You are on page 1of 10

Articles

Prognostic effect of chromosomal abnormalities in


childhood B-cell precursor acute lymphoblastic leukaemia:
results from the UK Medical Research Council ALL97/99
randomised trial
Anthony V Moorman, Hannah M Ensor, Sue M Richards, Lucy Chilton, Claire Schwab, Sally E Kinsey, Ajay Vora, Chris D Mitchell,
Christine J Harrison

Summary
Background Chromosomal abnormalities in childhood acute lymphoblastic leukaemia are well established disease Lancet Oncol 2010; 11: 429–38
markers and indicators of outcomes. However, the long-term prognosis and independent prognostic effect of some Published Online
abnormalities has been questioned. Also, little is known about the association between cytogenetics and the April 20, 2010
DOI:10.1016/S1470-
characteristics of relapse (eg, time and site of relapse) that are known to predict outcome after relapse.
2045(10)70066-8
See Reflection and Reaction
Methods We analysed cytogenetic data from 1725 children with B-cell precursor acute lymphoblastic leukaemia who page 403
were included in the UK Medical Research Council ALL97/99 study and followed up for a median time of 8·2 years. Leukaemia Research
Univariate and multivariate analysis were done to examine risk of relapse, event-free survival, and overall survival Cytogenetics Group, Northern
associated with 21 chromosomal abnormalities and three cytogenetic risk groups constructed from these data. Institute for Cancer Research,
Newcastle University,
Newcastle upon Tyne, UK
Findings Two chromosomal abnormalities were associated with a significantly better outcome (ETV6–RUNX1, hazard (A V Moorman PhD,
ratio [HR] 0·51, 95% CI 0·38–0·70 and high hyperdiploidy, 0·60, 0·47–0·78), whereas five abnormalities were H M Ensor MSc, L Chilton PhD,
associated with an increased risk of relapse (intrachromosomal amplification of chromosome 21 [iAMP21], 6·04, C Schwab BSc,
Prof C J Harrison FRCPath);
3·90–9·35; t(9;22), 3·55, 2·21–5·72; MLL translocations, 2·98, 1·71–5·20; abnormal 17p, 2·09, 1·30–3·37; and loss
Clinical Trial Service Unit,
of 13q, 1·87, 1·09–3·20). Multivariate analysis incorporating age, white-cell count, and treatment parameters showed University of Oxford, Oxford,
that six cytogenetic abnormalities (ETV6–RUNX1, high hyperdiploidy, iAMP21, t(9;22), loss of 13q, and abnormal UK (S M Richards PhD);
17p) retained their significance for effect on relapse risk. Based on these data, patients were classified into good, Department of Paediatric
Oncology, St James University
intermediate, and poor cytogenetic risk groups. Slow early treatment response correlated with cytogenetic risk group:
Hospital, Leeds, UK
34 of 460 (7%) in the good-risk group, 22 of 211 (10%) in the intermediate-risk group, and 27 of 95 (28%) in the poor- (Prof S E Kinsey FRCPath);
risk group had a slow response (p<0·0001). Additionally, the proportion of patients with a very early (<18 months) Department of Haematology,
relapse varied by cytogenetic risk group: eight of 129 (6%) patients in the good-risk group had a very early relapse, Sheffield Children s Hospital,
Sheffield, UK
compared with 24 of 98 (24%) in the intermediate-risk group, and 37 of 82 (45%) in the poor-risk group (p<0·0001).
(Prof A Vora FRCPath); and
However, there was no difference in the site of relapse by cytogenetic risk group. Department of Paediatric
Oncology, John Radcliffe
Interpretation Individual chromosomal abnormalities are strong independent indicators of outcome, especially risk Hospital, Oxford, UK
(C D Mitchell FRCP)
of relapse. Diagnostic cytogenetics identifies patients with a higher rate of relapse and those who are likely to have a
Correspondence to:
high-risk relapse.
Dr Anthony Moorman,
Leukaemia Research
Funding Leukaemia and Lymphoma Research (LLR). Cytogenetics Group, Northern
Institute for Cancer Research,
Newcastle University, Queen
Introduction four decades.2 Despite these advances, 15–20% of children
Victoria Road, Newcastle upon
Acute lymphoblastic leukaemia (ALL) is a heterogeneous with ALL have disease relapse.3 Continued refinement Tyne NE1 4LP, UK
disease at the cytogenetic and genetic levels.1 Many and monitoring of prognostic factors is warranted to anthony.moorman@ncl.ac.uk
acquired genetic abnormalities have been described in update risk-stratification algorithms in light of new
the bone-marrow cells of patients with ALL; including discoveries and to assess the role of these algorithms in
chromosomal translocations, aneuploidy, deletions, and the context of revised and developing protocols.
amplifications. Several genetic aberrations are Studies have suggested that the strongest risk factors
pathognomic of the disease and can be used to monitor for survival after a first relapse are length of first remission
the patient’s response to therapy. Along with treatment and site of relapse.3 Several clinical study groups have
regimen, age, white-cell count (WCC), and minimal proposed relapse risk classifications based on these factors
residual disease (MRD) detection, the genetic profile of and on immunophenotype.3 Increasingly, these relapse
leukaemia is a main determinant of clinical outcome, risk groups are being used to direct post-relapse therapy,
especially the risk of relapse.2 Improvements in the particularly the use of stem-cell transplantation. However,
design and delivery of frontline protocols for paediatric there are no studies examining the association between
ALL have increased survival rates steadily over the past cytogenetics and relapse risk groups.

www.thelancet.com/oncology Vol 11 May 2010 429


Articles

In this study, we present outcome data from the UK on age, sex, and WCC) or by the presence of t(9;22)
Medical Research Council (MRC) ALL97/99 paediatric (q34;q11.2)/BCR–ABL1, near haploidy (<30 chromosomes),
trial stratified by specific chromosomal abnormalities low hypodiploidy (30–39 chromosomes), or MLL/11q23
and by cytogenetic risk group. Additionally, we examine translocations (younger than 2 years) and were transferred
the association between presentation cytogenetics and to a more intensive protocol.5 In the ALL99 phase, children
relapse risk group. were stratified on the basis of age (<10 years) and WCC
(<50x10⁹ cells per L) to regimen A or (all others) regimen
Methods B. Patients received a three or four (regimen A or B) drug
Patients induction and were classified as a slow early responder if
Between April, 1997, and June, 2002, 1725 children aged the day 15 or 8 (regimen A or B) marrow contained 25%
1–18 years with B-cell precursor ALL (BCP-ALL) received blasts or higher. Patients who failed to remit, were slow
treatment in the ALL97 and ALL99 phases of the MRC early responders, or had t(9;22), near haploidy, low
ALL97/99 trial (figure 1). Infants younger than 1 year hypodiploidy, or MLL/11q23 translocations (<2 years old)
were not eligible for this trial. Participating centres were transferred to the more intensive regimen C. After
obtained approval from the local ethical committee and induction, patients received consolidation, two interim
written informed consent from patients or parents. maintenance blocks, two delayed-intensification blocks,
and continuing therapy for up to 2 years (girls) or 3 years
Procedures (boys). In the ALL97 phase, all patients who received
Full treatment protocols and overall results have been treatment on HR1 were eligible for sibling allogeneic
published.4–6 Both phases, ALL97 and ALL99, included a transplantation at first complete response, but only failure
steroid and purine randomisation: prednisolone or to achieve remission at day 29 and presence of t(9;22)
dexamethasone and mercaptopurine or thioguanine in (q34;q11.2)/BCR–ABL1 were indications for allogeneic
induction and maintenance. In the ALL97 phase, patients transplantation in the ALL99 phase.
received a four-drug induction followed by two or three Cytogenetic analyses were done on the pretreatment
intensification blocks, appropriate CNS-directed treatment, bone marrow or blood samples of 1694 of 1725 patients
and continuing therapy for a total of 2 years. High-risk (98%) by 30 member laboratories of the UK Cancer
patients were identified by the Oxford Hazard Score (based Cytogenetics Group, whose performance was monitored

ALL97
Standard risk Induction Short CNS-directed Short BFM-like
(3 drugs) intensification therapy intensification intensification* CMT
All non-high risk patients

High risk
Induction (3 drugs Intensifi- Intensifi- Transplant or further intensification
High Oxford Hazard Score Intensification CMT
+methotrexate) cation cation with high-dose methotrexate
High risk cytogenetics

ALL99
Regimen A—CCG modified BFM+DDI
Standard risk
Age <10 years Induction
CNS-directed therapy+IM1 DI1 IM2 DI2 CT
WCC <50×109/L (3 drugs)
Day 15 rapid early response

Regimen B—Standard BFM+DDI


Intermediate risk
Age ≥10 years Induction BFM+CNS-directed
IM1 DI1 IM2 DI2 CT
WCC <50×109/L (4 drugs) therapy
Day 8 rapid early response

Regimen C—Augmented BFM


High risk
High risk cytogenetics Induction BFM+CNS-directed Capizzi IM1 DI1 Capizzi DI2 CT
Day 8/15 slow early response (4 drugs) therapy IM2

Day 0 Day 28

Figure 1: ALL97/99 treatment regimens


CMT=continuing maintenance therapy. WCC=white-cell count. CCG=Children’s Cancer Study Group. BFM=Berlin Frankfurt Munster consolidation. DDI=double delayed intensification. IM=interim
maintenance. DI=delayed intensification. CT=continuing therapy. *Third block was randomised in the first year. Day 0 randomisation—dexamethasone vs prednisolone. Day 28 randomisation
mercaptopurine vs thioguanine.

430 www.thelancet.com/oncology Vol 11 May 2010


Articles

by the UK National External Quality Assessment Service model and inspecting its effect on the log likelihood.
for Clinical Cytogenetics.7 The results were collected Only the following cytogenetic variables were considered
centrally by the Leukaemia Research Cytogenetics in the model: high hyperdiploidy (51–65 chromosomes),
Research Group (LRCG).8 Karyotypes were not routinely ETV6–RUNX1, MLL translocations, intrachromosomal
analysed centrally, but were reviewed for accuracy in amplification of chromosome 21 [iAMP21], t(9;22),
description of the structural and numerical clonal abnormal 17p, and loss of 13q. Not all samples were
chromosomal abnormalities, which were reported in screened for all possible abnormalities (see above).
accordance with the International System for Human Because complete data is required to include a case in the
Cytogenetic Nomenclature.9 Analysis of fewer than model, we adopted two strategies—imputing missing
20 normal metaphases was classified as a failure, which values and reducing the dataset to cases with complete
occurred in 275 of 1694 samples (16%). A normal karyotype information. Analysis was done with both strategies,
(20 or more normal metaphases) was present in 219 of since each has the possibility of bias; the results obtained
1419 samples (15%). A clonal abnormality was detected in were very similar. We chose to present the results from
1200 of 1419 samples (85%). Fluorescence in-situ the imputed analysis, because these results were based
hybridisation (FISH) testing was done locally or centrally on more samples and therefore generated more precise
by the LRCG as previously described.10 FISH for ETV6– estimates. Among the 1546 cases used for the modelling,
RUNX1, BCR–ABL1, and MLL was done on 1451 (84%), 1233 (80%) had complete information. Imputing was
1448 (84%), and 1431 (83%) of 1725 samples, respectively, based on the assumption of mutual exclusivity, which is
using commercial fusion or break-apart probes as supported by the data in the webappendix p 1. Samples See Online for webappendix
previously described.10 The Multiprobe-I system (Cytocell, with one primary chromosomal abnormality were
Banbury, Oxfordshire, UK) for the detection of aneuploidy classified as negative for the presence of other primary
was done on 265 samples with failed or incomplete chromosomal abnormalities. The number of samples
cytogenetics or a normal karyotype. Each patient was imputed as absent were: high hyperdiploidy (n=79),
classified according to whether each chromosomal ETV6–RUNX1 (n=188), t(9;22) (n=5), iAMP21 (n=219),
abnormality was present, absent, or had not been and MLL translocations (n=9). Additionally, 127 samples
appropriately tested. Chromosomal abnormalities were with failed cytogenetics were classified as negative for
classified as primary or secondary based on previous abnormal 17p and loss of 13q. Although a more stringent
knowledge.1 Analysis of secondary abnormalities was multiple-comparisons adjustment might be applicable in
restricted to those that were present in ten or more cases. a classical setting, because of the investigative nature of
Event-free survival (EFS) was defined as the time from this analysis, all tests were conducted at the 1%
the start of treatment to relapse or death, and overall significance level. All analyses were done using
survival as the time to death. Relapse-free survival (RFS) Intercooled Stata 11.0 for Windows.
was only determined for patients who achieved a
complete remission, and was defined as the time from Role of the funding source
the date of complete remission until relapse; with deaths The funding source had no role in the study design,
in first remission being censored. Patients without an collection, analysis, and interpretation of data, or writing
event of interest were censored at the date of last contact of the report. The corresponding author had full access
or date of second neoplasm, whichever was earlier. to all data and had final responsibility for the decision to
A second neoplasm was diagnosed in 11 patients (<1%). submit the manuscript for publication.

Statistical analysis Results


Estimates of RFS, EFS, and overall survival were made Survival estimates for the entire cohort of 1725 patients at
using the Kaplan-Meier method. Hazard ratios comparing 5 years were: RFS 81% (95% CI 79–83), EFS 78% (76–80),
RFS, EFS, and overall survival between subgroups were and overall survival 87% (86–89) after a median follow-up
calculated using univariate Cox models. Multivariate Cox time of 8·1, 8·3, and 8·3 years respectively (table 1). In
regression modelling was done for RFS, EFS and overall total there were 351 (20%) relapses, 16 (1%) non-remitters,
survival using a forward-selection stepwise modelling and 257 (15%) deaths, including 57 (3%) in first remission.
process; the difference in the log likelihood (–2×log Patients in the ALL99 phase of the trial had a significantly
likelihood) was used, along with an adjustment for better outcome than those in the ALL97 phase: RFS 84%
steroid randomisation (dexamethasone vs prednisolone) (95% CI 81–87) versus 78% (75–81; p=0·003), EFS 81%
and phase of trial (ALL99 vs ALL97). Age and a log- (78–83) versus 75% (72–78; p=0·005), and overall survival
transformed WCC factor were modelled as continuous 90% (87–91) versus 85% (83–88; p=0·014).
variables, and all other factors were categorical variables. On the basis of cytogenetic and FISH data, 1269 of
All variables in the model were linear and conformed to 1694 patients (75%) were assigned to one (n=983, 78%),
the proportional hazards assumption. Interaction two (n=228, 18%), three (n=53, 4%), or four (n=5, <1%) of
between treatment protocol and cytogenetic abnormalities the cytogenetic subgroups listed in table 1. Of the
was explored by including an interaction term in the final remaining 425 patients (25%), 152 (9%) had some other

www.thelancet.com/oncology Vol 11 May 2010 431


Articles

Total (%) No Deaths in first Relapse, n Relapse-free Event-free Overall


remission, remission, n (%) survival survival survival
n (%) (%)
At 5 years Hazard ratio At 5 years Hazard ratio At 5 years Hazard ratio
(95% CI) (95% CI)* (95% CI) (95% CI)* (95% CI) (95% CI)*
Total
All patients 1725 (100%) 16 (1%) 57 (3%) 351 (20%) 81% (79–83) ·· 78% (76–80) ·· 87% (86–89) ··
Primary abnormalities
High hyperdiploidy 562 (38%) 3 (1%) 11 (2%) 85 (15%) 86% (83–89) 0·60 (0·47–0·78)† 84% (81–87) 0·58(0·46–0·74)† 93% (91–95) 0·45(0·33–0·63)†
ETV6–RUNX1 368 (25%) 2 (1%) 5 (1%) 47 (13%) 89% (86–92) 0·51 (0·38–0·70)† 89% (85–91) 0·48(0·36–0·65)† 96% (94–98) 0·29(0·18–0·47)†
t(1;19)(q23;p13) 50 (4%) 0 (0%) 4 (8%) 6 (12%) 87% (74–94) 0·60 (0·27–1·35) 80% (66–89) 0·85(0·45–1·59) 84% (71–92) 1·29(0·66–2·52)
t(9;22)(q34;q11·2) 43 (3%) 1 (2%) 7 (16%) 18 (42%) 56% (38–70) 3·55 (2·21–5·72)† 44% (29–58) 4·10(2·75–6·11)† 58% (42–71) 4·48(2·80–7·16)†
iAMP21 29 (2%) 1 (3%) 0 (0%) 22 (76%) 27% (12–44) 6·04 (3·90–9·35)† 26% (12–43) 4·91(3·21–7·0)† 69% (49–82) 3·57(2·00–6·40)†
MLL translocations 30 (2%) 0 (0%) 2 (7%) 13 (43%) 55% (35–71) 2·98 (1·71–5·20)‡ 50% (31–66) 2·79(1·66–4·67)† 60% (41–75) 3·34(1·87–5·98)†
Near haploidy 10 (1%) 0 (0%) 0 (0%) 6 (60%) 44% (14–72) ·· 40% (12–67) ·· 40% (12–67) ··
Low hypodiploidy 8 (1%) 0 (0%) 0 (0%) 4 (50%) 50% (15–78) ·· 50% (15–78) ·· 50% (15–78) ··
IGH–CEBP 7 (NA) 0 (0%) 0 (0%) 2 (29%) 71% (26–92) ·· 71% (26–92) ·· 86% (33–98) ··
IGH–ID4 3 (NA) 0 (0%) 0 (0%) 0 (0%) 100% ·· 100% ·· 100% ··
t(17;19)(q23;p13) 2 (0·1%) 0 (0%) 0 (0%) 2 (100%) ·· ·· ·· ·· ·· ··
Secondary abnormalities
del(6q) 77 (5%) 0 (0%) 2 (3%) 17 (22%) 80% (69–87) 1·09 (0·67–1·78) 79% (68–87) 1·02(0·64–1·62) 87% (77–93) 1·08(0·61–1·94)
Abnormal 9p 160 (11%) 2 (1%) 5 (3%) 38 (24%) 77% (70–83) 1·32 (0·93–1·85) 74% (67–80) 1·28(0·94–1·75) 86% (79–90) 1·06(0·69–1·63)
Abnormal 11q 68 (5%) 1 (2%) 3 (4%) 12 (18%) 81% (70–89) 0·89 (0·50–1·58) 78% (66–86) 0·93(0·56–1·56) 87% (76–93) 1·14(0·62–2·10)
Abnormal 17p 50 (4%) 1 (2%) 0 (0%) 18 (36%) 65% (50–77) 2·09 (1·30–3·37)§ 64% (49–76) 1·81(1·14–2·88) 76% (61–86) 1·96(1·12–3·44)
Loss of 13q 44 (3%) 2 (5%) 1 (2%) 14 (32%) 68% (52–80) 1·87 (1·09–3·20) 66% (50–78) 1·88(1·15–3·06) 82% (67–91) 1·35(0·67–2·73)
dup(1q) 41 (3%) 1 (2%) 0 (0%) 8 (20%) 82% (66–91) 0·93 (0·46–1·88) 80% (64–90) 0·88(0·46–1·71) 95% (82–99) 0·48(0·15–1·50)
-7 23 (2%) 1 (4%) 0 (0%) 7 (30%) 77% (54–90) 1·61 (0·76–3·40) 74% (51–87) 1·53(0·76–3·08) 87% (65–96) 0·92(0·30–2·88)
dic(9;20)(p13;q11) 13 (1%) 0 (0%) 1 (8%) 3 (23%) 77% (44–92) 1·13 (0·36–3·53) 77% (44–92) 1·26(0·47–3·38) 92% (57–99) 1·02(0·25–4·10)
dic(9;12)(p11– 15 (1%) 0 (0%) 0 (0%) 3 (20%) 80% (50–93) 1·04(0·33–3·24) 80% (50–93) 0·87(0·28–2·0) 87% (56–97) 0·95(0·24–3·81)
21;p11–13)

High hyperdiploidy=51–65 chromosomes. iAMP21=intrachromosomal amplification of chromosome 21. Near haploidy=less than 30 chromosomes. Low hypodiploidy=30–39 chromosomes. *Hazard ratio is
from a univariate Cox regression model comparing the risk of the event of interest in patients with and without the chromosomal abnormality. †p<0·0001. ‡p<0·001. §p<0·01. Percentages in total column
calculated using the total number of patients tested for each abnormality by cytogenetics, FISH, or RT-PCR: N=1486 for high hyperdiploidy; N=1451 for ETV6-RUNX1;N=1420 for t(1;19), abnormal 11q, and
dic(9;12); N=1419 for t(17;19), del(6q), abnormal 9p, abnormal 17p, loss of 13q, dup(1q), -7, and dic(9;20); N=1633 for t(9;22); N=1449 for iAMP21, N=1627 for other MLL translocations and t(4;11); N=1434 for
near haploidy and low hypodiploidy. No percentages given for IGH-CEBP and IGHID4 since both these abnormalities were identified by selected FISH screening.

Table 1: Outcome of children with B-cell precursor acute lymphoblastic leukaemia by chromosomal abnormality

chromosomal abnormality, 131 (8%) had a normal webappendix (pp 3,6,7). There was little correlation
karyotype, and 142 (8%) did not have a cytogenetic result. between cytogenetics and failure to achieve a remission,
With the exception of seven samples where t(9;22) or or death in first remission; six of seven patients with
ETV6–RUNX1 was observed in a high hyperdiploid t(9;22) who died in first remission underwent bone-
(51–65 chromosomes) karyotype, primary chromosomal marrow transplantation. ETV6–RUNX1 and high
abnormalities did not coexist in the same karyotype hyperdiploidy were associated with an improved outcome,
(webappendix p 1). By contrast, secondary abnormalities whereas t(9;22), iAMP21, MLL translocations, abnormal
often occurred together and in a non-random pattern. 17p, and loss of 13q were associated with a higher risk of
There was no significant variation in the ratio of boys to relapse, death, or both. Although the number of patients
girls by chromosomal abnormality (webappendix p 2). with near haploidy, low hypodiploidy, or t(17;19)(q23;p13)
Low hypodiploidy (30–39 chromosomes), iAMP21, was too small to test formally, the absolute number of
IGH–CEBP, abnormal 9p, and abnormal 17p were relapses and deaths observed strongly suggests they are
associated with older age. Patients with t(1;19)(q23;p13), associated with a poor outcome. There were too few
t(9;22), MLL translocations, near haploidy patients with an IGH–CEPB or IGH–ID4 fusion to assess
(<30 chromosomes), and abnormal 9p had higher WCCs their prognosis accurately.
at presentation, whereas high hyperdiploid and iAMP21 Of the 50 patients with abnormal 17p, 27 (54%) also had
patients had lower WCCs. high hyperdiploidy (n=21) or ETV6–RUNX1 (n=6;
The outcome of patients with different chromosomal webappendix p 1). Abnormalities included i(17q) (n=17),
abnormalities is described in table 1, figure 2, and the del(17p) (n=7), der (17p)/dic(17p) (n=11), add(17p) (n=8) and

432 www.thelancet.com/oncology Vol 11 May 2010


Articles

-17 (n=7). At least 42 (84%) of these 17p abnormalities


resulted in loss of 17p13. The presence of abnormal 17p 100

among patients with ETV6–RUNX1 or high hyperdiploidy


did not adversely affect their relapse risk (HR 1·69, 95% CI 80

Relapse-free survival (%)


0·74–3·85, p=0·210), whereas abnormal 17p conferred a
three times increased risk of relapse for patients without 60

ETV6–RUNX1 or high hyperdiploidy (3·17, 1·76–5·74,


p=0·0001). Given this difference and the fact that i(17q) 40
High hyperdiplody
correlates with high hyperdiploidy,1 we examined the type ETV6–RUNX1
of 17p abnormality across cytogenetic subgroups. We found 20 t(9;22)(q34;q11.2) Abnormal17p
iAMP21 Loss of 13q
some evidence of an association between high MLL translocations Other
0
hyperdiploidy and i(17q): 11 of 21 (52%) with high 0 2 4 6 8 10
hyperdiploidy also had i(17q) versus six of 29 (21%) without Years from diagnosis
high hyperdiploidy (p=0·03) but did not observe any other Number at risk
High hyperdiplody 551 529 470 434 249 98
association that might explain the differential effect of ETV6–RUNX1 365 355 331 301 167 47
abnormal 17p. t(9;22)(q34;q11.2) 42 22 19 14 8 2
iAMP21 28 21 14 6 1 0
44 patients had either a del(13q) (n=27), monosomy 13 MLL translocations 30 20 16 14 9 1
(n=16), or both (n=1). These abnormalities were clearly Abnormal17p 18 15 9 8 7 2
secondary and often coexisted with a known primary Loss of 13q 24 19 17 15 9 4
Other 644 567 483 440 258 105
abnormality (24 of 44 patients [55%]), particularly
ETV6–RUNX1 (n=11), high hyperdiploidy (n=5), and
Figure 2: Kaplan-Meier graphs of relapse-free survival for individual chromosomal abnormalities that were
t(1;19) (n=5; webappendix p 1). There was no correlation significant in univariate analysis
between del(13q)/-13 and cytogenetic subgroup. Among For the purposes of drawing this graph patients were classified hierarchically in the order ETV6–RUNX1, t(9;22),
22 patients with a deletion and defined breakpoints, MLL translocations, iAMP21, high hyperdiploidy, abnormal 17p, loss of 13q. A total of 23 patients were not
16 (73%) involved 13q14. As with abnormal 17p, loss of included due to failure to achieve a complete remission (n=16) or a missing date of remission (n=7).

13q within the ETV6–RUNX1 and high hyperdiploid


groups did not have a significant effect (HR 1·46, 95% CI hyperdiploidy) in the overall survival model. MLL
0·46–4·60, p=0·518). Although loss of 13q among translocations were not prognostic in any of the models.
patients with other abnormalities was associated with an There was no statistical interaction between ETV6–RUNX1
increased risk of relapse, the effect was not significant or high hyperdiploidy and the phase of trial (ALL97 vs
(1·78, 0·94–3·39, p=0·078). ALL99) in the RFS, EFS, or overall survival multivariate
Previous studies have found triple trisomy (+4,+10,+17)11 models. Individual poor-risk chromosomal abnormalities
or trisomy 18 (+18)12 to be associated with an improved were too rare to reliably test for interaction.
outcome. In this study, 419 of 562 (75%) and 484 of 562 The MLL subgroup was cytogenetically heterogeneous
(86%) of patients could be accurately classified by triple and contained patients with t(4;11)(q21;q23)/MLL–AF4
trisomy and +18 status, respectively. Only 253 of (n=17), t(9;11)(p21–22;q23)/MLL–AF9 (n=5), t(11;19)
409 patients (62%) that were classified by both criteria (q23;p13)/MLL–ENL (n=1), t(10;11)(p12–14;q23)/MLL–AF10
were concordant—ie, triple trisomy and +18 (n=190) or (n=1), and unknown MLL partners (n=6). Patients with
neither (n=63)—suggesting that the criteria do not t(4;11) (n=17) were significantly older (median age 8·6 vs
identify the same patient populations. There was no 1·6 years, p=0·002) than other MLL patients (n=13), but
difference in outcome by triple trisomy status (HR 0·80, there was no difference with respect to sex or WCC (data
95% CI 0·49–1·33, p=0·395; webfigure 3). However, not shown). Most MLL patients had a common or pre-B-
high-hyperdiploid patients with +18 had a lower risk of cell immunophenotype (18 of 29, 62%), including seven
relapse than did those with two copies of chromosome 18 t(4;11) patients. The remaining 11 patients—nine t(4;11),
(0·44, 0·26–0·74, p=0·002). Excluding patients on the one t(11;19), and one unknown MLL partner—had a null
ALL97 phase of the trial, who were included in our or pro-B-cell immunophenotype. Because the median age
previous study,12 gave a nearly identical result (0·37, of patients with an MLL translocation was 4 years, we
0·18–0·76, p=0·006). examined the prognostic effect of MLL status in younger
A multivariate Cox proportional hazards model was and older patients. The presence of an MLL translocation
used to assess the significance of each cytogenetic was associated with a higher risk of relapse in patients
abnormality that had been found to be significant in younger than 4 years (HR 8·75; 95% CI 4·55–16·84;
univariate analysis, within the context of other risk factors. p<0·0001), but not in patients 4 years or older (0·91;
Table 2 and the webappendix (pp 4,5) show that six 0·29–2·83; p=0·865). Because of the strong correlation
cytogenetic variables (iAMP21, t(9;22), ETV6–RUNX1, loss between t(4;11) and age in this cohort, similar risks were
of 13q, abnormal 17p, and high hyperdiploidy) retained noted when examining the risk of relapse associated with
their significance in the RFS and EFS models and five t(4;11) and other MLL translocations: HR 5·94 (3·06–11·52;
(iAMP21, t(9;22), ETV6–RUNX1, abnormal 17p, and high p=0·0001) and 1·37 (0·51–3·68, p=0·529), respectively.

www.thelancet.com/oncology Vol 11 May 2010 433


Articles

into these three risk groups and the 178 patients who
Hazard ratio (95% CI) p value
could not be assessed because of failed or missing
ALL99 vs ALL97 0·71 (0·56–0·89) 0·0031
cytogenetic data (webappendix p 3). We used outcome
Dexamethasone vs prednisolone 0·62 (0·49–0·79) 0·0001
data from this study to derive the classification, with
iAMP21 4·69 (2·94–7·48) <0·0001
exceptions. The decision to classify near haploidy, low
WCC† 1·23 (1·13–1·35) <0·0001 hypodiploidy, and t(17;19) as poor risk despite not being
Age 1·05 (1·02–1·08) 0·0003 able to formally test these groups is supported by the fact
t(9;22) 2·65 (1·62–4·35) 0·0001 that the low EFS and overall survival observed is consistent
ETV6–RUNX1 0·53 (0·38–0·74) 0·0002 with several previous studies;1 both near haploidy and low
High hyperdiploidy 0·68 (0·51–0·90) 0·0065 hypodiploidy were included in the risk-stratification
Abnormal 17p 2·21 (1·37–3·57) 0·0012 algorithm as high-risk features. The MLL translocation
Loss of 13q 1·99 (1·16–3·43) 0·0130 group was included in the poor-risk group despite not
Variables are listed in the order in which they entered the model.
retaining its significance in the multivariate model,
iAMP21=intrachromosomal amplification of chromosome 21. WCC=white-cell because the univariate analysis was clearly significant and
count. High hyperdiploidy=51–65 chromosomes. *Sex and MLL translocations patients younger than 2 years with an MLL translocation
were assessed in the stepwise multivariate analysis but were not included in
were treated as high risk in this trial. This, and other
the final model because they did not explain any of the variance. †WCC was
transformed to ln(WCC+1). high-risk criteria, meant that 18 of 30 patients (60%) with
an MLL translocation received the intensified HR1
Table 2: Final multivariate Cox models of relapse-free survival for
protocol or regimen C treatment. Poor-risk abnormalities
individual chromosomal abnormalities*
correlated with older age and higher WCC
(webappendix p 2). Both univariate and multivariate
Panel: Definition of cytogenetic risk groups analysis found a strong correlation between cytogenetic
risk group and outcome (tables 3 and 4; webappendix
Good risk* pp 4,5). There was no statistical interaction between
• High hyperdiploidy (51–65 chromosomes) cytogenetic risk group and the phase of trial (ALL97 vs
• ETV6–RUNX1 ALL99) in the RFS, EFS, or overall survival multivariate
Intermediate risk models (data not shown).
• t(1;19)(q23;p13) Among 766 patients treated in the ALL99 phase who
• IGH–CEBP were assessed for early response and could be classified
• IGH–ID4 into a cytogenetic risk group, 83 patients (11%) were slow
• del(6q) early responders. The proportion of slow early responders
• Abnormal 9p varied significantly by cytogenetic risk group: 34 of
• Abnormal 11q 460 (7%) in the good-risk group, 22 of 211 (10%) in the
• dup(1q) intermediate-risk group, and 27 of 95 (28%)
• -7 in the poor-risk group (p<0·0001).
• dic(9;20)(p13;q11) We used this classification to examine the association
• dic(9;12)(p11–21;p11–13) between cytogenetics and relapse characteristics (table 5).
• Any other abnormality There was a strong correlation between cytogenetic risk
• Normal karyotype group and the time to relapse. Patients in the poor-risk
group were more likely to relapse within 18 months of
Poor risk† diagnosis, whereas those in the good-risk group were
• t(9;22)(q34;q11.2) more likely to have later, off-treatment, relapses
• iAMP21 (p<0·0001). However, there was no correlation between
• MLL translocations the site of relapse and cytogenetic risk group, whether by
• Near haploidy (<30 chromosomes) individual site (table 5) or according to marrow
• Low hypodiploidy (30–39 chromosomes) involvement (data not shown). Finally, we investigated the
• t(17;19)(q23;p13) association between cytogenetic risk group and relapse
• Abnormal 17p risk group as defined by the UKALLR2 trial.13 Table 5
• Loss of 13q shows that relapses arising from the poor cytogenetic risk
iAMP21= intrachromosomal amplification of chromosome 21. *Irrespective of the
group were five times more likely to be classified as a
presence of poor-risk abnormalities, except t(9;22)(q34;q11). †In the absence of high-risk relapse compared with those occurring in the
good-risk abnormalities, except in the situation of t(9;22) with high hyperdiploidy. good cytogenetic risk group (p<0·0001).

We classified 1547 patients hierarchically into three Discussion


cytogenetic risk groups (the panel, tables 3 and 4, and To the best of our knowledge, this dataset represents one
figure 3). There was no significant difference in RFS, of the largest and most comprehensive cytogenetic
EFS, or overall survival between the 1547 patients classified studies of childhood BCP-ALL. The results provide

434 www.thelancet.com/oncology Vol 11 May 2010


Articles

Total (%) No Deaths in first Relapse, n (%) Relapse-free Event-free Overall


remission, remission, survival survival survival
n (%) n (%)
At 5 years Hazard ratio At 5 years Hazard ratio At 5 years Hazard ratio
(95% CI) (95% CI)* (95% CI) (95% CI)* (95% CI) (95% CI)*
Good risk 923 5 (1%) 15 (2%) 129 (14%) 88% (85–90) 0·59 (0·45–0·77)† 86% (84–88) 0·55(0·43–0·69)† 94% (93–96) 0·37(0·27–0·52)†
Intermediate risk 458 5 (1%) 23 (5%) 98 (21%) 80% (76–84) 1 76% (72–80) 1 85% (81–88) 1
Poor risk 166 4 (2%) 10 (6%) 82 (49%) 50% (42–58) 3·19 (2·38–4·29)† 45% (37–52) 2·91(2·23–3·80)† 61% (53–68) 2·82(2·04–3·88)†

*Hazard ratio is from a univariate Cox regression model; for the good and poor cytogenetic risk groups, each has been compared to the intermediate group. †p<0·0001.

Table 3: Outcome of children with B-cell precursor acute lymphoblastic leukaemia by cytogenetic risk group

compelling evidence for the prognostic significance of


Hazard ratio (95% CI) p value
cytogenetics in this disease. Since patients in the ALL99
phase had better outcomes than those in ALL97, we ALL99 vs ALL97 0·70 (0·55–0·87) 0·0019
assessed whether there was any interaction between Dexamethasone vs prednisolone 0·62 (0·49–0·79) 0·0001
cytogenetic abnormalities and protocol. No statistical Good vs intermediate risk 0·70 (0·53–0·92) 0·0110
interaction was observed, suggesting that the improved Poor vs intermediate risk 3·01 (2·23–4·06) <0·0001
outcome seen in ALL99 benefited all patients irrespective WCC* 1·16 (1·06–1·26) 0·0010
of cytogenetic risk group. We conclusively verified the Age 1·05 (1·02–1·08) 0·0004
excellent prognosis associated with ETV6–RUNX1 fusion
Variables are listed in the order in which they entered the model. *WCC was
and high hyperdiploidy.12,14–16 Multivariate analysis transformed to ln(WCC+1).
confirms that these associations are independent of other
Table 4: Final multivariate Cox models of relapse-free survival for
risk factors. Moreover, by contrast with reports from
cytogenetic risk groups
other groups, we did not find an increased risk of late
relapses associated with ETV6–RUNX1.17–19 The crude
relapse rate for patients with ETV6–RUNX1 was problems, and in the future more accurate datasets will
substantially lower in our study compared with others, hopefully resolve this ongoing question.
suggesting our treatment protocol might be averting the This study confirms the poor outcome associated with
late relapses seen, for example, in the NOPHO study.17 t(9;22), near haploidy, low hypodiploidy, MLL
Outcome heterogeneity in high hyperdiploidy by specific translocations, t(17;19), and iAMP21.1 This poor outcome
trisomies has been debated for many years.1 We could was despite patients with t(9;22), near haploidy, low
not confirm the results of the Children’s Oncology Group hypodiploidy, and MLL translocations (<2 years) receiving
study11 suggesting that high-hyperdiploid patients with more intensive treatment. However, these cytogenetic
triple trisomy (+4,+10,+17) have a better outcome. Rather, subgroups had an extremely poor outcome in the
this analysis confirms our previous findings regarding preceding UKALLXI trial20—5-year EFS was 0% for near
the favourable outcome of high hyperdiploid patients haploidy, 13% for t(4;11), and 27% for t(9;22)—suggesting
with +18.12 There are several differences between the two that the risk stratification of these rare cytogenetic
studies that might account for this discrepancy. First, the subgroups in the current trial was beneficial. Although
Children’s Oncology Group analysis was not limited to patients with an MLL translocation had a worse overall
high-hyperdiploid patients, thus their non-triple-trisomy outcome, there was clear evidence of outcome
group included patients with other abnormal and normal heterogeneity within this group. Our analysis suggests
karyotypes. Although this approach includes very rare that age and t(4;11) might be important factors. Because
non-high-hyperdiploid patients with triple trisomy, it of the small number of patients in this subgroup and the
introduces substantial heterogeneity into the analysis, strong correlation between t(4;11) and age, we were not
making the results more difficult to interpret. By contrast, able to determine which factor is the main cause of the
our analysis allows straightforward comparison within heterogeneity. The situation is further confounded by the
the high-hyperdiploid subgroup between patients with fact that 18 of 30 patients were treated as high risk, on the
and without a triple trisomy. Second, although the basis of an MLL translocation or other risk features.
Children’s Oncology Group study included more than Although it is clear from previous studies that age is an
5000 patients, they were treated over a longer period of important prognostic factor for patients with MLL
time (11 years) and in a different theraputic era (1988–99). translocations, most studies have compared infants
Third, poor chromosome morphology results in (<1 year) with children (>1 year), or have only considered
incomplete interpretation of karyotypes and chromosome infants,21–23 and our study did not include infants.
misidentification, both of which are likely contributing Heterogeneity of outcome by MLL partner, with t(4;11)
factors. FISH screening will improve these two technical and t(9;11) associated with a poor outcome, has also been

www.thelancet.com/oncology Vol 11 May 2010 435


Articles

We found that patients with abnormal 17p had a


100
significantly worse outcome than those without this
abnormality, in particular a higher rate of relapse, but
80
only for patients without one of the good-risk abnormalities
Relapse-free survival (%)

of ETV6–RUNX1 and high hyperdiploidy. Similarly, in


60
acute myeloid leukaemia (AML), the presence of poor-risk
cytogenetic abnormalities does not affect the good
40
prognosis of patients with t(15;17), t(8;21), or inv(16).25 We
have recently reported that abnormal or loss of 17p is an
20 Good
Intermediate
independent adverse risk factors in AML.25 Studies of
Poor childhood ALL have been limited to i(17q) within the
0
0 2 4 6 8 10 high-hyperdiploidy subgroup.1 This study confirms our
Years from diagnosis previous observations that abnormal 17p does not negate
Number at risk
Good 916 884 801 735 416 145 the good prognosis associated with high hyperdiploidy.12
Intermediate 451 401 346 316 188 77 In adult ALL, abnormal 17p was not a risk factor in BCP-
Poor 161 107 82 64 38 10
ALL, but results in T-cell ALL were strongly suggestive of
Figure 3: Kaplan-Meier graph of relapse-free survival for cytogentic risk groups
an association with poor outcome.26,27 In AML,
A total of 19 patients were not included due to failure to achieve a complete remission (n=13) or a missing date of abnormalities of 17p correlate with complex karyotypes
remission (n=6). and TP53 inactivation or mutation.28 Although TP53
mutations are rare in childhood ALL, it has been suggested
Total, n(%)* Cytogenetic risk group† that they occur more frequently in high-risk disease.29
Independent studies are required to confirm these
Good, n(%) Intermediate, n(%) Poor, n(%)
observations and unravel the nature of these aberrations.
Total In 2000, Heerema and colleagues30 reported that
Total relapses 309 (20%) 129 (100%) 98 (100%) 82 (100%) del(13q) was associated with an increased risk of relapse
Time of relapse in childhood ALL, although the risk was not independent
<18 months 69 (22%) 8 (6%) 24 (25%) 37 (45%) in multivariate analysis and did not extend to overall
>18 months and <6 months EOT 67 (22%) 28 (22%) 25 (26%) 14 (17%) survival. We included patients with monosomy 13 in our
>6 months EOT 173 (56%) 93 (72%) 49 (50%) 31 (38%) analysis and found that the increased risk of relapse was
Site of relapse independent of other risk factors—although, again,
Isolated marrow 180 (58%) 69 (54%) 57 (58%) 54 (66%) overall survival was not affected. Although it is tempting
Isolated CNS 51 (17%) 22 (17%) 19 (19%) 10 (12%) to speculate that the RB1 gene is the target of this
Isolated other 15 (5%) 8 (6%) 4 (4%) 3 (4%) abnormality, this requires verification.
Combined 63 (20%) 30 (23%) 18 (18%) 15 (18%) Historically, several groups have reported that patients
Relapse risk group‡ with t(1;19) have a worse outcome unless given more
Standard risk 19 (6%) 13 (10%) 3 (3%) 3 (4%) intensive treatment.1 However, in this trial and in our
Intermediate risk 217 (70%) 104 (81%) 72 (74%) 41 (50%) previous study, UKALLXI,20 outcomes for patients with
High risk 73 (24%) 12 (9%) 23 (24%) 38 (46%) t(1;19) were slightly but not significantly better than the
average (table 1). The St Jude Children’s Research
EOT=end of frontline treatment.*Excludes patients with failed or no cytogenetics. †See table 3 for definition of
cytogenetic risk group. ‡Standard risk: isolated extramedullary relapses occurring 6 months or more after EOT.
Hospital recently reported that t(1;19) patients have an
Intermediate risk: isolated extramedullary relapses occurring on therapy or within 6 months of EOT, combined relapses increased risk of CNS relapse.16,31 Of the 50 patients with
occurring 18 months or more after diagnosis, and isolated marrow relapses occurring 6 months after EOT. High risk: t(1;19) in this study, only six (12%) had a relapse, of which
combined relapses occurring within 18 months of diagnosis and isolated marrow relapses occurring on therapy or
within 6 months of EOT.
three involved the CNS. However, with so few relapses
occurring in this subgroup it is impossible to conclude
Table 5: Association between cytogenetic risk group and time of relapse, site of relapse, and relapse that this rate is truly higher than the overall CNS relapse
risk group rate of 110 of 351 (31%).
The prognostic relevance of dic(9;20) is currently the
suggested, particularly among children older than subject of debate, with several groups reporting an
1 year.21,22 A recent study reported a very good outcome for association with other poor-risk features and increased
t(4;11) patients, although the number of patients included incidence in high-risk cohorts.32,33 Our data, however, do
was very small.16 This important clinical issue will need not show evidence of an increased risk of relapse or
to be addressed using a larger cohort of children. worse outcome in this subgroup. This abnormality is
However, a recent gene-expression study that reported a both rare and heterogeneous at the molecular level;34
link between differential expression of HOXA genes and these two factors make it very difficult to assign a reliable
outcome among infants with t(4;11)24 raises the possibility prognosis to patients with dic(9;20).
that secondary genetic alterations could be responsible The literature is contradictory with respect to the
for this heterogeneity. prognostic relevance of 9p abnormalities and CDKN2A

436 www.thelancet.com/oncology Vol 11 May 2010


Articles

deletions in childhood ALL.20,35–40 The evidence from predicts those who are less likely to respond well to
paediatric T-cell ALL cohorts is more consistent for an treatment after relapse. Indeed, in the forthcoming NCRI
association between CDKN2A or 9p deletions and a poor ALL2010 paediatric trial t(9;22), iAMP21, MLL
outcome.41,42 However, not all patients with an abnormality translocations, near haploidy, low hypodiploidy, and
of 9p have a CDKN2A deletion, and vice versa.1,35,43 t(17;19) will be classified as high-risk abnormalities.
Therefore, the terms are not mutually exclusive, despite Contributors
considerable overlap. Our cytogenetics-based observations AVM and CJH were responsible for the conception and design of the
are consistent with the recent European Organisation for study. AVM and HME did the data analysis and interpretation, and
wrote the manuscript. AVM, LC, CS, and CJH collected and classified
Research and Treatment of Cancer (EORTC) study38 of cytogenetic and FISH data. SMR, SEK, AV, and CDM were responsible
paediatric BCP-ALL, which used molecular methods to for trial coordination and provision of clinical and outcome data.
assess CDKN2A copy number and found no evidence for All authors critically reviewed and approved the final manuscript.
an association between CDKN2A deletion and outcome. Conflicts of interests
Many studies in the past 5 years have shown that The authors declared no conflicts of interest.
outcome after relapse is highly heterogeneous.3 Although Acknowledgments
there is a consensus regarding the most relevant risk We thank Leukaemia and Lymphoma Research (formerly Leukaemia
factors affecting post-relapse survival (initial response, Research, UK) for financial support, member laboratories of the UK
Cancer Cytogenetic Group (UKCCG) for providing cytogenetic data and
time of relapse, and site of relapse), little is known about material, and past and present members of the Leukaemia Research
the association between cytogenetics and these factors. Cytogenetics Group (LRCG) for their contribution to establishing this
Using a cytogenetic risk index outlined in the panel, we dataset. The UK Medical Research Council funded the original ALL97/99
found a strong correlation between poor-risk cytogenetics trial and supported the Childhood Leukaemia Working Party. We thank
all members of the Childhood Leukaemia Working Party, especially the
and slow early response, as well as early relapse. chairs—Tim O B Eden (1991–2001) and Brenda E S Gibson (2001–10).
Furthermore, there was a strong correlation between Finally, we thank all the clinicians who entered patients into the trial and
cytogenetic risk group and the relapse risk groups used in the children and families who agreed to take part.
UKALLR2.13 These findings could be used in conjunction References
with data on MRD to identify patients at greatest risk of 1 Harrison CJ, Johannson B. Acute lymphoblastic leukaemia. In:
Heim S, Mitelman F, eds. Cancer Cytogenetics, 3rd edn. New
multiple treatment failure who would benefit most from Jersey: Wiley-Blackwell, 2009: 233–96.
early intervention with alternative therapies. 2 Pui CH, Evans WE. Treatment of acute lymphoblastic leukemia.
This study has many strengths, including size, long N Engl J Med 2006; 354: 166–78.
3 Bailey LC, Lange BJ, Rheingold SR, Bunin NJ. Bone-marrow relapse
follow-up, and quality and extent of cytogenetic analysis; in paediatric acute lymphoblastic leukaemia. Lancet Oncol 2008;
however, it does have some limitations. The cytogenetic 9: 873–83.
risk groups for childhood ALL proposed in the panel 4 Mitchell CD, Richards SM, Kinsey SE, Lilleyman J, Vora A,
require validation in independent cohorts. Recent array Eden TO. Benefit of dexamethasone compared with prednisolone
for childhood acute lymphoblastic leukaemia: results of the UK
studies have highlighted many new focal deletions of Medical Research Council ALL97 randomized trial.
B-cell differentiation and cell-cycle genes in childhood Br J Haematol 2005; 129: 734–45.
ALL.32,44 Unfortunately, we could not include data on these 5 Mitchell C, Payne J, Wade R, et al. The impact of risk stratification
by early bone-marrow response in childhood lymphoblastic
abnormalities in this study because of a lack of suitable leukaemia: results from the United Kingdom Medical Research
material. Although IKZF1 deletions have been reported Council trial ALL97 and ALL97/99. Br J Haematol 2009; 146: 424–36.
to confer a higher risk of relapse, the largest effect was 6 Vora A, Mitchell CD, Lennard L, et al. Toxicity and efficacy of
6-thioguanine versus 6-mercaptopurine in childhood lymphoblastic
seen in patients with t(9;22), which is already a well leukaemia: a randomised trial. Lancet 2006; 368: 1339–48.
established poor-risk subgroup.44 Additionally, den Boer 7 UK NEQAS. United Kingdom National External Quality
and colleagues32 reported that many of the microdeletions Assessment Scheme in Clinical Cytogenetics. http://www.ccneqas.
org.uk/ (accessed June, 2002).
correlated with the t(9;22)/BCR-ABL1 subgroup or the 8 Harrison CJ, Martineau M, Secker-Walker LM. The Leukaemia
newly described BCR-ABL1-like subgroup. However, the Research Fund/United Kingdom Cancer Cytogenetics Group
BCR-ABL1-like subgroup did not correlate with any of karyotype database in acute lymphoblastic leukaemia: a valuable
resource for patient management. Br J Haematol 2001; 113: 3–10.
the main cytogenetic subgroups; therefore, although it
9 An International System for Human Cytogenetic Nomenclature.
will be important to assess the prognostic relevance of Basel, Switzerland: Karger, 2005.
these microdeletions in the context of these results; we 10 Harrison CJ, Moorman AV, Barber KE, et al. Interphase molecular
do not think that the absence of such data substantially cytogenetic screening for chromosomal abnormalities of
prognostic significance in childhood acute lymphoblastic
changes the key findings in this study. leukaemia: a UK Cancer Cytogenetics Group Study.
The findings from this large and comprehensively Br J Haematol 2005; 129: 520–30.
characterised cohort of childhood ALL show the 11 Sutcliffe MJ, Shuster JJ, Sather HN, et al. High concordance from
independent studies by the Children’s Cancer Group (CCG) and
importance of cytogenetics in the management of patients Pediatric Oncology Group (POG) associating favorable prognosis
with this disease. We have confirmed that individual with combined trisomies 4, 10, and 17 in children with NCI
chromosomal abnormalities are strong independent standard-risk B-precursor acute lymphoblastic leukemia: a Children’s
Oncology Group (COG) initiative. Leukemia 2005; 19: 734–40.
predictors of outcome, especially risk of relapse. Moreover, 12 Moorman AV, Richards SM, Martineau M, et al. Outcome
we have shown that diagnostic cytogenetics not only heterogeneity in childhood high-hyperdiploid acute lymphoblastic
identifies patients with a higher rate of relapse, but also leukemia. Blood 2003; 102: 2756–62.

www.thelancet.com/oncology Vol 11 May 2010 437


Articles

13 Roy A, Cargill A, Love S, et al. Outcome after first relapse in 29 Marks DI, Kurz BW, Link MP, et al. High incidence of potential
childhood acute lymphoblastic leukaemia—lessons from the United p53 inactivation in poor outcome childhood acute lymphoblastic
Kingdom R2 trial. Br J Haematol 2005; 130: 67–75. leukemia at diagnosis. Blood 1996; 87: 1155–61.
14 Shurtleff SA, Buijs A, Behm FG, et al. TEL/AML1 fusion resulting 30 Heerema NA, Sather HN, Sensel MG, et al. Abnormalities of
from a cryptic t(12;21) is the most common genetic lesion in chromosome bands 13q12 to 13q14 in childhood acute
pediatric ALL and defines a subgroup of patients with an excellent lymphoblastic leukemia. J Clin Oncol 2000; 18: 3837–44.
prognosis. Leukemia 1995; 9: 1985–89. 31 Jeha S, Pei D, Raimondi SC, et al. Increased risk for CNS relapse in
15 Rubnitz JE, Wichlan D, Devidas M, et al. Prospective analysis of pre-B cell leukemia with the t(1;19)/TCF3-PBX1. Leukemia 2009;
TEL gene rearrangements in childhood acute lymphoblastic 23: 1406–09.
leukemia: a Children’s Oncology Group study. J Clin Oncol 2008; 32 Den Boer ML, van Slegtenhorst M, De Menezes RX, et al. A subtype
26: 2186–91. of childhood acute lymphoblastic leukaemia with poor treatment
16 Pui CH, Campana D, Pei D, et al. Treating childhood acute outcome: a genome-wide classification study. Lancet Oncol 2009;
lymphoblastic leukemia without cranial irradiation. 10: 125–34.
N Engl J Med 2009; 360: 2730–41. 33 Forestier E, Gauffin F, Andersen MK, et al. Clinical and cytogenetic
17 Forestier E, Heyman M, Andersen MK, et al. Outcome of ETV6/ features of pediatric dic(9;20)(p13.2;q11.2)-positive B-cell precursor
RUNX1-positive childhood acute lymphoblastic leukaemia in the acute lymphoblastic leukemias: a Nordic series of 24 cases and
NOPHO-ALL-1992 protocol: frequent late relapses but good overall review of the literature. Genes Chromosomes Cancer 2008; 47: 149–58.
survival. Br J Haematol 2008; 140: 665–72. 34 An Q, Wright SL, Moorman AV, et al. Heterogeneous breakpoints
18 Seeger K, Buchwald D, Peter A, et al. TEL-AML1 fusion in relapsed in patients with acute lymphoblastic leukemia and the dic(9;20)
childhood acute lymphoblastic leukemia. Blood 1999; 94: 374–76. (p11-13;q11) show recurrent involvement of genes at 20q11.21.
19 Tsang KS, Li CK, Chik KW, et al. TEL/AML1 rearrangement and the Haematologica 2009; 94: 1164–69.
prognostic significance in childhood acute lymphoblastic leukemia 35 Heerema NA, Sather HN, Sensel MG, et al. Association of
in Hong Kong. Am J Hematol 2001; 68: 91–98. chromosome arm 9p abnormalities with adverse risk in childhood
20 Hann I, Vora A, Harrison G, et al. Determinants of outcome after acute lymphoblastic leukemia: a report from the children’s cancer
intensified therapy of childhood lymphoblastic leukaemia: results group. Blood 1999; 94: 1537–44.
from Medical Research Council United Kingdom acute lymphoblastic 36 Carter TL, Watt PM, Kumar R, et al. Hemizygous p16(INK4A)
leukaemia XI protocol. Br J Haematol 2001; 113: 103–14. deletion in pediatric acute lymphoblastic leukemia predicts
21 Pui CH, Chessells JM, Camitta B, et al. Clinical heterogeneity in independent risk of relapse. Blood 2001; 97: 572–74.
childhood acute lymphoblastic leukemia with 11q23 37 Dalle JH, Fournier M, Nelken B, et al. p16INK4a
rearrangements. Leukemia 2003; 17: 700–06. immunocytochemical analysis is an independent prognostic factor
22 Pui CH, Gaynon PS, Boyett JM, et al. Outcome of treatment in in childhood acute lymphoblastic leukemia. Blood 2002;
childhood acute lymphoblastic leukaemia with rearrangements of 99: 2620–23.
the 11q23 chromosomal region. Lancet 2002; 359: 1909–15. 38 Mirebeau D, Acquaviva C, Suciu S, et al. The prognostic
23 Pieters R, Schrappe M, De Lorenzo P, et al. A treatment protocol for significance of CDKN2A, CDKN2B and MTAP inactivation in
infants younger than 1 year with acute lymphoblastic leukaemia B-lineage acute lymphoblastic leukemia of childhood: results of the
(Interfant-99): an observational study and a multicentre randomised EORTC studies 58881 and 58951. Haematologica 2006; 91: 881–85.
trial. Lancet 2007; 370: 240–50. 39 van Zutven LJ, van Drunen E, de Bont JM, et al. CDKN2 deletions
24 Stam RW, Schneider P, Hagelstein JA, et al. Gene expression have no prognostic value in childhood precursor-B acute
profiling-based dissection of MLL translocated and MLL germline lymphoblastic leukaemia. Leukemia 2005; 19: 1281–84.
acute lymphoblastic leukemia in infants. Blood 2010; 115: 2835–44. 40 Graf Einsiedel H, Taube T, Hartmann R, et al. Deletion analysis of
25 Grimwade D, Hills RK, Moorman AV, et al. Refinement of p16INKa and p15INKb in relapsed childhood acute lymphoblastic
cytogenetic classification in acute myeloid leukemia: determination leukemia. Blood 2002; 99: 4629–31.
of prognostic significance of rare recurring chromosomal 41 Fizzotti M, Cimino G, Pisegna S, et al. Detection of homozygous
abnormalities amongst 5876 younger adult patients treated in the deletions of the cyclin-dependent kinase 4 inhibitor (p16) gene in
UK Medical Research Council trials. Blood 2009; published online acute lymphoblastic leukemia and association with adverse
April 12, 2010; DOI 10.1182/blood-2009-11-254441. prognostic features. Blood 1995; 85: 2685–90.
26 Marks DI, Paietta EM, Moorman AV, et al. T-cell acute 42 Diccianni MB, Batova A, Yu J, et al. Shortened survival after relapse
lymphoblastic leukemia in adults: clinical features, in T-cell acute lymphoblastic leukemia patients with p16/p15
immunophenotype, cytogenetics and outcome from the large deletions. Leuk Res 1997; 21: 549–58.
randomised prospective trial (UKALL XII/ECOG 2993). 43 Sulong S, Moorman AV, Irving JA, et al. A comprehensive analysis
Blood 2009; 114: 5136–45. of the CDKN2A gene in childhood acute lymphoblastic leukaemia
27 Moorman AV, Harrison CJ, Buck GA, et al. Karyotype is an reveals genomic deletion, copy number neutral loss of
independent prognostic factor in adult acute lymphoblastic heterozygosity and association with specific cytogenetic subgroups.
leukemia (ALL): analysis of cytogenetic data from patients treated Blood 2009; 113: 100.
on the Medical Research Council (MRC) UKALLXII/Eastern 44 Mullighan CG, Su X, Zhang J, et al. Deletion of IKZF1 and
Cooperative Oncology Group (ECOG) 2993 Trial. prognosis in acute lymphoblastic leukemia. N Engl J Med 2009;
Blood 2007; 109: 3189–97. 360: 470–80.
28 Schoch C, Kern W, Kohlmann A, Hiddemann W, Schnittger S,
Haferlach T. Acute myeloid leukemia with a complex aberrant
karyotype is a distinct biological entity characterized by genomic
imbalances and a specific gene expression profile.
Genes Chromosomes Cancer 2005 43: 227–38.

438 www.thelancet.com/oncology Vol 11 May 2010

You might also like