Anda di halaman 1dari 11

D RUG TH ERA PY

Review Article

Drug Therapy cytes are available,10 only a few of these molecules


are truly lineage-specific. For this reason a panel of
antibodies is needed to establish the diagnosis and
A L A S T A I R J . J . W O O D , M. D. , Editor to distinguish among the immunologic subclasses.
The panel used at St. Jude Children’s Research
Hospital includes at least one marker that is highly
A CUTE L YMPHOBLASTIC L EUKEMIA sensitive (CD19 and CD7 for B-lineage and T-lin-
eage cells, respectively, and CD13 or CD33 for my-
CHING-HON PUI, M.D., AND WILLIAM E. EVANS, PHARM.D.
eloid cells) and one marker that is highly specific
(cytoplasmic CD79a and cytoplasmic CD3 for
B-lineage and T-lineage cells, respectively, and cyto-
plasmic myeloperoxidase for myeloid cells). On the
basis of these immunophenotypic analyses, a firm

A
CUTE lymphoblastic leukemia (ALL) is di-
agnosed in 3000 to 4000 persons in the
United States each year; two thirds of them
are children.1,2 The current rate of cure of nearly 80
percent in children attests to remarkable progress in Probability of Event-freeD 100
81±8%
the development of effective treatments for resistant XIIIA, XIIIB (n=366) 1991–1997
subtypes of the disease. Progress has been incremen- 80
Survival (%)

tal, from the introduction of combination chemo- 70±2% XI, XII (n=546) 1984–1991
60
therapy and central nervous system treatment for
53±2% X (n=428) 1979–1983
presymptomatic leukemia to newer, intensive treat-
40
ment regimens for patients at high risk for relapse 36±2%
V–IX (n=825) 1967–1979
(Fig. 1). In contrast, only 30 to 40 percent of adults
20
with ALL are cured.2,3 This discrepancy can be at- I–IV (n=90) 1962–1966
tributed in part to the higher frequency of adverse 9±3%
0
genetic abnormalities in the leukemic lymphoblasts 0 5 10 15 20 25 30 35
of adults.4-8 Here, we review advances in the classifi-
cation of lymphoblasts and in clinical management 100
that have contributed to recent gains in therapeutic
Probability of OverallD

86±6%
XIIIA, XIIIB (n=366) 1991–1997
outcome or that hold promise for the future. 80 81±2%
XI, XII (n=546) 1984–1991
Survival (%)

BIOLOGIC CHARACTERIZATION 74±2% X (n=428) 1979–1983


OF LEUKEMIC CELLS 60
V–IX (n=825) 1967–1979
ALL can develop from any lymphoid cell blocked 40 48±2%
at a particular stage of development, including prim-
itive cells with multilineage potential.8,9 In contrast 20
to acute myeloid leukemic cells, which can be readily 21±4%
I–IV (n=90) 1962–1966

identified in most instances by the presence of Auer 0


rods, myeloperoxidase, or monocyte-associated ester- 0 5 10 15 20 25 30 35
ases, leukemic lymphoblasts lack specific morpho- Years after Diagnosis
logic or cytochemical features, so that the diagnosis
of ALL depends on immunophenotyping. Although Figure 1. Kaplan–Meier Analyses of Event-free Survival (Top
Panel) and Overall Survival (Bottom Panel) in 2255 Children
monoclonal antibodies against 166 different cluster- with ALL in 13 Consecutive Studies Conducted at St. Jude
of-differentiation (CD) molecules on human leuko- Children’s Research Hospital from 1962 to 1997.
The results demonstrate improvement in survival with the
introduction of therapy for subclinical disease of the central
nervous system (studies V to IX, 1967 to 1979); with early inten-
sification of systemic chemotherapy, including high-dose meth-
From St. Jude Children’s Research Hospital (C.-H.P., W.E.E.) and
the Colleges of Medicine (C.-H.P., W.E.E.) and Pharmacy (W.E.E.), Uni-
otrexate (study X, 1979 to 1983, and studies XI and XII, 1984 to
versity of Tennessee, Memphis. Address reprint requests to Dr. Pui at St. 1991); and with reinduction treatment, early intensification of
Jude Children’s Research Hospital, 332 N. Lauderdale, Memphis, TN intrathecal chemotherapy, and pulsed therapy with dexameth-
38105-0318. asone and vincristine (studies XIIIA and XIIIB, 1991 to 1997).
©1998, Massachusetts Medical Society. The mean (±SE) five-year survival probabilities are shown.

Vol ume 33 9 Numb e r 9 · 605

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
The Ne w E ng l a nd Jo u r na l o f Me d ic i ne

diagnosis can be made in 99 percent of cases. Al- RISK ASSESSMENT


though cases involving B-lineage and T-lineage cells Although investigators agree that stringent evalu-
can be further subclassified according to the recog- ation of the risk of relapse is needed at the time of
nized steps of normal B- and T-cell maturation,8 the diagnosis to avert undertreatment or overtreatment,
only distinctions with therapeutic importance are there is considerable disagreement about risk criteria
those between the B-cell–precursor immunopheno- and the terminology for defining risk subgroups. For
type and the T-cell or mature–B-cell immunophe- the purposes of this review, three categories of risk
notype. — low, standard (average), and high — are consid-
Depending on the criteria used and the number of ered. Many presenting clinical and biologic variables
antigens tested, myeloid-associated–antigen expres- initially thought to be useful predictors of outcome
sion can be detected in as many as one fourth of chil- have proved to be of little value as treatment has im-
dren and one third of adults with ALL.7,8,11 This fea- proved.16 For example, patients with T-cell or B-cell
ture has no prognostic or therapeutic implications,11-13 ALL, once thought to have a very poor prognosis,
but it can be useful in immunologic monitoring for now have as favorable an outcome as patients with
minimal residual leukemia.14 The few patients with standard-risk B-cell–precursor ALL.2,3,12,17-22 Like-
leukemic cells that express both lymphoid-associated wise, in two independent trials of more intensive
molecules (usually CD2 and CD7) and myeloid- therapy,23,24 the poor prognosis previously associat-
restricted molecules (such as myeloperoxidase) re- ed with adolescent age or black race was not found.
quire treatment directed toward both lineages.11 Participants at a recent workshop25 agreed on cri-
Specific genetic abnormalities (e.g., chromosomal teria for defining a low risk of recurrent disease in
gains or losses, resulting in hyperdiploidy or hypodip- children with newly diagnosed B-cell–precursor ALL:
loidy, respectively; chromosomal translocations, lead- an age of one to nine years and a leukocyte count of
ing to the formation of transforming fusion genes or less than 50,000 per cubic millimeter. Patients in oth-
dysregulation of gene expression; and deletion or er age groups or those with higher leukocyte counts
functional inactivation of tumor-suppressor genes) were considered to be at high risk.25 These prognostic
are found in the blast cells of 60 to 75 percent of pa- relations can be explained to a large extent by the
tients with ALL (Fig. 2). The recognition of these presence of specific genetic abnormalities.
abnormalities has contributed greatly to our under- For example, among children less than one year
standing of the pathogenesis and prognosis of the old, in whom the prognosis is poor, 70 to 80 per-
disease.4,15 cent have rearrangements of the MLL gene.26,27 In

HyperdiploidyD
(>50 chromosomes)D
E2A–PBX1D 6% ETV6–CBFA2D
t(1;19) t(12;21)D
2%
5% MYCD 3%
HyperdiploidyD 2% t(8;14), t(2;8), t(8;22) 4%
(>50 chromosomes)D
25% 4% TCRad 6%
14q11
3% 2% RandomD
ETV6–CBFA2D TCRbD 41%
t(12;21)D 7q35
22% 1% 4%
HypodiploidyD
(<45 chromosomes)

RandomD 6% MLL rearrangementsD 7% BCR–ABLD


28% t(4;11), t(11;19), t(1;11) t(9;22)D
25%
BCR–ABLD
t(9;22)D
4%
Children Adults
Figure 2. Estimated Frequencies of Specific Genotypes among Children and Adults with ALL.
The data are from studies at St. Jude Children’s Research Hospital4,8 and from the Groupe Français de Cytogénétique Héma-
tologique,5 Chessells et al.,6 and Copelan and McGuire.7

606 · Augus t 2 7 , 19 9 8

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
D R UG TH ERA PY

adolescent and adult patients, the frequencies of whom remission is identified solely on the basis of
MLL rearrangements and of BCR–ABL fusion, an- morphologic criteria. On the other hand, the results
other genetic abnormality associated with a poor of qualitative assessment of some leukemia-specific
prognosis, are high.5,6,28 In contrast, two favorable fusion transcripts in patients in remission do not re-
genetic abnormalities — hyperdiploidy (>50 chro- liably predict subsequent relapse.35 Additional stud-
mosomes per cell) and ETV6–CBFA2 (TEL–AML1) ies are needed to establish the clinical utility of de-
fusion — occur mainly in children one to nine years termining drug sensitivity and growth characteristics
old; hyperdiploidy is associated with a low leukocyte of leukemic cells in vitro.36-38
count as well.9,29,30 We propose a risk-classification system (Fig. 4) that
A risk-assignment system based on primary genet- considers the blast-cell immunophenotype and geno-
ic abnormalities has great intuitive appeal; however, type along with the presenting clinical features and
as shown in Figure 3, the predictive value of these degree of early responsiveness to treatment. Although
abnormalities is not high. For example, as many as the presence of BCR–ABL fusion in leukemic cells
20 percent of children with ALL who have hyper- usually indicates high-risk leukemia warranting hem-
diploidy or the ETV6–CBFA2 fusion gene eventual- atopoietic stem-cell transplantation, patients with this
ly have a relapse.4,29,30 A useful measure in risk assess- genetic abnormality who have low leukocyte counts
ment is the rate of clearance of leukemic cells from at diagnosis or good early responses to prednisone
the blood or bone marrow during the early phase of therapy may be cured by intensive chemotherapy
therapy. In patients with B-lineage or T-lineage ALL, alone.39,40 Patients with near-haploid or markedly hy-
slow clearance of the cells has proved to be an indi- podiploid leukemic cells tend to have a poor progno-
cator of poor prognosis.18,31,32 sis and, regardless of their age or initial leukocyte
Another approach is to use the polymerase chain count, should undergo treatment at least as aggressive
reaction or immunologic methods to measure min- as that given to patients with standard-risk leukemia.41
imal residual disease soon after the induction of a Patients with B-cell–precursor ALL with MLL gene
clinical remission, when some patients may still have rearrangements, especially infants with the t(4;11)
as many as 10 billion leukemic cells.14,33,34 Patients translocation and MLL–AF4 fusion, usually have a
who have a “molecular” or “immunologic” remission, poor response to chemotherapy.28,42 However, there
defined as leukemic involvement of less than 0.01 is a subgroup of patients with MLL rearrangements
percent of nucleated bone marrow cells, are predict- who have a good response42 (Fig. 3): those with
ed to have a better clinical outcome than those in T-cell leukemia with the t(11;19) translocation and

100
HyperdiploidyD
(>50 chromosomes)D
(n=193)
80 ETV6–CBFA2 (n=82)
Event-free Survival (%)

E2A–PBX1 (n=34)

60

BCR–ABL (n=30)
40
MLL rearrangements (n=30)

20

0
0 2 4 6 8 10 12 14

Years after Diagnosis

Figure 3. Kaplan–Meier Analysis of Event-free Survival According to Genetic Features of Blast Cells
in 369 Children with ALL Treated at St. Jude Children’s Research Hospital from 1984 to 1997.
The relatively good survival of patients with BCR–ABL fusion reflects successful treatment of a sub-
group with low leukocyte counts at diagnosis. The E2A–PBX1 abnormality, once associated with poor
survival,8 now is associated with the same favorable prognosis as the ETV6–CBFA2 (TEL–AML1)
abnormality.

Vol ume 33 9 Numb e r 9 · 607

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
The Ne w E ng l a nd Jo u r na l o f Me d ic i ne

T cell B-cell precursor

Other genotypes orD Hyperdiploidy (>50D


BCR–ABL fusion orD CNS or testicularD chromosomes) orD
MLL rearrangement leukemia ETV6–CBFA2 fusion

BCR–ABL fusion andD Age <1 or »10 years,D


Age 1 to 9 yearsD
leukocyte countD OtherD leukocyte countD
and leukocyte countD
>25,000/mm3 or MLLD presentations »50,000/mm3, or CNSD
<50,000/mm3
rearrangement in infants or testicular leukemia

ProvisionalD ProvisionalD ProvisionalD


standard risk standard risk low risk

Poor earlyD
response orD Good earlyD Failure ofD Poor earlyD Good earlyD Failure ofD Poor earlyD Good earlyD
failure ofD response induction response response induction response response
induction

High risk Standard risk Low risk

Figure 4. Proposed Risk-Classification System for ALL According to Immunophenotype and Genotype, Age, Leukocyte Count, Early
Response to Treatment, and Presence or Absence of Extramedullary Disease (in B-Cell–Precursor Disease).
All patients with T-cell ALL are considered to have a standard risk of relapse, except those without a good response to remission-
induction therapy. Among patients with B-cell–precursor ALL, the high-risk group is defined by the presence of the BCR–ABL fusion
gene and a high leukocyte count or a poor early response to chemotherapy induction, MLL rearrangements (in infants), or lack of
response to induction therapy. Patients with low-risk ALL have a B-cell–precursor immunophenotype with hyperdiploidy (>50 chro-
mosomes), the ETV6–CBFA2 (TEL–AML1) fusion gene, or an age of one to nine years and a leukocyte count of less than 50,000 per
cubic millimeter, with a favorable early response to chemotherapy. All other patients with B-cell–precursor ALL are considered to
have a standard risk. Antimetabolite therapy with dexamethasone and vincristine pulses is sufficient for treatment of low-risk
leukemia, whereas standard-risk leukemia requires more intensive multidrug therapy. Patients with high-risk ALL are candidates for
hematopoietic stem-cell transplantation. Gradations of color within a box indicate the estimated proportions of patients in each of
the three risk categories. CNS denotes central nervous system. Patients with mature–B-cell ALL are not included in this classifica-
tion because of their requirement for highly specialized therapy.

MLL–ENL fusion (unpublished data). Although


T-cell ALL is treated as standard-risk leukemia at TABLE 1. COMPARISON OF TREATMENT OUTCOMES ACCORDING TO
DISEASE SUBTYPE IN ADULTS AND CHILDREN WITH ACUTE
virtually all centers, patients with high leukocyte LYMPHOBLASTIC LEUKEMIA.*
counts (>100,000 per cubic millimeter) or delayed
early responses require even more intensive central
nervous system–specific therapy than do other pa- SUBTYPE ESTIMATED EVENT-FREE SURVIVAL (%)

tients with this immunophenotype.43 The presence ADULTS CHILDREN

of any leukemic cells in the cerebrospinal fluid at di- B cell (rearranged MYC ) 50–55 at 4 yr 75–85 at 5 yr
agnosis may indicate that intensive intrathecal treat- B-cell precursor†
Hyperdiploidy (>50 chromosomes) 30–50 at 3 yr 80–90 at 5 yr
ment is needed to prevent relapse of central nervous ETV6–CBFA2 (TEL–AML1) Unknown 85–90 at 5 yr
system disease.44 E2A–PBX1 20–40 at 3 yr 70–80 at 5 yr
In adults, the frequency of the BCR–ABL fusion BCR–ABL <10 at 3 yr 20–40 at 5 yr
MLL–AF4 10–20 at 3 yr 10–35 at 5 yr
gene is higher and the frequency of favorable genetic Hypodiploidy (<45 chromosomes) 10 at 3 yr 25–40 at 3 yr
changes is lower than in children (Fig. 2).2,3,5-7 Even T cell 50 at 3 yr 65–75 at 5 yr
for the same genetic subtypes of ALL, survival rates
*This analysis reflects the results of nine studies.4-12
are lower in adults than in children (Table 1). Thus,
†The four fusion genes listed here arise from the following alterations:
although several risk models have been proposed for ETV6–CBFA2, t(12;21)(p13;q22); E2A–PBX1, t(1;19)(q23;p13); BCR–
adults with ALL,2 the prevailing view is that most af- ABL, t(9;22)(q34;q11); and MLL–AF4, t(4;11)(q21;q23).

608 · Augus t 2 7 , 19 9 8

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
D R UG TH ERA PY

fected adults should be considered to have a high even before chemotherapy is initiated. These pa-
risk of recurrence and should be treated accordingly. tients should be given intravenous hydration, sodi-
Adults younger than 30 years of age have a better um bicarbonate to alkalize the urine, allopurinol to
prognosis than those 30 to 59 years old, who in turn treat hyperuricemia, and aluminum hydroxide or
have a better prognosis than patients who are 60 or calcium carbonate (if the serum calcium concentra-
older.2,3,6 Adults with some types of ALL require tion is low) to treat hyperphosphatemia. Allopuri-
treatment modification: those with B-cell disease, nol, by inhibiting purine synthesis in leukemic blast
because of its unique sensitivity to therapy19,20; those cells, may reduce the peripheral blast-cell count
with Philadelphia-chromosome–positive disease, be- before chemotherapy is begun.47 Nonrecombinant
cause of its resistance to chemotherapy alone5,6; those urate oxidase, available in France and Italy, converts
with pre–T-cell (CD7+, CD2¡, CD5¡) leukemia, uric acid to allantoin (a readily excreted metabolite
because the response to chemotherapy is not as that is 5 to 10 times as soluble as uric acid) and
good as that in patients with leukemia of a more ma- decreases the serum uric acid concentration more
ture T-cell phenotype7; and elderly patients, who tol- rapidly than allopurinol; however, it can cause acute
erate treatment poorly.3 hypersensitivity reactions and, in patients with glu-
cose-6-phosphate dehydrogenase deficiency, can cause
SUPPORTIVE CARE methemoglobinemia or hemolytic anemia.48
At least half the patients with ALL present with For patients with extreme leukocytosis (leukocyte
fever. Often the fever is induced by pyrogenic cyto- count, >200,000 per cubic millimeter), either leu-
kines released from leukemic cells, including inter- kapheresis or exchange transfusion (in small chil-
leukin-1, tumor necrosis factor, and interleukin-6,45 dren) may be used to reduce the burden of leukemic
but in about a third of the patients it is due to infec- cells,49 although the short- and long-term benefits
tion. Therefore, broad-spectrum antibiotic therapy of these procedures remain in question.50 Emergen-
should be initiated in patients with fever, especially cy cranial irradiation has no therapeutic role in these
those with neutropenia, until a diagnosis of infec- patients.49,50 Other supportive care measures, includ-
tious disease has been excluded. At most centers, all ing the use of indwelling catheters and psychosocial
patients are treated prophylactically for Pneumocystis support, are important, but their discussion is be-
carinii pneumonia with trimethoprim–sulfameth- yond the scope of this review.
oxazole, given three days per week.46 Alternative
treatments for patients with intolerance to trimeth- TREATMENT
oprim–sulfamethoxazole include aerosolized pen- The improved rate of cure of ALL can be attrib-
tamidine, dapsone, and atovaquone. uted in large measure to the development of more
In patients with B-cell or T-cell ALL or B-cell– effective multidrug regimens in well-designed clinical
precursor leukemia with a large burden of leukemic trials. Table 2 summarizes several treatment strate-
cells, hyperuricemia, hyperkalemia, and hyperphos- gies that have been found to have a particularly im-
phatemia with secondary hypocalcemia are common, portant effect on outcome.

TABLE 2. TREATMENT STRATEGIES ASSOCIATED WITH IMPROVED OUTCOMES IN CLINICAL STUDIES OF ALL.

STRATEGY TYPE OF DISEASE REFERENCE

Fractionated high-dose cyclophosphamide, Mature–B-cell ALL in chil- Soussain et al.,19 Hoelzer et al.,20 Reiter et al.,21 Patte 22
high-dose methotrexate (3 to 8 g/m2), dren and adults
and cytarabine
Intensive induction treatment with four ALL in children Reiter et al.,18 Rivera et al.,51 Niemeyer et al.,52 Gaynon et al.53
or more drugs
Dexamethasone for subclinical central ALL in children Bostrom et al.,54 Veerman et al.55
nervous system involvement
Intensification (consolidation) therapy ALL in children and young Schorin et al.,17 Reiter et al.,18 Rivera et al.,51 Niemeyer et al.,52
shortly after induction of remission adults Gaynon et al.,53 Veerman et al.,55 Rohatiner et al.,56 Ma-
honey et al.,57 Chessells et al.,58 Hoelzer et al.59
High-dose methotrexate (5 g/m2) T-cell ALL in children Reiter et al.18
Increased exposure to systemic metho- ALL in children Evans et al.60,61
trexate
Reinduction treatment ALL in children and adults Reiter et al.,18 Hoelzer et al.,59 the Childhood ALL Collabo-
rative Group,62 Tubergen et al.,63 Nachman et al.64
Early and intensive intrathecal chemother- ALL in children and adults Reiter et al.,18 Veerman et al.,55 Cortes et al.,65 Nachman
apy for subclinical central nervous sys- et al.,66 Pui et al.67
tem involvement

Vol ume 33 9 Numb e r 9 · 609

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
The Ne w E ng l a nd Jo u r na l o f Me d ic i ne

At virtually all centers, patients with mature–B-cell nisone against relapse of central nervous system
ALL are treated with short-term (two-to-eight- disease in children with ALL.54,55 Three forms of as-
month) regimens of intensive chemotherapy that in- paraginase, each with a different pharmacokinetic
clude fractionated high-dose cyclophosphamide, high- profile, are available. In one randomized trial, the
dose methotrexate, and cytarabine.19-22 Some studies outcome in patients treated with asparaginase de-
have suggested that the addition of ifosfamide, eto- rived from Escherichia coli was better than that in pa-
poside, or both to these regimens may improve the tients treated with Erwinia carotovora asparaginase,
cure rate further.20-22 In a French study of 102 chil- which has a shorter half-life in plasma.7 The efficacy
dren, a seven-drug regimen including high-dose of various anthracyclines in adults with ALL has
methotrexate (8 g per square meter of body-surface been similar.73,74
area), cytarabine, cyclophosphamide, and etoposide
led to a cure rate of 85 percent,22 a standard against Intensification (Consolidation) Therapy
which other trial results are now measured. Because With restoration of normal hematopoiesis, pa-
of their poor prognosis, infants with ALL are often tients whose disease is in remission become candi-
considered a unique subgroup and treated with mul- dates for intensification (consolidation) therapy. Such
tiple drugs given at high dosages, but no cranial ir- treatment, administered shortly after the induction
radiation.26 of remission, includes several drugs, most often meth-
For all other patients, the basic approach to ther- otrexate given in high doses with or without 6-mer-
apy consists of a relatively brief remission-induction captopurine18,51,55,57; asparaginase given in high doses
phase, followed by intensification (consolidation) for an extended period17,52; an epipodophyllotoxin
treatment and then prolonged continuation therapy. plus cytarabine51,58; or a combination of vincristine,
Treatment for subclinical leukemia of the central dexamethasone, asparaginase, doxorubicin, and thio-
nervous system is initiated early and is given for guanine given with or without cyclophosphamide.18,53
varying lengths of time, depending on the treatment This phase of therapy has been credited with im-
protocol. proving outcomes, even in patients with low-risk
ALL.58 Very high doses of methotrexate (5 g per
Induction of Remission square meter) appear to improve the outcome in pa-
The first goal of therapy in patients with leukemia tients with T-cell ALL.18 This conclusion is con-
is to induce complete remission with restoration of sistent with data indicating that T-lineage blast cells
normal hematopoiesis. The induction regimen in- accumulate methotrexate polyglutamates (active me-
variably includes a glucocorticoid (prednisone or tabolites of the parent compound) less avidly than do
dexamethasone) and vincristine, as well as asparagin- B-lineage blast cells,75 so that higher serum concen-
ase for children or an anthracycline for adults.2,3,7,16 trations of methotrexate are needed for adequate ac-
With improvements in chemotherapy and support- cumulation of methotrexate polyglutamates.76 Indeed,
ive care, the rate of complete remission now ranges the conventional dose of methotrexate of 1 g per
from 97 to 99 percent in children and from 75 to square meter may be too low for many patients with
90 percent in adults. Nonetheless, attempts are be- B-cell–precursor ALL.60
ing made to intensify induction therapy, especially The value of intensification treatment is less cer-
for patients with standard-risk or high-risk disease, tain in adults with ALL because randomized trials
on the premise that more rapid and complete reduc- have not been performed and because some studies
tion of the leukemic-cell burden may forestall drug have used suboptimal continuation regimens.2,3 How-
resistance in leukemic cells, leading to improvements ever, the results of several nonrandomized studies
in long-term outcome. Indeed, intensive induction have suggested that intensive multidrug therapy be-
regimens with four or more drugs have been credit- gun after induction of remission results in longer-
ed with improving outcomes in several trials in chil- lasting remissions, especially in young adults; that
dren.18,51-53 Efforts to intensify induction therapy in patients with T-cell ALL benefit from cyclophospha-
adults have been limited by severe drug toxicity2,3,7; mide and cytarabine therapy; and that high-dose cy-
however, in one study, high-dose cytarabine may tarabine therapy improves outcome in patients with
have preferentially improved the outcome in adults standard-risk or high-risk ALL.2,12,56,59
with T-cell ALL.56 Administration of granulocyte
colony-stimulating factor may hasten recovery from Continuation Treatment
neutropenia and reduce the complications of inten- With the exception of those with mature B-cell
sive chemotherapy, but it does not improve the event- leukemia, children with ALL require prolonged con-
free survival rate in either children or adults.68-70 tinuation treatment for reasons that are poorly un-
Perhaps because of its increased penetration into derstood. Perhaps long-term drug exposure or the
cerebrospinal fluid and its longer half-life,71 dexa- host immune system is needed to kill residual, slowly
methasone, when used in induction and continua- dividing leukemic cells or to suppress their growth,
tion regimens, provides better protection than pred- allowing programmed cell death to occur.16 In one

610 · Augus t 2 7 , 19 9 8

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
D RUG TH ERA PY

trial, attempts to shorten the duration of moderately four months). This treatment, which relies on the
intensive chemotherapy to 18 months or less result- same drugs that were given during the first induction
ed in a high rate of relapse after the cessation of phase, has improved results in children and in adults
treatment; however, according to a meta-analysis of with ALL.18,59,62 Prolonged intensification including
42 studies, there is no advantage to prolonging a second reinduction phase or rotational administra-
treatment beyond three years.62 Hence, the general tion of non–cross-resistant drug pairs during contin-
rule is to discontinue all therapy in children whose uation treatment may further improve outcome in
disease remains in remission two and a half to three patients with standard-risk or high-risk ALL.64,84
years after therapy is begun. It remains unclear
whether the duration of therapy can be shortened Therapy Directed toward the Central Nervous System
for patients receiving intensified chemotherapy. It is Realization that the central nervous system can be
also uncertain whether adults with ALL require pro- a sanctuary for leukemic cells has prompted the devel-
longed continuation therapy. In two trials of the ef- opment of presymptomatic therapy directed toward
ficacy of post-remission treatment given for 5 to 10 the central nervous system in patients with ALL. Be-
months, the median durations of remission ranged cause of concern that cranial irradiation can involve
from 9 to 12 months.73,77 These poor results may re- substantial neurotoxicity and can occasionally cause
flect inadequate treatment for induction of remission brain tumors, many therapists instead administer in-
or inadequate consolidation treatment. tensive intrathecal or systemic chemotherapy early in
A combination of methotrexate administered week- the treatment course. The results of chemotherapy
ly and mercaptopurine administered daily constitutes are excellent: rates of relapse of central nervous sys-
the usual continuation regimen for children with tem disease of 2 percent or less have been achieved
ALL. Accumulation of higher intracellular concen- in several studies.43,55,65-67
trations of the active metabolites of methotrexate Whether certain groups of patients at high risk
and mercaptopurine and administration of this com- should be treated with cranial irradiation is unclear.
bination to the limits of tolerance (as indicated by In one study, among children with T-cell ALL and
low leukocyte counts) have been associated with an a leukocyte count of at least 100,000 per cubic mil-
improved clinical outcome.78-80 limeter, those receiving intensive intrathecal therapy
A few children (1 in 300) have an inherited defi- alone had an outcome that was inferior to that of
ciency of thiopurine S-methyltransferase, the en- children given cranial irradiation,43 but the chemo-
zyme that catalyzes the S-methylation (inactivation) therapy in the two groups was not identical. None-
of mercaptopurine. In these children the standard theless, in the context of effective systemic chemo-
doses of mercaptopurine have potentially fatal hema- therapy, a radiation dose of 12 Gy, rather than the
tologic side effects, but the drug can be given safely conventional dose of 18 Gy, appears to provide ade-
in much smaller doses.81 Furthermore, about 10 per- quate protection against central nervous system leu-
cent of children are heterozygous for this deficiency kemia, even in patients at high risk.43
and thus have intermediate levels of enzyme activity;
they may require moderate dose reduction to avert Allogeneic Stem-Cell Transplantation
side effects. Recent identification of the genetic basis Transplantation of allogeneic stem cells is usually
of this autosomal codominant trait has made possi- indicated for patients who do not have a response to
ble the molecular diagnosis of these cases.82 In addi- the initial induction treatment and those who have
tion, because mercaptopurine has an apparent circa- a second remission after a hematologic relapse.85
dian effect, treatment outcome improves when the However, for children with late relapses after anti-
drug is given in the evening.83 With methotrexate, metabolite treatment (more than 36 months after
the advantages of oral as compared with parenteral induction), transplantation may be deferred until a
administration are uncertain, but the parenteral subsequent relapse, because these patients have a
route circumvents problems of decreased bioavail- reasonable chance of cure with retrieval chemother-
ability and poor compliance. Prolonging exposure apy alone.86,87 Transplantation during the first remis-
to methotrexate by giving it orally in divided doses sion remains controversial. However, because of their
over a 36-hour period has proved to be less effective unfavorable prognosis, patients with the BCR–ABL
than giving a higher-dose intravenous infusion over or MLL–AF4 fusion gene are commonly treated
a 24-hour period.57 with allogeneic stem-cell transplantation during the
The addition of intermittent pulses of vincristine first remission.7,39,40,85,88
and a glucocorticoid to the antimetabolite continu- For patients who require allogeneic stem-cell
ation regimen has been shown to improve results62 transplantation but lack suitable family donors, al-
and has been widely adopted in the treatment of child- lografts from matched unrelated donors are a rea-
hood ALL. Another integral component of many sonable option and have yielded encouraging re-
protocols is reinduction therapy introduced early af- sults.89,90 In one study, transplantation of grafts from
ter the first induction of remission (for example, at unrelated donors was as effective as transplantation

Vol ume 33 9 Numb e r 9 · 611

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
The Ne w E ng l a nd Jo u r na l o f Me d ic i ne

of matched-sibling–donor grafts.90 Transplantation covorin rescue is inadequate.92,93 Although thought


of umbilical-cord blood or autologous bone marrow to have few life-threatening side effects, prednisone
has also been attempted.7,85,91 Preliminary results in- was implicated as a central nervous system toxin in
dicate that transplantation of cord-blood stem cells one recent study.94
is feasible, especially in children; an advantage of this Acute myeloid leukemia may develop in some pa-
approach is that it does not require the same degree tients who have received intensive treatment with
of histocompatibility as transplantation of hemato- topoisomerase II inhibitors, primarily etoposide and
poietic stem cells from adults.91 Whether the lower teniposide. The latency period is relatively short (me-
risk of graft-versus-host disease might be offset by a dian, three years), and the risk appears to be related
diminished graft-versus-leukemia effect and hence a to the treatment schedule and concomitant use of
higher risk of relapse remains to be determined. other drugs.95,96 The long-term survival rate of pa-
tients with this complication is very low, even with
LATE SEQUELAE allogeneic stem-cell transplantation.96 Treatment with
Each year, at least 1500 children in the United anthracyclines at high cumulative doses and rapid
States become long-term survivors of ALL, a group rates of administration can result in cardiomyopathy,
at risk for the late adverse effects of treatment (Table especially in young girls.97 Studies are under way to
3). Cranial irradiation has been implicated in the de- determine whether cardioprotective agents such as
velopment of brain tumors (with a median latency dexrazoxane can reduce the frequency of this com-
of 10 years), neuropsychological deficits, and endo- plication without loss of the therapeutic benefits of
crinopathy leading to short stature, obesity, preco- anthracyclines. There have been no indications that
cious puberty, and osteoporosis.4 Many children the incidence of cancer or birth defects is increased
with profound growth retardation due to cranial ir- among the offspring of adult survivors of childhood
radiation or intensive chemotherapy receive growth ALL.98-100
hormone therapy after completing antileukemic
therapy. An adequate final height can be attained in FUTURE CONSIDERATIONS
most cases with growth hormone therapy, which Despite the high rate of cure of childhood ALL,
does not appear to increase the risk of leukemic re- resistant forms of the disease still represent a leading
lapse (unpublished data). However, the long-term cause of cancer-related deaths in children. Efforts are
consequences of growth hormone deficiency in being made to identify new antileukemic drugs and
these patients and the extent to which these effects new approaches to therapy. Arabinosylguanine has
can be reversed by continued replacement therapy shown considerable promise, inducing complete re-
remain to be determined. missions in 44 percent of adults and children with
Encephalopathy, with or without seizures, can be refractory T-cell ALL in one small series.101 Recent
induced by methotrexate, usually after intravenous insights into the mechanisms by which tumor-specif-
administration of high doses but in some cases after ic cytolytic cells are produced have opened the way
oral administration of low doses (e.g., 25 mg per for the development of targeted immunotherapy.102
square meter every six hours for four doses), if leu- Several strategies for gene therapy have also been
developed and include the use of antisense oligonu-
cleotides and ribozymes to disrupt oncogene expres-
sion and the use of genes encoding cytokines and
human leukocyte antigens to induce an immune re-
TABLE 3. LATE COMPLICATIONS OF INTENSIVE CHEMOTHERAPY sponse against leukemic cells.103 In another poten-
FOR ALL. tially useful approach, small cell-permeable mole-
cules may be designed to inhibit the transcription of
COMPLICATION ASSOCIATED TREATMENT specific genes.104 The recent finding that ALL is
angiogenic suggests that antiangiogenic drugs also
Brain tumor Cranial irradiation
Acute myeloid leukemia Epipodophyllotoxins, alkylating
may have a therapeutic role.105
agents, anthracyclines These and related therapies may soon offer treat-
Cardiomyopathy Anthracyclines ment options that are more specific and hence less
Encephalopathy Cranial irradiation, methotrexate, toxic than conventional chemotherapy. However,
glucocorticoids
many remarkable advances in curative therapy for
Avascular necrosis of bone Glucocorticoids, local irradiation
childhood ALL (Fig. 1) have been achieved in the
Osteoporosis Cranial irradiation, glucocorticoids,
antimetabolites past 20 years through more effective use of antileu-
Short stature Cranial irradiation, glucocorticoids, kemic drugs that first became available decades ago.
intensive chemotherapy Indeed, it may be possible to improve existing ther-
Obesity Cranial irradiation apeutic regimens further by adjusting dosages on the
Thyroid dysfunction Cranial (and neck) irradiation, inten-
sive chemotherapy
basis of pharmacokinetic characteristics in individual
patients.61 Thus, further modifications of contempo-

612 · Augus t 2 7 , 19 9 8

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
D RUG TH ERA PY

rary treatment protocols, coupled with systematic of age at diagnosis entered on Childrens Cancer Group acute lymphoblastic
leukemia and acute myeloblastic leukemia protocols: results of treatment.
evaluation of novel biologic strategies, may provide Cancer 1993;71:Suppl:3377-85.
uniformly effective therapy for children with ALL 24. Pui C-H, Boyett JM, Hancock ML, Pratt CB, Meyer WH, Crist WM.
and begin to improve the generally unfavorable out- Outcome of treatment for childhood cancer in black as compared with
white children: the St. Jude Children’s Research Hospital experience, 1962
look for adults with this disease. through 1992. JAMA 1995;273:633-7.
25. Smith M, Arthur D, Camitta B, et al. Uniform approach to risk clas-
sification and treatment assignment for children with acute lymphoblastic
Supported in part by grants (PO1 CA-20180, R37 CA36401, RO1 CA- leukemia. J Clin Oncol 1996;14:18-24.
78224, and P30 CA-21765) from the National Cancer Institute, by a 26. Pui C-H, Kane JR, Crist WM. Biology and treatment of infant leuke-
Center of Excellence grant from the state of Tennessee, and by the Amer- mias. Leukemia 1995;9:762-9.
ican Lebanese Syrian Associated Charities. 27. Rubnitz JE, Link MP, Shuster JJ, et al. Frequency and prognostic sig-
nificance of HRX rearrangements in infant acute lymphoblastic leukemia:
We are indebted to Dieter F. Hoelzer, M.D., Hagop Kantarjian, a Pediatric Oncology Group study. Blood 1994;84:570-3.
28. Behm FG, Raimondi SC, Frestedt JL, et al. Rearrangement of the
M.D., and Richard A. Larson, M.D., for helpful comments regard- MLL gene confers a poor prognosis in childhood acute lymphoblastic leu-
ing the manuscript, and to J.R. Gilbert for medical editing. kemia, regardless of presenting age. Blood 1996;87:2870-7.
29. Rubnitz JE, Downing JR, Pui C-H, et al. TEL gene rearrangement in
REFERENCES acute lymphoblastic leukemia: a new genetic marker with prognostic sig-
nificance. J Clin Oncol 1997;15:1150-7.
1. Parker SL, Tong T, Bolden S, Wingo PA. Cancer statistics, 1997. CA 30. Borkhardt A, Cazzaniga G, Viehmann S, et al. Incidence and clinical
Cancer J Clin 1997;47:5-27. [Erratum, CA Cancer J Clin 1997;47:68.] relevance of TEL/AML1 fusion genes in children with acute lymphoblastic
2. Cortes JE, Kantarjian HM. Acute lymphoblastic leukemia: a comprehen- leukemia enrolled in the German and Italian multicenter therapy trials.
sive review with emphasis on biology and therapy. Cancer 1995;76:2393-417. Blood 1997;90:571-7.
3. Laport GF, Larson RA. Treatment of adult acute lymphoblastic leuke- 31. Gajjar A, Ribeiro R, Hancock ML, et al. Persistence of circulating
mia. Semin Oncol 1997;24:70-82. blasts after 1 week of multiagent chemotherapy confers a poor prognosis
4. Pui C-H. Acute lymphoblastic leukemia. Pediatr Clin North Am 1997; in childhood acute lymphoblastic leukemia. Blood 1995;86:1292-5.
44:831-46. 32. Gaynon PS, Desai AA, Bostrom BC, et al. Early response to therapy
5. Groupe Français de Cytogénétique Hématologique. Cytogenetic abnor- and outcome in childhood acute lymphoblastic leukemia: a review. Cancer
malities in adult acute lymphoblastic leukemia: correlations with hemato- 1997;80:1717-26.
logic findings and outcome: a collaborative study of the Groupe Français 33. Coustan-Smith E, Behm FG, Sanchez J, et al. Immunological detec-
de Cytogénétique Hématologique. Blood 1996;87:3135-42. [Erratum, tion of minimal residual disease in children with acute lymphoblastic leu-
Blood 1996;88:2818.] kaemia. Lancet 1998;351:550-4.
6. Chessells JM, Hall E, Prentice HG, Durrant J, Bailey CC, Richards SM. 34. Cavé H, van der Werff ten Bosch J, Suciu S, et al. Clinical significance
The impact of age on outcome in lymphoblastic leukaemia: MRC UKALL of minimal residual disease in childhood acute lymphoblastic leukemia.
X and XA compared: a report from the MRC Paediatric and Adult Working N Engl J Med 1998;339:591-8.
Parties. Leukemia 1998;12:463-73. 35. Hunger SP, Fall MZ, Camitta BM, et al. E2A-PBX1 chimeric tran-
7. Copelan EA, McGuire EA. The biology and treatment of acute lympho- script status at end of consolidation is not predictive of treatment outcome
blastic leukemia in adults. Blood 1995;85:1151-68. in childhood acute lymphoblastic leukemias with a t(1;19)(q23;p13): a Pe-
8. Pui C-H, Behm FG, Crist WM. Clinical and biologic relevance of im- diatric Oncology Group study. Blood 1998;91:1021-8.
munologic marker studies in childhood acute lymphoblastic leukemia. 36. Kaspers GJL, Veerman AJP, Pieters R, et al. In vitro cellular drug re-
Blood 1993;82:343-62. sistance and prognosis in newly diagnosed childhood acute lymphoblastic
9. Pui C-H. Childhood leukemias. N Engl J Med 1995;332:1618-30. leukemia. Blood 1997;90:2723-9.
10. Kishimoto T, Goyert S, Kikutani H, et al. CD antigens 1996. Blood 37. Uckun FM. Severe combined immunodeficient mouse models of hu-
1997;89:3502. man leukemia. Blood 1996;88:1135-46.
11. Pui C-H, Raimondi SC, Head DR, et al. Characteristics of childhood 38. Kumagai M, Manabe A, Pui C-H, et al. Stroma-supported culture in
acute leukemia with multiple myeloid and lymphoid markers at diagnosis childhood B-lineage acute lymphoblastic leukemia cells predicts treatment
and at relapse. Blood 1991;78:1327-37. outcome. J Clin Invest 1996;97:755-60.
12. Larson RA, Dodge RK, Burns CP, et al. A five-drug remission induction 39. Ribeiro RC, Broniscer A, Rivera GK, et al. Philadelphia chromosome-
regimen with intensive consolidation for adults with acute lymphoblastic leu- positive acute lymphoblastic leukemia in children: durable responses to
kemia: Cancer and Leukemia Group B Study 8811. Blood 1995;85:2025-37. chemotherapy associated with low initial white blood cell counts. Leuke-
13. Uckun FM, Sather HN, Gaynon PS, et al. Clinical features and treatment mia 1997;11:1493-6.
outcome of children with myeloid antigen positive acute lymphoblastic leu- 40. Schrappe M, Aricò M, Harbott J, et al. Ph+ childhood acute lympho-
kemia: a report from the Children’s Cancer Group. Blood 1997;90:28-35. blastic leukemia: good initial steroid response allows early prediction of a
14. Campana D, Pui C-H. Detection of minimal residual disease in acute favorable treatment outcome. Blood (in press).
leukemia: methodologic advances and clinical significance. Blood 1995;85: 41. Pui C-H, Carroll AJ, Raimondi SC, et al. Clinical presentation, kary-
1416-34. otypic characterization, and treatment outcome of childhood acute lym-
15. Look AT. Oncogenic transcription factors in the human acute leuke- phoblastic leukemia with a near-haploid or hypodiploid <45 line. Blood
mias. Science 1997;278:1059-64. 1990;75:1170-7.
16. Pui C-H, Crist WM. Biology and treatment of acute lymphoblastic 42. Pui C-H, Frankel LS, Carroll AJ, et al. Clinical characteristics and
leukemia. J Pediatr 1994;124:491-503. treatment outcome of childhood acute lymphoblastic leukemia with the
17. Schorin MA, Blattner S, Gelber RD, et al. Treatment of childhood t(4;11)(q21;q23): a collaborative study of 40 cases. Blood 1991;77:440-7.
acute lymphoblastic leukemia: results of Dana-Farber Cancer Institute/ 43. Conter V, Schrappe M, Aricó M, et al. Role of cranial radiotherapy for
Children’s Hospital Acute Lymphoblastic Leukemia Consortium Protocol childhood T-cell acute lymphoblastic leukemia with high WBC count and
85-01. J Clin Oncol 1994;12:740-7. good response to prednisone. J Clin Oncol 1997;15:2786-91.
18. Reiter A, Schrappe M, Ludwig W-D, et al. Chemotherapy in 998 un- 44. Mahmoud HH, Rivera GK, Hancock ML, et al. Low leukocyte
selected childhood acute lymphoblastic leukemia patients: results and con- counts with blast cells in cerebrospinal fluid of children with newly diag-
clusions of the multicenter trial ALL-BFM 86. Blood 1994;84:3122-33. nosed acute lymphoblastic leukemia. N Engl J Med 1993;329:314-9.
19. Soussain C, Patte C, Ostronoff M, et al. Small noncleaved cell lym- 45. Dinarello CA, Bunn PA Jr. Fever. Semin Oncol 1997;24:288-98.
phoma and leukemia in adults: a retrospective study of 65 adults treated 46. Hughes WT, Rivera GK, Schell MJ, Thornton D, Lott L. Successful
with the LMB pediatric protocols. Blood 1995;85:664-74. intermittent chemoprophylaxis for Pneumocystis carinii pneumonitis.
20. Hoelzer D, Ludwig W-D, Thiel E, et al. Improved outcome in adult N Engl J Med 1987;316:1627-32.
B-cell acute lymphoblastic leukemia. Blood 1996;87:495-508. 47. Masson E, Synold TW, Relling MV, et al. Allopurinol inhibits de novo
21. Reiter A, Schrappe M, Ludwig W-D, et al. Favorable outcome of purine synthesis in lymphoblasts of children with acute lymphoblastic
B-cell acute lymphoblastic leukemia in childhood: a report of three con- leukemia. Leukemia 1996;10:56-60.
secutive studies of the BFM group. Blood 1992;80:2471-8. 48. Pui C-H, Relling MV, Lascombes F, et al. Urate oxidase in prevention
22. Patte C. Non-Hodgkin’s lymphoma. Eur J Cancer 1998;34:359-63. and treatment of hyperuricemia associated with lymphoid malignancies.
23. Nachman J, Sather HN, Buckley JD, et al. Young adults 16-21 years Leukemia 1997;11:1813-6.

Vol ume 33 9 Numb e r 9 · 613

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
The Ne w E ng l a nd Jo u r na l o f Me d ic i ne

49. Bunin NJ, Pui C-H. Differing complications of hyperleukocytosis in ferences in cerebrospinal fluid penetration of corticosteroids: possible relation-
children with acute lymphoblastic or acute nonlymphoblastic leukemia. ship to the prevention of meningeal leukemia. J Clin Oncol 1987;5:202-7.
J Clin Oncol 1985;3:1590-5. 72. Otten J, Suciu S, Lutz P, et al. The importance of L-asparaginase
50. Nelson SC, Bruggers CS, Kurtzberg J, Friedman HS. Management of (A’ase) in the treatment of acute lymphoblastic leukemia (ALL) in children:
leukemic hyperleukocytosis with hydration, urinary alkalinization, and al- results of the EORTC 58881 randomized phase III trial showing greater
lopurinol: are cranial irradiation and invasive cytoreduction necessary? Am efficiency of Escherichia coli (E. coli) as compared to Erwinia (Erw) A’ase.
J Pediatr Hematol Oncol 1993;15:351-5. Blood 1996;88:Suppl 1:669a. abstract.
51. Rivera GK, Raimondi SC, Hancock ML, et al. Improved outcome in 73. Cuttner J, Mick R, Budman DR, et al. Phase III trial of brief intensive
childhood acute lymphoblastic leukemia with reinforced early treatment treatment of adult acute lymphoblastic leukemia comparing daunorubicin
and rotational combination chemotherapy. Lancet 1991;337:61-6. and mitoxantrone: a CALGB study. Leukemia 1991;5:425-31.
52. Niemeyer CM, Gelber RD, Tarbell NJ, et al. Low-dose versus high- 74. Fière D, Lepage E, Sebban C, et al. Adult acute lymphoblastic leuke-
dose methotrexate during remission induction in childhood acute lympho- mia: a multicentric randomized trial testing bone marrow transplantation
blastic leukemia (Protocol 81-01 update). Blood 1991;78:2514-9. as postremission therapy. J Clin Oncol 1993;11:1990-2001.
53. Gaynon PS, Steinherz PG, Bleyer WA, et al. Improved therapy for chil- 75. Synold TW, Relling MV, Boyett JM, et al. Blast cell methotrexate-poly-
dren with acute lymphoblastic leukemia and unfavorable presenting fea- glutamate accumulation in vivo differs by lineage, ploidy, and methotrexate
tures: a follow-up report of the Childrens Cancer Group Study CCG-106. dose in acute lymphoblastic leukemia. J Clin Invest 1994;94:1996-2001.
J Clin Oncol 1993;11:2234-42. 76. Galpin AJ, Schuetz JD, Masson E, et al. Differences in folylpoly-
54. Bostrom B, Gaynon PS, Sather H, et al. Dexamethasone (DEX) de- glutamate synthetase and dihydrofolate reductase expression in human
creases central nervous system (CNS) relapse and improves event-free sur- B-lineage versus T-lineage leukemic lymphoblasts: mechanisms for lineage
vival (EFS) in lower risk acute lymphoblastic leukemia (ALL). Proc Am Soc differences in methotrexate polyglutamylation and cytotoxicity. Mol Phar-
Clin Oncol 1998;17:527A. macol 1997;52:155-63.
55. Veerman AJP, Hählen K, Kamps WA, et al. High cure rate with a 77. Cassileth PA, Andersen JW, Bennett JM, et al. Adult acute lymphocyt-
moderately intensive treatment regimen in non-high-risk childhood acute ic leukemia: the Eastern Cooperative Oncology Group experience. Leuke-
lymphoblastic leukemia: results of protocol ALL VI from the Dutch Child- mia 1992;6:Suppl 2:178-81.
hood Leukemia Study Group. J Clin Oncol 1996;14:911-8. 78. Whitehead VM, Vuchich MJ, Lauer SJ, et al. Accumulation of high
56. Rohatiner AZ, Bassan R, Battista R, et al. High dose cytosine arabi- levels of methotrexate polyglutamates in lymphoblasts from children with
noside in the initial treatment of adults with acute lymphoblastic leu- hyperdiploid (>50 chromosomes) B-lineage acute lymphoblastic leukemia:
kaemia. Br J Cancer 1990;62:454-8. a Pediatric Oncology Group study. Blood 1992;80:1316-23.
57. Mahoney DH Jr, Shuster JJ, Nitschke R, et al. Intermediate-dose in- 79. Lennard L, Lilleyman JS, Van Loon J, Weinshilboum RM. Genetic
travenous methotrexate with intravenous mercaptopurine is superior to re- variation in response to 6-mercaptopurine for childhood acute lymphoblas-
petitive low-dose oral methotrexate with intravenous mercaptopurine for tic leukaemia. Lancet 1990;336:225-9.
children with lower-risk B-lineage acute lymphoblastic leukemia: a Pediat- 80. Chessells JM, Harrison G, Lilleyman JS, Bailey CC, Richards SM.
ric Oncology Group phase III trial. J Clin Oncol 1998;16:246-54. Continuing (maintenance) therapy in lymphoblastic leukaemia: lessons
58. Chessells JM, Bailey C, Richards SM. Intensification of treatment and from MRC UKALL X. Br J Haematol 1997;98:945-51.
survival in all children with lymphoblastic leukaemia: results of UK Medical 81. Evans WE, Horner M, Chu YQ, Kalwinsky D, Roberts WM. Altered
Research Council trial UKALL X. Lancet 1995;345:143-8. mercaptopurine metabolism, toxic effects, and dosage requirement in a thi-
59. Hoelzer D, Thiel E, Ludwig WD, et al. The German multicentre trials opurine methyltransferase-deficient child with acute lymphocytic leukemia.
for treatment of acute lymphoblastic leukemia in adults: the German Adult J Pediatr 1991;119:985-9.
ALL Study Group. Leukemia 1992;6:Suppl 2:175-7. 82. Yates CR, Krynetski EY, Loennechen T, et al. Molecular diagnosis of
60. Evans WE, Crom WR, Abromowitch M, et al. Clinical pharmacody- thiopurine S-methyltransferase deficiency: genetic basis for azathioprine
namics of high-dose methotrexate in acute lymphoblastic leukemia: identi- and mercaptopurine intolerance. Ann Intern Med 1997;126:608-14.
fication of a relation between concentration and effect. N Engl J Med 83. Schmiegelow K, Glomstein A, Kristinsson J, Salmi T, Schrøder H,
1986;314:471-7. Björk O. Impact of morning versus evening schedule for oral methotrexate
61. Evans WE, Relling MV, Rodman JH, Crom WR, Boyett JM, Pui C-H. and 6-mercaptopurine on relapse risk for children with acute lymphoblastic
Conventional compared with individualized chemotherapy for childhood leukemia. J Pediatr Hematol Oncol 1997;19:102-9.
acute lymphoblastic leukemia. N Engl J Med 1998;338:499-505. 84. Sackmann-Muriel F, Felice MS, Zubizarreta P, Chantada G, Barbieri
62. Childhood ALL Collaborative Group. Duration and intensity of main- MAF, Rossi J. Improved outcome in higher-risk acute lymphoblastic leuke-
tenance chemotherapy in acute lymphoblastic leukaemia: overview of 42 mia with a hybrid (Berlin-Frankfurt-Münster/St. Jude’s) therapy. Int J Pe-
trials involving 12 000 randomised children. Lancet 1996;347:1783-8. diatr Hematol Oncol 1996;3:439-49.
63. Tubergen DG, Gilchrist GS, O’Brien RT, et al. Improved outcome 85. Appelbaum FR. Allogeneic hematopoietic stem cell transplantation for
with delayed intensification for children with acute lymphoblastic leukemia acute leukemia. Semin Oncol 1997;24:114-23.
and intermediate presenting features: a Childrens Cancer Group phase III 86. Barrett AJ, Horowitz MM, Pollock BH, et al. Bone marrow trans-
trial. J Clin Oncol 1993;11:527-37. plants from HLA-identical siblings as compared with chemotherapy for
64. Nachman JB, Sather HN, Sensel MG, et al. Augmented post-induc- children with acute lymphoblastic leukemia in a second remission. N Engl
tion therapy for children with high-risk acute lymphoblastic leukemia and J Med 1994;331:1253-8.
a slow response to initial therapy. N Engl J Med 1998;338:1663-71. 87. Uderzo C, Valsecchi MG, Bacigalupo A, et al. Treatment of childhood
65. Cortes J, O’Brien SM, Pierce S, Keating MJ, Freireich EJ, Kantarjian acute lymphoblastic leukemia in second remission with allogeneic bone
HM. The value of high-dose systemic chemotherapy and intrathecal ther- marrow transplantation and chemotherapy: ten-year experience of the Ital-
apy for central nervous system prophylaxis in different risk groups of adult ian Bone Marrow Transplantation Group and the Italian Pediatric Hema-
acute lymphoblastic leukemia. Blood 1995;86:2091-7. tology Oncology Association. J Clin Oncol 1995;13:352-8.
66. Nachman J, Sather HN, Cherlow JM, et al. Response of children with 88. Sebban C, Lepage E, Vernant J-P, et al. Allogeneic bone marrow
high-risk acute lymphoblastic leukemia treated with and without cranial ir- transplantation in adult acute lymphoblastic leukemia in first complete re-
radiation: a report from the Children’s Cancer Group. J Clin Oncol 1998; mission: a comparative study. J Clin Oncol 1994;12:2580-7.
16:920-30. 89. Sierra J, Radich J, Hansen JA, et al. Marrow transplants from unrelated
67. Pui C-H, Mahmoud HH, Rivera GK, et al. Early intensification of donors for treatment of Philadelphia chromosome-positive acute lympho-
intrathecal chemotherapy virtually eliminates central nervous system re- blastic leukemia. Blood 1997;90:1410-4.
lapse in children with acute lymphoblastic leukemia. Blood 1998;92:411- 90. Hongeng S, Krance RA, Bowman LC, et al. Outcomes of transplan-
5. tation with matched-sibling and unrelated-donor bone marrow in children
68. Ottmann OG, Hoelzer D, Gracien E, et al. Concomitant granulocyte with leukaemia. Lancet 1997;350:767-71.
colony-stimulating factor and induction chemoradiotherapy in adult acute 91. Gluckman E, Rocha V, Boyer-Chammard A, et al. Outcome of cord-
lymphoblastic leukemia: a randomized phase III trial. Blood 1995;86:444- blood transplantation from related and unrelated donors. N Engl J Med
50. 1997;337:373-81.
69. Pui C-H, Boyett JM, Hughes WT, et al. Human granulocyte colony- 92. Winick NJ, Bowman WP, Kamen BA, et al. Unexpected acute neuro-
stimulating factor after induction chemotherapy in children with acute logic toxicity in the treatment of children with acute lymphoblastic leuke-
lymphoblastic leukemia. N Engl J Med 1997;336:1781-7. mia. J Natl Cancer Inst 1992;84:252-6.
70. Geissler K, Koller E, Hubmann E, et al. Granulocyte colony-stimulating 93. Mahoney DH Jr, Shuster JJ, Nitschke R, et al. Acute neurotoxicity
factor as an adjunct to induction chemotherapy for adult acute lymphoblas- in children with B-precursor acute lymphoid leukemia: an association with
tic leukemia — a randomized phase-III study. Blood 1997;90:590-6. intermediate-dose intravenous methotrexate and intrathecal triple therapy
71. Balis FM, Lester CM, Chrousos GP, Heideman RL, Poplack DG. Dif- — a Pediatric Oncology Group study. J Clin Oncol 1998;16:1712-22.

614 · Augus t 2 7 , 19 9 8

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.
DRUG THERAPY

94. Waber DP, Tarbell NJ, Fairclough D, et al. Cognitive sequelae of treat- 100. Sankila R, Olsen JH, Anderson H, et al. Risk of cancer among
ment in childhood acute lymphoblastic leukemia: cranial radiation requires offspring of childhood-cancer survivors. N Engl J Med 1998;338:1339-
an accomplice. J Clin Oncol 1995;13:2490-6. 44.
95. Pui C-H, Ribeiro RC, Hancock ML, et al. Acute myeloid leukemia in 101. Kurtzberg J, Keating M, Moore JO, et al. 2-Amino-9-B-D-arabinosyl-
children treated with epipodophyllotoxins for acute lymphoblastic leukemia. 6-methoxy-9H-guanine (GW 506U; compound 506U) is highly active in
N Engl J Med 1991;325:1682-7. patients with T-cell malignancies: results of a phase I trial in pediatric and
96. Pui C-H, Relling MV, Rivera GK, et al. Epipodophyllotoxin-related adult patients with refractory hematological malignancies. Blood 1996;88:
acute myeloid leukemia: a study of 35 cases. Leukemia 1995;9:1990-6. Suppl 1:669a. abstract.
97. Lipshultz SE, Lipsitz SR, Mone SM, et al. Female sex and higher drug 102. Hart I, Colaco C. Fusion induces tumour rejection. Nature 1997;
dose as risk factors for late cardiotoxic effects of doxorubicin therapy for 388:626-7.
childhood cancer. N Engl J Med 1995;332:1738-43. 103. Braun SE, Chen K, Battiwalla M, Cornetta K. Gene therapy strate-
98. Hawkins MM, Draper GJ, Winter DJ. Cancer in the offspring of sur- gies for leukemia. Mol Med Today 1997;3:39-46.
vivors of childhood leukaemia and non-Hodgkin lymphomas. Br J Cancer 104. Gottesfeld JM, Neely L, Trauger JW, Baird EE, Dervan PB. Regula-
1995;71:1335-9. tion of gene expression by small molecules. Nature 1997;387:202-5.
99. Kenney LB, Nicholson HS, Brasseux C, et al. Birth defects in offspring 105. Perez-Atayde AR, Sallan SE, Tedrow U, Connors S, Allred E, Folk-
of adult survivors of childhood acute lymphoblastic leukemia: a Childrens man J. Spectrum of tumor angiogenesis in the bone marrow of children
Cancer Group/National Institutes of Health report. Cancer 1996;78:169-76. with acute lymphoblastic leukemia. Am J Pathol 1997;150:815-21.

Vo l u m e 3 3 9 Nu m b e r 9 · 615

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF SOUTH CAROLINA on July 8, 2013. For personal use only. No other uses without permission.
Copyright © 1998 Massachusetts Medical Society. All rights reserved.

Anda mungkin juga menyukai