ArticlePDF AvailableLiterature Review

Why and how to quantify minimal residual disease in acute lymphoblastic leukemia? Leukemia

Authors:

Abstract and Figures

Several studies have demonstrated that monitoring of minimal residual disease (MRD) in childhood and adult acute lymphoblastic leukemia (ALL) significantly correlates with clinical outcome. MRD detection is particularly useful for evaluation of early treatment response and consequently for improved front-line therapy stratification. MRD information is also significant for children undergoing allogeneic hematopoietic stem cell transplantation and those with relapsed ALL. Currently, three highly specific and sensitive methodologies for MRD detection are available, namely multiparameter flow cytometric immunophenotyping, real-time quantitative polymerase chain reaction (RQ-PCR)-based detection of fusion gene transcripts or breakpoints, and RQ-PCR-based detection of clonal immunoglobulin and T-cell receptor gene rearrangements. In this review, characteristics, pitfalls, advantages and disadvantages of each MRD technique are critically discussed. The special emphasis is put on interlaboratory standardization, especially in view of the results obtained within the European collaborative BIOMED-1, BIOMED-2, and Europe Against Cancer projects and recent developments by European Study Group on MRD detection in ALL and EuroFlow Consortium. Standardized MRD techniques form the basis for stratification of patients into the risk groups in new treatment protocols mainly in childhood ALL. Only the results of these studies can answer the question whether MRD-based treatment intervention is associated with improved outcome.
Content may be subject to copyright.
CONCISE REVIEW
Why and how to quantify minimal residual disease in acute lymphoblastic leukemia?
T Szczepan
´ski
1,2
1
Department of Pediatric Hematology and Oncology, Silesian Medical Academy, Zabrze, Poland and
2
Department of
Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
Several studies have demonstrated that monitoring of minimal
residual disease (MRD) in childhood and adult acute lympho-
blastic leukemia (ALL) significantly correlates with clinical
outcome. MRD detection is particularly useful for evaluation
of early treatment response and consequently for improved
front-line therapy stratification. MRD information is also
significant for children undergoing allogeneic hematopoietic
stem cell transplantation and those with relapsed ALL.
Currently, three highly specific and sensitive methodologies
for MRD detection are available, namely multiparameter flow
cytometric immunophenotyping, real-time quantitative poly-
merase chain reaction (RQ-PCR)-based detection of fusion
gene transcripts or breakpoints, and RQ-PCR-based detection
of clonal immunoglobulin and T-cell receptor gene rearrange-
ments. In this review, characteristics, pitfalls, advantages and
disadvantages of each MRD technique are critically discussed.
The special emphasis is put on interlaboratory standardization,
especially in view of the results obtained within the European
collaborative BIOMED-1, BIOMED-2, and Europe Against Can-
cer projects and recent developments by European Study
Group on MRD detection in ALL and EuroFlow Consortium.
Standardized MRD techniques form the basis for stratification
of patients into the risk groups in new treatment protocols
mainly in childhood ALL. Only the results of these studies can
answer the question whether MRD-based treatment interven-
tion is associated with improved outcome.
Leukemia (2007) 21, 622–626. doi:10.1038/sj.leu.2404603;
published online 15 February 2007
Keywords: acute lymphoblastic leukemia; minimal residual
disease; flow cytometric immunophenotyping; real-time polymerase
chain reaction; immunoglobulin and T-cell receptor genes; fusion
genes
The rationale for detection of MRD in ALL
The concept of minimal residual disease (MRD) detection in
acute lymphoblastic leukemias (ALL) is inherently associated
with the progress in treatment of these malignancies.
1
More than
80% of childhood and 35% of adult ALL patients can be cured
with modern chemotherapy supplemented with hematopoietic
stem cell transplantation (HSCT) in high risk patients (reviewed
by Hoelzer et al.
2
). Still, a substantial number of ALL patients
relapse and the prediction of relapse with conventional
prognostic factors such as age, blast count at diagnosis,
immunophenotype at diagnosis, presence of chromosome
aberrations, response to steroid prophase and classical clinical
risk group assignment is far from optimal. Also microarray-based
gene expression profiling could not identify gene signatures,
typically associated with high risk of relapse. Tracing residual
leukemic cells during early phases of treatment provides
prognostic information superior to all known classical prog-
nostic factors. Several prospective studies in childhood ALL
demonstrated that the most relevant information comes from
detection of MRD in bone marrow at the early phases of
treatment, particularly at the end of induction treatment
(reviewed by Szczepan
´ski et al.
3
and Cazzaniga and Biondi
4
).
Children with undetectable MRD at the end of induction have
an excellent prognosis and are good candidates for treatment
de-intensification or at least should not be subjected to further
treatment intensification, particularly not to HSCT.
5–9
In child-
hood ALL, a group of ultrafast responders could be identified
with successful clearance of MRD in bone marrow even within
first 2 weeks of the induction.
10,11
In contrast, children with
high MRD levels at the end of induction treatment are in urgent
need for treatment intensification or even for novel treatment
approaches, particularly when such high MRD levels persist into
the consolidation treatment.
5–9
Children with high-risk primary
ALL and children with relapsed ALL planned for allogeneic
HSCT can also profit from MRD monitoring.
12–17
It is now
generally accepted that one of the prerequisites for successful
allo-HSCT is maximal reduction of MRD before start of the
transplant procedure. Patients with high pretransplant MRD
levels are at very high risk for ALL relapse.
12,14,15
Currently, the
ongoing studies aim at assessment of possible approaches to
lower pretransplant MRD levels and/or attune graft-versus-host
reaction according to post-transplant MRD levels by modifying
the immunosuppression to improve the outcome after HSCT.
Finally, it was recently demonstrated that treatment stratification
of standard risk adult ALL patients can be substantially improved
when including MRD information.
18
In conclusion, based on the above-summarized significant
results, several prospective treatment protocols, mainly in
childhood ALL have been initiated including MRD-based
treatment intervention as an essential part. Only the results of
these studies can answer the question whether MRD-based
treatment intervention is associated with improved outcome.
The methods for quantification of MRD in ALL
Optimal MRD techniques should be characterized by patient
specificity (or at least leukemia specificity), satisfactory sensi-
tivity (at least 10
4
, i.e., one malignant cell among 10 000
normal cells), applicability for the vast majority of patients under
the study, feasibility (easy standardization and rapid collection
of results for clinical application) as well as intralaboratory and
interlaboratory reproducibility. Another prerequisite for reliable
MRD technique is precise quantification of MRD levels. The
stringent criteria described above are for the greater part met by
three approaches, namely multiparameter flow cytometric
immunophenotyping, real-time quantitative polymerase chain
Received 7 January 2007; accepted 10 January 2007; published
online 15 February 2007
Correspondence: Dr T Szczepan
´ski, Department of Pediatric Hema-
tology and Oncology, Silesian Medical Academy, Ul. 3 Maja 13/15,
41-800 Zabrze, Poland.
E-mail: szczep57@poczta.onet.pl
Leukemia (2007) 21, 622–626
&2007 Nature Publishing Group All rights reserved 0887-6924/07 $30.00
www.nature.com/leu
reaction (RQ-PCR)-based detection of fusion gene transcripts or
breakpoints, and RQ-PCR-based detection of clonal immuno-
globulin (Ig) and T-cell receptor (TCR) gene rearrangements
(Table 1).
19–21
Quantification with multiparameter flow cytometric
immunophenotyping
Precise quantification is an inherent feature of flow cytometry,
which measures single cells. Up till now, immunophenotypic
MRD detection in ALL was based on 3–4 color flow cytometry.
This methodology relies on tracing the leukemia-specific
immunophenotypes as the result of cross-lineage antigen
expression, maturational asynchronous expression of antigens,
antigen overexpression, absence of antigen expression, ectopic
antigen expression, and various combinations of above-men-
tioned features bringing the ALL blasts into the ‘empty spaces’
between normal lymphoid differentiation (summarized by
Campana
19
and Szczepan
´ski et al.
20
). Still the detection limit
of this technique is not lower than 10
3
–10
4
, which never-
theless should be sufficient for identifying high-risk ALL patients.
Another pitfall of flow cytometry is the modulation of antigen
expression occurring during the treatment, which can change
the leukemia-specific immunophenotype into a phenotype that
resembles that of normal lymphoid precursors.
22,23
Moreover,
immunophenotypic changes might occur between diagnosis
and relapse.
24
Therefore, following of at least two leukemia-
specific immunophenotypes per patient has been recommended
to prevent false-negative results. It is striking that the most signi-
ficant MRD studies in ALL employing flow cytometric immuno-
phenotyping were based on a single expert laboratory.
7,9
This
raises the issue of interlaboratory standardization of flow
cytometric reagents and procedures, which is essential for
international multicenter treatment protocols. With the advent
of bench top X6-color flow cytometers, higher sensitivity for
MRD monitoring should be achieved, but with the increase of
technical complexity the need for interlaboratory standardiza-
tion becomes even more urgent. In Europe this need for
innovation and standardization is supported by the European
Commission via the EuroFlow Consortium aiming at standardi-
zation of ‘Flow cytometry for fast and sensitive diagnosis and
follow-up of hematological malignancies’.
25
RQ-PCR-based quantification of leukemia-associated
fusion genes
Leukemia-associated fusion genes resulting from chromosomal
translocations are directly linked to leukemogenesis and there-
fore represent very good and stable disease-specific markers.
After numerous single-center studies, the uniform primers and
Table 1 Characteristics of the techniques currently employed for MRD detection in ALL
Flow cytometric
immunophenotyping
PCR analysis of chromosome
aberrations (mainly detection of fusion
gene transcripts)
PCR analysis of Ig/TCR genes
(junctional region specific approach)
Sensitivity 10
3
–10
4
10
4
–10
6
10
4
–10
5
Applicability
Precursor-B-ALL 495% 40–45%
a
90–95%
T-ALL 495% 15–35%
b
90–95%
Advantages Applicable for most patients
Relatively cheap
Rapid: 1–2 days
Relatively easy and cheap
Sensitive and leukemia-specific
Stable target during disease course
Rapid: 2–3 days
Suitable for monitoring of uniform
patient groups (e.g. BCR-ABL+ ALL)
Applicable for virtually all patients, if
IGH,IGK-Kde, TCRG, and TCRD gene
rearrangements are used as targets
Sensitive and patient specific
Rapid during follow-up: 2–3 days
(if junctional region is identified and
if RQ-PCR is used)
Disadvantages Limited sensitivity
Need for preferably two aberrant
immunophenotypes per patient,
because of chance of
immunophenotypic shifts
Drug-induced modulation of the
immunophenotype might influence
the levels of antigenic expression
Useful in only a minority of patients
Not patient-specific cross-
contamination of PCR products might
lead to false-positive results (even at
diagnosis)
Differences in fusion transcript
expression levels between the patients
Stability of fusion gene transcripts
decreases over time
Time consuming at diagnosis:
identification of the junctional regions
and sensitivity testing
Relatively expensive
Need for preferably two PCR targets
per patient, because of chance of
clonal evolution
Two sensitive targets (p10
4
) available
in B80% of patients
Recent developments and
standardization in European
networks
X6-color cytometry gives
promises of increased sensitivity
and specificity; currently under
development in the European
EuroFlow Consortium
Largely standardized thanks to pan-
European collaboration within the
BIOMED-1 (fusion transcript detection)
and EAC project (RQ-PCR)
Methods for identification of fusion
gene breakpoints at the DNA level
provide patient-specific targets
Target identification standardized
within the European BIOMED-1 and
BIOMED-2 networks
RQ-PCR for MRD detection
standardized by the European Study
Group for MRD detection in ALL
(ESG-MRD-ALL)
Abbreviations: B-ALL, B-cell acute lymphoblastic leukemia; EAC, Europe Against Cancer; Ig, immunoglobulin; IGH, immunoglobulin heavy chain
gene; IGK, immunoglobulin kappa light chain gene; MRD, minimal residual disease; RQ-PCR, real-time quantitative polymerase chain reaction;
PCR, polymerase chain reaction; T-ALL, T-cell acute lymphoblastic leukemia; TCR, T-cell receptor; TCRD, T-cell receptor delta gene, TCRG, T-cell
receptor gamma gene.
a
In childhood ALL this particularly concerns t(12;21)(TEL-AML1) and in adult ALL particularly t(9;22)(BCR-ABL).
b
This mainly concerns del(1)(p32 p32) with SIL-TAL1 fusion and t(5;14) with aberrant HOX11L2 expression, together occurring in 25–35% of
childhood T-ALL and in 15-20% of adult ALL (Graux
48
).
Why and how to quantify MRD
T Szczepan
´ski et al
623
Leukemia
protocols for reverse transcription (RT)-PCR for identification of
the most frequent fusion transcripts in ALL: t(1;19)(q23;p13) with
the E2A-PBX1 fusion gene, t(4;11)(q21;q23) with the MLL-AF4
fusion gene, the two main types of t(9;22)(q34;q11) with
BCR-ABL fusion genes, t(12;21)(p13;q22) with the TEL-AML1
fusion gene, and the intrachromosomal microdeletion on 1p32
with the SIL-TAL1 fusion gene, were standardized within the
European BIOMED-1 and Europe Against Cancer networks.
26–28
However, these leukemia specific markers can be identified in
not more than half of ALL patients, which limits their application
as MRD markers for general cohorts of ALL patients. Still, several
translocations are significant prognostic markers and identify
‘homogenous’ ALL subgroups. Both in childhood and adult ALL,
t(9;22) with BCR-ABL fusion gene is associated with a dismal
outcome, although rare cases with a very good sensitivity to
chemotherapy could be also identified in this generally drug-
resistant subgroup of ALL.
29
With the addition of targeted
treatment such as imatinib mesylate to current chemotherapy
regimens for BCR-ABL
þ
ALL, BCR-ABL transcripts have become
the first-choice marker for MRD monitoring by RT-RQ-PCR.
30,31
In contrast, TEL-AML1 fusion gene occurring in approximately
25% of children with ALL identifies a subgroup with a very good
prognosis for which the advantage of MRD monitoring is not
yet fully proven.
The protocols for detection and quantification of fusion gene
transcripts based on RT-RQ-PCR employing TaqMan technology
have been developed thanks to European EAC network.
27
These
MRD assays are characterized by reproducibly high sensitivity
of 10 plasmid molecules or 10
4
RNA cell line dilution for the
majority of the targets. The EAC study also selected appropriate
reference genes to correct variations in RNA quality and
quantity and to calculate the sensitivity of each measurement.
28
These ‘control’ genes are characterized by highly stable
expression in blood and bone marrow of normal and patient
samples.
Because of the high sensitivity of PCR techniques, cross-
contamination of RT-PCR products between patient samples is a
major pitfall in RT-PCR-mediated MRD studies, resulting in up
to 20% of false-positive results.
32
Such cross-contamination is
difficult to recognize, as fusion gene transcripts, although
leukemia-specific, are not patient-specific markers. Also the
levels of fusion gene transcripts can vary significantly between
patients. This is in contrast to PCR products obtained from
genomic breakpoint fusions, which can be identified by use
of patient-specific oligonucleotide probes in RQ-PCR assays.
Unfortunately, in most translocations occurring in ALL the
breakpoints are widely over multiple intronic sequences and
their precise identification is rather complex. Nevertheless, the
group of Marschalek was able to develop a single standardized
approach to identify the vast majority of breakpoint fusions in
the many different chromosome translocations involving the
MLL gene on chromosome 11q23. The DNA breakpoints of MLL
fusions were shown to be highly specific and sensitive RQ-PCR
markers for MRD detection in MLL-rearranged infant and adult
ALL.
33,34
Such DNA-based approaches for precise breakpoint
identification and MRD monitoring should ideally be developed
for the other major ALL subgroups with specific chromosome
translocations.
RQ-PCR-based quantification of junctional regions of Ig
and TCR gene rearrangements
RQ-PCR-based detection and quantification of junctional
regions of clonal Ig and TCR gene rearrangements is by far the
most widely employed strategy of MRD monitoring in ALL.
Although this MRD strategy is the most laborious, expensive and
time consuming, it is the most reproducible approach not only
within the same laboratory but also between different labora-
tories. The junctional regions of clonal Ig and TCR gene
rearrangements are fingerprint-like sequences for each lymphoid
malignancy and can be identified in the vast majority of
ALL patients using the standardized primer sets established
through the European collaboration within the BIOMED-1 and
-2 frameworks.
35–37
The immunobiologic studies identified and
characterized oligoclonality and clonal evolution of Ig/TCR
gene rearrangements between diagnosis and relapse.
38–42
Therefore, it is widely accepted that preferably at least two
Ig/TCR targets should be followed per patient.
Although initial MRD studies employing different semiquan-
titative approaches, revealed significant results,
5,8,43
only the
introduction of RQ-PCR was the major step towards the wider
dissemination of this MRD strategy.
21,44
This issue of Leukemia
contains an excellent summary of Van der Velden et al.
45
on
interpretation of RQ-PCR data of MRD monitoring by Ig/TCR
gene rearrangements based on the experience of 30 MRD-PCR
laboratories forming the European Study Group on MRD
detection in ALL (ESG-MRD-ALL). This international initiative
has resulted in significantly improved standards, definitions
and guidelines for RQ-PCR-based MRD detection via Ig/TCR
gene rearrangements. Based on extensive experimentation, this
study clearly established definitions of MRD positivity and MRD
quantitative range, within which MRD levels can be expressed
in a reproducible way. As shown by earlier reports, this
quantitative range is usually until 10
4
and 10
5
, depending
on the type of gene rearrangement and the size of junctional
region. Another important guideline concerns the discrimination
between low MRD positivity outside the quantitative range and
false positivity resulting from nonspecific background amplifica-
tion. Clearly, some Ig/TCR gene rearrangement junctions
(particularly of TCRG), although unique for a particular patient,
are so similar to rearrangements in the normal Ig/TCR repertoire
that nonspecific amplification might occur from non-leukemic
lymphocytes at low or even moderate levels. Therefore, the
sensitivity of this MRD approach cannot easily be further
improved. Realizing this, the studies demonstrating frequent
MRD positivity in remission samples of children with ALL at the
levels of 10
7
should be interpreted with extreme caution.
Finally, the report contains additional criteria for interpreta-
tion of RQ-PCR data, which take into account whether the
clinical protocol aims at therapy intensification or treatment
reduction. The conclusion that the ESG-MRD-ALL guidelines for
interpretation of RQ-PCR MRD data enable uniform interpreta-
tion of MRD data between different MRD laboratories is
therefore fully justified.
45
Choice of MRD technique
The three major MRD techniques provide information expressed
as seemingly identical MRD levels. Nevertheless, while flow
cytometry relies on protein expression, fusion genes are
generally detected at messenger RNA levels, and Ig/TCR
junctions are RQ-PCR targets at the DNA level. As summarized
in Table 1, the three methodologies also differ in their sensitivity
and applicability. Therefore, MRD data obtained with different
techniques are hardly interchangeable. Small single-center
studies claimed that results of MRD detection by flow cytometry
and quantitative PCR of patient-specific Ig/TCR gene rearrange-
ments are largely comparable.
46,47
Indeed in 70–80% of
samples with MRD levels 410
3
both techniques seem to give
comparable results (p3-fold difference). However, in samples
Why and how to quantify MRD
T Szczepan
´ski et al
624
Leukemia
with MRD levels p10
3
, many discrepancies between the two
techniques have been found; this hampers the recognition of
low-risk patients.
46,47
Consequently, usage of different MRD
techniques for different patients within the same treatment
protocol should be avoided.
What would be the future of MRD studies in ALL?
The story of MRD is one of the most exciting examples of
translational research, where complicated basic research was
transferred into high-technology laboratory diagnostics. It is
beyond doubt that MRD diagnostics will be included in all ALL
treatment protocols, as MRD data provide so far the most
optimal reflection of the in vivo response to treatment, which
gives the clinician a better knowledge and control of the clinical
course in individual patients. It is fair to assume that such
individualized medicine will finally translate into improved
outcome of ALL patients. This is already obvious in case of
high-risk ALL patients, particularly those undergoing allogeneic
HSCT and still remains to be proved for general cohorts of
patients. Introduction of novel, preferably targeted therapy will
create additional applications of MRD monitoring. Still ongoing
international collaborative efforts are necessary to ensure that all
diagnostic MRD laboratories speak the same ‘MRD language’.
Acknowledgements
The author is supported by scientific Grants number 2P054 095 30
and 3PO5E 094 25 from the Polish Ministry of Science and Higher
Education.
References
1 Szczepan
´ski T, Orfao A, van der Velden VHJ, San Miguel JF, van
Dongen JJM. Minimal residual disease in leukaemia patients.
Lancet Oncol 2001; 2: 409–417.
2 Hoelzer D, Gokbuget N, Ottmann O, Pui CH, Relling MV,
Appelbaum FR et al. Acute lymphoblastic leukemia. Hematology
(Am Soc Hematol Educ Program) 2002, 162–192.
3 Szczepan
´ski T, Flohr T, van der Velden VHJ, Bartram CR, van
Dongen JJM. Molecular monitoring of residual disease using
antigen receptor genes in childhood acute lymphoblastic leukae-
mia. Best Pract Res Clin Haematol 2002; 15: 37–57.
4 Cazzaniga G, Biondi A. Molecular monitoring of childhood acute
lymphoblastic leukemia using antigen receptor gene rearrange-
ments and quantitative polymerase chain reaction technology.
Haematologica 2005; 90: 382–390.
5 Van Dongen JJM, Seriu T, Panzer-Grumayer ER, Biondi A, Pongers-
Willemse MJ, Corral L et al. Prognostic value of minimal residual
disease in acute lymphoblastic leukaemia in childhood. Lancet
1998; 352: 1731–1738.
6 Cave H, van der Werff ten Bosch J, Suciu S, Guidal C, Waterkeyn
C, Otten J et al. Clinical significance of minimal residual disease
in childhood acute lymphoblastic leukemia. N Engl J Med 1998;
339: 591–598.
7 Coustan-Smith E, Behm FG, Sanchez J, Boyett JM, Hancock ML,
Raimondi SC et al. Immunological detection of minimal residual
disease in children with acute lymphoblastic leukaemia. Lancet
1998; 351: 550–554.
8 Nyvold C, Madsen HO, Ryder LP, Seyfarth J, Svejgaard A, Clausen
Net al. Precise quantification of minimal residual disease at
day 29 allows identification of children with acute lymphoblastic
leukemia and an excellent outcome. Blood 2002; 99: 1253–1258.
9 Dworzak MN, Froschl G, Printz D, Mann G, Potschger U,
Muhlegger N et al. Prognostic significance and modalities of flow
cytometric minimal residual disease detection in childhood acute
lymphoblastic leukemia. Blood 2002; 99: 1952–1958.
10 Panzer-Grumayer ER, Schneider M, Panzer S, Fasching K, Gadner
H. Rapid molecular response during early induction chemotherapy
predicts a good outcome in childhood acute lymphoblastic
leukemia. Blood 2000; 95: 790–794.
11 Coustan-Smith E, Sancho J, Behm FG, Hancock ML, Razzouk BI,
Ribeiro RC et al. Prognostic importance of measuring early
clearance of leukemic cells by flow cytometry in childhood acute
lymphoblastic leukemia. Blood 2002; 100: 52–58.
12 Knechtli CJ, Goulden NJ, Hancock JP, Grandage VL, Harris EL,
Garland RJ et al. Minimal residual disease status before allogeneic
bone marrow transplantation is an important determinant of
successful outcome for children and adolescents with acute
lymphoblastic leukemia. Blood 1998; 92: 4072–4079.
13 Eckert C, Biondi A, Seeger K, Cazzaniga G, Hartmann R, Proba J
et al. Prognostic value of minimal residual disease in relapsed
childhood acute lymphoblastic leukaemia. Lancet 2001; 358:
1239–1241.
14 Van der Velden VHJ, Joosten SA, Willemse MJ, van Wering ER,
Lankester A, van Dongen JJM et al. Real-time quantitative PCR for
detection of minimal residual disease before allogeneic stem cell
transplantation predicts outcome in children with acute lympho-
blastic leukemia. Leukemia 2001; 15: 1485–1487.
15 Bader P, Hancock J, Kreyenberg H, Goulden NJ, Niethammer D,
Oakhill A et al. Minimal residual disease (MRD) status prior to
allogeneic stem cell transplantation is a powerful predictor for post
transplant outcome in children with ALL. Leukemia 2002; 16:
1668–1672.
16 Krejci O, van der Velden VHJ, Bader P, Kreyenberg H, Goulden N,
Hancock J et al. Level of minimal residual disease prior to
haematopoietic stem cell transplantation predicts prognosis in
paediatric patients with acute lymphoblastic leukaemia: a report of
the Pre-BMT MRD Study Group. Bone Marrow Transplant 2003;
32: 849–851.
17 Coustan-Smith E, Gajjar A, Hijiya N, Razzouk BI, Ribeiro RC,
Rivera GK et al. Clinical significance of minimal residual disease in
childhood acute lymphoblastic leukemia after first relapse.
Leukemia 2004; 18: 499–504.
18 Bruggemann M, Raff T, Flohr T, Gokbuget N, Nakao M, Droese J
et al. Clinical significance of minimal residual disease quantifica-
tion in adult patients with standard-risk acute lymphoblastic
leukemia. Blood 2006; 107: 1116–1123.
19 Campana D. Determination of minimal residual disease in
leukaemia patients. Br J Haematol 2003; 121: 823–838.
20 Szczepan
´ski T, van der Velden VH, van Dongen JJM. Flow-
cytometric immunophenotyping of normal and malignant lympho-
cytes. Clin Chem Lab Med 2006; 44: 775–796.
21 Van der Velden VHJ, Hochhaus A, Cazzaniga G, Szczepan
´ski T,
Gabert J, Van Dongen JJM. Detection of minimal residual disease in
hematologic malignancies by real-time quantitative PCR: principles,
approaches, and laboratory aspects. Leukemia 2003; 17: 1013–1034.
22 Gaipa G, Basso G, Maglia O, Leoni V, Faini A, Cazzaniga G et al.
Drug-induced immunophenotypic modulation in childhood ALL:
implications for minimal residual disease detection. Leukemia
2005; 19: 49–56.
23 Van der Sluijs-Gelling AJ, van der Velden VHJ, Roeffen ETJM,
Veerman AJP, van Wering ER. Immunophenotypic modulation in
childhood precursor-B-ALL can be mimicked in vitro and is related
to the induction of cell death. Leukemia 2005; 19: 1845–1847.
24 Van Wering ER, Beishuizen A, Roeffen ET, van der Linden-
Schrever BE, Verhoeven MA, Hahlen K et al. Immunophenotypic
changes between diagnosis and relapse in childhood acute
lymphoblastic leukemia. Leukemia 1995; 9: 1523–1533.
25 Orfao A, Lo
´pez A, Flores J, Almeida J, Vidriales B, Perez J et al.
Diagnosis of hematological malignancies: new applications for
flow cytometry. Hematology (EHA Educ Program) 2006; 2: 6–13.
26 Van Dongen JJM, Macintyre EA, Gabert JA, Delabesse E, Rossi V,
Saglio G et al. Standardized RT-PCR analysis of fusion gene
transcripts from chromosome aberrations in acute leukemia for
detection of minimal residual disease. Report of the BIOMED-1
Concerted Action: investigation of minimal residual disease in
acute leukemia. Leukemia 1999; 13: 1901–1928.
27 Gabert J, Beillard E, Van der Velden VHJ, Bi W, Grimwade D,
Pallisgaard N et al. Standardization and quality control studies of
‘real-time’ quantitative reverse transcriptase polymerase chain
reaction of fusion gene transcripts for residual disease detection in
Why and how to quantify MRD
T Szczepan
´ski et al
625
Leukemia
leukemia a Europe Against Cancer program. Leukemia 2003; 17:
2318–2357.
28 Beillard E, Pallisgaard N, van der Velden VH, Bi W, Dee R, van der
Schoot E et al. Evaluation of candidate control genes for diagnosis
and residual disease detection in leukemic patients using ‘real-
time’ quantitative reverse-transcriptase polymerase chain reaction
(RQ-PCR) a Europe against cancer program. Leukemia 2003; 17:
2474–2486.
29 Arico M, Valsecchi MG, Camitta B, Schrappe M, Chessells J,
Baruchel A et al. Outcome of treatment in children with
Philadelphia chromosome-positive acute lymphoblastic leukemia.
N Engl J Med 2000; 342: 998–1006.
30 Scheuring UJ, Pfeifer H, Wassmann B, Bruck P, Gehrke B,
Petershofen EK et al. Serial minimal residual disease (MRD)
analysis as a predictor of response duration in Philadelphia-
positive acute lymphoblastic leukemia (Ph+ALL) during imatinib
treatment. Leukemia 2003; 17: 1700–1706.
31 Wassmann B, Pfeifer H, Stadler M, Bornhauser M, Bug G,
Scheuring UJ et al. Early molecular response to posttransplantation
imatinib determines outcome in MRD+ Philadelphia-positive
acute lymphoblastic leukemia (Ph+ ALL). Blood 2005; 106:
458–463.
32 Gleissner B, Rieder H, Thiel E, Fonatsch C, Janssen LA, Heinze B
et al. Prospective BCR-ABL analysis by polymerase chain reaction
(RT-PCR) in adult acute B-lineage lymphoblastic leukemia:
reliability of RT-nested-PCR and comparison to cytogenetic data.
Leukemia 2001; 15: 1834–1840.
33 Meyer C, Schneider B, Jakob S, Strehl S, Attarbaschi A, Schnittger S
et al. The MLL recombinome of acute leukemias. Leukemia 2006;
20: 777–784.
34 Burmeister T, Marschalek R, Schneider B, Meyer C, Gokbuget N,
Schwartz S et al. Monitoring minimal residual disease by
quantification of genomic chromosomal breakpoint sequences
in acute leukemias with MLL aberrations. Leukemia 2006; 20:
451–457.
35 Pongers-Willemse MJ, Seriu T, Stolz F, d’Aniello E, Gameiro P,
Pisa P et al. Primers and protocols for standardized MRD detection
in ALL using immunoglobulin and T cell receptor gene rearrange-
ments and TAL1 deletions as PCR targets. Report of the BIOMED-1
Concerted Action: investigation of minimal residual disease in
acute leukemia. Leukemia 1999; 13: 110–118.
36 Van Dongen JJM, Langerak AW, Bruggemann M, Evans PAS,
Hummel M, Lavender FL et al. Design and standardization of PCR
primers and protocols for detection of clonal immunoglobulin and
T-cell receptor gene recombinations in suspect lymphoprolifera-
tions: report of the BIOMED-2 Concerted Action BMH4-CT98-
3936. Leukemia 2003; 17: 2257–2317.
37 Szczepan
´ski T, Van der Velden VHJ, Hoogeveen PG, de Bie M,
Jacobs DC, van Wering ER et al. Vdelta2-Jalpha rearrangements
are frequent in precursor-B-acute lymphoblastic leukemia but rare
in normal lymphoid cells. Blood 2004; 103: 3798–3804.
38 Szczepan
´ski T, Willemse MJ, van Wering ER, Weerden JF,
Kamps WA, van Dongen JJM. Precursor-B-ALL with DH-JH gene
rearrangements have an immature immunogenotype with a high
frequency of oligoclonality and hyperdiploidy of chromosome 14.
Leukemia 2001; 15: 1415–1423.
39 De Haas V, Verhagen OJ, von dem Borne AE, Kroes W, van den
Berg H, van der Schoot CE. Quantification of minimal residual
disease in children with oligoclonal B-precursor acute lympho-
blastic leukemia indicates that the clones that grow out during
relapse already have the slowest rate of reduction during induction
therapy. Leukemia 2001; 15: 134–140.
40 Szczepan
´ski T, Willemse MJ, Brinkhof B, van Wering ER, van der
Burg M, van Dongen JJM. Comparative analysis of Ig and TCR gene
rearrangements at diagnosis and at relapse of childhood precursor-
B-ALL provides improved strategies for selection of stable PCR
targets for monitoring of minimal residual disease. Blood 2002; 99:
2315–2323.
41 Germano G, del Giudice L, Palatron S, Giarin E, Cazzaniga G,
Biondi A et al. Clonality profile in relapsed precursor-B-ALL
children by GeneScan and sequencing analyses. Consequences
on minimal residual disease monitoring. Leukemia 2003; 17:
1573–1582.
42 Szczepan
´ski T, Van der Velden VHJ, Raff T, Jacobs DC, van
Wering ER, Bruggemann M et al. Comparative analysis of T-cell
receptor gene rearrangements at diagnosis and relapse of T-cell
acute lymphoblastic leukemia (T-ALL) shows high stability of
clonal markers for monitoring of minimal residual disease and
reveals the occurrence of second T-ALL. Leukemia 2003; 17:
2149–2156.
43 Brisco MJ, Condon J, Hughes E, Neoh SH, Sykes PJ, Seshadri R
et al. Outcome prediction in childhood acute lymphoblastic
leukaemia by molecular quantification of residual disease at the
end of induction. Lancet 1994; 343: 196–200.
44 Pongers-Willemse MJ, Verhagen OJHM, Tibbe GJM, Wijkhuijs JM,
De Haas V, Roovers E et al. Real-time quantitative PCR for the
detection of minimal residual disease in acute lymphoblastic
leukemia using junctional regions specific TaqMan probes.
Leukemia 1998; 12: 2006–2014.
45 Van der Velden VHJ, Cazzaniga G, Schrauder A, Hancock J, Bader
P, Panzer-Grumayer R et al. Analysis of minimal residual disease
by Ig/TCR gene rearrangements: guidelines for interpretation of
real-time quantitative PCR data. Leukemia 2007; 21, (in press).
46 Malec M, Bjorklund E, Soderhall S, Mazur J, Sjogren AM, Pisa P
et al. Flow cytometry and allele-specific oligonucleotide PCR are
equally effective in detection of minimal residual disease in ALL.
Leukemia 2001; 15: 716–727.
47 Neale GAM, CoustanSmith E, Pan Q, Chen X, Gruhn B, Stow P
et al. Tandem application of flow cytometry and polymerase chain
reaction for comprehensive detection of minimal residual disease
in childhood acute lymphoblastic leukemia. Leukemia 1999; 13:
1221–1226.
48 Graux C, Cools J, Michaux L, Vandenberghe P, Hagemeijer A.
Cytogenetics and molecular genetics of T-cell acute lymphoblastic
leukemia: from thymocyte to lymphoblast. Leukemia 2006; 20:
1496–1510.
Why and how to quantify MRD
T Szczepan
´ski et al
626
Leukemia
... Measurement of MRD is performed at different time points during and after treatment with prognostic value at the cutoff level of 0.01% or more MRD cells indicating a risk for leukemia relapse. [3,4] The study is an overview on the latest research methods and trends for minimal residual disease diagnostics in acute lymphoblastic leukemia by molecular approaches or flow cytometry with an emphasis on the latter. ...
... It is characterized by a high sensitivity (10 −5 -10 −6 ), but has some disadvantages such as lack of standardization, complex methodology, high cost, and application in less than half of the cases. [4][5][6] Reverse transcription polymerase chain reaction (RT-PCR) ...
... Limitations: mRNA instability, quantitative errors, high cost, low specificity, cross-contamination of products, false-positive results in up to 20% of cases, and need for detection of chromosomal abnormalities at diagnosis for follow up. [4,5,7] Digital droplet polymerase chain reaction A modern method that allows for absolute quantification of the target DNA without the need of calibration curves. It is applicable in 95% of cases, but there is no standardization. ...
Article
Full-text available
Minimal residual disease refers to a leukemia cell population that is resistant to chemotherapy or radiotherapy and leads to disease relapse. The assessment of MRD is crucial for making an accurate prognosis of the disease and for the choice of optimal treatment strategy. Here, we review the advantages and disadvantages of the available genetic and phenotypic methods and focus on the multiparametric flow cytometry as a promising method with greater sensitivity, speed, and standardization options. In addition, we discuss how the application of automated data analysis outweighs the use of complex combinations of windows and gates in classical analysis, thus eliminating subjective evaluation.
... However, heterogeneity is a common phenomenon in BCP-ALL [4,5], raising concerns that identified IG/TR rearrangements are not uniformly present in all leukemic blasts at the time of diagnosis. The preservation of the IG/TR gene rearrangements is found in only 70% of BCP-ALL and 90% of T-ALL patients at the time of relapse [6,7], either because of continuing V(D)J recombinase activity in the malignant cells, or due to the limited sensitivity of the available methods for identifying IG/TR genes that cause minor subpopulations to be overlooked at the time of diagnosis. Of note, the metastasis-like dissemination of ALL cells to the central nervous system is a common cause of relapse in ALL [8], and in some patients, the IG gene rearrangements that dominantly present in the ALL cells in the CNS are found only at very low levels in the bone marrow at the time of diagnosis [9]. ...
Article
Full-text available
Next-generation sequencing (NGS) methods have been introduced for immunoglobulin (IG)/T-cell receptor (TR) gene rearrangement analysis in acute lymphoblastic leukemia (ALL) and lymphoma (LBL). These methods likely constitute faster and more sensitive approaches to analyze heterogenous cases of ALL/LBL, yet it is not known whether gene rearrangements constituting low percentages of the total sequence reads represent minor subpopulations of malignant cells or background IG/TR gene rearrangements in normal B-and T-cells. In a comparison of eight cases of B-cell precursor ALL (BCP-ALL) using both the EuroClonality NGS method and the IdentiClone multiplex-PCR/gene-scanning method, the NGS method identified between 29% and 139% more markers than the gene-scanning method, depending on whether the NGS data analysis used a threshold of 5% or 1%, respectively. As an alternative to using low thresholds, we show that IG/TR gene rearrangements in subpopulations of cancer cells can be discriminated from background IG/TR gene rearrangements in normal B-and T-cells through a combination of flow cytometry cell sorting and multiple displacement amplification (MDA)-based whole genome amplification (WGA) prior to the NGS. Using this approach to investigate the clonal evolution in a BCP-ALL patient with double relapse, clonal TR rearrangements were found in sorted leukemic cells at the time of second relapse that could be identified at the time of diagnosis, below 1% of the total sequence reads. These data emphasize that caution should be exerted when interpreting rare sequences in NGS experiments and show the advantage of employing the flow sorting of malignant cell populations in NGS clonality assessments.
... Control parameters related to drug, δ j with j ∈ J, have been selected by searching for values that implies that the patient responds to treatment correctly, i.e. considering an amount of blasts less than 10 6 blasts/ml in blood in day +8 and MRD< 0.01% in bone marrow in day +15 [52]. In preliminary simulations we find that if δ j q j < 10 0 , the dose administered is ineffective. ...
Article
Full-text available
Acute Lymphoblastic Leukemia (ALL) accounts for the 80% of leukemias when coming down to pediatric ages. Survival of these patients has increased by a considerable amount in recent years. However, around 15−20% of treatments are unsuccessful. For this reason, it is definitely required to come up with new strategies to study and select which patients are at higher risk of relapse. Thus the importance to monitor the amount of leukemic cells to predict relapses in the first treatment phase. In this work we develop a mathematical model describing the behavior of ALL, examining the evolution of a leukemic clone when treatment is applied. In the study of this model it can be observed how the risk of relapse is connected with the response in the first treatment phase. This model is able to simulate cell dynamics without treatment, representing a virtual patient bone marrow behavior. Furthermore, several parameters are related to treatment dynamics, therefore proposing a basis for future works regarding childhood ALL survival improvement.
... The limitation of prognostic at 0.01% is based on the immunohistochemical detection of 3-4-color flow cytometers. The clinical significance of the 0.01% MRD cutoff level refers to a patient has cellular MRD levels ≥0.01% in a bone marrow sample at important measurement time points during therapy, if MRD levels are less than 0.01% means the patient will have a significantly higher risk for leukemia relapse (Szczepański, 2007;Campana, 2010;Short and Jabbour, 2017). Immunophenotypes characteristic of leukemic cells are distinguished from normal cells by flow cytometry (FC). ...
Article
Full-text available
Patients with acute leukemia port 10 malignant cells at presentation. Following chemotherapy or stem cell transplant, patients in complete remission by conventional analyses may still harbor 106/108 malignant cells below the detection limit of standard clinical assessment. Minimal residual disease (MRD) monitoring is one of the most powerful predictors of disease-free and overall survival, particularly for children with acute lymphoblastic leukemia (cALL), the percent annual of cALL increase in the incidence of cALL in Saudi Arabia. Breakpoint fusion regions of chromosomal aberrations can be used as tumor-specific targets for MRD detection by polymerase chain reaction. Levels of MRD, measured at critical time points, significantly correlate with clinical outcomes. Previous works investigated the prognostic significance of leukemia-associated immunophenotypes (LAIPs) as an assessment of the index of MRD in 125 adult B-ALL patients by eight-colour flow cytometry. More advanced molecular and genetics studies are so necessary to identify the mechanisms and cellular structure of the minimal-level disease. Selecting molecular methods for minimal residual disease detection have a much higher sensitivity and precision (100-fold or more) than others. This review highlights the minimal residual disease molecular detection to demonstrate the characterization of the lymphoblastic leukemia gene. Precise MRD monitoring predicts disease relapse after chemotherapy or SCT, provides early intervention, and may result in the rescue of many patients and improvement in the probability of long-term disease-free survival.
... The study of all estimated parameters is presented in Supplementary Information document. Control parameters related to drug, δ j with j ∈ J, have been selected by searching for values that implies that the patient responds to treatment correctly, i.e. considering an amount of blasts less than 10 6 blasts/ml in blood in day +8 and MRD< 0.01% in bone marrow in day +15 [53]. In preliminary simulations we find that if δ j q j < 10 0 , the dose administered is ineffective. ...
Preprint
Full-text available
Acute Lymphoblastic Leukemia (ALL) accounts for the 80% of leukemias when coming down to pediatric ages. Survival of these patients has increased by a considerable amount in recent years. However, around 15-20% of treatments are unsuccessful. For this reason, it is definitely required to come up with new strategies to study and select which patients are at higher risk of relapse. Thus the importance to monitor the amount of leukemic cells to predict relapses in the first treatment phase. In this work we develop a mathematical model describing the behavior of ALL, examining the evolution of a leukemic clone when treatment is applied. In the study of this model it can be observed how the risk of relapse is connected with the response in the first treatment phase. This model is able to simulate cell dynamics without treatment, representing a virtual patient bone marrow behavior. Furthermore, several parameters are related to treatment dynamics, therefore proposing a basis for future works regarding childhood ALL survival improvement.
... The main cause of cancer-related death is relapse, which is known to arise from small numbers of drug-resistant minimal residual disease (MRD) cells throughout therapy at levels below morphologic detection (Brüggemann et al., 2012). Therefore, early detection of MRD is critical for evaluating treatment response, allowing the stratification of patients with high-risk disease to receive more intensive treatment regimens (Szczepański, 2007;Della Starza et al., 2019). ...
Article
Full-text available
The measurement of circulating tumor DNA (ctDNA) has gained increasing prominence as a minimally invasive tool for the detection of cancer-specific markers in plasma. In adult cancers, ctDNA detection has shown value for disease-monitoring applications including tumor mutation profiling, risk stratification, relapse prediction, and treatment response evaluation. To date, there are ctDNA tests used as companion diagnostics for adult cancers and it is not understood why the same cannot be said about childhood cancer, despite the marked differences between adult and pediatric oncology. In this review, we discuss the current understanding of ctDNA as a disease monitoring biomarker in the context of pediatric malignancies, including the challenges associated with ctDNA detection in liquid biopsies. The data and conclusions from pediatric cancer studies of ctDNA are summarized, highlighting treatment response, disease monitoring and the detection of subclonal disease as applications of ctDNA. While the data from retrospective studies highlight the potential of ctDNA, large clinical trials are required for ctDNA analysis for routine clinical use in pediatric cancers. We outline the requirements for the standardization of ctDNA detection in pediatric cancers, including sample handling and reproducibility of results. With better understanding of the advantages and limitations of ctDNA and improved detection methods, ctDNA analysis may become the standard of care for patient monitoring in childhood cancers.
... MRD detection is an already well-established, independent prognostic indicator in ALL and one of the key parameters in contemporary ALL treatment protocols used for patient stratification [5,6,8,9]. Currently, two methods of MRD monitoring are employed: molecular technique based on clone-specific quantitative PCR amplification of immunoglobulin/T-cell receptor genes and flow cytometry (FC). The latter technique successfully competes with more time-consuming PCR-based techniques, and its application in a multiparametric (≥ 8-parameter) setting can provide the same sensitivity levels (down to 10 -5 ) [10][11][12]. ...
Article
Full-text available
Flow cytometry (FCM) is a precise and well-established tool to assess the minimal residual disease (MRD) level in childhood acute lymphoblastic leukemia (ALL). It is crucial to distinguish leukemic cells from their normal counterparts; thus new markers should be evaluated, to increase the accuracy of the analysis. The expression of CD73 on blast cells was measured and compared at the day of diagnosis and at days 15 and 33 of treatment. To determine antigen expression levels, a normalized scale based on median fluorescence intensity (nMFI) was used. The study group consisted of 188 patients from the Polish Pediatric Leukemia and Lymphoma Study Group. From 177 patients with positive MRD at day 15 of treatment, in 147 (83.1%) cases an increase of CD73 expression was observed (mean increase of +17 nMFI units). In addition, an increase of CD73 expression was noted in 26 of 31 (83.9%) patients at day 33 of treatment. In turn, a decrease of CD73 expression was observed only in 13/177 (7.3%) and 1/31 (3.2%) cases at days 15 and 33 of treatment, respectively. In 17 (9.6%) patients no change in expression of CD73 between diagnosis and day 15 of treatment was observed. In the great majority of cases the expression of CD73 is not only stable but increases during the early stages of treatment, which makes it a very useful marker to be used for MRD monitoring in childhood B-cell precursor (BCP)-ALL patients.
Article
Treatment algorithms differ for adult patients with Philadelphia-negative (Ph-) and Philadelphia-positive (Ph+) acute lymphoblastic leukemia (ALL). For Ph- ALL intensive induction-consolidation chemotherapy using “pediatric-inspired” protocols is a standard of care. Allogeneic hematopoietic cell transplantation (allo-HCT) from either an HLA-matched sibling, unrelated or haploidentical donor should be considered for patients with high estimated risk of relapse. Inadequate response at the level of measurable residual disease (MRD) is the strongest adverse prognostic factor. Patients with B-ALL and detectable MRD should be treated with blinatumomab. In the future, the use of blinatumomab and/or inotuzumab ozogamycin in addition to first-line chemotherapy may become a new standard of care reducing the role of allo-HCT. For patients with Ph+ ALL, tyrosine kinase inhibitors (TKI) are the most important components of treatment protocols, while the intensity of chemotherapy may be reduced. Allo-HCT is recommended for all patients treated with imatinib along with low-intensity chemotherapy. Results of phase-II studies using front-line dasatinib or ponatinib in sequence or in combination with blinatumomab are very promising. Such a strategy may allow the avoidance of systemic chemotherapy. The future role of allo-HCT in this context appears uncertain.
Article
Quantification of measurable residual disease (MRD) in acute lymphoblastic leukemia (ALL) is a well-established clinical tool used to risk stratify patients during the course of chemotherapy, immunotherapy, and/or transplant therapy. As technologies evolve, the sensitivity for quantifying exceptionally low disease burden using either next generation sequencing (NGS) or next generation flow cytometry (NGF) has improved. It is now possible to detect MRD and quantify it precisely in patients who would previously have been deemed MRD negative by older, lower sensitivity methods. Persistence or recurrence of ALL disease burden above 10⁻⁴ (0.01%) is accepted as the minimum threshold for making clinical decisions, but with NGS and NGF, clinicians now confront decision-making with disease burdens sometimes quantified to as low as 10⁻⁶ (0.0001%, or one leukemia cell in a million leukocytes). Emerging data suggest these higher sensitivity methods are superior for identifying patients at lowest risk for relapse, but it remains controversial whether to institute therapies such as blinatumomab or chimeric antigen receptor (CAR)-T cells or move patients to allogeneic hematopoietic cell transplant (alloHCT) when they have quantifiable disease burden less than 10⁻⁴. With additional evidence to facilitate integration of highly sensitive MRD quantification into clinical care and to contextualize MRD within the genotype of individual patients, it will likely be increasingly possible to identify patients able to avoid alloHCT and potentially even de-escalate therapy.
Article
Full-text available
Prospective studies on the detection of minimal residual disease (MRD) in acute leukemia patients have shown that large-scale MRD studies are feasible and that clinically relevant MRD-based risk group classification can be achieved and can now be used for designing new treatment protocols. However, multicenter international treatment protocols with MRD-based stratification of treatment need careful standardization and quality control of the MRD techniques. This was the aim of the European BIOMED-1 Concerted Action ‘Investigation of minimal residual disease in acute leukemia: international standardization and clinical evaluation’ with participants of 14 laboratories in eight European countries (ES, NL, PT, IT, DE, FR, SE and AT). Standardization and quality control was performed for the three main types of MRD techniques, ie flow cytometric immunophenotyping, PCR analysis of antigen receptor genes, and RT-PCR analysis of well-defined chromosomal aberrations. This study focussed on the latter MRD technique. A total of nine well-defined chromosome aberrations with fusion gene transcripts were selected: t(1;19) with E2A-PBX1, t(4;11) with MLL-AF4, t(8;21) with AML1-ETO, t(9;22) with BCR-ABL p190 and BCR-ABL p210, t(12;21) with TEL-AML1, t(15;17) with PML-RARA, inv (16) with CBFB-MYH11, and microdeletion 1p32 with SIL-TAL1. PCR primers were designed according to predefined criteria for single PCR (external primers A ↔ B) and nested PCR (internal primers C ↔ D) as well as for ‘shifted’ PCR with a primer upstream (E5′ primer) or downstream (E3′ primer) of the external A ↔ B primers. The ‘shifted’ E primers were designed for performing an independent PCR together with one of the internal primers for confirmation (or exclusion) of positive results. Various local RT and PCR protocols were compared and subsequently a common protocol was designed, tested and adapted, resulting in a standardized RT-PCR protocol. After initial testing (with adaptations whenever necessary) and approval by two or three laboratories, the primers were tested by all participating laboratories, using 17 cell lines and patient samples as positive controls. This testing included comparison with local protocols and primers as well as sensitivity testing via dilution experiments. The collaborative efforts resulted in standardized primer sets with a minimal target sensitivity of 10−2 for virtually all single PCR analyses, whereas the nested PCR analyses generally reached the minimal target sensitivity of 10−4. The standardized RT-PCR protocol and primer sets can now be used for molecular classification of acute leukemia at diagnosis and for MRD detection during follow-up to evaluate treatment effectiveness.
Article
Full-text available
The implications of the detection of residual disease after treatment of acute lymphoblastic leukemia (ALL) are unclear. We conducted a prospective study at 11 centers to determine the predictive value of the presence or absence of detectable residual disease at several points in time during the first six months after complete remission of childhood ALL had been induced. Junctional sequences of T-cell-receptor or immunoglobulin gene rearrangements were used as clonal markers of leukemic cells. Residual disease was quantitated with a competitive polymerase-chain-reaction (PCR) assay. Of 246 patients enrolled at diagnosis and treated with a uniform chemotherapy protocol, 178 were monitored for residual disease with one clone-specific probe (in 74 percent) or more than one probe (in 26 percent). The median follow-up period was 38 months. The presence or absence and level of residual leukemia were significantly correlated with the risk of early relapse at each of the times studied (P<0.001). PCR measurements identified patients at high risk for relapse after the completion of induction therapy (those with > or =10(-2) residual blasts) or at later time points (those with > or =10(-3) residual blasts). Multivariate analysis showed that as compared with immunophenotype, age, risk group (standard or very high risk), and white-cell count at diagnosis, the presence or absence and level of residual disease were the most powerful independent prognostic factors. Residual leukemia after induction of a remission is a powerful prognostic factor in childhood ALL. Detection of residual disease by PCR should be used to identify patients at risk for relapse and should be taken into account in considering alternative treatment.
Article
Full-text available
A total of 28 children and nine adults with relapsed T-ALL were analyzed for the configuration of their T-cell receptor (TCR) and TAL1 genes at diagnosis and relapse to evaluate their stability throughout the disease course. A total of 150 clonal TCR and TAL1 gene rearrangements were identified in the 37 patients at diagnosis. In 65% of cases all rearrangements and in 27% of cases most rearrangements found at diagnosis were preserved at relapse. Two children with unusually late T-ALL recurrences displayed completely different TCR gene rearrangement sequences between diagnosis and relapse. This indicates that a proportion of very late T-ALL recurrences might represent second T-ALL. Specifically, 88% of clonal rearrangements identified at diagnosis in truly relapsed T-ALL were preserved at relapse. This is significantly higher as compared to previously studied precursor-B-ALL ( approximately 70%). Thus, from biological point of view, immunogenotype of T-ALL is more stable as compared with precursor-B-ALL. The overall stability of TCR gene rearrangements was higher in adult T-ALL (97%) than in childhood T-ALL (86%). Based on the stability of TCR gene rearrangements, we propose a strategy for PCR target selection (TCRD+TAL1 --> TCRB --> TCRG), which probably allows reliable minimal residual disease detection in all T-ALL patients.
Article
Full-text available
Assessment of minimal residual disease (MRD) by flow cytometry is considered to be based on the reproducibility of the leukemic immunophenotype detected at diagnosis. However, we previously noticed modulation of surface antigen expression in acute lymphoblastic leukemia (ALL) during the early treatment. Hence, we investigated this in 30 children with B-cell precursor ALL consecutively enrolled in the AIEOP-BFM ALL 2000 protocol. Quantitative expression of seven antigens useful in MRD monitoring was studied at diagnosis and compared to that measured at different time points of remission induction therapy. Downmodulation in the expression of CD10 and CD34 occurred at follow-up. By contrast, upmodulation of CD19, CD20, CD45RA, and CD11a was observed, while the expression of CD58 remained stable. Despite this, we could unambiguously discriminate leukemic cells from normal residual B cells. This holds true when bone marrow (BM) samples from similarly treated T-ALL patients, but not from healthy donors, were used as reference. Our results indicate that immunophenotypic modulation occurs in ALL during the early phases of BFM-type protocols. However, the accuracy of MRD detection by flow cytometry seems not negatively affected if adequate analysis protocols are employed. Investigators should take this phenomenon into account in order to avoid pitfalls in flow cytometric MRD studies.
Article
The efficacy of allografting in acute lymphoblastic leukemia (ALL) is heavily influenced by remission status at the time of transplant. Using polymerase chain reaction (PCR)-based minimal residual disease (MRD) analysis, we have investigated retrospectively the impact of submicroscopic leukemia on outcome in 64 patients receiving allogeneic bone marrow transplantation (BMT) for childhood ALL. Remission BM specimens were taken 6 to 81 days (median, 23) before transplant. All patients received similar conditioning therapy; 50 received grafts from unrelated donors and 14 from related donors. Nineteen patients were transplanted in first complete remission (CR1) and 45 in second or subsequent CR. MRD was analyzed by PCR of Ig or T-cell receptor δ or γ rearrangements, electrophoresis, and allele-specific oligoprobing. Samples were rated high-level positive (clonal band evident after electrophoresis; sensitivity 10−2 to 10−3), low-level positive (MRD detected only after oligoprobing; sensitivity 10−3 to 10−5), or negative. Excluding 8 patients transplanted in CR2 for isolated extramedullary relapse (all MRD−), MRD was detected at high level in 12 patients, low level in 11, and was undetectable in 33. Two-year event-free survival for these groups was 0%, 36%, and 73%, respectively (P < .001). Follow-up in patients remaining in continuing remission is 20 to 96 months (median, 35). These results suggest that MRD analysis could be used routinely in this setting. This would allow identification of patients with resistant leukemia (who may benefit from innovative BMT protocols) and of those with more responsive disease (who may be candidates for randomized trials of BMT versus modern intensive relapse chemotherapy).
Article
This is a comprehensive overview on the most recent developments in diagnosis and treatment of acute lymphoblastic leukemia (ALL). Dr. Dieter Hoelzer and colleagues give an overview of current chemotherapy approaches, prognostic factors, risk stratification, and new treatment options such as tyrosine kinase inhibitors and monoclonal antibodies. Furthermore the role of minimal residual disease (MRD) for individual treatment decisions in prospective clinical studies in adult ALL is reviewed. Drs. Ching-Hon Pui and Mary Relling discuss late treatment sequelae in childhood ALL. The relation between the risk of second cancer and treatment schedule, pharmacogenetics, and gene expression profile studies is described. Also pathogenesis, risk factors, and management of other complications such as endocrinopathy, bone demineralization, obesity, and avascular necrosis of bone is reviewed. Dr. Fred Appelbaum addresses long-term results, late sequelae and quality of life in ALL patients after stem cell transplantation. New options for reduction of relapse risk, e.g., by intensified conditioning regimens or donor lymphocyte infusions, for reduction of mortality and new approaches such as nonmyeloablative transplantation in ALL are discussed. Drs. Jacques van Dongen and Tomasz Szczepanski demonstrate the prognostic value of MRD detection via flow cytometry or PCR analysis in childhood ALL. They discuss the relation between MRD results and type of treatment protocol, timing of the follow-up samples, and the applied technique and underline the importance of standardization and quality control. They also review MRD-based risk group definition and clinical consequences.
Chapter
Acute lymphoblastic leukemia (ALL) is a malignant hematologic disease characterized by the accumulation of immature cells within the marrow space that are arrested at the lymphoblast stage of development, with consequent suppression of normal hematopoiesis. ALL is not a biologically uniform disease; rather, it is a collection of heterogeneous entities characterized by distinct phenotypic, cytogenetic, and molecular-genetic profiles. The characterization and study of these subtypes has led to a risk-adapted approach to therapy, an approach pioneered by clinicians involved with the study of ALL in children, which has now been extended to the diagnosis and treatment of adult ALL. Much progress has been made over the years, particularly in pediatric ALL where cure rates are now in the 80% range. In contrast, for adults with ALL, only approximately one-third of patients achieve long-term disease-free survival. Utilization of newer molecular techniques, such as gene expression profiling, will lead to an improved understanding of the biology of the disease, as well as identification of new drug targets and unique drug susceptibility and resistance profiles. Ultimately, these insights will lead to a more refined and targeted approach to therapy with subsequent significant improvement in the cure rates for adults afflicted with this disease.
Article
To get more insight into the phenotypic changes of childhood acute lymphoblastic leukemia (ALL) at relapse, a detailed morphological and immunophenotypic study in 40 childhood ALL cases (32 precursor B-ALL and 8 T-ALL) was performed. Expression patterns of non-lineage specific markers (terminal deoxynucleotidyl transferase (TdT), CD34, and HLA-DR), B-lineage markers (CD10, CD19, CD20, and CD22), T-lineage markers (CD1, CD2, CD3, CD4, CD5, CD7, and CD8), and cross-lineage myeloid markers (CD14, CD15, and CD33) were compared at diagnosis and relapse. In case of low blast counts (< or = 70%) at relapse, double labeling for membrane markers and TdT was used in order to define the precise immunophenotype of the TdT+ leukemic cells. An immunological marker-shift was defined as either a conversion from positive to negative and vice versa or a difference in positivity of > or = 50%. Morphological differences between diagnosis and relapse were detected in 34% of precursor B-ALL and 14% of T-ALL. Differences in immunological marker expression were found in 72% of precursor B-ALL and in 75% of T-ALL, and generally concerned minor shifts with loss or acquisition of a few markers. The morphological shifts and immunophenotypic shifts were not correlated. Immunophenotypic shifts were found for all markers tested in precursor B-ALL, except for HLA-DR. Shifts in CD10 expression (16% of cases) were only observed in relapses occurring 30 months or more after diagnosis. In four precursor B-ALL an intra-lineage shift was found at relapse (one common ALL to null ALL and three pre-B-ALL to common ALL or null ALL) and two precursor B-ALL cases were diagnosed as acute non-lymphocytic leukemia at relapse based on morphology and immunophenotype. In T-ALL, neither intra-lineage nor inter-lineage shifts were observed, although shifts were detected in all T cell markers tested, except for the lineage specific CD3 and T cell receptor (TcR) markers. In conclusion, immunophenotypic shifts at relapse frequently occur in precursor B-ALL and T-ALL, in a small percentage leading to an intra-lineage shift (10%) or inter-lineage shift (5%). Therefore immunophenotypic monitoring of minimal residual disease in ALL patients should be based on multiple marker combinations, preferably together with polymerase chain reaction analysis of rearranged immunoglobulin and/or TcR genes or chromosome aberrations.
Article
Methods to detect and quantify minimal residual disease (MRD) after chemotherapy for acute lymphoblastic leukaemia (ALL) could improve treatment by identifying patients who need more or less intensive therapy. We have used a clone-specific polymerase chain reaction to detect rearranged immunoglobulin heavy-chain gene from the leukaemic clone, and quantified the clone by limiting dilution analysis. MRD was successfully quantified, by extracting DNA from marrow slides, from 88 of 181 children with ALL, who had total leucocyte counts below 100 x 10(9)/L at presentation and were enrolled in two clinical trials, in 1980-84 and 1985-89. Leukaemia was detected in the first remission marrow of 38 patients, in amounts between 6.7 x 10(-2) and 9.9 x 10(-7) cells; 26 of these patients relapsed. Of 50 patients with no MRD detected, despite study of 522-496,000 genomes, only 6 relapsed. The association between MRD detection and outcome was significant for patients in each trial. In the first trial, patients relapsed at all levels of detected MRD, whereas in the later trial, in which treatment was more intensive and results were better, the extent of MRD was closely related to the probability of relapse (5 of 5 patients with > 10(-3) MRD, 4 of 10 with 10(-3) to 2 x 10(-5), 0 of 3 with levels below 2 x 10(-5), and 2 of 26 with no MRD detected). Early quantification of leukaemic cells after chemotherapy may be a successful strategy for predicting outcome and hence individualizing treatment in childhood ALL, because the results indicate both in-vivo drug sensitivity of the leukaemia and the number of leukaemic cells that remain to be killed by post-induction therapy.