You are on page 1of 16

PEDIATRIC ONCOLOGY 0031-3955/97 $0.00 + .

20

ACUTE LYMPHOBLASTIC
LEUKEMIA
Ching-Hon h i , MD

Childhood acute lymphoblastic leukemia (ALL), one of the first malignant


diseases for which large-scale therapeutic trials were conducted, has served as
a paradigm for cancer research for over four decades. At present, two thirds or
more of children with ALL can be cured with contemporary treatment plans
(Table 1).6*This remarkable success began with the identification of effective
antineoplastic drugs in the late 1940s and 1950s, followed by the development
of combination chemotherapy and subclinical central nervous system (CNS)
leukemia therapy in the 1960s. Refinements in the design and analysis of clinical
trials during the 1970s led to the recognition of clinical and laboratory features
that connote a poor prognosis, setting the stage for risk-directed therapy. In the
1980s, the following became clear: certain antileukemic agents, as well as cranial
irradiation, can produce serious late sequelae; many leukemias arise from non-
random cytogenetic abnormalities with therapeutic implications; intensive che-
motherapy can abolish the prognostic significance of certain adverse features;
and some cases of refractwy leukemia can be cured with allogeneic hematopoi-
etic stem cell transplantation. Together, these discoveries resulted in greater
refinement of risk-oriented therapy that, in time, advanced cure rates to their
present high levels. We recently entered the era of molecular medicine, in which
genetic probes are being used to recognize new disease categories and improve
treatment selection. Ultimately, dissection of ALL at the molecular level should
pinpoint sensitive and highly specific targets for therapeutic intervention, lead-
ing to treatments that are both safe and uniformly effective. This article empha-
sizes only recent advances in the management of childhood ALL.

This work was partially supported by grants PO1 CA 20180 and P30 CA 21765 from
the National Cancer Institute and by the American Lebanese Syrian Associated Charities.

From the Department of Hematology/Oncology, St. Jude Children’s Research Hospital;


and the Department of Pediatrics, University of Tennessee, Memphis, College of
Medicine, Memphis, Tennessee

~ ~ ~~~~~~~

PEDIATRIC CLINICS OF NORTH AMERICA

VOLUME 44 NUMBER 4 * AUGUST 1997 831


832 PUI

EPIDEMIOLOGY

Acute lymphoblastic leukemia is the most common pediatric malignancy,


accounting for three fourths of all newly diagnosed leukemias and one fourth
of all cancers in childhood.33The incidence of childhood ALL in the United
States is approximately 3.4 cases per 100,000 children younger than 15 years of
age? with the peak incidence occurring between the ages of 3 and 4 years.i3 A
study by the Surveillance, Epidemiology, and End Results (SEER) Program,
which surveys cancer registries accounting for about 10% of the US population,
indicated a 20% cumulative increase in the incidence of childhood ALL from
1973 to 1991.Rh This finding was attributed to increased childhood or parental
exposure to environmental or occupational hazards, such as electromagnetic
fields, dietary nitrites, pesticides, and ionizing radiation, although none of these
factors have been conclusively linked to the development of ALL, to date.", 92
Part of the explanation may lie in changes in diagnostic specificity during
that survey period, resulting in recognition of not-otherwise-specified forms of
lymphoid leukemias as ALL.62 In the United States, the incidence of ALL is
higher in whites than in blacks (1.8 to 1) and in boys than in girls (1.2 to 1).
Worldwide, the frequency, age distribution, and subtypes of ALL show striking
geographic differences that could reflect variability in genetic susceptibility,
environmental factors, or 57 One study suggested an increased incidence

of infant leukemia in Greece owing to low-level intrauterine exposure to ionizing


radiation from Chern~byl?~ whereas larger investigations failed to show an
increased risk of childhood acute leukemia in regions affected by the nuclear
reactor
Numerous constitutional genetic defects (e.g., trisomy 21, congenital immu-
nodeficiency, and ataxia telangiectasia) have been linked to an increased risk of

Table 1. TREATMENT OUTCOME IN CHILDHOOD ACUTE LYMPHOBLASTIC


LEUKEMIA (ALL): RESULTS OF 11 INTERNATIONAL STUDIES

ChiIdren, Event-free Survival


Study n ( f SE), %
AIEOP ALL88 (1988-1992) 396 66.6 (2.4) at 5 y
BFM (1986-1 990) 998 72 (2) at 6 y
CCG 100 series (1983-1 989) 3465 67 (2) at 5 y
Dana-Farber 85-01 (1985-1987) 220 78 (3) at 7 y
DFW (1986-1 992) 243 66.6 (6.5) at 5 y
Dutch (1984-1 988) 291 72 (3) at 6 y
FRALLE 83 (1983-1 987) 559 57 (2) at 7 y
MRC UKALL X (1985-1990) 1612 62 (2) at 5 y
POG ALinC + T-cell +
infant protocols (1986-1990) 2404 66.4 (2.4) at 4 y
SJCRH XI (1984-1 988) 358 71% (4) at 8 y
TCCSG L84-11 (1984-1 989) 490 63.8% (3.2) at 7y

SE, Standard error; AIEOP. Associazione ltaliana Ematologia Oncologia Pediatrica; BFM, Berlin-
Frankfurt-Munster; CCG, Children's Cancer Group (Arcadia, CA); Dana-Farber, Dana-Farber Cancer
InstitutelChildren's Hospital Acute Lymphoblastic Leukemia Consortium (Boston, MA); DFW, Dallas-Ft.
Worth; Dutch, Dutch Childhood Leukemia Study Group; FRALLE, French Acute Lymphoblastic Leukemia
Cooperative Group; MRC, Medical Research Council (United Kingdom); UKALL X, United Kingdom
Acute Lymphoblastic Leukemia X; POG, Pediatric Oncology Group (Chicago, IL); ALinC, Acute Leuke-
mia in Childhood (protocol); SJCRH, St. Jude Children's Research Hospital (Memphis, TN); TCCSG,
Tokyo Children's Cancer Study Group.
From Pui C-H: Acute leukemia in children. Curr Opinion Hematol 3:249,1996; with permission.
ACUTE LYMPHOBLASTIC LEUKEMIA 833

ALL in children.6’ However, in most instances, cases of ALL lack a plausible


causal explanation. A provocative suggestion is that childhood leukemias are
the consequence of rare, abnormal immunologic response to a common infection,
perhaps preceded by an initiating mutational event in utero.30,47 Finally, siblings
of children with ALL have a two to four fold increased risk of developing ALL
than do children in the general population, and the risk is much higher among
monozygotic twins, especially during the first few years of life. Intrauterine
metastasis through the shared placental circulation has been established as the
mechanism for the latter observation. Interestingly, a pair of identical twins
developed a late-onset concordant T-cell malignancy of monoclonal origin at 9
and 11 years of age, respecti~ely.~~This finding supports our speculation that
many ALLs arise in utero, with the most aggressive forms presenting clinically
in infants and the remainder appearing in later childhood.6L

CELL CLASSIFICATION BASED ON BIOLOGIC FEATURES

lmmunophenotype

Morphologic examination and cytochemical staining of blast cells are usu-


ally sufficient to distinguish ALL from acute myeloid leukemia; however, these
methods do not establish the cell’s immunologic subtype, a necessary consider-
ation in the comprehensive diagnosis. Most leukemic cells share features with
normal hematopoietic progenitors. By recognizing patterns of developmentally
relevant gene expression, one can classify leukemic cells according to their
normal maturation sequences. Because, with a few exceptions, cell-differentia-
tion antigens lack absolute lineage specificity, immunologic classification is usu-
ally based on reactivity with a panel of lineage-associated antibodieP (Table 2).
By this approach, ALL is broadly considered to have two lineages, B and T. B-
lineage cases can be subdivided into four distinct subtypes: early pre-B, pre-B,
transitional pre-B, and mature B ce1L6’Likewise, T-lineage ALL can be subclassi-
fied according to the stage of thymocyte differentiation or the stage of expression
65 For therapeutic purposes, however, one need
of the T-cell receptor pi-~tein.’~,
only distinguish T-cell and mature B-cell cases from those originating in B-cell
precursors.”’ Expression of ”myeloid-associated” antigens on otherwise typical
lymphoblasts has been recognized for over a decade. Although some investiga-

Table 2. IMMUNOLOGIC SUBCLASSES OF ACUTE LYMPHOBLASTIC LEUKEMIA


(ALL)
Frequency
Type Antigen Expression (%.)
Early pre-B cCD22+,CD79a, CD19+,CD22+,clg-(mu),slg- 57
Pre-B clg+(mu) 22
Transitional pre-B clg+(mu),slg’, slg-(kappa),slg-(lambda) 4
B cell slg’, slg+(kappa),or slg+(lambda) 2
T cell cCD3+,CD7+,CD5+,or CD2’ 15

”Based on 200 cases of newly diagnosed ALL studied at St. Jude Children’s Research Hospital
from 1993 to 1996.
cCD22, Cytoplasmic CD22; clg, cytoplasmic immunoglobulin; slg, surface immunoglobulin; mu,
mu heavychain protein; kappa, kappa light-chain protein; lambda, lambda light-chain protein; cCD3,
cytoplasmic CD3.
834 PUI

tors believe that myeloid-antigen positive ALL is associated with a poor


progn0sis,2~,~’~
we and others have found that this type of leukemia responds
well to contemporary risk-directed therapy.” b7, 78,I M

Genetic Features
Because gene alterations are at the base of all leukemia^,'^,^^ a cell classifica-
tion system may eventually become purely genetic. In fact, childhood ALL can
be readily classified according to specific chromosomal changes, now identified
in over 90% of cases.71Many of these abnormalities, especially the reciprocal
translocations, have been well-characterized molecularly and are thought to
have a central role in leukemogenesis. The clear correlation between nonrandom
chromosomal anomalies and blast cell biologic properties and presenting clinical
features (Table 3) attests to their prognostic significance, and hence their ultimate
worth in cell classification. Even so, there are compelling reasons to focus on
the molecular defects in leukemic cells. First, cases with clinically important
genetic rearrangements may be missed by routine karyotyping, even in centers
with a high rate of successful analysis. For example, whereas molecular re-
arrangements of the MLL gene (located at llq23) occur in approximately 80% of
infants with ALL, 60% or fewer have detectable llq23 chromosomal ab-
normalities.66,&s Second, rearrangements affecting the same chromosomal region
may involve different genes and represent clinically and biologically diverse
entities (e.g., gene rearrangements produced by the 1;19 translocation or 11q23
abnormalitie~).~~. 82 Third, molecular analyses can identify several critical submi-
croscopic genetic alterations not visible by karyotyping. In fact, TEL/”LI
fusion (also known as ETV6/CBFA2 fusi0n)9~and rearrangements of the TALI
gene2-the most common abnormality in B-lineage and T-lineage ALL,
respectively-as well as deletions of tumor suppressor genes such as ~ 2 are 6 ~ ~
generally detectable only at the molecular level. By combining cytogenetic and
molecular findings, one can successfully classify 75% or more of newly diag-
nosed ALL cases into prognostic and therapeutically relevant subgroups (Table 3).

MOLECULAR PATHOGENESIS
There are two general mechanisms of leukemia induction. The first involves
the activation of a proto-oncogene or the creation of a fusion gene with onco-
genic properties. In either case, the protein products are often transcription
factors, suggesting that alterations in master regulator proteins that control
hematopoietic growth and differentiation play a major role in leukemic transfor-
mation. Interestingly, various genetic rearrangements may exert their effects on
common signal transduction pathways. The second mechanism involves the loss
or inactivation of genes whose proteins suppress leukemia. Proto-oncogene
activation and loss of tumor suppressor genes either endow the leukemic cell
with a proliferative advantage or prevent its normal differentiation and pro-
grammed cell death. Readers are referred to several excellent reviews for more
91
detailed discussion of ALL molecular pathogene~is.’~,%,

PROGNOSTIC FACTORS AND THERAPEUTICALLY


RELEVANT RISK GROUPS
The cornerstone of the modem approach to ALL therapy has been careful
assessment of the failure risk, so that only high-risk cases are treated aggres-
Table 3. CLINICALLY AND BIOLOGICALLY RELEVANT GENETIC SUBGROUPS OF ACUTE LYMPHOBLASTIC LEUKEMIA (ALL)’
~~ ~~

Estimated 5-y
Frequency Molecular Genetic Event-free
Chromosomal Abnormality (“/4 Alterations Associated Features Survival (%)
Hyperdiploidy (>50chromosomes) 27 Unknown 6-cell precursor phenotype; favorable age 80-90
(between 1 and 10 y); low leukocyte count
t(12;21)(p12-13;q22)t 21-24 . TEUAML7 fusion (also 6-cell precursor phenotype; pseudodiploidy; 85-90
known as E N 6 / favorable age (between 1 and 10 y)
CBFAP fusion)
t(1;19)(q23;p13.3) 5-6 E2MP6Xl fusion Pre-6 phenotype; pseudodiploidy; increased 70-80
leukocyte count
t(4;l l)(q21;q23) and other 1lq23 4-8 MLUAF4 fusion and CD10- 6-cell precursor phenotype; infant 20-30
translocations other MLL age group; hyperleukocytosis
rearrangements
t(9;22)(q34;ql1) 3 4 BCWA6L fusion 6-cell precursor phenotype; older age; 25-35
increased leukocyte count
t(1;14)(p34;qll) and TAL’ deletiont 3 TAL1 (SCL) CD10- T-cell phenotype; male 60-70
rearrangement predominance; hyperleukocytosis
t(8;14)(q24;q32.3), t(2;8)(ql2;q24), or 2 MYC (IgH, Ig[kappa], or B-cell phenotype; L3 morphology; male 70-80
t(8;22)(q24;qll) Ig[lambda]) predominance
t(l1;14)(p13;qll) 1 7 7 G X C R (delta) fusion T-cell phenotype; male predominance; 60
hyperleukocytosis; extramedullary disease
dic(9;12)(pl1-12;?~12) 1 Unknown 6-cell precursor phenotype; male 90
predominance
Hypodiploidy <45 (chromosomes) 1 Unknown None 20-30
and near-haploidy

‘Less common subgroups not listed.


tCryptic rearrangement requiring molecular detection.
$A local DNA deletion requiring molecular detection.
836 PUI

-
.-
a
others
(n=106)
2 40-
2
a 2o
- P<0.0001
I I I I I
0
0 2 4 6 8 10
Years From Diagnosis

Figure 1. Kaplan-Meier analysis of event-free survival according to presenting age and


leukocyte count in childhood B-cell precursor ALL cases treated in Total Therapy Study XI
(1984-1988) at St. Jude Children's Research Hospital.

sively and less toxic therapy, usually antimetabolites, is reserved for lower-risk
cases. Of the many variables that have been linked to ALL prognosis, treatment
has emerged as the most infl~ential.~~ For example, T-cell and B-cell ALL, once
associated with a dire prognosis, now have long-term response rates of 70% or
better with the use of intensive chem~therapy.~~, 83, e4, 93
Age and leukocyte count continue to be used for risk classification in
virtually every clinical trial involving B-cell precursor ALL. In a recent workshop
sponsored by the National Cancer Institute, participants agreed on a presenting
age between 1 and 9 years and a leukocyte count less than 50 X 109/L as the
minimal criteria for lower-risk ALL, with all other cases considered higher-ri~k.~~
By this standard, 60% to 70% of B-cell precursor cases would be classified as
lower-risk, with a long-term event-free survival probability of 70% to 85%; the
remaining 30% to 40% would have higher-risk disease and a survival probability
of 50% to 60% (Fig. 1).
Using genetic features, 'childhood B-cell precursor ALL can be classified
into three distinct subgroups (Fig. 2). Lower-risk ALL cases include patients
with hyperdiploidy (>50 chromo~omes),7~~ Io4 TELIAMLI fusion,51,89, 90 or the
dic(9;12).5All three genetic subgroups respond favorably to antimetabolite-based

3 4ol \
20
, 71 i 12

'I...........36i13
i
...............

''*
EA-PBX7 (n=14)

..BCR-ABL
*. ...............
**
(n=ll)

Reananged M L L (n=9)
I I I I 1
0'
0 2 4 6 8 10 12
Years From Diagnosis

Figure 2. Kaplan-Meier analysis of event-free survival according to selected genetic fea-


tures in childhood B-cell precursor ALL cases treated in Total Therapy Studies XI (1984-
1988) and XI1 (1988-1991) at St. Jude Children's Research Hospital.
ACUTE LYMPHOBLASTIC LEUKEMIA 837

therapy. The biologic basis for the favorable prognosis of hyperdiploidy (>50
chromosomes) may stem from the tendency of hyperdiploid blasts to accumulate
increased amounts of methotrexate and its active polyglutamate metabo-
lites,10z,113
as well as the marked propensity of these cells for programmed cell
death (apoptosis)." The reason for a favorable outcome in cases with TEL/
A M L l fusion or the dic(9;12) is unknown. High-risk ALL cases are those with
rearranged MLL? 72 BCWABL fusion (Philadelphia chrornosome),2' or hypodip-
loidy (<45 c ~ ~ o ~ o s oThe ~ ~remaining
s ) . ~ ~cases, including those with the
t(1;19),8I can be classified as having intermediate-risk ALL.
The prognostic impact of age and, to a lesser extent, leukocyte count can be
explained by their association with specific genetic abnormalities. For example,
the overall poor prognosis of infants less than 12 months of age is due to the
very high frequency of MLL rearrangement in this age 88 It is well
recognized that hyperdiploidy (>50 chromosomes) ALL preferentially occurs in
children aged 1 to 9 years.79Recently, TEL/AMLl fusion rearrangements, another
highly favorable genetic abnormality, was found almost exclusively in this age
g r o ~ p . *Altogether,
~,~~ 70% of patients 1 to 9 years of age have one or the
other of these genetic abnormalities in their blast cells (Pui C-H, Unpublished
observation). Finally, rearranged MLL genes and BCWABL fusions are more
common in older children and typically are associated with high leukocyte
counts on pre~entation.~, 21, 72

CELL GROWTH AND DRUG-SENSITIVITY TESTING

The drug sensitivity of leukemic cells early in the clinical course is a


useful predictor of treatment outcome. Strategies used to assess early treatment
responsiveness include measurement of the clearance rate of circulating blasts
after single-agent or multiagent induction chemotherapy and the percentage of
bone marrow blasts during the early phase of remission induction therapy.',
26* loo Using semiquantitative polymerase chain reaction analyses or immunologic
methods, some investigators have correlated higher levels of minimal residual
leukemia in the immediate postinduction period with an increased likelihood of
relapse.14,
Recent studies suggest that leukemic cell growth in model systems has
prognostic significance in B-cell precursor ALL. In one study, patients whose
leukemic cells grew in mice with severe combined immunodeficiency had a
significantly higher relapse rate than did those whose cells failed to grow.'07 We
have found that higher rates of leukemic cell recovery after culture on stromal
cell layers confer a poor prognosis." Both systems can also be used to test the
drug sensitivity of leukemic cells. Other methods for testing drug sensitivity
include the 3-[4,5-dimethyl-thiazo1-2-yl]-2,5-diphenyltetrazolium bromide
(MTT) test, in which viable cells are identified by their ability to reduce MTT to
a colored formazan,60and the differential-staining cytotoxicity assay, in which
the staining properties of centrifuged samples of treated cells are used to distin-
guish viable from nonviable cells.9 The relative advantages and disadvantages
of these techniques remain to be established.

RISK-DIRECTED THERAPY

With the exception of B-cell ALL, which can be treated with short-term (<6
months) but very intensive chemotherapy, including high-dose cyclophospha-
mide, doxorubicin, vincristine, methotrexate, and cytarabine,'", 58, ** subtypes of
ALL require prolonged treatment for 2 to 2.5 ~ e a r s . 6Although
~ specific ap-
proaches to therapy may differ from center to center, contemporary protocols
invariably comprise four major elements: remission induction, intensification-
consolidation, prevention of overt CNS disease, and continuation of remission.
Induction treatment typically includes administration of a glucocorticoid (pred-
nisone and dexamethasone), vincristine, and L-asparaginase. The substitution of
dexamethasone for prednisone could reduce the risk of CNS relapse.40, In
theory, cancer chemotherapy would be curative if early treatment were sufficient
to eradicate malignant cells before they could acquire drug resistance. This
concept led to a successful series of clinical trials testing aggressive early therapy
with an increased number of agents, especially in patients with intermediate-
risk or high-risk ALL.* Contemporary treatment should induce complete remis-
sions in 98% to 99% of children with newly diagnosed ALL.
Most current trials specify a brief period immediately or soon after remis-
sion induction when patients receive intensification or consolidation therapy
designed to eradicate residual blast cells. This approach has been widely used
for high-risk patients and was recently credited with an improved outcome that
extended to intermediate-risk and lower-risk patients.I7,83, Io5
Effective therapy directed to subclinical CNS leukemia is an integral part of
curative treatment for ALL. Growing concern that cranial irradiation can cause
significant neurotoxicity and occasional brain tumors, especially in girls and
young children,18,63, 97, *lo has led many leukemia therapists to replace this mo-
dality with additional doses of intrathecal chemotherapy and more intensive
systemic treatment. Two recent studies suggested that this strategy is effective
in eradicating subclinical CNS leukemia, even in patients with intermediate-risk
or high-risk leukemia.20,35 Currently, cranial irradiation is reserved for patients
who have CNS leukemia at diagnosis or other very high-risk features (eg.,
hyperleukocytosis and Philadelphia chromosome).
Although continuation treatment with standard methotrexate and 6-mercap-
topurine secures long-term disease-free survival in most children with B-cell
precursor ALL, higher-risk cases and those with T-cell ALL benefit from more
aggressive therapy.70 Recent improvement in the outcome of such cases was
attributed to intensive use of L-asparaginase and doxorubicin in the Dana-Farber
85-01 to the introduction of high-dose methotrexate (5 g/mZ as a 24-
hour infusion) in the Berlin-Frankfurt-Miinster 86 ~ t u d y , "and~ to the addition
of the epipodophyllotoxins, cyclophosphamide and cytarabine, in the St. Jude
study Despite numerous innovations in postremission treatment, patients
with the Philadelphia chromosome and high leukocyte count,'2 and those with
the MLL rearrangement: 72 continue to have a dismal prognosis. Ongoing stud-
ies are testing the efficacy of allogeneic stem cell transplantation for this subset
of patients.

TREATMENT FOR RELAPSE

The bone marrow remains the most common site of relapse in ALL; in other
sites, such as the CNS and testes, the frequency of leukemia relapse has de-
creased to 5% or less. Available data suggest that for patients who develop
hematologic relapse on therapy or shortly thereafter, allogeneic hematopoietic

*References 13, 20, 27, 83,87, 93, and 101.


ACUTE LYMPHOBLASTIC LEUKEMIA 839

stem cell transplantation is the treatment of choice,52,Io8 and that autologous


transplantation offers no substantial advantage over chemotherapy as postinduc-
tion treatment.6 For patients without histocompatible related donors, matched
unrelated transplantation is a reasonable option and has yielded some encourag-
ing results.22The efficacy of remission retrieval therapy for patients with an
isolated CNS relapse depends largely on whether they have received prior CNS
irradiation. Intensive chemotherapy and craniospinal irradiation can be expected
to secure long-term second complete remissions in about two thirds of the
previously unirradiated g r o ~ p . 485~ .Among patients with earlier exposure to
prophylactic radiation, this rate generally does not exceed 30%, even when
intensive chemotherapy and CNS irradiation are used to retrieve remis~ions.~~
Hematopoietic stem cell transplantation offers a reasonable option, especially if
the child has received intensive systemic therapy before relapse. Historically,
one third of patients with early testicular relapse and two thirds with late
recurrences in this site became long-term survivors with salvage chemotherapy
and bilateral testicular irradiati~n.~" Whether this experience can be extrapo-
lated to children who have received contemporary intensive treatment remains
uncertain. It will be difficult to test the efficacy of transplantation versus chemo-
therapy in a randomized trial because increasingly smaller numbers of patients
are relapsing in the testes.

LATE SEQUELAE

Each year approximately 1500 childhood ALL patients in the United States
become long-term survivors of their disease, emphasizing the need for a better
understanding of the pattern and magnitude of late complications. This informa-
tion is important not only to pediatric oncologists developing new protocols,
but also to primary physicians who care for these patients in their later life.
Few complications are as devastating as a second cancer, especially brain
tumors or acute myeloid leukemia. Children who received cranial irradiation at
5 years of age or younger are most susceptible to the development of brain
tumors.53, Acute myeloid leukemia has been linked to intensive treatment
with the epipodophyllotoxins (teniposide and etoposide), with the risk of disease
development apparently depending on treatment schedule and the concomitant
use of other agents.75,77
Treatment with the anthracycline components has been associated with
cardiomyopathy, especially when these agents are given to young girls at high
cumulative doses.47, Cranial irradiation has been implicated in a number of
late sequelae other than the development of brain tumors, including neuropsy-
chological deficitsI8,97, and endocrine dysfunction resulting in obesity, short
stature, precocious puberty, and osteoporosis.Z8,
38, w, 69,96 In general, these compli-
cations are seen in girls more often than in boys, and in young children more
often than in older ones. Many children with profound growth retardation due
to leukemia or its treatment are receiving growth hormone replacement therapy.
Recent emphasis on the intensive use of methotrexate and glucocorticoids has
led to an increased frequency of neurotoxicitp 115 and aseptic necrosis of
respectively, underscoring the need for judicious use of even seemingly benign
agents. As newer and more intensive regimens enter clinical trials, close monitor-
ing for long-term side effects will assume even greater importance, as indicated
by the numerous complications associated with allogeneic hematopoietic stem
840 PUI

cell transplantation, with or without total-body irradiation.* Effective counseling


and early psychoeducational intervention are also needed because adult survi-
vors of childhood ALL are at risk of emotional disturbance and unemploy-
ment.lI9
To close on a positive note, there have been no indications of increased
birth defects or cancers among offspring of adult survivors of childhood
ALL.32, 16. 37.42

SUMMARY

Advances in the molecular and immunologic characterization of leukemic


cells have greatly aided the diagnosis and risk assignment of ALL, as well as
the monitoring of bone marrow samples for minimal residual disease. Currently,
75% of childhood cases have biologically and therapeutically relevant genetic
abnormalities. Although gene discoveries in ALL have not been directly trans-
lated into effective therapy, there is every reason to believe that this disease will
eventually yield to molecular intervention.", 91 In the meantime, efforts are
being made to enhance the efficacy of existing regimens while reducing their
toxic side effects. We have learned, for example, the following: high-dose metho-
trexate is more effective than lower-dose methotrexate, especially for T-cell
ALL50,Io2; patients who need drastic adjustment of mercaptopurine dosage due
to thiopurine S-methyltransferase deficiency can be prospectively identified1I8;
dexrazoxane (ICRF-187)could reduce anthracycline cardiotoxicity1I2;granulocyte
colony-stimulating factor can shorten hospital stays for febrile neutropenia after
intensive remission induction therapy6R;and prolonged low-dose epipodophyllo-
toxin treatment may reduce the risk of therapy-induced acute myeloid leukemia
without compromising treatment efficacy.I6 The challenge now is to identify
specific treatments for genetically defined subtypes of ALL.

References

1. Arich M, Basso G, Mandelli F, et a1 Good steroid response in vivo predicts a favorable


outcome in children with T-cell acute lymphoblastic leukemia. Cancer 75:1684, 1995
2. Bash RO, Hall S, Timmons CF, et a1 Does activation of the TALl gene occur in a
majority of patients with T-cell acute lymphoblastic leukemia? A Pediatric Oncology
Group study. Blood 86:666, 1995
3. Bearman SI: The syndrome of hepatic veno-occlusive disease after marrow trans-
plantation. Blood 85.3005, 1995
4. Behm FG, Raimondi SC, Frestedt JL, et a1 Rearrangement of the M L L gene confers a
poor prognosis in childhood acute lymphoblastic leukemia, regardless of presenting
age. Blood 87:2870, 1996
5. Behrendt H, Charrin C, Gibbons B, et al: Dicentric (9;12) in acute lymphocytic
leukemia and other hematological malignancies: Report from a dic(9;12) study group.
Leukemia 9:102, 1995
6. Borgmann A, Schmid H, Hartmann R, et al: Autologous bone-marrow transplants
compared with chemotherapy for children with acute lymphoblastic leukaemia in a
second remission: A matched-pair analysis. Lancet 3462373, 1995
7. Borgstrom 8, Bolme P: Thyroid function in children after allogeneic bone marrow
transplantation. Bone Marrow Transplantation 13:59, 1994

"References 3, 7, 29, 39, 61, 94, 99, and 103.


ACUTE LYMFHOBLASTIC LEUKEMIA 841

8. Borowitz MJ, Shuster JJ, Land VJ, et al: Myeloid-antigen expression in childhood
acute lymphoblastic leukemia. N Engl J Med 325:1379, 1991
9. Bosanquet AG: Correlations between therapeutic response of leukaemias and in vitro
drug-sensitivity assay. Lancet 337711, 1991
10. Bowman WP, Shuster JJ, Cook B, et al: Improved survival for children with B-cell
acute lymphoblastic leukemia and stage N small noncleaved-cell lymphoma: A
Pediatric Oncology Group study. J Clin Oncol 14:1252,1996
11. Braun S, Chen K, Battiwalla M, et al: Gene therapy strategies for leukemia. Mol Med
Today 3:39, 1997
12. Broniscer A, Ribeiro RC, Rivera GK, et al: Intensive chemotherapy for Philadelphia-
chromosome-positive childhood acute lymphoblastic leukemia: Prognostic impor-
tance of initial white blood cell count. Leukemia, in press
13. Camitta B, Mahoney D, Leventhal B, et al: Intensive intravenous methotrexate and
mercaptopurine treatment of higher-risk non-T, non-B acute lymphocytic leukemia:
A Pediatric Oncology Group Study. J Clin Oncol 121383, 1994
14. Campana D, Pui C-H Detection of minimal residual disease in acute leukemia:
Methodologic advances and clinical significance. Blood 85:1416, 1995
15. Campana D, van Dongen JJM, Mehta A, et al: Stages of T-cell receptor protein
expression in T-cell acute lymphoblastic leukemia. Blood 771546, 1991
16. Chen C-L, Fuscoe JC, Liu Q, et al: Relationship between cytotoxicity and site-specific
DNA recombination after in vitro exposure of leukemia cells to etoposide. J Natl
Cancer lnst 88:1840, 1996
17. Chessells JM, Bailey C, Richards S M Intensification of treatment and survival in all
children with lymphoblastic leukaemia: Results of UK Medical Research Council trial
UKALL X. Lancet 345:143, 1995
18. Christie D, Leiper AD, Chessells JM, et a1 Intellectual performance after presympto-
matic cranial radiotherapy for leukaemia: Effects of age and sex. Arch Dis Child
73:136, 1995
19. Cline MJ: The molecular basis of leukemia. N Engl J Med 330:328, 1994
20. Conter V, Aricb M, Valsecchi MG, et al: Extended intrathecal methotrexate may
replace cranial irradiation for prevention of CNS relapse in children with intermedi-
ate-risk acute lymphoblastic leukemia treated with Berlin-Frankfurt-Munster-based
intensive chemotherapy. J Clin Oncol 13:2497, 1995
21. Crist W, Carroll A, Shuster J, et al: Philadelphia chromosome positive childhood
acute lymphoblastic leukemia: Clinical and cytogenetic characteristics and treatment
outcome. A Pediatric Oncology Group study. Blood 76:489, 1990
22. Davies SM, Wagner JE, SKU X-0, et a1 Unrelated donor bone marrow transplantation
for children with acute leukemia. J Clin Oncol 15:557, 1997
23. Fink F-M, Koller U, Mayer H, et al: Prognostic significance of myeloid-associated
antigen expression on blast cells in children with acute lymphoblastic leukemia. Med
Pediatr Oncol21:340, 1993
24. Finklestein JZ, Miller DR, Feusner J, et al: Treatment of overt isolated testicular
relapse in children on therapy for acute lymphoblastic leukemia. A Report from the
Children's Cancer Group. Cancer 73219, 1994
25. Ford AM, Pombo-de-Oliveria MS, McCarthy KP, et al: Monoclonal origin of concor-
dant T-cell malignancy in identical twins. Blood 89:281, 1997
26. Gajjar A, Ribeiro R, Hancock ML, et al: Persistence of circulating blasts after 1
week of multiagent chemotherapy confers a poor prognosis in childhood acute
lymphoblastic leukemia. Blood 861292, 1995
27. Gaynon PS, Steinherz PG, Bleyer WA, et al: Improved therapy for children with acute
lymphoblastic leukemia and unfavorable presenting features: A follow-up report of
the Childrens Cancer Group Study CCG-106. J Clin Oncol 11:2234, 1993
28. Gilsanz V, Carlson ME, Roe TF, et al: Osteoporosis after cranial irradiation for acute
lymphoblastic leukemia. J Pediatr 117238, 1990
29. Giorgiani G, Bozzola M, Locatelli F, et al: Role of busulfan and total body irradiation
on growth of prepubertal children receiving bone marrow transplantation and results
of treatment with recombinant human growth hormone. Blood 86:825, 1995
30. Greaves MF: Aetiology of acute leukaemia. Lancet 349:344, 1997
042 PUI

31. Greaves MF, Colman SM, Beard MEJ, et al: Geographical distribution of acute lym-
phoblastic leukaemia subtypes: Second report of the Collaborative Group Study.
Leukemia 727, 1993
32. Green DM, Zevon MA, Lowrie G, et al: Congenital anomalies in children of patients
who received chemotherapy for cancer in childhood and adolescence. N Engl J Med
325:141, 1991
33. Gurney JG, Severson RK, Davis S, et al: Incidence of cancer in children in the United
States. Cancer 75:2186, 1995
34. Hanif I, Mahmoud H, Pui C-H. Avascular femoral head necrosis in pediatric cancer
patients. Med Pediatr Oncol 21:655, 1993
35. Hasle H, Helgestad J, Christensen JK, et al: Prolonged intrathecal chemotherapy
replacing cranial irradiation in high-risk acute lymphatic leukaemia: Long-term follow
up with cerebral computed tomography scans and endocrinological studies. Eur J
Pediatr 1 x 2 4 , 1995
36. Hawkins MM: Pregnancy outcome and offspring after childhood cancer: Data show
no evidence of mutagenesis of the germ cells. BMJ 309:1034, 1994
37. Hawluns MM, Draper GJ, Winter D L Cancer in the offspring of survivors of child-
hood leukaemia and non-Hodgkin lymphomas. Br J Cancer 71:1335, 1995
38. Hokken-Koelega ACS, van Doom JWD, Hahlen K, et al: Long-term effects of treat-
ment for acute lymphoblastic leukemia with and without cranial irradiation on
growth and puberty: A comparative study. Pediatr Res 33:577, 1993
39. Huma Z, Boulad F, Black P, et al: Growth in children after bone marrow transplanta-
tion for acute leukemia. Blood 86:819, 1995
40. Jones B, Freeman AI, Shuster JJ, et al: Lower incidence of meningeal leukemia when
prednisone is replaced by dexamethasone in the treatment of acute lymphocytic
leukemia. Med Pediatr Oncol 19:269, 1991
41. Karp JE, McCaffrey R E New avenues of translational research in leukemia and
lymphoma: Outgrowth of a Leukemia Society of America-National Cancer Institute
Workshop. J Natl Cancer Inst 861196, 1994
42. Kenney LB, Nicholson HS, Brasseux C, et al: Birth defects in offspring of adult
survivors of childhood acute lymphoblastic leukemia. A Childrens Cancer Group/
National Institutes of Health Report. Br J Cancer 71:1335, 1995
43. Kinlen LJ, Dickson M, Stiller CA: Childhood leukaemia and non-Hodgkin’s lym-
phoma near large rural construction sites, with a comparison with Sellafield nuclear
site. BMJ 310:763, 1995
44. Kumagai M, Manabe A, Pui C-H, et al: Stroma-supported culture of childhood B-
lineage acute lymphoblastic leukemia cells predicts treatment outcome. J Clin Invest
97755, 1996
45. Kumar P, Kun LE, H u s h HO, et al: Survival outcome following isolated central
nervous system relapse treated with additional chemotherapy and craniospinal irradi-
ation in childhood acute lymphoblastic leukemia. Int J Radiat Oncol Biol Phys
31:477, 1995
46. Latchman DS: Transcription-factor mutations and disease. N Engl J Med 334:28,1996
47. Lipshultz SE, Colan SD, Gelber RD, et al: Late cardiac effects of doxorubicin therapy
for acute lymphoblastic leukemia in childhood. N Engl J Med 324:808, 1991
48. Lipshultz SE, Lipsitz SR, Mone SM, et al: Female sex and higher drug dose as risk
factors for late cardiotoxic effects of doxorubicin therapy for childhood cancer. N
Engl J Med 332:1738, 1995
49. Mahoney DH, Nitschke R, Lauer S, et al: Neurotoxicity (NT) in children with acute
lymphoblastic leukemia (ALL) receiving intensive methotrexate (MTX) schedules. A
Pediatric Oncology Group (POG) Study [abstract]. Proc Am Soc Clin Oncol 15:366,
1996
50. Masson E, Relling MV, Synold TW, et al: Accumulation of methotrexate polygluta-
mates in lymphoblasts is a determinant of antileukemic effects in vivo. A rationale
for high-dose methotrexate. J Clin Invest 9773, 1996
51. McLean TW, Ringold S, Neuberg D, et al: TEL-AML-1 dimerizes and is associated
with a favorable outcome in childhood acute lymphoblastic leukemia. Blood
88:4252, 1996
ACUTE LYMPHOBLASTIC LEUKEMIA 843

52. Moussalem M, Esperou Bourdeau H, Devergie A, et al: Allogeneic bone marrow


transplantation for childhood acute lymphoblastic leukemia in second remission:
Factors predictive of survival, relapse and graft-versus-host disease. Bone Marrow
Transplantation 15:943, 1995
53. Neglia JP, Meadows AT, Robison LL, et al: Second neoplasms after acute lymphoblas-
tic leukemia in childhood. N Engl J Med 325:1330, 1991
54. Odame I, Reilly JJ, Gibson BES, et al: Patterns of obesity in boys and girls after
treatment for acute lymphoblastic leukaemia. Arch Dis Child 71:147, 1994
55. Okuda T, Shurtleff SA, Valentine MB, et al: Frequent deletion of p26”Kk/MTS2 and
plSNK4b/MTS2 in pediatric acute lymphoblastic leukemia. Blood 85:2321, 1995
56. Parkin DM, Clayton D, Black RJ, et al: Childhood leukaemia in Europe after Chemo-
byl: 5 year follow-up. Br J Cancer 731006, 1996
57. Parkin DM, Stiller CA, Draper GJ, et al: The international incidence of childhood
cancer. Int J Cancer 42:511, 1988
58. Patte C, Leverger G, Rubie H, et al: High cure rate in B-cell (Burkitt’s) leukemia in
the LMB 89 protocol of the French Pediatric Oncology Society [abstract]. Proc Am
SOCClin Oncol 12:317, 1993
59. Petridou E, Trichopoulos D, Dessypris N, et al: Infant leukaemia after in utero
exposure to radiation from Chemobyl. Nature 382:352, 1996
60. Pieters R, Huismans DR, Loonen AH, et al: Relation of cellular drug resistance
to long-term clinical outcome in childhood acute lymphoblastic leukaemia. Lancet
338:399, 1991
61. Pihkala J, Saarinen UM, Lundstrom U, et al: Effects of bone marrow transplantation
on myocardial function in children. Bone Marrow Transplantation 13:149, 1994
62. Pui C-H: Acute leukemia in children. Curr Opinion Hematol3:249, 1996
63. Pui C-H: Childhood leukemias. N Engl J Med 332:1618, 1995
64. Pui C-H: Childhood leukemias [reply to letter]. N Engl J Med 333:1286, 1995
65. Pui C-H, Behm FG, Crist WM: Clinical and biologic relevance of immunologic marker
studies in childhood acute lymphoblastic leukemia. Blood 82343, 1993
66. Pui C-H, Behm FG, Downing JR, et al: llq23/MLL rearrangement confers a poor
prognosis in infants with acute lymphoblastic leukemia. J Clin Oncol 12:909, 1994
67. Pui C-H, Behm FG, Singh B, et al: Myeloid-associated antigen expression lacks
prognostic value in childhood acute lymphoblastic leukemia treated with intensive
multiagent chemotherapy. Blood 75:198, 1990
68. Pui C-H, Boyett JM, Hughes WT, et a1 Human granulocyte colony-stimulating factor
__ ~-
after induction chemotherapy in children with acute lymphoblastic
J Med, in press *
leukemia. N Engl
-
69. Pui C-H, Crist WM: Biology and treatment of acute lymphoblastic leukemia. J Pediatr
124491, 1994
70. Pui C-H, Crist WM: Treatment of childhood leukemias. Curr Opin Oncol 736, 1995
71. Pui C-H, Crist WM, Look AT: Biology and clinical significance of cytogenetic abnor-
malities in childhood acute lymphoblastic leukemia. Blood 76:1449, 1990
72. Pui C-H, Frankel LS, Carroll AJ, et al: Clinical characteristics and treatment outcome
of childhood acute lymphoblastic leukemia with the t(4;11)(q21;q23): A collaborative
study of 40 cases. Blood 77440, 1991
73. Pui C-H, Raimondi SC, Dodge RK, et a1 Prognostic importance of structural chromo-
somal abnormalities in children with hyperdiploid (>50 chromosomes) acute lym-
phoblastic leukemia. Blood 73:1963, 1989
74. Pui C-H, Raimondi SC, Hancock ML, et a 1 Immunologic, cytogenetic, and clinical
characterization of childhood acute lymphoblastic leukemia with the t(1;19)(q23;p13)
or its derivative. J Clin Oncol 12:2601, 1994
75. Pui C-H, Relling M V , Rivera GK, et al: Epipodophyllotoxin-related acute myeloid
leukemia: A study of 35 cases. Leukemia 9:1990, 1995
76. Pui C-H, Ribeiro RC, Campana D, et a1 Prognostic factors in the acute lymphoid and
myeloid leukemias of infants. Leukemia 10:952, 1996
77. Pui C-H, Ribeiro RC, Hancock ML, et al: Acute myeloid leukemia in children treated
with epipodophyllotoxins for acute lymphoblastic leukemia. N Engl J Med 325:1682,
1991
78. Pui C-H, Schell MJ, Raimondi SC, et al: Myeloid-antigen expression in childhood
acute lymphoblastic leukemia. N Engl J Med 325:1378, 1991
79. Pui C-H, Williams DL, Roberson PK, et al: Correlation of karyotype and immunophe-
notype in childhood acute lymphoblastic leukemia. J Clin Oncol6:56, 1988
80. Rabbitts TH: Chromosomal translocations in human cancer. Nature 372143, 1994
81. Raimondi SC, Behm FG, Roberson PK, et al: Cytogenetics of pre-B-cell acute lympho-
blastic leukemia with emphasis on prognostic implications of the t(1;19). J Clin Oncol
8:1380, 1990
82. Raimondi SC, Frestedt JL, Pui C-H, et al: Acute lymphoblastic leukemias with deletion
of llq23 or a novel inversion (ll)(p13q23) lack MLL gene rearrangements and have
favorable clinical features. Blood 86:1881, 1995
83. Reiter A, Schrappe M, Ludwig W-D, et al: Chemotherapy in 998 unselected childhood
acute lymphoblastic leukemia patients. Results and conclusions of the Multicenter
Trial ALL-BFM 86. Blood W3122, 1994
84. Reiter A, Schrappe M, Ludwig W-D, et al: Favorable outcome of B-cell acute lympho-
blastic leukemia in childhood: A report of three consecutive studies of the BFM
Group. Blood 802471, 1992
85. Ribeiro RC, Rivera GK, Hudson M, et al: An intensive re-treatment protocol for
children with an isolated CNS relapse of acute lymphoblastic leukemia. j Clin Oncol
13:333, 1995
86. Ries LAG, Miller BA, Hankey BF, et a1 (eds): SEER Cancer Statistics Review 1973-
1991: Tables and Graphs. Betkesda, M D National Cancer Institute, 1994, publication
#NIH-2789
87. Rivera GK, Raimondi SC, Hancock ML, et al: Improved outcome in childhood acute
lymphoblastic leukaemia with reinforced early treatment and rotational combination
chemotherapy. Lancet 33761, 1991
88. Rubnitz JE, Behm FG, Downing J R llq23 rearrangements in acute leukemia. Leuke-
mia 10:74, 1996
89. Rubnitz JE, Downing JR, Pui C-H, et al: TEL gene rearrangement in acute lymphoblas-
tic leukemia: A new genetic marker with prognostic significance. J Clin Oncol
15:1150, 1997
90. Rubnitz JE, Shuster JJ, Land VJ, et a1 Case-control study suggests a favorable impact
of TEL rearrangement in patients with B-lineage acute lymphoblastic leukemia treated
with antimetabolite-based therapy. A Pediatric Oncology Group study. Blood
89:1143, 1997
91. Sawyers C L Molecular genetics of acute leukemia. Lancet 349:196, 1997
92. Sandler DP: Recent studies in leukemia epidemiology. Curr Opin Oncol 712, 1995
93. Schorin MA, BIattner S, Gelber RD, et al: Treatment of childhood acute lymphoblastic
leukemia: Results of Dana-Farber Cancer Institute/Children’s Hospital acute lym-
phoblastic leukemia consortium protocol 85-01. J Clin Oncol 12740, 1994
94. Schultz KR, Green GJ, Wensley D, et al: Obstructive lung disease in children after
allogeneic bone marrow transplantation. Blood 84:3212, 1994
95. Shurtleff SA, Buijs A, Behm FG, et al: TEL/AMLl fusion resulting from a cryptic
t(12;21) is the most common genetic lesion in pediatric ALL and defines a subgroup
of patients with an excellent prognosis. Leukemia 9:1985, 1995
96. Sklar C, Mertens A, Walter A, et al: Final height after treatment for childhood acute
lymphoblastic leukemia: Comparison of no cranial irradiation with 1800 and 2400
centigrays of cranial irradiation. J Pediatr 123:59, 1993
97. Smibert E, Anderson V, Godber T, et a1 Risk factors for intellectual and educational
sequelae of cranial irradiation in childhood acute lymphoblastic leukaemia. Br J
Cancer 73825, 1996
98. Smith M, Arthur D, Camitta 8, et al: Uniform approach to risk classification and
treatment assignment for children with acute lymphoblastic leukemia. J Clin Oncol
1418, 1996
99. Socie G, Selimi F, Sedel I: Avascular necrosis of bone after allogeneic bone marrow
transplantation: Clinical findings, incidence and risk factors. Br J Haematol 86:624,
1994
100. Steinherz PG, Gaynon PS, Breneman JC, et al: Cytoreduction and prognosis in acute
ACUTE LYMPHOBLASTIC LEUKEMIA 845

lymphoblastic leukemia: The importance of early marrow response: Report from the
Childrens Cancer Group. J Clin Oncol 14:389, 1996
101. Steinherz PG, Redner A, Steinherz L, et a1 Development of a new intensive therapy
for acute lymphoblastic leukemia in children at increased risk of early relapse. Cancer
723120, 1993
102. Synold TW, Relling MV, Boyett JM, et al: Blast cell methotrexate-polyglutamate
accumulation in vivo differs by lineage, ploidy, and methotrexate dose in acute
lymphoblastic leukemia. J Clin Invest 94:1996, 1994
103. Tichelli A, Grahvohl A, Egger T, et al: Cataract formation after bone marrow trans-
plantation. Ann Intern Med 119:1175, 1993
104. Trueworthy R, Shuster J, Look T, et al: Ploidy of lymphoblasts is the strongest
predictor of treatment outcome in B-progenitor cell acute lymphoblastic leukemia of
childhood: A Pediatric Oncology Group study. J Clin Oncol 10606, 1992
105. Tubergen DG, Gilchrist GS, OBrien RT, et a1 Improved outcome with delayed
intensification for children with acute lymphoblastic leukemia and intermediate pre-
senting features: A Childrens Cancer Group Phase I11 Trial. J Clin Oncol 11:527,
1993
106. Uckun FM, Sather H, Gaynon P, et al: Clinical features and treatment outcome of
children with myeloid antigen positive acute lymphoblastic leukemia: A report from
the Childrens Cancer Group. Blood, in press
107. Uckun FM, Sather H, Reaman G, et al: Leukemic cell growth in SCID mice as
a predictor of relapse in high-risk 8-lineage acute IympKoblastic leukemia. Blood
85:873, 1995
108. Uderzo C, Valsecchi MG, Bacigalupo A, et al: Treatment of childhood acute lympho-
blastic leukemia in second remission with allogeneic bone marrow transplantation
and chemotherapy: Ten-year experience of the Italian bone marrow transplantation
group and the Italian Pediatric Hematology Oncology Association. J Clin Oncol
13:352, 1995
109. Veerman AJP, Hahlen K, Kamps WA, et al: High cure rate with a moderately
intensive treatment regimen in non-high-risk childhood acute lymphoblastic leuke-
mia: Results of Protocol ALL VI from the Dutch Childhood Leukemia Study Group.
J Clin Oncol 14911, 1996
110. Waber DP, Tarbell NJ, Fairclough D, et al: Cognitive sequelae of treatment in child-
hood acute lymphoblastic leukemia: Cranial radiation requires an accomplice. J Clin
Oncol 13:2490, 1995 *
111. Wasserman R, Galili N, Ito Y, et al: Residual disease at the end of the induction
therapy as a predictor of relapse during therapy in childhood B-lineage acute lymph-
oblastic leukemia. J Clin Oncol 10:1879, 1992
112. Wexler LH, Andrich MP, Venzon D, et al: Randomized trial of the cardioprotective
agent ICRF-187 in pediatric sarcoma patients treated with doxorubicin. J Clin Oncol
14362, 1996
113. Whitehead VM, Vuchich MJ, Lauer SJ, et al: Accumulation of high levels of methotrex-
ate polyglutamates in lymphoblasts from children with hyperdiploid (>50 chromo-
somes) B-lineage acute lymphoblastic leukemia: A Pediatric Oncology Group study.
Blood 80:1316, 1992
114. Wiersma SR, Ortega J, Sobel E, et al: Clinical importance of myeloid-antigen expres-
sion in acute lymphoblastic leukemia of childhood. N Engl J Med 324:800, 1991
115. Winick NJ, Bowman WP, Kamen BA, et al: Unexpected acute neurologic toxicity in
the treatment of children with acute lymphoblastic leukemia. J Natl Cancer Inst
84252,1992
116. Witherspoon RP, Fisher LD, Schoch G, et al: Secondary cancers after bone marrow
transplantation for leukemia or aplastic anemia. N Engl J Med 321:784, 1989
117. Wofford MM, Smith SD, Shuster JJ, et al: Treatment of occult or late overt testicular
relapse in children with acute lymphoblastic leukemia: A Pediatric Oncology Group
Study. J Clin Oncol 10624,1992
118. Yates CR, Krynetski EY, Loennechen T, et al: Molecular diagnosis of thiopurine
S-methyltransferase deficiency: Genetic basis for azathioprine and mercaptopurine
intolerance. Ann Intern Med, 126:608, 1997
846 PUI

119. Zeltzer LK, Chen E, Weiss R, et al: Comparison of psychologic outcome in adult
survivors of childhood acute lymphoblastic leukemia versus sibling controls: A coop-
erative Children’s Cancer Group and National Institutes of Health study. J Clin Oncol
15:547, 1997

Address reprint requests to


Ching-Hon Pui, MD
Department of Hematology/Oncology
St. Jude Children’s Research Hospital
332 North Lauderdale Street
Memphis, TN 38105

You might also like