Sie sind auf Seite 1von 24

GENES, CHROMOSOMES & CANCER 48:637660 (2009)

High Hyperdiploid Childhood Acute


Lymphoblastic Leukemia
Kajsa Paulsson
*
and Bertil Johansson
*
Department of Clinical Genetics,Lund University Hospital, Lund, Sweden
High hyperdiploidy (5167 chromosomes) is the most common cytogenetic abnormality pattern in childhood B-cell precur-
sor acute lymphoblastic leukemia (ALL), occurring in 2530% of such cases. High hyperdiploid ALL is characterized cytoge-
netically by a nonrandom gain of chromosomes X, 4, 6, 10, 14, 17, 18, and 21 and clinically by a favorable prognosis.
Despite the high frequency of this karyotypic subgroup, many questions remain regarding the epidemiology, etiology, pres-
ence of other genetic changes, the time and cell of origin, and the formation and pathogenetic consequences of high hyper-
diploidy. However, during the last few years, several studies have addressed some of these important issues, and these, as
well as previous reports on high hyperdiploid childhood ALL, are reviewed herein. V VC
2009 Wiley-Liss, Inc.
DELINEATION OF HIGH HYPERDIPLOID
CHILDHOOD ALL
The high hyperdiploid subgroup was identied
rst in the early 1980s, when Kaneko et al. (1981)
described a distinct difference between the
distribution of modal chromosome numbers
between adult and childhood acute lymphoblastic
leukemia (ALL), emphasizing that hyperdiploidy
with modes between 50 and 59 was only seen in
pediatric cases. Furthermore, the vast majority of
the children were below the age of 4, the white
blood cell (WBC) counts were generally low, and
the blasts were non-T and non-B, i.e., B-cell pre-
cursor in modern nomenclature. In addition, an
excellent response to chemotherapy, as well as
long remissions, was noted. Although we will
return to the prognostic impact of high hyperdi-
ploidy, it should be stressed that Fritz Lampert,
performing DNA measurements and chromosome
studies of childhood ALL, already in 1967 reported
that some of them were characterized by a hyper-
diploid DNA content corresponding to 5059 chro-
mosomes and that survival was particularly long in
such cases, suggesting that the high hyperdiploid
cells were particularly sensitive to chemotherapy
because of their increased DNA content (Lampert,
1967). It is surprising that these seminal and early
ndings rarely are referred to in publications focus-
ing on high hyperdiploid ALL, something hope-
fully rectied in the present review.
At about the same time as the report by Kaneko
et al. (1981), the Third International Workshop on
Chromosomes in Leukemia (1981a,b) also found
an association between high hyperdiploidy and
pediatric ALL. Shortly afterward, several corrobora-
tive studies appeared, using either karyotyping or
DNA content measurements to identify this sub-
group (Kaneko et al., 1982; Look et al., 1982; Wil-
liams et al., 1982; Heerema et al., 1985; Kowalczyk
et al., 1985; Smets et al., 1985). Thus, by the mid
1980s, it had become clear that high hyperdiploidy
is a characteristic subgroup of childhood ALL.
DEFINITION OF THE HIGH
HYPERDIPLOID SUBGROUP
Whereas the term hyperdiploidy is clearly
dened in the cytogenetic nomenclature as 4757
chromosomes (ISCN, 2005), there is no consensus
as regards which cases that should be included in
the high hyperdiploid subgroup of childhood B-
cell precursor ALL. The decision to dene this
subset as more than 50 chromosomes, something
that has become universally accepted, may seem
arbitrary. However, Williams et al. (1982) noted
that this was a natural division because of the
bimodal distribution of chromosome numbers
(Fig. 1) and the clear differences in treatment
response between the >50 and 4750 chromo-
some groups. The upper limit of high hyper-
diploidy, however, varies among different
studies. Originally, all cases with more than 50
Supported by: Swedish Childhood Cancer Foundation; Swedish
Cancer Society; Swedish Research Council.
*Correspondence to: Kajsa Paulsson, Department of Clinical
Genetics, University Hospital, SE-221 85, Lund, Sweden. E-mail:
kajsa.paulsson@med.lu.se or Bertil Johansson, Department of Clin-
ical Genetics, University Hospital, SE-221 85, Lund, Sweden.
E-mail: bertil.johansson@med.lu.se
Received 4 March 2009; Accepted 31 March 2009
DOI 10.1002/gcc.20671
Published online 4 May 2009 in
Wiley InterScience (www.interscience.wiley.com).
REVIEW ARTICLE
V VC
2009 Wiley-Liss, Inc.
chromosomes were included; thus, no distinction
was made between hyperdiploid, near-triploid,
and near-tetraploid ALL. Although some investi-
gators still adhere to that tradition, it has recently
become apparent that cases with chromosome
numbers in the tetraploid range are genetically
distinct, with the vast majority harboring the
t(12;21)(p13;q22) [ETV6/RUNX1 fusion] (Attarba-
schi et al., 2006; Raimondi et al., 2006). Near-tet-
raploid cases should hence not be part of the
high hyperdiploid group. Conversely, based on
the presence of specic trisomies and tetrasomies
(see below) as well as on the modal number dis-
tribution (Fig. 1), it is quite clear that most hypo-
triploid cases should be included. In practice, the
upper limit has been variably dened as 61 (For-
estier et al., 2008a), 65 (Moorman et al., 2003;
Sharathkumar et al., 2008), and 67 (Raimondi
et al., 1996; Heerema et al., 2007). In most
instances, no reasons are given for choosing a par-
ticular cutoff, but in a recent analysis of trisomies
and tetrasomies at different modal numbers,
Heerema et al. (2007) concluded that the chro-
mosomal patterns differed between cases with
5167 chromosomes and those with >67 chromo-
somes. Considering that this is, to the best of our
knowledge, the only delineation of high hyperdi-
ploidy based on a clear-cut difference between
two neighboring modal numbers (67 and 68), we
have chosen to use an upper limit of 67 chromo-
somes. Therefore, high hyperdiploidy is dened
as 5167 chromosomes in the present review.
EPIDEMIOLOGY
Frequency of High Hyperdiploidy in ALL
High hyperdiploidy is generally found in 25
30% of pediatric B-lineage ALL (Table 1). In
contrast, modal numbers of 5167 are much less
common in adult B-lineage ALL and are rarely
seen in T-cell ALL or Burkitt leukemia/lym-
phoma (Fig. 2). Thus, high hyperdiploidy is
strongly associated with childhood ALL.
Although the reported frequencies of high
hyperdiploid cases are quite similar in different
studies, it should be stressed that the patient
cohorts may differ, for several reasons. First, the
denition of high hyperdiploidy has varied. Sec-
ond, also the age range has differed; adolescents
above the age of 15 and/or infants have been
included in some studies, but not in others. Con-
sidering the typical age distribution of high
hyperdiploidy (Fig. 3) this may introduce a fre-
quency variation. Third, and perhaps most impor-
tant, cytogenetic analyses of high hyperdiploid
ALL are notoriously difcult, and there is ample
evidence that many cases go undetected if only
chromosome banding is used. Already during the
1980s, it was shown that ow cytometry analysis
could identify an aneuploid DNA index, compati-
ble with high hyperdiploidy, in cases with a nor-
mal karyotype (Look et al., 1982; Smets et al.,
1985). Subsequently, interphase FISH analyses
were shown to increase the detection rate of high
hyperdiploid cases quite substantially (Berger
et al., 1994; Martin et al., 1996; Kasprzyk and
Secker-Walker, 1997; Ritterbach et al., 1998;
Nordgren et al., 2002; Pere z-Vera et al., 2004).
TABLE 1. Frequency of High Hyperdiploidy in Larger Series
of Pediatric B-Lineage ALL
Origin Frequency
a
Reference
Austria 24% (71/299) Attarbaschi et al. (2004)
France 31% (104/334) Groupe Francais de
Cytogenetique
Hematologique (1993)
Germany 27% (173/650) Lampert et al. (1991)
Germany/Italy 17% (48/286) Borkhardt et al. (1997)
Japan 25% (47/191) Kobayashi et al. (1994)
Netherlands 38% (100/266) Smets et al. (1995)
Nordic
countries
25% (168/679) Forestier et al. (2008a)
UK 27% (183/680) Chessells et al. (1997)
UK 30% (303/1013) UK Childhood Cancer
Study Investigators
(2000)
USA 33% (41/125) Williams et al. (1982)
USA 30% (55/183) Rubnitz et al. (1997)
USA 24% (54/222) Borowitz et al. (1998)
USA 28% (332/1178) Heerema et al. (2000)
USA 28% (38/135) Sharathkumar
et al. (2008)
a
P < 0.001 (chi square).
Figure 1. Modal chromosome number distribution of 637 cytoge-
netically abnormal pediatric (<18 years) B-lineage ALL reported in
the literature (Mitelman et al., 2009). Only unselected cases are
included. Two incidence peaks are clearly seen, one corresponding to
near-diploid cases and one to high hyperdiploid cases.
638 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
Thus, such analyses have proved to be a valuable
adjunct to conventional cytogenetic studies, with
FISH having the additional advantage of provid-
ing information on which chromosomes that are
gained, in contrast to ow cytometry. Since
hidden high hyperdiploidy has been detected by
FISH in one third of cytogenetically normal
childhood ALL and in close to 60% in karyotypi-
cally failed cases (Harrison et al., 2005), inter-
phase FISH analyses should denitely be
undertaken in pediatric ALL with uninformative
cytogenetic results.
Frequency of High Hyperdiploidy in Relation to
Geographic or Ethnic Background
Among larger series of pediatric B-lineage ALL
from different countries, the frequencies of high
hyperdiploidy vary between 17% and 38% (Table
1). Although this variation is statistically signicant,
it should be emphasized that in the vast majority
of studies the prevalence is 2530%, strongly indi-
cating that there is no pronounced geographic fre-
quency heterogeneity. However, larger cytogenetic
studies have only been reported from rather afu-
ent countries; thus, whether the frequency of high
hyperdiploid ALL may be different in developing
countries is presently unknown.
As regards a possible impact of ethnic back-
ground, only a few studies, all from the US, have
specically addressed this topic. Pui et al.
(1988b) noted that high hyperdiploid ALL was
signicantly more common in white than in
nonwhite children and subsequently reported
that this type of ALL was underrepresented
among African-Americans (Raimondi et al., 1996;
Pui et al., 2003). Similarly, Pollock et al. (2000)
found a lower frequency of high hyperdiploid
ALL in African-Americans. On the other hand,
Bhatia et al. (2002) did not observe a signicant
frequency difference of the various ploidy levels
in different ethnic groups; however, the lowest
incidence of high hyperdiploidy was seen among
African-Americans. Thus, ethnicity may play a
role in the occurrence of high hyperdiploid ALL,
but whether this is due to genetic or environmen-
tal factorsnature versus nurtureremains
unknown.
Frequency of High Hyperdiploidy in
Relation to Age
As seen in Figure 3, high hyperdiploid ALL
displays a pronounced incidence peak at 24
Figure 2. Modal chromosome number distribution of (A) 143
adult (18 years) B-lineage ALL, (B) 571 T-ALL, and (C) 286 Burkitt
leukemias/lymphomas with cytogenetic abnormalities reported in the
literature (Mitelman et al., 2009). Only unselected cases are included.
As seen, the incidence of high hyperdiploid cases is very low in all
three disorders.
Figure 3. Age distribution of high hyperdiploid B-lineage ALL
reported in the literature (Mitelman et al., 2009).
HIGH HYPERDIPLOID CHILDHOOD ALL 639
Genes, Chromosomes & Cancer DOI 10.1002/gcc
years, being very rare in infant ALL and also
quite infrequent in patients above the age of 7;
the median age has been reported to be 3.63.9
years in larger studies (Table 2). In fact, high
hyperdiploid cases comprise the largest karyo-
typic subgroup of the frequency peak at 27
years of all childhood ALL. Together with ETV6/
RUNX1-positive cases, they constitute 50% of
all childhood ALL and almost 80% of ALL
occurring in the 27 years age peak (Forestier
and Schmiegelow, 2006).
Frequency of High Hyperdiploidy in Relation
to Gender
Retrieving all cytogenetically characterized B-
lineage ALL with modal chromosome numbers
between 51 and 67 in children/adolescents below
the age of 18 reported in the literature (Mitelman
et al., 2009) reveals a male/female ratio of 1.3.
That high hyperdiploidy may be more common
in boys has also been reported in some previous
studies; however, the sex ratio is close to 1.0 in
all larger series (Table 2). Thus, if there is really
a male preponderance, it is not particularly
pronounced.
ETIOLOGY
Although there is a seemingly never-ending list
of possible risk factors for childhood ALL,
including parental, in utero, and postnatal expo-
sures as well as genetic susceptibility, it seems
safe to conclude that many are called, but few
are chosen. Furthermore, and unfortunately,
most studies addressing this important issue treat
ALL as a single entity, not taking into account
the diverse clinical, phenotypic, and genetic fea-
tures that characterize this heterogeneous dis-
easeas succinctly stated by Greaves (1997):
different biological subtypes of leukaemia may
have distinct causal mechanisms.
Considering that high hyperdiploidy is the
most common cytogenetic abnormality pattern in
pediatric ALL, surprisingly little is known about
its etiology. As discussed below, several studies
have provided circumstantial evidence that this
karyotypic feature arises in a single mitosis al-
ready before birth. This suggests that exposure to
transforming agents during pregnancy, probably
in cooperation with genetic risk factors, is impor-
tant and that postnatal second hits, which also
may be caused by environmental agents and
modulated by constitutional factors, are necessary
for overt leukemia. Thus, to further our under-
standing of the etiology of high hyperdiploid
ALL both genetic risk factors as well as prenatal
and postnatal exposures warrant investigation.
Genetic Risk Factors
Down syndrome (DS) has long been known
to be associated with an 20 fold increased risk
of ALL (Krivit and Good, 1957; Hasle, 2001).
The proportion of high hyperdiploid DS-ALL
has been shown to be signicantly lower
(10%) than in non-DS-ALL patients (25
30%). This notwithstanding, it is clear that a
constitutional trisomy 21 still increases the risk
for high hyperdiploid ALL (Forestier et al.,
2008b). High hyperdiploid DS-ALL are charac-
terized by gains of the same chromosomes as
non-DS ALL, strongly suggesting the same eti-
ology and/or pathogenesis of high hyperdiploidy
in children with or without DS (Forestier et al.,
2008b). No other constitutional chromosome ab-
normality has been correlated with high hyper-
diploid ALL.
Despite the fact that several Mendelian disor-
ders have been associated with acute leukemia
(Online Mendelian Inheritance in Man, 2009),
they do not seem to increase the risk for high
hyperdiploid ALL; there are, as yet, no known
highly penetrant recessive or dominant genes
linked to the development of this ALL subtype.
Attention has instead been focused on suscepti-
bility due to low penetrance genes. However,
analyses of genetic polymorphisms as a risk fac-
tor for high hyperdiploid ALL have thus far
been quite unsuccessful. Chen et al. (1997)
identied glutathione S-transferase deletions as
a risk factor, but those with this genotype more
often had non-high hyperdiploid ALL. On the
other hand, the HLA-DPB1*0201 allele, belong-
ing to the HLA class II genes that are important
in adaptive immune responses to infection, has
been shown to be signicantly more common in
children with hyperdiploid ALL than in healthy
infants, suggesting that it increases the risk for
this type of ALL (Taylor et al., 2002). Hence,
there may be genetic risk factors for high hyper-
diploid ALL, but based on available evidence,
in particular that siblingstwins excludedto
children with ALL do not have an increased
risk of ALL (Schmiegelow and Hjalgrim, 2006),
constitutional genetics does not seem to be a
major etiologic factor.
640 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
T
A
B
L
E
2
.
C
l
i
n
i
c
a
l
F
e
a
t
u
r
e
s
a
t
D
i
a
g
n
o
s
i
s
o
f
H
i
g
h
H
y
p
e
r
d
i
p
l
o
i
d
P
e
d
i
a
t
r
i
c
B
-
L
i
n
e
a
g
e
A
L
L
R
e
f
e
r
e
n
c
e
N
o
.
o
f
c
a
s
e
s
M
/
F
A
g
e
i
n
y
e
a
r
s
a
H
e
m
o
g
l
o
b
i
n
a
(
g
/
l
)
P
l
a
t
e
l
e
t
s
a

1
0
9
/
l
W
B
C
c
o
u
n
t
a

1
0
9
/
l
P
B
b
l
a
s
t
s
a
(
%
)
B
M
b
l
a
s
t
s
a
(
%
)
M
e
d
i
a
s
t
i
n
a
l
m
a
s
s
(
%
)
C
N
S
(
%
)
K
a
n
e
k
o
e
t
a
l
.
(
1
9
8
1
)
5
0
.
7
4
(
2

4
)
5
8
(
2
9

7
2
)
4
6
(
8

2
6
9
)
1
0
(
4
.
3

7
5
)
4
8
(
0

8
2
)
N
R
N
R
N
R
T
I
W
C
L
b
(
1
9
8
1
b
)
2
2
1
.
0
3
7
0
6
3
6
3
9
9
9
0
5
K
a
n
e
k
o
e
t
a
l
.
(
1
9
8
2
)
9
0
.
8
4
(
2

1
5
)
6
3
(
2
9

7
7
)
7
4
(
8

3
2
6
)
4
.
3
(
1
.
1

7
5
)
2
7
(
0

8
2
)
N
R
0
0
W
i
l
l
i
a
m
s
e
t
a
l
.
(
1
9
8
2
)
4
1
0
.
9
3
.
5
N
R
N
R
5
.
6
N
R
N
R
2
5
H
e
e
r
e
m
a
e
t
a
l
.
(
1
9
8
5
)
1
4
1
.
3
3
(
2

1
0
)
7
3
(
3
3

1
2
2
)
2
9
(
2

1
2
3
)
5
.
2
(
2
.
6

1
1
8
)
N
R
9
6
(
7
2

9
9
)
0
N
R
H
e
i
n
o
n
e
n
e
t
a
l
.
(
1
9
8
8
)
2
0
1
.
0
5
.
4
(
1
.
7

1
2
)
N
R
N
R
8
.
4
N
R
N
R
N
R
0
P
r
i
g
o
g
i
n
a
e
t
a
l
.
(
1
9
8
8
)
2
6
1
.
6
4
.
4
(
1
.
7

1
2
)
N
R
N
R
4
.
7
(
1
.
0

6
5
)
3
1
(
0

8
9
)
1
0
0
(
5
1

1
0
0
)
1
2
N
R
F
e
n
a
u
x
e
t
a
l
.
(
1
9
8
9
)
1
3
0
.
4
1
0
8
2
N
R
8
.
9
N
R
N
R
N
R
N
R
P
u
i
e
t
a
l
.
(
1
9
8
9
)
1
3
8
1
.
0
3
.
6
(
0
.
9

1
7
)
7
6
(
1
0

1
4
3
)
4
6
(
0

8
3
1
)
6
.
6
(
0
.
8

3
3
2
)
N
R
N
R
2
4
S
e
c
k
e
r
-
W
a
l
k
e
r
e
t
a
l
.
(
1
9
8
9
)
1
7
1
.
4
4
.
0
N
R
N
R
<
2
0
N
R
N
R
N
R
N
R
H
a
y
a
s
h
i
e
t
a
l
.
(
1
9
9
1
)
1
3
1
.
2
3
(
1

8
)
N
R
N
R
1
4
(
1
.
9

7
5
)
N
R
N
R
N
R
N
R
W
h
i
t
e
h
e
a
d
e
t
a
l
.
(
1
9
9
2
)
1
3
1
.
2
3
.
8
(
1
.
3

8
.
9
)
N
R
N
R
5
.
0
(
1
.
5

7
9
)
N
R
N
R
N
R
N
R
G
F
C
H
c
(
1
9
9
3
)
1
1
0
0
.
9
3
.
8
8
1
6
2
6
.
5
N
R
N
R
4
N
R
H
e
e
r
e
m
a
e
t
a
l
.
(
1
9
9
3
)
8
1
.
0
3
.
5
(
2
.
3

8
.
5
)
N
R
N
R
1
9
(
2
.
5

3
3
)
N
R
9
4
(
8
5

9
9
)
N
R
N
R
H
e
i
n
o
n
e
n
a
n
d
M
a
h
l
a
m
a
k
i
(
1
9
9
6
)
9
2
.
0
4
(
2

1
5
)
9
5
(
7
9

1
2
9
)
1
1
9
(
5
8

2
2
6
)
2
.
6
(
1
.
5

1
5
)
N
R
9
5
(
9
0

1
0
0
)
N
R
N
R
R
a
i
m
o
n
d
i
e
t
a
l
.
(
1
9
9
6
)
1
8
2
1
.
0
3
.
7
(
0
.
9

1
8
)
N
R
N
R
6
.
7
(
0
.
8

3
3
2
)
N
R
N
R
1
.
7
N
R
H
a
a
s
e
t
a
l
.
(
1
9
9
8
)
1
4
1
.
8
2
(
1

1
4
)
N
R
N
R
1
3
(
2
.
0

1
2
0
)
5
6
(
1

9
2
)
9
2
(
7
5

9
8
)
N
R
N
R
F
o
r
e
s
t
i
e
r
e
t
a
l
.
(
2
0
0
0
b
)
3
0
4
N
R
3
.
7
(
0
.
7

1
5
)
N
R
N
R
7
(
1

3
2
0
)
N
R
N
R
2
2
H
e
e
r
e
m
a
e
t
a
l
.
(
2
0
0
0
)
4
8
0
1
.
0
N
R
N
R
N
R
N
R
N
R
N
R
4
0
F
o
r
e
s
t
i
e
r
e
t
a
l
.
(
2
0
0
8
a
)
1
6
8
1
.
0
3
.
9
(
1
.
1

1
5
)
N
R
N
R
6
.
2
(
0
.
5

1
6
9
)
N
R
N
R
0
.
5
0
.
5
S
h
a
r
a
t
h
k
u
m
a
r
e
t
a
l
.
(
2
0
0
8
)
3
8
1
.
4
3
.
8
(
1
.
2

1
6
)
N
R
N
R
8
.
9
(
0
.
9

4
2
)
N
R
N
R
N
R
N
R
B
M
,
b
o
n
e
m
a
r
r
o
w
;
C
N
S
,
c
e
n
t
r
a
l
n
e
r
v
o
u
s
s
y
s
t
e
m
;
F
,
f
e
m
a
l
e
;
M
,
m
a
l
e
;
N
R
,
n
o
t
r
e
p
o
r
t
e
d
;
P
B
,
p
e
r
i
p
h
e
r
a
l
b
l
o
o
d
;
W
B
C
,
w
h
i
t
e
b
l
o
o
d
c
e
l
l
.
a
M
e
d
i
a
n
v
a
l
u
e
s
;
r
a
n
g
e
(
i
f
r
e
p
o
r
t
e
d
)
i
s
g
i
v
e
n
i
n
p
a
r
e
n
t
h
e
s
e
s
.
b
T
h
i
r
d
I
n
t
e
r
n
a
t
i
o
n
a
l
W
o
r
k
s
h
o
p
o
n
C
h
r
o
m
o
s
o
m
e
s
i
n
L
e
u
k
e
m
i
a
.
c
G
r
o
u
p
e
F
r
a
n
c
a
i
s
d
e
C
y
t
o
g
e
n
e
t
i
q
u
e
H
e
m
a
t
o
l
o
g
i
q
u
e
.
HIGH HYPERDIPLOID CHILDHOOD ALL 641
Genes, Chromosomes & Cancer DOI 10.1002/gcc
Environmental Risk Factors
No parental exposure has been specically
associated with an increased risk of having a child
developing high hyperdiploid ALL. However, it
has been suggested that maternal and/or paternal
exposure to organic solvents or use of mind-
altering drugs, such as amphetamine, marijuana,
and cocaine, correlates with the presence of RAS
mutations in pediatric ALL (Shu et al., 2004).
Considering that RAS mutations have been
shown to be relatively common in high hyperdi-
ploid ALL (Wiemels et al., 2005; Paulsson et al.,
2008b), this may indirectly suggest that use of
such drugs may increase the risk for high hyper-
diploidy. On the other hand, Wiemels et al.
(2005) reported that high hyperdiploid ALL is
signicantly less common in children prenatally
exposed to paternal smoking, speculating that
smoking-associated mutagens may be toxic to the
hyperdiploid clone. Although one could thus
argue that a reasonable advice to parents-to-be, in
order to decrease their risk of having a child
developing high hyperdiploid ALL, would be to
stop using mind-altering drugs and to start smok-
ing, we would like to stress that further studies
are needed to conrm or refute these very pre-
liminary associations.
Turning from drugs to vitamins, an increased
risk for childhood ALL, albeit not specically
for the high hyperdiploid subtype, has been
associated with low folate intake during preg-
nancy (Thompson et al., 2001), although this
was not conrmed in a subsequent study (Dock-
erty et al., 2007). Interestingly, folate deciency
has been shown to induce aneuploidy of chro-
mosomes 17 and 21 in cultured lymphocytes,
suggesting a possible association between dis-
turbances in this metabolic pathway and hyper-
diploidy (Wang et al., 2004). Wiemels et al.
(2001) found that constitutional homozygosity
for certain polymorphisms in the MTHFR gene,
coding for the enzyme methylenetetrahydrofo-
late reductase involved in the folate metabolic
pathway, is less common in hyperdiploid cases.
In fact, these MTHFR alleles have been associ-
ated with a reduced risk of pediatric ALL in
general (Krajinovic et al., 2004). Further support
for involvement of folic acid metabolism comes
from the study by Gast et al. (2007) who
showed that different haplotypes of the MTRR
gene, also involved in this pathway, correlate
with increased or decreased risk of childhood
ALL. Unfortunately, they did not analyze dif-
ferent cytogenetic subgroups separately.
It has been hypothesized, based on data on
population mixing, international incidence-rate
comparisons, time trends, and socioeconomic sta-
tus, that pediatric B-lineage ALL occurring in the
childhood peak arises as a consequence of a rare,
abnormal response to a common infection
(Greaves, 1988; Kinlen, 1988). Support for this
hypothesis has emerged from case-control studies
showing that daycare attendance, used as a proxy
for early infections, is associated with a reduced
risk of ALL (Greaves, 2006). However, this has
not, as far as we know, been specically linked to
the high hyperdiploid subgroup.
After a meta-analysis of several epidemiological
studies, Hjalgrim et al. (2003) concluded that
children weighing more than 4 kg at birth were
at increased risk of developing acute leukemia, in
particular ALL. In a later population-based case-
control study, the same group not only conrmed
this association but also showed that birth weight
was a risk factor for high hyperdiploid as well as
for t(12;21)-positive ALL (Hjalgrim et al., 2004).
The underlying mechanisms are unknown, but
could involve levels of insulin-like growth factor
1 and/or increased bone marrow volume, the lat-
ter yielding more cells vulnerable to leukemic
transformation (Hjalgrim et al., 2004).
CLINICAL CHARACTERISTICS
Peripheral Blood Values
High hyperdiploid ALL is generally associated
with low WBC counts; the median is <10 10
9
/l
in most series (Table 2). In cases from our own
department, the median is 5.9 10
9
/l (range,
1.3129), with 35% of cases having >10 10
9
/l
and 10% having >50 10
9
/l (unpublished data).
Thus, based on WBC counts quite few patients
would be stratied into high risk protocols. Data
on peripheral blast percentages are scarce, but
have been reported to be 3060% (Table 2);
these rather low frequencies should denitely be
taken into consideration when performing, for
example, array-based analyses of DNA extracted
from peripheral blood.
The patients are generally moderately anemic
at the time of diagnosis, with the median hemo-
globin levels generally varying between 60 and
75 g/l (Table 2). Most patients are also thrombo-
cytopenic, usually having median levels <50
10
9
/l. In our own data set, the median level is
42 10
9
/l (range, 5432), but it should be noted
642 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
that 15% of the patients have normal platelet
levels (unpublished data).
Extramedullary Leukemia
Extramedullary leukemia in the form of media-
stinal mass or central nervous system (CNS)
involvement, at the time of diagnosis is rare in
patients with high hyperdiploid ALL; as seen in
Table 2, the frequencies are less than 5%. Thus,
extramedullary leukemia is not a signicant factor
in this patient cohort.
Bone Marrow Morphology
In most instances, high hyperdiploid cases dis-
play a very high bone marrow blast percentage,
with the median being 95100% (Table 2). How-
ever, Knulst et al. (1993) reported a few patients
who presented with a smoldering high hyperdi-
ploid ALL where acute leukemia could not be
established by conventional morphological analy-
sis, i.e., the blast frequency was too low. The
prevalence of this phenomenon within the high
hyperdiploid group is unknown, but we are aware
of one such case at our department during the
last few years.
Several early studies showed that high hyperdi-
ploid ALL more often are of the FAB type L1
than L2, i.e., the cells are usually small and uni-
form and more rarely large and varied; they are in
all practice never L3 (Third International Work-
shop on Chromosomes in Leukemia, 1981b;
Williams et al., 1982; Heerema et al., 1985; Pui
et al., 1989; Secker-Walker et al., 1989; Kaspers
et al., 1995; Smets et al., 1995). However, there
are no characteristic morphological features that
can be used to deduce the presence of high
hyperdiploidy.
Immunophenotypic Features
Already in the initial report delineating high
hyperdiploid pediatric ALL, Kaneko et al. (1981)
noted that all cases had non-T-cell and non-
mature-B-cell markers, i.e., they belonged to the
group that today would be denoted B-cell precur-
sor. Subsequent studies during the 1980s showed
that the high hyperdiploid cases were character-
ized by the expression of the common (CD10)
ALL antigen (Third International Workshop on
Chromosomes in Leukemia, 1981a,b; Kaneko
et al., 1982; Williams et al., 1982; Heerema et al.,
1985; Smets et al., 1985; Harbott et al., 1987;
Heinonen et al., 1988; Michael et al., 1988; Pui
et al., 1988b; Fenaux et al., 1989; Secker-Walker
et al., 1989; Uckun et al., 1989). As more surface
markers were identied and used in immunophe-
notypic analyses, it gradually became apparent
that high hyperdiploid lymphoblasts typically are
positive for TdT, HLA-DR, CD10, CD19,
CD22, CD24, CD34, and CD66c and negative
for CD13, CD33, CD45, CD65, cIg, and sIg (Bor-
owitz et al., 1990; Behm et al., 1992; Groupe
Francais de Cytoge ne tique He matologique, 1993;
Lavabre-Bertrand et al., 1994; Raimondi et al.,
1996; Hrusa k et al., 1998; Hru sa k and Porwit-
MacDonald, 2002). However, in contrast to some
genetic subtypes, the immunophenotype cannot
be used to predict high hyperdiploidy.
OUTCOME
Lampert (1967) investigated 10 children with
ALL and noted that the four patients with 5059
chromosomes responded very well to induction
therapy and that their survival was particularly
long. Further support for a favorable outcome of
hyperdiploid ALL in general (>46 chromosomes)
was supplied a decade later by Secker-Walker
et al. (1978, 1979), who reported that such
patients had signicantly longer rst remissions
and better survival than those with diploid or
hypodiploid modal numbers. Subsequently,
Kaneko et al. (1981) described six children who
all achieved complete remission (CR) after induc-
tion treatment comprised only of vincristine and
prednisolone and who remained in CR for several
years. During the following years, numerous stud-
ies conrmed the favorable outcome of this cyto-
genetic subgroup (Kaneko et al., 1982; Williams
et al., 1982; Harbott et al., 1987; Heinonen et al.,
1988; Michael et al., 1988; Prigogina et al., 1988;
Bloomeld et al., 1989; Fenaux et al., 1989;
Secker-Walker et al., 1989), and by the end of
the 1980s, the favorable prognosis of high hyper-
diploid ALL had become widely recognized.
Event-Free and Overall Survival
As seen in Table 3, complete morphological
remission is achieved in the vast majority of high
hyperdiploid cases, i.e., initial treatment failure is
very rare. It is clear that this karyotypic feature is
associated with quite favorable event-free survival
(EFS) rates, often above 80% (Table 3). How-
ever, more dismal frequencies have been
reported, such as 73% at 5 years (Moorman et al.,
2003), and although late relapses are rare, they
HIGH HYPERDIPLOID CHILDHOOD ALL 643
Genes, Chromosomes & Cancer DOI 10.1002/gcc
have been noted to occur several years after ces-
sation of treatment (Dastugue et al., 1992). Very
recently, bone marrow brosis at diagnosis has
been associated with an increased risk of relapses
in the high hyperdiploid subgroup (Nore n-
Nystro m et al., 2008). The relapses most com-
monly involve the bone marrow (7080%), with
extramedullary involvement, mainly of the CNS,
occurring in a minority of cases (Pui et al., 1989;
Raimondi et al., 1996). However, it should be
noted that a trend toward an overrepresentation
of extramedullary relapses in this subgroup has
been reported (Sharathkumar et al., 2008).
Today, the overall survival (OS) rate of chil-
dren with high hyperdiploid ALL is 90% (Ta-
ble 3). Considering that the OS frequencies are
notably higher than the corresponding EFS rates
(Table 3), it is apparent that the treatment of
relapses generally is successful.
Possible Explanations for the Favorable Outcome
It has long been known that high hyperdiploid
ALL is particularly sensitive to chemotherapy,
especially to corticosteroids, mercaptopurine,
thioguanine, cytarabine, L-asparaginase (ASP),
and methotrexate (MTX) (Smets et al., 1987;
Synold et al., 1994; Kaspers et al., 1995). As
regards ASP, it has been shown that high hyper-
diploid blasts often display low expression of the
gene encoding asparaginase synthetase, some-
thing that may well explain the benecial effects
of asparagine depletion (Iwamoto et al., 2007).
The sensitivity to MTX is due to the increased
accumulation of MTX polyglutamates in the high
hyperdiploid blast cells (Whitehead et al., 1992,
1998; Synold et al., 1994). Intriguingly, the
SLC19A1 gene, which codes for the reduced fo-
late carrier that transports MTX into the cell, is
located at chromosome 21, which is trisomic or
tetrasomic in virtually all high hyperdiploid ALL.
Thus, the increased MTX accumulation may be
ascribed to additional copies of SLC19A1. In fact,
gain of chromosome 21 has been shown to be
associated with up-regulation of this gene in sev-
eral studies (Zhang et al., 1998; Belkov et al.,
1999; Cheng et al., 2005).
The successful response to chemotherapy is
most likely also linked to the activation of apo-
ptotic pathways in high hyperdiploid cells that
are already poised to die (Kersey, 1997).
Indeed, hyperdiploid blasts have been reported
to have quite a marked propensity to undergo
spontaneous apoptosis, both in vitro in stromal
TABLE 3. Outcome of Children with High Hyperdiploid B-Lineage ALL
Reference No. of cases CR (%) EFS (%) OS (%)
Kaneko et al. (1981) 6 100 NR NR
Kaneko et al. (1982) 9 100 NR NR
Williams et al. (1982) 41 95 NR NR
Heerema et al. (1985) 15 87 NR NR
Heinonen et al. (1988) 20 100 NR NR
Michael et al. (1988) 31 100 NR NR
Prigogina et al. (1988) 26 89 NR 40 (5 yr)
Bloomeld et al. (1989) 22 95 NR NR
Fenaux et al. (1989) 13 100 NR NR
Fletcher et al. (1989) 35 NR 80 (5 yr) 88 (5 yr)
Secker-Walker et al. (1989) 17 100 75 (5 yr) NR
Jackson et al. (1990) 146 100 75 (5 yr) NR
Hayashi et al. (1991) 13 100 NR NR
Rubin et al. (1991) 29 NR 93 (4 yr) NR
GFCH
a
(1993) 110 NR 85 (4 yr) NR
Kobayashi et al. (1994) 47 NR 74 (4 yr) NR
Raimondi et al. (1996) 105/63
b
NR 72/86 (5 yr) NR
Chessells et al. (1997) 195 NR 71 (5 yr) NR
Rubnitz et al. (1997) 55 NR 81 (5 yr) NR
Heerema et al. (2000) 480 NR 79 (8 yr) NR
Moorman et al. (2003) 807 NR 73 (5 yr) 91 (5 yr)
Moghrabi et al. (2007) 82 NR 86 (5 yr) NR
Forestier et al. (2008a) 168 NR 81 (10 yr) 89 (10 yr)
Sharathkumar et al. (2008) 38 NR 71 (5 yr) 93 (5 yr)
CR, complete remission; EFS, event-free survival; NR, not reported; OS, overall survival.
a
Groupe Francais de Cytogenetique Hematologique.
b
Cases with 5155 and 5667 chromosomes, respectively.
644 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
cultures and in vivo in bone marrow (Kumagai
et al., 1996; Ito et al., 1999; Zhang et al., 2002).
This may to some extent be explained by the rel-
atively high expression of the proapoptotic CAS-
P8AP2 gene at 6q15 in high hyperdiploid ALL
(Flotho et al., 2006).
Cytogenetic Features Inuencing the Outcome
Several studies have suggested that certain kar-
yotypic patterns and aberrations have a major
prognostic impact within the high hyperdiploid
subgroup, including modal numbers, specic tris-
omies, and structural changes.
Some investigators have reported a superior
outcome for cases with higher (>53 or >55)
modal chromosome numbers (Raimondi et al.,
1996; Heerema et al., 2000; Moorman et al.,
2003). However, Moorman et al. (2003) con-
cluded that this effect may simply be due to the
fact that such ALL are more likely to harbor 4,
10, and 18.
That certain trisomies might be associated with
prognosis was initially proposed by Jackson et al.
(1990), who correlated the presence of 6 with
improved EFS; however, their investigation also
included cases with 4750 chromosomes and was
hence not restricted to the high hyperdiploid sub-
group. Furthermore, an association between gain
of chromosome 6 and outcome has not been con-
rmed in subsequent studies. Harris et al. (1992)
reported increased EFS for high hyperdiploid
cases with both 4 and 10 compared with those
with trisomy of only one or none of these chro-
mosomes. However, Raimondi et al. (1996) could
not conrm that 4 and 10 inuence the prog-
nosis. Heerema et al. (2000) concluded that
whereas 4 and 6 did not affect the prognosis,
concurrent 10 and 17 were associated with a
superior outcome and 5 with a poor prognosis.
On the other hand, Moorman et al. (2003) found
an association between improved outcome and
trisomies 4, 10, and 18, but with only 4 and
18 having an independent impact in multivari-
ate analysis. Although these results seem conict-
ing, most studies have detected specic effects of
trisomies 4, 10, 17, and 18 in univariate analyses,
while the conclusions drawn from the multivari-
ate analyses have differed (Moorman et al., 2003).
In a study of >5000 pediatric B-cell precursor
ALL, Sutcliffe et al. (2005) showed that simulta-
neous trisomies 4, 10, and 17 were associated
with a very favorable outcome. Such triple triso-
mies are now used to stratify ALL as low stand-
ard risk in the US (Schultz et al., 2007).
However, to the best of our knowledge, specic
trisomies are not risk stratifying in European
treatment protocols.
A small subset of high hyperdiploid ALL har-
bors both near-haploid and high hyperdiploid
clones, with the latter representing a doubling of
the former (see below). It should be stressed that
such cases share the dismal outcome of near-hap-
loid ALL in general and that they should hence
be distinguished from the classic high hyperdi-
ploid cases (Harrison and Johansson, 2009).
As regards structural chromosome changes in
high hyperdiploid ALL, Pui et al. (1989) reported
that the presence of such aberrations in general
was associated with an increased risk of treatment
failure. However, this was not conrmed in sub-
sequent, larger series (Raimondi et al., 1996;
Moorman et al., 2003). Among the common struc-
tural abnormalities in high hyperdiploid ALL,
only i(17q) has been correlated with poor out-
come in some (Pui et al., 1988a; Raimondi et al.,
1996) but not all studies (Forestier et al., 2000b;
Moorman et al., 2003). Furthermore, there
are rare instances of high hyperdiploid cases har-
boring the ALL-specic rearrangements t(1;19)
(q23;p13), t(9;22)(q34;q11), or 11q23 transloca-
tions. In such cases, the prognostic impact of the
translocation is believed to override the benecial
effect of the high hyperdiploidy, and they are
hence usually not included in this karyotypic sub-
group (Forestier et al., 2000b; Moorman et al.,
2003).
GENETIC FEATURES
Obtaining Cytogenetic Data
As alluded to above, high hyperdiploid blast
cells are notoriously difcult to culture in vitro,
something that explains the fact that many cases
present as karyotypically normal or cytogenetic
failures (Harrison et al., 2005). Thus, to identify
all high hyperdiploid ALL alternative methods,
such as interphase FISH and DNA index analy-
ses, are mandatory (Look et al., 1982; Smets
et al., 1985; Berger et al., 1994; Martin et al.,
1996; Kasprzyk and Secker-Walker, 1997; Ritter-
bach et al., 1998; Nordgren et al., 2002; Pere z-
Vera et al., 2004; Nygaard et al., 2006). The dif-
culties in culturing also explain the lack of high
hyperdiploid cell lines; to the best of our knowl-
edge, there is only one established cell line
HIGH HYPERDIPLOID CHILDHOOD ALL 645
Genes, Chromosomes & Cancer DOI 10.1002/gcc
available (MHH-CALL-2) (Tomeczkowski et al.,
1995). Unfortunately, this precludes many in-
depth functional studies of high hyperdiploid
ALL.
Numerical Chromosome Aberrations
On the basis of conventional chromosome
banding analyses, it is quite clear that there is a
nonrandom pattern of chromosomal gains in high
hyperdiploid ALL, with additional copies of chro-
mosomes X, 4, 6, 10, 14, 17, 18, and 21 being
present in more than half of the cases (Fig. 4).
Chromosomes 8 and 5 are gained in 40% and
30%, respectively, whereas all other chromosomes
are supernumerary in only 1020%. As seen in
Figure 4, chromosome 21 is most frequently
gained; three or more copies are found in 90%
of cases. However, it should be emphasized that
even this high frequency is an underestimate:
FISH analyses reveal 21 in 100% of cases
(Moorman et al., 1996; Elghezal et al., 2001;
Paulsson et al., 2005). Hence, we would like to
suggest that gain of this chromosome is a sine
qua non for high hyperdiploid ALL.
By chromosome banding analysis, chromosome
21 is tetrasomic, sometimes even pentasomic, in
60% of cases (Fig. 4); using FISH this frequency
increases to 85% (Elghezal et al., 2001; Paulsson
et al., 2005). The remaining numerical changes
are usually trisomies, but tetrasomies do occur, in
particular involving chromosomes X, 14, and 18
(Heerema et al., 2007; Fig. 4). In contrast, monos-
omies are seldom, if ever, seen (Mitelman et al.,
2009).
The rare high hyperdiploid ALL originating
via a near-haploid clone (see below) harbor only
disomies and tetrasomies, with the latter most
often being tetrasomies X, 14, 18, and 21. They
can hence be distinguished cytogenetically quite
easily, even in the absence of the near-haploid
clone (Onodera et al., 1992a).
Structural Chromosome Aberrations
Approximately 50% of high hyperdiploid ALL
harbor cytogenetically identiable structural chro-
mosome aberrations in addition to the chromo-
somal gains (Groupe Francais de Cytoge ne tique
He matologique, 1993; Raimondi et al., 1996;
Forestier et al., 2000a; Moorman et al., 2003). In
addition, further structural changes are frequently
found when cases are analyzed with multicolor
FISH (Elghezal et al., 2001; Nordgren et al.,
2001; Paulsson et al., 2005).
Balanced chromosome aberrations
In the Mitelman Database of Chromosome
Aberrations in Cancer (Mitelman et al., 2009), 5%
of high hyperdiploid ALL carry balanced abnor-
malities, mainly translocations. In approximately
half of these, well-known ALL-associated tran-
slocations, most often t(1;19)(q23;p13) or t(9;22)
(q34;q11), are seen; such cases are associated
with the same chromosomal gains as high hyper-
diploid ALL in general, with the exception that
trisomy 2 is quite frequent in t(9;22)-positive
high hyperdiploid ALL (Mitelman et al., 2009).
It should be noted that none of the remaining
balanced rearrangements identied to date have
Figure 4. Chromosome gains in 414 high hyperdiploid B-lineage ALL (<18 years, modal chromosome
number 5167, not including incomplete karyotypes) reported in the literature (Mitelman et al., 2009).
Xf denotes gain of the X chromosome in females; Xm in males. Trisomy and Tetrasomy of Xm and
Y correspond to one and two extra copies of these chromosomes, respectively.
646 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
been recurrent, something that may argue against
the presence of a common fusion gene in this
cytogenetic subgroup.
Unbalanced chromosome aberrations
Most structural chromosome changes in high
hyperdiploid ALL are unbalanced, i.e., result in
gain or loss of genetic material. The most com-
mon are partial gains of 1q, deletions of 6q, and
isochromosomes of 7q and 17q (Pui et al., 1992;
Raimondi et al., 1996; Moorman et al., 2003).
Gain of 1q, via either duplications or unbal-
anced translocations, is found in 1015% of high
hyperdiploid cases (Groupe Francais de Cytoge -
ne tique He matologique, 1993; Raimondi et al.,
1996; Moorman et al., 2003). Recently, detailed
genetic and expression analyses revealed up-regu-
lation of ve genes (B4GALT3, DAP3, RGS16,
MEM183A, and UCK2) located in the minimally
duplicated 57 Mb segment at 1q2232.3 (Davids-
son et al., 2007), but it remains to be seen
whether this overexpression is pathogenetically
important.
Approximately 5% of high hyperdiploid ALL
harbor deletions involving the long arm of chro-
mosome 6 (Raimondi et al., 1996; Moorman
et al., 2003). Interestingly, Moorman et al. (2003)
noted that 6q deletions were usually independent
of 1q gains, which could suggest that these abnor-
malities represent alternative leukemogenic path-
ways. As of yet, no minimally deleted region on
6q has been identied in high hyperdiploid ALL
using G-banding or FISH. Furthermore, genome-
wide array-based analyses have not disclosed any
recurrent microdeletions on 6q (Paulsson et al.,
2006; Davidsson et al., 2007; Kuiper et al., 2007;
Mullighan et al., 2007; Strefford et al., 2007).
Thus, the biological impact of 6q deletions
remains unclear.
An i(7q) is present in 12% of high hyper-
diploid cases (Pui et al., 1992; Martineau et al.,
1996). Although usually denoted isochromosome,
FISH mapping has shown that it actually is an
isodicentric aberration with breakpoints in
7p11.2. No known gene is located in the break-
point region, indicating that i(7q) does not result
in a fusion gene and that the functional outcome
is the resulting loss of 7p and/or 7q duplication
(Schaad et al., 2006).
Between 2% and 5% of high hyperdiploid ALL
have been reported to harbor i(17q) (Martineau
et al., 1996; Raimondi et al., 1996; Moorman
et al., 2003). Since trisomy 17 is one of the char-
acteristic numerical anomalies in high hyperdi-
ploid cases (Fig. 4), it is noteworthy that 17 and
i(17q) seems to be mutually exclusive (Mitelman
et al., 2009). This may indicate that the pathoge-
netic consequence of trisomy 17 is gain of the
long arm.
Fusion Genes
As mentioned earlier, ALL-specic transloca-
tions resulting in fusion genes are occasionally
detected in high hyperdiploid cases. To date,
only four different chimeric genes, namely BCR/
ABL1 (Heerema et al., 2004), ETV6/RUNX1 (For-
estier et al., 2007), TCF3/HLF (Inukai et al.,
2007), and TCF3/PBX1 (Troussard et al., 1995),
have been identied in high hyperdiploid ALL.
It should be emphasized that cases with these
fusions are extremely rare, most likely comprising
less than 1% of the high hyperdiploid subgroup.
Interestingly, as regards TCF3/PBX1 several
investigators have noted that high hyperdiploid
ALL with t(1;19)(q23;p13) or der(19)t(1;19)
(q23;p13) rarely expresses this fusion transcript,
in contrast to the vast majority of t(1;19)-positive
non-high hyperdiploid ALL (Privitera et al.,
1992; Gaynon et al., 1997; Barber et al., 2007).
The reason for this remains unknown, but may
be due to different genomic breakpoints in the
TCF3 and PBX1 genes, as has been detected in a
few t(1;19)-positive ALL (Paulsson et al., 2007).
Alternatively, the 1q23 and 19p13 breakpoints
may involve other genes.
Thus, there is no known fusion gene speci-
cally associated with high hyperdiploid ALL to
date. Whether this reects evidence of absence
or absence of evidence is presently not clear.
The fact that there are no recurrent translocations
in high hyperdiploid ALL, with the exceptions
mentioned earlier, could suggest a true absence
of chimeric genes in this subgroup. Furthermore,
breakpoint mapping of inversions, insertions, and
translocationsbalanced as well as unbalanced
in high hyperdiploid cases reveals no pronounced
clustering of breaks, apart from the ones associ-
ated with t(1;19), t(9;22), and t(12;21) (Fig. 5).
However, the presence of fusion genes generated
through as yet unknown rearrangements in high
hyperdiploid cases cannot be excluded.
Microdeletions
With the advent of various array-based high-
resolution screening techniques, it has become
HIGH HYPERDIPLOID CHILDHOOD ALL 647
Genes, Chromosomes & Cancer DOI 10.1002/gcc
increasingly apparent that microdeletions are a
characteristic feature of ALL in general (Kuiper
et al., 2007; Mullighan et al., 2007; Paulsson
et al., 2008a). This also holds true for high hyper-
diploid cases, although the overall frequency of
such cryptic deletions seems to be lower than in
most other genetic ALL subgroups (Mullighan
et al., 2007). Using FISH or genome-wide arrays,
the loci thus far shown to be recurrent targets of
microdeletions in high hyperdiploid ALL are
located at 7p12.2 (IKZF1), 9p21.3 (CDKN2A),
9p13.2 (PAX5), 12p13.2 (ETV6), 13q14.2 (RB1),
and 19p13.3 (TCF3) (Mart nez-Ram rez et al.,
2001; Nordgren et al., 2002; Paulsson et al., 2006;
Figure 5. Breakpoint distribution of inversions, insertions, and
translocationsbalanced as well as unbalancedin 505 high hyperdi-
ploid B-lineage ALL (<18 years, modal chromosome number 5167)
reported in the literature (Mitelman et al., 2009). The breakpoint
clusters seen at 1q23, 9q34, 12p13, 19p13, 21q22, and 22q11 corre-
spond to the well-known ALL-associated translocations
t(1;19)(q23;p13), t(9;22)(q34;q11), and t(12;21)(p13;q22).
648 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
Barber et al., 2007; Davidsson et al., 2007; Mul-
lighan et al., 2007; Strefford et al., 2007; Sulong
et al., 2009).
The chromosomes with recurrent microdele-
tions, i.e., chromosomes 7, 9, 12, 13, and 19, are
rarely gained in high hyperdiploid ALL, making
the deletions easily identiable by single nucleo-
tide polymorphism (SNP) array analyses. In
contrast, a microdeletion in one of the three
homologues of a trisomic chromosome could
prove difcult to detect by this technique. Thus,
the frequency of microdeletions in high hyperdi-
ploid ALL may well be an underestimate
because of the multiple gains present in these
cases.
Gene Mutations
Mutations in the FLT3, KRAS, NRAS, and
PTPN11 genes have been shown to be relatively
frequent in high hyperdiploid ALL, with activat-
ing FLT3 mutations being found in 1025%,
KRAS/NRAS mutations in 1530%, and PTPN11
mutations in 1015% (Armstrong et al., 2004;
Taketani et al., 2004; Tartaglia et al., 2004; Wie-
mels et al., 2005; Stam et al., 2007; Paulsson
et al., 2008b). The mutations are usually mutually
exclusive, something that may be due to the fact
that all four genes are involved in the RTK-RAS
signaling pathway. Taken together, a substantial
proportionapproximately one thirdof high
hyperdiploid cases harbors a FLT3, KRAS, NRAS,
or PTPN11 mutation (Paulsson et al., 2008b).
Copy Neutral Loss of
Heterozygosity/Uniparental Isodisomy
The term UPID, originally coined in the eld
of constitutional genetics, denotes the presence
of two identicalmaternal or paternalhomolo-
gous chromosomes. Quite recently, this phenom-
enon has also been observed in neoplastic cells,
where it is detectable with SNP array analyses as
LOH without concurrent copy number changes
(Raghavan et al., 2005). These acquired aberra-
tions may either involve whole chromosomes
(wUPID) or parts of chromosomes (pUPID). In
acute myeloid leukemia (AML) and chronic mye-
loproliferative disorders, UPIDs have been shown
to lead to homozygosity for mutations in genes
such as CEBPA, FLT3, and JAK2 (Fitzgibbon
et al., 2005; Baxter et al., 2005). In ALL, a similar
mechanism renders CDKN2A deletions homozy-
gous in 9p (Mullighan et al., 2007; Kawamata
et al., 2008; Paulsson et al., 2008a).
Although there are a few examples of pUPIDs
in high hyperdiploid ALL, in particular for 9p
(Mullighan et al., 2007), they seem to be less
common in this subtype as compared with pediat-
ric ALL in general (Kawamata et al., 2008). The
reason for this is unknown. In contrast, there
appears to be an overrepresentation of wUPIDs
in high hyperdiploid cases (Kawamata et al.,
2008). As discussed below, this most likely
reects the underlying mechanism behind the
hyperdiploidy and may not be pathogenetically
important as such.
Epigenetic Changes
The eld of epigenetics in cancer, including
DNA methylation and histone modications, has
expanded dramatically in the past few years.
Although several groups have investigated hyper-
methylation of tumor suppressor genes in child-
hood ALL, only few have stratied data
according to genetic subtypes; no histone modi-
cation study has been published. Thus far, the
promoters of the following 19 genes have been
reported to be targeted by CpG methylation in
some high hyperdiploid ALL: APC, CADM1,
CD44, CDH13, CDKN1A, CDKN2B, CHFR,
DKK3, ESR1, FHIT, GATA5, MSH6, PAX6,
RARB, THBS1, TIMP3, TP73, WNT5A, and WT1
(Roman-Gomez et al., 2002, 2004, 2007; Zheng
et al., 2004; Paulsson et al., 2009). Of these,
CADM1, ESR1, FHIT, RARB, and WNT5A are
hypermethylated in more than 50% of cases.
Taken together, aberrant methylation of tumor
suppressor gene promoters seems to be a com-
mon phenomenon in high hyperdiploid ALL
(Paulsson et al., 2009).
Is High Hyperdiploid ALL Karyotypically
Stable or Not?
Chromosomal instability, detectable as cell-to-
cell variation in chromosomal content and the
presence of multiple subclones, is a characteristic
feature of many malignant disorders. On the basis
of chromosome banding analyses, subclones
usually differing from the stemline by one to
three additional chromosomes or structural rear-
rangementsare seen in 1520% of high hyper-
diploid ALL (Raimondi et al., 1996; Mitelman
et al., 2009), but since only one subclone is found
in most instances they probably reect clonal
HIGH HYPERDIPLOID CHILDHOOD ALL 649
Genes, Chromosomes & Cancer DOI 10.1002/gcc
evolution rather than karyotypic instability. In
contrast, there are some reports suggesting that
cell-to-cell variation, as detected with interphase
FISH analyses of the most common trisomies and
tetrasomies, is quite frequent (Betts et al., 2001;
Blandin et al., 2008). However, our own experi-
ence does not support this conclusion; only rarely
do we nd signicant differences in interphase
FISH signal patterns in high hyperdiploid ALL
(unpublished observation). Hence, further studies
are clearly needed to determine whether high
hyperdiploid ALL is karyotypically stable or not.
It should be noted that investigations of IGH@
rearrangements, performed as part of minimal re-
sidual disease analyses, quite often detect appa-
rent oligoclonality. However, in the vast majority
of cases such results can be explained by the fre-
quent occurrence of trisomy or tetrasomy 14,
leading to additional IGH@ alleles. Thus, mini-
mal residual disease analyses do generally not
provide any evidence for true oligoclonality in
high hyperdiploid ALL (Csinady et al., 2009).
PRENATAL ORIGIN
Almost half a century ago, MacMahon and
Levy (1964) reported that childhood leukemia
could have a prenatal origin, a proposition based
on twin studies showing that monozygous twins
are at substantial risk of concordance for this dis-
ease. Clarkson and Boyse (1971) developed this
idea further by suggesting that the high concord-
ance rate could represent only one occurrence of
leukemianot twowith the leukemic cells
circulating into the other twin through shared
placental vessels. This has since been clearly
demonstrated by the identication of identical
clonotypic breakpoints in concordant leukemias
characterized by various ALL-associated fusion
genes. In addition, chimeric genes have also been
detected in retrospective analyses of archived
neonatal blood spots from children who later
developed ALL. Thus, many pediatric ALL, or
rather preleukemic clones, arise in utero, with
additional postnatal events, most likely genetic
changes, being necessary for overt disease
(Greaves, 2005).
As regards high hyperdiploid ALL, the lack of
leukemia-specic fusion genes precludes similar
analyses of clone-specic breakpoints. However,
there is ample evidence for a prenatal origin of
also this genetic subgroup. First, Yagi et al.
(2000), Taub et al. (2002), Maia et al. (2004), and
Gruhn et al. (2008) demonstrated clonotypic
IGH@ rearrangements in Guthrie cards from
patients who later developed high hyperdiploid
ALL. Second, Panzer-Gru mayer et al. (2002), tak-
ing into account the fact that the IGH@ gene is
located on chromosome 14, showed that three
different IGH@ rearrangements and hence 14
were present already at the time of birth, strongly
suggesting that trisomy of at least this chromo-
some had taken place in utero. Third, Maia et al.
(2003) reported shared IGH@ rearrangements,
and similar chromosomal gains, in a pair of mono-
zygous twins with high hyperdiploid ALL.
Finally, Maia et al. (2004) supplied direct evi-
dence for the presence of trisomies in utero.
They investigated a child with high hyperdiploid
ALL who, by chance, happened to have had his
cord blood cells collected and frozen at birth,
and showed by interphase FISH analyses that
CD34CD19 cord blood cells harbored triso-
mies for chromosomes 15 and 17, as did the
blasts at diagnosis of ALL. Taken together, these
ndings show that hyperdiploidy is a prenatal
event in at least some cases of childhood ALL
and provide strong support for the possibility that
all such cases arise in utero.
CELL OF ORIGIN
Cancer stem cells, the existence of which was
suggested already in the early 1960s (Bruce and
Van Der Gaag, 1963), have received much atten-
tion during recent years, and it is now generally
accepted that hematologic malignancies are sus-
tained by leukemic stem cells capable of both
initiating and maintaining the disease. In leuke-
mias, the presence of different gene fusions, for
example ETV6/RUNX1, in well-dened cell popu-
lations has been used to identify the affected lin-
eages, hence providing circumstantial evidence
for candidate leukemia stem cells (Castor et al.,
2005).
Only few studies have focused on the cell of
origin in high hyperdiploid ALL. Larramendy
et al. (1995), using the morphology-antibody-
chromosomes technique and interphase FISH for
trisomic chromosomes, investigated ve high
hyperdiploid ALL and found that the gains were
restricted to cells expressing lymphoid markers in
four cases. In the remaining ALL, also glyco-
phorin A-positive (GPA) cells harbored the tris-
omy. Since these cells were morphologically
normal maturing erythroblasts, aberrant GPA
expression on lymphoblasts was considered
unlikely. They thus concluded that high
650 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
hyperdiploid ALL, at least in some instances,
may be a stem cell disease. Support for this con-
clusion was provided by Quijano et al. (1997),
who analyzed immunophenotypically character-
ized populations with interphase FISH for one or
two of the gained chromosomes and found that
CD34CD33CD38CD19 cells, i.e., the
primitive compartment that in normal bone mar-
row contains the stem cells, were trisomic in 25%
of the high hyperdiploid cases. However, Kaspr-
zyk et al. (1999), investigating B (CD19), T
(CD3), myeloid (CD13), and erythroid
(GPA) cells with interphase FISH, detected
trisomies only in the B cells, suggesting that high
hyperdiploidy arises in a lymphoid-committed
progenitor cell, although a pluripotent stem cell
origin with subsequent lineage restriction could
not be excluded. Although the latter possibility
still remains and further studies are denitely
needed, one should note that the high cure rate
of high hyperdiploid ALL with chemotherapy
alone would indicate that it does not originate in
a hematopoietic stem cell.
FORMATION
An important as well as intriguing question is
how the cell becomes high hyperdiploid. Theo-
retically, hyperdiploidy may arise through four
different mechanisms: (a) by initial near-haploidy
followed by doubling of the chromosomes, (b) by
tetraploidization with subsequent chromosome
losses, (c) by sequential gains of chromosomes in
consecutive cell divisions, or (d) by a simultane-
ous gain of chromosomes in a single abnormal mi-
tosis (Fig. 6) (Onodera et al. 1992b). Since we
cannot directly observe a high hyperdiploid ALL
in statu nascendi, evidence for and against the
above mechanisms is by necessity circumstantial;
investigations of allelic ratios of loci on tetrasomic
chromosomes and of wUPID frequencies may
provide valuable information (Fig. 6).
Near-Haploid Pathway
Near-haploidy (2529 chromosomes) is a rare
but recurrent cytogenetic feature of childhood B-
lineage ALL that is similar to high hyperdiploid
ALL in that a specic pattern of numerical
changes is seen, with most cases retaining diso-
mies for 21, X/Y, 14, and 18 (Harrison and
Johansson, 2009). However, in contrast to clas-
sic high hyperdiploid cases, near-haploid ALL is
associated with a dismal prognosis (Brodeur et al.,
1981; Gibbons et al., 1991; Harrison and Johans-
son, 2009).
Since the mid 1970s, it has been known that
near-haploid cases may harbor two clones: one
with a near-haploid chromosomal content and
one with a high hyperdiploid karyotype repre-
senting an exact duplicate of the stemline
(Kessous et al., 1975; Oshimura et al., 1977). Sub-
sequent investigations have shown that such
related clones are quite common in near-haploid
ALL; for example, Harrison et al. (2004) reported
a frequency of 65%. This has raised the possibil-
ity that some high hyperdiploid cases may origi-
nate as near-haploid, with subsequent duplication
of chromosomes and loss of the original stemline,
at least at the cytogenetic level (Brodeur et al.,
1981; Gibbons et al., 1991; Onodera et al., 1992a).
High hyperdiploid ALL arising via a near-hap-
loid state is cytogenetically distinct, being charac-
terized by the presence of only disomies and
tetrasomies; trisomies are rarely, if ever, seen
(Mitelman et al., 2009). Furthermore, such cases
have wUPIDs for all disomic chromosomes and
2:2 allelic ratios on all tetrasomic chromosomes
(Fig. 6A). Onodera et al. (1992a) showed that all
loci at disomic chromosomes displayed LOH, in-
dicative of wUPIDs, in a high hyperdiploid case;
a similar case has also been reported by us
(Paulsson et al., 2005). Furthermore, analysis of
the data set published by Mullighan et al. (2007),
including 39 high hyperdiploid ALL, reveals one
additional case. Also, FISH studies have revealed
near-haploid clones in interphase cells in some
high hyperdiploid cases (Ma et al., 1998; Stark
et al., 2001). Hence, it is clear that a proportion
of high hyperdiploid ALL originates through a
near-haploid pathway. However, it is equally
clear that most cases do not. As described in
detail below, the vast majority of high hyperdi-
ploid cases display retained heterozygosity of the
disomic chromosomes, in effect excluding a near-
haploid route. Furthermore, most gains in high
hyperdiploid ALL are trisomies (Fig. 4).
Finally, we would like to stress that the clinical
differences between near-haploid and high hyper-
diploid ALL makes it incumbent upon diagnostic
laboratories to identify the rare near-haploid cases
masquerading as high hyperdiploid ALL to
ensure correct risk stratication.
Tetraploid Pathway
A second possible mechanism leading to high
hyperdiploidy is initial tetraploidy with subsequent
HIGH HYPERDIPLOID CHILDHOOD ALL 651
Genes, Chromosomes & Cancer DOI 10.1002/gcc
Figure 6. Four possible mechanisms for the formation of high
hyperdiploidy. (A) Doubling of a near-haploid set of chromosomes,
resulting in whole chromosome uniparental isodisomies (wUPIDs) for
all disomies and 2:2 allelic ratios for all loci on tetrasomic chromo-
somes. (B) Initial tetraploidization with subsequent loss of chromo-
somes, resulting in wUPIDs for approximately one third of the
disomic chromosomes and 2:2 allelic ratios for all loci on tetrasomic
chromosomes. (C) Sequential gains of chromosomes in consecutive
cell divisions, resulting in no wUPIDs and 3:1 allelic ratios, corre-
sponding to triplication of one of the homologues and retention of
the other, for approximately two thirds of the tetrasomic chromo-
somes. (D) Simultaneous gain of chromosomes in a single abnormal
mitosis, resulting in no wUPIDs and 2:2 allelic ratios for all loci on
tetrasomic chromosomes. This gure was originally published in
Blood. Paulsson K, Panagopoulos I, Knuutila S, Jee KJ, Garwicz S,
Fioretos T, Mitelman F, Johansson B. Formation of trisomies and their
parental origin in hyperdiploid childhood acute lymphoblastic leukae-
mia. Blood. 2003;102:30103015, VC
the American Society of
Hematology.
652 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
loss of chromosomes. The allelic ratios of loci on
tetrasomic chromosomes will be 2:2 since both
homologues have been duplicated. Furthermore,
when four homologues are reduced to two, on av-
erage one third of the resulting disomies will be
wUPIDs in the absence of selective forces (Fig.
6B).
Virtually all reported allelic ratios of tetrasomic
chromosomes in high hyperdiploid cases have
been 2:2 (Onodera et al., 1992b; Paulsson et al.,
2003, 2005; Haas et al., 2004). Since this is
expected also in cases arising via near-haploidy
and by a simultaneous gain of chromosomes (Fig.
6), this nding cannot be used to identify cases
originating by the tetraploid pathway. Hence, the
pattern of wUPIDs is essential. A total of 52 high
hyperdiploid cases with SNP array or microsatel-
lite data on all disomies have been reported (Irv-
ing et al., 2005; Paulsson et al., 2005; Mullighan
et al., 2007). Of these, 30% displayed a pattern
of wUPIDs consistent with a tetraploid pathway,
but this does not by necessity reect the true fre-
quency of cases originating via tetraploidy. First,
a case arising through a different mechanism may
subsequently acquire wUPIDs, in particular if
they provide a selective advantage. However, the
reported wUPIDs have generally involved differ-
ent chromosomes, making selection for a specic
mutated gene or an imprinted region unlikely.
Second, in cases with high modal chromosome
numbers, lack of wUPIDs may simply reect the
fact that only few disomies are present, making it
impossible to exclude a tetraploid pathway.
Nevertheless, the observed frequency of 30% is
the best estimate of the occurrence of this
mechanism.
Sequential Gains
Another putative pathway to hyperdiploidy is
sequential gains of chromosomes in consecutive
cell divisions (Fig. 6C). This is an attractive pos-
sibility, because it ts well with the current para-
digmatic view of clonal evolution in malignant
disorders. However, it is not supported by nd-
ings at the cytogenetic level; as mentioned ear-
lier, most high hyperdiploid ALL do not display
subclones (Raimondi et al., 1996; Mitelman et al.,
2009). At the molecular level, high hyperdiploidy
formed via this pathway is expected to display
allelic ratios of 3:1corresponding to triplication
of one of the homologues and retention of the
otherfor approximately two thirds of the tetra-
somic chromosomes and no wUPIDs (Fig. 6C).
As regards the latter, 70% of high hyperdiploid
cases do not harbor any wUPIDs (Irving et al.,
2005; Paulsson et al., 2005; Mullighan et al.,
2007), something that would agree with sequen-
tial gains. However, we know of only two cases
where 3:1 allelic ratios for tetrasomies have been
observed (Onodera et al., 1992b; Paulsson et al.,
2005). Taken together, while there may be rare
cases arising through sequential gains in consecu-
tive cell divisions, this mechanism can be
excluded in the vast majority of cases.
Simultaneous Gain
Intuitively, the fourth and nal pathway
simultaneous gain of all extra chromosomes in a
single abnormal cell divisionmay seem quite
unlikely, not least considering the very charact-
eristic and nonrandom pattern of additional
chromosomes. However, available data actually
support this mechanism for the majority of high
hyperdiploid ALL: the expected pattern would
be a 2:2 allelic ratio of loci on tetrasomic chromo-
somes and no wUPIDs (Fig. 6D), and this is
what is found in most cases (Onodera et al.,
1992b; Paulsson et al., 2003, 2005; Haas et al.,
2004; Irving et al., 2005; Mullighan et al., 2007).
Thus, in all practice high hyperdiploidy arises
either via a tetraploid pathway (30% of cases)
or by a simultaneous gain (70% of cases).
PATHOGENETIC IMPACT OF
HIGH HYPERDIPLOIDY
As for trisomies and tetrasomies in hematologic
malignancies in general, next to nothing is known
about how high hyperdiploidy results in ALL. In
fact, there is evidence to suggest that it is not by
itself sufcient for leukemogenesis; additional
genetic alterations may be needed for overt dis-
ease. Indeed, it is not even known whether all
trisomies/tetrasomies are necessary; the variation
in chromosomal content may suggest that they
are not, as would the fact that sequential gains
are very rare (see above). Perhaps some of the
gains, in particular the less frequent ones, are epi-
phenomena in the sense of innocent bystanders
that are tolerated but do not provide a selective
edge. Even so, some of the extra chromosomes
are undoubtedly pathogenetically important, with
possible leukemogenic consequences including
mutated genes on the duplicated chromosomes,
gains of imprinted loci, and gene dosage effects.
HIGH HYPERDIPLOID CHILDHOOD ALL 653
Genes, Chromosomes & Cancer DOI 10.1002/gcc
High Hyperdiploidy is Not Sufcient to
Cause Leukemia
As discussed earlier, several lines of evidence
comprising clonotypic IGH@ rearrangements, con-
cordant high hyperdiploid ALL in twins, and the
presence of trisomic cells in cord bloodstrongly
suggest that high hyperdiploidy frequently, possi-
bly always, occurs in utero (Yagi et al., 2000; Pan-
zer-Gru mayer et al., 2002; Taub et al., 2002; Maia
et al., 2003, 2004; Gruhn et al., 2008). However,
among the 22 patients in whom a prenatal origin
has been demonstrated, the median age at ALL di-
agnosis has been 2 years and 10 months, with the
oldest being 14 years old. Although we cannot
exclude the possibility that the cells investigated
in the aforementioned studies had not yet acquired
high hyperdiploidyfor most of the cases only clo-
notypic IGH@ rearrangements were demon-
stratedthe rather long latency period
nevertheless indicates that high hyperdiploidy is
not sufcient to cause leukemia; additional genetic
aberrations are necessary for overt disease. These
may include structural chromosome aberrations,
microdeletions, gene mutations, and epigenetic
changes. However, the temporal order of the vari-
ous genetic hits in relation to the high hyperdi-
ploidy remains to be elucidated.
Pathogenetic ImpactDuplication of
Gene Mutations?
Some neoplasia-associated trisomies have been
shown to be associated with duplication of a
mutated gene residing on the gained chromo-
some, namely 4 and KIT in AML, 7 and Hras
in mouse skin squamous cell carcinomas, 7 and
MET in hereditary papillary renal carcinomas,
and 10 and RET in pheochromocytomas (Bian-
chi et al., 1990; Zhuang et al., 1998; Huang et al.,
2000; Beghini et al., 2004). However, we deem
this an unlikely pathogenetic basis for high
hyperdiploidy because it would imply that each
trisomic chromosome should harbor a mutated
gene. Furthermore, the fact that the vast majority
of tetrasomies in high hyperdiploid ALL display
a 2:2 allelic ratio, i.e., duplication of both parental
homologues (Onodera et al., 1992b; Paulsson
et al., 2003, 2005; Haas et al., 2004), strongly
argues against duplication of a specic mutation.
Pathogenetic ImpactGains of Imprinted Loci?
That imprinting effects may be of importance
in high hyperdiploid ALL was rst suggested by
Haas (1996), who hypothesized that gains of
maternally or paternally derived chromosomes
could result in deregulation of imprinted loci. If
correct, this would be detectable as a skewed
parental origin of the extra chromosomes (Haas
1996). We previously addressed this issue in a
series of high hyperdiploid ALL, including paren-
tal samples, and found no evidence for preferen-
tial duplication of maternal or paternal
chromosomes (Paulsson et al., 2003, 2005), a nd-
ing subsequently conrmed by Wilson and Mori-
son (2005). Thus, gains of imprinted loci can be
excluded as a pathogenetically important mecha-
nism in high hyperdiploid ALL.
Pathogenetic ImpactGene Dosage Effects?
A quite likely pathogenetic outcome of the
additional chromosomes in high hyperdiploid
ALL is gene deregulation, i.e., increased expres-
sion of genes located on the trisomic and tetraso-
mic chromosomes. In fact, most global gene
expression analyses have revealed a general up-
regulation of such genes in this cytogenetic sub-
group (Ross et al., 2003; Gruszka-Westwood
et al., 2004; Andersson et al., 2005). However, it
should be noted that not all genes behave in this
manner; some even display decreased expression
(Gruszka-Westwood et al., 2004; Andersson et al.,
2005). Furthermore, it remains unknown whether
the high transcription really translates into higher
protein levels. This notwithstanding, high hyper-
diploid ALL displays a specic gene expression
signature that allows a very high classication
accuracy (Yeoh et al., 2002; Qiu et al., 2003; Ross
et al., 2003; Andersson et al., 2005; van Delft
et al., 2005). Interestingly, a large proportion of
the class-dening genes is located on chromo-
somes 21 and X (Yeoh et al., 2002; Andersson
et al., 2005).
At present, next to nothing is known about
which pathways that are affected by the global
gene expression changes or their contribution to
the leukemogenesis of high hyperdiploid ALL.
However, considering the high frequency of
FLT3 mutations and the possible involvement of
folic acid metabolism in high hyperdiploid cases
(see above), it is noteworthy that FLT3 overex-
pression as well as a specic expression pattern of
genes involved in the folate pathway have been
reported in this genetic subgroup (Brown et al.,
2005; Kager et al., 2005). Thus, further studies
are denitely warranted. In addition, alternative
mechanisms for gene deregulation, such as global
654 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
epigenetic changes or aberrant microRNA expres-
sion proles, may also be important, but have not
been investigated in high hyperdiploid ALL to
date.
CONCLUSIONS AND FUTURE DIRECTIONS
Since the high hyperdiploid subgroup of child-
hood ALL was rst delineated in the early 1980s,
it has become clear that it comprises 2530% of
all pediatric B-lineage ALL; that it is associated
with a very favorable outcome; that the chromo-
some gains are nonrandom and most commonly
involve X, 4, 6, 10, 14, 17, 18, and 21; that addi-
tional genetic abnormalities are frequent, agree-
ing well with a multistep leukemogenic scenario;
that the preleukemic clone oftenperhaps
alwaysis present already at birth; that the high
hyperdiploid pattern may arise via different
mechanisms but most commonly through a single
abnormal cell division; and that deregulated
gene expression probably is pathogenetically
important.
However, many questions remain. Why does
high hyperdiploidy display such a strong correla-
tion with young age? What are the prenatal and
postnatal etiologic factors associated with high
hyperdiploidy and subsequent overt leukemia?
Why do 1520% of cases relapse? What is the
temporal order of the various cooperating
genetic eventstrisomies, structural chromosome
changes, microdeletions, mutations et cetera? Are
there other, as yet unidentied, genetic or epige-
netic changes, such as cryptic fusion genes or
methylation hotspots, promoting leukemogenesis?
At which stage of the hematopoietic hierarchy
does high hyperdiploidy arise? What are the exact
mechanisms underlying the formation of the high
hyperdiploid pattern? Are there biological or clin-
ical differences between cases originating via a
tetraploid pathway and by a simultaneous gain of
chromosomes? To answer all these questions is
truly a Herculean task, and this great challenge
will undoubtedly occupy scientists from different
elds of study for many years to come.
REFERENCES
Andersson A, Olofsson T, Lindgren D, Nilsson B, Ritz C, Ede n
P, Lassen C, Ra de J, Fontes M, Mo rse H, Heldrup J, Behrendtz
M, Mitelman F, Ho glund M, Johansson B, Fioretos T. 2005.
Molecular signatures in childhood acute leukemia and their cor-
relations to expression patterns in normal hematopoietic subpo-
pulations. Proc Natl Acad Sci USA 102:1906919074.
Armstrong SA, Mabon ME, Silverman LB, Li A, Gribben JG, Fox
EA, Sallan SE, Korsmeyer SJ. 2004. FLT3 mutations in child-
hood acute lymphoblastic leukemia. Blood 103:35443546.
Attarbaschi A, Mann G, Ko nig M, Dworzak MN, Trebo MM,
Mu hlegger N, Gadner H, Haas OA. 2004. Incidence and rele-
vance of secondary chromosome abnormalities in childhood
TEL/AML1 acute lymphoblastic leukemia: An interphase
FISH analysis. Leukemia 18:16111616.
Attarbaschi A, Mann G, Ko nig M, Steiner M, Dworzak MN, Gad-
ner H, Haas OA. 2006. Near-tetraploidy in childhood B-cell pre-
cursor acute lymphoblastic leukemia is a highly specic feature
of ETV6/RUNX1-positive leukemic cases. Genes Chromosomes
Cancer 45:608611.
Barber KE, Harrison CJ, Broadeld ZJ, Stewart ARM, Wright SL,
Martineau M, Strefford JC, Moorman AM. 2007. Molecular
cytogenetic characterization of TCF3 (E2A)/19p13.3 rearrange-
ments in B-cell precursor acute lymphoblastic leukemia. Genes
Chromosomes Cancer 46:478486.
Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swan-
ton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N, Scott MA,
Erber WN, Green AR. 2005. Acquired mutation of the tyrosine
kinase JAK2 in human myeloproliferative disorders. Lancet
365:10541061.
Beghini A, Ripamonti CB, Cairoli R, Cazzaniga G, Colapietro P,
Elice F, Nadali G, Grillo G, Haas OA, Biondi A, Morra E, Lar-
izza L. 2004. KIT activating mutations: Incidence in adult and
pediatric acute myeloid leukemia, and identication of an inter-
nal tandem duplication. Haematologica 89:920925.
Behm FG, Raimondi SC, Schell MJ, Look AT, Rivera GK, Pui
C-H. 1992. Lack of CD45 antigen on blast cells in childhood
acute lymphoblastic leukemia is associated with chromosomal
hyperdiploidy and other favorable prognostic features. Blood
79:10111016.
Belkov VM, Krynetski EY, Schuetz JD, Yanishevski Y, Masson E,
Mathew S, Raimondi S, Pui C-H, Relling MV, Evans WE.
1999. Reduced folate carrier expression in acute lymphoblastic
leukemia: A mechanism for ploidy but not lineage differences
in methotrexate accumulation. Blood 93:16431650.
Berger A, Strehl S, Hekele A, Ambros PF, Haas OA, Gadner H.
1994. Interphase cytogenetic study of childhood acute lympho-
blastic leukemia. Med Pediatr Oncol 23:413421.
Betts DR, Riesch M, Grotzer MA, Niggli FK. 2001. The investi-
gation of karyotypic instability in the high-hyperdiploidy sub-
group of acute lymphoblastic leukemia. Leuk Lymphoma
42:187193.
Bhatia S, Sather HN, Heerema NA, Trigg ME, Gaynon PS,
Robison LL. 2002. Racial and ethnic differences in survival of
children with acute lymphoblastic leukemia. Blood 100:1957
1964.
Bianchi AB, Aldaz CM, Conti CJ. 1990. Nonrandom duplication
of the chromosome bearing a mutated Ha-ras-1 allele in mouse
skin tumors. Proc Natl Acad Sci USA 87:69026906.
Blandin AT, Mu hlematter D, Bougeon S, Gogniat C, Porter S,
Beyer V, Parlier V, Beckmann JS, van Melle G, Jotterand M.
2008. Automated four-color interphase uorescence in situ
hybridization approach for the simultaneous detection of spe-
cic aneuploidies of diagnostic and prognostic signicance in
high hyperdiploid acute lymphoblastic leukemia. Cancer Genet
Cytogenet 186:6977.
Bloomeld CD, Secker-Walker LM, Goldman AI, Van Den
Berghe H, de la Chapelle A, Ruutu T, Alimena G, Garson OM,
Golomb HM, Rowley JD, Kaneko Y, Whang-Peng J, Prigogina
E, Philip P, Sandberg AA, Lawler SD, Mitelman F. 1989. Six-
year follow-up of the clinical signicance of karyotype in acute
lymphoblastic leukemia. Cancer Genet Cytogenet 40:171185.
Borkhardt A, Cazzaniga G, Viehmann S, Valsecchi MG, Ludwig
WD, Burci L, Mangioni S, Schrappe M, Riehm H, Lampert F,
Basso G, Masera G, Harbott J, Biondi A. 1997. Incidence and
clinical relevance of TEL/AML1 fusion genes in children with
acute lymphoblastic leukemia enrolled in the German and Ital-
ian multicenter therapy trials. Blood 90:571577.
Borowitz MJ, Shuster JJ, Civin CI, Carroll AJ, Look AT, Behm
FG, Land VJ, Pullen DJ, Crist WM. 1990. Prognostic signi-
cance of CD34 expression in childhood B-precursor acute lym-
phocytic leukemia: A Pediatric Oncology group study. J Clin
Oncol 8:13891398.
Borowitz MJ, Rubnitz J, Nash M, Pullen DJ, Camitta B. 1998.
Surface antigen phenotype can predict TEL-AML1 rearrange-
ment in childhood B-precursor ALL: A Pediatric Oncology
group study. Leukemia 12:17641770.
Brodeur GM, Williams DL, Look AT, Bowman WP, Kalwinsky
DK. 1981. Near-haploid acute lymphoblastic leukemia: A
unique subgroup with a poor prognosis? Blood 58:1419.
HIGH HYPERDIPLOID CHILDHOOD ALL 655
Genes, Chromosomes & Cancer DOI 10.1002/gcc
Brown P, Levis M, Shurtleff S, Campana D, Downing J, Small D.
2005. FLT3 inhibition selectively kills childhood acute lympho-
blastic leukemia cells with high levels of FLT3 expression.
Blood 105:812820.
Bruce WR, Van Der Gaag H. 1963. A quantitative assay for the
number of murine lymphoma cells capable of proliferation
in vivo. Nature 199:7980.
Castor A, Nilsson L, A

strand-Grundstro m I, Buitenhuis M, Ram-


irez C, Anderson K, Stro mbeck B, Garwicz S, Be ka ssy AN,
Schmiegelow K, Lausen B, Hokland P, Lehmann S, Juliusson
G, Johansson B, Jacobsen SEW. 2005. Distinct patterns of
hematopoietic stem cell involvement in acute lymphoblastic
leukemia. Nat Med 11:630637.
Chen C-L, Liu Q, Pui C-H, Rivera GK, Sandlund JT, Ribeiro R,
Evans WE, Relling MV. 1997. Higher frequency of glutathione
S-transferase deletions in black children with acute lymphoblas-
tic leukemia. Blood 89:17011707.
Cheng Q, Yang W, Raimondi SC, Pui C-H, Relling MV, Evans
WE. 2005. Karyotypic abnormalities create discordance of germ-
line genotype and cancer cell phenotypes. Nat Genet 37:878
882.
Chessells JM, Swansbury GJ, Reeves B, Bailey CC, Richards SM.
1997. Cytogenetics and prognosis in childhood lymphoblastic
leukaemia: Results of MRC UKALL X. Br J Haematol 99:93
100.
Clarkson BD, Boyse EA. 1971. Possible explanation of the high
concordance for acute leukaemia in monozygotic twins. Lancet
1:699701.
Csinady E, van der Velden VHJ, Joas R, Fischer S, de Vries JF,
Beverloo HB, Ko nig M, Po tschger U, van Dongen JJM, Mann
G, Haas OA, Panzer-Gru mayer ER. Chromosome 14 copy num-
ber-dependent IGH gene rearrangement patterns in high hyper-
diploid childhood B-cell precursor ALL: Implications for
leukemia biology and minimal residual disease analysis. Leuke-
mia (in press).
Dastugue N, Robert A, Payen C, Cle ment D, Kessous A, Demur
C, Rubie H, Plaisancie H, Bourrouillou G, Colombies P. 1992.
Prognostic signicance of karyotype in a twelve-year follow-up
in childhood acute lymphoblastic leukemia. Cancer Genet Cyto-
genet 64:4955.
Davidsson J, Andersson A, Paulsson K, Heidenblad M, Isaksson
M, Borg A

, Heldrup J, Behrendtz M, Panagopoulos I, Fioretos


T, Johansson B. 2007. Tiling resolution array comparative
genomic hybridization, expression and methylation analyses of
dup(1q) in Burkitt lymphomas and pediatric high hyperdiploid
acute lymphoblastic leukemias reveal clustered near-centro-
meric breakpoints and overexpression of genes in 1q2232.3.
Hum Mol Genet 16:22152225.
Dockerty JD, Herbison P, Skegg DCG, Elwood M. 2007. Vitamin
and mineral supplements in pregnancy and the risk of child-
hood acute lymphoblastic leukaemia: A case-control study.
BMC Public Health 7:136.
Elghezal H, Le Guyader G, Radford-Weiss I, Perot C, Van Den
Akker J, Eydoux P, Vekemans M, Romana SP. 2001. Reassess-
ment of childhood B-lineage lymphoblastic leukemia karyotypes
using spectral analysis. Genes Chromosomes Cancer 30:383
392.
Fenaux P, Lai JL, Morel P, Nelken B, Taboureau O, Deminatti
M, Bauters F. 1989. Cytogenetics and their prognostic value in
childhood and adult acute lymphoblastic leukemia (ALL)
excluding L3. Hematol Oncol 7:307317.
Fitzgibbon J, Smith L-L, Raghavan M, Smith ML, Debernardi S,
Skoulakis S, Lillington D, Lister TA, Young BD. 2005. Associa-
tion between acquired uniparental disomy and homozygous gene
mutation in acute myeloid leukemia. Cancer Res 65:91529154.
Fletcher JA, Kimball VM, Lynch E, Donnelly M, Pavelka K,
Gelber RD, Tantravahi R, Sallan SE. 1989. Prognostic implica-
tions of cytogenetic studies in an intensively treated group of
children with acute lymphoblastic leukemia. Blood 74:2130
2135.
Flotho C, Coustan-Smith E, Pei D, Iwamoto S, Song G, Cheng C,
Pui C-H, Downing JR, Campana D. 2006. Genes contributing
to minimal residual disease in childhood acute lymphoblastic
leukemia: Prognostic signicance of CASP8AP2. Blood
108:10501057.
Forestier E, Schmiegelow K. 2006. The incidence peaks of the
childhood acute leukemias reect specic cytogenetic aberra-
tions. J Pediatr Hematol Oncol 28:486495.
Forestier E, Johansson B, Borgstro m G, Kerndrup G, Johannsson
J, Heim S. 2000a. Cytogenetic ndings in a population-based
series of 787 childhood acute lymphoblastic leukemias from the
Nordic countries. Eur J Haematol 64:194200.
Forestier E, Johansson B, Gustafsson G, Borgstro m G, Kerndrup
G, Johannsson J, Heim S. 2000b. Prognostic impact of karyo-
typic ndings in childhood acute lymphoblastic leukaemia: A
Nordic series comparing two treatment periods. Br J Haematol
110:147153.
Forestier E, Andersen MK, Autio K, Blennow E, Borgstro m G,
Golovleva I, Heim S, Heinonen K, Hovland R, Johannsson JH,
Kerndrup G, Nordgren A, Rosenquist R, Swolin B, Johansson
B. 2007. Cytogenetic patterns in ETV6/RUNX1-positive pediat-
ric B-cell precursor acute lymphoblastic leukemia: A Nordic
series of 245 cases and review of the literature. Genes Chromo-
somes Cancer 46:440450.
Forestier E, Heyman M, Andersen MK, Autio K, Blennow E,
Borgstro m G, Golovleva I, Heim S, Heinonen K, Hovland R,
Johannsson JH, Kerndrup G, Nordgren A, Rosenquist R, Swolin
B, Johansson B. 2008a. Outcome of ETV6/RUNX1-positive
childhood acute lymphoblastic leukaemia in the NOPHO-ALL-
1992 protocol: Frequent late relapses but good overall survival.
Br J Haematol 140:665672.
Forestier E, Izraeli S, Beverloo B, Haas O, Pession A, Michalova
K, Stark B, Harrison CJ, Teigler-Schlegel A, Johansson B.
2008b. Cytogenetic features of acute lymphoblastic and myeloid
leukemias in pediatric patients with Down syndrome: An
iBFM-SG study. Blood 111:15751583.
Gast A, Bermejo JL, Flohr T, Stanulla M, Burwinkel B,
Schrappe M, Bartram CR, Hemminki K, Kumar R. 2007.
Folate metabolic gene polymorphisms and childhood acute
lymphoblastic leukemia: A case-control study. Leukemia 21:
320325.
Gaynon PS, Crotty ML, Sather HN, Bostrom BC, Nachman JB,
Steinherz PG, Heerema NA, Sarquis M, Tuel-Ahlgren L,
Uckun FM. 1997. Expression of BCR-ABL, E2A-PBX1, and
MLL-AF4 fusion transcripts in newly diagnosed children with
acute lymphoblastic leukemia: A Childrens Cancer group initia-
tive. Leuk Lymphoma 26:5765.
Gibbons B, MacCallum P, Watts E, Rohatiner AZS, Webb D,
Katz FE, Secker-Walker LM, Temperley IJ, Harrison CJ,
Campbell RHA, Nash R, Broadbent V, Chessells JM. 1991.
Near-haploid acute lymphoblastic leukemia: Seven new cases
and a review of the literature. Leukemia 5:738743.
Greaves MF. 1988. Speculations on the cause of childhood acute
lymphoblastic leukemia. Leukemia 2:120125.
Greaves MF. 1997. Aetiology of acute leukaemia. Lancet
349:344349.
Greaves M. 2005. In utero origins of childhood leukaemia. Early
Hum Dev 81:123129.
Greaves M. 2006. Infection, immune responses and the aetiology
of childhood leukaemia. Nat Rev Cancer 6:193203.
Groupe Francais de Cytoge ne tique He matologique. 1993. Collab-
orative study of karyotypes in childhood acute lymphoblastic
leukemias. Leukemia 7:1019.
Gruhn B, Taub JW, Ge Y, Beck JF, Zell R, Ha fer R, Hermann
FH, Debatin K-M, Steinbach D. 2008. Prenatal origin of child-
hood acute lymphoblastic leukemia, association with birth
weight and hyperdiploidy. Leukemia 22:16921697.
Gruszka-Westwood AM, Horsley SW, Martinez-Ramirez A, Harri-
son CJ, Kempski H, Moorman AV, Ross FM, Grifths M,
Greaves MF, Kearney L. 2004. Comparative expressed sequence
hybridization studies of high-hyperdiploid childhood acute lym-
phoblastic leukemia. Genes Chromosomes Cancer 41:191202.
Haas OA. 1996. Is genomic imprinting involved in the pathogene-
sis of hyperdiploid and haploid acute lymphoblastic leukemia of
childhood? Acta Genet Med Gemellol 45:239242.
Haas OA, Henn T, Romanakis K, du Manoir S, Lengauer C.
1998. Comparative genomic hybridization as part of a new diag-
nostic strategy in childhood hyperdiploid acute lymphoblastic
leukemia. Leukemia 12:474481.
Haas OA, Zeitlhofer P, Strehl S, Pfeilstoecker M, Koenig M,
Weinhaeusel A. 2004. A novel DNA/RNA FISH X inactivation
assay reveals a nonrandom, ploidy-dependent acquisition of the
active and inactive X chromosomes in childhood hyperdiploid
acute lymphoblastic leukemia (ALL) and non-Hodgkin lym-
phoma (NHL). ASH Annu Meet Abstr 104:1080.
Harbott J, Budde M, Creutzig U, Engel R, Fengler R, Rudolph
B, Lampert F. 1987. Prognostic meaning of chromosome aberra-
tions in acute lymphocytic leukemia and acute nonlymphocytic
leukemia patients of the BFM Study group. Haematol Blood
Transfus 30:497503.
656 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
Harris MB, Shuster JJ, Carroll A, Look AT, Borowitz MJ, Crist
WM, Nitschke R, Pullen J, Steuber CP, Land VJ. 1992. Tris-
omy of leukemic cell chromosomes 4 and 10 identies children
with B-progenitor cell acute lymphoblastic leukemia with a very
low risk of treatment failure: A Pediatric Oncology group study.
Blood 79:33163324.
Harrison CJ, Johansson B. 2009. Acute lymphoblastic leukemia.
In: Heim S, Mitelman F, editors. Cancer Cytogenetics, 3rd ed.
New York: Wiley-Blackwell (in press).
Harrison CJ, Moorman AV, Broadeld ZJ, Cheung KL, Harris RL,
Reza Jalali G, Robinson HM, Barber KE, Richards SM, Mitch-
ell CD, Eden TOB, Hann IM, Hill FGH, Kinsey SE, Gibson
BES, Lilleyman J, Vora A, Goldstone AH, Franklin IM, Durrant
J, Martineau M. 2004. Three distinct subgroups of hypodiploidy
in acute lymphoblastic leukaemia. Br J Haematol 125:552559.
Harrison CJ, Moorman AV, Barber KE, Broadeld ZJ, Cheung
KL, Harris RL, Jalali GR, Robinson HM, Strefford JC, Stewart
A, Wright S, Grifths M, Ross FM, Harewood H, Martineau M.
2005. Interphase molecular cytogenetic screening for chromo-
somal abnormalities of prognostic signicance in childhood
acute lymphoblastic leukaemia: A UK Cancer Cytogenetics
group study. Br J Haematol 129:520530.
Hasle H. 2001. Pattern of malignant disorders in individuals with
Downs syndrome. Lancet Oncol 2:429436.
Hayashi Y, Hanada R, Yamamoto K. 1991. Chromosome abnor-
malities and prognosis in childhood acute leukemia. Acta Pae-
diatr Jpn 33:497506.
Heerema NA, Palmer CG, Baehner RL. 1985. Karyotypic and
clinical ndings in a consecutive series of children with acute
lymphocytic leukemia. Cancer Genet Cytogenet 17:165179.
Heerema NA, Argyropoulos G, Weetman R, Tricot G, Secker-
Walker LM. 1993. Interphase in situ hybridization reveals mini-
mal residual disease in early remission and return of the diag-
nostic clone in karyotypically normal relapse of acute
lymphoblastic leukemia. Leukemia 7:537543.
Heerema NA, Sather HN, Sensel MG, Zhang T, Hutchinson RJ,
Nachman JB, Lange BJ, Steinherz PG, Bostrom BC, Reaman
GH, Gaynon PS, Uckun FM. 2000. Prognostic impact of triso-
mies of chromosomes 10, 17, and 5 among children with acute
lymphoblastic leukemia and high hyperdiploidy (> 50 chromo-
somes). J Clin Oncol 18:18761887.
Heerema NA, Harbott J, Galimberti S, Camitta BM, Gaynon PS,
Janka-Schaub G, Kamps W, Basso G, Pui C-H, Schrappe M,
Auclerc M-F, Carroll AJ, Conter V, Harrison CJ, Pullen J, Rai-
mondi SC, Richards S, Riehm H, Sather HN, Shuster JJ, Silver-
man LB, Valsecchi MG, Arico` M. 2004. Secondary cytogenetic
aberrations in childhood Philadelphia chromosome positive
acute lymphoblastic leukemia are nonrandom and may be asso-
ciated with outcome. Leukemia 18:693702.
Heerema NA, Raimondi SC, Anderson JR, Biegel J, Camitta BM,
Cooley LD, Gaynon PS, Hirsch B, Magenis RE, McGavran L,
Patil S, Pettenati MJ, Pullen J, Rao K, Roulston D, Schneider
NR, Shuster JJ, Sanger W, Sutcliffe MJ, van Tuinen P, Watson
MS, Carroll AJ. 2007. Specic extra chromosomes occur in a
modal number dependent pattern in pediatric acute lympho-
blastic leukemia. Genes Chromosomes Cancer 46:684693.
Heinonen K, Mahlama ki E. 1996. Detection of numerical chromo-
some abnormalities by FISH in childhood acute lymphoblastic
leukemia. Cancer Genet Cytogenet 87:123126.
Heinonen K, Rautonen J, Siimes MA, Knuutila S. 1988. Cytoge-
netic study of 105 children with acute lymphoblastic leukemia.
Eur J Haematol 41:237242.
Hjalgrim LL, Westergaard T, Rostgaard K, Schmiegelow K, Mel-
bye M, Hjalgrim H, Engels EA. 2003. Birth-weight as a risk fac-
tor for childhood leukemia: A meta-analysis of 18 epidemiologic
studies. Am J Epidemiol 158:724735.
Hjalgrim LL, Rostgaard K, Hjalgrim H, Westergaard T, Thomas-
sen H, Forestier E, Gustafsson G, Kristinsson J, Melbye M,
Schmiegelow K. 2004. Birth weight and risk for childhood
leukemia in Denmark, Sweden, Norway, and Iceland. J Natl
Cancer Inst 96:15491556.
Hrsa k O, Porwit-MacDonald A. 2002. Antigen expression patterns
reecting genotype of acute leukemias. Leukemia 16:1233
1258.
Hrusa k O, Trka J, Zuna J, Houskova J, Bartu n kova J, Stary J.
1998. Aberrant expression of KOR-SA3544 antigen in childhood
acute lymphoblastic leukemia predicts TEL-AML1 negativity.
Leukemia 12:10641070.
Huang SC, Koch CA, Vortmeyer AO, Pack SD, Lichtenauer UD,
Mannan P, Lubensky IA, Chrousos GP, Gagel RF, Pacak K,
Zhuang Z. 2000. Duplication of the mutant RET allele in tris-
omy 10 or loss of the wild-type allele in multiple endocrine
neoplasia type 2-associated pheochromocytomas. Cancer Res
60:62236226.
Inukai T, Hirose K, Inaba T, Kurosawa H, Hama A, Inada H,
Chin M, Nagatoshi Y, Ohtsuka Y, Oda M, Goto H, Endo M,
Morimoto A, Imaizumi M, Kawamura N, Miyajima Y, Ohtake
M, Miyaji R, Saito M, Tawa A, Yanai F, Goi K, Nakazawa S,
Sugita K. 2007. Hypercalcemia in childhood acute lymphoblas-
tic leukemia: Frequent implication of parathyroid hormone-
related peptide and E2A-HLF from translocation 17;19. Leuke-
mia 21:288296.
Irving JAE, Bloodworth L, Bown NP, Case MC, Hogarth LA,
Hall AG. 2005. Loss of heterozygosity in childhood acute lym-
phoblastic leukemia detected by genome-wide microarray single
nucleotide polymorphism analysis. Cancer Res 65:30533058.
ISCN 2005. An international system for human cytogenetic
nomenclature. Shaffer LG, Tommerup N, editors. Basel: S.
Karger.
Ito C, Kumagai M, Manabe A, Coustan-Smith E, Raimondi SC,
Behm FG, Murti KG, Rubnitz JE, Pui C-H, Campana D. 1999.
Hyperdiploid acute lymphoblastic leukemia with 51 to 65 chro-
mosomes: A distinct biological entity with a marked propensity
to undergo apoptosis. Blood 93:315320.
Iwamoto S, Mihara K, Downing JR, Pui C-H, Campana D. 2007.
Mesenchymal cells regulate the response of acute lymphoblastic
leukemia cells to asparaginase. J Clin Invest 117:10491057.
Jackson JF, Boyett J, Pullen J, Brock B, Patterson R, Land V, Bor-
owitz M, Head D, Crist W. 1990. Favorable prognosis associated
with hyperdiploidy in children with acute lymphocytic leukemia
correlates with extra chromosome 6. A Pediatric Oncology
Group study. Cancer 66:11831189.
Kager L, Cheok M, Yang W, Zaza G, Cheng Q, Panetta JC, Pui
C-H, Downing JR, Relling MV, Evans WE. 2005. Folate path-
way gene expression differs in subtypes of acute lymphoblastic
leukemia and inuences methotrexate pharmacodynamics.
J Clin Invest 115:110117.
Kaneko Y, Hayashi Y, Sakurai M. 1981. Chromosomal ndings
and their correlation to prognosis in acute lymphocytic leuke-
mia. Cancer Genet Cytogenet 4:227235.
Kaneko Y, Rowley JD, Variakojis D, Chilcote RR, Check I,
Sakurai M. 1982. Correlation of karyotype with clinical fea-
tures in acute lymphoblastic leukemia. Cancer Res 42:2918
2929.
Kaspers GJL, Smets LA, Pieters R, Van Zantwijk CH, Van Wer-
ing ER, Veerman AJP. 1995. Favorable prognosis of hyperdi-
ploid common acute lymphoblastic leukemia may be explained
by sensitivity to antimetabolites and other drugs: Results of an
in vitro study. Blood 85:751756.
Kasprzyk A, Secker-Walker LM. 1997. Increased sensitivity of
minimal residual disease detection by interphase FISH in acute
lymphoblastic leukemia with hyperdiploidy. Leukemia 11:429
435.
Kasprzyk A, Harrison CJ, Secker-Walker LM. 1999. Investigation
of clonal involvement of myeloid cells in Philadelphia-positive
and high hyperdiploid acute lymphoblastic leukemia. Leukemia
13:20002006.
Kawamata N, Ogawa S, Zimmermann M, Kato M, Sanada M,
Hemminki K, Yamatomo G, Nannya Y, Koehler R, Flohr T,
Miller CW, Harbott J, Ludwig W-D, Stanulla M, Schrappe M,
Bartram CR, Koefer HP. 2008. Molecular allelokaryotyping of
pediatric acute lymphoblastic leukemias by high-resolution sin-
gle nucleotide polymorphism oligonucleotide genomic microar-
ray. Blood 111:776784.
Kersey JH. 1997. Fifty years of studies of the biology and therapy
of childhood leukemia. Blood 90:42434251.
Kessous A, Corberand J, Grozdea J, Colombies P. 1975. Clone cel-
lulaire a 27 chromosomes dans une leuce mie aigue humaine.
Nouv Rev Fr Hematol 15:7382.
Kinlen L. 1988. Evidence for an infective cause of childhood leu-
kaemia: Comparison of a Scottish new town with nuclear
reprocessing sites in Britain. Lancet 2:13231327.
Knulst AC, Adriaansen HJ, Ha hlen K, Stigter JC, van den Beemd
MW, Hagemeijer A, van Dongen JJ, Hooijkaas H. 1993. Early
diagnosis of smoldering acute lymphoblastic leukemia using
immunological marker analysis. Leukemia 7:532536.
Kobayashi H, Maseki N, Homma C, Sakurai M, Kaneko Y.
1994. Clinical signicance of chromosome abnormalities in
childhood acute lymphoblastic leukemia in Japan. Leukemia
8:19441950.
HIGH HYPERDIPLOID CHILDHOOD ALL 657
Genes, Chromosomes & Cancer DOI 10.1002/gcc
Kowalczyk JR, Grossi M, Sandberg AA. 1985. Cytogenetic nd-
ings in childhood acute lymphoblastic leukemia. Cancer Genet
Cytogenet 15:4764.
Krajinovic M, Lamothe S, Labuda D, Lemieux-Blanchard E

,
The ore t Y, Moghrabi A, Sinnett D. 2004. Role of MTHFR
genetic polymorphisms in the susceptibility to childhood acute
lymphoblastic leukemia. Blood 103:252257.
Krivit W, Good RA. 1957. Simultaneous occurrence of mongolism
and leukemia. Report of a nationwide survey. AMA J Dis Child
94:289293.
Kuiper RP, Schoenmakers EFPM, van Reijmersdal SV, Hehir-
Kwa JY, Guerts van Kessel A, van Leeuwen FN, Hoogerbrugge
PM. 2007. High-resolution genomic proling of childhood ALL
reveals novel recurrent genetic lesions affecting pathways
involved in lymphocyte differentiation and cell cycle progres-
sion. Leukemia 21:12581266.
Kumagai M, Manabe A, Pui C-H, Behm FG, Raimondi SC, Han-
cock ML, Mahmoud H, Crist WM, Campana D. 1996. Stroma-
supported culture of childhood B-lineage acute lymphoblastic
leukemia cells predicts treatment outcome. J Clin Invest
97:755760.
Lampert F. 1967. Cellula rer DNS-Gehalt und Chromosomenzahl
bei der akuten Leuka mie im Kindesalter und ihre Bedeutung
fu r Chemotherapie und Prognose. Klin Wschr 45:763768.
Lampert F, Harbott J, Ritterbach J. 1991. Chromosomenaberratio-
nen bei akuten Leuka mien im Kindesalter: Analyse von 1009
Patienten. Klin Pa diatr 203:311318.
Larramendy M, El-Rifai W, Saarinen U, Alitalo R, Luomahaara S,
Knuutila S. 1995. Myeloid lineage involvement in acute lym-
phoblastic leukemia: A morphology antibody chromosomes
(MAC) study. Exp Hematol 23:15631567.
Lavabre-Bertrand T, Janossy G, Ivory K, Peters R, Secker-Walker
L, Porwit-MacDonald A. 1994. Leukemia-associated changes
identied by quantitative ow cytometry. I. CD10 expression.
Cytometry 18:209217.
Look AT, Melvin SL, Williams DL, Brodeur GM, Dahl GV, Kal-
winsky DK, Murphy SB, Mauer AM. 1982. Aneuploidy and per-
centage of S-phase cells determined by ow cytometry correlate
with cell phenotype in childhood acute leukemia. Blood
60:959967.
Ma SK, Chan GCF, Wan TSK, Lam CK, Ha SY, Lau YL, Chan LC.
1998. Near-haploid common acute lymphoblastic leukaemia of
childhood with a second hyperdiploid line: A DNA ploidy and u-
orescence in-situ hybridization study. Br J Haematol 103:750755.
MacMahon B, Levy MA. 1964. Prenatal origin of childhood leuke-
mia. Evidence from twins. N Engl J Med 270:10821085.
Maia AT, van der Velden VHJ, Harrison CJ, Szczepanski T, Wil-
liams MD, Grifths MJ, van Dongen JJM, Greaves MF. 2003.
Prenatal origin of hyperdiploid acute lymphoblastic leukemia in
identical twins. Leukemia 17:22022206.
Maia AT, Tussiwand R, Cazzaniga G, Rebulla P, Colman S,
Biondi A, Greaves M. 2004. Identication of preleukemic pre-
cursors of hyperdiploid acute lymphoblastic leukemia in cord
blood. Genes Chromosomes Cancer 40:3843.
Martin PL, Look AT, Schnell S, Harris MB, Pullen J, Shuster JJ,
Carroll AJ, Pettenati MJ, Rao PN. 1996. Comparison of uores-
cence in situ hybridization, cytogenetic analysis, and DNA
index analysis to detect chromosomes 4 and 10 aneuploidy in
pediatric acute lymphoblastic leukemia: A Pediatric Oncology
group study. J Pediatr Hematol Oncol 18:113121.
Martineau M, Clark R, Farrell DM, Hawkins JM, Moorman AV,
Secker-Walker LM. 1996. Isochromosomes in acute lymphoblas-
tic leukaemia: i(21q) is a signicant nding. Genes Chromo-
somes Cancer 17:2130.
Mart nez-Ram rez A, Urioste M, Contra T, Cantalejo A, Tavares
A, Portero JA, Lo pez-Ibor B, Bernacer M, Soto C, Cigudosa JC,
Ben tez J. 2001. Fluorescence in situ hybridization study of
TEL/AML1 fusion and other abnormalities involving TEL and
AML1 genes. Correlation with cytogenetic ndings and prog-
nostic value in children with acute lymphoblastic leukemia.
Haematologica 86:12451253.
Michael PM, Garson OM, Ekert H, Tauro G, Rennie GC,
Pilkington GR. 1988. Prospective study of childhood acute
lymphocytic leukemia: Hematologic, immunologic, and cytoge-
netic correlations. Med Pediatr Oncol 16:153161.
Mitelman F, Johansson B, Mertens F. 2009. Mitelman database of
chromosome aberrations in cancer. Available at: http://cgap.nci.
nih.gov/Chromosomes/Mitelman.
Moghrabi A, Levy DE, Asselin B, Barr R, Clavell L, Hurwitz C,
Samsom Y, Schorin M, Dalton VK, Lipshultz SE, Neuberg DS,
Gelber RD, Cohen HJ, Sallan SE, Silverman LB. 2007. Results
of the Dana-Farber Cancer Institute ALL Consortium Protocol
9501 for children with acute lymphoblastic leukemia. Blood
109:896904.
Moorman AV, Clark R, Farrell DM, Hawkins JM, Martineau M,
Secker-Walker LM. 1996. Probes for hidden hyperdiploidy in
acute lymphoblastic leukaemia. Genes Chromosomes Cancer
16:4045.
Moorman AV, Richards SM, Martineau M, Cheung KL, Robinson
HM, Jalali GR, Broadeld ZJ, Harris RL, Taylor KE, Gibson
BES, Hann IM, Hill FGH, Kinsey SE, Eden TOB, Mitchell
CD, Harrison CJ. 2003. Outcome heterogeneity in childhood
high-hyperdiploid acute lymphoblastic leukemia. Blood 102:
27562762.
Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith E,
Dalton JD, Girtman K, Mathew S, Ma J, Pounds SB, Su X, Pui
C-H, Relling MV, Evans WE, Shurtleff SA, Downing JR. 2007.
Genome-wide analysis of genetic alterations in acute lympho-
blastic leukaemia. Nature 446:758764.
Nordgren A, Farnebo F, Johansson B, Holmgren G, Forestier E,
Larsson C, So derha ll S, Nordenskjo ld M, Blennow E. 2001.
Identication of numerical and structural chromosome aberra-
tions in 15 high hyperdiploid childhood acute lymphoblastic
leukemias using spectral karyotyping. Eur J Haematol 66:297
304.
Nordgren A, Heyman M, Sahle n S, Schoumans J, So derha ll S,
Nordenskjo ld M, Blennow E. 2002. Spectral karyotyping and
interphase FISH reveal abnormalities not detected by conven-
tional G-banding. Implications for treatment stratication of
childhood acute lymphoblastic leukaemia: Detailed analysis of
70 cases. Eur J Haematol 68:3141.
Nore n-Nystro m U, Roos G, Bergh A, Botling J, Lo nnerholm G,
Porwit A, Heyman M, Forestier E. 2008. Bone marrow brosis
in childhood acute lymphoblastic leukemia correlates to biologi-
cal factors, treatment response and outcome. Leukemia 22:504
510.
Nygaard U, Larsen J, Kristensen TD, Wesenberg F, Jonsson OG,
Carlsen NT, Forestier E, Kirchhoff M, Larsen JK, Schmiegelow
K, Christensen IJ. 2006. Flow cytometric DNA index, G-band
karyotyping, and comparative genomic hybridization in detec-
tion of high hyperdiploidy in childhood acute lymphoblastic
leukemia. J Pediatr Hematol Oncol 28:134140.
Online Mendelian inheritance in man. 2009. McKusick-Nathans
Institute of Genetic Medicine, Johns Hopkins University, Balti-
more, MD and National Center for Biotechnology Information,
National Library of Medicine, Bethesda, MD. Available at:
http://www.ncbi.nlm.nih.gov/omim/.
Onodera N, McCabe NR, Nachman JB, Johnson FL, Le Beau
MM, Rowley JD, Rubin CM. 1992a. Hyperdiploidy arising from
near-haploidy in childhood acute lymphoblastic leukemia.
Genes Chromosomes Cancer 4:331336.
Onodera N, McCabe NR, Rubin CM. 1992b. Formation of a
hyperdiploid karyotype in childhood acute lymphoblastic leuke-
mia. Blood 80:203208.
Oshimura M, Freeman AI, Sandberg AA. 1977. Chromosomes and
causation of human cancer and leukemia. XXIII. Near-haploidy
in acute leukemia. Cancer 40:11431148.
Panzer-Gru mayer ER, Fasching K, Panzer S, Hettinger K, Schmitt
K, Sto ckler-Ipsiroglu S, Haas OA. 2002. Nondisjunction of chro-
mosomes leading to hyperdiploid childhood B-cell precursor
acute lymphoblastic leukemia is an early event during leukemo-
genesis. Blood 100:347349.
Paulsson K, Panagopoulos I, Knuutila S, Jee KJ, Garwicz S, Fiore-
tos T, Mitelman F, Johansson B. 2003. Formation of trisomies
and their parental origin in hyperdiploid childhood acute lym-
phoblastic leukemia. Blood 102:30103015.
Paulsson K, Mo rse H, Fioretos T, Behrendtz M, Stro mbeck B,
Johansson B. 2005. Evidence for a single-step mechanism in the
origin of hyperdiploid childhood acute lymphoblastic leukemia.
Genes Chromosomes Cancer 44:113122.
Paulsson K, Heidenblad M, Mo rse H, Borg A

, Fioretos T, Johans-
son B. 2006. Identication of cryptic aberrations and characteri-
zation of translocation breakpoints using array CGH in high
hyperdiploid childhood acute lymphoblastic leukemia. Leuke-
mia 20:20022007.
Paulsson K, Jonson T, O

ra I, Olofsson T, Panagopoulos I,
Johansson B. 2007. Characterisation of genomic translocation
breakpoints and identication of an alternative TCF3/PBX1
fusion transcript in t(1;19)(q23;p13)-positive acute lymphoblastic
leukaemias. Br J Haematol 138:196201.
658 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc
Paulsson K, Cazier J-B, Macdougall F, Stevens J, Stasevich I,
Vrcelj N, Chaplin T, Lillington DM, Lister TA, Young BD.
2008a. Microdeletions are a general feature of adult and adoles-
cent acute lymphoblastic leukemia: Unexpected similarities
with pediatric disease. Proc Natl Acad Sci USA 105:67086713.
Paulsson K, Horvat A, Stro mbeck B, Nilsson F, Heldrup J, Behrendtz
M, Forestier E, Andersson A, Fioretos T, Johansson B. 2008b. Muta-
tions of FLT3, NRAS, KRAS, and PTPN11 are frequent and possibly
mutually exclusive in high hyperdiploid childhood acute lympho-
blastic leukemia. Genes Chromosomes Cancer 47:2633.
Paulsson K, An Q, Moorman AV, Parker H, Molloy G, Davies T,
Grifths M, Ross FM, Irving J, Harrison CJ, Young BD, Stref-
ford JC. 2009. Methylation of tumour suppressor gene pro-
moters in the presence and absence of transcriptional silencing
in high hyperdiploid acute lymphoblastic leukaemia. Br J Hae-
matol 144:838847.
Pere z-Vera P, Fr as S, Carnevale A, Betancourt M, Mu jica M,
Rivera-Luna R, Ort z R. 2004. A strategy to detect chromosomal
abnormalities in children with acute lymphoblastic leukemia.
J Pediatr Hematol Oncol 26:294300.
Pollock BH, DeBaun MR, Camitta BM, Shuster JJ, Ravindranath
Y, Pullen DJ, Land VJ, Mahoney DH, Lauer SJ Jr, Murphy SB.
2000. Racial differences in the survival of childhood B-precursor
acute lymphoblastic leukemia: A Pediatric Oncology Group
study. J Clin Oncol 18:813823.
Prigogina EL, Puchkova GP, Mayakova SA. 1988. Nonrandom
chromosomal abnormalities in acute lymphoblastic leukemia of
childhood. Cancer Genet Cytogenet 32:183203.
Privitera E, Kamps MP, Hayashi Y, Inaba T, Shapiro LH, Rai-
mondi SC, Behm F, Hendershot L, Carroll AJ, Baltimore D,
Look AT. 1992. Different molecular consequences of the 1;19
chromosomal translocation in childhood B-cell precursor acute
lymphoblastic leukemia. Blood 79:17811788.
Pui C-H, Raimondi SC, Williams DL. 1988a. Isochromosome 17q
in childhood acute lymphoblastic leukemia: An adverse cytoge-
netic feature in association with hyperdiploidy? Leukemia
2:222225.
Pui C-H, Williams DL, Roberson PK, Raimondi SC, Behm FG,
Lewis SH, Rivera GK, Kalwinsky DK, Abromowitch M, Crist
WM, Murphy SB. 1988b. Correlation of karyotype and immuno-
phenotype in childhood acute lymphoblastic leukemia. J Clin
Oncol 6:5661.
Pui C-H, Raimondi SC, Dodge RK, Rivera GK, Fuchs LAH,
Abromowitch M, Look AT, Furman WL, Crist WM, Williams
DL. 1989. Prognostic importance of structural chromosomal
abnormalities in children with hyperdiploid (>50 chromosomes)
acute lymphoblastic leukemia. Blood 73:19631967.
Pui C-H, Carroll AJ, Raimondi SC, Schell MJ, Head DR, Shuster
JJ, Crist WM, Borowitz MJ, Link MP, Behm FG, Steuber CP,
Land VJ. 1992. Isochromosomes in childhood acute lymphoblastic
leukemia: A collaborative study of 83 cases. Blood 79:23842391.
Pui C-H, Sandlund JT, Pei D, Rivera GK, Howard SC, Ribeiro
RC, Rubnitz JE, Razzouk BI, Hudson MM, Cheng C, Rai-
mondi SC, Behm FG, Downing JR, Relling MV, Evans WE.
2003. Results of therapy for acute lymphoblastic leukemia in
black and white children. JAMA 290:20012007.
Qiu J, Gunaratne P, Peterson LE, Khurana D, Walsham N, Loul-
seged H, Karni RJ, Roussel E, Gibbs RA, Margolin JF, Gingras
MC. 2003. Novel potential ALL low-risk markers revealed by
gene expression proling with new high-throughput SSH-CCS-
PCR. Leukemia 17:18911900.
Quijano CA, Moore II D, Arthur D, Feusner J, Winter SS, Pallavi-
cini MG. 1997. Cytogenetically aberrant cells are present in the
CD34

CD33
-
38
-
19
-
marrow compartment in children with acute
lymphoblastic leukemia. Leukemia 11:15081515.
Raghavan M, Lillington DM, Skoulakis S, Debernardi S, Chaplin
T, Foot NJ, Lister TA, Young BD. 2005. Genome-wide single
nucleotide polymorphism analysis reveals frequent partial uni-
parental disomy due to somatic recombination in acute myeloid
leukemias. Cancer Res 65:375378.
Raimondi SC, Pui C-H, Hancock ML, Behm FG, Filatov L,
Rivera GK. 1996. Heterogeneity of hyperdiploid (5167) child-
hood acute lymphoblastic leukemia. Leukemia 10:213224.
Raimondi SC, Zhou Y, Shurtleff SA, Rubnitz JE, Pui C-H, Behm
FG. 2006. Near-triploidy and near-tetraploidy in childhood acute
lymphoblastic leukemia: association with B-lineage blast cells car-
rying the ETV6-RUNX1 fusion, T-lineage immunophenotype,
and favorable outcome. Cancer Genet Cytogenet 169:5057.
Ritterbach J, Hiddemann W, Beck JD, Schrappe M, Janka-Schaub
G, Ludwig W-D, Harbott J, Lampert F. 1998. Detection of
hyperdiploid karyotypes (>50 chromosomes) in childhood acute
lymphoblastic leukemia (ALL) using uorescence in situ hybrid-
ization (FISH). Leukemia 12:427433.
Roman-Gomez J, Castillejo JA, Jimenez A, Gonzalez MG, Moreno
F, Rodriguez MC, Barrios M, Maldonado J, Torres A. 2002. 5
0
CpG island hypermethylation is associated with transcriptional
silencing of the p21
CIP1/WAF1/SDI1
gene and confers poor progno-
sis in acute lymphoblastic leukemia. Blood 99:22912296.
Roman-Gomez J, Jimenez-Velasco A, Agirre X, Castillejo JA, Nav-
arro G, Barrios M, Andreu EJ, Prosper F, Heiniger A, Torres A.
2004. Transcriptional silencing of the Dickkopfs-3 (Dkk-3) gene
by CpG hypermethylation in acute lymphoblastic leukaemia. Br
J Cancer 91:707713.
Roman-Gomez J, Jimenez-Velasco A, Cordeu L, Vilas-Zornoza A,
San Jose-Eneriz E, Garate L, Castillejo JA, Martin V, Prosper
F, Heiniger A, Torres A, Agirre X. 2007. WNT5A, a putative
tumour suppressor of lymphoid malignancies, is inactivated by
aberrant methylation in acute lymphoblastic leukaemia. Eur J
Cancer 43:27362746.
Ross ME, Zhou X, Song G, Shurtleff SA, Girtman K, Williams
WK, Liu H-C, Mahfouz R, Raimondi SC, Lenny N, Patel A,
Downing JR. 2003. Classication of pediatric acute lymphoblas-
tic leukemia by gene expression proling. Blood 102:29512959.
Rubin CM, Le Beau MM, Mick R, Bitter MA, Nachman J,
Rudinsky R, Appel HJ, Morgan E, Suarez CR, Schumacher
HR, Subramanian U, Rowley JD. 1991. Impact of chromosomal
translocations on prognosis in childhood acute lymphoblastic
leukemia. J Clin Oncol 9:21832192.
Rubnitz JE, Behm FG, Pui C-H, Evans WE, Relling MV, Rai-
mondi SC, Harrison PL, Sandlund JT, Ribeiro RC, Grosveld G,
Downing JR. 1997. Genetic studies of childhood acute lympho-
blastic leukemia with emphasis on p16, MLL, and ETV6 gene
abnormalities: Results of St Jude Total Therapy Study XII.
Leukemia 11:12011206.
Schaad K, Stro mbeck B, Mandahl N, Andersen MK, Heim S, Mert-
ens F, Johansson B. 2006. FISH mapping of i(7q) in acute leuke-
mias and myxoid liposarcoma reveals clustered breakpoints in
7p11.2: Implications for formation and pathogenetic outcome of
the idic(7)(p11.2). Cytogenet Genome Res 114:126130.
Schmiegelow K, Hjalgrim H. 2006. Is the risk of acute lympho-
blastic leukemia reduced in siblings to children with the
disease? A novel hypothesis explored by international collabora-
tion. Leukemia 20:12061208.
Schultz KR, Pullen J, Sather HN, Shuster JJ, Devidas M, Borowtiz
MJ, Carroll AJ, Heerema NA, Rubnitz JE, Loh ML, Raetz EA,
Winick NJ, Hunger SP, Carroll WL, Gaynon PS, Camitta BM.
2007. Risk- and response-based classication of childhood B-pre-
cursor acute lymphoblastic leukemia: A combined analysis of
prognostic markers from the Pediatric Oncology Group (POG)
and Childrens Cancer Group (CCG). Blood 109:926935.
Secker-Walker LM, Lawler SD, Hardisty RM. 1978. Prognostic
implications of chromosomal ndings in acute lymphoblastic
leukaemia at diagnosis. Br Med J 2:15291530.
Secker-Walker LM, Swansbury GJ, Lawler SD, Hardisty RM.
1979. Bone marrow chromosomes in acute lymphoblastic leu-
kaemia: A long-term study. Med Pediatr Oncol 7:371385.
Secker-Walker LM, Chessells JM, Stewart EL, Swansbury GJ,
Richards S, Lawler SD. 1989. Chromosomes and other prognos-
tic factors in acute lymphoblastic leukaemia: A long-term
follow-up. Br J Haematol 72:336342.
Sharathkumar A, DeCamillo D, Bhambhani K, Cushing B,
Thomas R, Mohamed AN, Ravindranath Y, Taub JW. 2008.
Children with hyperdiploid but not triple trisomy (4,10,17)
acute lymphoblastic leukemia have an increased incidence of
extramedullary relapse on current therapies: A single institution
experience. Am J Hematol 83:3440.
Shu XO, Perentesis JP, Wen W, Buckley JD, Boyle E, Ross JA,
Robison LL. 2004. Parental exposure to medications and hydro-
carbons and ras mutations in children with acute lymphoblastic
leukemia: A report from the Childrens Oncology Group. Can-
cer Epidemiol Biomark Prev 13:12301235.
Smets LA, Slater RM, Behrendt H, Vant Veer MB, Homan-Blok
J. 1985. Phenotypic and karyotypic properties of hyperdiploid
acute lymphoblastic leukaemia of childhood. Br J Haematol
61:113123.
Smets LA, Homan-Blok J, Hart A, de Vaan G, Behrendt H,
Ha hlen K, de Waal FJ. 1987. Prognostic implication of hyperdi-
ploidy as based on DNA ow cytometric measurement in child-
hood acute lymphocytic leukemiaA multicenter study.
Leukemia 1:163166.
HIGH HYPERDIPLOID CHILDHOOD ALL 659
Genes, Chromosomes & Cancer DOI 10.1002/gcc
Smets LA, Slater R, van Wering ER, van der Does-van den Berg
A, Hart AAM, Veerman AJP, Kamps WA. 1995. DNA index and
%S-phase cells determined in acute lymphoblastic leukemia of
children: A report from studies ALL V, ALL VI, and ALL VII
(19791991) of the Dutch Childhood Leukemia Study Group
and the Netherlands Workgroup on Cancer Genetics and Cyto-
genetics. Med Pediatr Oncol 25:437444.
Stam RW, den Boer ML, Schneider P, Meier M, Beverloo HB,
Pieters R. 2007. D-HPLC analysis of the entire FLT3 gene in
MLL rearranged and hyperdiploid acute lymphoblastic leuke-
mia. Haematologica 92:15651568.
Stark B, Jeison M, Gobuzov R, Krug H, Glaser-Gabay L, Luria D,
El-Hasid R, Harush MB, Avrahami G, Fisher S, Stein J, Zaizov R,
Yaniv I. 2001. Near-haploid childhood acute lymphoblastic leuke-
mia masked by hyperdiploid line: Detection by uorescence in
situ hybridization. Cancer Genet Cytogenet 128:108113.
Strefford JC, Worley H, Barber K, Wright S, Stewart ARM, Rob-
inson HM, Bettney G, van Delft FW, Atherton MG, Davies T,
Grifths M, Hing S, Ross FM, Talley P, Saha V, Moorman AV,
Harrison CJ. 2007. Genome complexity in acute lymphoblastic
leukemia is revealed by array-based comparative genomic
hybridization. Oncogene 26:43064318.
Sulong S, Moorman AV, Irving JAE, Strefford JC, Konn ZJ, Case
MC, Minto L, Barber KE, Parker H, Wright SL, Stewart ARM,
Bailey S, Bown NP, Hall AG, Harrison CJ. 2009. A comprehen-
sive analysis of the CDKN2A gene in childhood acute lympho-
blastic leukemia reveals genomic deletion, copy number neutral
loss of heterozygosity, and association with specic cytogenetic
subgroups. Blood 113:100107.
Sutcliffe MJ, Shuster JJ, Sather HN, Camitta BM, Pullen J,
Schultz KR, Borowitz MJ, Gaynon PS, Carroll AJ, Heerema
NA. 2005. High concordance from independent studies by the
Childrens Cancer Group (CCG) and Pediatric Oncology Group
(POG) associating favorable prognosis with combined trisomies
4, 10, and 17 in children with NCI standard-risk B-precursor
acute lymphoblastic leukemia: a Childrens Oncology Group
(COG) initiative. Leukemia 19:734740.
Synold TW, Relling MV, Boyett JM, Rivera GK, Sandlund JT,
Mahmoud H, Crist WM, Pui C-H, Evans WE. 1994. Blast cell
methotrexate-polyglutamate accumulation in vivo differs by
lineage, ploidy, and methotrexate dose in acute lymphoblastic
leukemia. J Clin Invest 94:19962001.
Taketani T, Taki T, Sugita K, Furuichi Y, Ishii E, Hanada R,
Tsuchida M, Sugita K, Ida K, Hayashi Y. 2004. FLT3 mutations
in the activation loop of tyrosine kinase domain are frequently
found in infant ALL with MLL rearrangements and pediatric
ALL with hyperdiploidy. Blood 103:10851088.
Tartaglia M, Martinelli S, Cazzaniga G, Cordeddu V, Iavarone I,
Spinelli M, Palmi C, Carta C, Pession A, Arico` M, Masera G,
Basso G, Sorcini M, Gelb BD, Biondi A. 2004. Genetic evi-
dence for lineage-related and differentiation stage-related con-
tribution of somatic PTPN11 mutations to leukemogenesis in
childhood acute leukemia. Blood 104:307313.
Taub JW, Konrad MA, Ge Y, Naber JM, Scott JS, Matherly LH,
Ravindranath Y. 2002. High frequency of leukemic clones in
newborn screening blood samples of children with B-precursor
acute lymphoblastic leukemia. Blood 99:29922996.
Taylor GM, Dearden S, Ravetto P, Ayres M, Watson P, Hussain
A, Greaves M, Alexander F, Eden OS. 2002. Genetic suscepti-
bility to childhood common acute lymphoblastic leukaemia is
associated with polymorphic peptide-binding pocket proles in
HLA-DPB1*0201. Hum Mol Genet 11:15851597.
Third International Workshop on chromosomes in leukemia.
1981a. Chromosomal abnormalities in acute lymphoblastic leu-
kemia: Structural and numerical changes in 234 cases. Cancer
Genet Cytogenet 4:101110.
Third International Workshop on chromosomes in leukemia. 1981b.
Clinical signicance of chromosomal abnormalities in acute lym-
phoblastic leukemia. Cancer Genet Cytogenet 4:111137.
Thompson JR, Gerald PF, Willoughby MLN, Armstrong BK.
2001. Maternal folate supplementation in pregnancy and protec-
tion against acute lymphoblastic leukaemia in childhood: A
case-control study. Lancet 358:19351940.
Tomeczkowski J, Yakisan E, Wieland B, Reiter A, Welte K,
Sykora K-W. 1995. Absence of G-CSF receptors and absent
response to G-CSF in childhood Burkitts lymphoma and B-
ALL cells. Br J Haematol 89:771779.
Troussard X, Rimokh R, Valensi F, Leboeuf D, Fenneteau O,
Guitard A-M, Manel A-M, Schillinger F, Leglise C, Brizard A,
Gardais J, Lessard M, Flandrin G, Macintyre E. 1995. Hetero-
geneity of t(1;19)(q23;p13) acute leukaemias. Br J Haematol
89:516526.
Uckun FM, Gajl-Peczalska KJ, Provisor AJ, Heerema NA. 1989.
Immunophenotype-karyotype associations in human acute lym-
phoblastic leukemia. Blood 73:271280.
UK Childhood Cancer Study Investigators. 2000. The United
Kingdom Childhood Cancer Study: Objectives, materials and
methods. Br J Cancer 82:10731102.
van Delft FW, Bellotti T, Luo Z, Jones LK, Patel N, Yiannikouris
O, Hill AS, Hubank M, Kempski H, Fletcher D, Chaplin T,
Foot N, Young BD, Hann IM, Gammerman A, Saha V. 2005.
Prospective gene expression analysis accurately subtypes acute
leukaemia in children and establishes a commonality between
hyperdiploidy and t(12;21) in acute lymphoblastic leukaemia. Br
J Haematol 130:2635.
Wang X, Thomas P, Xue J, Fenech M. 2004. Folate deciency
induces aneuploidy in human lymphocytes in vitroEvidence
using cytokinesis-blocked cells and probes specic for chromo-
somes 17 and 21. Mutat Res 551:167180.
Whitehead VM, Vuchich MJ, Lauer SJ, Mahoney D, Carroll AJ,
Shuster JJ, Esseltine DW, Payment C, Look AT, Akabutu J,
Bowen T, Taylor LD, Camitta B, Pullen DJ. 1992. Accumula-
tion of high levels of methotrexate polyglutamates in lympho-
blasts from children with hyperdiploid (>50 chromosomes)
B-lineage acute lymphoblastic leukemia: A Pediatric Oncology
Group study. Blood 80:13161323.
Whitehead VM, Vuchich MJ, Cooley LD, Lauer SJ, Mahoney DH,
Shuster JJ, Payment C, Koch PA, Akabutu JJ, Bowen T, Kamen BA,
Ravindranath Y, Emami A, Look AT, Beardsley GP, Pullen DJ,
Camitta B. 1998. Accumulation of methotrexate polyglutamates,
ploidy and trisomies of both chromosomes 4 and 10 in lymphoblasts
from children with B-progenitor cell acute lymphoblastic leukemia:
A Pediatric Oncology Group study. Leuk Lymphoma 31:507519.
Wiemels JL, Smith RN, Taylor GM, Eden OB, Alexander FE,
Greaves MF. 2001. Methylenetetrahydrofolate reductase
(MTHFR) polymorphisms and risk of molecularly dened sub-
types of childhood acute leukemia. Proc Natl Acad Sci USA
98:40044009.
Wiemels JL, Zhang Y, Chang J, Zheng S, Metayer C, Zhang L,
Smith MT, Ma X, Selvin S, Bufer PA, Wiencke JK. 2005. RAS
mutation is associated with hyperdiploidy and parental charac-
teristics in pediatric acute lymphoblastic leukemia. Leukemia
19:415419.
Williams DL, Tsiatis A, Brodeur GM, Look AT, Melvin SL, Bow-
man WP, Kalwinsky DK, Rivera G, Dahl GV. 1982. Prognostic
importance of chromosome number in 136 untreated children
with acute lymphoblastic leukemia. Blood 60:864871.
Wilson JL, Morison IM. 2005. No evidence for preferential mater-
nal origin of duplicated chromosome 14 in hyperdiploid ALL.
Blood 105:1837.
Yagi T, Hibi S, Tabata Y, Kuriyama K, Teramura T, Hashida T,
Shimizu Y, Takimoto T, Todo S, Sawada T, Imashuku S. 2000.
Detection of clonotypic IGH and TCR rearrangements in the
neonatal blood spots of infants and children with B-cell precur-
sor acute lymphoblastic leukemia. Blood 96:264268.
Yeoh E-J, Ross ME, Shurtleff SA, Williams WK, Patel D, Mah-
fouz R, Behm FG, Raimondi SC, Relling MV, Patel A, Cheng
C, Campana D, Wilkins D, Zhou X, Li J, Liu H, Pui C-H,
Evans WE, Naeve C, Wong L, Downing JR. 2002. Classica-
tion, subtype discovery, and prediction of outcome in pediatric
acute lymphoblastic leukemia by gene expression proling.
Cancer Cell 1:133143.
Zhang L, Taub JW, Williamson M, Wong SC, Hukku B, Pullen J,
Ravindranath Y, Matherly LH. 1998. Reduced folate carrier gene
expression in childhood acute lymphoblastic leukemia: Relationship
to immunophenotype and ploidy. Clin Cancer Res 4:21692177.
Zhang Y, Lu J, van den Berghe J, Lee S-H. 2002. Increased inci-
dence of spontaneous apoptosis in the bone marrow of hyperdi-
ploid childhood acute lymphoblastic leukemia. Exp Hematol
30:333339.
Zheng S, Ma X, Zhang L, Gunn L, Smith MT, Wiemels JL,
Leung K, Bufer PA, Wiencke JK. 2004. Hypermethylation of
the 5
0
CpG island of the FHIT gene is associated with hyperdi-
ploid and translocation-negative subtypes of pediatric leukemia.
Cancer Res 64:20002006.
Zhuang Z, Park W-S, Pack S, Schmidt L, Vortmeyer AO, Pak E,
Pham T, Weil RJ, Candidus S, Lubensky IA, Linehan WM,
Zbar B, Weirich G. 1998. Trisomy 7-harbouring non-random
duplication of the mutant MET allele in hereditary papillary
renal carcinomas. Nat Genet 20:6669.
660 PAULSSON AND JOHANSSON
Genes, Chromosomes & Cancer DOI 10.1002/gcc

Das könnte Ihnen auch gefallen