 2000

69
1375

FCM: TNC, PCR: MNC


16
ALL-BFM-95

mayer,
Dworzak and Gru
Leukemia and
Lymphoma 2003
Veltroni et al. (58)
Neale et al. (100)

84

ACKNOWLEDGMENTS
We thank Ms. Elaine Coustan-Smith for helpful discussions and providing Figure 2, Dr. Barbara Buldini, Dr.
Andrea Zangrando, and Mr. Oscar Maglia for their support in this study. We also thank the I-BFM-ALL-FCMMRD-Study Group.

24

95.6%
96.7%
4
4
1024
1025
Not reported
1024

Any level
0.01%

83.0%
3
Not reported
Not reported

0.01%

78.0%
3
102321025
FCM: TNC, PCR: MNC
89
NOPHO 92

23

assessment of efficacy. As a caveat to this use, a recent


trial demonstrated a significantly better EFS for ALL
relapsed patients treated in induction with mitoxantrone
vs idarubicine but no apparent difference in MRD at the
end of induction (119). Whether a single MRD positive
finding during or off treatment is sufficient to define
relapse and initiate retrieval therapy, and whether it
would be of benefit to initiate treatment at such stage
instead of waiting for overt relapse remains unclear. The
finding of MRD, however, should trigger frequent
monitoring.
In sum, MRD studies are a key tool towards the development of personalized treatment for children with ALL
and FCM has significantly contributed to widen the
applicability of this valuable test.

LITERATURE CITED

Malec et al. (101)

Study

6 (early and
late remission
induction)
>2

102321025

Any level

Concordance
rate
Cut-off
for
positivity
FCM
colors
(n)
PCR
sensitivity
FCM
sensitivity
N of time
points
studied
Source of starting
material
No. of
samples
No. of
patients
studied
Clinical
protocol

Table 2
Studies Directly Comparing MRD Detection by Flow Cytometry (FCM) and Polymerase Chain Reaction (PCR) Amplification of Antigen Receptor Genes

MRD IN PEDIATRIC ALL

1. Pui C-H, Evans WE. Acute lymphoblastic leukemia. N Engl J Med


2006;354:166167.
2. Bradstock KF, Janossy G, Tidman N, Papageorgiou ES, Prentice HG,
Willoughby M, Hoffbrand AV. Immunological monitoring of residual disease in treated thymic acute lymphoblastic leukaemia. Leuk
Res 1981;5:301309.
3. Szczepa
nski T, Orf~ao A, van der Velden VH, San Miguel JF, van
Dongen JJ. Minimal residual disease in leukaemia patients. Lancet
Oncol 2001;2:409417.
4. Campana D. Determination of minimal residual disease in leukaemia patients. Br J Haematol 2003;121:823838.
5. Brisco MJ, Condon J, Hughes E, Neoh SH, Sykes PJ, Seshadri R,
Toogood I, Waters K, Tauro G, Ekert H. Outcome prediction in
childhood acute lymphoblastic leukaemia by molecular quantification of residual disease at the end of induction. Lancet 1994;343:
196200.
6. van Dongen JJ, Seriu T, Panzer-Gr
umayer ER, Biondi A, PongersWillemse MJ, Corral L, Stolz F, Schrappe M, Masera G, Kamps WA,
et al. Prognostic value of minimal residual disease in acute lymphoblastic leukaemia in childhood. Lancet 1998;352:17311738.
7. Coustan-Smith E, Sancho J, Behm FG, Hancock ML, Razzouk BI,
Ribeiro RC, Rivera GK, Rubnitz JE, Sandlund JT, Pui CH, et al.
Prognostic importance of measuring early clearance of leukemic
cells by flow cytometry in childhood acute lymphoblastic leukemia. Blood 2002;100:5258.
8. Cav
e H, van der Werff ten Bosch J, Suciu S, Guidal C, Waterkeyn
C, Otten J, Bakkus M, Thielemans K, Grandchamp B, Vilmer E.
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;339:591598.
9. Eckert C, Biondi A, Seeger K, Cazzaniga G, Hartmann R,
Beyermann B, Pogodda M, Proba J, Henze G. Prognostic value of
minimal residual disease in relapsed childhood acute lymphoblastic
leukaemia. Lancet 2001;358:12391241.
10. Dworzak MN, Fr
oschl G, Printz D, Mann G, P
otschger U,
M
uhlegger N, Fritsch G, Gadner H; Austrian Berlin-FrankfurtM
unster Study Group. Prognostic significance and modalities of
flow cytometric minimal residual disease detection in childhood
acute lymphoblastic leukemia. Blood 2002; 99:19521958.
11. Borowitz MJ, Devidas M, Hunger SP, Bowman WP, Carroll AJ,
Carroll WL, Linda S, Martin PL, Pullen DJ, Viswanatha D, et al;
Childrens Oncology Group. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: A Childrens Oncology Group
study. Blood 2008;111:54775485.

366

GAIPA ET AL.

12. Conter V, Bartram CR, Valsecchi MG, Schrauder A, PanzerGr


umayer R, M
oricke A, Aric
o M, Zimmermann M, Mann G, De
Rossi G, 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;115:32063214.
13. Pui CH, Campana D, Evans WE. Childhood acute lymphoblastic
leukaemia. Current status and future perspectives. Lancet Oncol
2001;2:597607.
14. Cazzaniga G, Gaipa G, Rossi V, Biondi A. Minimal residual disease
as a surrogate marker for risk assignment to ALL patients. Rev Clin
Exp Hematol 2003;7:292323.
15. van der Velden VH, Hochhaus A, Cazzaniga G, Szczepanski T,
Gabert J, van Dongen JJ. Detection of minimal residual disease in
hematologic malignancies by real-time quantitative PCR: Principles,
approaches, and laboratory aspects. Leukemia 2003;17:10131034.
16. van der Velden VH, Cazzaniga G, Schrauder A, Hancock J, Bader P,
Panzer-Grumayer ER, Flohr T, Sutton R, Cave H, Madsen HO,
Cayuela JM, et al; European Study Group on MRD detection in ALL
(ESG-MRD-ALL). Analysis of minimal residual disease by Ig/TCR
gene rearrangements: Guidelines for interpretation of real-time
quantitative PCR data. Leukemia 2007;21:604611.
17. Campana D. Status of minimal residual disease testing in childhood
haematological malignancies. Br J Haematol 2008;143:481489.
18. Flohr T, Schrauder A, Cazzaniga G, Panzer-Gr
umayer R, van der
Velden V, Fischer S, Stanulla M, Basso G, Niggli FK, Schafer BW, et al;
International BFM Study Group (I-BFM-SG). Minimal residual diseasedirected 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;22:771782.
19. Foroni L, Harrison CJ, Hoffbrand AV, Potter MN. Investigation of
minimal residual disease in childhood and adult acute lymphoblastic leukaemia by molecular analysis. Br J Haematol. 1999;105:724.
20. Pongers-Willemse MJ, Seriu T, Stolz F, dAniello E, Gameiro P, Pisa
P, Gonzalez M, Bartram CR, Panzer-Gr
umayer ER, Biondi A, et al.
Primers and protocols for standardized detection of minimal residual disease in acute lymphoblastic leukemia using immunoglobulin
and T cell receptor gene rearrangements and TAL1 deletions as
PCR targets: Report of the BIOMED-1 CONCERTED ACTION:
Investigation of minimal residual disease in acute leukemia. Leukemia 1999;13:110118.
21. Campana D, Coustan-Smith E. Advances in the immunological
monitoring of childhood acute lymphoblastic leukaemia. Best Pract
Res Clin Haematol 2002;15:119.
22. van der Velden VH, Panzer-Gr
umayer ER, Cazzaniga G, Flohr T,
Sutton R, Schrauder A, Basso G, Schrappe M, Wijkhuijs JM, Konrad
M, et al. Optimization of PCR-based minimal residual disease diagnostics for childhood acute lymphoblastic leukemia in a multicenter setting. Leukemia 2007;21:706713.
23. Dworzak MN, Gaipa G, Ratei R, Veltroni M, Schumich A, Maglia O,
Karawajew L, Benetello A, P
otschger U, Husak Z, et al. Standardization of flow cytometric minimal residual disease evaluation in
acute lymphoblastic leukemia: Multicentric assessment is feasible.
Cytometry B Clin Cytom 2008;74B:331340.
24. Cazzaniga G, Valsecchi MG, Gaipa G, Conter V, Biondi A. Defining
the correct role of minimal residual disease tests in the management
of acute lymphoblastic leukaemia. Br J Haematol 2011;155:4552.
25. Bendall SC, Nolan GP, Roederer M, Chattopadhyay PK. A deep profilers guide to cytometry. Trends Immunol 2012;33:32332.
26. Baumgarth N, Roederer M. A practical approach to multicolor flow
cytometry for immunophenotyping. J Immunol Methods 2000;243:
7797.
27. Lansdorp PM, Smith C, Safford M, Terstappen LW, Thomas TE. Single laser three color immunofluorescence staining procedures
based on energy transfer between phycoerythrin and cyanine 5.
Cytometry 1991;12:723730.
28. Roederer M, Kantor AB, Parks DR, Herzenberg LA. Cy7PE and
Cy7APC: Bright new probes for immunofluorescence. Cytometry
1996; 24:191197.
29. De Rosa SC, Brenchley J, Roederer M. Beyond six colors: A new
era in flow cytometry. Nature Med 2003;9:112117.
30. Service RF. Getting a charge out of plastics. Science 2000;290:
425427.
31. Chattopadhyay PK, Price DA, Harper TF, Betts MR, Yu J, Gostick E,
Perfetto SP, Goepfert P, Koup RA, De Rosa SC, et al. Quantum dot
semiconductor nanocrystals for immunophenotyping by polychromatic flow cytometry. Nature Med 2006;12:972977.

32. Preffer F, Dombkowski D. Advances in complex multiparameter


flow cytometry technology: Applications in stem cell research.
Cytometry B Clin Cytom 2009;76B:295314.
33. Bj
orklund E, Gruber A, Mazur J, Martensson A, Hansson M, Porwit
A. CD341 cell subpopulations detected by 8-color flow cytometry
in bone marrow and in peripheral blood stem cell collections:
Application for MRD detection in leukemia patients. Int J Hematol
2009;90:292302.
34. Kamnesh RP, Mund JA, Johnson C, Vik TA, Ingram DA, Case J. Polychromatic flow cytometry identifies novel subsets of circulating
cells with angiogenic potential in pediatric solid tumors. Cytometry B Clin Cytom 2011;80B:335338.
35. Mund JA, Estes ML, Yoder MC, Ingram DA Jr, Case J. Flow cytometric identification and functional characterization of immature and
mature circulating endothelial cells. Arterioscler Thromb Vasc Biol
2012;32:10451053.
36. Preijers FW, Huys E, Moshaver B. OMIP-010: A new 10-color monoclonal antibody panel for polychromatic immunophenotyping of
small hematopoietic cell samples. Cytometry A 2012;81A:453455.
37. Basso G, Veltroni M, Valsecchi MG, Dworzak MN, Ratei R, Silvestri
D, Benetello A, Buldini B, Maglia O, Masera G, 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;27:51685174.
38. Lorenzana R, Coustan-Smith E, Antillon F, Ribeiro RC, Campana D.
Simple methods for the rapid exchange of flow cytometric data
between remote centers. Leukemia 2000; 14:336337.
39. Perfetto SP, Ambrozak D, Nguyen R, Chattopadhyay P, Roederer M.
Quality assurance for polychromatic flow cytometry. Nat Protoc
2006;1:15221530.
40. Perfetto SP, Ambrozak D, Nguyen R, Chattopadhyay PK, Roederer
M. Quality assurance for polychromatic flow cytometry using a
suite of calibration beads. Nat Protoc 2012;7:20672079.
41. Lanza F, Moretti S, Castagnari B, Montanelli F, Latorraca A, Ferrari
L, Bardi A, Dominici M, Campioni D, Dabusti M, et al. Assessment
of distribution of CD34 epitope classes in fresh and cryopreserved
peripheral blood progenitor cells and acute myeloid leukemic
blasts. Haematologica 1999;84:969977.
42. Rothe G, Schmitz G. Consensus protocol for the flow cytometric
immunophenotyping of hematopoietic malignancies. Working
Group on Flow Cytometry and Image Analysis. Leukemia 1996;10:
877895.
43. DiGiuseppe JA, Cardinali J. Improved compensation of the fluorochrome AmCyan using cellular controls. Cytometry B Clin Cytom
2011;80B:191194.
44. Rimsza LM, Larson RS, Winter SS, Foucar K, Chong YY, Garner
KW, Leith CP. Benign hematogone-rich lymphoid proliferations can
be distinguished from B-lineage acute lymphoblastic leukemia by
integration of morphology, immunophenotype, adhesion molecule
expression, and architectural features. Am J Clin Pathol 2000;114:
6675.
45. McKenna RW, Washington LT, Aquino DB, Picker LJ, Kroft SH.
Immunophenotypic analysis of hematogones (B-lymphocyte precursors) in 662 consecutive bone marrow specimens by 4-color
flow cytometry. Blood 2001;98:24982507.
46. Hurwitz CA, Loken MR, Graham ML, Karp JE, Borowitz MJ, Pullen
DJ, Civin CI. Asynchronous antigen expression in B lineage acute
lymphoblastic leukemia. Blood 1988;72:299307.
47. L
ucio P, Parreira A, van den Beemd MW, van Lochem EG, van
Wering ER, Baars E, Porwit-MacDonald A, Bjorklund E, Gaipa G,
Biondi A, et al. Flow cytometric analysis of normal B cell differentiation: A frame of reference for the detection of minimal residual
disease in precursor-B- ALL. Leukemia 1999;13:419427.
48. Campana D, Coustan-Smith E. Detection of minimal residual disease in acute leukemia by flow cytometry. Cytometry 1999;38:
139152.
49. Ciudad J, San Miguel JF, L
opez-Berges MC, Vidriales B, Valverde B,
Ocqueteau M, Mateos G, Caballero MD, Hernandez J, Moro MJ,
et al. Prognostic value of immunophenotypic detection of minimal
residual disease in acute lymphoblastic leukemia. J Clin Oncol
1998;16:37743781.
50. Coustan-Smith E, Ribeiro RC, Stow P, Zhou Y, Pui CH, Rivera GK,
Pedrosa F, Campana D. A simplified flow cytometric assay identifies
children with acute lymphoblastic leukemia who have a superior
clinical outcome. Blood 2006;108:97102.
51. Wells DA, Sale GE, Shulman HM, Myerson D, Bryant EM, Gooley T,
Loken MR. Multidimensional flow cytometry of marrow can differentiate leukemic from normal lymphoblasts and myeloblasts after

Cytometry Part B: Clinical Cytometry

MRD IN PEDIATRIC ALL

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

63.

64.

65.

66.

67.

chemotherapy and bone marrow transplantation. Am J Clin Pathol


1998;110:8494.
Pui CH, Campana D, Pei D, Bowman WP, Sandlund JT, Kaste SC,
Ribeiro RC, Rubnitz JE, Raimondi SC, Onciu M, et al. Treating
childhood acute lymphoblastic leukemia without cranial irradiation. N Engl J Med 2009;360:27302741.
Lucio P, Gaipa G, van Lochem EG, van Wering ER, PorwitMacDonald A, Faria T, Bjorklund E, Biondi A, van den Beemd MW,
Baars E, et al; BIOMED-I. BIOMED-I concerted action report: Flow
cytometric immunophenotyping of precursor B-ALL with standardized triple-stainings. BIOMED-1 Concerted Action Investigation of
Minimal Residual Disease in Acute Leukemia: International Standardization and Clinical Evaluation. Leukemia 2001;15:11851192.
Muzzafar T, Medeiros LJ, Wang SA, Brahmandam A, Thomas DA,
Jorgensen JL. Aberrant underexpression of CD81 in precursor Bcell acute lymphoblastic leukemia: Utility in detection of minimal
residual disease by flow cytometry. Am.J Clin Pathol 2009;132:
692698.
DiGiuseppe JA, Fuller SG, Borowitz MJ. Overexpression of CD49f
in precursor B-cell acute lymphoblastic leukemia: Potential usefulness in minimal residual disease detection. Cytometry B Clin
Cytom 2009;76B:150155.
Rhein P, Mitlohner R, Basso G, Gaipa G, Dworzak MN, KirschnerSchwabe R, Hagemeier C, Stanulla M, Schrappe M, Ludwig WD,
et al. CD11b is a therapy resistance- and minimal residual diseasespecific marker in precursor B-cell acute lymphoblastic leukemia.
Blood 2010;115:37633771.
Chen JS, Coustan-Smith E, Suzuki T, Neale GA, Mihara K, Pui CH,
Campana D. Identification of novel markers for monitoring minimal residual disease in acute lymphoblastic leukemia. Blood 2001;
97:21152120.
Veltroni M, De Zen L, Sanzari MC, Maglia O, Dworzak MN, Ratei R,
Biondi A, Basso G, Gaipa G; I-BFM-ALL-FCM-MRD-Study Group.
Expression of CD58 in normal, regenerating and leukemic bone
marrow B cells: Implications for the detection of minimal residual
disease in acute lymphocytic leukemia. Haematologica 2003;88:
12451252.
Gaipa G, Basso G, Maglia O, Leoni V, Faini A, Cazzaniga G, Bugarin
C, Veltroni M, Michelotto B, Ratei R, Coliva T, et al; I-BFM-ALLFCM-MRD Study Group. Drug-induced immunophenotypic modulation in childhood ALL: Implications for minimal residual disease
detection. Leukemia 2005;19:4956.
Lee RV, Braylan RC, Rimsza LM. CD58 expression decreases as
nonmalignant B cells mature in bone marrow and is frequently
overexpressed in adult and pediatric precursor B-cell acute lymphoblastic leukemia. Am.J Clin Pathol 2005;123:119124.
Mejstrkova E, Fronkova E, Kalina T, Omelka M, Batinic D,
Dubravcic K, Pospsilova K, Vaskova M, Luria D, Cheng SH, et al.
Detection of residual B precursor lymphoblastic leukemia by uniform gating flow cytometry. Pediatr. Blood Cancer 2010;54:6270.
Coustan-Smith E, Song G, Clark C, Key L, Liu P, Mehrpooya M,
Stow P, Su X, Shurtleff S, Pui CH, et al. New markers for minimal
residual disease detection in acute lymphoblastic leukemia. Blood
2011;117:62676276.
Solly F, Angelot F, Garand R, Ferrand C, Seille`s E, Schillinger F,
Decobecq A, Billot M, Larosa F, Plouvier E, et al. CD304 is preferentially expressed on a subset of B-lineage acute lymphoblastic leukemia and represents a novel marker for minimal residual disease
detection by flow cytometry. Cytometry A 2012;81A:1724.
Djokic M, Bj
orklund E, Blennow E, Mazur J, S
oderhall S, Porwit A.
Overexpression of CD123 correlates with the hyperdiploid genotype in acute lymphoblastic leukemia. Haematologica 2009;94:
10161019.
Ratei R, Basso G, Dworzak M, Gaipa G, Veltroni M, Rhein P, Biondi
A, Schrappe M, Ludwig WD, Karawajew L; AIEOP-BFM-FCM-MRDStudy Group. Monitoring treatment response of childhood precursor B-cell acute lymphoblastic leukemia in the AIEOP-BFM-ALL
2000 protocol with multiparameter flow cytometry: Predictive
impact of early blast reduction on the remission status after induction. Leukemia 2009;23:528534.
Bj
orklund E, Matinlauri I, Tierens A, Axelsson S, Forestier E,
Jacobsson S, Ahlberg AJ, Kauric G, Mantymaa P, Osnes L, et al. Quality control of flow cytometry data analysis for evaluation of minimal
residual disease in bone marrow from acute leukemia patients during treatment. J Pediatr Hematol Oncol 2009;31:406415.
Irving J, Jesson J, Virgo P, Case M, Minto L, Eyre L, Noel N,
Johansson U, Macey M, Knotts L, et al; UKALL Flow MRD Group;
UK MRD steering Group. Establishment and validation of a

Cytometry Part B: Clinical Cytometry

68.

69.

70.

71.

72.

73.

74.

75.

76.

77.

78.

79.

80.

81.

82.

367

standard protocol for the detection of minimal residual disease in


B lineage childhood acute lymphoblastic leukemia by flow cytometry in a multi-center setting. Haematologica 2009;94:870874.
Pierzchalski A, Mittag A, Tarnok A. Methods in cell biology, Introduction A: Recent advances in cytometry instrumentation, probes,
and methodsReview. Methods Cell Biol 2011;102:121.
van Dongen JJ, Lhermitte L, B
ottcher S, Almeida J, van der Velden
VH, Flores-Montero J, Rawstron A, Asnafi V, L
ecrevisse Q, Lucio P,
et al. EuroFlow antibody panels for standardized n-dimensional
flow cytometric immunophenotyping of normal, reactive and
malignant leukocytes. Leukemia 2012;26:19081975.
van Dongen JJ and Wolvers-Tettero IL. Analysis of immunoglobulin
and T cell receptor genes. II. Possibilities and limitations in the
diagnosis and management of lymphoproliferative diseases and
related disorders. Clin Chim Acta 1991;198:93174.
Beishuizen A, de Bruijn MA, Pongers-Willemse MJ, Verhoeven MA,
van Wering ER, Hahlen K, Breit TM, de Bruin-Versteeg S, Hooijkaas
H, van Dongen JJ. Heterogeneity in junctional regions of immunoglobulin kappa deleting element rearrangements in B cell leukemias: A new molecular target for detection of minimal residual disease. Leukemia 1997;11:22002207.
van der Velden VH, Szczepanski T, Wijkhuijs JM, Hart PG,
Hoogeveen PG, Hop WC, van Wering ER, van Dongen JJ. Agerelated patterns of immunoglobulin and T-cell receptor gene rearrangements in precursor-B-ALL: Implications for detection of minimal residual disease. Leukemia 2003;17:18341844.
Wu D, Sherwood A, Fromm JR, Winter SS, Dunsmore KP, Loh ML,
Greisman HA, Sabath DE, Wood BL, Robins H. High-throughput
sequencing detects minimal residual disease in acute T lymphoblastic leukemia. Sci Transl Med 2012;4:134ra63.
Willemse MJ, Seriu T, Hettinger K, dAniello E, Hop WC, PanzerGr
umayer ER, Biondi A, Schrappe M, Kamps WA, Masera G, et al.
Detection of minimal residual disease identifies differences in treatment response between T-ALL and precursor B-ALL. Blood 2002;
99:43864393.
Schrappe M, Valsecchi MG, Bartram CR, Schrauder A, PanzerGr
umayer R, M
oricke A, Parasole R, Zimmermann M, Dworzak M,
Buldini B, et al. Late MRD response determines relapse risk overall
and in subsets of childhood T-cell ALL: Results of the AIEOP-BFMALL 2000 study. Blood 2011;118:20772084.
Van der Velden VH, Corral L, Valsecchi MG, Jansen MW, De
Lorenzo P, Cazzaniga G, Panzer-Gr
umayer ER, Schrappe M,
Schrauder A, Meyer C, et al; Interfant-99 Study Group. Prognostic
significance of minimal residual disease in infants with acute lymphoblastic leukemia treated within the Interfant-99 protocol. Leukemia 2009;23:10731079.
Mussolin L, Pillon M, Bonato P, Leszl A, Franceschetto G, Di Meglio
A, dAmore ES, Sainati L, Rosolen A. Cytogenetic analysis of pediatric anaplastic large cell lymphoma. Pediatr Blood Cancer 2010;55:
446451.
Attarbaschi A, Mann G, Panzer-Gr
umayer R, R
ottgers S, Steiner M,
K
onig M, Csinady E, Dworzak MN, Seidel M, Janousek D, et al.
Minimal residual disease values discriminate between low and
high relapse risk in children with B-cell precursor acute lymphoblastic leukemia and an intrachromosomal amplification of chromosome 21: The Austrian and German acute lymphoblastic leukemia Berlin-Frankfurt-Munster (ALL-BFM) trials. J Clinical Oncol
2008;26:30463050.
Faham M, Zheng J, Moorhead M, Carlton VE, Stow P, CoustanSmith E, Pui CH, Campana D. Deep sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia.
Blood 2012;120:51735180.
Wassmann B, Pfeifer H, Stadler M, Bornha
user M, Bug G,
Scheuring UJ, Br
uck P, Stelljes M, Schwerdtfeger R, Basara N, et al.
Early molecular response to posttransplantation imatinib determines outcome in MRD1 Philadelphia-positive acute lymphoblastic leukemia (Ph1 ALL). Blood 2005;106:458463.
Pane F, Cimino G, Izzo B, Camera A, Vitale A, Quintarelli C, Picardi
M, Specchia G, Mancini M, Cuneo A, et al; GIMEMA group. (2005)
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;19:628635.
Coustan-Smith E, Behm FG, Sanchez J, Boyett JM, Hancock ML,
Raimondi SC, Rubnitz JE, Rivera GK, Sandlund JT, Pui CH, et al.
Immunological detection of minimal residual disease in children
with acute lymphoblastic leukaemia. Lancet 1998;351:550554.

368

GAIPA ET AL.

83. Coustan-Smith E, Sancho J, Hancock ML, Boyett JM, Behm FG,


Raimondi SC, Sandlund JT, Rivera GK, Rubnitz JE, Ribeiro RC,
et al. Clinical importance of minimal residual disease in childhood
acute lymphoblastic leukemia. Blood 2000;96:26912696.
84. Zhou J, Goldwasser MA, Li A, Dahlberg SE, Neuberg D, Wang H,
Dalton V, McBride KD, Sallan SE, Silverman LB, et al; Dana-Farber
Cancer Institute ALL Consortium. 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;110:16071611.
85. Sutton R, Venn NC, Tolisano J, Bahar AY, Giles JE, Ashton LJ, Teague
L, Rigutto G, Waters K, Marshall GM, et al; Australian and New Zealand Childrens Oncology Group. Clinical significance of minimal
residual disease at day 15 and at the end of therapy in childhood
acute lymphoblastic leukaemia. Br J Haematol 2009;146:292299.
86. Stow P, Key L, Chen X, Pan Q, Neale GA, Coustan-Smith E,
Mullighan CG, Zhou Y, Pui CH, Campana D. Clinical significance
of low levels of minimal residual disease at the end of remission
induction therapy in childhood acute lymphoblastic leukemia.
Blood 2010;115:46574663.
87. Yamaji K, Okamoto T, Yokota S, Watanabe A, Horikoshi Y, Asami
K, Kikuta A, Hyakuna N, Saikawa Y, Ueyama J, et al; Japanese
Childhood Cancer Leukemia Study Group. Minimal residual
disease-based augmented therapy in childhood acute lymphoblastic
leukemia: A report from the Japanese Childhood Cancer and Leukemia Study Group. Pediatr Blood Cancer 2010;55:12871295.
88. Katsibardi K, Moschovi MA, Braoudaki M, Papadhimitriou SI,
Papathanasiou C, Tzortzatou-Stathopoulou F. Sequential monitoring
of minimal residual disease in acute lymphoblastic leukemia: 7-year
experience in a pediatric hematology/oncology unit. Leuk Lymphoma 2010; 51:846852.
89. Meleshko AN, Savva NN, Fedasenka UU, Romancova AS, Krasko
OV, Eckert C, von Stackelberg A, Aleinikova OV. Prognostic value
of MRD-dynamics in childhood acute lymphoblastic leukemia
treated according to the MB-2002/2008 protocols. Leuk Res 2011;
35:13121320.
90. Coustan-Smith E, Gajjar A, Hijiya N, Razzouk BI, Ribeiro RC, Rivera
GK, Rubnitz JE, Sandlund JT, Andreansky M, Hancock ML, et al.
Clinical significance of minimal residual disease in childhood acute
lymphoblastic leukemia after first relapse. Leukemia 2004;18:
499504.
91. Paganin M, Zecca M, Fabbri G, Polato K, Biondi A, Rizzari C,
Locatelli F, Basso G. Minimal residual disease is an important predictive factor of outcome in children with relapsed high-risk
acute lymphoblastic leukemia. Leukemia 2008;22:21932200.
92. Raetz EA, Borowitz MJ, Devidas M, Linda SB, Hunger SP, Winick
NJ, Camitta BM, Gaynon PS, Carroll WL. Reinduction platform for
children with first marrow relapse in acute lymphoblastic lymphoma. J Clin Oncol 2008;26:39713978.
93. Krejci O, van der Velden VH, Bader P, Kreyenberg H, Goulden N,
Hancock J, Schilham MW, Lankester A, Rev
esz T, Klingebiel T, van
Dongen JJ. Level of minimal residual disease prior to haematopoietic stem cell transplantation predicts prognosis in paediatric
patients with acute lymphoblastic leukaemia: A report of the PreBMT MRD Study Group. Bone Marrow Transplant 2003;32:
849851.
94. Bader P, Kreyenberg H, Henze GH, Eckert C, Reising M, Willasch
A, Barth A, Borkhardt A, Peters C, Handgretinger R, et al; ALL-REZ
BFM Study Group. Prognostic value of minimal residual disease
quantification before allogeneic stem-cell transplantation in
relapsed childhood acute lymphoblastic leukemia: The ALL-REZ
BFM Study Group. J Clin Oncol 2009;27:377384.
95. Leung W, Campana D, Yang J, Pei D, Coustan-Smith E, Gan K,
Rubnitz JE, Sandlund JT, Ribeiro RC, Srinivasan A, et al. High success rate of hematopoietic cell transplantation regardless of donor
source in children with very high-risk leukemia. Blood 2011;118:
223230.
96. Zhao XS, Liu YR, Zhu HH, Xu LP, Liu DH, Liu KY, Huang XJ. Monitoring MRD with flow cytometry: An effective method to predict
relapse for ALL patients after allogeneic hematopoietic stem cell
transplantation. Ann Hematol 2012;91:183192.
97. Bowman WP, Larsen EL, Devidas M, Linda SB, Blach L, Carroll AJ,
Carroll WL, Pullen DJ, Shuster J, Willman CL, et al. Augmented
therapy improves outcome for pediatric high risk acute lymphocytic leukemia: Results of Childrens Oncology Group trial P9906.
Pediatr Blood Cancer 2011;57:569577.
98. Panzer-Grumayer ER, Schneider M, Panzer S, Fasching K, Gadner
H. Rapid molecular response during early induction chemotherapy

99.

100.

101.

102.

103.

104.

105.

106.

107.

108.

109.

110.

111.

112.

predicts a good outcome in childhood acute lymphoblastic leukemia. Blood 2000;95790794.


Neale GA, Coustan-Smith E, Pan Q, Chen X, Gruhn B, Stow P,
Behm FG, Pui CH, Campana D. Tandem application of flow cytometry and polymerase chain reaction for comprehensive detection
of minimal residual disease in childhood acute lymphoblastic leukemia. Leukemia 1999;13:12211226.
Neale GA, Coustan-Smith E, Stow P, Pan Q, Chen X, Pui CH,
Campana D. Comparative analysis of flow cytometry and polymerase chain reaction for the detection of minimal residual disease in
childhood acute lymphoblastic leukemia. Leukemia 2004;18:
934938.
Malec M, van der Velden VH, Bj
orklund E, Wijkhuijs JM, S
oderhall
S, Mazur J, Bj
orkholm M, Porwit-MacDonald A. 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;18:16301636.
Kerst G, Kreyenberg H, Roth C, Well C, Dietz K, Coustan-Smith E,
Campana D, Koscielniak E, Niemeyer C, Schlegel PG, 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;128:774782.
Ryan J, Quinn F, Meunier A, Boublikova L, Crampe M, Tewari P,
OMarcaigh A, Stallings R, Neat M, OMeara A, et al. Minimal residual disease detection in childhood acute lymphoblastic leukaemia
patients at multiple time points reveals high levels of concordance
between molecular and immunophenotypic approaches. Br J Haematol 2009;144:107115.
Gaipa G, Cazzaniga G, Valsecchi MG, Panzer-Gr
umayer R, Buldini
B, Silvestri D, Karawajew L, Maglia O, Ratei R, Benetello A, et al.
Time point-dependent concordance of flow cytometry and RQPCR in minimal residual disease detection in childhood acute lymphoblastic leukemia. Haematologica 2012;97:15821593.
van der Velden VH, Cazzaniga G, Schrauder A, Hancock J, Bader P,
Panzer-Grumayer ER, Flohr T, Sutton R, Cave H, Madsen HO, et al;
European Study Group on MRD detection in ALL (ESG-MRD-ALL).
Analysis of minimal residual disease by Ig/TCR gene rearrangements: Guidelines for interpretation of real-time quantitative PCR
data. Leukemia 2007;21:604611.
Steward CG, Goulden NJ, Katz F, Baines D, Martin PG, Langlands
K, Potter MN, Chessells JM, Oakhill A. A polymerase chain reaction study of the stability of Ig heavy-chain and T-cell receptor
delta gene rearrangements between presentation and relapse of
childhood B-lineage acute lymphoblastic leukemia. Blood 1994;83:
13551362.
Beishuizen A, Verhoeven MA, Van Wering ER, Hahlen K, Hooijkaas
H, van Dongen JJ. Analysis of Ig and T-cell receptor genes in 40
childhood acute lymphoblastic leukemias at diagnosis and subsequent relapse: Implications for the detection of minimal residual
disease by polymerase chain reaction analysis. Blood 1994;83:
22382247.
Baruchel A, Cayuela JM, MacIntyre E, Berger R, Sigaux F. Assessment of clonal evolution at Ig/TCR loci in acute lymphoblastic leukaemia by single-strand conformation polymorphism studies and
highly resolutive PCR derived methods: Implication for a general
strategy of minimal residual disease detection. Br J Haematol 1995;
90:8593.
Steenbergen EJ, Verhagen OJ, van den Berg H, van Leeuwen EF,
Behrendt H, Slater RR, von dem Borne AE, van der Schoot CE.
Rearrangement status of the malignant cell determines type of secondary IgH rearrangement (V-replacement or V to DJ joining) in
childhood B precursor acute lymphoblastic leukemia. Leukemia
1997;11:12581265.
Szczepanski T, Willemse MJ, Brinkhof B, Van Wering ER, van der
BM, van Dongen JJ. Comparative analysis of Ig and TCR gene rearrangements at diagnosis and at relapse of childhood precursor-BALL provides improved strategies for selection of stable PCR targets for monitoring of minimal residual disease. Blood 2002;99:
23152323.
Li A, Zhou J, Zuckerman D, Rue M, Dalton V, Lyons C, Silverman
LB, Sallan SE, Gribben JG. Sequence analysis of clonal immunoglobulin and T-cell receptor gene rearrangements in children with
acute lymphoblastic leukemia at diagnosis and at relapse: Implications for pathogenesis and for the clinical utility of PCR-based
methods of minimal residual disease detection. Blood 2003;102:
45204526.
Dworzak MN, Fritsch G, Fleischer C, Printz D, Fr
oschl G,
Buchinger P, Mann G, Gadner H. Multiparameter phenotype

Cytometry Part B: Clinical Cytometry

MRD IN PEDIATRIC ALL

113.

114.

115.

116.

mapping of normal and post-chemotherapy B lymphopoiesis in


pediatric bone marrow. Leukemia 1997;11:12661273.
van Wering ER, van der Linden-Schrever BE, Szczepa
nski T,
Willemse MJ, Baars EA, van Wijngaarde-Schmitz HM, Kamps WA,
van Dongen JJ. Regenerating normal B-cell precursors during and
after treatment of acute lymphoblastic leukaemia: Implications for
monitoring of minimal residual disease. Br J Haematol 2000;110:
139146.
van Wering ER, Beishuizen A, Roeffen ET, van der Linden-Schrever
BE, Verhoeven MA, Hahlen K, Hooijkaas H, van Dongen JJ. Immunophenotypic changes between diagnosis and relapse in childhood
acute lymphoblastic leukemia. Leukemia 1995;9:152333.
Borowitz MJ, Pullen DJ, Winick N, Martin PL, Bowman WP,
Camitta B. Comparison of diagnostic and relapse flow cytometry
phenotypes in childhood acute lymphoblastic leukemia: Implications for residual disease detection: A report from the childrens
oncology group. Cytometry B Clin Cytom 2005;68B:1824.
Dworzak MN, Gaipa G, Schumich A, Maglia O, Ratei R, Veltroni M,
Husak Z, Basso G, Karawajew L, Gadner H, et al. Modulation of
antigen expression in B-cell precursor acute lymphoblastic

Cytometry Part B: Clinical Cytometry

369

leukemia during induction therapy is partly transient: Evidence for


a drug-induced regulatory phenomenon. Results of the AIEOPBFM-ALL-FLOW-MRD-Study Group. Cytometry B Clin Cytom 2010;
78B:147153.
117. Coustan-Smith E, Sancho J, Hancock ML, Razzouk BI, Ribeiro RC,
Rivera GK, Rubnitz JE, Sandlund JT, Pui CH, Campana D. Use of
peripheral blood instead of bone marrow to monitor residual disease in children with acute lymphoblastic leukemia. Blood 2002;
100:23992402.
118. Th
orn I, Forestier E, Botling J, Thuresson B, Wasslavik C,
Bj
orklund E, Li A, Lindstr
om-Eriksson E, Malec M, Gr
onlund E,
et al. Minimal residual disease assessment in childhood acute lymphoblastic leukaemia: A Swedish multi-centre study comparing
real-time polymerase chain reaction and multicolour flow cytometry. Br J Haematol 2011;152:743753.
119. Parker C, Waters R, Leighton C, Hancock J, Sutton R, Moorman AV,
Ancliff P, Morgan M, masurekar A, Goulden N, et al. Effect of
mitoxantrone on outcome of children with first relapse of acute
lymphoblastic leukemia (ALL R3): An open-label randomised trial.
The Lancet 2010;376:20092017.

Copyright of Cytometry. Part B is the property of John Wiley & Sons, Inc. and its content
may not be copied or emailed to multiple sites or posted to a listserv without the copyright
holder's express written permission. However, users may print, download, or email articles for
individual use.

Anda mungkin juga menyukai