Case Study

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Acute leukemia is the most common childhood malignancy, representing 30% of all

cancer in American children under the age of 15 years and 12% of cancer cases in those
ages 15 to 19 years old. In the United States, approximately 2500 new cases are
diagnosed annually; 80% of these are acute lymphoblastic leukemia, 15% are acute
myelogenous leukemia, and 5% belong to the chronic leukemia category. 1 The survival
rates of children with acute leukemia have increased dramatically in the last 40 years. 2-5
The most success in outcome has occurred in acute lymphoblastic leukemia, although
improvement is also being reported in acute myelogenous leukemia in the past f e w years.
Progress comes from treatment strategy modifications on the basis of observations made
in sequential large-scale therapeutic trials, an approach tha't serves as a paradigm for
research in other malignant diseases.

.11.

Acute Lymphoblastic Leukemia

Ka Wah Chan, MD

he incidence of childhood acute lymphoblastic liferation and infiltration of the leukemia cells (lymph-
leukemia (ALL) in the United States is approx- adenopathy, hepatosplenomegaly, and bone pain).
imately 3.4 cases per 100,000 children younger These features are nonspecific in nature, and none is
than 15 years of age, with the peak incidence occur- found in more than two thirds of the patients. How-
ring between 2 and 5 years of age. It is more common ever, the constellation of abnormal findings should
in males and in whites. The incidence of ALL and of raise the suspicion and prompt the ordering of labora-
the leukemia of infancy, but not other types of leuke- tory testing. Ultimately a marrow aspirate is required
mia in childhood, increased in the past two decades, to establish a definite diagnosis.
a change that appears to affect white children only. Because a broad spectrum of peripheral blood ab-
Other than prenatal exposure to ionizing radiation and normalities can be found at diagnosis, some common
specific genetic syndromes such as Down syndrome pitfalls should be avoided. For example, not all pa-
and ataxia telangiectasia, little is known about the tients have elevated blood counts; only half of the
cause of ALL. Epidemiologic studies so far have patients have a leukocyte count of more than 10,000/
failed to implicate any environmental factor (including /zL. Thrombocytopenia is often present although
electromagnetic fields, radon exposure, pesticides, and rarely as an isolated finding. Idiopathic thrombocyto-
maternal smoking) as causative for this disease. 6"7 penic purpura (ITP), a fairly common condition affect-
The clinical manifestations of childhood ALL are ing young children, is an important differential diag-
protean. Symptoms and signs reflect the effects of nosis of childhood ALL. This condition may even be
dysfunctional hematopoiesis (anemia, abnormal white associated with an increase in the proportion of "im-
cell counts, fever, thrombocytopenia), and clonal pro- mune" lymphocytes in the peripheral blood. However,
a normal hemoglobin and differential leukocyte count,
and absence of hepatomegaly and lymphadenopathy
From the Division of Pediatrics, The University of Texas MD Anderson (splenomegaly may occur in ITP) should be reassur-
Cancer Center, Houston, Texas. ing. The practice guidelines from the American Soci-
Curt Probl Pediatr 2002;32:40-49.
ety of Hematology suggested that a bone marrow
Copyright © 2002 by Mosby, Inc.
0045-9380/2002/$35.00 + 0 53/1/121790 examination is not necessary in the initial workup of
doi:10.lO67/mps.2002.121790 ITP. s

40 Curr Probl Pediatr, February 2002


Biologic Features of ALL this phenomenon does not appear to carry any prog-
nostic implication. Jl
Morphologic and Cytochemical Classification
The morphologic and cytochemical features of acute Cytogenetics
lymphoblastic leukemia are not pathognomic. The A number of nonrandom genetic abnormalities have
French-American-British working group had previ- been identified, and they bear significance in the
ously subdivided ALL into 3 types: L1, L2, and L3. pathobiologic condition of childhood ALL. t2 The
Although early trials suggested that L2 structure is advent of fluorescent in situ hybridization and molec-
associated with a poorer prognosis, this distinction has ular diagnostic techniques has allowed more sensitive
disappeared as more intensive therapy has been and accurate detection of these genetic aberrations.
used. 5"9 The L3 designation describes lymphoblasts of For example, it has recently been recognized that the
mature B-cell stage that require a different treatment most common cytogenetic abnormality in childhood
strategy, namely therapy specifically for the Burkitt's ALL is the cryptic translocation t(12;22), not detect-
type lymphoma. able by routine karyotyping analysis. ~3 This abnormal-
ity confers a good prognosis, as does hyperdiploidy
Immunophenoh/ping with 51 to 65 chromosomes. The numeric increase
Leukemia is a clonal disease resulting from the most often affects chromosomes 4, 10, 17, and 21.12 In
genetic mutation and transformation of a single hema- the leukemia cells of these patients, intracellular ac-
topoietic progenitor cell during its process of normal cumulation of methotrexate polyglutamates is in-
maturation. Therefore the phenotypic characteristics creased, which results in prolonged retention of the
of the malignant population reflect the expression of antifolate activity of methotrexate. ~4 The leukemia
lineage-associated antigens of that stage. Approxi- cells also have a propensity to undergo apoptosis. ~5
mately 85% of cases of ALL belong to the B-cell Both of these cytogenetic subtypes of ALL are more
lineage. Of these, 2% to 3% are mature B-cell ALL frequently found in children between 2 and 9 years of
that express surface immunoglobulin and are CD 20+. age and are associated with a low leukocyte count.
Another 20% to 30% of cases have a pre-B-cell These are also favorable prognostic factors and may
phenotype: presence of cytoplasmic, but not surface account for the high cure rate in these patients. On the
immunoglobulin. This represents an intermediate other hand, hypodiploidy with less than 45 chromo-
stage of B-cell differentiation. The rest of the cases somes, particularly 24 to 28 chromosomes (near-
belong to the early pre-B-cell type (B-precursor ALL). haploidy) is a significant adverse risk factor in spite of
They lack immunoglobulin expression but can be contemporary intensive therapies. 16
identified by the common ALL antigen (CD10) and A number of structural abnormalities of the chromo-
terminal deoxynucleotidyl transferase (CD19) positiv- some band 1 lq23 have been identified, most of which
ity. Likewise, the 15% of ALL derived from the T-cell convey a worse prognosis. ~7 Seventy percent of in-
lineage reflects the stages of thymic differentiation. fants with ALL TM and 85% of those with secondary
Most cases are from the early thymocyte stage; this is leukemia after epipodophyllotoxin exposure have this
different from T-cell lymphoma, which tends to dem- abnormality ~9 and carry a dismal prognosis. Other
onstrate an intermediate or a mature phenotype. T-cell children with this cytogenetic aberration have the
ALL is most commonly found in teenage males with same prognosis as those in the same risk category.
leukocytosis, meningeal involvement, and mediastinal Other cytogenetic features that portend a poor prog-
lymphadenopathy. Once associated with a poor prog- nosis and require different therapy are the transloca-
nosis, this subtype of ALL now has a prognosis equal tions involving protooncogenes on chromosome 8, 9,
to that of B-cell precursor disease when treated with and 22. The hallmark of mature B-cell ALL is the
appropriate risk-directed therapy. ~° With sensitive translocation of the MYC protooncogene from chro-
monoclonal antibody technology, leukemia cells can mosome 8 to an immunoglobulin gene, either the
often be shown to express markers of different hema- heavy chain on chromosome 14 or the light chains on
topoietic lineages. Between 7% and 25% of cases of chromosome 2 and 22, respectively. Patients with
otherwise typical ALL also express myeloid markers. B-cell ALL fare poorly with conventional ALL ther-
With modem chemotherapy regimens, the presence of apy but have an 80% event-free survival rate when

CUrT Probl Pediatr, February 2002 41


TABLE 1. Prognosticfactors for childhood ALL
Favorable Unfavorable
Age 1-9 yrs <1, >10 yrs
WBC <50,O00/#L >50,O00//~L"
Sex Female Male
Race White Black, Hispanic
Fulky disease" Absent Present
Extramedullary leukemia" Absent Present
Morphology (FAB)" Lz L2, L3
Immunophenotype" B-Precursor B, T
Cytogenetics t(12:21) Hypodiploidy (<45 chromosomes)
Hyperdiploidy (54-68 chromosomes) Philadelphia chromosome
11q23
t(1:19)"
Response to early treatment Rapid Slow"
(Day 7 or 14)
"Prognosticvalue modifiedby treatment.

treated with 3 to 6 months of rapid-sequence, intensive if not the most, important prognostic factors and is the
chemotherapy. 2° only one that can be modified. The current therapeutic
The Philadelphia (Ph) chromosome, with its t(9: approach is stratification according to risk factors at
22)(q34;ql 1) translocation, is found in 3% to 5% of diagnosis and early changes in therapy for those who
childhood ALL and about 20% of adult ALL. The do not respond well to induction. The concept of
translocation process leads to the juxtaposition of the altering therapy for patients who respond slowly, on
ABL and the BCR genes and the production of a the basis of laboratory findings during the first 7, 14, or
fusion protein (p185) with tyrosine kinase activity. It 28 days of induction therapy, has become a standard
is associated with a less than 20% disease-free survival practice and is described further in the Treatment
rate, even with intensive conventional chemothera- Strategies section b e l o w . 5"23"24 This principle applies
py.2~ Allogeneic hematopoietic stem cell transplanta- to all subgroups of ALL, with the exception of mature
tion has recently been identified as the only current B-cell-ALL, for which patients are treated with short-
therapy that has altered the long-term outcome of this term (less than 6 months) regimens of intensive
type of ALL. chemotherapy. On the other hand, the distinction of
Another translocation of prognostic significance is "lymphoma/leukemia" syndrome (for patients with
the translocation t (l:19)(q23;p13) found in 25% of bulky lymphadenopathy and massive hepatospleno-
cytoplasmic immunoglobulin M-positive pre-B-cell megaly) has been dropped, and patients are treated
ALL. 22 It is the most common cytogenetic abnormal- with identical risk-directed protocols.
ity of this subtype and is associated with a high There is no international consensus about risk as-
incidence of treatment failure if treated with a protocol signment, and various groups have used different and
based on the patent's age and white blood cell (WBC) overlapping risk classification schema for treatment
count alone. However, intensification of chemotherapy decisions. As a result, it is often difficult to compare
has improved the outcome of this subset of patients to outcome data from one group with another. The
a rate similar to those without the cytogenetic abnor- National Cancer Institute sponsored a risk classifica-
malities. tion workshop in 1993, and an agreement was reached
to use age and WBC count at diagnosis as the starting
point for risk assignment and reporting. 25 Children
Principles of Risk-directed Therapy ages 1 through 9 and with WBC count <50,000/~L
Childhood ALL is a heterogeneous disease. A num- are considered to be at standard risk. This constitutes
ber of host-related and disease-related prognostic fac- about two thirds of all cases and has an estimated
tors have been identified (Table 1); however, the event-free survival rate of more than 80%. The rest of
predictive strength of many of the original factors has the patients (those younger than 1 year or older than 10
been overcome by modern programs of chemotherapy. years of age or with a WBC count >50,000//zL)
The type of treatment has emerged as one of the most, belong to the high-risk group. They also generally

42 Curr Probl Pediatr, February 2002


have unfavorable cytogenetic and immunophenotypic complications. 27 Intrathecal administration of one or
characteristics. Some, but not all, staging classifica- more chemotherapeutic agents at regular intervals
tions also include genetic markers such as chromo- throughout the duration of leukemia therapy have been
somal analysis and leukemia cell DNA index to further shown to be as efficacious as radiation for CNS
determine risk assignment. prophylaxis. 28"29 It is also apparent that the CNS
relapse rate is, in part, dependent on the intensity of
concurrently administered systemic therapy. 3° For ex-
Treatment Strategies ample, high-dose intravenous methotrexate and cytar-
The standard elements of ALL treatment include abine penetrate into the spinal fluid and help to control
remission induction, prevention of leukemic infiltra- leukemia cells in this and other sanctuary sites. Use of
tion of the central nervous system, and prolonged dexamethasone instead of prednisone during induction
maintenance therapy (for 2 to 3 years). This basic improved event-free survival rates and affords better
approach has remained essentially unchanged for more CNS protection. 3~ This is attributed to dexametha-
than 30 years, but a number of modifications have sone's longer half-life, increased affinity for steroid
been incorporated that account for the steady improve- receptors, and better CNS penetration. Cranial irradi-
ment of treatment outcome. Current emphasis is fo- ation is now reserved for patients with overt CNS
cusing on intensification of chemotherapy in the early infiltration at diagnosis, or other very high-risk fea-
postremission period on the premise that early and tures (eg, extreme hyperleukocytosis, and presence of
extensive reduction of the leukemic burden will fore- the Philadelphia chromosome, slow response to initial
stall the development of drug resistance. therapy). The overall rate of relapse in the CNS is now
less than 5%. 28"29
Induction The definition of CNS leukemia has also been a
With a glucocorticoid, vincristine, and L-asparagi- point of controversy because of morphologic distor-
nase, with or without daunorubicin, all but 1% to 3% tion of white blood cells in the cerebrospinal fluid.
of children with ALL achieve morphologic marrow Five or more white blood cells//xL of cerebrospinal
remission after 4 weeks. Early rapid response as fluid and the presence of blast cells is generally
assessed by the rate of clearance of circulating or bone accepted as diagnostic. 32 With intensive systemic
marrow blasts after only l to 2 weeks of single-agent therapy and CNS radiotherapy, overt CNS leukemia at
or combination chemotherapy has a much better pre- presentation no longer has an adverse prognosis,
dictive value of long-term outcome than does response analogous to the diminished importance of many other
after 4 weeks. Schrappe et a126 showed that among prognostic factors with better treatment strategies.
patients with >1000 blasts//xL of blood after l week
of steroids and l dose of intrathecal methotrexate
Intensification
event-free survival was 43% points lower than among Randomized trials have shown that giving one or
the "early responders." Similarly, patients with >25% more courses of rotational, multiagent chemotherapy
blasts in the bone marrow 7 days from the beginning during the first 6 to 8 months from diagnosis signifi-
of treatment have a 5-year event-free survival rate of cantly reduces the risk of relapse. The approach was
only 42%. 24 These observations led to the strategy of first found to work in patients with high-risk fea-
modifying treatment intensity according to the "early tures 33'34 but subsequent studies showed that patients
response" criterion. of all risk categories benefited, including those with
clinical features suggestive of a high probability of
Central Nervous System Preventive Therapy c u r e . 9"35 There is wide variation among protocols in
Treatment directed to eradicate occult leukemia cells the choice of drugs, the dose intensity, and the timing
in the central nervous system (CNS) early after remis- of intensification treatments. Some introduce this
sion reduces the incidence of recurrence in this site. phase immediately after remission is achieved (con-
Preemptive cranial irradiation is effective but produces solidation) whereas others offer intensification after
long-term adverse neurodevelopmental and endocrine completion of a short period of intrathecal prophylaxis
sequelae. A reduction of the dose of cranial irradiation and oral maintenance (delayed intensification). How-
from 24 Gy to 18 Gy was equally effective, but ever, there is no definitive evidence that any specific
unfortunately modification did not prevent these late regimen is superior. It has been suggested recently that

Curr Probl Pediatr, February 2002 43


an additional course of intensive treatment (double association with t (4;11)]. Other patients who do poorly
delayed intensification) at 9 to 10 months after diag- include those who take more than 4 to 6 weeks to achieve
nosis may improve the outcome of patients with poor remission, and those with near-haploidy cytogenetic
prognostic factors such as older adolescents and slow abnormalities. Allogeneic HSC transplantation has been
"early responders" during induction. 33"34 tried with some success in these cases. 42"43 Recent
encouraging results with matched unrelated donors
Continuation Therapy (including umbilical cord blood units) suggest this ap-
Prolonged maintenance therapy is still an integral proach can be considered when a sibling donor is not
part of any ALL protocols. Timed from the end of available. 44.45
intensification, girls require 2 years of maintenance
therapy. For boys, this duration results in a higher
relapse rate. 36 When a third year of maintenance is
included for boys, males and females have similar Treatment of Relapse
outcomes. 9 Attempts to reduce the duration of treat- Although the outlook of childhood ALL has im-
ment to less than 2 years, even with aggressive proved in recent years, because of its relative fre-
chemotherapy up front, have been unsuccessful. 37 On quency, relapsed ALL remains a major problem in
the other hand, prolongation of maintenance to beyond pediatric oncology. Actually the number of such cases
3 years provides no advantage. There is no evidence is comparable to the number of children with newly
that treatment intensification at this stage is any more diagnosed brain tumors, the second most common
effective than low-dose antimetabolite therapy. How- malignancy in childhood. Because most of the effec-
ever, because of the variable bioavailability of oral tive antileukemic agents are given during initial ther-
agents, maintaining a low neutrophil count of 750 to apy, retrieval therapy with essentially the same drugs
1500//zL as a surrogate of adequate drug level is
has a low success rate. Duration of first remission,
important. 38 The addition of a dose of vincristine and
intensity of original therapy, and the site of relapse
a 5-day course of steroid monthly improve the overall
appear predictive of the duration of second remission.
outcomes, 39 whereas parenteral administration of 6
Bone marrow relapse is the principle form of treatment
mercaptopurine or methotrexate has not been shown to
failure. Patients who experience a relapse within the
confer any advantage. 4° Current research focuses on
first 18 months of diagnosis have little to no chance of
the choice of antimetabolite (mercaptopurine versus
survival with conventional chemotherapy.44'45 Even
thioguanine) and individualization of chemotherapy
patients who relapse more than 3 years after initial
dosage on the basis of drug clearance.41
diagnosis have an event-free survival rate of only
Treatment of Very High-risk Patients in First 30%. 46,47 Patients with combined marrow and ex-
Complete Remission tramedullary relapse have been reported to have a
Alternative treatment strategies have been advo- somewhat better prognosis. 48
cated for certain groups of patients with very high Thirty percent of all relapses appear to be isolated to
risk of treatment failure, defined as an anticipated extramedullary sites, most commonly in CNS, testic-
event-free survival rate of less than 40% in spite of ular, or ocular locations. 49 The prognosis of these
modern therapy. For example, allogeneic hemato- patients is better than that of patients with marrow
poietic stem cell (HSC) transplantation resulted in relapse, but many do have development of subsequent
the best outcome in patients with Philadelphia- systemic disease. The outcome is worse for CNS
positive ALL. 21 In addition it has been suggested relapse that occurs within 18 months of diagnosis,
that the graft-versus-leukemia effect may be more similar to that of marrow relapse. Patients with iso-
prominent in this condition. lated testicular relapse fare the best with an event-free
Acute lymphoblastic leukemia in infants is com- survival rates exceeding 60%. 50 Because the incidence
monly associated with hyperleukocytosis and a pro- of testicular relapse has decreased with contemporary
pensity to have meningeal disease at diagnosis. Increased intensified systemic therapy and because of the lack of
treatment intensity has not affected the prognosis of the predictive value of random testicular biopsy at the
70% to 80% of infants who have the mixed lineage conclusion of the maintenance phase, this procedure is
leukemia (MLL) gene rearrangement [most often in no longer recommended.

44 Curr Probl Pediatr, February 2002


AIIogeneic HSC Transplantation Recent Advances
The role of HSC transplantation in ALL remains the Monitoring of Minimal ResidualDisease
subject of continued debate. Because of the excellent
With the success of risk-directed therapy, a logical
results of modern chemotherapy, HSC transplantation
next step is to explore the ability to identify those
with its associated toxicity is not indicated in first
patients destined to have a relapse in spite of being in
remission, with the exceptions of the conditions dis-
apparent morphologic remission. Techniques such as
cussed above. Once the leukemia has relapsed, the
flow cytometry (to enumerate cells expressing leuke-
prognosis is drastically different. Chemotherapy pro- mic-associated antigens) and polymerase chain reac-
duces an event-free survival rate of only 10% for tion (to examine "patient-specific" T cell or immuno-
patients with bone marrow relapse, and 25% for those globulin gene receptor rearrangement pattern) can
with CNS relapse that occurred within 3 years from detect one leukemic cell among 104 tO 105 cells,
diagnosis. High-dose chemoradiotherapy and allo- several logs less in magnitude than that found at frank
geneic HSC transplantation has been shown to be relapse. 56 Cave et a157 recently reported that detectable
more effective in preventing further leukemia recur- minimal residual disease (MRD) at levels higher than
rence. 4648 Most series report a better event-free sur- l in 100 at the end of induction was associated with a
vival rate for recipients of matched sibling transplants 16 times higher risk of subsequent relapse compared
for early marrow relapses, although its benefit is less with lower MRD burden. After consolidation the
definite for patients with relapses occurring more than breakpoint of MRD burden for increased risk of
6 months after discontinuation of therapy. 47"51 Cur- treatment failure was l in 1000. 57 The prognostic
rently most centers have reserved transplantation for significance of MRD was independent of other known
patients with late relapses only if they respond slowly risk factors for relapse. Clinical trials are being de-
to retrieval chemotherapy. signed for patients with detectable MRD at defined
The risk of bone marrow transplantation relates to time points to receive augmented therapy. MRD is
toxicity of the preparative regimen and the harmful also related to the outcome of HSC transplantation for
effect of graft-versus-host disease. These complica- ALL. Patients with negative, low, and high levels of
tions are more pronounced with unrelated and par- MRD immediately before transplantation had an
tially mismatched family donor grafts, the only stem event-free survival rate of 73%, 36%, and 0%, respec-
cell source available to three quarters of the patients tively. 58 In another study the presence of MRD in the
needing transplantation. 52"53 A high transplantation- first 3 months after transplantation increased the like-
related mortality rate with these donors might ne- lihood of subsequent relapse by 9-fold. 59
gate the antileukemic potential of the procedure, Genetic Polymorphisms
leading to greater uncertainty with regard to
Race- and sex-associated genetic polymorphisms
whether HSC transplantation as a treatment modal-
can affect drug metabolism and detoxification enzyme
ity can alter the overall outcome of relapsed ALL. 5
activities. For instance, genotype variations of N-
Recent technologic advances in alternative donor
acetyltransferase, glutathione-S-transferase, and the
HSC transplantation may have improved the patient cytochrome P450 system enzymes may alter chemo-
outcome to the same extent as in the sibling donor therapeutic agent disposition and account for drug
group. 54,55
resistance and toxicity. 6° This phenomenon may ex-
Autologous HSC transplantation, with the patient's plain the prognostic differences in individual patients,
stem cells harvested in remission, is an alternative for as well as among racial groups and sexes. A recent
patients without an allogeneic donor. Purging with development is the screening of gene expression
chemotherapeutic agents or with antibody against profiles by use of microarray techniques to better
leukemia-associated antigens is often performed on define the prognosis and treatment strategies for indi-
the stem cell collection to reduce tumor contamina- vidual patients. 61
tion. The procedure carries a lower morbidity rate, but
evidence so far has not shown any evidence that Complications of Treatment
autologous HSC transplantation is of benefit in re- Complications of antileukemia treatment can be
lapsed ALL. acute or of belated onset. Advances in supportive care

Curr Probl Pediatr, February 2002 45


measures have helped to reduce the mortality and and treatment given for the primary tumor. Solid
morbidity rates of ALL therapy. Platelet and erythro- tumors, especially brain tumors, are the most common
cyte transfusions are often required, and all cellular second malignancies in patients with ALL. One per-
products should be irradiated to avoid the risk of cent of patients who received cranial irradiation will
transfusion-related graft-versus-host disease. On the have development of a brain tumor. The latency period
other hand, administration of granulocyte colony- is 9 years for high-grade glioma and 19 years for
stimulating factor during the intensive phase of che- meningioma. 65 Secondary AML has developed after
motherapy does not significantly reduce the duration regimens that include topoisomerase II inhibitor (epi-
of neutropenia, rate of hospitalization, or delay in podophyllotoxin) or alkylating agents. The risk for
therapy. 62 topoisomerase II inhibitor-induced leukemia was
Metabolic derangement as a result of tumor lysis is shown to be related to the treatment schedule of the
particularly common in patients with marked leukocy- offending agents and concomitant use of other
tosis (>100,000//~L), massive mediastinal lymphade- drugs. ~9 The prognosis of secondary AML is ex-
nopathy, and T- or B-cell disease. Urate oxidase, a tremely poor, and only allogeneic HSC transplantation
recombinant enzyme that breaks down uric acid to provides any chance of cure.
allantoin has been shown to rapidly and markedly The neurocognitive development and academic
lower uric acid levels and avoid renal insufficiency.63 achievement of childhood leukemia survivors are
However, the nephrology service should be consulted lower than that of control subjects and their normal
should dialysis become necessary. Induction chemo- siblings. Cranial irradiation, especially in patients 5
therapy should not be unduly delayed waiting for the years of age and younger, has the most deleterious
complete correction of the metabolic abnormalities. effect on neuropsychologic development. A dose-
Infections are the most common cause of death in response relationship has been observed between re-
remission for children with ALL. This happens during duction in intelligence quotient points and the dose of
induction and intensification phase of chemotherapy. irradiation. 27 On the other hand, infants exposed to a
Prompt administration of broad-spectrum antibiotic very high dose of systemic methotrexate and intrathe-
therapy is essential for the treatment of the patient with cal chemotherapy for CNS prophylaxis showed neu-
febrile neutropenia (<500 granulocytes/~L). During rodevelopmental testing scores normal for age. 66 Ra-
maintenance therapy, patients are susceptible to pneu- diation-induced hypothalamic-pituitary dysfunction
monits by Pneumocvstis carinii, although the risk can results in growth hormone deficiency but can also be
be completely eliminated by prophylactic use of tri- associated with accelerated skeletal maturation and
methoprim-sulfamethoxazole (given on 3 sequential epiphyseal fusion. A complex of short stature, obesity,
days per week). Disseminated varicella is also a and precocious puberty is recognized and is particu-
serious complication of ALL therapy. Administration larly common among girls. Although most ALL sur-
of zoster immunoglobulin within 72 to 96 hours of vivors have normal sexual development, gonadal dam-
exposure may attenuate the severity of subsequent age can be demonstrated in some patients. This may
infection. Acyclovir is effective in limiting the sever- become more prevalent because the total dose of
ity of an established varicella infection. Immunization cyclophosphamide has been escalated in the more
of nonimmune household contact with the live, atten- intensive regimens. Currently there is no evidence that
uated varicella vaccine is encouraged. Its use in the offspring of patients with leukemia have an increased
patient with leukemia remains controversial. Measur- risk of congenital abnormalities.
able antibody production against the virus is found, Avascular necrosis of bone, with destruction of
but maintenance chemotherapy has to be withheld for weight-bearing joints, is a side effect of glucocorticoid
several weeks before immunization. (especially high-dose dexamethasone) treatment. 67 It
Late effects of disease and therapy have become affects mainly female patients older than 10 years of
increasingly recognized in survivors of childhood age. Magnetic resonance imaging is the most sensitive
ALL. 64 The cumulative incidence of second malig- diagnostic study for this complication. Decreased bone
nancy 15 years after the diagnosis of childhood leu- mineral density and osteoporosis has been observed in
kemia is low, only 1.6% in the trials of the large U.S. all phases of treatment up to 20 years after diagnosis. 68
national cooperative groups. This occurs as a result of This leads to musculoskeletal pain, compression frac-
an interaction between predisposing genetic factors tures, kyphosis, and lordosis.

46 Curr Probl Pediatr, February 2002


Each year more than 1500 children in this country 14. Whitehead VM, Vuchich MJ, Lauer SJ, et al. Accumulation of
become long-term survivors of childhood ALL. They high levels of methotrexate polyglutamates in lymphoblasts
from children with hyperdiploid (greater than 50 chromo-
return to the community and are monitored by their
somes) B-lineage acute lymphoblastic leukemia: a Pediatric
local physicians. Late side effects are common among Oncology Group Study. Blood 1992;80:1316-23.
these survivors, and the manifestations may be subtle 15. Ito C, Kumagai M, Manabe A, et al. Hyperdiploid acute
and take years to develop. Careful and regular (prob- lymphoblastic leukemia with 51 to 65 chromosomes: a distinct
ably life-long) surveillance of these leukemia-free biological entity with a marked propensity to undergo apopto-
children has become essential. sis. Blood 1999;93:315-20.
16. Pui CH, Carroll AJ, Raimondi SC, et al. Clinical presentation,
karyotypic characterization, and treatment outcome of child-
References hood lymphoblastic leukemia with near-haploid or hypodip-
1. Smith MA, Gloeckler Ries LA, Gurney JG, et al. Leukemia. loid less than 45 line. Blood 1990;75:1170-7.
In: Ries LAG, Smith MA, Gurney JG, Linet M, Tamra T, 17. Uckun FM, Herman-Hatten K, Crotty ML, et al. Clinical
Young JL, Bunin GR, editors. Cancer incidence and survival significance of MLL-AF4 fusion transcript expression in the
among children and adolescents. Bethesda (MD): National absence of a cytogenetically detectable t(4:l l)(q21 ;q23) chro-
Cancer Institute, SEER Program; 1999. p. 17-34. mosomal translocation. Blood 1998;93:1106-7.
2. Pui CH, Evans WE. Acute lymphoblastic leukemia. N Engl 18. Reaman GH, Sposto R, Sensel MG, et al. Treatment outcome
J Med 1998;339:605-14. and prognostic factors for infants with acute lymphoblastic
3. Gaynon PS, Trigg ME, Heerema NA, et al. Children's cancer leukemia treated on two consecutive trials of the Children's
group trials in childhood acute lymphoblastic leukemia: 1983- Cancer Group. J Clin Oncol 1999;17:445-55.
1995, Leukemia 2000; 14:2223-33. 19. Felix CA. Secondary leukemias induced by topoisomerase-
4. Pui CH, Boyett JM, Rivera GK, et al. Long-term results of targeted drugs. Biochimica et Biophysica Acta 1998;1400:
total therapy studies II, 12 and 13A for childhood acute 233-55.
lymphoblastic leukemia at St Jude Children's Research Hos- 20. Atra A, Gerrard M, Hobson R, et al. Improved cure rate in
pital. Leukemia 2000; 14:2286-94.
children with B-cell acute lymphoblastic leukemia (B-ALL)
5. Reiter A, Schrappe M, Ludwig WD, et at. Chemotherapy in
and stage IV B-cell non-Hodgkin's lymphoma (B-NHL)--
998 unselected childhood acute lymphoblastic leukemia
results of UKCCSG 9003 protocol. Brit J Cancer 1998;77:
patients: results and conclusions of the multicenter trial
2281-5.
ALL-BFM 86. Blood 1994;84:3122-33.
21. Arico M, Valsecchi MG, Camitta B, et al. Outcome of
6. Lubin JH, Linet MS, Boice JD Jr, et al. Case-control study of
treatment in children with Philadelphia chromosome-positive
childhood acute lymphoblastic leukemia and residential radon
acute lymphoblastic leukemia. N Engl J Med 2000;342:998-
exposure. J Natl Cancer lnst 1998;90:294-300.
I006.
7. Linet MS, Hatch EE, Kleinerman RA, et al. Residential
22. Pui CH, Raimondi SC, Hancock ML, et al. Immunologic,
exposure to magnetic fields and acute lymphoblastic leukemia
cytogenetic, and clinical characterization of childhood acute
in children. N Engl J Med 1997;337:1-7.
lymphoblastic leukemia with the t(l ;19)(q23;p13) or its de-
8. George JN, Woolf SH, Raskob GE, et al. Idiopathic throm-
bocytopenic purpura: a practice guideline developed by ex- rivative. J Clin Oncol 1994;12:2601-6.
plicit methods for the American Society of Hematology. 23. Steinherz PG, Gaynon PS, Breneman JC, et al. Cytoreduction
and prognosis in acute lymphoblastic leukemia--the impor-
Blood 1996;88:3-40.
9. Schrappe M, Reiter A, Zimmermann M, et al. Long-term tance of early marrow response: report from the Children's
results of tbur consecutive trials in childhood ALL performed Cancer Group. J Clin Oncol 1996;14:389-98.
by the ALL-BFM study group from 1981-1995. Leukemia 24. Gaynon PS, Desai AA, Bostrom BC, et al. Early response to
2000; 14:2205-22. therapy and outcome in childhood acute lymphoblastic
10. Uckun FM, Reaman G, Steinherz PG, et al. Improved clinical leukemia: a review. Cancer 1997;80:1717-26.
outcome for children with T-lineage acute lymphoblastic 25. Smith M, Arthur D, Camitta B, et al. Uniform approach to risk
leukemia after contemporary chemotherapy: a Children's classification and treatment for children with acute lympho-
Cancer Group study. Leuk Lymphoma 1996;24:570-70. blastic leukemia. J Clin Oncol 1996;14:18-24.
I I. Uckun FM, Sather HN, Gaynon PS, et al. Clinical features and 26. Schrappe M, Reiter A, Sauter S, et al. Risk-oriented treatment
treatment outcome of children with myeloid antigen positive of childhood ALL: favorable outcome despite reduced inten-
acute lymphoblastic leukemia: a report from the Children's sity of treatment in trial ALL-BFM 90. Blood 1997;90(suppl
Cancer Group. Blood 1997;90:28-35. 1):2448.
12. Ferrando AA, Look AT. Clinical implications of recurring 27. Mulhern RK, Fairclough D, Ochs J, et al. A prospective
chromosomal and associated molecular abnormalities in acute comparison of neuropsychological performance of children
lymphoblastic leukemia. Sem Hematol 2000;37:381-95. surviving leukemia who survived 18-Gy, 24-Gy, or no cranial
13. Shurtleff SA, Buijs A, Behm FG, et al. TEL/AML1 fusion irradiation. J Clin Oncol 1991;9:1348-56.
resulting from a cryptic t(12;21) is the most common genetic 28. Pullen J, Boyett J, Shuster J, et al. Extended triple intrathecal
lesion in pediatric ALL and defines a subgroup of patients chemotherapy trial for prevention of CNS relapse in good-risk
with an excellent prognosis. Leukemia 1995;5:1985-9. and poor-risk patients with B-progenitor acute lymphoblastic

Curr Probl Pediatr, February 2002 47


leukemia: a Pediatric Oncoiogy Group study. J Clin Oncol treatment for infants with acute lymphoblastic leukemia. Med
1993;11:839-49. Pediatr Oncol 1999;32:1-6.
29. Tubergen DG, Gilchrsit GS, O'Brien RT, et al. Prevention of 43. Wheeler KA, Richards SM, Bailey CC, et al. Bone marrow
CNS disease in intermediate-risk acute lymphoblastic transplantation versus chemotherapy in the treatment of very
leukemia: comparison of cranial radiation and intrathecal high-risk childhood acute lymphoblastic leukemia in first
methotrexate and the importance of systemic therapy: a remission: results from Medical Research Council UK ALL X
Children's Cancer Group report. J Clin Oncol 1993;11:520-6. and XI. Blood 2000;96:2412-8.
30. Tubergen DG, Gilchrist GS, O'Brien RT, et al. Improved 44. Gaynon PS, Qu RP, Chappell RJ, et al. Survival after relapse
outcome with delayed intensification for children with acute in childhood acute lymphoblastic leukemia: impact of site and
lymphoblastic leukemia and intermediate presenting features: time to first relapse--the Children's Cancer Group experience.
a Children's Cancer Group phase III trial. J Clin Oncol Cancer 1998;82:1387-95.
1993;11:527-37. 45. Rivera GK, Hudson MM, Liu Q, et al. Effectiveness of
31. Balls FM, Lester CM, Chrousos GP, et al. Differences in intensified rotational combination chemotherapy for late he-
cerebrospinal fluid penetration of corticosteroids: possible matologic relapse of childhood lymphoblastic leukemia.
relationship to the prevention of meningeal leukemia. J Clin Blood 1996;88:83 I-7.
Oncol 1987;5:202-7. 46. Barrett AJ, Horowitz MM, Pollock BH, et al. Bone marrow
32. Tubergen DG, Cullen JW, Boyett JM, et al. Blasts in the CSF transplants from HLA-identical siblings as compared with
with a normal cell count do not justify alternation of therapy chemotherapy for children with acute lymphoblastic leukemia
of acute lymphoblastic leukemia in remission: a Children's in second remission. N Engl J Med 1994;331:1253-8.
47. Uderzo C, Valsecchi MG, Bacigalupo A, et al. Treatment of
Cancer Group study. J Clin Oncol 1994;12:273-8.
childhood acute lymphoblastic leukemia in second remission
33. Nachman JB, Sather HN, Sensel MG, et al. Augmented
with allogeneic bone marrow transplantation and
post-induction therapy for children with acute lymphoblastic
chemotherapy: ten-year experience of the Italian bone marrow
leukemia and a slow response to initial therapy. N Engl J Med
transplantation group and the Italian Pediatric Hematology
1998;338:1663-71.
Oncology Association. J Clin Oncol 1995;13:352-8.
34. Chessells JM. The management of high-risk lymphoblastic
48. Buhrer C, Hartmann R, Fengler R, et al. Superior prognosis in
leukemia in children. Br J Haematol 2000; 108:204-16.
combined to isolated bone marrow relapse in salvage therapy
35. Chessells JM, Bailey C, Richards SM, et al. Intensification of
of childhood acute lymphoblastic leukemia. Med Pediatr
treatment and survival in all children with lymphoblastic
Oncol 1993;21:470-6.
leukemia: results of UK Medical Research Council trial
49. Winick NJ, Smith SD, Shuster J, et al. Treatment of CNS
UKALL X. Medical Research Council Working Party on
relapse in children with acute lymphoblastic leukemia: a
Childhood Leukemia. Lancet 1995;345:143-8.
Pediatric Oncology Group study. J Clin Oncol 1993; 11:271-8.
36. Pui CH, Boyett JM, Relling MV, et al. Sex differences in
50. Buchanan GR, Boyett JM, Pollock BH, et al. Improved
prognosis for children with acute lymphoblastic leukemia.
treatment results in boys with overt testicular relapse during or
J Clin Oncol 1999;17:818-24. shortly after initial therapy for acute lymphoblastic leukemia:
37. Toyoda Y, Manabe A, Tsuchida M, et al. Six months of a Pediatric Oncology Group study. Cancer 1991;68:48-55.
maintenance chemotherapy after intensified treatment for 51. Wheeler K, Richards S, Bailey C, et al. Comparison of bone
acute lymphoblastic leukemia of childhood. J Clin Oncol marrow transplant and chemotherapy for relapsed childhood
2000;18:1508-16. acute lymphoblastic leukemia: the MRC UKALL X experi-
38. Chessells JM, Harrison G, Lilleyman JS, et al. Continuing ence. Medical Research Council Working Party on Childhood
(maintenance) therapy in lymphoblastic leukemia: lessons Leukemia. Br J Haematol 1998;101:94-103.
from MRC UKALL X Medical Research Council Working 52. Davies SM, Wagner JE, Shu XO, et al. Unrelated donor bone
Party in Childhood Leukemia. Br J Haematol 1997;98:945-5 I. marrow transplantation for children with acute leukemia.
39. Bleyer WA, Sather HN, Nickerson HJ, et al. Monthly pulses J Clin Oncol 1997;15:557-65.
of vincristine and prednisone prevent bone marrow and 53. Balduzzi A, Gooley T, Anasetti C, et al. Unrelated donor
testicular relapse in low-risk childhood acute lymphoblastic marrow transplantation in children. Blood 1995;86:3247-56.
leukemia: a report of the CCG 161 study by the Children's 54. Oakhill A, Pamphilon DH, Potter MN, et al. Unrelated donor
Cancer Study Group. J Clin Oncol 1991;9:1012-21. bone marrow transplantation for children with relapsed acute
40. Lange BJ, Blatt J, Sather HN, et al. Randomized comparison lymphoblastic leukemia in second complete remission. Br J
of moderate-dose methotrexate infusions to oral methotrexate Haematol 1996;94:574-8.
in children with intermediate risk acute lymphoblastic 55. Saarinen-Pihkala UM, Gustafsson G, Ringden O, et al. No
leukemia: a Children's Cancer Group study. Med Pediatr disadvantage in outcome of using matched unrelated donors as
Oncol 1996;27:15-20. compared with matched sibling donors for bone marrow
41. Evans WE, Relling MV, Rodman JH, et al. Conventional transplantation in children with acute lymphoblastic leukemia
compared with individualized chemotherapy for childhood in second remission. J Clin Oncol 2001;19:3406-14.
acute lymphoblastic leukemia. N Engl J Med 1998;338:499- 56. Coustan-Smith, Sancho J, Hancock ML, et al. Clinical impor-
505. tance of minimal residual disease in childhood acute lympho-
42. Pirich L, Haut P, Morgan E, et al. Total body irradiation, blastic leukemia. Blood 2000;96:2691-6.
cyclophosphamide, and etoposide with stem cell transplant as 57. Cave H, van der Werfften Bosch J, Suciu S, et al. Clinical

48 Curr Probl Pediatr, February 2002


significance of minimal residual disease in childhood acute children with acute lymphoblastic leukemia. N Engl J Med
lymphoblastic leukemia. European Organization for Research 1997 ;336:1781-7.
and Treatment of Cancer-Childhood Leukemia Cooperative 63. Pui CH, Jeha S, Camitta B. Recombinant urate oxidase
Group. N Engl J Med 1998;339:591-8. (Rasburicase) in the prevention and treatment of malignancy-
58. Knechtli CJC, Goulden NJ, Hancock JP, et al. Minimal associated hyperuricemia: the compassionate use experience
residual disease status before allogeneic bone marrow trans- abstract. Blood 2000;96:719a.
plantation is an important determinant of successful outcome 64. Shusterman S, Meadows AT. Long term survivors of child-
for children and adolescents with acute lymphoblastic leuke- hood leukemia. Curr Opin Hematol 2000;7:217-20.
mia. Blood 1998;92:4072-9. 65. Walter AW, Hancock ML, Pui CH, et al. Secondary brain
59. Knechtli CJC, Goulden NJ, Hancock JP, et al. Minimal tumors in children treated for acute lymphoblastic leukemia at
residual disease status as a predictor of relapse after allogeneic
St Jude Children's Research Hospital. J Clin Oncol 1998;16:
bone marrow transplantation for children with acute lympho-
3761-7.
blastic leukemia. Br J Haematol 1998;102:860-71.
66. Kaleita TA, Reaman GH, MacLean WE, et al. Neurodevelop-
60. Stanulla M, Schrappe M, Brechlin AM, et al. Polymorphisms
mental outcome of infants with acute lymphoblastic leukemia:
with glutathione-S-transferase genes (GSTMI, GSTI'I,
a Children's Cancer Group report. Cancer 1999;85:1859-65.
GSTPI) and risk of relapse in childhood B-precursor acute
lymphoblastic leukemia: a case-controlled study. Blood 2000; 67. Mattano LA Jr, Sathere HN, Trigg ME, et al. Osteonecrosis as
95:1222-8. a complication of treating acute lymphoblastic leukemia in
61. Chen JS, Coustan-Smith E, Suzuki T, et al. Identification of children: a report from the Children's Cancer Group. J Clin
novel markers for the monitoring minimal residual disease in Oncol 2000;18:3262-72.
acute lymphoblastic leukemia. Blood 2001;97:2115-20. 68. Haddy TB, Mosher RB, Reaman GH. Osteoporosis in survi-
62. Pui CH, Boyett JM, Hughes WT, et al. Human granulocyte vors of acute lymphoblastic leukemia. The Oncologist 2001;
colony-stimulating factor after induction chemotherapy in 6:278-85.

Curr Probl Pediatr, February 2002 49

You might also like