You are on page 1of 8

Indian J Pediatr (September 2015) 82(9):817824

DOI 10.1007/s12098-015-1695-5

REVIEW ARTICLE

Guest Editor: Bhim S. Pandhi

Leukemias in Children
Rachna Seth & Amitabh Singh

Received: 6 June 2014 / Accepted: 7 January 2015 / Published online: 15 February 2015
# Dr. K C Chaudhuri Foundation 2015

Abstract Childhood cancers are rare but an important cause of the unregulated proliferation of the malignant clone and bone
morbidity and mortality in children younger than 15y of age. marrow failure.
Common childhood malignancies include leukemias There are two main subtypes of acute leukemias in chil-
(commonest, 3040%), brain tumors (20%) and lymphoma dren; the commoner acute lymphoblastic leukemia (ALL) and
(12%) followed by neuroblastoma, retinoblastoma and tumors acute myeloid leukemia (AML). Acute leukemia constitutes
arising from soft tissues, bones and gonads. Leukemias, the about 40% of all childhood cancers. Chronic leukemias are
commonest childhood cancer, arise from clonal proliferation more common in adults than in children. They tend to grow
of abnormal hematopoietic cells leading to disruption of normal more slowly than acute leukemias and are harder to treat.
marrow function and marrow failure. The various clinical man- Chronic leukemias are divided into two main types: chronic
ifestations of leukemia result from unregulated proliferation of myelogenous leukemia (CML) and chronic lymphocytic leu-
the malignant clone and bone marrow failure. There are two kemia (CLL). CML is rare in children and is treatable as in
main subtypes, the commoner, acute lymphoblastic leukemia adults. CLL on the other hand is extremely rare in children.
(ALL) and acute myeloid leukemia (AML). A small proportion Juvenile myelomonocytic leukemia (JMML) is a form of leu-
may have chronic myeloid leukemia (CML) and juvenile kemia that is neither chronic nor acute and occurs most often
myelomonocytic leukemia (JMML). A systematic approach is in children under the age of four. JMML begins from myeloid
necessary for diagnosis. Treatment should be initiated as early cells, but is not as fast-growing as AML or as slow as CML.
as possible to avoid complications. A timely referral to a cancer
center must be done if facilities for diagnosis/treatment, man-
agement of complications and provision for supportive care are Acute Lymphoblastic Leukemia (ALL)
not available at the treating center.
ALL is the most common childhood malignancy accounting
Keywords Leukemia . Children for one-fourth of all childhood cancers and three-fourths of all
newly diagnosed patients with acute leukemia [1]. Its inci-
dence is approximately 34 cases per 100,000 children below
15y of age in USA. In India, leukemia is the most common
childhood cancer (2540%) with higher proportion of T cells
Introduction (2050%) compared to west (1020%) [2]. Boys have higher
rates than girls, especially in adolescents with T-cell ALL [3].
Leukemia is the most common cancer in children. It is a ma- The etiology of ALL remains unknown in a majority of cases.
lignancy that arises from proliferation of hematopoietic cells However, several genetic syndromes like Down syndrome have
leading to disruption of normal marrow function and marrow been associated with an increased risk of leukemia [1, 4, 5].
failure. The clinical manifestations of leukemia are result of
Morphology
R. Seth (*) : A. Singh
Division of Oncology, Department of Pediatrics, All India Institute of
Medical Sciences, Ansari Nagar, New Delhi 110029, India The classification of ALL is based on morphology, cytochem-
e-mail: drrachnaseth@yahoo.co.in istry, immunephenotyping, karyotyping and molecular
818 Indian J Pediatr (September 2015) 82(9):817824

biology techniques. ALL cells can be classified using the Table 1 Co-relation of ALL subtypes with surface markers
French-American-British (FAB) criteria into morphologic Pro B/Early Pre B CD34+/CD19+
subtypes. L1 morphology lymphoblasts are the most common
subtype (8085%), and are associated with a better prognosis. Pre B cell CD79a, CD10+, Presence of CD10
L2 subtype accounts for 15% cases. Only 12% patients with CD18+,CD19+, (common ALL
CD20+,CD21+, antigen-CALLA)
ALL show L3 morphology. The lymphoblast has high HLADR+ represents a
nuclear/cytoplasmic (N/C) ratio, clumped chromatin com- favorable prognosis
pared to the myeloblast which has low N/C ratio and a spongy Absence of CD10 is
nuclear chromatin. Myeloperoxidase positivity is characteris- associated with MLL
translocations, particularly
tic of myeloblast (Fig. 1). t(4;11) and a poor
outcome.
Immunophenotype Mature B cell CD19+,CD20+, Correlates with L3
CD21+,sIg+ leukemia -Burkitts
leukemia, needs
Immunophenotype classification describes ALL as either B
intensified regime
cell derived or T cell derived. 8085% ALL are progenitor B
Pre T cell CD 2+/CD5+/CD8+
cell derived, 15% are derived from T cells and 12% from
T cell CD3+,CD5+, Associated with older
mature B cells (Table 1). CD7+,CD2 age, high initial
white cell count,
Cytogenetics mediastinal mass
and risk for CNS
involvement of disease
Genetic abnormalities found in the leukemic clone greatly
impact the therapy and prognosis of ALL. Conventional cy-
togenetics and fluorescence in-situ hybridization (FISH) The most common cytogenetic abnormalities in B cell ALL
should be performed on the bone marrow specimen to look are hyperdiploidy25 %, t(12;21)25%, MLL5 %, hypodip-
for common genetic alterations in ALL. These should be per- loidy15 % and t(9;22)4%.
formed for diagnostic as well as follow-up specimens during Mature B cell ALL has poorer prognosis than earlier B
treatment. The presence of hyperdiploidy (chromosome num- lineage subgroups but distinction between pre B cell and early
ber >50, DNA index >1.16) is associated with good prognosis pre B cell is not relevant from prognosis point of view.
in contrast to the poor prognosis in patients with hypodiploidy
(chromosome number <45 per cell) [6]. Specific chromosom-
al translocations in ALL, including t (8; 14, associated with Prognostic Factors and Risk Assessment
Burkitt leukemia) in B-cell ALL, t(4;11) in infant leukemia
and t(9;22) translocation, that forms the Philadelphia chromo- The improvement in survival in ALL is partly due to recogni-
some, are associated with a poor prognosis. Certain chromo- tion that all cases are not same. Risk adapted therapy of ALL,
somal abnormalities are associated with a favorable prognosis recognizing standard, intermediate and high risk groups has led
like t(12;21) and simultaneous presence of trisomy 4 and 10. to different treatment approaches. Host factors like age, sex,
ethnicity and disease characteristics like initial total white blood
count, immunophenotype, cytogenetic abnormality and treat-
ment response are used to define these three groups. The two
most important prognostic factors include age at diagnosis and
the initial leukocyte count. Children less than 1-y-old have an
unsatisfactory prognosis; infant leukemia is often associated
with t(4;11) translocation and high leukocyte counts. Children
between the ages of 1- and 9-y do well. The presence of leu-
kocyte count more than 50,000/ cu mm at diagnosis is associ-
ated with a bad prognosis. Relapse rates are higher in boys.
Patients showing hyperdiploidy have a good prognosis, while
presence of hypodiploidy is associated with an unsatisfactory
Fig. 1 Bone marrow from a child with acute lymphoblastic leukemia outcome. Philadelphia positive t(9;22) ALL and translocation
shows reduced normal marrow elements which are replaced by t(4;11) which is present in infant leukemia are associated with
lymphoblasts. The neoplastic lymphoblasts are slightly larger than
lymphocytes and have round or convoluted nuclei, fine chromatin,
poor prognosis. Response to treatment with prednisone is con-
often with a smudged appearance, inconspicuous nucleoli and scant, sidered a prognostic factor; patients showing 1000/cu mm
faintly basophilic cytoplasm blasts in peripheral blood following 7d treatment with
Indian J Pediatr (September 2015) 82(9):817824 819

prednisone and an intrathecal dose of methotrexate are likely to Central nervous system involvement, diagnosed by CSF
have an adverse outcome [7, 8]. Treatment response is influ- examination is rare (25%) at diagnosis; and may present with
enced by the drug sensitivity of leukemic cells and host phar- features of raised intracranial pressure. Overt testicular leuke-
macodynamics and pharmacogenomics (Table 2). mia presenting as firm, painless, unilateral or bilateral swell-
ing may be seen in about 1% of cases. Other rare sites of
extramedullary involvement include heart, lungs, kidneys,
Clinical Presentation ovaries, skin, eye or the gastrointestinal tract.

The clinical features of ALL are attributed to bone marrow Diagnosis and Differential Diagnosis
infiltration with leukemic cells (bone marrow failure) and
extramedullary involvement. Common features include man- Children may present with pancytopenia or
ifestations of anemia, thrombocytopenia and neutropenia. hyperleukocytosis. The diagnosis is confirmed by peripheral
These include pallor and fatigue, petechiae or purpura and smear examination and/or bone marrow aspirate / biopsy.
infections. Lymphadenopathy, hepatomegaly and splenomeg- Higher white blood cell counts are more common with T-
aly are present in more than 60% patients. Bone or joint pain cell ALL. The bone marrow smear where >25% of bone mar-
and tenderness may occur due to leukemic involvement of the row cells are leukemic lymphoblasts, is diagnostic for ALL.
periosteum of bones or joints. Infants and young children may While morphology of the leukemic blasts can give important
present with a limp or refusal to walk. Tachypnea and respi- clues to the diagnosis, it needs to be confirmed by
ratory distress may be present secondary to severe anemia immunephenotyping of the bone marrow. Immunophenotype
leading to congestive heart failure or secondary to the pres- differentiates the cellular lineages of ALL into pre-B, T cell
ence of mediastinal mass leading to tracheal compression (su- and mature B cell. This distinction has therapeutic implica-
perior mediastinal syndrome). A large mediastinal mass may tions. Evaluation of CSF for presence of leukemic blasts to
sometimes cause superior vena cava syndrome with facial determine CNS involvement is important for staging of leu-
edema and plethora, throbbing headache, conjunctival con- kemia. The first spinal tap must be performed ideally with
gestion and dilated neck veins. Patients with high tumor bur- platelet count close to 100,000/cu mm. Children with CNS
den can occasionally present with very high total white cell leukemia require more intensive CNS directed therapy
count (hyperleukocytosis, TLC >1,00,000/cu mm) or tumor The clinical profile of acute lymphoblastic leukemia
lysis syndrome with decreased urine output and azotemia sec- may mimic many other clinical conditions like infectious
ondary to uric acid nephropathy. The elevated number of leu- mononucleosis, acute infectious lymphocytosis, idiopathic
kemia blast cells in hyperleukocytosis may sludge in the vas- thrombocytopenic purpura, aplastic anemia and viral in-
cular supply to brain, lungs, and liver producing symptoms of fections like cytomegalovirus and others that result in leu-
organ dysfunction. kemoid reactions and pancytopenia. Idiopathic thrombo-
cytopenic purpura (ITP) is the most common cause of
Table 2 Prognostic features in acute lymphoblastic leukemia
acute onset of petechiae and purpura in children.
Feature Standard risk High risk Ordinarily children with ITP have no evidence of anemia
and have normal total and differential leukocyte count.
Age 210 y Less than 1 y Bone marrow smear reveals normal hematopoiesis and
Greater than 10 y
normal or increased number of megakaryocytes. A bone
Sex Female Male
marrow examination is recommended prior to initiation of
Initial white cell count <50,000/cu mm >50,000/cu mm
steroid treatment for presumed ITP, in order to exclude
Hepatosplenomegaly Absent Massive leukemia. ALL must be differentiated from aplastic ane-
Lymphadenopathy Absent Massive mia, juvenile rheumatoid arthritis and other malignancies
Mediastinal mass Absent Present (Neuroblastoma, Non-Hodgkin lymphoma,
Central nervous Absent Present Rhabdomyosarcoma, Ewing sarcoma and
system (CNS) leukemia
Phenotype Pre B (T- cell Mature B - cell Retinoblastoma). Morphologic, cytochemical,
intermediate) immunophenotypic and cytogenetic characteristics of the
Ploidy Hyperdiploidy Hypodiploidy malignant cells should be done. Occasionally, patients
Cytogenetics t(12;21),trisomy t(9;22), with ALL may present with hypereosinophilia or as an
4 and 10 t(4;11),t(8;14) emergency with very high white cell count
Response to treatment Good early response Poor early (hyperleukocytosis, TLC>100,000/cu mm), life threaten-
response
ing infections, hemorrhage, organ dysfunction secondary
Minimal residual disease Negative Positive
(MRD) after first induction
to leukostasis or signs and symptoms of superior vena
cava/superior mediastinal syndrome.
820 Indian J Pediatr (September 2015) 82(9):817824

Management and magnitude of disappearance of peripheral blasts after pro-


phase is highly predictive of outcome.
Improvement in supportive care and use of combination che-
motherapy has led to survival rate of more than 80% overall CNS Preventive Therapy
and greater than 95% in children with low risk disease, in
western countries. Treatment is determined by the risk of re- The concept of CNS preventive therapy is based on the fact
lapse in each patient. that most children with leukemia have subclinical CNS in-
Risk based approach allows use of modest therapy for chil- volvement at the time of diagnosis and this acts as a sanctuary
dren who have historically had very good outcome thereby site where leukemic cells are protected from systemic chemo-
avoiding the toxic adverse effects of high intensity therapy. therapy because of the blood brain barrier. The early institu-
The three most important determinants of this risk are age at tion of CNS prophylaxis is essential to eradicate leukemic
presentation, total WBC count at presentation and response to cells which have passed the blood brain barrier. CNS prophy-
initial therapy. laxis has enabled increased survival rates in leukemia. Most
The successful treatment of ALL requires the control of children in the past received a combination of intrathecal
bone marrow or systemic disease, as well as treatment (or methotrexate and cranial irradiation. However, there is consid-
prevention) of extramedullary disease in sanctuary sites, par- erable concern regarding long term neurotoxicity and risk of
ticularly the central nervous system (CNS). The cornerstone development of brain tumors following this therapy. In order
of this strategy is systemic combination chemotherapy togeth- to achieve effective CNS prophylaxis while minimizing neu-
er with CNS prophylaxis. Different centers use different pro- rotoxicity, experts now recommend a lower dose of cranial
tocols for childhood ALL. Most European and American pro- irradiation (1800cGy) with intrathecal methotrexate.
tocols use aggressive chemotherapy regimens with 5y survival Intrathecal methotrexate, given periodically starting from
rates above 8085% [9]. day 1 of induction, provides good CNS protection and has
The treatment of ALL is divided into 4 stages. (i) Induction good prognosis. Other alternative regimens for CNS prophy-
therapy (to attain remission) (ii) CNS prophylaxis or CNS laxis include the use of triple intrathecal therapy consisting of
preventive therapy, (iii) Intensification (consolidation) and methotrexate, hydrocortisone and cytarabine without cranial
(iv) Maintenance therapy (continuation). The intensification irradiation. Others propose that cranial irradiation should be
(consolidation) phase, following induction of remission, may limited for patients with high risk features at diagnosis, includ-
not be required in low risk patients, though recent studies ing T-cell ALL with leukocyte count >1000,000/cu mm,
suggest benefits in long-term survival with intensification Philadelphia chromosome positive and presence of CNS
therapy in both low risk and high risk patients. The average leukemia.
duration of treatment in ALL ranges between 2 and 2.5 y.
Intensification (Consolidation) Therapy
Induction Therapy
This is a period of intensified treatment, administered shortly
The goal of this phase is to eradicate leukemia from the bone after remission induction with administration of new chemo-
marrow such that at end of this phase there are <5% leukemic therapeutic agents to tackle the problem of drug resistance
blasts in the bone marrow by morphology. Patients who [10]. Commonly used agents include high dose methotrexate,
achieve rapid early remission (<5% blasts in marrow) by L-asparginase, epipodophyllotoxin, cyclophosphamide and
day 7 or 14 of induction have a better prognosis than slow cytarabine.
responders. Failure to achieve this at end of induction is asso-
ciated with high risk of relapse. Induction therapy generally Maintenance (Continuing) Therapy
consists of 46 wk of therapy and current induction regimens
include vincristine, prednisone, L-asparaginase and an It has been estimated that approximately two to three logs of
anthracycline, with remission achieved in 9598% of cases. leukemic blasts are killed during the induction therapy, leav-
Basic two drug regimen of vincristine and steroid achieves ing a leukemic cell burden in the range of 10 9 10 10 .
remission in 85% of children and addition of L-asparginase, Additional therapy is therefore necessary to prevent a relapse.
an anthracycline or both improves remission induction to Once remission is achieved, maintenance therapy is continued
95%. Because of the complications related with for an additional 22.5 y. The main agents used include 6-
anthracyclines, some protocols use four drug induction for mercaptopurine daily and methotrexate once a week given
high risk patients only. Three drug induction i.e., steroid, vin- orally, with or without pulses of vincristine and prednisone
cristine and L-asparginase is used for standard risk patients in or other cytostatic drugs. Monthly pulses of vincristine and
these protocols. Some protocols also use seven day predniso- prednisolone appear to be beneficial in terms of better event
lone prophase along with intrathecal methotrexate. The rate free survival. For better outcome, methotrexate and 6-
Indian J Pediatr (September 2015) 82(9):817824 821

mercaptopurine should be given to the limits of tolerance as More than 80% of children with ALL are long term
determined by absolute neutrophil counts. survivors in the developed countries. However, survival
Philadelphia chromosome positive (ph+) ALL requires remains poor in the developing nations. Several factors
addition of tyrosine kinase inhibitor (TKI) like imatinib to are responsible for this difference. There is increased
chemotherapy regimen. Second generation TKIs, e.g., infection related mortality due to lack of adequate sup-
dasatinib and nilotinib, show activity against most of the portive care. Malnutrition also increases mortality in
bcr-abl tyrosine kinase domain (TKD) mutations involved children with ALL. Also, there is higher incidence of
in acquired imatinib resistance, but clinical benefit is gen- T-cell ALL in India leading to poorer survival.
erally short-lived. Accordingly, stem cell transplantation
(SCT) in first complete remission (CR) is considered to Treatment after Relapse
be the best curative option.
Stem cell transplantation, also called bone marrow trans- Despite the success of modern treatment, 2030% of
plantation (BMT), is performed only in patients who have children with ALL relapse [13]. The main cause of
abnormal cytogenetic, or high-risk ALL features in first or treatment failure in leukemia is relapse of the disease.
second remission. Cytogenetics is the most important compo- The most common site of relapse is in the bone marrow
nent of deciding whether or not a person should have a bone (20%), followed by CNS (5%) and testis (3%). The
marrow transplant for ALL. Patients with the Philadelphia prognosis for a child with ALL who relapses depends
chromosome positive ALL or young infants with the translo- on the site and time of relapse. Early bone marrow
cation involving chromosomes 4 and 11(MLL gene rearrange- relapse before completing maintenance therapy has the
ment), are candidates for BMT. worst prognosis and longtime survival of only 1020%
while late relapses occurring after cessation of mainte-
nance therapy have a better prognosis (3040% surviv-
Supportive Care
al). Relapse in extramedullary sites, particularly testes,
is more favorable in terms of survival. The treatment of
Because of the potential complications encountered with treat-
relapse must be more aggressive than the first line ther-
ment and the need for aggressive supportive care like blood
apy with use of new drugs to overcome the problem of
component therapy, monitoring and management of tumor
drug resistance.
lysis syndrome, detection and management of infections, nu-
Allogenic bone marrow transplantation offers a better
tritional and metabolic needs and psychosocial support, these
chance of cure than conventional chemotherapy for chil-
children should be treated by a specialist in a cancer center or
dren with ALL who enter a second remission after he-
hospital with all support facilities. These children should be
matologic relapse. In addition to the treatment related
given cotrimoxazole as prophylaxis against Pneumocystis
morbidity and mortality, lack of a suitable matched do-
carinii pneumonia. They should be vaccinated against hepati-
nor limits widespread application of allergenic transplan-
tis B infection and screened for HIV infection. Oral hygiene
tation. Facilities for bone marrow transplantation are
should be taken care of. Facilities for blood component ther-
available in all major centers in India including
apy should be available [11]. The survival rates of children
Bombay, Vellore, Chennai and Delhi.
with malignancies are enhanced through access to state-of-
the-art treatment given according to well defined protocols
Late Effects of Treatment
in specialized centers.
Long term effects of treatment, which may take years to
Prognosis become apparent, are of particular concern. Continued
evaluation of these patients for prolonged periods is
Hypodiploidy, Philadelphia chromosome positivity, T-cell necessary. Patients who have received cranial irradiation
ALL, MLL rearrangement, IKZF1 gene deletion, age <1y at a younger age are at risk for cognitive and intellec-
and >10y, leukocyte count >50,000/cu mm and presence of tual impairment [14] and development of CNS neo-
CNS disease are poor prognostic features. plasms. There is a risk of development of secondary
Assessment of minimal residual disease (MRD) by PCR AML after the intensive use of epipodophyllotoxin
assay using immunoglobulin/ T- cell receptor gene rearrange- (etoposide or teniposide) therapy [15]. Endocrine dys-
ments or by flow cytometry has been shown to be an impor- function leading to short stature, obesity [16], preco-
tant determinant of outcome. These methods can detect one cious puberty, osteoporosis, thyroid dysfunction and
leukemic cell in 10,000 to 100,000 normal cells. Patients with growth retardation (growth hormone deficiency) are re-
MRD <0.01% on day 29 of induction are at low risk of relapse ported. Patients having received treatment with an
[12]. anthracycline are at risk of cardiac toxicity [17, 18].
822 Indian J Pediatr (September 2015) 82(9):817824

Acute Myeloid Leukemia neutropenia. Unlike ALL, lymphadenopathy and massive


hepatosplenomegaly is not very common in AML. However,
Acute myeloid leukemia (AML) or acute non-lymphoblastic infants and toddlers with AML have more organomegaly, high
leukemia, the second most common type of leukemia in chil- leukocyte counts and CNS disease at diagnosis. Disseminated
dren, accounts for 1520% of leukemia in children. AML intravascular coagulation may occur with any subgroup, but is
results from clonal proliferation of hematopoietic precursors common in acute promyelocytic leukemia (M3). Chloromas
of myeloid, erythroid and megakaryocytic lineage. Intensive are localized collections of leukemic cells seen exclusively in
myelosuppressive induction and post-remission therapy en- patients with AML. They may occur at any site including
ables long term survival in 4050% patients only. CNS, neck, bones (typically orbit) and skin (Fig. 3).
Gingival hypertrophy may be present (Fig. 4). Patients with
Epidemiology high WBC count may present with signs of leukostasis such as
pulmonary infiltrates causing respiratory distress or stroke.
AML can occur at any age but the incidence is more during Central nervous system involvement may occur in up to
adolescence; males are affected as frequently as females. 15% of patients.
Congenital leukemia (occurring during first 4wk of life) is Diagnosis is ascertained as in acute lymphoblastic leuke-
mostly AML. While the etiology of AML is not known, there mia by a peripheral smear and bone marrow examination.
is an association following exposure to ionizing radiation.
Down syndrome is the most common genetic predisposing
factor associated with risk of developing AML during the first Treatment
three years of life.
The long term survival for children with AML has increased
Classification from less than 10% to almost 50% during the past two de-
cades. This is due to intensification of therapy along with
AML can be divided into several subgroups according to the improved supportive care. However compared to ALL, the
FAB classification: M0, undifferentiated; M1, acute myelo- cure rate is hampered by a lower remission rate, an increased
blastic leukemia with minimal maturation; M2, acute myelo- relapse rate due to the development of resistance to multiple
blastic leukemia with maturation; M3, acute promyelocytic chemotherapeutic drugs and a greater risk of death in remis-
leukemia; M4, acute myelomonocytic leukemia; M5, acute sion due to infections and hemorrhage [19]. The main drugs
monoblastic leukemia; M6, erythroblastic leukemia and M7, used for induction therapy are combination of cytosine arabi-
acute megakaryoblastic leukemia (Fig. 2). Several genetic ab- noside and an anthracycline (doxorubicin or daunorubicin)
normalities have been identified in AML. with or without additional drugs (etoposide, thioguanine).
Post remission therapy (consolidation therapy) includes high
dose chemotherapy including cytosine arabinoside and
Clinical Features
etoposide. Risk based approach is also used for treatment of
AML. Various prognostic factors in childhood AML have
The clinical presentation of AML is similar to ALL but it is
been identified: older age, obesity, M0 and M7 subtype of
more likely to have higher white cell count and incidence of
infections at the time of presentation. Most patients with AML
present with pallor, fatigue, bleeding or fever as manifesta-
tions of underlying anemia, thrombocytopenia and

Fig. 2 Peripheral smear of a 6-y-old child diagnosed with acute myeloid


leukemia (AML-M2) showing a myeloblast containing an Auer rod (pink Fig. 3 A child presenting with proptosis (chloroma) and diagnosed
colored aggregated lysosome) as AML
Indian J Pediatr (September 2015) 82(9):817824 823

Adult Variety of Chronic Myeloid Leukemia

Though the adult variety of CML is one of the commonest


leukemias in adults, it is rare in children accounting for 35%
cases. The natural history is divided into chronic, accelerated
and blast phases. In the chronic phase, patient has nonspecific
symptoms such as fever, malaise and weight loss.
Occasionally, patients present with acute symptoms such as
bone or joint pain or priapism. Splenomegaly is the most
common physical finding and is usually massive. Mild hepa-
tomegaly and lymphadenopathy may be present. Symptoms
Fig. 4 A child diagnosed as AML showing gum hypertrophy of leukostasis such as headache, dizziness and visual distur-
bances may occur rarely. Leukocytosis is present in all cases
and 80% patients have leukocyte counts above 100,000/cu mm.
AML FAB classification, presence of CNS disease at diagno- The differential count shows all forms of myeloid cells from
sis, absence of minimal residual disease and cytogenetics promyelocytic to polymorphonuclear leukocytes; basophilia
characteristics like Inv16, t(8;21) and t(15;17) and are associ- is common. Mild anemia and thrombocytosis are common,
ated with a favorable outcome. M3 subtype and AML with but thrombocytopenia is rare. Bone marrow examination
Down syndrome are associated with a favorable outcome. shows extreme hypercellularity. Myeloid blasts and
Patients with unfavorable genetic alterations undergo stem promyelocytes constitute <20% of cells in the marrow in
cell transplantation in 1st remission. Since patients with chronic phase. Diagnosis is made by detection of
AML are at high risk for myelosuppression following chemo- Philadelphia (Ph) chromosome on cytogenetics or by FISH
therapy, they require intensive blood component support in- or RT-PCR. Chronic phase typically progresses to blast crisis
cluding platelet transfusions and broad spectrum antibiotics which shows sudden rise in leukocytes and blast count and is
for control of infection which is one of the reason that not indistinguishable from acute leukemia. Leukocyte alkaline
all centers treat AML. phosphatase activity is low. Philadelphia chromosome, which
involves a reciprocal translocation between long arms of chro-
Acute Promyelocytic Leukemia (APL) Acute promyelocytic mosomes 22 and 9; t(9;22) is present in 90% cases.
leukemia (M3), accounts for about 1015% of patients of Allogeneic transplantation from an HLA matched sibling
AML, and are treated differently with all trans retinoic donor can cure 80% of patients with CML [20, 21]. The aim of
acid or arsenic and systemic chemotherapy. All trans treatment during chronic phase is to control the increasing
retinoic acid (ATRA) significantly improved the outcome white cell counts. This can usually be achieved by single agent
in patients with APL. These patients are treated with chemotherapy with either busulfan or hydroxyurea. However,
intensive anthracyclines and high dose cytarabine and these agents are being replaced with -interferon and more
ATRA. Arsenic trioxiode is also now used in combination recently developed tyrosine kinase inhibitor, imatinib
with ATRA for this disease.

Chronic Myeloid Leukemia

Chronic myeloid leukemia (CML), a myeloproliferative dis-


order, is primarily a disease of middle age; the peak incidence
is in the fourth and fifth decade. However, it may occur at any
age including infants and young children. Two main types of
well differentiated myelogenous leukemia have been recog-
nized. One is clinically and rheumatologically comparable
with the adult form of chronic myeloid leukemia and occurs
in children above the age of 4y. The other presents in infancy
and early childhood usually below the age of 4y, called juve-
nile chronic myelogenous leukemia (JCML) has a much more Fig. 5 A 1-y-old boy presented with fever, rashes, anemia, significant
rapid course. splenomegaly and hepatomegaly. He was diagnosed as JMML
824 Indian J Pediatr (September 2015) 82(9):817824

mesylate. Majority of patients achieve complete hematologic References


and cytogenetic response with this therapy and the rate of
progression to accelerated or blast crisis is decreased. 1. Stat bite: Estimated new leukemia cases in 2008. J Natl Cancer Inst.
Dasatinib, a newer tyrosine kinase inhibitor approved for use 2008;100:531.
in adults with CML is currently under clinical trials in chil- 2. Arora RS, Eden TOB, Kapoor G. Epidemiology of childhood cancer
in India. Indian J Cancer. 2009;46:26473.
dren. The blood counts return to normal or near normal in
3. Pui CH, Relling MV, Downing JR. Acute lymphoblastic leukemia. N
almost all patients within 68 wk. Spleen size also decreases. Engl J Med. 2004;350:153548.
With the conventional treatment, the average survival is 34 y. 4. Zwaan MC, Reinhart D, Hitzler J, Vyas P. Acute leukemias in children
Survival after development of accelerated phase is usually less with Down syndrome. Hematol Oncol Clin N Am. 2010;24:1934.
5. Moloney WC. Leukemia in survivors of atomic bombing. N Engl J
than a year and after blast transformation only a few months.
Med. 1955;253:8890.
Allogeneic stem cell transplantation is now used for patients 6. Look AT, Roberson PK, Williams DL, Rivera G, Bowman WP, Pui
who do not respond to tyrosine kinase inhibitors. CH, et al. Prognostic importance of blast cell DNA content in child-
hood acute lymphoblastic leukemia. Blood. 1985;65:107986.
7. Arya LS. Childhood cancerchallenges and opportunities. Indian J
Juvenile Chronic Myeloid Leukemia Pediatr. 2003;70:15962.
8. Pui CH, Robison LL, Look TA. Acute lymphoblastic leukemia.
Lancet. 2008;371:103043.
JCML, also termed as juvenile myelomonocytic leukemia, is 9. Margolin JF, Rabin KR, Steuber CP, Poplack DG. Acute lympho-
an uncommon hematological malignancy accounting for less blastic leukemia. In: Pizzo PA, Poplack DG, editors. Principles and
than 2% leukemias in children. Patients with neurofibromato- Practice of Pediatric Oncology. Philadelphia: Lippincott Williams &
Wilkins; 2011. p. 51865.
sis are at high risk for development of JCML. Compared to
10. Aur RJ, Simone JV, Hustu HO, Verzosa MS. A comparative study of
ACML, JCML, a disease of infancy and early childhood, of- central nervous system irradiation and intensive chemotherapy early
t e n p r e s e n t s w i t h l y m p h a d e n o p a t h y, a n e m i a , in remission of childhood acute lymphocytic leukemia. Cancer.
hepatosplenomegaly, skin involvement (eczema, xanthoma 1972;29:38191.
11. Slats AM, Egeler RM, van-der Does-van den Berg A, Korbijn C,
and caf-au-lait spots), infection and thrombocytopenia
Hhlen K, Kamps WA, et al. Causes of deathother than-
(Fig. 5). progressive leukemia in childhood acute lymphoblastic leukemia
Peripheral smear shows leukocytosis (usually less than and myeloid leukemia: the Dutch Childhood Oncology Group expe-
1000,000/mm3) with the full spectrum of granulocytic rience. Leukemia. 2005;19:53744.
12. Campana D. Minimal residual disease in acute lymphoblastic leuke-
precursors and increased normoblasts; monocytosis is of-
mia. Semin Hematol. 2009;46:1006.
ten striking. Thrombocytopenia and anemia are common. 13. Bleyer WA, Poplack DG. Prophylaxis and treatment of leukemia in
The leukocyte alkaline phosphatase score is normal or the central nervous system and other sanctuaries. Semin Oncol.
low and fetal hemoglobin levels are elevated. Bone mar- 1985;12:13148.
row aspirates show an increased cellularity with predom- 14. Harila-Saari AH, Lhteenmki PM, Pukkala E, Kyyrnen P, Lanning
M, Sankila R. Scholastic achievements of childhood leukemia pa-
inance of granulocytic cells in all stages of maturation; tients: a nationwide, register-based study. J Clin Oncol. 2007;25:
megakaryocytes are normal or decreased. Most patients 351824.
have normal karyotypes or nonspecific chromosomal ab- 15. Davies SM. Subsequent malignant neoplasms in survivors of child-
normalities. Philadelphia chromosome is negative; mono- hood cancer: Childhood Cancer Survivor Study (CCSS) studies.
Pediatr Blood Cancer. 2007;48:72730.
somy 7 is found in 30% patients [22]. 16. Ross JA, Oeffinger KC, Davies SM, Mertens AC, Langer EK,
JCML has a fulminant and rapidly fatal course. Kiffmeyer WR, et al. Genetic variation in the leptin receptor gene
Management involves supportive care including packed red and obesity in survivors of childhood acute lymphoblastic leukemia:
cell and platelet transfusions, treatment of infections and allo- a report from the Childhood Cancer Survivor Study. J Clin Oncol.
2004;22:355862.
geneic stem cell transplant if a matched sibling donor is pres- 17. Hill DE, Ciesielski KT, Hart BL, Jung RE. MRI morphometric and
ent. Even with transplant, there is 3050 % event-free survival neuropsychologic correlates of long term memory in survivors of
rate at 3y. Cis retinoic acid has been tried with some benefit in childhood leukemia. Pediatr Blood Cancer. 2004;42:6117.
JMML. 18. Katz JA, Pulock BH, Jacaruso D, Morad A. Final attained height in
patients successfully treated for childhood acute lymphoblastic leu-
kemia. J Pediatr. 1993;123:5964.
19. Pui CH. Childhood leukemias. N Engl J Med. 1995;332:161830.
20. Marin T, Utturini A, Kantarjian H, Sokal J, Mickey MR, Gale R.
Survival of children with chronic myeloid leukemia. Am J Pediatr
Contributions RS: Wrote the article; AS: Did literature search, edito- Hematol Oncol. 1992;14:22932.
rial corrections and assisted in revising the manuscript. RS will act as 21. Goldman JM. How I, treat chronic myeloid leukemia in the imatinib
guarantor for this paper. era? Blood. 2007;110:282837.
22. Altman AJ, Cecilia FU. Chronic leukemias of childhood. In: Pizzo
Conflict of Interest None. PA, Poplack DG, editors. Principles and Practice of Pediatric
Oncology. Philadelphia: Lippincot Williams & Wilkins; 2011. p.
Source of Funding None. 61137.

You might also like