ArticlePDF Available

Quantification of minimal residual disease in T-lineage acute lymphoblastic with the TAL-1 deletion using a standardized real-time PCR assay

Authors:

Abstract and Figures

Hematologic relapse remains the greatest obstacle to the cure of children with acute lymphoblastic leukemia (ALL). Recent studies have shown that patients with increased risk of relapse can be identified by measuring residual leukemic cells, called minimal residual disease (MRD), during clinical remission. Current PCR methods, however, for measuring MRD are cumbersome and time-consuming. To improve and simplify MRD assessment, we developed a real-time quantitative PCR (RQ-PCR) assay for detection of leukemic cells that harbor the TAL-1 deletion. We studied serial dilutions of leukemic DNA and found the assay had a sensitivity of detection of one leukemic cell among 100,000 normal cells. We then investigated 23 samples from eight children with ALL in clinical remission. We quantified residual leukemic cells by using the TAL-1 RQ-PCR assay and by using limiting dilution analysis. In 17 samples, both methods detected MRD levels > or =0.001%. The percentages of leukemic cells measured by the two methods correlated well (r2 = 0.926). In the remaining six samples, both methods detected fewer than 0.001% leukemic cells. We conclude the TAL-1 RQ-PCR assay can be used for rapid, sensitive and accurate assessment of MRD in T-lineage ALL with the TAL-1 deletion.
Content may be subject to copyright.
Leukemia (2001) 15, 166–170
2001 Nature Publishing Group All rights reserved 0887-6924/01 $15.00
www.nature.com/leu
BIO-TECHNICAL METHODS SECTION (BTS)
BTS
Leukemia
Quantification of minimal residual disease in T-lineage acute lymphoblastic leukemia
with the TAL-1 deletion using a standardized real-time PCR assay
X Chen
1
, Q Pan
1
, P Stow
1
, FG Behm
2
, R Goorha
3,4
, C-H Pui
1,2,4
and GAM Neale
1,2,4
Departments of
1
Hematology/Oncology,
2
Pathology, and
3
Virology and Molecular Biology, St Jude Children’s Research Hospital, Memphis,
TN; and
4
University of Tennessee College of Medicine, Memphis, TN, USA
Hematologic relapse remains the greatest obstacle to the cure
of children with acute lymphoblastic leukemia (ALL). Recent
studies have shown that patients with increased risk of relapse
can be identified by measuring residual leukemic cells, called
minimal residual disease (MRD), during clinical remission. Cur-
rent PCR methods, however, for measuring MRD are cumber-
some and time-consuming. To improve and simplify MRD
assessment, we developed a real-time quantitative PCR (RQ-
PCR) assay for detection of leukemic cells that harbor the
TAL-
1
deletion. We studied serial dilutions of leukemic DNA and
found the assay had a sensitivity of detection of one leukemic
cell among 100 000 normal cells. We then investigated 23
samples from eight children with ALL in clinical remission. We
quantified residual leukemic cells by using the
TAL-1
RQ-PCR
assay and by using limiting dilution analysis. In 17 samples,
both methods detected MRD levels 0.001%. The percentages
of leukemic cells measured by the two methods correlated well
(
r
2
0.926). In the remaining six samples, both methods
detected fewer than 0.001% leukemic cells. We conclude the
TAL-1
RQ-PCR assay can be used for rapid, sensitive and
accurate assessment of MRD in T-lineage ALL with the
TAL-1
deletion.
Leukemia
(2001) 15, 166–170.
Keywords: real-time PCR quantification; MRD; ALL;
TAL-1
Introduction
With a cure rate of 80% in childhood acute lymphoblastic
leukemia (ALL), great effort has been expended to precisely
define risk assessment so that intensive treatment is given to
children at high risk, whereas less toxic therapy is reserved
for those with excellent prognosis.
1,2
Several recent prospective studies have shown that the level
of leukemic cells during clinical remission, known as minimal
residual disease (MRD), is predictive of patient outcome.
3–5
The consensus from MRD studies is that clinical evaluation of
MRD should improve the overall cure rate of ALL by ident-
ifying the patients with highest risk of relapse thereby allowing
timely therapeutic intervention.
6,7
The most common method for quantifying leukemic cells
in ALL is polymerase chain reaction (PCR) amplification of
leukemia-specific targets.
6,8
Of the various PCR techniques,
limiting dilution analysis
9–11
and comparative hybridization
Correspondence: G Neale, Department of Hematology/Oncology, St
Jude Children’s Research Hospital, 332 N Lauderdale, Memphis TN
38105, USA; Fax: 901 495 3749
Received 31 July 2000; accepted 26 September 2000
methods
12–14
have been used the most. However, these
methods are cumbersome and time-consuming. Therefore,
PCR-based methods must be simplified and improved before
they can be routinely applied in the clinical setting. With that
goal in mind, we investigated the ability of real-time quantitat-
ive PCR (RQ-PCR) to quantify leukemic cells harboring the
TAL-1 deletion.
15
RQ-PCR permits accurate quantification by
measuring PCR products during the exponential phase of the
PCR reaction,
16
and reduces time and labor by eliminating
post-PCR processing. The TAL-1 deletion is present in
approximately 25% of cases of T-lineage ALL, but it is not
found in normal T cells.
15,17,18
Therefore, the TAL-1 deletion
provides a leukemia-specific target for MRD assessment in a
significant number of patients with ALL. We developed a Taq-
Man RQ-PCR assay for detection and quantification of leu-
kemic cells harboring the TAL-1 deletion. We applied this
assay to analyze clinical samples from ALL patients. To assess
the accuracy of our TAL-1 RQ-PCR assay, we compared RQ-
PCR data with those obtained by limiting dilution analysis.
Materials and methods
Cells and cell culture
Bone marrow and peripheral blood samples were obtained
from patients diagnosed with T-lineage ALL. Diagnostic DNA
samples were tested by PCR amplification for SIL
db1
-TAL
db1
and SIL
db1
-TAL
db2
deletions using the primers described.
19
Each case in this report had a SIL
db1
-TAL
db1
deletion. All
samples analyzed by RQ-PCR and limiting dilution were
obtained during clinical remission. Patient samples were
obtained with appropriate consent, and with the approval of
the institutional review board. Normal peripheral blood cells
were obtained from healthy volunteers. Mononuclear cells
were isolated by centrifugation on a density gradient
(Lymphoprep; Life Technologies, Gaithersburg, MD, USA) and
washed three times in phosphate buffered saline. The human
T-lineage ALL cell line CEM-C7 was obtained from Dr Dario
Campana (Department of Hematology/Oncology, St Jude
Children’s Research Hospital). Cells were maintained in
RPMI-1640 culture medium (Life Technologies) containing
10% fetal bovine serum (Hyclone, Logan, UT, USA),
2m
ML
-glutamine and antibiotics, and incubated at 37°Cina
humidified atmosphere containing 5% CO
2
.
Real-time MRD assay using
TAL-1
deletion
X Chen
et al
167
Quantification of leukemic cells by RQ-PCR
DNA samples were prepared by using the QIAamp blood kit
(Qiagen, Valencia, CA, USA). To construct standard curves for
quantification of leukemic cells with the TAL-1 deletion, DNA
from the CEM-C7 cell line was diluted into DNA prepared
from normal peripheral blood mononuclear cells (PBMC).
Each DNA sample was analyzed in triplicate using an ABI
Prism 7700 sequence detection system. PCR reactions con-
tained 600 ng of DNA, 1 ×TaqMan Universal Master Mix
(Perkin Elmer, Foster City, CA, USA), 0.1
M
of TAL-1 TaqMan
probe, and 0.25
M
TAL-1 TaqMan amplification primers
(Table 1) in a total volume of 50
l. The amplification con-
ditions consisted of an initial incubation at 95°C for 10 min,
then 50 cycles consisting of 15 s at 95°C and 60 s at 60°C.
To control for the number of amplifiable genomes in the
assay, we tested each sample in a parallel TaqMan assay to
quantify the number of N-ras copies. PCR reactions contained
200 ng of DNA, 1 ×TaqMan Universal Master Mix (Perkin
Elmer, Foster City, CA, USA), 0.1
M
of N-ras TaqMan probe,
and 0.25
M
N-ras TaqMan amplification primers (Table 1) in
a total volume of 50
l. The amplification conditions con-
sisted of an initial incubation at 95°C for 10 min, then 50
cycles consisting of 15 s at 95°C and 60 s at 60°C. An N-ras
standard curve was constructed by making serial dilutions of
normal PBMC DNA in water. The number of amplifiable gen-
omes in each sample varied by less than two-fold (data not
shown). Consequently, we did not adjust our TAL-1 TaqMan
data.
Quantification of leukemic cells with TAL-1 deletion
by limiting dilution analysis
A two-round PCR assay was used to detect a single copy of
the TAL-1 deletion among 10
5
normal genomes. In the first
round, 600 ng of genomic DNA was amplified for 25 cycles
by using the SIL-db-5primer and the TAL-db1–3primer
(Table 1) in a 50
l reaction. In the second round, the first
round amplification mixture was diluted 1:100, and 1
l was
amplified for 45 cycles by using the same amplification pri-
mers in a 25
l reaction. Amplification mixtures contained 1
Table 1 Sequences of RQ-PCR TaqMan probes and amplification primers
Application Oligonucleotide Oligonucleotide sequence
name
TAL-1
RQ-PCR
TAL-1
probe ctctttcacataccttagctcagatgatacccaa
a
SIL
-bp-5gagctagtgggagaaattaagcagtc
b
TAL
-bp1-3ccgcccacagttctcatga
c
N-ras
RQ-PCR
N-ras
probe tctcatggcactgtactcttcttggtccagct
d
N-ras
-5aacctgtttgttggacatactggata
e
N-ras
-3cgcctgtcctcatgtattggt
e
TAL-1
limiting
SIL
-db-5aaggggagctagtgggagaaa
f
dilution analysis
TAL
-dbl-3agagcctgtcgccaagaa
f
a
The
TAL-1
TaqMan probe spans part of exon 1b and the following intron within the
SIL
locus (accession Y07540). The 5nucleotide is
located 107 bp upstream of the
SIL
-db1 breakpoint. The oligonucleotide was modified with 6-FAM reporter dye at 5-end and TAMRA
quencher dye at 3end.
b
The
SIL
-bp-5primer is located within exon 1b of the
SIL
locus; the 5nucleotide is 177 bp upstream of the
SIL
-db1 breakpoint.
c
The
TAL
-bp1–3primer is located within exon 1b of the
TAL-1
locus (accession S53698); the 5nucleotide is 77 bp downstream of the
TAL
-db1 breakpoint.
d
The
N-ras
TaqMan is located within exon 2 of the N-ras gene (accession L00041). The oligonucleotide was modified with VIC reporter
dye at 5-end and TAMRA quencher dye at 3-end.
e
The
N-ras
primers amplify an 80 bp product within exon 2 of the human N-ras gene.
f
Sequences of these oligonucleotides are from Pongers-Willemse
et al.
19
Leukemia
×PCR buffer (PE Applied Biosystems), 1.5 mmMgCl
2
, 200
M
dNTPs, 0.25
mprimers, and 1 unit of AmpliTaq Gold (PE
Applied Biosystems). The amplification conditions consisted
of an initial incubation at 95°C for 10 min, then cycling
between 94°C for 20 s and 60°C for 40 s, followed by a final
incubation at 72°C for 5 min. PCR products were analyzed
by electrophoresis on a 3% agarose gel. MRD was quantified
by limiting dilution analysis using 10 replicates and Poisson
statistics.
20
The sensitivity of detection was 2 ×10
6
for every
sample tested.
Results
The TAL-1 deletion, present in approximately 25% of T-lin-
eage cases of ALL, arises via site-specific recombination
between the SIL and TAL-1 loci on chromosome 1p32. There
are three breakpoints tightly clustered within the SIL locus and
six breakpoints distributed within the TAL-1 locus.
21,22
The
two most frequently observed TAL-1 deletions involve the
SIL
db1
breakpoint rearranged to either the TAL
db1
or the TAL
db2
breakpoints (Figure 1a). In approximately 85% of cases, the
SIL
db1
breakpoint is rearranged with the TAL
db1
break-
point.
17,18
Using sequence data from the SIL locus (accession
Y07540) and the TAL-1 locus (accession S53698), we con-
structed amplification primers and a TaqMan probe for detec-
tion of the SIL
db1
-TAL
db1
deletion (Table 1, Figure 1b). We syn-
thesized the TaqMan probe complementary to sequences
within the SIL locus so that a single probe could potentially
be used with deletions arising from both major breakpoints
within the TAL-1 locus. Further, the 5-end of the probe was
oriented toward the TAL-1 locus to minimize non-specific
hydrolysis of the probe that might contribute to background
fluorescence.
We tested the sensitivity of the TAL-1 RQ-PCR assay using
the CEM-C7 cell line which contains the TAL-1 deletion. DNA
from the CEM-C7 cells was diluted in normal PBMC DNA
and analyzed by RQ-PCR. An excellent correlation was found
between the input of leukemic cells and the fluorescence
threshold (Ct) value (Figure 2). In every experiment, with input
DNA spanning five orders of magnitude, the correlation coef-
ficient always exceeded 0.99. At a dilution of 10
5
, there is
Real-time MRD assay using
TAL-1
deletion
X Chen
et al
168
Leukemia
Figure 1 Schematic design of the TAL-1 RQ-PCR assay. (a) The SIL and TAL-1 loci are separated by approximately 90 kb on chromosome
1. Breakpoints identified
21,22
within the SIL and TAL-1 loci are shown by vertical arrows; the large vertical arrows indicate the most frequently
used breakpoints. The relative positions of the TAL-1 TaqMan probe (horizontal bar) and primers (horizontal arrows) are indicated. (b) The
SIL
db1
-TAL
db1
deletion occurs in approximately 85% of cases with TAL-1 deletion. After deletion of intervening sequences, and insertion of N-
region nucleotides (N), the SIL and TAL-1 loci are juxtaposed. Amplification using the SIL-bp-5and TAL-bp1–3primers generates a PCR
product of approximately 220 bp in size. The TAL-1 TaqMan probe, containing reporter (R) and quencher (Q) dyes, (left panel) was synthesized
complementary to the SIL locus (see Table 1 for details). During amplification (right panel) the TaqMan probe is hydrolyzed, resulting in
increased fluorescence.
Figure 2 Real-time detection of leukemic cells with TAL-1
deletion. Standard curve showing the threshold cycle (Ct) vs the input
of CEM-C7 cells. Each point represents the mean of triplicate PCR
reactions. Three out of four replicates using input DNA equivalent to
a single CEM-C7 cell gave positive results.
an average of one genome per reaction. By using multiple
replicates of the 10
5
dilution, we found the RQ-PCR assay
could detect a single leukemic genome.
We then used the TAL-1 RQ-PCR assay to investigate MRD
in 23 bone marrow and peripheral blood samples from eight
patients during clinical remission. To determine the accuracy
of the TAL-1 RQ-PCR assay, we also investigated the same
samples by using a limiting dilution assay for the TAL-1
deletion. Of the 23 samples, 17 samples had greater than
0.001% leukemic cells by both methods (Figure 3). A good
correlation (r
2
=0.926) was found between the percentage of
leukemic cells determined by both methods. In the remaining
six samples, both methods detected fewer than 0.001% leu-
kemic cells. Two samples had undetectable leukemic cells by
both methods. In four samples, extremely low levels of MRD
(0.0002–0.0009% leukemic cells) were detected by the limit-
ing dilution method but not by the RQ-PCR assay. This appar-
ent difference in sensitivity could arise from the greater mass
of DNA being interrogated by the limiting dilution assay.
Discussion
We have developed a real-time PCR assay for detection and
quantification of leukemic cells that contain the TAL-1
deletion. This assay, applicable to a significant number of ALL
patients is rapid, sensitive, accurate, and less labor-intensive
than current methods for MRD assessment. This assay will
improve and simplify the assessment of MRD in patients with
T-lineage ALL.
Recent studies have shown the predictive value of MRD
assessment to identify ALL patients with increased risk of
relapse. However, current PCR methods are not optimal for
clinical evaluation of MRD. The most frequently used
methods for MRD assessment are comparative hybridization
and limiting dilution analysis.
6
Comparative hybridization is
time-consuming, taking 2–3 days to complete. Further, data
from these analyses are semi-quantitative. MRD levels are esti-
mated by comparison with standards, and it is difficult to
Real-time MRD assay using
TAL-1
deletion
X Chen
et al
169
Figure 3 Comparison of MRD data obtained by TAL-1 RQ-PCR
and limiting dilution analysis in clinical remission T-ALL samples. Six
samples had 10
5
leukemic cells by both methods.
establish an endpoint of amplification in which each standard
is still within the logarithmic phase. Limiting dilution analysis
provides quantitative MRD estimates in the absence of stan-
dards. However, the overall process is laborious due to the
multiple replicates analyzed, and the two rounds of PCR
amplification performed. Both methods require post-PCR
handling and analysis.
Competitive PCR is another method used to quantify mol-
ecular targets. Hosler and colleagues
23
recently reported the
use of that approach to quantify leukemic cells with the TAL-
1deletion. The authors synthesized an internal calibration
standard, a known quantity of which was added to PCR reac-
tions. After amplification, the number of leukemic cells was
calculated by comparing the intensity of the PCR products of
the endogenous TAL-1 deletion and the calibrator. This assay
was found to be precise and accurate, but analysis of multiple
dilutions of the unknown samples was often needed to
achieve the highest accuracy.
23
RQ-PCR using TaqMan technology affords significant
advantages over current PCR methods. First, the TaqMan
probe provides the sensitivity and specificity required for
MRD assessment. The probe hybridizes to a known sequence
between the amplification primers. It is cleaved, releasing
fluorescence into the reaction, only when the primers direct
amplification to that target sequence. Second, measurement
of fluorescence in real time during the PCR reaction permits
extrapolation of data back to the exponential phase of the
amplification, thus avoiding problems associated with other
comparative hybridization methods. Third, post-PCR pro-
cessing of samples is eliminated, thereby reducing the overall
time and labor of the assay.
We demonstrated that the TAL-1 RQ-PCR assay has a sensi-
tivity of detection of one leukemic cell among 10
5
normal
cells. This sensitivity is sufficient for discriminating ALL
patients with high and low risk of hematologic relapse.
7
Because data are obtained within hours of sample collection,
RQ-PCR analysis permits timely clinical intervention for
patients with increased risk of relapse. In addition, excellent
Leukemia
concordance was found between MRD estimates determined
by RQ-PCR and by limiting dilution analysis. The agreement
between methods provides confidence in the accuracy of the
TAL-1 RQ-PCR assay.
Other recent reports have confirmed the potential of RQ-
PCR to improve quantification of leukemic cells in ALL.
24–27
Those reports, however, used antigen-receptor gene
rearrangements for quantification of leukemic cells, a strategy
that requires leukemic-specific assays for each patient. An
additional way to simplify MRD assessment is to develop
assays that are applicable to a group of patients. This strategy
has been used to quantify MRD in patients with chronic mye-
logenous leukemia,
28,29
acute myelogenous leukemia
30–32
acute promyelocytic leukemia
33
and lymphoma.
34,35
In ALL,
the TAL-1 deletion is present in approximately 5% of total
patients and therefore our RQ-PCR assay will simplify the
assessment of MRD in those cases.
We have developed an RQ-PCR assay for quantification of
leukemic cells harboring the TAL-1 deletion. This standard
assay, applicable to a significant number of patients with ALL,
provides sensitive, accurate and timely assessment of MRD.
Application of this assay in clinical studies will improve
assessment of MRD in ALL, and should identify patients with
increased risk of relapse.
Acknowledgements
This work was supported in part by the following NIH grants:
R01 CA52259, R01 CA43237 and P30 CA21765; by a Center
of Excellence grant from the State of Tennessee, and by the
American Lebanese Syrian Associated Charities (ALSAC). We
thank Ms Sharon Naron for editorial assistance with the
manuscript.
References
1 Pui CH, Evans WE. Acute lymphoblastic leukemia. N Engl J Med
1998; 339: 605–615.
2 Pui CH. Acute lymphoblastic leukemia in children. Curr Opin
Oncol 2000; 12: 3–12.
3 Coustan-Smith E, Behm FG, Sanchez J, Boyett JM, Hancock ML,
Raimondi SC, Rubnitz JE, Rivera GK, Sandlund JT, Pui CH, Cam-
pana D. Immunological detection of minimal residual disease in
children with acute lymphoblastic leukaemia. Lancet 1998; 351:
550–554.
4 Cave 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 Cooper-
ative Group. N Engl J Med 1998; 339: 591–598.
5 van Dongen JJ, Seriu T, Panzer-Grumayer ER, Biondi A, Pongers-
Willemse MJ, Corral L, Stolz F, Schrappe M, Masera G, Kamps
WA, Gadner H, van Wering ER, Ludwig WD, Basso G, de Bruijn
MA, Cazzaniga G, Hettinger K, van der Does-van den Berg A, Hop
WC, Riehm H, Bartram CR. Prognostic value of minimal residual
disease in acute lymphoblastic leukaemia in childhood. Lancet
1998; 352: 1731–1738.
6 Foroni L, Harrison CJ, Hoffbrand AV, Potter MN. Investigation of
minimal residual disease in childhood and adult acute lympho-
blastic leukaemia by molecular analysis. Br J Haematol 1999; 105:
7–24.
7 Pui CH, Campana D. New definition of remission in childhood
acute lymphoblastic leukemia. Leukemia 2000; 14: 783–785.
8 Campana D, Pui CH. Detection of minimal residual disease in
acute leukemia: methodologic advances and clinical significance.
Blood 1995; 85: 1416–1434.
Real-time MRD assay using
TAL-1
deletion
X Chen
et al
170
Leukemia
9 Brisco MJ, Condon J, Sykes PJ, Neoh SH, Morley AA. Detection
and quantitation of neoplastic cells in acute lymphoblastic leu-
kaemia, by use of the polymerase chain reaction. Br J Haematol
1991; 79: 211–217.
10 Sykes PJ, Neoh SH, Brisco MJ, Hughes E, Condon J, Morley AA.
Quantitation of targets for PCR by use of limiting dilution. Biotech-
niques 1992; 13: 444–449.
11 Ouspenskaia MV, Johnston DA, Roberts WM, Estrov Z, Zipf TF.
Accurate quantitation of residual B-precursor acute lymphoblastic
leukemia by limiting dilution and a PCR-based detection system:
a description of the method and the principles involved. Leukemia
1995; 9: 321–328.
12 Yokota S, Hansen-Hagge TE, Ludwig WD, Reiter A, Raghavachar
A, Kleihauer E, Bartram CR. Use of polymerase chain reactions to
monitor minimal residual disease in acute lymphoblastic leukemia
patients. Blood 1991; 77: 331–339.
13 Biondi A, Yokota S, Hansen-Hagge TE, Rossi V, Giudici G, Maglia
O, Basso G, Tell C, Masera G, Bartram CR. Minimal residual dis-
ease in childhood acute lymphoblastic leukemia: analysis of
patients in continuous complete remission or with consecutive
relapse. Leukemia 1992; 6: 282–288.
14 Nizet Y, Van Daele S, Lewalle P, Vaerman JL, Philippe M, Ver-
mylen C, Cornu G, Ferrant A, Michaux JL, Martiat P. Long-term
follow-up of residual disease in acute lymphoblastic leukemia
patients in complete remission using clonogeneic IgH probes and
the polymerase chain reaction. Blood 1993; 82: 1618–1625.
15 Brown L, Cheng JT, Chen Q, Siciliano MJ, Crist W, Buchanan G,
Baer R. Site-specific recombination of the tal-1 gene is a common
occurrence in human T cell leukemia. EMBO J 1990; 9: 3343–
3351.
16 Heid CA, Stevens J, Livak KJ, Williams PM. Real time quantitative
PCR. Genome Res 1996; 6: 986–994.
17 Bash RO, Crist WM, Shuster JJ, Link MP, Amylon M, Pullen J,
Carroll AJ, Buchanan GR, Smith RG, Baer R. Clinical features and
outcome of T-cell acute lymphoblastic leukemia in childhood
with respect to alterations at the TAL1 locus: a Pediatric Oncology
Group study. Blood 1993; 81: 2110–2117.
18 Breit TM, Beishuizen A, Ludwig WD, Mol EJ, Adriaansen HJ, van
Wering ER, van Dongen JJ. tal-1 deletions in T-cell acute lym-
phoblastic leukemia as PCR target for detection of minimal
residual disease. Leukemia 1993; 7: 2004–2011.
19 Pongers-Willemse MJ, Seriu T, Stolz F, d’Aniello E, Gameiro P,
Pisa P, Gonzalez M, Bartram CR, Panzer-Grumayer ER, Biondi A,
San Miguel JF, van Dongen JJ. Primers and protocols for stan-
dardized detection of minimal residual disease in acute lympho-
blastic 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: 110–118.
20 Taswell C. Limiting dilution assays for the determination of immu-
nocompetent cell frequencies. I. Data analysis. J Immunol 1981;
126: 1614–1619.
21 Aplan PD, Lombardi DP, Reaman GH, Sather HN, Hammond GD,
Kirsch IR. Involvement of the putative hematopoietic transcription
factor SCL in T-cell acute lymphoblastic leukemia. Blood 1992;
79: 1327–1333.
22 Delabesse E, Bernard M, Landman-Parker J, Davi F, Leboeuf D,
Varet B, Valensi F, Macintyre EA. Simultaneous SIL-TAL1 RT-PCR
detection of all tal(d) deletions and identification of novel tal(d)
variants. Br J Haematol 1997; 99: 901–907.
23 Hosler GA, Bash RO, Bai X, Jain V, Scheuermann RH. Develop-
ment and validation of a quantitative polymerase chain reaction
assay to evaluate minimal residual disease for T-cell acute lym-
phoblastic leukemia and follicular lymphoma. Am J Pathol 1999;
154: 1023–1035.
24 Pongers-Willemse MJ, Verhagen OJ, Tibbe GJ, Wijkhuijs AJ, de
Haas V, Roovers E, van der Schoot CE, van Dongen JJ. Real-time
quantitative PCR for the detection of minimal residual disease in
acute lymphoblastic leukemia using junctional region specific
TaqMan probes. Leukemia 1998; 12: 2006–2014.
25 Eckert C, Landt O, Taube T, Seeger K, Beyermann B, Proba J,
Henze G. Potential of LightCycler technology for quantification of
minimal residual disease in childhood acute lymphoblastic leuke-
mia. Leukemia 2000; 14: 316–323.
26 Donovan JW, Ladetto M, Zou G, Neuberg D, Poor C, Bowers D,
Gribben JG. Immunoglobulin heavy-chain consensus probes for
real-time PCR quantification of residual disease in acute lym-
phoblastic leukemia. Blood 2000; 95: 2651–2658.
27 Kwan E, Norris MD, Zhu L, Ferrara D, Marshall GM, Haber M.
Simultaneous detection and quantification of minimal residual dis-
ease in childhood acute lymphoblastic leukaemia using real-time
polymerase chain reaction. Br J Haematol 2000; 109: 430–434.
28 Mensink E, van de Locht A, Schattenberg A, Linders E, Schaap N,
Geurts van Kessel A, De Witte T. Quantitation of minimal residual
disease in Philadelphia chromosome positive chronic myeloid leu-
kaemia patients using real-time quantitative RT-PCR. Br J Haema-
tol 1998; 102: 768–774.
29 Eder M, Battmer K, Kafert S, Stucki A, Ganser A, Hertenstein B.
Monitoring of BCR-ABL expression using real-time RT-PCR in
CML after bone marrow or peripheral blood stem cell transplan-
tation. Leukemia 1999; 13: 1383–1389.
30 Marcucci G, Livak KJ, Bi W, Strout MP, Bloomfield CD, Caligiuri
MA. Detection of minimal residual disease in patients with
AML1/ETO-associated acute myeloid leukemia using a novel
quantitative reverse transcription polymerase chain reaction assay.
Leukemia 1998; 12: 1482–1489.
31 Sugimoto T, Das H, Imoto S, Murayama T, Gomyo H, Chakraborty
S, Taniguchi R, Isobe T, Nakagawa T, Nishimura R, Koizumi T.
Quantitation of minimal residual disease in t(8;21)-positive acute
myelogenous leukemia patients using real-time quantitative RT-
PCR. Am J Hematol 2000; 64: 101–106.
32 Fujimaki S, Funato T, Harigae H, Imaizumi M, Suzuki H, Kaneko
Y, Miura Y, Sasaki T. A quantitative reverse transcriptase poly-
merase chain reaction method for the detection of leukaemic cells
with t(8;21) in peripheral blood. Eur J Haematol 2000; 64: 252–
258.
33 Cassinat B, Zassadowski F, Balitrand N, Barbey C, Rain JD, Fenaux
P, Degos L, Vidaud M, Chomienne C. Quantitation of minimal
residual disease in acute promyelocytic leukemia patients with
t(15;17) translocation using real-time RT-PCR. Leukemia 2000; 14:
324–328.
34 Luthra R, McBride JA, Cabanillas F, Sarris A. Novel 5exonucle-
ase-based real-time PCR assay for the detection of
t(14;18)(q32;q21) in patients with follicular lymphoma. Am J
Pathol 1998; 153: 63–68.
35 Olsson K, Gerard CJ, Zehnder J, Jones C, Ramanathan R, Reading
C, Hanania EG. Real-time t(11;14) and t(14;18) PCR assays pro-
vide sensitive and quantitative assessments of minimal residual
disease (MRD). Leukemia 1999; 13: 1833–1842.
... However, in group IV, patients with SIL-TALI1 were characterized by higher ESF (90%) in comparison to others without fusion (57.1%) [8]. TAL1 gene deletion is a common abnormality, observed with a frequency of 25% of patients with T-ALL [65]. Wang et al. achieved similar results. ...
... SIL-TAL1 deletion occurred in every fifth patient with T-ALL [66]. Both Chen and Wang suggested that TAL-1 gene deletion detection may be useful in assessing MRD in T-ALL patients [65,66]. In another study, deletion of TAL1 was observed in 3 out of 39 patients under 18 years of age and was associated with a higher WBC count [67]. ...
Article
Full-text available
Acute lymphoblastic leukaemia (ALL) is a relevant form of childhood neoplasm, as it accounts for over 80% of all leukaemia cases. T-cell ALL constitutes a genetically heterogeneous cancer derived from T-lymphoid progenitors. The diagnosis of T-ALL is based on morphologic, immunophenotypic, cytogenetic, and molecular features, thus the results are used for patient stratification. Due to the expression of surface and intracellular antigens, several subtypes of T-ALL can be distinguished. Although the aetiology of T-ALL remains unclear, a wide spectrum of rearrangements and mutations affecting crucial signalling pathways has been described so far. Due to intensive chemotherapy regimens and supportive care, overall cure rates of more than 80% in paediatric T-ALL patients have been accomplished. However, improved knowledge of the mechanisms of relapse, drug resistance, and determination of risk factors are crucial for patients in the high-risk group. Even though some residual disease studies have allowed the optimization of therapy, the identification of novel diagnostic and prognostic markers is required to individualize therapy. The following review summarizes our current knowledge about genetic abnormalities in paediatric patients with T-ALL. As molecular biology techniques provide insights into the biology of cancer, our study focuses on new potential therapeutic targets and predictive factors which may improve the outcome of young patients with T-ALL.
... 134 The detection of TAL1 deletions at the DNA level has already been described. [143][144][145] A recent report described a TaqManbased RQ-PCR method for the detection of TAL1 deletions at the DNA level in T-ALL patients. 145 In that report, the forward primer and probe were positioned in SIL exon 1b (and part of the following intron) and the reverse primer was located in TAL1 exon 1b. ...
... [143][144][145] A recent report described a TaqManbased RQ-PCR method for the detection of TAL1 deletions at the DNA level in T-ALL patients. 145 In that report, the forward primer and probe were positioned in SIL exon 1b (and part of the following intron) and the reverse primer was located in TAL1 exon 1b. Using the CEM cell line, a sensitivity of 10 À5 could be obtained, which is equivalent to a single leukemic genome. ...
... As indicated in Table 1, this includes studies showing that the upregulation of AnxA6 is an indicator of the progression of ovarian carcinomas [107], women's thyroid cancer [108], polycystic ovarian syndrome [109], pancreatic cancer [96] and esophageal adenocarcinoma [110]. Other studies have unambiguously demonstrated that AnxA6 may be useful to detect minimal residual disease in B-lineage acute lymphoblastic leukemia [111], the progression of melanomas [112] and squamous cervical cancer carcinogenesis [113]. Meanwhile, some studies have shown that AnxA6 is downregulated in the highly malignant forms of gastric cancer [114], hepatocellular carcinomas [115], cervical cancer [116] and breast cancer [22]. ...
Article
Full-text available
The calcium (Ca2+)-dependent membrane-binding Annexin A6 (AnxA6), is a multifunctional, predominantly intracellular scaffolding protein, now known to play relevant roles in different cancer types through diverse, often cell-type-specific mechanisms. AnxA6 is differentially expressed in various stages/subtypes of several cancers, and its expression in certain tumor cells is also induced by a variety of pharmacological drugs. Together with the secretion of AnxA6 as a component of extracellular vesicles, this suggests that AnxA6 mediates distinct tumor progression patterns via extracellular and/or intracellular activities. Although it lacks enzymatic activity, some of the AnxA6-mediated functions involving membrane, nucleotide and cholesterol binding as well as the scaffolding of specific proteins or multifactorial protein complexes, suggest its potential utility in the diagnosis, prognosis and therapeutic strategies for various cancers. In breast cancer, the low AnxA6 expression levels in the more aggressive basal-like triple-negative breast cancer (TNBC) subtype correlate with its tumor suppressor activity and the poor overall survival of basal-like TNBC patients. In this review, we highlight the potential tumor suppressor function of AnxA6 in TNBC progression and metastasis, the relevance of AnxA6 in the diagnosis and prognosis of several cancers and discuss the concept of therapy-induced expression of AnxA6 as a novel mechanism for acquired resistance of TNBC to tyrosine kinase inhibitors.
... Standard DNA samples collected at the first clinical visit were serially diluted with DNA from a single healthy subject to 10 -5 -10 -1 copies and then amplified by RQ-PCR, with N-ras as the internal reference. 7 Detection was performed following guidelines established by the European Study Group on MRD Detection in ALL (ESG-MRD-ALL) 8 and by our previous study. 9 Detection results were defined as quantifiable, positive but unquantifiable, and negative respectively according to this guideline. ...
Article
Full-text available
Importance: By demonstrating with TEL-AML1, this study indicated that mRNAs transcribed from fusion genes are ideal targets for minimal residual disease (MRD) monitoring in childhood acute lymphoblastic leukemia, and that different thresholds are needed to apply them into the risk stratification. Objective: TEL-AML1 expression was measured at three time points to 1) determine cut-off values for predicting acute lymphoblastic leukemia (ALL) relapse; 2) investigate the prognostic value of this method and how well the results at these time points correlated; 3) determine the correlation between MRD levels assessed using this marker and that determined by immunoglobulin/T-cell receptor (Ig/TCR) rearrangement detection. Methods: TEL- AML1 expression in 62 children with ALL was quantitated by real-time quantitative PCR at day 15, day 33, and month 3. The relationship between patient outcome and TEL-AML1 level was analyzed at each time point. The correlation between the MRD levels determined by TEL-AML1 or Ig/TCR rearrangements was also analyzed. Results: For day 33, 6.68 TEL-AML1 copies/104ABL copies was determined to be the best cut-off value. Higher levels were correlated with relapse (P = 0.001). For day 15 and month 3, the best cut-off values were 336.5 and 0.85 copies/104ABL copies respectively; patients with higher expression levels had lower RFSs (day 15: P = 0.027; month 3: P = 0.023). For days 15 and 33, MRD levels assessed using TEL-AML1 or Ig/TCR rearrangements were strongly correlated [Spearman rank correlation coefficient (ρ) = 0.729 (day 15), 0.719 (day 33); P < 0.001 (both)], and both methods were equally effective at predicting relapse. At month 3, there was moderate correlation between the results derived from the two markers (ρ = 0.418, P = 0.003); however, receiver operating characteristic curve analysis showed that TEL-AML1 was a better prognostic marker. Interpretation: TEL-AML1 is an effective marker for MRD assessment and relapse prediction in children with ALL.
... The potential for AnxA6 as a biomarker for chronic diseases has been investigated in a number of studies. These include studies demonstrating that detection of AnxA6 may be useful to discriminate acute (AnxA6-low) from chronic (AnxA6-high) myocarditis (48), as a serum biomarker for esophageal adenocarcinoma (49) and to detect minimal residual disease in Blineage acute lymphoblastic leukemia (50). Given that assays to reliably detect the development of resistance against EGFR-TKIs remain a major challenge, it seems probable that detection of AnxA6 along with cholesterol or other lipid raft markers before and after treatment with these drugs may be a reliable indicator of whether or not tumor cells can develop resistance to these drugs. ...
Article
Full-text available
The epidermal growth factor receptor (1) is a major oncogene in triple negative breast cancer (TNBC), but the use of EGFR targeted tyrosine kinase inhibitors (TKI) and therapeutic monoclonal antibodies is associated with poor response and acquired resistance. Understanding the basis for the acquired resistance to these drugs and identifying biomarkers to monitor the ensuing resistance remains a major challenge. We previously showed that reduced expression of annexin A6 (AnxA6), a calcium dependent membrane binding tumor suppressor, not only promoted the internalization and degradation of activated EGFR but also sensitized TNBC cells to EGFR TKIs. Here, we demonstrate that prolong (>3 days) treatment of AnxA6-low TNBC cells with lapatinib led to AnxA6 up-regulation and accumulation of cholesterol in late endosomes. Basal ERK1/2 activation was EGFR-independent and significantly higher in lapatinib resistant MDA-MB-468 (LAP-R) cells. These cells were more sensitive to cholesterol depletion than untreated control cells. Inhibition of lapatinib-induced up-regulation of AnxA6 by RNA interference (A6sh) or withdrawal lapatinib from LAP-R cells not only reversed the accumulation of cholesterol in late endosomes but also led to enrichment of plasma membranes with cholesterol, restored EGFR-dependent activation of ERK1/2 and sensitized the cells to lapatinib. These data suggest that lapatinib induced AnxA6 expression and accumulation of cholesterol in late endosomes constitute an adaptive mechanism for EGFR expressing TNBC cells to overcome prolong treatment with EGFR-targeted TKIs, and can be exploited as an option to inhibit and/or monitor the frequently observed acquired resistance to these drugs.
Article
Introduction: Relapse remained the major obstacle to improving the prognosis of children with acute lymphoblastic leukemia (ALL). This study aimed to investigate the changing patterns of Ig/TCR gene rearrangements between diagnosis and relapse and the clinical relevance and to explore the mechanism of leukemic relapse. Methods: Clonal Ig/TCR gene rearrangements were screened by multiplex PCR amplification in 85 paired diagnostic and relapse bone marrow (BM) samples from children with ALL. The new rearrangements presented at relapse were quantitatively assessed by the RQ-PCR approach targeting the patient-specific junctional region sequence in 19 diagnostic samples. The relapse clones were further back-traced to diagnostic and follow-up BM samples from 12 patients. Results: Comparison of Ig/TCR gene rearrangements between diagnosis and relapse showed that 40 (57.1%) B-ALL and 5 (33.3%) T-ALL patients exhibited a change from diagnosis to relapse, and 25 (35.7%) B-ALL patients acquired new rearrangements at relapse. The new relapse rearrangements were present in 15 of the 19 (78.9%) diagnostic samples as shown by RQ-PCR, with a median level of 5.26 × 10-2 . The levels of minor rearrangements correlated with B immunophenotype, WBC counts, age at diagnosis, and recurrence time. Furthermore, back-tracing rearrangements in 12 patients identified three patterns of relapse clone dynamics, which suggested the recurrence mechanisms not only through clonal selection of pre-existing subclones but also through an ongoing clonal evolution during remission and relapse. Conclusion: Backtracking Ig/TCR gene rearrangements in relapse clones of pediatric ALL revealed complex patterns of clonal selection and evolution for leukemic relapse.
Article
Minimal residual disease (MRD) monitoring is of prognostic importance in childhood acute lymphoblastic leukemia (ALL). The detection of immunoglobulin and T-cell receptor gene rearrangements by real-time quantitative PCR (RT-PCR) is considered the gold standard for this evaluation. However, more accessible methods also show satisfactory performance. This study aimed to compare MRD analysis by four-color flow cytometry (FC) and qualitative standard PCR on days 35 and 78 of chemotherapy and to correlate these data with patients’ clinical characteristics. Forty-two children with a recent diagnosis of ALL, admitted to a public hospital in Brazil for treatment in accordance with the Brazilian Childhood Cooperative Group for ALL Treatment (GBTLI LLA-2009), were included. Bone marrow samples collected at diagnosis and on days 35 and 78 of treatment were analyzed for the immunophenotypic characterization of blasts by FC and for the detection of clonal rearrangements by standard PCR. Paired analyses were performed in 61/68 (89.7%) follow-up samples, with a general agreement of 88.5%. Disagreements were resolved by RT-PCR, which evidenced one false-negative and four false-positive results in FC, as well as two false-negative results in PCR. Among the prognostic factors, a significant association was found only between T-cell lineage and MRD by standard PCR. These results show that FC and standard PCR produce similar results in MRD detection of childhood ALL and that both methodologies may be useful in the monitoring of disease treatment, especially in regions with limited financial resources.
Article
Full-text available
Introduction: Minimal residual disease is an important independent prognostic factor that can identify poor responders among patients with acute lymphoblastic leukemia. Objective: The aim of this study was to analyze minimal residual disease using immunoglobulin (Ig) and T-cell receptor (TCR) gene rearrangements by conventional polymerase chain reaction followed by homo-heteroduplex analysis and to compare this with real-time polymerase chain reaction at the end of the induction period in children with acute lymphoblastic leukemia. Methods: Seventy-four patients diagnosed with acute lymphoblastic leukemia were enrolled. Minimal residual disease was evaluated by qualitative polymerase chain reaction in 57 and by both tests in 44. The Kaplan-Meier and multivariate Cox methods and the log-rank test were used for statistical analysis. Results: Nine patients (15.8%) were positive for minimal residual disease by qualitative polymerase chain reaction and 11 (25%) by real-time polymerase chain reaction considering a cut-off point of 1×10(-3) for precursor B-cell acute lymphoblastic leukemia and 1×10(-2) for T-cell acute lymphoblastic leukemia. Using the qualitative method, the 3.5-year leukemia-free survival was significantly higher in children negative for minimal residual disease compared to those with positive results (84.1%±5.6% versus 41.7%±17.3%, respectively; p-value=0.004). There was no significant association between leukemia-free survival and minimal residual disease by real-time polymerase chain reaction. Minimal residual disease by qualitative polymerase chain reaction was the only variable significantly correlated to leukemia-free survival. Conclusion: Given the difficulties in the implementation of minimal residual disease monitoring by real-time polymerase chain reaction in most treatment centers in Brazil, the qualitative polymerase chain reaction strategy may be a cost-effective alternative.
Article
Full-text available
The polymerase chain reaction (PCR) has revolutionized the detection of DNA and RNA, allowing the detection of as little as a single copy of a given sequence. PCR is largely used in immunogenetics studies for a variety of purposes, such as the detection of viral load in infectious diseases, identification of markers that can predict survival of patients undergoing organ transplantation1 and, in general, to assess the expression of immune related genes. Theoretically, there is a quantitative relationship between the amount of starting material and the PCR product at any cycle. However, it is a common experience for replicate reactions to yield different amounts of PCR product. Furthermore, at the end of a PCR reaction in which the number of cycles are empir- ically pre-determined, different amounts of starting material can yield similar amounts of amplification product due to the consumption of the reagents. As a consequence, conventional PCR cannot be considered a quantitative assay and the results are usually expressed in terms of positivity/negativity. A semi- quantitative evaluation is possible on the basis of the appear- ance of the electrophoretic band (weak/strong positivity), but this approach is highly subjective and the sensitivity is very low. To address the issue of PCR product quantitation, methods such as competitive PCR,2 sequence analysis gene expression (SAGE) 3 and high-throughput microarray 4 have been pro- posed. These techniques are cumbersome, time consuming, and require multiple manipulations of the samples, thus increasing the risk of carry-over contamination. Quantitative real-time PCR (qr-PCR) allows a highly sensitive quantification of transcriptional levels of the gene of interest in a few hours with minimal handling of the samples.5,6 Here we describe the principles and the technical aspects of this tech- nique, and review its main applications in the field of immuno- genetics.
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
The SCL gene, initially discovered at the site of a translocation breakpoint associated with the development of a stem cell leukemia, encodes a protein that contains the highly conserved basic helix-loop-helix (bHLH) motif found in a large array of eukaryotic transcription factors. Recently, we have described a nonrandom, site-specific SCL rearrangement in several T-cell acute lymphoblastic leukemia (ALL) cell lines that juxtaposes SCL with a distinct transcribed locus, SIL. The SIL/SCL rearrangement was found in leukemic blasts from 11 of 70 (16%) newly diagnosed T-cell ALL patients, a prevalence substantially higher than that of the t(11;14) translocation, which has previously been reported as the most frequent nonrandom chromosomal abnormality in T-cell ALL. We did not detect the SIL/SCL rearrangement in the leukemic blasts from 30 patients with B-cell precursor ALL, indicating that the rearrangement was specific for T-cell ALL. Analysis of RNA from these patients indicated that an SIL/SCL fusion mRNA was formed, joining SIL and SCL in a head-to-tail fashion. The fusion occurs in the 5' untranslated region (UTR) of both genes, preserving the SCL coding region. The net result of this rearrangement is that SCL mRNA expression becomes regulated by the SIL promoter, leading to inappropriate SCL expression. The resultant inappropriate expression of this putative transcription factor may then contribute to leukemic transformation in T-cell ALL.
Article
T-cell receptor (TCR) delta gene rearrangements are observed in more than 80% of acute lymphoblastic leukemia (ALL) patients. Moreover, a preferential usage of specific genetic elements has been shown in different ALL subtypes: V delta 1 DJ delta 1 rearrangements predominate in T-ALL, while most B-precursor ALLs show a recombination of V delta 2 to D delta 3. Recently we have proposed a strategy for the detection of minimal residual disease (MRD) based on the isolation of clonospecific probes following the in vitro amplification of V delta 1 DJ delta 1 junctions by polymerase chain reaction (PCR) and now have adapted this method to the preparation of specific V delta 2 D delta 3 fragments. In the present study, clonospecific probes were generated from 11 T-ALL and 16 cALL patients (21 children, 6 adults). The sensitivity of these 27 probes in detecting residual leukemia cells varied between 10(-4) to 10(-6) as determined by semiquantitative evaluation of dilution experiments. PCR analysis of 55 bone marrow (BM) and peripheral blood (PB) samples obtained from the 27 patients during complete clinical remission showed the following results: (1) Evidence for MRD was obtained in the BM of all patients (eight of eight) investigated 2 to 6 months after remission induction and also in 6 of 11 cases on maintenance therapy 7 to 19 months after diagnosis. (2) In contrast, all patients but one (10 of 11) analyzed 6 to 41 months after the termination of treatment lacked apparent evidence for leukemia DNA; the exception was a girl exhibiting 10(-4) to 10(-5) residual cells in her PB 5.5 years after diagnosis. (3) Longitudinal analysis in nine patients disclosed marked individual differences in the intervals between achievement of clinical remission and complete eradication of the leukemia cell clone. (4) Differences in the duration of MRD were not associated with distinct clinical-hematologic features. (5) Detection of residual disease by PCR proceeded clinical relapse in two cases.
Article
Tumor-related immunoglobulin heavy-chain (IgH) rearrangements are markers for polymerase chain reaction (PCR) detection of minimal residual disease (MRD) in B-cell malignancies. Nested PCR with patient IgH allele-specific oligonucleotide primers can detect 1 tumor cell in 104 to 106 normal cells. In childhood acute lymphoblastic leukemia (ALL), persistence of PCR-detectable disease is associated with increased risk of relapse. The clinical significance of qualitative PCR data can be limited, however, because patients can harbor detectable MRD for prolonged periods without relapse. Recent studies indicate that a quantitative rise in tumor burden identifies patients who are at high risk for relapse. Therefore, an efficient and reliable PCR method for MRD quantification is needed for ALL patients. We have developed a real-time PCR method to quantify MRD with IgH VH gene family consensus fluorogenically labeled probes. With this method, a small number of probes can be used to quantify MRD in a large number of different patients. The assay was found to be both accurate and reproducible over a wide range and capable of detecting approximately 1 tumor cell in 5 × 104 normal cells. We demonstrate that this methodology can discriminate between patients with persistence of MRD who relapse and those who do not. This technique is generally applicable to B-cell malignancies and is currently being used to quantify MRD in a number of prospective clinical studies at our institution.
Article
The SCL gene, initially discovered at the site of a translocation breakpoint associated with the development of a stem cell leukemia, encodes a protein that contains the highly conserved basic helix-loop- helix (bHLH) motif found in a large array of eukaryotic transcription factors. Recently, we have described a nonrandom, site-specific SCL rearrangement in several T-cell acute lymphoblastic leukemia (ALL) cell lines that juxtaposes SCL with a distinct transcribed locus, SIL. The SIL/SCL rearrangement was found in leukemic blasts from 11 of 70 (16%) newly diagnosed T-cell ALL patients, a prevalence substantially higher than that of the t(11;14) translocation, which has previously been reported as the most frequent nonrandom chromosomal abnormality in T- cell ALL. We did not detect the SIL/SCL rearrangement in the leukemic blasts from 30 patients with B-cell precursor ALL, indicating that the rearrangement was specific for T-cell ALL. Analysis of RNA from these patients indicated that an SIL/SCL fusion mRNA was formed, joining SIL and SCL in a head-to-tail fashion. The fusion occurs in the 5′ untranslated region (UTR) of both genes, preserving the SCL coding region. The net result of this rearrangement is that SCL mRNA expression becomes regulated by the SIL promoter, leading to inappropriate SCL expression. The resultant inappropriate expression of this putative transcription factor may then contribute to leukemic transformation in T-cell ALL.
Article
We sequentially studied bone marrow (BM) samples of 25 patients in complete remission of an acute lymphoblastic leukemia (ALL) using a simplified polymerase chain reaction (PCR) strategy (direct use of the PCR product as a clonogenic probe recognizing rearranged Ig heavy chain sequences) as a first approach. BM aspirates were serially investigated after obtention of a complete response. When sensitivity was less than 1:10(4), the PCR fragment was sequenced and a specific oligonucleotide was synthetized and used as a probe (five cases). Cases in which minimal residual disease (MRD) became undetectable were cross- controlled using either TCR delta rearrangement or a specific translocation to circumvent the problem of false-negative results arising from clonal evolution. The median follow-up was 30 months (3 to 51 months). Within the first 3 months of complete remission, MRD was detectable in 22 of 23 investigated patients and remained so in 19 of 21 patients examined at 6 months, regardless of the long-term clinical outcome. In patients remaining in complete remission at 30 months or more, two patterns of MRD emerged during the follow-up. Either it continuously decreased to ultimately become undetectable (five patients) or remained detectable (five patients) with an increase after discontinuation of treatment in two. In the eight patients who relapsed, MRD persisted throughout the clinical course, and eventually increased 3 to 12 months before relapse was clinically detectable. In one case, clonal evolution of the VDJ heavy chain region was observed and recurrence of MRD shown by the use of TCR delta rearrangement as a control. We conclude that the use of this simplified methodology is a valuable tool for the follow-up of MRD in a majority of ALL patients, though in a few cases, sequencing needs to be performed to achieve a relevant sensitivity. The possibility of clonal evolution requires a cross-control of any sample becoming negative whatever the initial rearrangement used to generate a probe. In patients on therapy, sequential search for MRD seems to be a good tool for predicting the long-term outcome. In addition, patients remaining positive at the time treatment is discontinued or with a high tumor burden after a few months therapy may be at a higher risk of subsequent relapse, although a longer follow-up is needed to answer this question.
Article
Alteration of the TAL1 locus is the most common nonrandom genetic defect in childhood T-cell acute lymphoblastic leukemia (T-ALL). To determine if rearrangements of the TAL1 proto-oncogene confer a distinct leukemic phenotype, we studied leukemic peripheral blood or bone marrow samples from 182 children with newly diagnosed T-ALL enrolled on Pediatric Oncology Group treatment protocols. Forty-eight (26%) of the samples had a local rearrangement of the TAL1 locus. Demographic and clinical features were compared for patient subgroups with and without TAL1 rearrangements. The only clinical correlates that were significantly associated with TAL1 gene rearrangements were higher white blood cell count (P = .017) and higher hemoglobin (P = .007) at diagnosis. Immunophenotypically, samples with altered TAL1 were more likely to be CD2+ (P = .001) and lack CD10 (cALLa) expression (P = .007) than those without the rearrangement. There was a trend toward improved event-free survival (EFS) in patients with TAL1 rearrangements (4-year EFS was 44% +/- 7% for patients without the rearrangements v 59% +/- 11% for those with rearrangements), but the difference was not significant (P = .34). The role of TAL1 in leukemogenesis has yet to be clearly defined, and the prognostic significance of TAL1 gene rearrangements in T-ALL deserves further study.
Article
As the overall long-term event-free survival rate in children with acute lymphoblastic leukemia approaches 80%, emphasis is being placed on risk-directed therapy so that patients are neither overtreated nor undertreated. It has become apparent that a risk assignment system based on primary genetic abnormalities is inadequate by itself. For example, leukemias with the MLL-AF4 or BCR-ABL fusion gene are, in fact, heterogeneous diseases. Many require allogeneic hematopoietic stem-cell transplantation; some, if the patient is of favorable age and has a low presenting leukocyte count, can be cured with chemotherapy alone. Measurement of early responses to therapy and extent of minimal residual disease can greatly improve the accuracy of risk assessment. Consideration of the variable effects of therapy on the prognostic significance of specific genetic abnormalities is also important. Therefore, TEL-AML1 fusion confers a favorable prognosis in some protocols of chemotherapy but not in others. Studies to identify genetic polymorphisms with pharmacokinetic and pharmacodynamic significance promise to guide further refinement of treatment strategies. This will allow maximization of anticancer effects without induction of unacceptable toxicity in individual patients.
Article
A number of prospective studies have indicated the clinical utility of measuring minimal residual disease (MRD) in childhood acute lymphoblastic leukaemia (ALL) and have highlighted the need for improved methodology for quantification of residual disease. We describe a novel real-time polymerase chain reaction (PCR) strategy for MRD analysis based on the exonuclease activity of Taq polymerase to cleave a fluorescently labelled probe. Using a consensus probe designed to the framework 2 region of the IgH gene, together with leukaemia-specific primers, the utility of this technique for simultaneous detection and quantification of MRD was demonstrated in samples from six ALL patients. This technique provides a rapid quantitative assay for determining MRD levels which lends itself to the routine monitoring of minimal residual leukaemia.