You are on page 1of 13

Seminar

Acute lymphoblastic leukaemia


Hiroto Inaba, Mel Greaves, Charles G Mullighan

Acute lymphoblastic leukaemia occurs in both children and adults but its incidence peaks between 2 and 5 years of age. Lancet 2013; 381: 1943–55
Causation is multifactorial and exogenous or endogenous exposures, genetic susceptibility, and chance have roles. Published Online
Survival in paediatric acute lymphoblastic leukaemia has improved to roughly 90% in trials with risk stratification by March 22, 2013
http://dx.doi.org/10.1016/
biological features of leukaemic cells and response to treatment, treatment modification based on patients’
S0140-6736(12)62187-4
pharmacodynamics and pharmacogenomics, and improved supportive care. However, innovative approaches are
Department of Oncology
needed to further improve survival while reducing adverse effects. Prognosis remains poor in infants and adults. (H Inaba MD) and Department of
Genome-wide profiling of germline and leukaemic cell DNA has identified novel submicroscopic structural genetic Pathology (C G Mullighan MD),
changes and sequence mutations that contribute to leukaemogenesis, define new disease subtypes, affect responsiveness St Jude Children’s Research
Hospital and University of
to treatment, and might provide novel prognostic markers and therapeutic targets for personalised medicine.
Tennessee Health Science
Center, Memphis, TN, USA; and
Introduction are based on reproducible data or are biologically Haemato-Oncology Research
An estimated 6000 new cases (male:female prevalence of plausible. Some of these candidate exposures are of Unit, Division of Molecular
Pathology, Institute of Cancer
roughly 1·3:1) of acute lymphoblastic leukaemia are public concern, especially ionising and non-ionising (eg,
Research, Sutton, UK
diagnosed yearly in the USA.1 Patients are mainly electromagnetic field) radiation. Ionising radiation is (Prof M Greaves PhD)
children; roughly 60% of cases occur in people aged an established causal exposure for childhood acute Correspondence to:
younger than 20 years.2–5 Survival in childhood acute lymphoblastic leukaemia, as evidenced by the effects of Dr Hiroto Inaba, St Jude
lymphoblastic leukaemia is approaching 90% (appen- the 1945 atomic bombs in Japan13 and the slightly but Children’s Research Hospital,
262 Danny Thomas Place,
dix),4,6 but treatment in infants (ie, children younger than significantly increased risks associated with x-ray
Memphis, TN 38105, USA
12 months) and adults needs improvement.5,7 We review pelvimetry during pregnancy.14 Such exposures are no hiroto.inaba@stjude.org
advances in the epidemiology, pathobiology, and clinical longer relevant, although some scientists argue that
management of acute lymphoblastic leukaemia. background or natural radiation could be important.15 See Online for appendix
Exposures to electromagnetic fields (eg, via power lines)
Epidemiology have been particularly controversial, and concern or
Acute lymphoblastic leukaemia, like cancer in general, confusion might be exacerbated by uncritical news
probably arises from interactions between exogenous or reporting. A meta-analysis16 suggests that high levels of
endogenous exposures, genetic (inherited) susceptibility, electromagnetic field radiation (ie, magnetic flux densities
and chance (figure 1). These factors account for the >0·2 μT) are associated with slightly increased risk, but
roughly 1 in 2000 risk of the disease in childhood the reliability of this finding is uncertain. To prove that
(0–15 years). The challenge is to identify the relevant exposure to electromagnetic fields never causes acute
exposures and inherited genetic variants and decipher lymphoblastic leukaemia is impossible, but at most such
how and when these factors contribute to the multistep radiation might be implicated in only a few cases.
natural history of acute lymphoblastic leukaemia from Furthermore, the likelihood that exposure to electro-
initiation (usually in utero) through the largely covert magnetic field radiation is causal in promotional or late-
evolution to overt disease.8 The rarity of the illness and stage leukaemia development is lessened by the absence
the existence of biologically distinct subtypes that might of any biological mechanism or credible modelling in
not share common causative mechanisms complicates vitro or in vivo.
matters.12 For example, in infants acute lymphoblastic Infection was the first suggested causal exposure for
leukaemia is usually associated with MLL rearrangement, childhood acute lymphoblastic leukaemia17 and remains
and the remarkably high concordance rate in monozygotic the strongest candidate. Two specific hypotheses have
twins (approaching 100% in those with a single or mono-
chorionic placenta) suggests that leukaemogenesis is
largely complete at birth.9 By contrast, incidence of non- Search strategy and selection criteria
MLL-rearranged B lymphoblastic leukaemia peaks We searched Medline and PubMed with the keywords “acute lymphoblastic leukaemia”,
between 2 and 5 years and has a concordance rate of “acute lymphocytic leukaemia”, and “acute lymphoid leukaemia” for articles published in
10–15%, suggesting that, although initiation in utero English between Jan 1, 2007, and Nov 30, 2012. Additional information was obtained
usually occurs, other so-called promotional exposures are from abstracts presented to the American Society of Hematology and American Society
probably necessary for disease emergence.9 of Clinical Oncology. We focused on publications from the past 5 years, but did not
exclude commonly referenced and highly regarded older publications. We also searched
Contributing exposures the reference lists of articles identified by this search strategy and selected those we
Exposures and their roles remain contentious. More than judged relevant. Review articles and book chapters are cited to provide readers with more
20 candidate exposures that contribute to childhood details and more references than this Seminar can provide. Our reference list was
disease have been identified through epidemiological modified on the basis of comments from peer reviewers.
and case-control studies,10 but very few of these findings

www.thelancet.com Vol 381 June 1, 2013 1943


Seminar

lymphoblastic leukaemia (roughly 40 fold at age 0–4 years)


Exposures and acute myeloid leukaemia.29 The seeming absence of
familial clustering of acute lymphoblastic leukaemia or
In-utero Postnatal
Acute greatly raised sibling risk (maximum 2 fold) does not,
Chance lymphoblastic
initiation promotion
leukaemia
however, argue against inherited susceptibility. Previous
attempts to identify inherited genetic susceptibility to the
disease have been based on a candidate gene approach.
Inherited genetic variation
Such studies30 identified some noteworthy potential
candidates implicated in, for example, folate metabolism
Figure 1: Composite causality of childhood acute lymphoblastic leukaemia
Exogenous (eg, infection) and endogenous (eg, inflammation, oxidative stress)
and the immune response, but most were statistically
exposures, normal allelic variation in inherited genes, and chance all have roles in underpowered or not consistently reproducible.
the covert natural history of childhood acute lymphoblastic leukaemia.8–10 Cancer Genome-wide association studies of childhood acute
causation is closely linked with chance11—eg, incidental external exposure, lymphoblastic leukaemia compare the whole genome
damage to a relevant oncogene in a relevant cell (stem or progenitor cell), chance
events at conception implicating parental gene shuffling and recombination.
(usually remission-blood-derived DNA) in large series of
patients to that in an ethnically matched control group
and focus on single nucleotide polymorphisms in DNA
been proposed, are often referred to by their eponymous sequences (with roughly 80% genome coverage).31–33
titles, and are supported by epidemiological data These studies need hundreds or thousands of patients
(table 1).11,17,18 Both postulate that the disease results from and controls, and, in view of the thousands of pan-
an abnormal response to a common infection. The genome markers being compared, a p of less than 10–⁷ is
hypotheses differ in detail but are not mutually exclusive generally needed to deem the result robust. Genome-
as explanations of rare time–space clusters of leukae- wide association studies should be validated in a second,
mia19,25 or acute lymphoblastic leukaemia in the general independent series of patients and independently
community.10 A unique or single transforming virus,10 confirmed by another research group. So far, common
which has been noted in leukaemia in some animal allelic variants in IKZF1, ARID5B, CEBPE, and CDKN2A
species, has not been detected in acute lymphoblastic have been significantly and consistently associated with
leukaemia.21 Rather, the illness is probably promoted childhood acute lymphoblastic leukaemia (appendix).31–33
indirectly by an abnormal or dysregulated immune Variants in other genes probably have odds ratios (or
response to one or more common infections (viral or impact) less than 1·2, but can be identified only in very
bacterial) in susceptible individuals. Influenza viruses large cooperative studies (3000–5000 cases).
are plausible candidates.26 In such a scenario, susceptible Gene variants have additive effects; someone inheriting
children would have minimum previous exposure to one copy of a variant will have a roughly 50% increased
infection during infancy, a persistent in-utero-generated risk whereas someone who inherits all four variants
preleukaemic clone,10 and a variable degree of genetic in homozygous form would have a roughly 10-fold
susceptibility. Exploration of the possible biological increased risk. The overall conclusion from these studies
mechanisms of infectious promotion should continue,27 is that the risk of childhood acute lymphoblastic
because it could lead to prophylactic interventions. leukaemia is affected by co-inheritance of several low-
risk variants. That the incidence of B lymphoblastic
Inherited susceptibility leukaemia with high hyperdiploidy is lower in African-
Very little evidence shows inherited predisposition via American children than in other children in the USA
highly penetrant mutations in children or adults.28 The might partly be a result of lower prevalence of the
high concordance in identical twin children has a non- ARID5B risk allele (appendix).34 Inherited allelic variation
genetic explanation (blood cell chimaerism).9 Infants might also affect response to treatment.35
born with constitutive trisomy 21 or Down’s syndrome The products of the four genes implicated by genome-
are, however, at substantially increased risk of acute wide association studies do not impinge directly on

Hypothesis Key notions Timing Agent Evidence


Kinlen population Unusual demographic mixing Herd immunity20 Possibly Possibly a single Transiently increased incidence
mixing of susceptible and infected Animal leukaemia precedents21 perinatal novel virus (roughly double) in several
hypothesis18,19 individuals situations of population mixing
or clusters19
Greaves delayed Delayed exposure to Mismatch between evolutionary Late One or (more Reduced risk associated with
infection common infections in programming of immune system promotional probably) several day-care attendance in infancy23,34
hypothesis10,22 childhood because of and modern (hygienic) lifestyle10 or triggering common infections
underexposure as infants Two-step prenatal and postnatal event10 (bacterial or viral)
natural history10

Table 1: Infection-based hypotheses for childhood acute lymphoblastic leukaemia

1944 www.thelancet.com Vol 381 June 1, 2013


Seminar

potential exposure pathways (eg, immune, liver detoxifi- Others (T-cell disease) 2%
cation), but rather are key regulators of blood cell TLX1 0·3%
development, proliferation, and differentiation. Acquired LYL1 1·4%
ETP 2% ETV6–RUNX1 22%
or somatic mutations of each of these genes have been TLX3 2·3%
TAL1 7%
detected in acute lymphoblastic leukaemia, suggesting
that the inherited gene variants contribute to the intrinsic
vulnerability of stem or precursor blood cells to trans-
forming events either in utero at initiation or with Others
(B-cell disease) 9%
subsequent postnatal promotion and clonal evolution, or
both (figure 1). Evidence suggests that the risk-conferring
gene variants have lowered expression of products,31 but ERG 3%
functional aspects remain to be explored. These data
provide valuable insights into the cause of childhood
disease, but do not have enough predictive value to merit (Other) 5·5% Hyperdiploid
BCR–ABL1-
like 9% (>50 chromosomes)
screening of all children. (CRLF2) 3·5% 20%

CRLF2 4%
Pathobiology
iAMP21 2%
Genetic basis MLL
rearrangements 6%
High-resolution profiling of genetic alterations has Hypodiploid (<44 chromosomes) 1%

transformed understanding of the genetic basis of acute Dicentric 3% TCF3–PBX1 4%


lymphoblastic leukaemia. That most childhood cases BCR–ABL1 2%
harbour gross chromosomal alterations has been known
for several decades (figure 2).36 In B-cell disease, these Figure 2: Cytogenetic and molecular genetic abnormalities in childhood acute lymphoblastic leukaemia
alterations include high hyperdiploidy with non-random Acute lymphoblastic leukaemia with rearrangement of CRLF2 but without the BCR–ABL1-like transcriptional profile
rarely presents with other classifying karyotypic alterations, but can be noted with high hyperdiploidy. Dicentric
gain of at least five chromosomes (including X, 4, 6, 10, cases might have a range of translocations, including classifying translocations (eg, ETV6–RUNX1).
14, 17, 18, and 21); hypodiploidy with fewer than iAMP21=intrachromosomal amplification of chromosome 21. ETP=early T-cell precursor.
44 chromosomes; and recurring translocations including
t(12;21)(p13;q22) encoding ETV6–RUNX1, t(1;19)(q23;p13)
encoding TCF3–PBX1, t(9;22)(q34;q11) encoding BCR–
ABL1, rearrangement of MLL at 11q23 with a wide range Initiation Lesion generation
of partner genes, and rearrangement of MYC into antigen Initiating lesion Aberrant RAG activity
receptor gene loci. Dysregulation of TAL1, TLX1, TLX3, (eg, ETV6–RUNX1, Developmental arrest
MLL rearrangement) Alterations in B-cell transcription
and LYL1, particularly by rearrangement into T-cell confers self-renewal factor genes (eg, PAX5, IKZF1, EBF1)
antigen receptor loci, often occurs in T lymphoblastic
leukaemia. These changes are of key importance in both
pathogenesis and clinical management (figure 3).
Many chromosomal rearrangements disrupt genes
that regulate normal haemopoiesis and lymphoid de-
velopment (eg, RUNX1, ETV6), activate oncogenes (eg, Haemopoietic Pro-B-cell/ Mature
MYC), or constitutively activate tyrosine kinases (eg, stem cell/ Pre-B-cell B cell
ABL1). Several are significantly associated with outcomes, lymphoid
progenitor Predisposition Cooperating events
particularly in B-cell disease, and are used in risk Inherited variants • Cell cycle and tumour suppressors
(eg, IKZF1, CEBPE (CDKN2A/CDKN2B [INK4/ARF], TP53, RB1)
stratification. High hyperdiploidy and ETV6–RUNX1 ARID5B, CDKN2A) • Cytokine receptor and kinases (CRLF2, JAK1, JAK2, ABL1, PDGFRB)
rearrangement are associated with favourable outcome, • Ras signalling (NRAS, KRAS, NF1, PTPN11)
whereas low hypodiploidy and MLL rearrangement • Lymphoid signalling (BTLA, TOX, CD200)
• Transcription factors, coregulators, coactivators
(especially in infants and adults) are associated with poor • Mutations in epigenetic regulators (CREBBP)
prognosis in both children and adults. • Other
However, many of these alterations alone do not induce Selective pressure
leukaemia in experimental models, and no gross chromo- (chemotherapy)

somal alteration is noted in many cases, suggesting that


additional submicroscopic genetic alterations contribute to
leukaemogenesis. High-resolution microarray profiling of
DNA copy number alterations (deletions and gains) and Genetic alterations that
confer resistance to treatment
sequencing have led to identification of several novel (subclonal at diagnosis, or acquired)
structural genetic alterations and sequence mutations Diagnosis (eg, IKZF1, CREBBP, TP53) Relapse
(some of which are being investigated as prognostic
markers or therapeutic targets) that define new subtypes of Figure 3: Genetic pathogenesis of B lymphoblastic leukaemia at diagnosis and relapse

www.thelancet.com Vol 381 June 1, 2013 1945


Seminar

Frequency Pathway and consequences Clinical relevance


PAX5
Focal deletions, 31·7% Transcription factor needed for B-lymphoid Not associated with adverse
translocations, sequence development; mutations impair DNA binding and outcome
mutations transcriptional activation
IKZF1
Focal deletions, sequence 15% of paediatric cases Transcription factor needed for development of Associated with poor outcomes
mutations More than 80% of BCR–ABL1 cases haemopoietic stem cells to lymphoid precursors;
and 66% of cases of chronic myeloid deletions and mutations result in loss of function or
leukaemia in lymphoid blast crisis dominant negative isoforms
A third of cases of high-risk Tripling of cumulative incidence
BCR–ABL1-negative disease of relapse
Inherited variants ·· ·· Increased risk of developing
disease
JAK1, JAK2
Pseudokinase and kinase 18–35% of Down’s-syndrome- Mutations cause constitutive JAK–STAT activation; ··
domain mutations associated cases, 11% of high-risk transforms mouse Ba/F3-EpoR haemopoietic cell
BCR–ABL1-negative cases line
CRLF2
Rearrangement as IGH@– 5–16% of paediatric and adult cases Associated with mutant JAK in as much as 50% of ··
CRLF2 or P2RY8–CRLF2 and >50% of cases associated with cases; CRLF2 and JAK mutations cotransform in
resulting in overexpression Down’s syndrome Ba/F3 cells, causing constitutive STAT activation
14% of paediatric high-risk cases of Associated with IKZF1 alteration and JAK mutations Associated with poor outcome
B lymphoblastic leukaemia
CREBBP
Focal deletion and sequence 19% of relapsed cases of B Impaired histone acetylation and transcriptional Mutations selected for at relapse
mutations lymphoblastic leukaemia* regulation and associated with
glucocorticoid resistance

A referenced version of this table is in the appendix. *Also mutated in non-Hodgkin lymphoma.

Table 2: Key genetic alterations in B lymphoblastic leukaemia, by gene

disease, contribute to leukaemogenesis, and affect treat- B-cell disease, and the CDKN2A/CDKN2B loci encoding
ment responsiveness.37–42 Importantly, many subtypes of the INK4/ARF tumour suppressors, which are implicated
acute lymphoblastic leukaemia are characterised by in more than 80% of T-cell cases. Although understanding
distinct constellations of structural genetic alterations that of sequence mutations is incomplete, data show that
together drive establishment of the leukaemic clone. several genes are changed by various mechanisms
(including deletion or amplification, sequence mutation,
Submicroscopic genetic alterations and translocation) that are highly gene-dependent—eg,
More than 50 regions of recurring DNA copy number structural alterations are more common than are
alteration have been identified in acute lymphoblastic sequence mutations in PAX5 and IKZF1 in B-cell disease,
leukaemia.37,38,43–47 Deletions are more common than is whereas WT1, PHF6, and NOTCH1 are frequently
amplification and are typically focal and often implicate targeted by sequence alteration in T-cell disease.
only one gene. The nature and frequency of these alter- Several genetic alterations have well established
ations are associated with cell lineage and cytogenetic roles in leukaemogenesis (eg, activating mutations in
disease subtype. Notably, MLL-rearranged acute lympho- NOTCH1). The roles of many other genes remain un-
blastic leukaemia, which is typically aggressive and arises known, partly because of the paucity of murine models
early in life, harbours, on average, roughly one additional that faithfully represent human acute lymphoblastic
genetic alteration per case, whereas diseases associatied leukaemia. However, several alterations disrupt the
with ETV6–RUNX1 and BCR–ABL1 translocations mani- activity of the encoded proteins in vitro or result in
fest later in childhood and typically harbour between at dominant negative activity, and loss-of-function muta-
least six and eight additional genetic alterations. Many of tions in PAX5 and IKZF1 accelerate the onset of
the implicated genes encode regulators of lymphoid B lymphoblastic leukaemia in murine models.48,49
development or cell cycle, tumour suppressors, or
lymphoid signalling molecules (figure 3, table 2). The Prognostic genetic alterations
most frequently altered genes are transcriptional regu- Because cytogenetic alterations alone do not accurately
lators of B-cell lymphoid development (eg, PAX5, IKZF1, predict the risk of relapse, relations between novel
EBF1), which are affected in more than two-thirds of genetic alterations and outcomes are of interest. The

1946 www.thelancet.com Vol 381 June 1, 2013


Seminar

most consistent association is between IKZF1 alterations As much as 50% of BCR–ABL1-like cases have CRLF2
and poor outcome (table 2). IKZF1 encodes IKAROS, rearrangements and JAK mutations. Next-generation
which is needed for lymphoid lineage development sequencing, including transcriptome and whole-genome
(figure 3). IKZF1 is altered in 15% of cases of sequencing, has shown that the remaining cases harbour
B lymphoblastic leukaemia and is a hallmark of two a diverse range of rearrangements, deletions, and
high-risk subtypes—namely, BCR–ABL1 lymphoblastic sequence mutations that activate cytokine-receptor and
leukaemia (either de novo or chronic myeloid leukaemia kinase signalling (eg, those implicating ABL1, EPOR,
in lymphoid blast crisis)38,50 and BCR–ABL1-like acute IL7R, JAK2, and PDGFRB); several of these alterations
lymphoblastic leukaemia.51,52 transform in vitro and activate kinase signalling in
IKZF1 alterations include loss-of-function deletions of primary leukaemic cells.58
the entire locus, focal deletions resulting in expression of Intrachromosomal amplification of chromosome 21
dominant negative IKZF1 alleles, and, less frequently, occurs in roughly 2% of cases of B lymphoblastic
sequence mutations. Alteration is associated with in- leukaemia (figure 2),59 and was originally defined by
creased risk of treatment failure and relapse in both fluorescence in-situ hybridisation as a gain of at least
BCR–ABL1-positive and BCR–ABL1-negative disease. three copies of RUNX1.60 Subsequent studies showed that
Although testing for IKZF1 alterations at diagnosis has the amplification region is typically large and complex
not yet been incorporated into frontline trials of acute and often accompanied by deletion of the subtelomeric
lymphoblastic leukemia, it is being assessed in regions of chromosome 21.61 The functional consequences
prospective clinical studies. of the amplication are largely unknown, but identification
is important because this subtype is associated with poor
Novel subtypes outcomes with standard-risk regimens.59,62
As much as 8% of childhood cases have CRLF2 rearrange-
ment at pseudoautosomal region 1 of Xp/Yp (figure 2; Genetics of relapse and clonal heterogeneity
table 2). CRLF2 encodes the receptor for thymic stromal Cytogenetic63 and genomic profiling of samples collected
lymphopoietin.40,41 Either rearrangement of CRLF2 into at diagnosis and relapse shows substantial changes in
the immunoglobulin heavy chain locus at 14q32 or a the nature of genetic alterations during the course of
focal deletion immediately upstream of the gene occurs, most cases and that relapse often arises from the
resulting in the novel fusions IGH@–CRLF2 and emergence of a minor subclone with genetic alterations
P2RY8–CRLF2, respectively. Both events dysregulate distinct from those of the predominant clone at diagnosis
CRLF2 expression, leading to increased expression by (figure 3).64–66 In most cases, the relapse clone shares
lymphoblasts that might be detected by diagnostic lesions with the predominant clone at diagnosis,
immunophenotyping. Less common is a p.Phe232Cys suggesting a common preleukaemic origin, but other
mutation that results in receptor dimerisation.53 CRLF2 lesions are discordant. Sensitive assays specific for
rearrangement is particularly common (>50% of cases) individual alterations show that the relapse clone is often
in cases associated with Down’s syndrome. present in low proportions at diagnosis, suggesting that
Roughly 50% of cases with CRLF2 rearrangement relapse alterations confer resistance to treatment.
harbour concomitant activating mutations in JAK1 or Furthermore, several of the alterations that most often
JAK2 (table 2).40,41,54 Particularly in disease not associated emerge at relapse are also associated with poor treatment
with Down’s syndrome, CRLF2 and JAK alterations are outcomes when present at diagnosis—eg, deletions of
associated with deleterious IKZF1 alterations and poor IKZF1, CDKN2A/CDKN2B. Less frequently, the relapse
outcomes.55 JAK mutations are located either at or near clone seems identical or completely dissimilar to that at
the active site of the JAK kinase domain or in the diagnosis. Similar findings were noted in candidate-
pseudokinase domains of JAK1 or 2, most commonly at gene-sequencing studies of relapsed disease, which
p.Arg683 of JAK2. Importantly, the JAK2 mutations that identified enrichment of loss-of-function mutations of
are common in acute lymphoblastic leukaemia are the transcriptional coactivator and acetyltransferase
distinct from those noted in myeloproliferative neo- CREBBP (encoding CREB-binding protein) and TP53
plasms.56 Coexpression of CRLF2 and JAK mutant (table 2, figure 3).42,67,68
alleles transforms model cell lines and activates down-
stream JAK-STAT signalling, suggesting that these Genome sequencing
alterations are cotransforming in B-cell disease.41 In Next-generation sequencing enables comprehensive
disease associated with Down’s syndrome, alterations of identification of the genetic changes in leukaemia.
IKZF1, but not of CRLF2 or JAK2, were associated with Simultaneous sequencing of hundreds of thousands of
poor prognosis.57 nucleic acids (so-called massively parallel sequencing)
10–12% of cases of B lymphoblastic leukaemia have a might be used to identify sequence mutations and
gene expression profile similar to that of BCR–ABL1 structural variants in the encoding portion of the
disease but are BCR–ABL1 negative, frequently show genome (exome sequencing), the transcriptome
IKZF1 alteration, and have poor outcomes (figure 2).51,52 (mRNA sequencing), or the entire genome. In 187 cases

www.thelancet.com Vol 381 June 1, 2013 1947


Seminar

of high-risk B lymphoblastic leukaemia, 120 candidate Risk assignment


genes and pathways targeted by DNA copy number Clinical and biological factors
alterations were sequenced; a high frequency of Age (infant or ≥10 years old), presenting leucocyte count
alterations targeting B lymphoid development (68%), (≥50 × 10⁹/L), race (Hispanic or black), male sex, and
the TP53–RB1 tumour suppressor pathway (54%), Ras T-cell immunophenotype are adverse clinical prognostic
signalling (50%), and Janus kinases (11%), and recurring factors in children, although their effect is diminished by
mutations in genes including ETV6, TBL1XR1, CREBBP, contemporary risk-adapted treatment and improved
MUC4, ASMTL, and ADARB2 were identified.69 supportive care.2–6 Infants with MLL rearrangements,
Early T-cell precursor acute lymphoblastic leukae- especially those younger than 6 months old with more
mia is an aggressive leukaemia characterised by an than 300 × 10⁹ leucocytes per L at diagnosis, still have a
immature immunophenotype reminiscent of the dismal prognosis.7
murine thymic early T-cell precursor,70 aberrant expres- Racial differences in prognosis have been linked not
sion of myeloid and stem cell markers, a distinct gene only to socioeconomic factors but also to differences in
expression profile, and dismal outcomes.71 Whole- genomic variarions.55,74,75 For example, germline single
genome sequencing of 12 cases, and mutational-recur- nucleotide polymorphisms of PDE4B and ARID5B are
rence testing in an additional 94 patients with T associated with Native American genetic ancestry,74,75 and
lymphoblastic leukaemia (52 of whom had the early somatic CRLF2 rearrangements in acute lymphoblastic
T-cell precursor form) showed that three pathways leukaemia blasts55 were over-represented in children from
frequently mutated in acute myeloid leukaemia were a Hispanic background; these alterations contributed to
mutated at high frequency in early T-cell precursor inferior outcomes in Hispanic children. Adverse prognosis
disease72—specifically, inactivating mutations targeting conferred by genetic ancestry was mitigated by adding a
haemopoietic and lymphoid development (including course of delayed intensification therapy.74
GATA3, ETV6, RUNX1, and IKZF1), mutations driving Adolescents and adults have a greater prevalence of
aberrant cytokine receptor and Ras signalling (NRAS, biologically high-risk leukaemia (eg, BCR–ABL1, MLL
KRAS, FLT3, JAK1, JAK3, and IL7R), and deleterious rearrangement), a lower incidence of favourable subtypes
mutations in chromatin-modifying genes, most notably (eg, ETV6–RUNX1, hyperdiploidy), and poorer adherence
components of polycomb repressor complex 2 (EZH2, to, and tolerance of, treatment than do pre-adolescent
EED, and SUZ12). Polycomb repressor complex 2 children.76 Older age (especially ≥60 years) and high
normally mediates trimethylation of H3K27, resulting presenting leucocyte count are poor prognostic factors in
in transcriptional repression; thus, these mutations are adults (appendix). Adolescents and adults seem to have
predicted to derepress transcription. These results better outcomes with paediatric than with adult
extend findings of PHF6 mutations in T-cell disease, regimens.76–80 Typically, paediatric regimens provide higher
which were obtained by sequencing X chromosome doses of non-myelosuppressive drugs, early and frequent
genes and explained the increased incidence in men intrathecal therapy, reinduction and long maintenance
and boys.73 phases, and strict oversight of adherence.
Whole-genome sequencing of all subtypes is needed
to identify all genetic alterations contributing to Response to treatment
leukaemogenesis. Additionally, studies of the nature of Early response is predictive of the risk of relapse and
non-coding genetic mutations and the interaction of is used to assign patients to subsequent risk-adapted
genetic, epigenetic, and transcriptomic factors will be treatment.81 Methods that track residual leukaemic cells
of great interest. by flow cytometry (for aberrant immunophenotypes) and
PCR amplification (for leukaemia-specific immuno-
Diagnosis globulin and T-cell receptor genes or fusion transcripts)
Morphological identification of lymphoblasts by micros- allow detection at proportions less than those detectable
copy and immunophenotypic assessment of lineage com- by microscopic morphological assessment—ie, minimal
mitment and developmental stage by flow cytometry are residual disease (MRD). MRD is the most powerful prog-
essential for diagnosis.2 Chromosomal analysis still has an nostic indicator in childhood and adult disease, even in
important role in the initial cytogenetic work-up. Reverse patients with low-risk features at presentation.6,82–86
transcriptase PCR, fluorescence in-situ hybridisation or The kinetics of MRD clearance in response to identical
multiplex ligation-dependent probe amplification, and remission-induction chemotherapy differs between
flow cytometry are used to identify leukaemia-specific disease subtypes; negative MRD on day 33 (after adminis-
translocations, submicroscopic chromosomal abnor- tration of four drugs) was the strongest prognostic factor
malities, and cellular DNA content, respectively. When in B-cell disease,83 whereas negative MRD on day 78 (after
genome-wide analysis becomes time effective and cost seven drugs) was also predictive in T-cell disease,
effective, it might replace many diagnostic techniques. irrespective of positive MRD status on day 33.84
Tests that have prognostic and therapeutic implications are PCR is typically more sensitive than is flow cytometry
listed in the appendix. for measurement of MRD (roughly 0·001% vs roughly

1948 www.thelancet.com Vol 381 June 1, 2013


Seminar

0·01%), and PCR-measurable low proportions of MRD recommended for adolescents with B lymphoblastic
(0·001–<0·01%) after remission-induction therapy have leukaemia during remission-induction therapy.96
prognostic importance in children.87 However, flow Three preparations of asparaginase are available. One
cytometry is faster, generally less expensive, and appli- is derived from Escherichia coli and one from Erwinia
cable to a larger proportion of patients,81 allowing early caratovora; the third is a monoethoxypolyethylene
tailoring of treatment.85 The sensitivity of flow cytometry glycol succinimidyl conjugate of E coli L-asparaginase
can be improved with multicolour combinations of (PEG-asparaginase).97 These formulations have dif-
additional leukaemia-associated markers identified ferent half-lives (PEG-asparaginase>E–coli-derived>E-
from differently expressed genes in acute lymphoblastic caratovora-derived), and maintenance of asparagine
leukaemia cells, yielding a detection threshold of depletion by optimising dose intensity and schedule is
roughly 0·001%.88 crucial. PEG-asparaginase has largely replaced the native
E coli product, because it provides at least 2 weeks of
Treatment therapeutic activity after a single dose and induces
Treatment typically spans 2–2·5 years, and comprises antibodies less frequently.98,99 Native-E-coli-asparaginase
three phases: induction of remission, intensification and PEG-asparaginase activity could be inversely related
(or consolidation), and continuation (or maintenance).2 to titres of anti-E-coli-asparaginase antibodies, although
Most of the drugs used were developed before 1970. PEG-asparaginase was inhibited only at high titres.100
However, their dosages and schedule in combination Therefore, PEG-asparaginase might be used when
chemotherapy have been optimised on the basis of antibody titres are low or intermediate, and E caratovora
leukaemic-cell biological features, response to treatment asparaginase when titres are high. Glucocorticoids and
(MRD), and pharmacodynamic and pharmacogenomic asparaginase have a noteworthy pharmacokinetic inter-
findings in patients, resulting in the high survival rate. action.101,102 High systemic exposure to asparaginase was
CNS-directed treatment prevents relapse due to associated with high exposure to dexamethasone,
sequestered leukaemia cells. Allogeneic haemopoietic presumably because of impaired hepatic synthesis of
stem cell transplantation is an option for patients at very proteins implicated in dexamethasone clearance. Thus,
high risk. We focus on the most important advances anti-asparaginase antibodies can reduce exposure to both
during the past 5 years. drugs and might increase relapse risk.102
Patients with BCR–ABL1-positive disease have poor
Remission-induction therapy prognoses but benefit from early treatment with tyrosine-
4–6 weeks of remission-induction therapy eradicates kinase inhibitors (eg, imatinib, dasatinib). When tyrosine-
the initial leukaemic cell burden and restores normal kinase inhibitors are added to multidrug chemotherapy,
haemopoiesis in 96–99% of children and 78–92% of complete remission rates are more than 90% and event-
adults with acute lymphoblastic leukaemia.2–5 Chemo- free survival is superior to that in historical controls.103,104
therapy generally includes a glucocorticoid (prednisone Unlike imatinib, dasatinib targets both ABL1 and Src
or dexamethasone), vincristine, and asparaginase, with kinases; it also has more potent activity against BCR–ABL1,
or without anthracycline. This regimen seems sufficient is active against imatinib-resistant BCR–ABL1 (except that
for standard-risk disease when intensified postremission with T315I mutation), and has better CNS penetration.104–106
treatment is given. Patients at high or very high risk
receive four or more drugs. Intensification (consolidation) therapy
Prednisone (or prednisolone, which is dose-equiva- Intensification (consolidation) therapy is given after
lent) has traditionally been used, but dexamethasone is remission-induction treatment, and eradicates residual
increasingly used.89 However, the optimum doses and leukaemic cells.2,3 High-dose (ie, 1–8 g/m²) methotrexate
bioequivalence of these drugs are unclear. In prospective with mercaptopurine is often given, as are frequent
randomised trials,90–93 dexamethasone provided better pulses of vincristine and glucocorticoids, uninterrupted
control of CNS leukaemia than did prednisone, and, at a asparaginase for 20–30 weeks, and reinduction therapy
prednisone-to-dexamethasone dose ratio of less than with drugs similar to those used during remission-
7 to 1, yielded better event-free survival, especially in induction therapy.
children with T-cell disease who responded well to The accumulation of the active metabolites of metho-
prednisone prephase treatment and children younger trexate—methotrexate polyglutamates (MTXPG1–7)—in
than 10 years with B-cell disease. However, when a leukaemic cells is associated with antileukaemic activity,
higher dose of prednisolone (dose ratio>7:1) was used, which can be affected by somatic and germline genetic
the two drugs showed no difference in efficacy.94,95 factors, dose and duration of methotrexate, and folinic acid
Glucocorticoids are associated with infection, osteo- rescue. Functional enzyme and somatic genetic studies
necrosis, fracture, psychosis, and myopathy; the inci- show that MTXPG1–7 accumulation varies widely between
dence of such side-effects is generally higher with subtypes of acute lymphoblastic leukaemia. Accumulation
dexamethasone than with prednisone. Thus, high- is low in TCF3–PBX1, T-cell, and ETV6–RUNX1 disease
dose dexamethasone (eg, 10 mg/m² per day) is not (and thus patients might benefit from high doses of

www.thelancet.com Vol 381 June 1, 2013 1949


Seminar

methotrexate), and high in hyperdiploid B-cell disease, to pronouncedly myelosuppressed when given thio-
especially that with gain of chromosomes 18 or 10.107–109 purines.121 They can develop secondary malignant disease,
Germline single nucleotide polymorphisms of the especially at high doses (eg, mercaptopurine 75 mg/m²
organic anion transporter polypeptide SLCO1B1 are per day).122 Adherence of less than 95% to planned
associated with high methotrexate clearance.110,111 In mercaptopurine doses is associated with relapse.123
patients with high-risk disease high-dose methotrexate Therefore, uninterrupted, pharmacogenetics-based mer-
(ie, 5 g/m² every 14 days for four doses) and mercapto- captopurine dosing is important.2
purine were more effective than were escalating-dose After thioguanine nucleotides are incorporated into
methotrexate (ie, initial dose of 100 mg/m², increasing by DNA, DNA mismatch repair enzymes exert cytotoxic
50 mg/m² every 10 days for five doses) plus PEG- effects. Deficiency of such enzymes (eg, MSH2) renders
asparaginase, and were not associated with increased leukaemic cells thiopurine-resistant.124 MSH2 expres-
acute toxic effects.112 Duration of effective serum concen- sion was low or undetectable in roughly 11% of children
trations of methotrexate is important; accumulation of with newly diagnosed acute lymphoblastic leukaemia
MTXPG1–7 was less with high-dose 4-h infusions than due to partial or complete somatic deletion of genes
with high-dose 24-h infusions.113 Folinic acid rescue is that regulate MSH2 degradation (MTOR, HERC1,
necessary after high-dose methotrexate; however, exces- PRKCZ, and PIK3C2B).124 These children had a high
sive use can counteract antileukaemic effects of metho- incidence of relapse.
trexate and increase the risk of relapse.114
Reinduction therapy is a crucial element of protocols for Haemopoietic stem cell transplantation and
acute lymphoblastic leukaemia. Intensified reinduction cellular therapy
therapy with vincristine and asparaginase improved the Allogeneic haemopoietic stem cell transplantation is an
outcome of patients with high-risk disease.115 However, an option for children with very-high-risk or persistent
identical second reinduction cycle did not improve the disease.125 Contemporary protocols with high-resolution
outcome of patients with high-risk disease and a rapid HLA typing, case-based conditioning, and improved
marrow response to 7 days of induction therapy or those supportive care have reduced relapse-related mortality,
with standard-risk disease, suggesting that residual regimen-related toxic effects, and infection.126,127 Further-
leukaemic clones after a course of reinduction therapy more, leukaemia-free survival is not affected by the
might represent intrinsic drug resistance.115,116 Whether a source of stem cells (matched related, matched unrelated,
second reinduction cycle offers benefits to patients with cord blood, or haploidentical donor).127–129
high-risk disease and a slow early response in the context In view of the development of disease detection and
of contemporary therapy is unclear. Osteonecrosis frontline therapies, indications for allogeneic haemo-
frequently occurs after reinduction therapy, especially in poietic stem cell transplantation should be reassessed
children aged 10 years or older. Alternate-week (10 mg/m² continuously. MRD of 1 cell in 10 000 or more before
per day on days 0–6 and 14–20) rather than continuous transplantation is strongly associated with relapse, and
(10 mg/m² per day on days 0–20) administration of new strategies are needed to reduce the disease burden
dexamethasone significantly reduced osteonecrosis before and after the procedure.130,131 Patients with BCR–
despite delivering a higher cumulative dose.117 ABL1-positive acute lymphoblastic leukaemia who go
into remission after multidrug chemotherapy with ABL1
Continuation therapy kinase inhibitors and young children (<6 years) with
Continuation therapy typically lasts 2 years or longer and B-cell disease in delayed remission after induction failure
comprises mainly daily mercaptopurine and weekly can be treated without transplantation.103,104,132 Is the
methotrexate with or without pulses of vincristine and procedure beneficial in infants? Its role, if any, is limited
dexamethasone. Mercaptopurine and tioguanine are struc- to a small high-risk group.133,134 Although haemopoietic
tural analogues of hypoxanthine and guanine, respectively, stem cell transplantation during first complete remission
and inhibit de-novo purine synthesis. Although tioguanine is a key element of treatment in many adult centres, the
forms the active-metabolite thioguanine nucleotides in greater application of paediatric regimens in adult
fewer steps and is more cytotoxic in vitro to lymphoblasts patients should decrease its use.5,76
than is mercaptopurine, randomised studies have not
consistently shown improvements in event-free118,119 or CNS-directed therapy
overall120 survival, and protracted doses of 40 mg/m² per Control of CNS disease is a key component of treatment.
day or more were associated with death during remission, Prophylactic cranial irradiation (12–18 Gy) is effective,
veno-occlusive disease, portal hypertension, and thrombo- but its use has been reduced or eliminated to prevent
cytopenia.118–120 Thus, mercaptopurine is preferred for acute neurotoxic effects, neurocognitive deficits, endo-
continuation therapy. crinopathies, secondary malignant disease, and excess
TPMT catalyses S-methylation of thiopurines to in- late mortality.135 In the St Jude Total XV and Dutch
active methylated metabolites. Patients with heterozygous Childhood Oncology Group acute lymphoblastic
or homozygous TPMT deficiency become moderately leukaemia-9 protocols,6,136 cranial irradiation is replaced

1950 www.thelancet.com Vol 381 June 1, 2013


Seminar

by triple intrathecal chemotherapy with methotrexate, sensitivity to concomitant chemotherapy. Such drugs
hydrocortisone, and cytarabine for all newly diagnosed could be used in infants with MLL-rearranged disease, in
patients. The 5 year cumulative risk of isolated CNS whom aberrant DNA and histone methylation are
relapse was 2·7% with the St Jude and 2·6% with the frequently observed,138,139 and in patients with CREBBP
Dutch Childhood Oncology Group protocol—ie, within mutations that encode histone acetyltransferase CREB-
the range achieved by prophylactic cranial irradiation binding protein.42
(1·5–4·5%). Patients at high risk of CNS relapse—ie, Mutations in early T-cell precursor acute lymphoblastic
those with any CNS disease (including leukaemia-cell leukaemia are also noted in acute myeloid leukaemia,
contamination as a result of traumatic spinal tap) or with suggesting that treatments for acute myeloid leukaemia or
T-cell disease6—should be given intensified intrathecal drugs that target JAK signalling might be beneficial.72
therapy during early remission-induction therapy. Monoclonal antibodies to surface antigens such as CD19,
Cranial irradiation can be reserved only for salvage CD20, CD22, and CD52 have been used in unconjugated
treatment, because the retrieval rate is high in patients form (eg, rituximab and epratuzumab), conjugated
with an isolated CNS relapse who did not receive to immunotoxins or chemotherapeutic drugs (eg, moxe-
irradiation with initial treatment.6 tumomab, inotuzumab ozogamicin), or in the form of
In a randomised study in standard-risk acute lympho- bispecific antibodies (blinatumomab).140 The incorporation
blastic leukaemia, triple-drug intrathecal treatment of rituximab into the hyper-CVAD regimen (fractionated
reduced the frequency of CNS relapse compared with cyclophosphamide, vincristine, doxorubicin, dexameth-
intrathecal methotrexate only, but was associated with asone) seems to improve outcome in adults younger than
increased risk of bone marrow and testicular relapse, 60 years with CD20-positive, BCR–ABL1-negative B
possibly because systemic therapy was less intensive.137 In lymphoblastic leukaemia.141 Blinatumomab, a bispecific,
the St Jude total XV protocol, not only excellent CNS single-chain antibody to CD19 and CD3ε, recruits and
outcomes but also excellent overall outcomes (5 year event- activates CD3 effector cytotoxic T cells and is cytotoxic to
free survival 85·6%, overall survival 93·5%) were CD19-expressing target cells bound to the other arm of the
achieved.6 Because CNS and haematological relapses are antibody; it was active against relapsed and refractory B
competing events, systemic chemotherapy with high-dose lymphoblastic leukaemia in an adult phase 2 study.142
methotrexate, intensive asparaginase, and dexamethasone, Emerging evidence shows that newly diagnosed acute
plus risk-based early intensive intrathecal chemotherapy, lymphoblastic leukaemia comprises several subclones
have substantial roles in prevention of CNS relapse.135 and that chemoresistance is frequently driven by sub-
populations harbouring genetic alterations that confer
Remaining questions and future directions resistance.64–66 Thus, efforts should be made to identify
Some subsets of acute lymphoblastic leukaemia still have patients at high risk of relapse through use of highly
an adverse prognosis. Further intensification of available sensitive methods to detect these subpopulations at
regimens is unlikely to substantially improve survival but diagnosis. Treatment should target these subpopulations
will increase short-term and long-term adverse effects. to improve the efficacy of therapeutic regimens while
Reduction of treatment intensity should be sought in reducing intensity.
patients at low risk. Studies of chronic health compli- Contributors
cations in long-term adult survivors will help to refine All authors contributed to the writing of this paper and have seen and
treatments, reducing the toxic effects of treatment. Func- approved the final version.
tional genomics and proteomics will improve under- Conflicts of interest
standing of the epidemiology and pathogenesis of HI receives unrelated research support from Bayer and Onyx. MG and
CGM declare that they have no conflicts of interest.
individual cases, allowing targeted personalised medicine
(appendix). Most epidemiologial and pathobiological Acknowledgments
Our work was supported in part by grants from the National Institutes of
studies have been developed for paediatric disease— Health (CA21765; HI, CGM), Leukaemia and Lymphoma Research UK
these should be expanded to adult patients. (MG), and the American Lebanese Syrian Associated Charities
Pharmacological inhibitors of Janus kinases (eg, (HI, CGM). CGM is a Pew Scholar in the Biomedical Sciences and a
ruxolitinib) are being investigated in childhood disease St Baldrick’s Scholar. These sponsors had no roles in writing of the
Seminar. We thank Ching-Hon Pui for critical review and Sharon Naron
harbouring CRLF2 and JAK alterations.39 Detailed for editing the manuscript.
preclinical studies in BCR–ABL1-like disease showed
References
that the ABL1 and PDGFRB inhibitors imatinib and 1 Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012.
dasatinib are effective against acute lymphoblastic CA Cancer J Clin 2012; 62: 10–29.
leukaemia cells with NUP214–ABL1 rearrangements and 2 Pui C-H, Robison LL, Look AT. Acute lymphoblastic leukaemia.
Lancet 2008; 371: 1030–43.
JAK inhibitors are effective against those with BCR– 3 Stanulla M, Schrappe M. Treatment of childhood acute
JAK2 or IL7R mutations; such cases might be candidates lymphoblastic leukemia. Semin Hematol 2009; 46: 52–63.
for targeted therapy.58 DNA and histone methyltransferase 4 Hunger SP, Lu X, Devidas M, et al. Improved survival for children
inhibitors and histone deacetylase inhibitors might and adolescents with acute lymphoblastic leukemia between
1990 and 2005: a report from the Children’s Oncology Group.
reactivate silenced tumour-suppressor genes or increase J Clin Oncol 2012; 30: 1663–69.

www.thelancet.com Vol 381 June 1, 2013 1951


Seminar

5 Bassan R, Hoelzer D. Modern therapy of acute lymphoblastic 33 Sherborne AL, Hosking FJ, Prasad RB, et al. Variation in CDKN2A
leukemia. J Clin Oncol 2011; 29: 532–43. at 9p21.3 influences childhood acute lymphoblastic leukemia risk.
6 Pui CH, Campana D, Pei D, et al. Treating childhood acute Nat Genet 2010; 42: 492–94.
lymphoblastic leukemia without cranial irradiation. N Engl J Med 34 Yang W, Trevino LR, Yang JJ, et al. ARID5B SNP rs10821936 is
2009; 360: 2730–41. associated with risk of childhood acute lymphoblastic leukemia in
7 Pieters R, Schrappe M, De Lorenzo P, et al. A treatment protocol for blacks and contributes to racial differences in leukemia incidence.
infants younger than 1 year with acute lymphoblastic leukaemia Leukemia 2010; 24: 894–96.
(Interfant-99): an observational study and a multicentre randomised 35 Yang JJ, Cheng C, Yang W, et al. Genome-wide interrogation of
trial. Lancet 2007; 370: 240–50. germline genetic variation associated with treatment response in
8 Greaves MF, Wiemels J. Origins of chromosome translocations in childhood acute lymphoblastic leukemia. JAMA 2009; 301: 393–403.
childhood leukaemia. Nat Rev Cancer 2003; 3: 639–49. 36 Harrison CJ. Cytogenetics of paediatric and adolescent acute
9 Greaves MF, Maia AT, Wiemels JL, Ford AM. Leukemia in twins: lymphoblastic leukaemia. Br J Haematol 2009; 144: 147–56.
lessons in natural history. Blood 2003; 102: 2321–33. 37 Mullighan CG, Goorha S, Radtke I, et al. Genome-wide analysis of
10 Greaves M. Infection, immune responses and the aetiology of genetic alterations in acute lymphoblastic leukaemia. Nature 2007;
childhood leukaemia. Nat Rev Cancer 2006; 6: 193–203. 446: 758–64.
11 Greaves M. Cancer. The evolutionary legacy. Oxford: Oxford 38 Mullighan CG, Miller CB, Radtke I, et al. BCR-ABL1 lymphoblastic
University Press, 2000. leukaemia is characterized by the deletion of Ikaros. Nature 2008;
12 Greaves MF. Aetiology of acute leukaemia. Lancet 1997; 349: 344–49. 453: 110–14.
13 Preston DL, Kusumi S, Tomonaga M, et al. Cancer incidence in 39 Mullighan CG, Zhang J, Harvey RC, et al. JAK mutations in
atomic bomb survivors. Part III: leukemia, lymphoma and high-risk childhood acute lymphoblastic leukemia.
multiple myeloma, 1950–1987. Radiat Res 1994; Proc Natl Acad Sci USA 2009; 106: 9414–18.
137 (suppl): S68–S97. 40 Russell LJ, Capasso M, Vater I, et al. Deregulated expression of
14 Doll R, Wakeford R. Risk of childhood cancer from fetal irradiation. cytokine receptor gene, CRLF2, is involved in lymphoid
Br J Radiol 1997; 70: 130–39. transformation in B-cell precursor acute lymphoblastic leukemia.
Blood 2009; 114: 2688–98.
15 Kendall G, Little MP, Wakeford R. Numbers and proportions of
leukemias in young people and adults induced by radiation of 41 Mullighan CG, Collins-Underwood JR, Phillips LA, et al.
natural origin. Leuk Res 2011; 35: 1039–43. Rearrangement of CRLF2 in B-progenitor- and Down
syndrome-associated acute lymphoblastic leukemia. Nat Genet
16 Schüz J. Exposure to extremely low-frequency magnetic fields and
2009; 41: 1243–46.
the risk of childhood cancer: update of the epidemiological
evidence. Progr Biophysics Mol Biol 2011; 107: 339–42. 42 Mullighan CG, Zhang J, Kasper LH, et al. CREBBP mutations in
relapsed acute lymphoblastic leukaemia. Nature 2011; 471: 235–39.
17 Ward G. The infective theory of acute leukaemia. Br J Child Dis
1917; 14: 10–20. 43 Kuiper RP, Schoenmakers EF, van Reijmersdal SV, et al.
High-resolution genomic profiling of childhood ALL reveals novel
18 Kinlen L. Evidence for an infective cause of childhood leukaemia:
recurrent genetic lesions affecting pathways involved in lymphocyte
comparison of a Scottish New Town with nuclear reprocessing sites
differentiation and cell cycle progression. Leukemia 2007; 21: 1258–66.
in Britain. Lancet 1988; 332: 1323–27.
44 Kawamata N, Ogawa S, Zimmermann M, et al. Molecular
19 Kinlen LJ. Epidemiological evidence for an infective basis in
allelokaryotyping of pediatric acute lymphoblastic leukemias by
childhood leukaemia. Br J Cancer 1995; 71: 1–5.
high-resolution single nucleotide polymorphism oligonucleotide
20 Anderson RM, May RM. Immunisation and herd immunity. Lancet genomic microarray. Blood 2008; 111: 776–84.
1990; 335: 641-45.
45 Strefford JC, Worley H, Barber K, et al. Genome complexity in acute
21 Goldman JM, Jarrett O. Mechanisms of viral leukaemogenesis. lymphoblastic leukemia is revealed by array-based comparative
Edinburgh: Churchill Livingstone, 1984. genomic hybridization. Oncogene 2007; 26: 4306–18.
22 Greaves MF. Speculations on the cause of childhood acute 46 Van Vlierberghe P, Homminga I, Zuurbier L, et al. Cooperative
lymphoblastic leukemia. Leukemia 1988; 2: 120–25. genetic defects in TLX3 rearranged pediatric T-ALL. Leukemia 2008;
23 Gilham C, Peto J, Simpson J, et al. Day care in infancy and risk of 22: 762–70.
childhood acute lymphoblastic leukaemia: findings from a UK 47 Van Vlierberghe P, Meijerink JP, Lee C, et al. A new recurrent 9q34
case-control study. BMJ 2005; 330: 1294–97. duplication in pediatric T-cell acute lymphoblastic leukemia.
24 Urayama KY, Buffler PA, Gallagher ER, Ayoob JM, Ma X. Leukemia 2006; 20: 1245–53.
A meta-analysis of the association between day-care attendance and 48 Miller C, Mullighan CG, Su X, et al. Pax5 haploinsufficiency
childhood acute lymphoblastic leukaemia. Int J Epidemiol 2010; cooperates with BCR-ABL1 to induce acute lymphoblastic leukemia.
39: 718–32. Blood 2008; 112: 293s.
25 Kinlen L, Doll R. Population mixing and childhood leukaemia: 49 Virely C, Moulin S, Cobaleda C, et al. Haploinsufficiency of the
Fallon and other US clusters. Br J Cancer 2004; 91: 1–3. IKZF1 (IKAROS) tumor suppressor gene cooperates with BCR-ABL
26 Kroll ME, Draper GJ, Stiller CA, Murphy MFG. Childhood in a transgenic model of acute lymphoblastic leukemia. Leukemia
leukemia incidence in Britain, 1974–2000: time trends and possible 2010; 24: 1200–04.
relation to influenza epidemics. J Natl Cancer Inst 2006; 98: 417–20. 50 Iacobucci I, Storlazzi CT, Cilloni D, et al. Identification and
27 Ford AM, Palmi C, Bueno C, et al. The TEL-AML1 leukemia fusion molecular characterization of recurrent genomic deletions on 7p12
gene dysregulates the TGFb pathway in early B lineage progenitor in the IKZF1 gene in a large cohort of BCR-ABL1-positive acute
cells. J Clin Invest 2009; 119: 826–36. lymphoblastic leukemia patients: on behalf of Gruppo Italiano
28 Taylor GM, Birch JM. The hereditary basis of human leukemia. Malattie Ematologiche dell’Adulto Acute Leukemia Working Party
In: Henderson ES, Lister TA, Greaves MF, eds. Leukemia. 6th edn. (GIMEMA AL WP). Blood 2009; 114: 2159–67.
Philadelphia: WB Saunders, 1996: 210–45. 51 Mullighan CG, Su X, Zhang J, et al. Deletion of IKZF1 and prognosis
29 Hasle H, Clemmensen IH, Mikkelsen M. Risks of leukaemia and in acute lymphoblastic leukemia. N Engl J Med 2009; 360: 470–80.
solid tumours in individuals with Down’s syndrome. Lancet 2000; 52 Den Boer ML, van Slegtenhorst M, De Menezes RX, et al. A subtype
355: 165–69. of childhood acute lymphoblastic leukaemia with poor treatment
30 Vijayakrishnan J, Houlston RS. Candidate gene association studies outcome: a genome-wide classification study. Lancet Oncol 2009;
and risk of childhood acute lymphoblastic leukemia: a systematic 10: 125–34.
review and meta-analysis. Haematologica 2010; 95: 1405–14. 53 Yoda A, Yoda Y, Chiaretti S, et al. Functional screening identifies
31 Papaemmanuil E, Hosking FJ, Vijayakrishnan J, et al. Loci on CRLF2 in precursor B-cell acute lymphoblastic leukemia.
7p12.2, 10q21.2 and 14q11.2 are associated with risk of childhood Proc Natl Acad Sci USA 2010; 107: 252–57.
acute lymphoblastic leukemia. Nat Genet 2009; 41: 1006–10. 54 Hertzberg L, Vendramini E, Ganmore I, et al. Down syndrome acute
32 Trevino LR, Yang W, French D, et al. Germline genomic variants lymphoblastic leukemia: a highly heterogeneous disease in which
associated with childhood acute lymphoblastic leukemia. Nat Genet aberrant expression of CRLF2 is associated with mutated JAK2:
2009; 41: 1001–05. a report from the iBFM Study Group. Blood 2010; 115: 1006–17.

1952 www.thelancet.com Vol 381 June 1, 2013


Seminar

55 Harvey RC, Mullighan CG, Chen IM, et al. Rearrangement of CRLF2 77 Boissel N, Auclerc MF, Lheritier V, et al. Should adolescents with
is associated with mutation of JAK kinases, alteration of IKZF1, acute lymphoblastic leukemia be treated as old children or young
Hispanic/Latino ethnicity, and a poor outcome in pediatric adults? Comparison of the French FRALLE-93 and LALA-94 trials.
B-progenitor acute lymphoblastic leukemia. Blood 2010; 115: 5312–21. J Clin Oncol 2003; 21: 774–80.
56 Levine RL, Gilliland DG. Myeloproliferative disorders. Blood 2008; 78 De Bont JM, Holt B, Dekker AW, van der Does-van den Berg A,
112: 2190–98. Sonneveld P, Pieters R. Significant difference in outcome for
57 Buitenkamp TD, Pieters R, Gallimore NE, et al. Outcome in adolescents with acute lymphoblastic leukemia treated on pediatric
children with Down’s syndrome and acute lymphoblastic leukemia: vs adult protocols in the Netherlands. Leukemia 2004; 18: 2032–35.
role of IKZF1 deletions and CRLF2 aberrations. Leukemia 2012; 79 Stock W, La M, Sanford B, et al. What determines the outcomes for
26: 2204–11. adolescents and young adults with acute lymphoblastic leukemia
58 Roberts KG, Morin RD, Zhang J, et al. Genetic alterations activating treated on cooperative group protocols? A comparison of Children’s
kinase and cytokine receptor signaling in high-risk acute Cancer Group and Cancer and Leukemia Group B studies. Blood
lymphoblastic leukemia. Cancer Cell 2012; 22: 153–66. 2008; 112: 1646–54.
59 Moorman AV, Ensor HM, Richards SM, et al. Prognostic effect of 80 Pui CH, Pei D, Campana D, et al. Improved prognosis for older
chromosomal abnormalities in childhood B-cell precursor acute adolescents with acute lymphoblastic leukemia. J Clin Oncol 2011;
lymphoblastic leukaemia: results from the UK Medical Research 29: 386–91.
Council ALL97/99 randomised trial. Lancet Oncol 2010; 11: 429–38. 81 Campana D. Minimal residual disease monitoring in childhood
60 Harewood L, Robinson H, Harris R, et al. Amplification of AML1 acute lymphoblastic leukemia. Curr Opin Hematol 2012; 19: 313–18.
on a duplicated chromosome 21 in acute lymphoblastic leukemia: 82 Borowitz MJ, Devidas M, Hunger SP, et al. Clinical significance of
a study of 20 cases. Leukemia 2003; 17: 547–53. minimal residual disease in childhood acute lymphoblastic
61 Robinson HM, Harrison CJ, Moorman AV, Chudoba I, Strefford JC. leukemia and its relationship to other prognostic factors:
Intrachromosomal amplification of chromosome 21 (iAMP21) may a Children’s Oncology Group study. Blood 2008; 111: 5477–85.
arise from a breakage–fusion–bridge cycle. 83 Conter V, Bartram CR, Valsecchi MG, et al. Molecular response to
Genes Chromosomes Cancer 2007; 46: 318–26. treatment redefines all prognostic factors in children and
62 Attarbaschi A, Mann G, Panzer-Grumayer R, et al. Minimal adolescents with B-cell precursor acute lymphoblastic leukemia:
residual disease values discriminate between low and high relapse results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood
risk in children with B-cell precursor acute lymphoblastic 2010; 115: 3206–14.
leukemia and an intrachromosomal amplification of chromosome 84 Schrappe M, Valsecchi MG, Bartram CR, et al. Late MRD response
21: the Austrian and German acute lymphoblastic leukemia determines relapse risk overall and in subsets of childhood T-cell ALL:
Berlin-Frankfurt-Munster (ALL-BFM) trials. J Clin Oncol 2008; results of the AIEOP-BFM-ALL 2000 study. Blood 2011; 118: 2077–84.
26: 3046–50. 85 Basso G, Veltroni M, Valsecchi MG, et al. Risk of relapse of
63 Raimondi SC, Pui CH, Head DR, Rivera GK, Behm FG. childhood acute lymphoblastic leukemia is predicted by flow
Cytogenetically different leukemic clones at relapse of childhood cytometric measurement of residual disease on day 15 bone
acute lymphoblastic leukemia. Blood 1993; 82: 576–80. marrow. J Clin Oncol 2009; 27: 5168–74.
64 Mullighan CG, Phillips LA, Su X, et al. Genomic analysis of the 86 Gokbuget N, Kneba M, Raff T, et al. Adult patients with acute
clonal origins of relapsed acute lymphoblastic leukemia. Science lymphoblastic leukemia and molecular failure display a poor
2008; 322: 1377–80. prognosis and are candidates for stem cell transplantation and
65 Yang JJ, Bhojwani D, Yang W, et al. Genome-wide copy number targeted therapies. Blood 2012; 120: 1868–76.
profiling reveals molecular evolution from diagnosis to relapse in 87 Stow P, Key L, Chen X, et al. Clinical significance of low levels of
childhood acute lymphoblastic leukemia. Blood 2008; 112: 4178–83. minimal residual disease at the end of remission induction therapy
66 Kawamata N, Ogawa S, Seeger K, et al. Molecular allelokaryotyping in childhood acute lymphoblastic leukemia. Blood 2010; 115: 4657–63.
of relapsed pediatric acute lymphoblastic leukemia. Int J Oncol 88 Coustan-Smith E, Song G, Clark C, et al. New markers for minimal
2009; 34: 1603–12. residual disease detection in acute lymphoblastic leukemia. Blood
67 Hof J, Krentz S, van Schewick C, et al. Mutations and deletions of 2011; 117: 6267–76.
the TP53 gene predict nonresponse to treatment and poor outcome 89 Inaba H, Pui CH. Glucocorticoid use in acute lymphoblastic
in first relapse of childhood acute lymphoblastic leukemia. leukaemia. Lancet Oncol 2010; 11: 1096–106.
J Clin Oncol 2011; 29: 3185–93. 90 Bostrom BC, Sensel MR, Sather HN, et al. Dexamethasone versus
68 Inthal A, Zeitlhofer P, Zeginigg M, et al. CREBBP HAT domain prednisone and daily oral versus weekly intravenous mercaptopurine
mutations prevail in relapse cases of high hyperdiploid childhood for patients with standard-risk acute lymphoblastic leukemia: a report
acute lymphoblastic leukemia. Leukemia 2012; 26: 1797–803. from the Children’s Cancer Group. Blood 2003; 101: 3809–17.
69 Zhang J, Mullighan CG, Harvey RC, et al. Key pathways are 91 Mitchell CD, Richards SM, Kinsey SE, Lilleyman J, Vora A,
frequently mutated in high-risk childhood acute lymphoblastic Eden TO. Benefit of dexamethasone compared with prednisolone
leukemia: a report from the Children’s Oncology Group. Blood 2011; for childhood acute lymphoblastic leukaemia: results of the UK
118: 3080–87. Medical Research Council ALL97 randomized trial. Br J Haematol
70 Rothenberg EV, Moore JE, Yui MA. Launching the T-cell-lineage 2005; 129: 734–45.
developmental programme. Nat Rev Immunol 2008; 8: 9–21. 92 Schrappe M, Zimmermann M, Moricke A, et al. Dexamethasone in
71 Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell induction can eliminate one third of all relapses in childhood acute
precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukemia (ALL): results of an international
lymphoblastic leukaemia. Lancet Oncol 2009; 10: 147–56. randomized trial in 3655 patients (trial AIEOP-BFM ALL 2000).
72 Zhang J, Ding L, Holmfeldt L, et al. The genetic basis of early Blood 2008; 112: 7s.
T-cell precursor acute lymphoblastic leukaemia. Nature 2012; 93 Winick N, Salzer W, Devidas M, et al. Dexamethasone (DEX)
481: 157–63. versus prednisone (PRED) during induction for children with
73 Van Vlierberghe P, Palomero T, Khiabanian H, et al. PHF6 mutations high-risk acute lymphoblastic leukemia (HR-ALL): a report from
in T-cell acute lymphoblastic leukemia. Nat Genet 2010; 42: 338–42. the Children’s Oncology Group study AALL0232. J Clin Oncol
2011; 29: 586s.
74 Yang JJ, Cheng C, Devidas M, et al. Ancestry and
pharmacogenomics of relapse in acute lymphoblastic leukemia. 94 Igarashi S, Manabe A, Ohara A, et al. No advantage of
Nat Genet 2011; 43: 237–41. dexamethasone over prednisolone for the outcome of standard- and
intermediate-risk childhood acute lymphoblastic leukemia in the
75 Xu H, Cheng C, Devidas M, et al. ARID5B genetic polymorphisms
Tokyo Children’s Cancer Study Group L95-14 protocol. J Clin Oncol
contribute to racial disparities in the incidence and treatment
2005; 23: 6489–98.
outcome of childhood acute lymphoblastic leukemia. J Clin Oncol
2012; 30: 751–57. 95 Bertrand Y, Suciu S, Benoit Y, et al. Dexamethasone(DEX)
(6mg/sm/d) and prednisolone(PRED)(60mg/sm/d) in induction
76 Schafer ES, Hunger SP. Optimal therapy for acute lymphoblastic
therapy of childhood ALL are equally effective: results of the
leukemia in adolescents and young adults. Nat Rev Clin Oncol 2011;
2nd interim analysis of EORTC Trial 58951. Blood 2008; 112: 8s.
8: 417–24.

www.thelancet.com Vol 381 June 1, 2013 1953


Seminar

96 Pui CH, Mullighan CG, Evans WE, Relling MV. Pediatric acute 115 Seibel NL, Steinherz PG, Sather HN, et al. Early postinduction
lymphoblastic leukemia: where are we going and how do we get intensification therapy improves survival for children and
there? Blood 2012; 120: 1165–74. adolescents with high-risk acute lymphoblastic leukemia: a report
97 Pieters R, Hunger SP, Boos J, et al. L-asparaginase treatment in from the Children’s Oncology Group. Blood 2008; 111: 2548–55.
acute lymphoblastic leukemia: a focus on Erwinia asparaginase. 116 Matloub Y, Bostrom BC, Hunger SP, et al. Escalating intravenous
Cancer 2011; 117: 238–49. methotrexate improves event-free survival in children with
98 Silverman LB, Supko JG, Stevenson KE, et al. Intravenous standard-risk acute lymphoblastic leukemia: a report from the
PEG-asparaginase during remission induction in children and Children’s Oncology Group. Blood 2011; 118: 243–51.
adolescents with newly diagnosed acute lymphoblastic leukemia. 117 Mattano LA, Devidas M, Nachman JB, et al. Effect of alternate-week
Blood 2010; 115: 1351–53. versus continuous dexamethasone scheduling on the risk of
99 Silverman LB, Gelber RD, Dalton VK, et al. Improved outcome for osteonecrosis in paediatric patients with acute lymphoblastic
children with acute lymphoblastic leukemia: results of Dana-Farber leukaemia: results from the CCG-1961 randomised cohort trial.
Consortium protocol 91-01. Blood 2001; 97: 1211–18. Lancet Oncol 2012; 13: 906–15.
100 Willer A, Gerss J, Konig T, et al. Anti-Escherichia coli asparaginase 118 Harms DO, Gobel U, Spaar HJ, et al. Thioguanine offers no
antibody levels determine the activity of second-line treatment with advantage over mercaptopurine in maintenance treatment of
pegylated E coli asparaginase: a retrospective analysis within the childhood ALL: results of the randomized trial COALL-92. Blood
ALL-BFM trials. Blood 2011; 118: 5774–82. 2003; 102: 2736–40.
101 Yang L, Panetta JC, Cai X, et al. Asparaginase may influence 119 Vora A, Mitchell CD, Lennard L, et al. Toxicity and efficacy of
dexamethasone pharmacokinetics in acute lymphoblastic leukemia. 6-thioguanine versus 6-mercaptopurine in childhood lymphoblastic
J Clin Oncol 2008; 26: 1932–39. leukaemia: a randomised trial. Lancet 2006; 368: 1339–48.
102 Kawedia JD, Liu C, Pei D, et al. Dexamethasone exposure and 120 Stork LC, Matloub Y, Broxson E, et al. Oral 6-mercaptopurine
asparaginase antibodies affect relapse risk in acute lymphoblastic versus oral 6-thioguanine and veno-occlusive disease in children
leukemia. Blood 2012; 119: 1658–64. with standard-risk acute lymphoblastic leukemia: report of the
103 Schultz KR, Bowman WP, Aledo A, et al. Improved early event-free Children’s Oncology Group CCG-1952 clinical trial. Blood 2010;
survival with imatinib in Philadelphia chromosome-positive acute 115: 2740–48.
lymphoblastic leukemia: a Children’s Oncology Group study. 121 Relling MV, Gardner EE, Sandborn WJ, et al. Clinical
J Clin Oncol 2009; 27: 5175–81. Pharmacogenetics Implementation Consortium guidelines for
104 Ravandi F, O’Brien S, Thomas D, et al. First report of phase 2 study thiopurine methyltransferase genotype and thiopurine dosing.
of dasatinib with hyper-CVAD for the frontline treatment of patients Clin Pharmacol Ther 2011; 89: 387–91.
with Philadelphia chromosome-positive (Ph+) acute lymphoblastic 122 Schmiegelow K, Al-Modhwahi I, Andersen MK, et al.
leukemia. Blood 2010; 116: 2070–77. Methotrexate/6-mercaptopurine maintenance therapy influences
105 Ottmann O, Dombret H, Martinelli G, et al. Dasatinib induces the risk of a second malignant neoplasm after childhood acute
rapid hematologic and cytogenetic responses in adult patients with lymphoblastic leukemia: results from the NOPHO ALL-92 study.
Philadelphia chromosome positive acute lymphoblastic leukemia Blood 2009; 113: 6077–84.
with resistance or intolerance to imatinib: interim results of a 123 Bhatia S, Landier W, Shangguan M, et al. Nonadherence to oral
phase 2 study. Blood 2007; 110: 2309–15. mercaptopurine and risk of relapse in Hispanic and non-Hispanic
106 Porkka K, Koskenvesa P, Lundan T, et al. Dasatinib crosses the white children with acute lymphoblastic leukemia: a report from
blood-brain barrier and is an efficient therapy for central nervous the Children’s Oncology Group. J Clin Oncol 2012; 30: 2094–101.
system Philadelphia chromosome-positive leukemia. Blood 2008; 124 Diouf B, Cheng Q, Krynetskaia NF, et al. Somatic deletions of genes
112: 1005–12. regulating MSH2 protein stability cause DNA mismatch repair
107 Whitehead VM, Vuchich MJ, Lauer SJ, et al. Accumulation of high deficiency and drug resistance in human leukemia cells. Nat Med
levels of methotrexate polyglutamates in lymphoblasts from 2011; 17: 1298–303.
children with hyperdiploid (greater than 50 chromosomes) 125 Balduzzi A, Valsecchi MG, Uderzo C, et al. Chemotherapy versus
B-lineage acute lymphoblastic leukemia: a Pediatric Oncology allogeneic transplantation for very-high-risk childhood acute
Group study. Blood 1992; 80: 1316–23. lymphoblastic leukaemia in first complete remission: comparison
108 Rots MG, Pieters R, Peters GJ, et al. Role of folylpolyglutamate by genetic randomisation in an international prospective study.
synthetase and folylpolyglutamate hydrolase in methotrexate Lancet 2005; 366: 635–42.
accumulation and polyglutamylation in childhood leukemia. Blood 126 Marks DI, Wang T, Perez WS, et al. The outcome of full-intensity
1999; 93: 1677–83. and reduced-intensity conditioning matched sibling or unrelated
109 French D, Yang W, Cheng C, et al. Acquired variation outweighs donor transplantation in adults with Philadelphia
inherited variation in whole genome analysis of methotrexate chromosome-negative acute lymphoblastic leukemia in first and
polyglutamate accumulation in leukemia. Blood 2009; second complete remission. Blood 2010; 116: 366–74.
113: 4512–20. 127 Leung W, Campana D, Yang J, et al. High success rate of
110 Trevino LR, Shimasaki N, Yang W, et al. Germline genetic variation hematopoietic cell transplantation regardless of donor source in
in an organic anion transporter polypeptide associated with children with very high-risk leukemia. Blood 2011; 118: 223–30.
methotrexate pharmacokinetics and clinical effects. J Clin Oncol 128 Eapen M, Rubinstein P, Zhang M-J, et al. Outcomes of
2009; 27: 5972–8. transplantation of unrelated donor umbilical cord blood and bone
111 Ramsey LB, Bruun GH, Yang W, et al. Rare versus common marrow in children with acute leukaemia: a comparison study.
variants in pharmacogenetics: SLCO1B1 variation and methotrexate Lancet 2007; 369: 1947–54.
disposition. Genome Res 2012; 22: 1–8. 129 Eapen M, Rocha V, Sanz G, et al. Effect of graft source on unrelated
112 Larsen E, Salzer WL, Devidas M, et al. Comparison of high-dose donor haemopoietic stem-cell transplantation in adults with acute
methotrexate (HD-MTX) with Capizzi methotrexate plus leukaemia: a retrospective analysis. Lancet Oncol 2010; 11: 653–60.
asparaginase (C-MTX/ASNase) in children and young adults with 130 Bader P, Kreyenberg H, Henze GH, et al. Prognostic value of
high-risk acute lymphoblastic leukemia (HR-ALL): a report from minimal residual disease quantification before allogeneic stem-cell
the Children’s Oncology Group study AALL0232. J Clin Oncol transplantation in relapsed childhood acute lymphoblastic
2011; 29: 3s. leukemia: the ALL-REZ BFM study group. J Clin Oncol 2009;
113 Mikkelsen TS, Sparreboom A, Cheng C, et al. Shortening infusion 27: 377–84.
time for high-dose methotrexate alters antileukemic effects: 131 Leung W, Pui CH, Coustan-Smith E, et al. Detectable minimal
a randomized prospective clinical trial. J Clin Oncol 2011; residual disease before hematopoietic cell transplantation is
29: 1771–78. prognostic but does not preclude cure for children with
114 Skarby TV, Anderson H, Heldrup J, Kanerva JA, Seidel H, very-high-risk leukemia. Blood 2012; 120: 468–72.
Schmiegelow K. High leucovorin doses during high-dose 132 Schrappe M, Hunger SP, Pui CH, et al. Outcomes after induction
methotrexate treatment may reduce the cure rate in childhood acute failure in childhood acute lymphoblastic leukemia. N Engl J Med
lymphoblastic leukemia. Leukemia 2006; 20: 1955–62. 2012; 366: 1371–81.

1954 www.thelancet.com Vol 381 June 1, 2013


Seminar

133 Dreyer ZE, Dinndorf PA, Camitta B, et al. Analysis of the role of 138 Stumpel DJ, Schneider P, van Roon EH, et al. Specific promoter
hematopoietic stem-cell transplantation in infants with acute methylation identifies different subgroups of MLL-rearranged
lymphoblastic leukemia in first remission and MLL gene infant acute lymphoblastic leukemia, influences clinical outcome,
rearrangements: a report from the Children’s Oncology Group. and provides therapeutic options. Blood 2009; 114: 5490–98.
J Clin Oncol 2011; 29: 214–22. 139 Bernt KM, Zhu N, Sinha AU, et al. MLL-rearranged leukemia is
134 Mann G, Attarbaschi A, Schrappe M, et al. Improved outcome with dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell
hematopoietic stem cell transplantation in a poor prognostic 2011; 20: 66–78.
subgroup of infants with mixed-lineage-leukemia (MLL)-rearranged 140 Hoelzer D. Novel antibody-based therapies for acute lymphoblastic
acute lymphoblastic leukemia: results from the Interfant-99 study. leukemia. Hematology Am Soc Hematol Educ Program 2011;
Blood 2010; 116: 2644–50. 2011: 243–49.
135 Pui C-H, Howard SC. Current management and challenges of 141 Thomas DA, O’Brien S, Faderl S, et al. Chemoimmunotherapy with
malignant disease in the CNS in paediatric leukaemia. Lancet Oncol a modified hyper-CVAD and rituximab regimen improves outcome
2008; 9: 257–68. in de novo Philadelphia chromosome-negative precursor B-lineage
136 Veerman AJ, Kamps WA, van den Berg H, et al. acute lymphoblastic leukemia. J Clin Oncol 2010; 28: 3880–89.
Dexamethasone-based therapy for childhood acute lymphoblastic 142 Topp MS, Kufer P, Gokbuget N, et al. Targeted therapy with the
leukaemia: results of the prospective Dutch Childhood Oncology T-cell-engaging antibody blinatumomab of chemotherapy-refractory
Group (DCOG) protocol ALL-9 (1997–2004). Lancet Oncol 2009; minimal residual disease in B-lineage acute lymphoblastic
10: 957–66. leukemia patients results in high response rate and prolonged
137 Matloub Y, Lindemulder S, Gaynon PS, et al. Intrathecal triple leukemia-free survival. J Clin Oncol 2011; 29: 2493–98.
therapy decreases central nervous system relapse but fails to
improve event-free survival when compared with intrathecal
methotrexate: results of the Children’s Cancer Group (CCG)
1952 study for standard-risk acute lymphoblastic leukemia, reported
by the Children’s Oncology Group. Blood 2006; 108: 1165–73.

www.thelancet.com Vol 381 June 1, 2013 1955

You might also like