Vous êtes sur la page 1sur 10

TREATMENT

BETSY M. BICKERT
OF COMMON CHILDHOOD MALIGNANCIES

Treatment of Common
Childhood Malignancies
Betsy M. Bickert, PharmD

Malignancy is the fourth leading cause of death in persons toxicity. This article will discuss the current treatment regi-
aged 1 to 19 years. The success of the pediatric oncology co- mens for several of the more common pediatric malignancies
operative group clinical trials has allowed a systematic ap- and their long-term complications developed through this
proach to the treatment of relatively rare malignancies. Each process.
trial builds on previous results in terms of both survival and
KEY WORDS: pediatric, oncology, cancer, chemotherapy.

M ALIGNANCY IS THE FOURTH leading cause of


death in persons aged 1 to 19 years. In 1998,
12,400 persons younger than 20 years of age were diag-
of all stages or grades of each tumor type unless other-
wise specified.

nosed with a malignancy, and it was the cause of death HEMATOLOGIC MALIGNANCIES
for an estimated 2500 children. Newborn males have a
1 in 300 chance of developing a malignancy by the age Leukemias are identified based on the cell of origin:
of 20, and the number is slightly higher for females.1 lymphoid or myeloid. The leukemias are the most com-
The incidence of the most common malignancies of mon malignancies in childhood and are the primary
childhood is presented in Figure 1. cause of cancer-related mortality in the United States.1
In a 1992 survey of protocol participation, 62% of Acute lymphoblastic leukemia (ALL) composes 75%
pediatric oncology patients treated at pediatric centers of childhood leukemia, and acute myelogenous leuke-
were enrolled onto a treatment protocol, whereas only mia composes approximately 19%.1 Infant leukemia is
2% of adults were enrolled on such protocols.2 This has a separate entity that has been reviewed elsewhere.3,4
allowed a systematic approach to the treatment of rela-
tively rare diseases by accruing large numbers of pa- Acute Lymphoblastic Leukemia
tients to consecutive protocols within a cooperative
group to compare therapeutic regimens. These re- Approximately 2400 persons younger than 20 years
search protocols allow the discovery of therapeutic of age are diagnosed with ALL annually in the United
successes and failures as well as characterizing treat- States.1 The peak incidence is between 2 and 5 years of
ment-related toxicity. This article will discuss the cur- age. ALL is 30% more common in males than in fe-
rent treatment regimens and their long-term complica- males, and white children are twice as likely to develop
tions for common pediatric malignancies developed ALL as black children. Children with Down syndrome,
through this process. Results are presented as averages neurofibromatosis, Schwachmann syndrome, Bloom
syndrome, ataxia telangiectasia, Langerhans cell
histiocytosis, and Klinefelter syndrome are at higher
Address correspondence to: Betsy M. Bickert, PharmD, Oncology
and Bone Marrow Transplant Clinical Specialist, Department of
risk of developing ALL.
Pharmacy Services, The Children’s Hospital of Philadelphia, 34th In the 1960s, a remission could be achieved in more
St and Civic Center Blvd, Philadelphia, PA 19104. E-mail: bickert@ than 90% of children treated with prednisone and
email.chop.edu vincristine, but resistance would develop within
months of continued therapy. At that time, 50% of chil-
JOURNAL OF PHARMACY PRACTICE, 15:1 42–51
© 2002 Sage Publications dren with ALL developed central nervous system
DOI: 10.1106/JYT9-MA70-6CP3-XGCT (CNS) involvement of the disease.5 The method of treat-
TREATMENT OF COMMON CHILDHOOD MALIGNANCIES

Other (20%)
Leukemia (25%)

Kidney (4%)

Sympathetic (5%)

Bone (6%)
Brain tumors (17%)

Soft Tissue (7%)


Figure 1. Cancer incidence
<20 years of age from 1975 to
Lymphoma (16%) 1995.1

ment and prevention of CNS relapse was cranial or Dexamethasone decreased the CNS relapse rate and in-
craniospinal irradiation. creased 3-year event-free survival by 5% over
In an effort to maintain remission, St. Jude prednisone in standard risk patients.13 Dexamethasone
Children’s Research Hospital developed the concept of was also associated with an increased incidence of
total therapy to include induction, consolidation, and hyperglycemia. The current standard risk protocol in-
maintenance. Induction is the initial therapy given to cludes dexamethasone in all phases of treatment.
induce a remission; consolidation targets leukemia As survival improved with new treatments, certain
cells in the CNS; and maintenance utilizes low-dose prognostic factors became apparent. Children with a
therapy to ensure clearance of residual leukemia cells white blood cell count greater than 100,000/mm3,
over a longer period of time. This approach achieved a bulky extramedullary disease, and age younger than 1
9-year, event-free survival rate of 51% in the 1970s.6 year and greater than 10 years had worse outcomes. At
In 1976, the Berlin/Frankfurt/Munster (BFM) group the same time, technological advances determined that
added a pulse of delayed intensification to enhance having ≥50 chromosomes is favorable and having <45
therapy against residual leukemia with novel agents is unfavorable. Nonrandom chromosomal transloca-
that achieved a 70% long-term survival rate.7 Delayed tions were also identified. Translocation (4;11) and the
intensification (DI) increased 5-year event-free survival Philadelphia chromosome t (9;22) are unfavorable and
from 61% to 73% in patients with intermediate risk place patients in the high-risk group. The translocation
ALL and increased 7-year event-free survival from 40% involving the MLL gene on chromosome 11 band q23 is
associated with infant ALL and a 5-year event-free sur-
to 63% for high-risk patients in the United States.8,9 The
vival of only 37%.1 Risk factors identified through the
addition of a second delayed intensification (double
delayed intensification [DDI]) has improved event-free
survival at 6 years from 87% with a single DI to 91% Table 1
with DDI in patients with intermediate risk ALL.10 The 1997 National Cancer Institute Acute
14
current standard risk protocol randomizes patients to Lymphoblastic Leukemia Risk Stratification
either one or two delayed intensification phases. White Blood
In 1983, a randomized trial showed that 3 years of Risk Age Cells
therapy was equivalent to 5 years of therapy.11 Miller
3
Group (years) (cells/mm ) Karyotype
and colleagues12 showed that for girls, 2 years of ther- Standard 1 to <10 <50,000 No t(9;22) or t(4;11)
apy was equivalent to 3 years of therapy, but that for High 1 to <10 ≥50,000
boys 3 years of therapy was better than 2 years, which <1 or ≥10 Any
Any Any t(9;22) or t(4;11)
remains the current standard length of treatment.12

JOURNAL OF PHARMACY PRACTICE 2002(15.1) • 43


BETSY M. BICKERT

Table 2
Standard Acute Lymphoblastic Leukemia Therapy

Phase (duration) Standard-risk Therapy High-risk Therapy

Induction (1 month) Intrathecal therapy, vincristine, steroid, Intrathecal therapy, vincristine, steroid,
asparaginase daunorubicin
Consolidation Mercaptopurine, vincristine, intrathecal Mercaptopurine intrathecal methotrexate,
(4 weeks/5 weeks) methotrexate cyclophosphamide, cytarabine, prednisone
Interim maintenance Methotrexate, mercaptopurine, vincristine, Methotrexate, mercaptopurine, intrathecal
(2 months) steroid, intrathecal methotrexate methotrexate
Delayed intensification Vincristine, dexamethasone, pegaspargase, Vincristine, dexamethasone, asparaginase,
(8 weeks/7 weeks) doxorubicin, cyclophosphamide, thioguanine, doxorubicin, cyclophosphamide, thioguanine,
cytarabine, intrathecal methotrexate cytarabine, intrathecal methotrexate
Maintenance Methotrexate, mercaptopurine, vincristine, Methotrexate, mercaptopurine, vincristine,
(12-week cycles) dexamethasone, intrathecal methotrexate prednisone, intrathecal methotrexate
Oral methotrexate, mercaptopurine, and thioguanine are most effective if given at bedtime on an empty stomach.

cooperative group trials resulted in the 1997 Na- kill residual leukemic cells while limiting develop-
tional Cancer Institute ALL Risk Stratification as seen ment of drug resistance by administering agents with
in Table 1.14 These criteria are now used to determine different mechanisms of action. Interim maintenance is
treatment for children presenting with newly diag- given in between intense cycles to maintain remission
nosed ALL. but to avoid toxicity from continuously intense ther-
Most chemotherapy agents do not readily cross the apy. Low-dose pulses of chemotherapy are given dur-
blood-brain barrier. This allows leukemic cells to use ing maintenance to further ensure the clearance of re-
the CNS as a sanctuary site from treatment. Intrathecal sidual leukemic clones. Patients with unfavorable
chemotherapy, cranial irradiation, and high-dose intra- cytogenetics or with slow early response after induc-
venous methotrexate or cytarabine are all effective tion receive augmented therapy with higher doses of
methods to prevent spread of the disease into the CNS. some agents, such as steroids, and the addition of other
Intrathecal therapy consists of cytarabine alone, agents to increase the probability of a remission.
methotrexate alone, or methotrexate, hydrocortisone, Children with high-risk ALL receive more intense
and cytarabine in combination. Tubergan and chemotherapy (Table 2). On Day 7 of treatment, the
colleagues9 found intrathecal methotrexate is equiva- bone marrow is assessed for residual leukemic blasts.
lent to cranial irradiation for CNS prophylaxis in chil- Patients with greater than 25% blasts are considered
dren younger than 10 years of age. Italian investigators slow early responders and receive even more intense
have also found that cranial irradiation can be replaced therapy. This stratification increased 5-year event-free
with 4 doses of intravenous methotrexate in consolida- survival from 55% to 75%.19
tion and with additional intrathecal methotrexate in Modifications based on study results have improved
maintenance therapy with a less than 1% incidence of survival rates by building on existing treatment regi-
CNS relapse at 6 years.15 Prophylactic cranial irradia- mens rather than relying on development of new
tion has been eliminated for patients without CNS dis- agents. Current therapy yields an 80% to 85% long-
ease at diagnosis in the United States due to the long- term survival.1 For patients with high-risk disease who
term sequelae of radiation. Long-term sequelae of cra- receive more intense therapy, the survival is only 50%
nial radiation include short stature, delayed learning to 78%.1,19 The 5-year survival for black children is only
deficits, thyroid dysfunction, obesity, and brain 64%.1
tumors.16-18 If a patient relapses after first-line therapy, a second
The current model of therapy for standard risk ALL remission may be induced in 82% to 95% of patients
includes induction, consolidation, 1 or 2 interim main- treated with an anthracycline, steroid, vincristine, and
tenance cycles, 1 or 2 delayed intensification cycles, and asparaginase/pegaspargase; however, the long-term
maintenance (Table 2). Standard 3-drug induction with prognosis is worse the second time around.20-21
vincristine, prednisone, or dexamethasone, and Matched sibling bone marrow transplantation (BMT) is
asparaginase achieves remission in 97% to 99% of chil- the treatment of choice for ALL in second remission.
dren. Patients also receive intrathecal chemotherapy. Five-year event-free survival after a matched sibling
Consolidation allows intensive treatment of the CNS to BMT in second remission is 40%, versus 17% with
prevent relapse. Delayed intensification is designed to chemotherapy alone.22 Patients who relapse more than

44 • JOURNAL OF PHARMACY PRACTICE 2002(15.1)


TREATMENT OF COMMON CHILDHOOD MALIGNANCIES

36 months after completion of initial therapy may mic blast cells in the bone marrow to less than 5%, and
benefit from chemotherapy alone, although allogeneic eliminate extramedullary disease.28 Current induction
transplantation is the best option, if available. Trans- regimens consist of two 4-day cycles of high-dose ther-
plantation during first remission is controversial. The apy separated by 1 week that allows recruitment and
exception is Philadelphia chromosome–positive pa- synchronization of blasts into S phase at the time of the
tients (3% to 5% of childhood ALL) who have a poor second pulse.34 These regimens consist of idarubicin or
prognosis and in whom benefit from matched related daunorubicin, etoposide, cytarabine, thioguanine, and
donor BMT in first remission has been demonstrated.23 dexamethasone. This requires a minimum of 1 month
Secondary acute myeloid leukemia occurs in about in the hospital.
5% of children following ALL treatment with high Following remission, it is recommended to proceed
doses and intensive schedules of etoposide or with a matched related BMT if a suitable donor is avail-
teniposide;24 therefore, these agents are no longer in- able. The 8-year survival with an allogeneic BMT is
cluded in the cooperative group trials. Corticosteroid 60% and only 53% with chemotherapy alone.35 If there
use has been associated with osteopenia, avascular ne- is no donor, then the patient proceeds with consolida-
crosis, and hyperglycemia.25 Native E coli asparaginase tion and intensification chemotherapy. Consolidation
causes hypersensitivity reactions in approximately with high-dose cytarabine has replaced maintenance
50% of ALL patients.26 therapy and increased survival at 5 years from 41%
In addition, anthracyclines used in high-risk or re- to 58%.36 Patients receiving autologous BMT do not
lapsed patients can cause cardiomyopathy. suffer from graft versus host disease; however, they
Doxorubicin may result in decreased left ventricular cannot benefit from the graft versus leukemia effect.
mass with subsequent increase in left ventricular Multiple studies have shown no benefit to autologous
afterload and decreased contractility. Congestive heart BMT.34,35,37,38
failure has been reported during or within 1 year of A role for monoclonal antibodies is currently being
therapy. Late cardiotoxic effects have been found 2 to sought for childhood AML. Gemtuzumab ozogamicin
15 years following therapy, with an incidence of 65% was recently marketed for the treatment of adult AML.
in survivors of childhood ALL.27 Risk factors include Gemtuzumab consists of a humanized anti-CD33 anti-
female sex, young age at diagnosis, rapid infusion rate, body linked to calicheamicin, a toxin. The antibody is
mantle irradiation, and cumulative dose greater than internalized after binding to CD33, which is expressed
400 mg/m2.27 on greater than 90% of childhood AML cells, and the
toxin is released intracellularly. The pediatric dose-
Acute Myelogenous Leukemia finding clinical trials are not yet completed, but there
are plans to include gemtuzumab ozogamicin in the
There is an increased incidence of Acute Myeloge- next randomized phase-3 pediatric AML clinical trial.
nous Leukemia (AML) in Hispanic children, patients With induction therapy, mortality risk is 10% to
who have received epidopophyllotoxins or alkylating 20%.28 The prolonged bone marrow suppression pre-
agents, and patients with trisomy 21, neuro- disposes patients to bacterial and fungal complications
fibromatosis Type I, or Kostmann syndrome.1,28 that are the leading cause of morbidity and mortality.
Children with Down syndrome have the best outcomes Viridans streptococcal infections are common in pa-
and now receive lower doses of chemotherapy agents tients receiving high-dose cytarabine.39 Prophylactic
secondary to excessive toxicity.29-31 Monosomy 7 or 7q penicillin and fluconazole have only short-lived suc-
deletions are associated with poor outcomes in both cess due to the development of resistance. Empiric
children and adults.32 Clinical trials have improved 5- amphotericin is started after only 3 days of persistent
year survival through incremental intensification of fever for these patients instead of the common 5 to 7
therapy from 17% in 1976 to 42% in 1996 for patients days. Many patients surviving childhood AML for
younger than 20 years of age.33 Children with Down more than 10 years developed growth abnormalities
syndrome have a 70% event-free survival at 5 years.30 (51%), endocrine dysfunction (16%), infertility (25%),
Unfortunately, treatment-induced morbidity limits fur- neurocognitive impairment (30%), cataracts (12%),
ther intensification of current therapy, and relapse rates and secondary malignant neoplasms (14%).40
remain unacceptable.
AML therapy consists of induction, consolidation, Hodgkin’s Disease
and intensification. Maintenance therapy is no longer
used in the United States. The goals of induction ther- In children younger than 15 years of age, Hodgkin’s
apy are to restore normal hematopoiesis, reduce leuke- disease (HD) is more common in males; it is more com-

JOURNAL OF PHARMACY PRACTICE 2002(15.1) • 45


BETSY M. BICKERT

mon in females after the age of 15 years. There is a bi- in males.1 Thirty-four percent of NHL cases are Bur-
modal age distribution, with peak incidence occurring kitt’s, 29% are lymphoblastic, and 27% are large cell.44
in the second and fifth decades of life in industrialized Two thirds of children have widespread disease at di-
countries.41 Clinical trials have improved 5-year sur- agnosis secondary to the rapid growth of NHL. Newer
vival for patients younger than 20 years of age with HD regimens have improved 5-year survival for NHL pa-
from 85% in 1976 to 93% in 1996.33 Since the survival tients younger than 20 years of age from 43% in 1976 to
rate is so high, the focus of most trials is to maintain ef- 74% in 1996; however, children with less extensive
ficacy while decreasing long-term sequelae, such as in- disease (that is, Stage I or II disease) have an 85% to
fertility and secondary malignancy. 95% long-term survival.33,44
Most pediatric protocols for HD utilize multiagent Chemotherapy is the main modality for treatment of
chemotherapy regimens with or without low-dose, NHL. Surgery is only used for diagnostic purposes un-
involved-field radiation. The addition of radiation in less there is an isolated gut mass. Radiation of the pri-
patients with bulky disease or B symptoms (that is, mary site is restricted to emergency situations.
high temperature for 3 days, drenching night sweats, Intrathecal therapy is an integral part of therapy to pre-
and ≥10% unexplained weight loss over 6 months) at vent or treat CNS disease for all types of childhood
diagnosis may reduce the risk of relapse and may allow NHL. Choice of chemotherapy differs by the cell of ori-
for fewer cycles of intensive chemotherapy. Adoles- gin, as discussed below. Large cell lymphoma has been
cents may receive high-dose radiation therapy for lo- treated with both strategies but only achieves long-term
calized disease. For low stage (I, II, IIIA) and favorable event-free survival in 50% to 70% of patients.44
disease, patients receive 4 to 6 courses of COPP (cyclo- B-cell lymphomas usually receive intensive, repeti-
phosphamide, vincristine, procarbazine, prednisone)/ tive therapy with alkylating agents and antimetabolites
ABV (doxorubicin, bleomycin, vinblastine) hybrid for 9 weeks to 8 months. Low-stage disease is treated
therapy and low-dose, involved field radiation.42 with COP (cyclophosphamide, vincristine, and predni-
Certain clinical risk factors predict a worse outcome. sone). Higher stage disease (that is, Stage III or IV) re-
These risk factors define unfavorable disease and in- ceives COP plus doxorubicin and high-dose
clude large mediastinal mass, massive splenic involve- methotrexate ± etoposide, ifosfamide, cytarabine, or
ment, extranodal involvement, elevated erythrocyte cisplatin.44 Current therapy aims to decrease duration
sedimentation rate, 3 involved lymph node areas, and of therapy, leading to decreased anthracycline-induced
anemia.42 Advanced-stage and unfavorable disease cardiomyopathy and alkylating agent-induced
have received MOPP-ABV (M = mechlorethamine) hy- sterility.
brid therapy with radiation.41 Currently, the BEACOPP T-cell lymphoblastic lymphomas are treated much
(bleomycin, etoposide, doxorubicin, cyclophos- like ALL for 12 to 32 months. Patients receive multiple
phamide, vincristine, prednisone, procarbazine) regi- agents in 3 phases: induction, consolidation, and main-
men shown to be effective in adults is being investi- tenance. Therapy in the United States is tailored after
gated in children.42 High-dose chemotherapy alone is the work of the BFM group. The NHL-BFM–90 regimen
usually avoided to prevent potential morbidity from treated patients with an 8 drug induction for 9 weeks,
myelosuppression, gonadal injury, and secondary ma- consolidation with high-dose methotrexate for 8
lignant neoplasms. Children treated with radiation ± weeks, and maintenance with oral methotrexate/
chemotherapy from 1935 until 1994 had an increased mercaptopurine to complete 24 months. High-risk pa-
risk of secondary malignancy 10 to 30 years following tients also received 8 weeks of intensification prior to
diagnosis.43 Attempts at decreasing the alkylating agent maintenance that included cranial radiation. The 5-
doses in order to decrease toxicity have resulted in in- year event-free survival was 90%.45 No patients died of
ferior survival rates.41 Cardiopulmonary toxicity has treatment-related toxicity, but one patient developed a
decreased secondary to careful monitoring and limita- secondary malignant neoplasm.
tion of cumulative doses of bleomycin and doxorubicin Patients with recurrent NHL and HD may respond to
in combination with radiation. DECAL (dexamethasone, etoposide, cisplatin,
cytarabine, and asparaginase). Sixty-five percent of HD
Non-Hodgkin’s Lymphoma patients and 42% of NHL patients had an initial re-
sponse, but the 5-year survival is only 26% and 23%,
The incidence of non-Hodgkin’s lymphoma (NHL) respectively.46 Late effects of NHL therapy include in-
begins to increase after 10 years of age, with a peak inci- fertility from alkylating agents, secondary malignant
dence at 15 to 19 years.1 Seventy percent of cases occur neoplasms from alkylating agents and radiation,

46 • JOURNAL OF PHARMACY PRACTICE 2002(15.1)


TREATMENT OF COMMON CHILDHOOD MALIGNANCIES

Table 3
47,55-57
Survival and Treatment of Common Pediatric Brain Tumors

Tumor Type 5-Year PFS Treatment Regimen


Standard risk PNET 60% Craniospinal XRT ± cisplatin/lomustine/vincristine
High-risk PNET 30% Triple peripheral blood stem cell transplant
Low-grade astrocytoma 70% XRT ± carboplatin/vincristine to delay XRT if <2 years
High-grade astrocytoma 20% Total resection is best; XRT + lomustine + vincristine
Brainstem glioma 5% 54-55 Gy XRT ± radiosensitizer such as topotecan
Ependymoma 30% Total resection is best therapy; cisplatin or cyclophosphamide
+ etoposide + vincristine + XRT
PFS = progression-free survival; PNET = primitive neuroectodermal tumor = medulloblastoma; XRT = radiation therapy

cardiomyopathy from doxorubicin, and radiation- short-lived depending on the areas resected, but pa-
induced growth abnormalities, renal dysfunction, or tients may experience permanent behavioral changes
pulmonary toxicity. or motor deficits. Radiation-induced injury is progres-
sive over time and correlates with the amount received,
SOLID TUMORS location of tumor, and age at time of treatment. There is
an inverse relationship between age and the severity of
Brain Tumors injury; thus, current treatment strategies include using
chemotherapy to delay radiation until the patient is
Central nervous system tumors are the second most older if possible.47 Brain irradiation is associated with
common malignancy of childhood. The incidence of hormonal deficits, cognitive deficits, hearing loss, and
invasive tumors is 18% higher in white than black chil- secondary malignant neoplasms. Spinal irradiation is
dren and more common in males than females.1 Brain associated with primary hypothyroidism, cardiac dam-
tumor incidence appears to have increased in the last age, restrictive pulmonary disease, and impaired verte-
20 years, but this may be the result of improved diag- bral growth. Some patients may benefit from
nostic imaging and not a true increase. Clinical trials methylphenidate 0.6 mg/kg/dose (maximum 20 mg) to
have improved 5-year survival for patients younger improve attention span.48 Less is known about long-
than 20 years of age from 56% in 1976 to 66% in 1996.33 term sequelae of the chemotherapy, but reports include
Treatment regimens depend on tumor pathology, tu- hearing loss from platinum agents, pulmonary fibrosis
mor location, and patient age. Certain tumors, such as from lomustine, and motor deficits from vincristine.
cerebellar pilocytic astrocytoma, may be cured by sur-
gical resection alone, whereas hypothalamic gliomas Ewing’s Sarcoma
require repeated resections. Others, such as germ cell
tumors, may respond to radiation therapy alone. Pa- The Ewing family of tumors may arise from either
tient age limits the amount of radiation a patient can re- bone or soft tissue of extremities or the central axis
ceive because younger patients are more susceptible to (45%).1 Ewing’s sarcoma (ES) is 9 times more common
the cognitive and developmental effects of in white than black children.1 Twenty percent to 25%
craniospinal irradiation. However, some remain inef- of patients have metastases at diagnosis, most com-
fectively treated using surgical resection, radiation, monly in the lung (50%), bone (25%), and bone mar-
and chemotherapy. Table 3 presents the most common row (25%).49 Clinical trials have improved 5-year sur-
brain tumors of childhood, their survival rate, and the vival for patients younger than 20 years of age with ES
regimen used most frequently following surgical resec- from 42% in 1975 to 58% in 1994.1
tion. The primary cause of treatment failure is the in- Surgical excision of the primary tumor is desirable
ability to achieve local control often due to surgical in- when in a feasible location. Radiation therapy is
accessibility. Strategies to improve medical therapy avoided in young children if possible due to its effects
include agents to disrupt the blood-brain barrier to in- on growth and a 10% to 20% incidence of secondary
crease the penetration of chemotherapy, antiangi- malignant neoplasms.49 However, local radiation is
ogenesis agents, and gene therapy. highly effective. Typical management consists of in-
Each treatment modality has associated short- and duction chemotherapy followed by local control (sur-
long-term side effects. Most neurosurgical deficits are gery and/or radiation) and then additional chemother-

JOURNAL OF PHARMACY PRACTICE 2002(15.1) • 47


BETSY M. BICKERT

apy. Standard chemotherapy consists of alternating mary histological subtypes are embryonal (75%) and
cycles of VAdriaC (vincristine, doxorubicin, cyclo- alveolar (worse prognosis). Twenty-nine percent of em-
phosphamide) ± (dactinomycin and ifosfamide/ bryonal RMS occurs in the head and neck region, 11%
etoposide). Womer and colleagues50 found that dose in- in the orbital, and 28% in the genital-urinary tract re-
tensification could be achieved by compressing the in- gion.1 Thirty-nine percent of alveolar RMS occurs in
terval between cycles of induction chemotherapy us- the extremities.1 Approximately 50% of patients have
ing filgrastim from 21 days to an average of 16 days readily resectable disease at diagnosis, and 15% have
during induction chemotherapy. This achieved a 73% metastatic disease.49 Clinical trials have improved 5-
event-free survival at 30 months. Long-term survival year survival for patients younger than 20 years old
for patients with metastatic disease remains a dismal with RMS from 25% in the 1960s to 75% in 1990.49
19% to 30%.49 Late effects include cardiomyopathy Primary tumor excision for RMS is desirable either
from doxorubicin ± chest irradiation, infertility, or de- before or after chemotherapy. Radiation therapy is
layed puberty secondary to cyclophosphamide/ highly effective for local control and administered to
ifosfamide, secondary malignant neoplasms, renal dys- virtually all patients once the tumor burden is de-
function from ifosfamide, and growth abnormalities or creased. If surgical resection is not possible due to tu-
functional defects at the primary site of disease. mor location, radiation may be administered prior to
chemotherapy. Chemotherapy is based on prognostic
Osteosarcoma factors such as tumor location, extent of disease, and
histology from the 4 International Rhabdomyosarcoma
Peak incidence for osteosarcoma (OS) is 15 years of Study Group trials from 1972 to 1998. Treatment lasts 8
age and corresponds to a time of rapid bone growth. OS to 12 months. Low-risk patients receive vincristine and
is more common in black than white children.1 Primary dactinomycin. Intermediate-risk patients receive VAC
sites are usually long bones, with 78% in the lower (vincristine, dactinomycin, cyclophosphamide). High-
limbs.1 Almost all patients have subclinical micro- risk patients have a 20% chance of long-term survival
scopic metastases at diagnosis, and 20% to 25% have following VAC with possible new agents or high-dose
detectable metastases (90% in the lung).49 Only 15% of chemotherapy and hematopoietic stem cell transplan-
patients with surgical resection alone will not have a tation. Late effects include infertility or delayed pu-
recurrence.49 Clinical trials have improved 5-year sur- berty secondary to cyclophosphamide, secondary ma-
vival for patients younger than 20 years of age with OS lignant neoplasms from cyclophosphamide or
from 20% in 1971 to 63% in 1994.1,51 radiation, cataracts from radiation to the head, and
Surgical excision of the primary tumor and meta- growth abnormalities or functional defects at the pri-
static lesions is crucial in OS due to the relative resis- mary site of disease.
tance to radiation. Amputation is often avoided in OS
in lieu of limb-salvage procedures with cadaveric bone Wilms Tumor
grafts, vascularized grafts, or prostheses following ef-
fective chemotherapy. Preoperative chemotherapy al- Wilms tumor composes 95% of renal malignancies
lows an assessment of early response from surgical in persons younger than 15 years of age.1 Approxi-
specimens. Chemotherapy continues after surgery and mately 500 cases are diagnosed annually in the United
consists of alternating regimens containing combina- States, with the highest incidence in children younger
tions of doxorubicin, high-dose methotrexate, ifos- than 5 years of age. Wilms tumor primarily occurs in
famide, and cisplatin. The use of high-dose only one kidney. Seven percent of patients have in-
methotrexate remains controversial.51 New agents, volvement of both kidneys, and 12% of patients have
such as MTP-PE, are targeting micrometastases.51 Late disease outside of the kidney, primarily in the lung.1
effects of therapy include cardiomyopathy from Survival and treatment depend on the stage and histol-
doxorubicin, secondary malignant neoplasms from ogy of the tumor. Clinical trials have improved 5-year
ifosfamide or radiation, and hearing loss, renal dys- survival for patients younger than 20 years of age with
function, and electrolyte wasting from cisplatin. Wilms tumor from 74% in 1976 to 93% in 1996.33
A summary of the results of the 4 completed Na-
Rhabdomyosarcoma tional Wilms Tumor Study (NWTS) Group trials is pre-
sented in Table 4. These trials defined a successful regi-
Rhabdomyosarcoma (RMS) accounts for more than men for low-stage disease and then refined the regimen
50% of soft-tissue sarcomas in children.49 The 2 pri- to maintain efficacy and decrease toxicity. Each trial

48 • JOURNAL OF PHARMACY PRACTICE 2002(15.1)


TREATMENT OF COMMON CHILDHOOD MALIGNANCIES

Table 4
58-59
Results of the National Wilms Tumor Study Group Trials

Study Results

NWTS 1 XRT not necessary for Group I <2 years; VCR + DAC > VCR = DAC. Preoperative chemotherapy had no effect on
(1969-1973) overall survival.
NWTS 2 6 months = 15 months chemo in Stage I; no postoperative XRT needed for FH Stage I/II; VCR + DAC + DOX > VCR
(1974-1978) + DAC for Stage II/III/IV FH disease.
NWTS 3 10 weeks = 24 weeks VCR + DAC in Stage I FH; DOX had no benefit on Stage II FH survival; no XRT = 20 Gy in
(1979-1986) Stage II FH; 10.8 Gy XRT = 20 Gy XRT in Stage III FH; DOX ↑ survival for Stage III FH; CPM did not ↑ survival
for Stage IV FH; CPM + VCR + DAC + DOX > VCR + DAC + DOX in Stages II-IV UH.
NWTS 4 6 months = 15 months chemo in Stage II/III/IV FH; pulse-intensive chemotherapy is equivalent to divided doses.
(1986-1994)
CPM = cyclophosphamide; DAC = dactinomycin; DOX = doxorubicin; FH = favorable histology; NWTS = National Wilms Tumor Study; UH = unfavorable histology;
VCR = vincristine; XRT = radiation.

Table 5
59
National Wilms Tumor Study Group 5 Treatment Regimens

Stage of Disease Chemotherapy Duration Radiation

Stage I/II FH, Stage I Anaplastic DAC + VCR, pulse-intensive 18 weeks None
Stage III/IV FH, Stage II-IV focal anaplasia DAC + VCR + DOX, pulse-intensive 24 weeks Abdominal; IV = abdomen + lungs
Stage II-IV diffuse anaplasia DAC + VCR + DOX + CPM + ETOP 24 weeks Tumor bed
CPM = cyclophosphamide; DAC = dactinomycin; DOX = doxorubicin; ETOP = etoposide; FH = favorable histology; VCR = vincristine. Prognosis: FH > focal
anaplasia > diffuse anaplasia.

has also improved success for patients with high-risk CONCLUSION


disease by intensifying therapy. Pulse-intensive therapy
(maximum tolerated doses at more frequent intervals) Extensive progress has been made in the treatment
yielded equal efficacy, increased dose intensity, de- of pediatric oncology patients. Pharmacists have a key
creased hematologic toxicity, fewer clinic visits, fewer role in ensuring patients receive the therapy outlined
days hospitalized than standard administration, and in the protocol. Having a basic understanding of the
consequently decreased cost.52 Following surgical re- common treatment strategies can facilitate this process.
section, children currently receive the treatment regi- Pharmacists can also play an integral role in the sup-
mens utilized in NWTS 5 as presented in Table 5. portive care used to treat and prevent adverse effects.
Overall, the therapy is quite effective with minimal Assisting the oncology team in making rational, cost-
late effects of treatment. Mantle radiation and effective choices and monitoring patients for adverse
doxorubicin are both associated with cardiomyopathy. effects over time can improve the care of these complex
Twenty years after diagnosis, the cumulative incidence patients.
of congestive heart failure for patients treated with
doxorubicin on NWTS 1 through 4 is 4.4%; however, it
Acknowledgments
is 17.4% among those treated with doxorubicin for
their first or subsequent relapse.53 Whole-lung radia-
I thank Dave Henry, MS, for his critical review of this
tion may decrease total lung capacity. Vincristine and
manuscript.
dactinomycin cause hepatotoxicity in 3% of recipients
of the combination.54 The combination of neph-
rectomy, local irradiation, and chemotherapy may REFERENCES
cause decreased renal function and hypertension. Pa-
tients receiving megavoltage radiation therapy and 1. Ries LAG, Smith MA, Gurney JG, et al, eds. Cancer Incidence
nephrectomy are at risk for spinal deformity. Ovarian and Survival among Children and Adolescents: United States
and testicular function are damaged by abdominal ra- SEER Program 1975-1995, National Cancer Institute, SEER Pro-
gram. NIH Pub. No. 99-4649. Bethesda, MD, 1999.
diation, and shielding should be used if possible. There 2. Shochat SJ, Fremgen AM, Murphy SB, et al. Childhood cancer:
is a 1.6% cumulative risk of developing a secondary patterns of protocol participation in a national survey. CA Can-
malignant neoplasm within 15 years of diagnosis.54 cer J Clin. 2001;51:119-130.

JOURNAL OF PHARMACY PRACTICE 2002(15.1) • 49


BETSY M. BICKERT

3. Felix CA, Lange BJ. Leukemia in infants. Oncologist. marrow relapse: a Pediatric Oncology Group study. Blood.
1999;4:225-240. 1988;72:1286-1292.
4. Biondi A, Cimino G, Pieters R, Pui CH. Biological and therapeu- 21. Leahey AM, Bunin NJ, Belasco JB, et al. Novel multiagent che-
tic aspects of infant leukemia. Blood. 2000;96(1):24-33. motherapy for bone marrow relapse of pediatric acute
5. Bleyer WA, Poplack DG. Prophylaxis and treatment of leukemia lymphoblastic leukemia. Med Pediatr Oncol. 2000;34:313-318.
in the central nervous system and other sanctuaries. Sem Oncol. 22. Barrett AJ, Horowitz MM, Pollock BH, et al. Bone marrow trans-
1985;12:131-148. plants from HLA-identical siblings as compared with chemo-
6. Pui CH, Simone JV, Hancock ML, et al. Impact of treatment therapy for children with acute lymphoblastic leukemia in a
intensification on acute lymphoblastic leukemia in children: second remission. N Engl J Med. 1994;331:1253-1258.
long-term results of St. Jude total therapy study X. Leukemia. 23. Arico M, Valsecchi MG, Camitta B, et al. Outcome of treatment
1992;6:150-157. in children with Philadelphia chromosome-positive acute
7. Henze G, Langermann HJ, Ritter J. Treatment strategy for differ- lymphoblastic leukemia. N Engl J Med. 2000;342:998-1006.
ent risk groups in childhood acute lymphoblastic leukemia: a 24. Pui C, Behm F, Raimondi S, et al. Secondary acute myeloid leu-
report from the BFM study group. Haematol Blut Transfus. kemia in children treated for acute lymphoid leukemia. N Engl J
1981;26:87-93. Med. 1989;321:136-142.
8. Gaynon PS, Steinherz PG, Bleyer WA, et al. Improved therapy 25. Mattano LA, Sather HN, Trigg ME, Nachman JB. Osteonecrosis
for children with acute lymphoblastic leukemia and unfavor- as a complication of treating acute lymphoblastic leukemia in
able presenting features: a follow-up report of the Children’s children: a report from the Children’s Cancer Group. J Clin
Cancer Study Group Study CCG 106. J Clin Oncol. Oncol. 2000;18(18):3262-3272.
1993;11:2234-2242. 26. Woo MH, Hak LJ, Storm MC, et al. Hypersensitivity or develop-
9. Tubergen DG, Gilchrist GS, O’Brien RT, et al. Improved outcome ment of antibodies to asparaginase does not impact treatment
with delayed intensification for children with acute outcome of childhood acute lymphoblastic leukemia. J Clin
lymphoblastic leukemia and intermediate presenting features: a Oncol. 2000;18(7):1525-1532.
Children’s Cancer Group Phase III trial. J Clin Oncol. 27. Lipschultz SE, Lipsitz SR, Mone SM, et al. Female sex and
1993;11:527-537. higher drug dose as risk factors for late cardiotoxic effects of
10. Lange BJ, Bostrom BC, Cherlow JM, et al. Double delayed inten- doxorubicin therapy for childhood cancer. N Engl J Med.
sification improves event-free survival for children with inter- 1995;332:1738-1743.
mediate risk acute lymphoblastic leukemia: a report from the 28. Langmuir PB, Aplenc RA, Lange BJ. Acute myeloid leukaemia
Children’s Cancer Group. Blood. In press. in children. Balliere’s Clin Haematol. 2001;14(1):77-93.
11. Nesbit M, Sather H, Robison L, et al. Randomized study of 3 29. Ravindranath Y, Abella E, Krischer J, et al. Acute myeloid leuke-
years vs 5 years of chemotherapy in childhood acute mia (AML) in Down’s syndrome is highly responsive to chemo-
lymphoblastic leukemia. J Clin Oncol. 1983;1:308-316. therapy: experience on pediatric oncology group AML study
12. Miller DR, Leikin SL, Albo VC, et al. Three versus five years of 8498. Blood. 1992;80:2210-2214.
maintenance therapy are equivalent in childhood acute 30. Craze JL, Harrison G, Wheatley K, et al. Improved outcome of
lymphoblastic leukemia: a report from the Children’s Cancer acute myeloid leukemia in Down’s syndrome. Arch Dis Child.
Study Group. J Clin Oncol. 1989;7:73-79. 1999;81:32-37.
13. Gayon PS,Carrel AL. Glucocorticosteroid therapy in childhood 31. Lange BJ, Kobrinsky N, Barnard DR, et al. Distinctive demogra-
acute lymphoblastic leukemia. Adv Exp Med Biol. phy, biology, and outcome of acute myeloid leukemia and
1999;457:593-605. myelodysplastic syndrome in children with Down syndrome:
14. Smith M, Arthur D, Camitta B, et al. Uniform approach to risk Children’s Cancer Group Studies 2861 and 2891. Blood.
classification and treatment assignment for children with acute 1998;91:608-615.
lymphoblastic leukemia. J Clin Oncol. 1996;14:18-24. 32. Woods WG, Nesbit ME, Buckley J, et al. Correlation of chromo-
15. Conter V, Arico M, Valsecchi MG, et al. Extended intrathecal some abnormalities with patient characteristics, histologic sub-
methotrexate may replace cranial irradiation for prevention of type, and induction success in children with acute
CNS relapse in children with intermediate-risk acute nonmyelocytic leukemia. J Clin Oncol. 1985;3(1):3-11.
lymphoblastic leukemia treated with Berlin-Frankfurt-Munster- 33. Ries LAG, Eisner MP, Kosary CL, et al, eds. SEER Cancer Statis-
based intensive chemotherapy. J Clin Oncol. 1995;13:2497- tics Review, 1973-1997, National Cancer Institute. NIH Pub. No.
2502. 00-2789. Bethesda, MD, 2000.
16. Meadows A, Massari D, Fergusson J, et al. Declines in IQ scores 34. Woods WG, Kobrinsky N, Buckley JD, et al. Timed-sequential
and cognitive dysfunctions in children with acute induction therapy improves postremission outcome in acute
lymphoblastic leukemia treated with cranial irradiation. Lancet. myeloid leukemia: a report from the Children’s Oncology
1981;10:1015-1018. Group. Blood. 1996;87:4979-4989.
17. Neglia J, Meadows A, Robison L, et al. Second neoplasms after 35. Woods WG, Neudorf S, Gold S, et al. A comparison of allogeneic
acute lymphoblastic leukemia in children. N Engl J Med. bone marrow transplantation, autologous marrow transplanta-
1991;325:1330-1336. tion, and aggressive chemotherapy in children with acute
18. Relling RV, Rubnitz JE, Rivera GK, et al. High incidence of sec- myeloid leukemia in remission: a report from the Children’s
ondary brain tumours after radiotherapy and antimetabolites. Cancer Group. Blood. 2001;97:56-62.
Lancet. 1999;354:34-39. 36. Wells R, Woods W, Lampkin B, et al. Impact of high-dose
19. Nachman JB, Sather HN, Sensel MG, et al. Augmented post- cytarabine and asparaginase intensification on childhood acute
induction therapy for children with high-risk acute myeloid leukemia: a report from the Children’s Cancer Group
lymphoblastic leukemia and a slow response to initial therapy. 213P. J Clin Oncol. 1993;11(3):538-545.
N Engl J Med. 1998;338:1663-1671. 37. Ravindranath Y, Yeager AM, Chang MN, et al. Autologous bone
20. Buchanan GR, Rivera GK, Boyett JM, et al. Reinduction therapy marrow transplantation versus intensive consolidation chemo-
in 297 children with acute lymphocytic leukemia in first bone

50 • JOURNAL OF PHARMACY PRACTICE 2002(15.1)


TREATMENT OF COMMON CHILDHOOD MALIGNANCIES

therapy for acute myeloid leukemia in childhood. Pediatric Oncol- 48. Thompson SJ, Leigh L, Christensen R, et al. Immediate
ogy Group. N Engl J Med. 1996;334:1428-1434. neurocognitive effects of methylphenidate on learning-impaired
38. Michel G, Leverger G, Leblanc T, et al. Allogeneic bone marrow survivors of childhood cancer. J Clin Oncol. 2001;19:1802-1808.
transplantation versus aggressive post-remission chemotherapy for 49. Arndt CAS, Crist WM. Common musculoskeletal tumors of child-
children with acute myeloid leukemia in first complete remission. A hood and adolescence. N Engl J Med. 1999;341:342-352.
prospective study from the French Society of Pediatric Hematology 50. Womer RB, Daller RT, Fenton JG, Miser JS. Granuloxyte colony stim-
and Immunology (SHIP). Bone Marrow Transplant. 1996;17:191- ulating factor permits dose intensification by interval compression
196. in the treatment of Ewing’s sarcomas and soft tissue sarcomas in chil-
39. Weisman SJ, Scoopo FJ, Johnson GM, et al. Septicemia in pediatric dren. Eur J Cancer. 2000;36:87-94.
oncology patients: the significance of viridans streptococcal infec- 51. Bramwell VHC. The role of chemotherapy in osteogenic sarcoma.
tions. J Clin Oncol. 1990;8:453-459. Crit Rev Oncol Hematol. 1995;20:61-85.
40. Leung W, Hudson MM, Strickland DK, et al. Late effects of treatment 52. Green DM, Breslow NE, Evans I, et al. The effect of chemotherapy
in survivors of childhood acute myeloid leukemia. J Clin Oncol. dose intensity on the hematological toxicity of the treatment for
2000;18:3273-3279. Wilms’ tumor: a report from the National Wilms’ Tumor Study. Am J
41. Hudson MM, Donaldson SS. Hodgkin’s disease. Pediatr Clin North Pediatr Hematol Oncol. 1994;16(3):207-212.
Am. 1997;44:891-907. 53. Green DM, Grigoriev YA, Nan B, et al. Congestive heart failure after
42. Josting A, Wolf J, Diehl V. Hodgkin disease: prognostic factors and treatment for Wilms’ tumor: a report from the National Wilms’
treatment strategies. Curr Opin Oncol. 2000;12:403-411. Tumor Study Group. J Clin Oncol. 2001;19:1926-1934.
43. Metayer C, Lynch CF, Clarke EA, et al. Second cancers among long- 54. Green DM, Donckerwolcke R, Evans AE, D’Angio GJ. Late effects of
term survivors of Hodgkin’s disease diagnosed in childhood and treatment for Wilms Tumor. Hematol Oncol Clin North Am.
adolescence. J Clin Oncol.. 2000;18:2435-2443. 1995;9(6):1317-1327.
44. Sandlund JT, Downing JR, Crist WM. Non-Hodgkin’s lymphoma in 55. Albright AL. Pediatric brain tumors. CA Cancer J Clin. 1993;43:272-
childhood. N Engl J Med 1996;334:1238-1248. 288.
45. Reiter A, Schrappe M, Ludwig WD, et al. Intensive ALL-type therapy 56. Freeman CR, Perilongo G. Chemotherapy for brain stem gliomas.
without local radiotherapy provides a 90% event-free survival for Child Nerv Syst. 1999;15:545-553.
children with T-cell lymphoblastic lymphoma: a BFM group report. 57. Bouffet E, Foreman N. Chemotherapy for intracranial ependy-
Blood. 2000;95:416-421. momas. Child Nerv Syst. 1999;15:563-570.
46. Kobrinsky NL, Sposto R, Shah NR, et al. Outcomes of treatment of 58. Green DM, Thomas PRM, Shochat S. The treatment of Wilms tumor:
children and adolescents with recurrent non-Hodgkin’s lymphoma results of the National Wilms Tumor Studies. Hematol Oncol Clin
and Hodgkin’s disease with dexamethasone, etoposide, cisplatin, North Am. 1995;9(6):1267-1274.
cytarabine, and L-asparaginase, maintenance chemotherapy, and 59. Neville HL, Ritchey ML. Wilms’ tumor: overview of National Wilms’
transplantation: Children’s Cancer Group Study 5912. J Clin Oncol. Tumor Study Group results. Urol Clin North Am. 2000;27(3):435-
2001;19:2390-2396. 438.
47. Siffert J, Greenleaf M, Mannis R, Allen J. Pediatric brain tumors.
Child Adol Pysch Clin North Am. 1999;8(4):879-903.

JOURNAL OF PHARMACY PRACTICE 2002(15.1) • 51

Vous aimerez peut-être aussi