You are on page 1of 9

Pediatrics International (2015) 57, 811819 doi: 10.1111/ped.

12758

Review Article

Recent progress in the treatment of infant acute lymphoblastic leukemia

Daisuke Tomizawa
Division of Leukemia and Lymphoma, Childrens Cancer Center, National Center for Child Health and Development, Tokyo, Japan

Abstract Treatment of infants with acute lymphoblastic leukemia (ALL), especially those with mixed lineage leukemia (MLL)
rearrangement (MLL-r), which account for approximately 80% of cases, is still a major challenge for pediatric
hematologists and oncologists worldwide. Continuing efforts by collaborative clinical study groups in Europe, North
America, and Japan have rescued approximately half of the MLL-r ALL patients with intensive chemotherapy with or with-
out allogeneic hematopoietic stem cell transplantation. Recent progress has claried the unique mechanism of MLL-r ALL:
the aberrant methylation and histone modications via DOT1L and other related molecules by MLL fusion proteins lead to
leukemogenetic gene expression, thus to overt leukemia. In order to overcome this dismal subtype of ALL, novel targeted
therapy based on leukemia biology is urgently needed. Due to the extreme rarity of the disease, collaboration between the
study groups in Europe (Interfant), North America (Childrens Oncology Group), and Japan (Japanese Pediatric
Leukemia/Lymphoma Study Group) is under way.

Key words acute lymphoblastic leukemia, epigenetics, hematopoietic stem cell transplantation, infant, mixed lineage leukemia.

Acute lymphoblastic leukemia (ALL), a malignant disorder of lym- approximately 80% of infant ALL; and the other with germline
phoid progenitor cells, is the most common type of malignant neo- MLL. Infant ALL with rearranged MLL (MLL-r) is a highly ag-
plasms in children and adolescents, with approximately 444532 gressive leukemia with high white blood cell (WBC) count and
cases diagnosed annually in Japan.1 Optimal use of old drugs, de- frequent involvement of extramedullary sites including the cen-
veloped between the 1950s and 1970s under successive collabora- tral nervous system (CNS) at diagnosis.9,11 In addition, MLL-r
tive clinical trials, has improved the outcome of childhood ALL to ALL has a very immature CD10-negative B-cell precursor phe-
achieve an event-free survival (EFS) rate of 80.487.2% and an notype in approximately two-thirds of cases and is frequently
overall survival (OS) rate of 91.893.5% in recently completed associated with co-expression of myeloid-specic antigens,
clinical trials.25 The progress in the treatment of infant ALL (diag- while mature B-cell or T-cell phenotype is extremely rare. This
nosed at age <12 months), however, is lagging behind compared suggests that infant MLL-r ALL arises from an immature pre-
with the treatment of children 1 year: EFS and OS remain at cursor hematopoietic cell that is not fully committed to lym-
41.750.9% and 44.860.5%, respectively (Table 1).9,1113 In this phoid lineage. In fact, several cases of lineage switch from
review, advances in the pathobiology and clinical management of B-cell lineage to monocytic lineage leukemia have been
ALL in infants is described. reported.14 In contrast, infant ALL involving germline MLL
(MLL-g) generally occurs in late infancy, has a more mature
Epidemiology and characteristics CD10-positive B-cell precursor phenotype, shares similar cyto-
genetic abnormalities with ALL in older children, and is associ-
Infant ALL accounts for <5% of childhood ALL, with 2025 new
ated with better outcome.8,15
cases diagnosed annually in Japan. In children 1 year, ALL com-
prises 7580% of all childhood leukemia because of the high peak
incidence of B-lineage ALL between the age of 2 and 5 years, with
Pathobiology
a predominance in boys. In contrast, the incidence of ALL in in-
fants is the same as that of acute myeloid leukemia (AML), with Pathogenesis of MLL rearrangement
a slight predominance in girls.1,11 The MLL rearrangement, a hallmark of most infant ALL, occurs as
Biologically and clinically, infant ALL consists of two dis- a result of balanced chromosomal translocations that fuse the
tinct subtypes: one involves rearrangements of mixed lineage N-terminus of MLL on chromosome 11q23 to the C-terminus of
leukemia (MLL also called KMT2A), which accounts for one of more than 70 known partner genes.16 The most common
in infant ALL is t(4;11)(q21;q23) or MLL-AF4 (MLL-AFF1),
Correspondence: Daisuke Tomizawa, MD PhD, Division of Leuke- which accounts for approximately half of the cases, followed
mia and Lymphoma, Childrens Cancer Center, National Center by t(11:19)(q23;p13.3) or MLL-ENL (MLL-MLLT1), and t(9;11)
for Child Health and Development, 2-10-1 Okura, Setagaya-ku,
(p22;q23) or MLL-AF9 (MLL-MLLT3). t(9;11)/MLL-AF9 occurs
Tokyo 157-8535, Japan. Email: tomizawa-d@ncchd.go.jp
Received 31 May 2015; revised 25 June 2015; accepted 10 in relatively older infants and is associated with more mature
July 2015. phenotypes compared with other subtypes of infant MLL-r ALL.

2015 Japan Pediatric Society


812 D Tomizawa

Table 1 Outcome of infant ALL in recent clinical trials

No. patients treated


Study Study acronym and No. patients with HSCT in
group inclusion time (MLL-r/MLL-g) rst CR CR (%) EFS (%) OS (%) Source
JILSG MLL96/98 102 (80/22) 49 94.1 4 years 50.9 4.9 4 years 60.5 4.8 Kosaka et al.6
(19952001) Isoyama et al.7
Nagayama et al.8
Tomizawa et al.9
JPLSG MLL03 62 (62/) 44 80.6 4 years 43.2 6.3 4 years 67.2 6.0 Koh et al.10
(20042009)
Interfant Interfant-99 482 (314/82) 37 94 4 years 47.0 2.6 4 years 55.3 2.7 Pieters et al.11
(19992005)
CCG CCG1953 115 (79/36) 37 82.5 5 years 41.7 9.2 5 years 44.8 5.6 Hilden et al.12
(19962000)
COG COG P9407 147 (100/35) 0 91.8 5 years 42.3 6.0 5 years 52.9 6.5 Dreyer et al.13
(20012006)
ALL, acute lymphoblastic leukemia; CCG, Childrens Cancer Group; COG, Childrens Oncology Group; CR, complete remission; EFS, event-
free survival; HSCT, hematopoietic stem cell transplantation; JILSG, Japan Infant Leukemia Study Group; JPLSG, Japanese Pediatric Leukemia/
Lymphoma Study Group; MLL, mixed lineage leukemia; MLL-g, germline MLL; MLL-r, rearranged MLL; OS, overall survival.

It is well recognized that the genetic event involving MLL takes potent driver and plays a dominant role in leukemogenesis of in-
place in utero. This is supported by evidence of the very early onset fant MLL-r ALL without cooperating mutations.
of infant ALL, the high rate of concordance of leukemia in mono-
zygotic twins when one twin developed leukemia in the rst year of
Disorder of epigenetic regulation in MLL fusions
life, and the prenatal origin of infant ALL conrmed directly by the
detection of unique MLL-AF4 fusion in neonatal blood spots on The MLL gene is a human homologue of Drosophila melanogaster
Guthrie cards from infants with MLL-AF4-positive ALL.17,18 trithorax, which encodes a DNA-binding protein that methylates
MLL rearrangements are often recognized in patients with histone H3 lysine 4 (H3K4) and positively regulates gene expres-
treatment-related AML arising after exposure to topoisomerase II sion including multiple Hox genes. As a result of inter-
inhibitors. As for infant ALL, epidemiologic studies indicated that chromosomal translocations, disrupted MLL in a breakpoint cluster
exposure to topoisomerase II-like materials (such as bioavonoids) region that spans exons 812 loses the H3K4 methyltransferase
via the maternal diet might be involved in leukemogenesis.19 (SET) domain, and is replaced by one of the translocation partner
genes. MLL fusion proteins, MLL-AF10, MLL-ENL, and MLL-
AF9, however, recruit and directly interact with histone H3 lysine
Genetics of infant ALL and MLL rearrangement 79 (H3K79) methyltransferase DOT1L (Fig. 1).23 Other major
The mechanism of leukemogenesis in infants after the initial MLL fusion proteins, MLL-AF4, MLL-AF5q31, and MLL-ELL1
genetic event in MLL has been a mystery for decades. On early recruit and interact directly with positive transcription elongation
gene expression proling, that of infant MLL-r ALL was highly factor b (pTEFb) that phosphorylates RNA polymerase II, and
distinct, consistent with an early hematopoietic progenitor express-
ing select multilineage markers and individual HOX genes, com-
pared with those of ALL diagnosed in older children and AML.
Notably, overexpression of wild-type fms-related tyrosine kinase
3 (FLT3) distinguished infant MLL-r ALL from ALL in older
children and AML.20
It is known that multiple stepwise mutations are necessary for
leukemogenesis in general. In mouse models of MLL-r leukemia,
expression of MLL chimeric gene alone is insufcient for full
transformation to leukemia.21 The St Jude Childrens Research
Hospital and Washington Universitys Pediatric Cancer Genome
Project, however, showed that for the genome, exome, RNA
and targeted DNA sequencing on MLL-r ALL, the majority of Fig. 1 Molecular actions induced by mixed lineage leukemia (MLL)
copy number alterations (CNA) and structural variations found fusion. Direct or indirect recruitment of aberrant DOT1L by MLL fu-
in infant MLL-r ALL was a direct consequence of MLL rear- sion proteins induces site-specic hyper-methylation and inappropriate
rangement, and only a mean of 1.3 non-silent mutations per case transcription of several leukemogenic genes, which leads to leukemia
subsequently. BRD4, bromodomain-containing protein 4; H3K79,
was detected in the dominant leukemic clone, which is one of the H3 lysine 79; H4K16, H4 lysine 16; LEDGF, lens epithelium-derived
lowest frequencies of somatic mutations of any sequenced cancer growth factor; PAFc, polymerase-associated factor c; pTEFb, positive
ever.22 These data clearly imply that the MLL fusion protein is a transcription elongation factor b; SEC, superelongation complex.

2015 Japan Pediatric Society


Treatment progress in infant ALL 813

interact indirectly with DOT1L.24 Most of the major fusion part- children, and has also been proven to be prognostic in infants.11,30
ners with MLL directly interact with superelongation complex Recently, detection of minimal residual disease (MRD) measured
(SEC). Together with polymerase-associated factor c (PAFc), by polymerase chain reaction (PCR) or ow cytometry has been
SEC binds bromodomain and extra-terminal (BET) family member shown to be a strong and independent prognostic factor in several
BRD4, which is able to bind acetylated histone through its clinical studies, and is currently considered to be the standard of
bromodomains.25 Direct or indirect interactions of DOT1L, care in ALL children aged >1 year.31,32 The Interfant-99 study,
pTEFb, SEC, PAFc, and/or BRD4 with MLL fusion proteins, en- an international collaborative infant ALL study that consisted
hance expression of leukemogenic genes, including HOXA9 and mainly of European countries, reported that MRD targeting the
MEIS1.26 rearranged MLL breakpoint is a powerful tool for better risk strati-
Although the exact mechanism of MLL-r leukemogenesis in in- cation of MLL-r ALL infants.33 The study also indicated that
fants is not fully understood, it is estimated that aberrant epigenetic MLL rearrangement was the preferred PCR MRD target over im-
regulation induced by MLL fusions brings about a unique prole of munoglobulin (Ig) or T-cell receptor (TCR) rearrangement, be-
gene expression, thus leading to leukemia without additional gene cause of the oligoclonality of leukemia clones, which is
mutations. frequently seen in infant MLL-r ALL. Although MLL rearrange-
ments are assumed to be present in the total leukemic clone, there
Leukemia-initiating cells in infant MLL-r ALL is a risk of detecting subclone only when targeting Ig/TCR rear-
There is increasing evidence suggesting that some leukemia, such rangements, resulting in underestimation of actual MRD load.
as AML, is maintained by self-renewing leukemia-initiating cells
Chemotherapy
(LIC), a leukemia cell of origin, which exists at the apex of a hier-
archy and is capable of differentiating into leukemic blasts.27 This Leukemic cells from infants with MLL-r ALL are more resistant
model suggests that therapy failure results from residual LIC after in vitro to prednisolone and L-asparaginase, which are the key
eradication of differentiated leukemic blasts with current treatment drugs for treating ALL, than cells from older children with ALL.
modalities; therefore, identication and characterization of LIC in In contrast, infant ALL cells are more sensitive to cytarabine
infant MLL-r ALL may lead to the development of effective thera- (Ara-C) irrespective of MLL status.34 The greater Ara-C sensitivity
peutic strategies. Using a xenotransplantation model, Aoki et al. is explained by the high expression of human equilibrative nucleo-
found that LIC in infant MLL-r ALL have distinct surface expres- side transporter 1 (hENT1) on infant ALL cells, which Ara-C
sion patterns of CD34, CD38, CD19, and CD33 according to the mainly depends on to permeate the cell membrane.35 From these
fusion subtypes: CD34+CD38+CD19+ and CD34 CD19+ cells ini- observations, a hybrid chemotherapy combining ALL-oriented
tiated leukemia in MLL-AF4 patients, CD34 CD19+ in MLL-AF9 drugs (e.g. corticosteroids, vincristine, L-asparaginase, and metho-
patients, and either CD34+ or CD34 depending on the pattern of trexate [MTX]) and AML-oriented drugs (e.g. anthracyclines,
CD34 expression in MLL-ENL patients.28 In addition, CD9, etoposide, and especially Ara-C) is considered effective and is of-
CD32, and CD24 were differentially overexpressed in MLL-r ten used for treating infants with ALL. Remission induction che-
LIC compared with normal hematopoietic stem cells. By targeting motherapy results in a high complete remission (CR) rate 90%
these surface molecules that are selectively expressed in MLL-r in general, but a high incidence of relapse occurs 45 months after
LIC with recently developed immunotherapeutic modalities such achieving CR. Attempt to improve outcome of infants with ALL by
as bi-specic T-cell engager (BiTE) antibodies or chimeric antigen intensifying delayed intensication with high-dose Ara-C and
receptor (CAR) T cells, it may be possible to treat MLL-r ALL high-dose MTX failed in the Interfant-99 study.11 Therefore, the
more effectively while minimizing treatment-related hematopoietic current ongoing Interfant-06 is investigating whether early
adverse events. intensication with AML-oriented chemotherapy with Ara-C,
daunorubicin or mitoxantrone, and etoposide will be superior to
Clinical management ALL-oriented chemotherapy with cyclophosphamide, Ara-C, and
6-mercaputopurine in MLL-r infants. Regardless, it is likely that
Prognostic factors medium-risk MLL-r infants can be effectively treated with chemo-
Among all the known prognostic factors, presence of MLL rear- therapy only. The Interfant-99 study noted a disease-free survival
rangement is the strongest.29 Recently completed clinical trials (DFS) rate of 53.8% with chemotherapy for MLL-r infants without
show similar outcomes among the subtypes of translocation or fu- additional poor prognostic factors (age <6 months at onset with ei-
sion partners of rearranged MLL.11 Other factors such as high ther WBC 300 109/L or poor prednisolone response).36 The
WBC, CNS involvement, and absence of CD10 expression on leu- Childrens Oncology Group (COG) in North America also showed
kemic cells are also prognostic but are highly correlated with MLL in the COG P9407 study that an EFS of 43.8% was achieved for in-
rearrangement. The second most prognostic is young age at onset fants >90 days old at onset of MLL-r ALL with only chemother-
of ALL: outcome of infants younger than 3 or 6 months is ex- apy.13 Given the experience in Europe and North America, a
tremely poor compared with older infants.9,11,12 Finally, treatment strategy omitting the indication of allogeneic hematopoietic stem
response evaluated in the early phase of the treatment has evolved cell transplantation (HSCT) but with the introduction of the
as a powerful prognostic factor. Classically, peripheral blood blast Interfant induction followed by COG post-remission chemotherapy
count after 8 days of prednisolone therapy (prednisolone response) for MLL-r infants 6 months old without CNS involvement, is
has been used for risk stratication in the therapy of ALL in older currently being evaluated in the Japanese nationwide MLL-10

2015 Japan Pediatric Society


814 D Tomizawa

trial (UMIN000004801) conducted by the Japanese Pediatric outcome of 22 infants with MLL-g ALL was excellent with
Leukemia/Lymphoma Study Group (JPLSG). chemotherapy only, as previously described.8
Infants with MLL-g ALL may be cured with a chemotherapy In 2003, a new Japanese nationwide clinical study group for
regimen relatively similar to the treatment regimen for older chil- childhood hematological malignancies, the JPLSG, was esta-
dren with ALL. Four year EFS and OS of 67% and 77%, respec- blished, as a result of the merging of the four existing study groups
tively, were achieved in the combined results of the Interfant-99 in Japan (Tokyo Childrens Cancer Study Group [TCCSG], Japan
study and the COG-P9407 study.15 Moreover, in Japan, 5 year Association of Childhood Leukemia Study [JACLS], Kyushu
EFS and OS rates of 95.5% were achieved in the combined results Yamaguchi Childrens Cancer Study Group [KYCCSG], and
of the MLL96 and MLL98 trials.8 Japanese Childhood Cancer and Leukemia Study Group
Irrespective of MLL status, special attention needs to be paid [CCLSG]). An encouraging result in the previous MLL98 study
when prescribing chemotherapeutic drugs for infants because of for 3 year post-transplantation EFS (64.4%) in 29 infants with
the following factors: higher percentage of total and extracellular MLL-r ALL transplanted at the rst CR prompted us to speculate
body water content than older children or adults, higher unbound whether an intervention with more effective chemotherapy and
active fraction of drugs because of lower afnity of drugs to serum HSCT in an earlier phase could prevent early relapse and produce
protein, lower P450 enzyme activity, lower tubular and glomerular a better outcome.6 Thus, the MLL03 study was conducted, which
function, lower bodyweight to body surface area ratio and so on. recruited 62 eligible infants with MLL-r ALL between 2004 and
Dose adjustments (e.g. 2/3 of the calculated dose for infants 2009.10 In that study, pre-transplant chemotherapy was intensied
<6 months old, 3/4 dose for 6<12 months old, and full dose for with high-dose Ara-C and all the patients at their rst CR were
12 months old, used in Interfant-99) are generally recommended assigned to receive HSCT within 4 months of initial induction. Be-
but these are somewhat arbitrary due to a lack of pharmacokinetic cause there are no differences in the outcome of HSCT for ALL in-
(PK) data for infants for many of the drugs. PK analysis of high- fants according to either donor source (cord blood vs unrelated
dose MTX (5 g/m2/dose) in the Interfant-99, applying the afore- bone marrow [BM] vs related BM) or conditioning regimen
mentioned dose reduction rule, indicated a slightly lower MTX (myeloablative busulfan [BU] based vs myeloablative total body ir-
clearance in younger infants, but the toxicity prole was similar radiation [TBI] based), the donors were restricted to either human
to that for older infants.37 leukocyte antigen 4/6 serologically matched unrelated cord blood
or human leukocyte antigen 5/6 matched related donor; condition-
HSCT ing was a non-TBI myeloablative regimen with BU (dose adjusted
according to individual PK test), etoposide and cyclophospha-
Clinical trials in Japan mide.9,40 The prophylaxis for graft-versus-host disease (GVHD)
The dismal outcome of 24 infants with ALL noted in a retrospec- was either cyclosporine or tacrolimus combined with short-term
tive survey conducted by Ishii et al. in the 1980s led to the rst in- MTX. As a result, 4 year EFS and OS were 43.2% and 67.2%,
fant ALL-specic clinical trial in Japan conducted by the respectively. The strategy of early HSCT effectively prevented
Aggressive Treatment (AT) Research Group and the Childhood early relapse and was feasible for infants with MLL-r ALL,
Leukemia Study Group of the Ministry of Health and Welfare.38 given that 44/50 (88%) of those who achieved CR were able
Between 1989 and 1995, 37 infants were registered and treated to receive HSCT in rst CR. Modest improvement in the out-
with intensive chemotherapy. As a result, the 3 year EFS rate was come has been observed through the MLL96, MLL98, and
33%, but an EFS rate of 75% was observed for infants with MLL03 studies (Fig. 2), but the fact that 18/44 (40%) of pa-
CD10-positive phenotype without 11q23/MLL involvement.39 tients in MLL03 still relapsed despite receiving transplantation
To address this poor prognosis, the Japanese Infant Leuke- in their rst CR indicates the limited anti-leukemic efcacy of
mia Study Group was established in cooperation with the allogeneic HSCT for infants with MLL-r ALL.
existing childhood leukemia study groups in Japan at that time,
and two consecutive clinical trials, designated MLL96
(19951998) and MLL98 (19982002), were performed with Clinical trials in North America and Europe
102 ALL infants (80 MLL-r and 22 MLL-g).69 These were Sanders et al. reported the outcome of allogeneic HSCT with TBI
the rst studies to stratify ALL infants according to MLL status conditioning for 40 infants with ALL (17 at rst CR, seven at sec-
conrmed on either Southern blot or split-signal uorescence in ond or third CR, and 16 at relapse) at the Fred Hutchinson Cancer
situ hybridization (FISH). Use of these molecular methods for Research Center between 1982 and 2003.41 Eleven of the 14 in-
the detection of MLL rearrangements is critical because 13/80 fants with MLL-r ALL transplanted at rst CR survived. This re-
MLL-r ALL patients (16.2%) had no 11q23 abnormalities on sult, however, should be interpreted with caution because many
cytogenetic analysis with the G-banding technique, which would of the patients were >6 months of age at diagnosis, and, if it were
have been missed otherwise. Given the poor outcome by che- present day, they would have been successfully treated with che-
motherapy for MLL-r patients as historically observed, all the motherapy without HSCT. In addition, analysis of HSCT outcome
infants with MLL-r ALL were assigned to receive allogeneic should take into account the waiting time from diagnosis to trans-
HSCT in rst CR. Due to the frequent treatment failure (27/49 plantation, especially for analyzing infants with MLL-r ALL be-
events) before HSCT, 5 year EFS and OS of the 80 infants with cause they tend to relapse at a very early stage. In fact, most of
MLL-r ALL remained at 38.6% and 50.8%, respectively.9 The the study reports that adjusted waiting time to HSCT found that

2015 Japan Pediatric Society


Treatment progress in infant ALL 815

MLL-r ALL who received allogeneic HSCT from 1996 to 2011;


the NRM rate of 5.6% for the recent period (20042011) was lower
than the 20.8% for the previous period (19962003).40 The causes
of overall non-relapse deaths in 15 cases were pulmonary compli-
cations (n = 6), infection (n = 5), GVHD (n = 3), and veno-
occlusive disease/sinusoidal obstruction syndrome (VOD/SOS;
n = 1). Given that BU-based myeloablative conditioning is widely
used as standard conditioning for transplantation in ALL infants,
one should be aware of pulmonary complications as well as hepatic
VOD/SOS. Kawashima et al. reported that infants with
BU-conditioning might be at risk of pulmonary VOD or pulmonary
hypertension, and the latter could be treated with sildenal and
beraprost.44 Although there was only one death due to VOD/SOS
in the Japanese HSCT registry data, the incidence of VOD/SOS
was 14% (6/44) in the JPLSG MLL03 trial.10 Therefore, in order,
at least, to reduce the risk of VOD/SOS, treosulphan, an alkylating
agent similar to BU but with lower risk of seizures, severe mucosi-
Fig. 2 Event-free survival (EFS) in infants with acute lymphoblastic tis, and hepatotoxicity, might be an alternative to BU.45
leukemia and mixed lineage leukemia (MLL) rearrangements registered In addition, late effects need to be borne in mind when treating
in the ( ) MLL96 (n = 42, 19951998), ( ) MLL98 infants with HSCT, although little is known about them. The
(n = 38, 19982002), and ( ) MLL03 (n = 62, 20042009)
in Japan. Japanese MLL96 and MLL98 studies noted no severe late effects
for MLL-g patients who received chemotherapy only, but various
late complications such as chronic GVHD, hypothyroidism, skin
allogeneic HSCT had a negative impact on clinical benet in in- abnormalities, ophthalmologic complications, pulmonary compli-
fants with ALL. Pui et al. conducted a retrospective analysis of cations, dental abnormalities, and neurocognitive problems were
497 ALL children (including 214 infants) and various 11q23 ab- observed in MLL-r patients who did receive allogeneic HSCT.9
normalities from 11 study groups and single institutions between The most frequently observed late effect was short stature, espe-
1983 and 1995, and concluded that any type of HSCT was associ- cially for those who received TBI conditioning, but ve of the 14
ated with signicantly worse DFS and OS compared with chemo- patients who received BU conditioning also had height SD score
therapy only in 256 patients with t(4;11) ALL.42 As for < -2.0. A likely reason why little attention has been paid to late
prospective studies, combined results from North American stud- effects in ALL infants is that their survival is poor. That said, as
ies, Childrens Cancer Group (CCG) 1953 and Pediatric Oncology the survival rate grows, issues such as pubertal problems should
Group (POG) 9407, showed a 5 year EFS rate of 48.8% in infants be claried in order to improve quality of life.
who received HSCT (n = 53) and 48.7% in infants who received
chemotherapy (n = 47; P = 0.60).43 In the following COG trials Treatment for refractory or relapsed disease
for infant ALL, amended COG P9407 (20042006) and Although we have encountered refractory or relapsed disease in
AALL0631 (20082014, NCT00557193) trials eliminated the in- approximately 50% of MLL-r ALL infants, few reports have
dication of HSCT in rst CR. Finally, a report from the Interfant- focused on clinical course and outcome. A Japanese report on
99 study compared 37 infants with MLL-r ALL who received 39 refractory or relapsed patients from the MLL96 and MLL98
HSCT and 240 who received chemotherapy only, and noted bet- studies showed a 40.5% CR rate with various chemotherapy
ter DFS (60.1% vs 46.8%, P = 0.03) and OS (65.6% vs 48.6%, regimens (ALL-oriented, AML-oriented, or ALL/AML hybrid)
P = 0.02) for the HSCT group. The advantage of HSCT, however, and a 25.6% nal OS rate.46 Notably, OS was 21.4% even in
was restricted to only the high-risk group, dened as MLL-r ALL patients with a history of prior HSCT. Achievement of CR after
infants aged <6 months, and either WBC 300 109/L or poor second-line treatment was the most signicant prognostic factor,
prednisolone response.36 In the current ongoing Interfant-06 study with a >50% chance of survival once CR was obtained.
(EudraCT No. 2005-004 599-19), HSCT is indicated only for high-
risk patients and medium-risk patients (other MLL-r ALL infants) Supportive care
with MRD 10 4 after consolidation chemotherapy. Sufcient supportive care is an essential part of treating infants with
ALL, because infants are particularly vulnerable to intensive cyto-
toxic therapy. In particular, during the remission induction phase,
Problematic issues of HSCT infants are at high risk of tumor lysis syndrome and intracranial
Recent progress in supportive therapy has resulted in substantial re- hemorrhage because of the very high tumor burden in MLL-r, to-
duction of the non-relapse mortality (NRM) rate related to alloge- gether with a risk of mucositis, severe infection, and pulmonary
neic HSCT for infants with ALL. According to the nationwide complications. In the Japanese MLL03 study, tumor lysis syn-
registry of the Japan Society for Hematopoietic Cell Transplanta- drome was observed in 40% of cases presumably because
tion, overall NRM rate was 12.0% among 132 infants with rasburicase, a recombinant urate oxidase, was not available in

2015 Japan Pediatric Society


816 D Tomizawa

Japan during the study period; the situation improved after authoriza- be achieved with this approach. Novel therapies are urgently re-
tion of rasburicase in Japan in December 2009.10,47 In the COG quired, especially for those with MLL rearrangements. New agents
P9407 study, an unacceptably high rate of early death from bacterial, with potential efcacy for infant ALL are listed in Table 2.
fungal, and viral infections prompted the investigators to amend the
protocol by substituting dexamethasone with prednisolone, changing Clofarabine
the delivery of daunorubicin from 48 h continuous infusion to 30 min Given that infant leukemia cells are sensitive to purine nucleoside
infusion, and introducing an enhanced supportive care guideline.48 analogues such as Ara-C, a newly developed nucleoside analogue,
These amendments led to a signicant reduction in early mortality clofarabine, might be a good candidate for use in treating infants
rate from 13.7% to 3.5% (P = 0.036). Respiratory syncytial virus with ALL.
(RSV) infection can cause serious lower respiratory complications
in infants, which may result in death. Palivizumab, a humanized FLT3 inhibitors
monoclonal antibody that targets RSV epitope, was approved for
use in Japan in August 2013 in young children (<2 years old) with High expression of wild-type FLT3 is observed in 80% of the in-
malignant disease who receive cytotoxic chemotherapy.49 Together fants with MLL-r ALL. In in vitro studies, high-level FLT3 expres-
with universal vaccination for RSV, which is currently in develop- sion in primary infant MLL-r ALL samples was associated with
ment, it is expected that RSV-induced pulmonary complications activated FLT3 and cytotoxic responsiveness to FLT3 inhibi-
could be effectively prevented in infants with ALL. tors.5052 The role of several small-molecule FLT3 inhibitors has
been investigated in North America and Europe.
Novel therapies
Intensication of conventional cytotoxic chemotherapeutic drugs Epigenetic modiers
with or without HSCT has enabled cure in approximately 50% of Currently, the most attractive class of agents for infant MLL-r ALL
the infants with ALL, but this is far from satisfactory and further is epigenetic modiers. Genome-wide methylation studies have
improvement in the outcome of this dismal disease is unlikely to shown that MLL-r infant ALL cells are characterized by aberrant

Table 2 New agents under investigation for infant ALL

Class Comments
Nucleoside analogues
Clofarabine Investigated within Total XVI study ongoing at SJCRH, a phase III study for pediatric ALL (NCT00549848).
Evaluating its role in early and delayed intensication in combination with cyclophosphamide
and etoposide for infant MLL-r ALL.
FLT3 inhibitors
Lestaurtinib (CEP-701) Phase III study, COG ALLL0631, for infant ALL (NCT00557193). Randomizing to receive or not to receive
lestaurtinib combined with conventional chemotherapy. Closed to accrual.
Midostaurin (PKC412) ITCC phase I/II study for children with R/R MLL-r ALL or FLT3 mutated AML (NCT00866281, EudraCT
No. 2008006 93111). Completed.
Quizartinib (AC220) TACL phase I study in combination with Ara-C and etoposide for children with R/R MLL-r ALL or FLT3
mutated AML (NCT01411267, T2009-004). Completed.
Epigenetic modiers
Demethylating agents TACL phase I study testing azacitidine followed by udarabine/Ara-C for children with R/R ALL or AML
(Azacitidine, decitabine) (NCT01861002). Infants were not included. Completed.
Ongoing TACL pilot study testing decitabine and vorinostat with chemotherapy (vincristine, dexamethasone,
mitoxantrone, pegasparaginase, methotrexate) for relapsed ALL (NCT01483690). Infants not included.
Histone deacetylase inhibitors Ongoing SJCRH phase II study, RELMLL, testing combination of vorinostat and bortezomib (proteasome
(Vorinostat, panobinostat) inhibitor) for R/R MLL-r hematologic malignancies (NCT02419755).
Ongoing TACL phase I study testing panobinostat in combination with Ara-C for children with refractory
hematological malignancies (NCT01321346, T2009-012). Infants not included.
DOT1L inhibitor (EPZ-5676) Inhibition of the DOT1L histone methyltransferase for R/R MLL-r leukemia including AML with MLL-PTD.
Ongoing pediatric phase I study in North America (NCT02141828).
BET protein inhibitors Inhibition of BET-proteins (e.g. BRD4) with BRD2/3/4 inhibitor (OTX015) may provide a new treatment
(OTX-015) strategy in infant ALL. Evaluated in adult phase I trial for hematological malignancies (NCT01713582).
Immunotherapy
CD19 BiTE (Blinatumomab) Phase I/II study testing BiTE antibodies targeting CD19 for children and adolescents with R/R B-precursor
ALL in North America and Europe (NCT01471782). Closed to accrual.
CD19 CAR T-cells Infusion of autologous T cells transduced with CD19-directed chimeric antigen receptor lentiviral vector
(CART19 etc.) for children and adolescents with R/R CD19 + ALL or lymphoma (NCT01626495). Infants not included.

ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; BET, bromodomain and extra-terminal; BiTE, bi-specic T-cell engager;
Ara-C, cytarabine; CAR, chimeric antigen receptor; FLT3, fms-related tyrosine kinase 3; ITCC, Innovative Therapy for Childhood Cancer;
MLL, mixed lineage leukemia; MLL-PTD, partial tandem duplication of MLL; MLL-r, rearranged MLL; R/R, relapsed or refractory; SJCRH,
St Jude Childrens Research Hospital; TACL, Therapeutic Advances in Childhood Leukemia Consortium.

2015 Japan Pediatric Society


Treatment progress in infant ALL 817

methylated genomic state and respond well to demethylating agents, 2 Conter V, Bartram CR, Valsecchi MG et al. Molecular response to
leading to apoptosis in these cells.53,54 In addition, leukemia- treatment redenes all prognostic factors in children and adoles-
cents with B-cell precursor acute lymphoblastic leukemia: Results
specic histone modications such as H3K79 dimethylation in- in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood
duced via DOT1L recruitment by MLL fusion proteins can be effec- 2010; 115: 320614.
tively modulated by histone deacetylase (HDAC) inhibitors.55 3 Hunger SP, Lu X, Devidas M et al. Improved survival for children
These ndings support the use of demethylating agents and/or and adolescents with acute lymphoblastic leukemia between 1990
HDAC inhibitors that might reverse the inherent resistance to che- and 2005: A report from the Childrens Oncology Group. J. Clin.
Oncol. 2012; 30: 16639.
motherapy for infant MLL-r ALL. A possible international collabo- 4 Pui C-H, Campana D, Pei D et al. Treating childhood acute lym-
rative trial for infant ALL involving this strategy is in development phoblastic leukemia without cranial irradiation. N. Engl. J. Med.
by the Interfant, COG, and JPLSG. Direct inhibition of DOT1L is 2009; 360: 273041.
also currently being investigated in an early clinical trial.56,57 5 Vora A, Goulden N, Wade R et al. Treatment reduction for
children and young adults with low-risk acute lymphoblastic
leukaemia dened by minimal residual disease (UKALL
Immunotherapy
2003): A randomised controlled trial. Lancet Oncol. 2013; 14:
Another attractive approach, especially for relapsed/refractory 199209.
cases, is immunotherapy targeting CD19, a B-lymphocyte antigen 6 Kosaka Y, Koh K, Kinukawa N et al. Infant acute lymphoblastic
leukemia with MLL gene rearrangements: Outcome following in-
universally expressed in infant MLL-r ALL cells. Recently, a phase tensive chemotherapy and hematopoietic stem cell
I/II study evaluating CD19-targeting BiTE antibodies for children transplantation. Blood 2004; 104: 352734.
and adolescents with relapsed/refractory ALL was closed to patient 7 Isoyama K, Eguchi M, Hibi S et al. Risk-directed treatment of in-
accrual.58 Another CD19-targeting immunotherapy using CAR- fant acute lymphoblastic leukaemia based on early assessment of
modied autologous T-cell therapy is also in development for MLL gene status: Results of the Japan Infant Leukaemia Study
(MLL96). Br. J. Haematol. 2002; 118: 9991010.
children and adolescents with relapsed/refractory CD19 + ALL or 8 Nagayama J, Tomizawa D, Koh K et al. Infants with acute lympho-
lymphoma, and early reports have been encouraging.59 blastic leukemia and a germline MLL gene are highly curable with
use of chemotherapy alone: Results from the Japan infant leukemia
Conclusions and future directions study group. Blood 2006; 107: 46635.
9 Tomizawa D, Koh K, Sato T et al. Outcome of risk-based therapy
Infant ALL, especially that involving MLL rearrangements, is one for infant acute lymphoblastic leukemia with or without an MLL
of the most difcult to cure among all the subtypes of childhood gene rearrangement, with emphasis on late effects: A nal report
acute leukemia. Efforts by clinical trial groups in Europe, North of two consecutive studies, MLL96 and MLL98, of the Japan in-
fant leukemia study group. Leukemia 2007; 21: 225863.
America, and Japan to overcome this difcult disease with inten-
10 Koh K, Tomizawa D, Moriya Saito A et al. Early use of allogeneic
sive chemotherapy with or without HSCT has improved EFS to hematopoietic stem cell transplantation for infants with MLL gene-
40-50%. Further improvement will not be achieved without the de- rearrangement-positive acute lymphoblastic leukemia. Leukemia
velopment of novel targeted therapies based on the unique biology 2015; 29: 2906.
of the type of leukemia. Unfortunately, the extreme rarity of the dis- 11 Pieters R, Schrappe M, de Lorenzo P et al. A treatment protocol for
infants younger than 1 year with acute lymphoblastic leukaemia
ease is a major obstacle to drug development. Close, proactive col-
(Interfant-99): An observational study and a multicentre
laboration between Interfant, COG, and JPLSG would be a key randomised trial. Lancet 2007; 370: 24050.
solution to this issue. 12 Hilden JM, Dinndorf PA, Meerbaum SO et al. Analysis of prog-
nostic factors of acute lymphoblastic leukemia in infants: Report
Acknowledgments on CCG 1953 from the Childrens Oncology Group. Blood 2006;
108: 44151.
The author acknowledges all the individuals who were involved in 13 Dreyer ZE, Hilden JM, Jones TL et al. Intensied chemotherapy
infant ALL studies in Japan, especially Drs Eiichi Ishii (Department without SCT in infant ALL: Results from COG P9407 (cohort 3).
of Pediatrics, Ehime University Graduate School of Medicine) and Pediatr. Blood Cancer 2015; 62: 41926.
14 Sakaki H, Kanegane H, Nomura K et al. Early lineage switch in an
Katsuyoshi Koh (Department of Hematology/Oncology, Saitama infant acute lymphoblastic leukemia. Int. J. Hematol. 2009; 90:
Childrens Medical Center) for their strong leadership. Infant 6535.
ALL studies in Japan are supported by Grants for Clinical Cancer 15 Moorman AV, Pieters R, Dreyer ZE et al. Cytogenetics and out-
Research and Grants-in-Aid for Cancer Research from the Ministry come of infants with acute lymphoblastic leukemia and absence
of Health, Labour, and Welfare of Japan. The author also acknowl- of MLL rearrangements. Leukemia 2014; 28: 42830.
16 Meyer C, Hofmann J, Burmeister T et al. The MLL recombinome
edges Dr Julian Tang of the Department of Education for Clinical of acute leukemias in 2013. Leukemia 2013; 27: 216576.
Research, National Center for Child Health and Development, for 17 Gale KB, Ford AM, Repp R et al. Backtracking leukemia to
critical comments and assistance with the manuscript. The author birth: Identication of clonotypic gene fusion sequences in neo-
declares that he has no conicts of interest. natal blood spots. Proc. Natl. Acad. Sci. U. S. A. 1997; 94:
139504.
18 Greaves MF, Maia AT, Wiemels JL, Ford AM. Leukemia in twins:
References Lessons in natural history. Blood 2003; 102: 232133.
1 Horibe K, Takimoto T, Tsuchida M et al. Incidence and survival 19 Spector LG, Xie Y, Robison LL et al. Maternal diet and infant leu-
rates of hematological malignancies in Japanese children and ado- kemia: The DNA topoisomerase II inhibitor hypothesis: A report
lescents (20062010): Based on registry data from the Japanese from the childrens oncology group. Cancer Epidemiol.
Society of Pediatric Hematology. Int. J. Hematol. 2013; 98: 7488. Biomarkers Prev. 2005; 14: 6515.

2015 Japan Pediatric Society


818 D Tomizawa

20 Armstrong SA, Staunton JE, Silverman LB et al. MLL transloca- 39 Nishimura S, Kobayashi M, Ueda K et al. Treatment of infant acute
tions specify a distinct gene expression prole that distinguishes a lymphoblastic leukemia in Japan. Childhood Leukemia Study
unique leukemia. Nat. Genet. 2002; 30: 417. Group of the Ministry of Health and Welfare (Kouseisho). Int. J.
21 Krivtsov AV, Armstrong SA. MLL translocations, histone modi- Hematol. 1999; 69: 24452.
cations and leukaemia stem-cell development. Nat. Rev. Cancer 40 Kato M, Hasegawa D, Koh K et al. Allogeneic
2007; 7: 82333. haematopoietic stem cell transplantation for infant acute lym-
22 Andersson AK, Ma J, Wang J et al. The landscape of somatic mu- phoblastic leukaemia with KMT2A (MLL) rearrangements: A
tations in infant MLL-rearranged acute lymphoblastic leukemias. retrospective study from the paediatric acute lymphoblastic
Nat. Genet. 2015; 47: 3307. leukaemia working group of the Japan society for
23 Bernt KM, Zhu N, Sinha AU et al. MLL-rearranged leukemia is haematopoietic cell transplantation. Br. J. Haematol. 2015;
dependent on aberrant H3K79 methylation by DOT1L. Cancer 168: 56470.
Cell 2011; 20: 6678. 41 Sanders JE, Im HJ, Hoffmeister PA et al. Allogeneic hematopoietic
24 Yokoyama A, Lin M, Naresh A, Kitabayashi I, Cleary ML. A cell transplantation for infants with acute lymphoblastic leukemia.
higher-order complex containing AF4 and ENL family proteins Blood 2005; 105: 374956.
with P-TEFb facilitates oncogenic and physiologic MLL- 42 Pui C-H, Gaynon PS, Boyett JM et al. Outcome of treatment in
dependent transcription. Cancer Cell 2010; 17: 198212. childhood acute lymphoblastic leukaemia with rearrangements of
25 Dawson MA, Prinjha RK, Dittmann A et al. Inhibition of BET re- the 11q23 chromosomal region. Lancet 2002; 359: 190915.
cruitment to chromatin as an effective treatment for MLL-fusion 43 Dreyer ZE, Dinndorf PA, Camitta B et al. Analysis of the role of
leukaemia. Nature 2011; 478: 52933. hematopoietic stem-cell transplantation in infants with acute lym-
26 Imamura T, Morimoto A, Takanashi M et al. Frequent co- phoblastic leukemia in rst remission and MLL gene
expression of HoxA9 and Meis1 genes in infant acute lymphoblas- rearrangements: A report from the Childrens oncology group.
tic leukaemia with MLL rearrangement. Br. J. Haematol. 2002; J. Clin. Oncol. 2011; 29: 21422.
119: 11921. 44 Kawashima N, Ikoma M, Sekiya Y et al. Successful treatment of
27 Bonnet D, Dick JE. Human acute myeloid leukemia is organized as pulmonary hypertension with beraprost and sildenal after cord
a hierarchy that originates from a primitive hematopoietic cell. Nat. blood transplantation for infantile leukemia. Int. J. Hematol.
Med. 1997; 3: 7307. 2013; 97: 14750.
28 Aoki Y, Watanabe T, Saito Y et al. Identication of CD34 45 Corker E, Astwood E, Williams J, Vora A. Treosulphan-based
+ and CD34- leukemia-initiating cells in MLL-rearranged radiation-free myeloablative conditioning for allogeneic transplant
human acute lymphoblastic leukemia. Blood 2014; 125: in infant acute lymphoblastic leukaemia. Br. J. Haematol. 2012;
96780. 159: 1046.
29 Taki T, Ida K, Hanada R et al. Frequency and clinical signicance 46 Tomizawa D, Koh K, Hirayama M et al. Outcome of recurrent or
of the MLL gene rearrangements in infant acute leukemia. Leuke- refractory acute lymphoblastic leukemia in infants with MLL gene
mia 1996; 10: 13037. rearrangements: A report from the Japan infant leukemia study
30 Drdelmann M, Reiter A, Borkhardt A et al. Prednisone response group. Pediatr. Blood Cancer 2009; 52: 80813.
is the strongest predictor of treatment outcome in infant acute lym- 47 Kikuchi A, Kigasawa H, Tsurusawa M et al. A study of
phoblastic leukemia. Blood 1999; 94: 120917. rasburicase for the management of hyperuricemia in pediatric
31 van Dongen JJ, Seriu T, Panzer-Grmayer ER et al. Prognostic patients with newly diagnosed hematologic malignancies at
value of minimal residual disease in acute lymphoblastic leukaemia high risk for tumor lysis syndrome. Int. J. Hematol. 2009;
in childhood. Lancet 1998; 352: 17318. 90: 492500.
32 Coustan-Smith E, Behm FG, Sanchez J et al. Immunological detec- 48 Salzer WL, Jones TL, Devidas M et al. Modications to induction
tion of minimal residual disease in children with acute therapy decrease risk of early death in infants with acute lympho-
lymphoblastic leukaemia. Lancet 1998; 351: 5504. blastic leukemia treated on Childrens Oncology Group P9407.
33 Van der Velden VHJ, Corral L, Valsecchi MG et al. Prognostic sig- Pediatr. Blood Cancer 2012; 59: 8349.
nicance of minimal residual disease in infants with acute 49 Mori M, Onodera M, Morimoto A et al. Palivizumab use in
lymphoblastic leukemia treated within the interfant-99 protocol. Japanese infants and children with immunocompromised
Leukemia 2009; 23: 10739. conditions. Pediatr. Infect. Dis. J. 2014; 33: 11835.
34 Pieters R, den Boer ML, Durian M et al. Relation between age, 50 Brown P, Levis M, Shurtleff S, Campana D, Downing J, Small D.
immunophenotype and in vitro drug resistance in 395 children with FLT3 inhibition selectively kills childhood acute lymphoblastic
acute lymphoblastic leukemia: Implications for treatment of in- leukemia cells with high levels of FLT3 expression. Blood 2005;
fants. Leukemia 1998; 12: 13448. 105: 81220.
35 Stam RW, den Boer ML, Meijerink JPP et al. Differential mRNA 51 Stam RW, den Boer ML, Schneider P et al. Targeting FLT3 in pri-
expression of Ara-C-metabolizing enzymes explains Ara-C sensi- mary MLL-gene-rearranged infant acute lymphoblastic leukemia.
tivity in MLL gene-rearranged infant acute lymphoblastic Blood 2005; 106: 248490.
leukemia. Blood 2003; 101: 12706. 52 Stam RW, Schneider P, de Lorenzo P, Valsecchi MG, den Boer ML,
36 Mann G, Attarbaschi A, Schrappe M et al. Improved outcome with Pieters R. Prognostic signicance of high-level FLT3 expression in
hematopoietic stem cell transplantation in a poor prognostic sub- MLL-rearranged infant acute lymphoblastic leukemia. Blood 2007;
group of infants with mixed-lineage-leukemia (MLL)-rearranged 110: 27745.
acute lymphoblastic leukemia: Results from the interfant-99 study. 53 Stumpel DJPM, Schneider P, van Roon EHJ et al. Specic pro-
Blood 2010; 116: 264450. moter methylation identies different subgroups of MLL-
37 Lnnerholm G, Valsecchi MG, de Lorenzo P et al. Pharma- rearranged infant acute lymphoblastic leukemia, inuences clinical
cokinetics of high-dose methotrexate in infants treated for outcome, and provides therapeutic options. Blood 2009; 114:
acute lymphoblastic leukemia. Pediatr. Blood Cancer 2009; 54908.
52: 596601. 54 Stumpel DJPM, Schneider P, van Roon EHJ, Pieters R, Stam RW.
38 Ishii E, Okamura J, Tsuchida M et al. Infant leukemia in Japan: Absence of global hypomethylation in promoter hypermethylated
Clinical and biological analysis of 48 cases. Med. Pediatr. Oncol. mixed lineage leukaemia-rearranged infant acute lymphoblastic
1991; 19: 2832. leukaemia. Eur. J. Cancer 2013; 49: 17584.

2015 Japan Pediatric Society


Treatment progress in infant ALL 819

55 Stumpel DJPM, Schneider P, Seslija L et al. Connectivity map- 58 Hoffman LM, Gore L. Blinatumomab, a bi-specic anti-CD19/
ping identies HDAC inhibitors for the treatment of t(4;11)- CD3 BiTE() antibody for the treatment of acute lymphoblastic
positive infant acute lymphoblastic leukemia. Leukemia 2012; leukemia: Perspectives and current pediatric applications. Front
26: 68292. Oncol 2014; 4: 63.
56 Daigle SR, Olhava EJ, Therkelsen CA et al. Selective killing of 59 Maude SL, Frey N, Shaw PA et al. Chimeric antigen receptor T
mixed lineage leukemia cells by a potent small-molecule DOT1L cells for sustained remissions in leukemia. N. Engl. J. Med. 2014;
inhibitor. Cancer Cell 2011; 20: 5365. 371: 150717.
57 Daigle SR, Olhava EJ, Therkelsen CA et al. Potent inhibition of
DOT1L as treatment of MLL-fusion leukemia. Blood 2013; 122:
101725.

2015 Japan Pediatric Society

You might also like