+ All Categories
Home > Documents > Acute Lymphoblastic Leukaemia Lancet 2008

Acute Lymphoblastic Leukaemia Lancet 2008

Date post: 14-Apr-2018
Category:
Upload: alexis-may-uc
View: 228 times
Download: 0 times
Share this document with a friend

of 14

Transcript
  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    1/14

    Seminar

    1030 www.thelancet.com Vol 371 March 22, 2008

    Acute lymphoblastic leukaemia

    Ching-Hon Pui, Leslie L Robison, A Thomas Look

    Acute lymphoblastic leukaemia, a malignant disorder of lymphoid progenitor cells, affects both children and adults,with peak prevalence between the ages of 2 and 5 years. Steady progress in development of effective treatments hasled to a cure rate of more than 80% in children, creating opportunities for innovative approaches that would preservepast gains in leukaemia-free survival while reducing the toxic side-effects of current intensive regimens. Advances inour understanding of the pathobiology of acute lymphoblastic leukaemia, fuelled by emerging molecular technologies,suggest that drugs specifically targeting the genetic defects of leukaemic cells could revolutionise management of thisdisease. Meanwhile, studies are underway to ascertain the precise events that take place in the genesis of acutelymphoblastic leukaemia, to enhance the clinical application of known risk factors and antileukaemic agents, and toidentify treatment regimens that might boost the generally low cure rates in adults and subgroups of children withhigh-risk leukaemia.

    IntroductionAddition of acute lymphoblastic leukaemia to the growinglist of cancers that have succumbed to effective treatmentis tempting. The decision would be easy to justify in viewof data showing cure rates higher than 80% for childrentreated in modern centres, most of whom will leadhealthy productive lives as long-term cancer survivors.13Thus, the future management of acute lymphoblasticleukaemia might be viewed as simply tweaking existingprotocols and devising alternative regimens for the fifthof patients who respond poorly to available agents. Thisscenario, however attractive, must be rejected on severalgrounds. It does not accommodate the poor prognosis

    for adults with acute lymphoblastic leukaemia or thecomplexity, expense, and toxic effects of contemporarymultiagent treatments.1,4Most importantly, it overlooksour rapidly increasing ability to analyse the genetic andepigenetic abnormalities of leukaemic cells and totranslate them into enhanced diagnostic methods andmolecularly targeted therapy.5,6 Although the molecularmedicine approach is still in its investigative stage, withmany new obstacles to overcome, it holds enormouspromise. Put simply, we are about to enter an era inwhich leukaemia patients will probably receiveindividualised treatment based on the genetic features oftheir malignant cells and their own unique geneticmake-up (so-called pharmacogenomics).7 Our intent in

    this Seminar is to review advances in both thefundamental understanding and clinical management ofacute lymphoblastic leukaemia in children and adults.

    Epidemiology and causeThe precise pathogenetic events leading to developmentof acute lymphoblastic leukaemia are unknown. Only afew cases (03 or 04 T) ofresidential, power-frequency magnetic fields.9,10

    Observations of a peak age of development of childhoodacute lymphoblastic leukaemia of 25 years, an associationof industrialisation and modern or affl uent societies withincreased prevalence of the disease, and the occasionalclustering of childhood leukaemia cases (especially in newtowns) have fuelled two parallel infection-based hypotheses

    by British investigators: Kinlens population-mixinghypothesis and Greaves delayed-infection hypothesis(figure 1).11,12 Kinlens hypothesis predicts that clusters ofchildhood cases of acute lymphoblastic leukaemia resultfrom exposure of susceptible (non-immune) individualsto common but fairly non-pathological infections afterpopulation-mixing with carriers. The delayed-infectionhypothesis of Greaves is based on a minimal two-hitsmodel and suggests that some susceptible individualswith a prenatally acquired preleukaemic clone had low orno exposure to common infections early in life becausethey lived in an affl uent hygienic environment. Suchinfectious insulation predisposes the immune system ofthese individuals to aberrant or pathological responses

    after subsequent or delayed exposure to commoninfections at an age commensurate with increasedlymphoid-cell proliferation.

    Retrospective identification of leukaemia-specificfusion genes, hyperdiploidy, or clonotypic rearrange-ments of immunoglobulin or T-cell-receptor loci in

    Lancet2008; 371: 103043

    Department of Oncology

    (Prof C-H Pui MD)and

    Department of Epidemiology

    and Cancer Control

    (Prof L L Robison PhD), St Jude

    Childrens Research Hospital

    and University of Tennessee

    Health Science Center,

    Memphis, TN, USA; and

    Department of Pediatric

    Oncology, Dana-Farber Cancer

    Institute and Harvard Medical

    School, Boston, MA, USA(Prof A T Look MD)

    Correspondence to:

    Prof Ching-Hon Pui, St Jude

    Childrens Research Hospital,

    332 N Lauderdale, Memphis,

    TN 38105, USA

    [email protected]

    Search strategy and selection criteria

    We searched Medline and PubMed for articles published in

    English dating from 2002, with the keywords acute

    lymphoblastic leukemia, acute lymphocytic leukemia, and

    acute lymphoid leukemia. In some instances, review articles

    were selected over original articles because of space constraints.

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    2/14

    Seminar

    www.thelancet.com Vol 371 March 22, 2008 1031

    archived neonatal blood spots (Guthrie cards) and

    studies of leukaemia in monozygotic twins indicateclearly a prenatal origin for some childhoodleukaemias.1215Screening of neonatal cord-blood sampleshas revealed a putative leukaemic clone with theTEL-AML1fusion gene (also known as ETV6-RUNX1)in1% of newborn babies, a frequency 100 times higherthan the prevalence of acute lymphoblastic leukaemiadefined by this fusion gene later in childhood.16 Thevariable incubation period and clinical outcome of suchcases, and the 10% concordance rate of leukaemia inidentical twins with this genotype, support the notionthat additional postnatal events are needed for fullleukaemic transformation.17 A recent study furtherestablished the presence of a preleukaemic clone with

    the TEL-AML1fusion.15Investigations have also focused on the genetic

    variability in xenobiotic metabolism, DNA repairpathways, and cell-cycle checkpoint functions that mightinteract with environmental, dietary, maternal, and otherexternal factors to affect development of acutelymphoblastic leukaemia. Although the number ofinvestigations and sample sizes are limited, data exist tosupport a possible causal role for polymorphisms ingenes encoding cytochrome P450, NAD(P)H quinoneoxidoreductase, glutathione S-transferases, methyl-enetetahydrofolate reductase, thymidylate synthase,serine hyroxymethyltransferase, and cell-cycle inhi-bitors.1823To date, however, no direct gene-environmentinteractions have been established convincingly.

    In view of the scarcity of causal insights from large-scaleepidemiological studies, some investigators have adopteda strategy that focuses on distinct subtypes of childhoodacute lymphoblastic leukaemia. An important example isthe study of infant acute lymphoblastic leukaemia withMLLrearrangement,24a genetic abnormality that has alsobeen associated with secondary leukaemia after exposureto a topoisomerase II inhibitor.25Thus, dietary, medical,and environmental exposures to substances that inhibittopoisomerases, and the reduced ability of fetuses ortheir mothers to detoxify such agents, could lead todevelopment of infant leukaemia.26,27

    PathobiologyAcute lymphoblastic leukaemia is thought to originatefrom various important genetic lesions in blood-progenitorcells that are committed to differentiate in the T-cell orB-cell pathway, including mutations that impart thecapacity for unlimited self-renewal and those that lead toprecise stage-specific developmental arrest.6,28 In somecases, the first mutation along the multistep pathway toovert acute lymphoblastic leukaemia might arise in ahaemopoietic stem cell possessing multilineage develop-mental capacity.29 The cells implicated in acutelymphoblastic leukaemia have clonal rearrangements intheir immunoglobulin or T-cell receptor genes andexpress antigen-receptor molecules and other

    differentiation-linked cell-surface glycoproteins thatlargely recapitulate those of immature lymphoidprogenitor cells within the early developmental stages ofnormal T and B lymphocytes.6,28,30The dominant themeof contemporary research in pathobiology of acutelymphoblastic leukaemia is to understand the outcomesof frequently arising genetic lesions, in terms of theireffects on cell proliferation, differentiation, and survival,and then to devise selectively targeted treatments againstthe altered gene products to which the leukaemic cloneshave become addicted.31

    Chromosomal translocationsChromosomal translocations that activate specific genesare a defining characteristic of human leukaemias andof acute lymphoblastic leukaemia in particular.6,28Gene-expression patterns studied in large series of newlydiagnosed leukaemias have substantiated the idea thatspecific chromosomal translocations identify uniquesubtypes of the disease.3235Usually, translocations activatetranscription-factor genes, which in many cases cancontrol cell differentiation (rather than cell division per

    se), are developmentally regulated, and frequently encodeproteins at the apex of important transcriptionalcascades.28These so-called master oncogenic transcriptionfactors, which can exert either positive or negative controlover downstream responder genes, are expressedaberrantly in leukaemic cells as one gene product or as aunique fusion protein combining elements from twodifferent transcription factors.6,28

    About 25% of cases of B-cell precursor acutelymphoblastic leukaemia, the most frequent form ofacute leukaemia in children, harbour the TEL-AML1fusion genegenerated by the t(12;21)(p13;q22)chromosomal translocation.6 Although the molecularpathogenesis of TEL-AML1-positive leukaemia remainsunclear, findings in mice establish the Tel gene as an

    Fetal development Birth Infancy early childhood

    Low exposure

    to pathogensresulting indecreased

    proliferativestress

    Rapid cellproliferation

    Exposure topathogens duringperiod of

    increased lymphoidproliferation

    Exposure to the new pathogens as a result ofincreased population mixing

    Delayed exposure to common pathogens

    Low or no

    exposure tonon-endemic

    pathogens

    Live in an environmentwith decreased

    exposure to commonpathogens

    Randommutations

    Greaves

    hypothesis

    Kinlens

    hypoth

    esis

    Figure :Infection-based models of leukaemia development

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    3/14

    Seminar

    1032 www.thelancet.com Vol 371 March 22, 2008

    important regulator of haemopoietic-cell development,

    essential for definitive haemopoiesis.36 Similarly, Aml1gene is essential for definitive embryonic haemo-poiesis.37,38Thus, the presence of the TEL-AML1 fusionprotein in B-cell progenitors seems to lead to disorderedearly B-lineage lymphocyte development, a hallmark ofleukaemic lymphoblasts. Analysis of TEL-AML1-inducedcord blood cells suggests that the fusion gene serves as afirst-hit mutation by endowing the preleukemic cell withaltered self-renewal and survival properties.15

    In adults, the most frequent chromosomal translocationis t(9;22), or the Philadelphia chromosome, which causesfusion of the BCR signalling protein to the ABLnon-receptor tyrosine kinase, resulting in constitutivetyrosine kinase activity and complex interactions of thisfusion protein with many other transforming elements,

    such as the signalling pathway for RAS (GTP-binding

    protein that activates target genes involved in celldifferentiation, proliferation, and survival).39 As anactivated kinase, BCR-ABL offers an attractive therapeutictarget, and imatinib mesilate, a small-molecule inhibitorof the ABL kinase, has proven effective against leukaemiasthat express BCR-ABL.40

    More than 50% of cases of T-cell acute lymphoblasticleukaemia have activating mutations that involveNOTCH1,41a gene encoding a transmembrane receptorthat regulates normal T-cell development.42 NOTCHreceptors become activated when ligands of theDelta-Serrate-Lag2 family of proteins bind to theextracellular portion of the transmembrane molecule.This interaction initiates a cascade of proteolytic cleavages,terminating in -secretase generation of intracellular

    pre-Notch

    NotchICN

    Heterodimerisation

    Glycosylation

    Fucosylation andendoplasmic reticulum exit

    Ubiquitination and

    proteosome degradation

    -secretasecomplex

    S3cleavage

    S2cleavage

    S1Cleavage

    ICN

    GSI ICN

    ICN

    ICN

    ICN

    MINTdnMAML NRARP

    Notch

    ADAMmetalloprotease

    pre-Notch

    OFUT 1

    Fringe

    Fringe

    Numb

    Furin

    DLL

    Jagged

    SEL 10

    CSL

    CSL

    CoA

    MAML

    n300

    CoR

    Deltex?

    NEURL

    MIB

    HES 1PRE TDELTEX

    NRARPCD25MYC

    Receiving cell

    Nucleus

    Golgi

    Endoplasmic

    reticulum

    Ligand

    endocytosis

    and

    degradation

    Sending cell

    A

    B

    C

    E

    F

    G

    Figure :NOTCH signalling in normal thymocytes

    The NOTCH signalling pathway is complex and involves the coordinated activities of many different molecules. Briefly, NOTCH is synthesised in the endoplasmic

    reticulum (A) as one protein consisting of an extracellular domain (pre-Notch) and an intracellular domain (ICN), which are transported in tandem to the Golgi (B),

    where several post-translational modifications take pl ace, including a proteolytic cleavage (S1) that separates the two domains from ea ch other. The resultant

    heterodimer is then transported to the cell membrane, where NOTCH interacts with ligands (C) and is cleaved twice (D) by the ADAM protease (S2) and a -secretase

    complex (S3), enabling the liberated ICN domain to translocate to the nucleus (E). Nuclear ICN forms a binding/activator complex with a group of cooperating

    proteins (F), resulting in transcriptional activation of several functionally important genes, including MYC and pre-T. Hyperphosphorylation of ICN via interaction of

    its PEST domain (polypeptide enriched in proline, glutamate, serine, and threonine) with CDK8, MAML, and p300 facilitates ubiquitylation (G) by SEL1 family

    members, targeting ICN to the proteosome. ADAM=a disintegrin and metalloproteinase domain. DLL=Delta ligand . hes1=hairy/enhancer of split. GSI=-secretase

    inhibition.MAML=mastermind-like proteins. MIB=mindbomb. NEURL=neuralised-like. Nrarp=gene encoding NOTCH-regulated ankyrin-repeat protein.

    OFUT1=O-fucosyltransferase1. Pre-T=pre-TCR. Deltex=positive regulator of Notch signalling pathway. CS1=DNA binding component. CoA=co-activators.

    CoR=co-repressors. Fringe=regulator of Notch ligand. SEL10=positive regulator of Notch. Numb=negative receptor of Notch.

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    4/14

    Seminar

    www.thelancet.com Vol 371 March 22, 2008 1033

    NOTCH1, which translocates to the nucleus and regulates

    by transcription a diverse set of responder genes,including the MYC oncogene (figure 2).43,44 The precisemechanisms by which aberrant NOTCH signalling (dueto mutational activation) causes T-cell acute lymphoblasticleukaemia are still unclear but probably entail constitutiveexpression of oncogenic responder genes, such as MYC,and cooperation with other signalling pathways (pre-TCR[T-cell receptor for antigen] and RAS, for example).Interference with NOTCH signalling by small-moleculeinhibition of -secretase activity has the potential toinduce remission of T-cell acute lymphoblasticleukaemia.

    Evidence suggests that the MYC oncoprotein is animportant downstream mediator of the pro-growth

    effects of NOTCH1 signalling in developing thym-ocytes.28,45 However, results of retroviral insertionalmutagenesis in murine models of transgenic T-cell acutelymphoblastic leukaemia show that Notch1 mutations,with outcomes similar to those in primary human T-cellacute lymphoblastic leukaemias, can potentiate theeffects of pre-existing MYCoverexpression,42,46suggestingthat NOTCH1 must have important transformationaltargets other than MYC. Activating mutations inNOTCH1 suffi cient to produce constitutive NOTCH1signalling can induce T-cell acute lymphoblasticleukaemia in experimental models and could be theinstigating event in most human T-cell leukaemias.28,42-secretase, a multicomponent membrane-associatedenzyme, is needed for NOTCH1 signalling throughmutant NOTCH receptors in T-cell acute lymphoblasticleukaemia, providing an attractive target for therapeuticintervention with newly developed -secretaseinhibitors.28,47

    Cooperating mutationsAlthough chromosomal abnormalities are a hallmark ofpathogenesis of acute lymphoblastic leukaemia, evidencesuggests that they must act in concert with several othergenetic lesions to induce overt leukaemia. A primeexample is the biallelic deletion or epigenetic silencing ofthe cyclin-dependent kinase inhibitor 2A gene (CDKN2A),

    which encodes both the tumour suppressors p16 INK4Aandp14ARFand whose inactivation neutralises both the TP53and retinoblastoma pathways in most cases of T-cell andmany cases of B-cell precursor acute lymphoblasticleukaemia.6 In a genome-wide analysis of 242 cases ofpaediatric acute lymphoblastic leukaemia usinghigh-resolution single nucleotide polymorphism arrays,deletions, amplifications, point mutations, and otherstructural rearrangements were identified in genesencoding regulators of B-lymphocyte development in40% of cases of B-cell precursor acute lymphoblasticleukaemia.48The PAX5 gene was the most frequent targetof somatic mutation, being altered in almost a third ofcases. Deletions were also detected in other B-celldevelopmental genes, such as TCF3(E2A), EBF1(EBF),

    LEF1, IKZF1 (Ikaros), and IKZF3 (Aiolos). Finally, in

    T-cell acute lymphoblastic leukaemia, at least fivemultistep mutational pathways leading to frank leukaemiahave been identified, and in some cases these pathwaysentail five or more documented genetic lesions.28,30,42

    Ongoing research to define the oncogenic contributionsof various classes of genetic lesions relies heavily onanimal models that accurately recapitulate the molecularpathogenesis of B-cell precursor or T-cell acutelymphoblastic leukaemia.49 Most studies undertaken todate have used genetically engineered mice to elucidatethe multistep transformation pathways leading to T-cellacute lymphoblastic leukaemia.50 Such models dependon breeding strategies to combine one or more geneticlesions and show synergy in transformation,51 whereas

    some investigators have also capitalised on the use ofretroviral insertional mutagenesis screens to uncovercollaborating oncogenes.46,52New models of T-cell acutelymphoblastic leukaemia in the zebrafish offer a powerfulalternative vertebrate system for leukaemia research,whose unique advantages complement those of extantmurine models.53Currently available zebrafish models ofacute lymphoblastic leukaemia include a myctransgene-driven system, in which lymphoblastsfaithfully reproduce the multistep oncogenic pathwaynoted in up to 60% of human T-cell acute lymphoblasticleukaemias,54,55 and a transgenic zebrafish model, inwhich the TEL-AML1 oncoprotein induces B-cellprecursor leukaemia.56

    The challenge now is to understand how thesecooperative genetic lesions and their affected pathwaysinteract to alter the proliferation, differentiation, andsurvival of lymphocyte progenitors leading to theirleukaemic conversion. This research will undoubtedlyprovide the molecular rationales needed to select newtherapeutic targets and to develop interfering smallmolecules or antibodies with high levels of antileukaemicspecificity and activity.57The table provides a partial list ofmolecularly targeted drugs now in clinical testing.

    DiagnosisPhenotype

    Immunophenotyping of leukaemic lymphoblasts by flowcytometry is essential to establish the correct diagnosisand define cell lineage. Although acute lymphoblasticleukaemia can be readily subclassified according to themany steps of normal B-cell and T-cell differentiation,the only findings with therapeutic importance are T-cell,mature B-cell, and B-cell precursor phenotypes.26,58

    Myeloid-associated antigen expression can be detected inas many as half the cases of acute lymphoblasticleukaemia. However, with contemporary treatment, thisso-called aberrant antigen expression has no prognosticimplications but can be used to distinguish leukaemiccells from normal progenitor cells, thereby enablingdetection of minimal (ie, submicroscopic) residualleukaemia.6,26

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    5/14

    Seminar

    1034 www.thelancet.com Vol 371 March 22, 2008

    GenotypeAlthough chromosomal analysis is still an integralcomponent of initial work-up of acute lymphoblasticleukaemia, other highly specific and sensitivetechniquessuch as RT-PCR, fluorescence in-situhybridisation, and flow cytometryare increasingly

    used to detect specific fusion transcripts, gain or loss ofcellular DNA content, or specific chromosomes withprognostic or therapeutic relevance.26,58 Although still aresearch technique, gene-expression profiling can notonly identify accurately the major subtypes of acutelymphoblastic leukaemia but also implicate single genesor signalling pathways as important determinants ofclinical outcome.3234,5961 Once this method has beenrefined and made cost effective, it will undoubtedlyreplace many current diagnostic techniques.

    PharmacogeneticsGenetic polymorphisms of drug transporters, receptors,targets, and drug-metabolising enzymes can affect theeffectiveness and toxic effects of antineoplastic drugs.7,62

    Traditionally, pharmacogenetic studies have focused on

    single genes identified on the basis of their influenceon the pharmacokinetics and pharmacological effectsof anticancer drugs. Findings of global gene-expressionprofiling studies have identified a growing number ofgenomic determinants of treatment responses thatcould allow development of polygenic models foroptimisation of treatment for acute lymphoblasticleukaemia.6,7,63

    Despite the promise of pharmacogenetic studies toenhance treatment outcome in acute lymphoblasticleukaemia, only polymorphisms and the activity ofthiopurine methyltransferasean enzyme that catalysesS-methylation (inactivation) of thiopurines such asmercaptopurine and thioguaninehave been useful in

    clinical practice.7,64,65 About 10% of the total populationinherit one wild-type gene encoding thiopurinemethyltransferase and one non-functional variant allele,resulting in intermediate enzyme activity, whereas 1 in300 people inherit two non-functional variant alleles withno enzyme activity. When treated with conventionaldoses of thiopurines, up to half of patients with theheterozygous deficiency and all homozygous-deficientpatients develop haemopoietic toxic effects, which can befatal in the homozygous group.7The enzyme deficiencyalso confers a high risk of developing therapy-relatedacute myeloid leukemia7 and radiation-induced braintumours, in the context of intensive thiopurinetreatment.66 Conversely, patients with high levels ofenzyme activity might be at greater risk of relapse owingto decreased exposure of leukaemic cells to active drugmetabolites.65 In most centres, studies of thiopurinemethyltransferase activity are undertaken only in peoplewith poor tolerance to antimetabolite-based therapy, andthe result is used to guide reductions in drug dosage.67We use a fairly high dose of mercaptopurine and, thus,study this enzyme prospectively in all patients, loweringthe dose of mercaptopurine in individuals with enzymedeficiency.64

    Risk assessmentCareful assessment of the risk of relapse in individual

    patients ensures that very intensive treatment is givenonly to high-risk cases, thus sparing people at lower riskfrom undue toxic effects. Although enhanced treatmenthas abolished the prognostic strength of many clinicaland biological risk factors identified in the past, wewould stress that even so-called low-risk patients need acertain degree of treatment intensification to avoidunacceptable rates of relapse. Findings have shown thatadolescents and young adults who were treated on adultprotocols fared significantly worse than the sameage-groups treated on paediatric protocols.6870 Thesuperior outcome achieved with paediatric regimenshas been attributed to more effective treatment and tobetter adherence by patients, parents, and doctors.6872To understand the actual basis for this difference in

    Mechanism of action Subtype of

    leukaemia targeted

    Clofarabine Inhibits DNA polymerase and ribonucleotide

    reductase; disrupts mitochrondria

    membrane

    All

    Nelarabine Inhibits ribonucleotide reductase and DNA

    synthesis

    T-cell

    Forodesine Inhibits purine nucleoside phosphorylase T-cell

    -sec retase inhibitors Inhibit -secretase , an enzyme req uired for

    NOTCH1 signalling

    T-cell

    Rituximab Anti-CD20 chimeric murine-human

    monoclonal antibody

    CD20-positive

    Epratuzumab Anti-CD22 humanised monoclonal antibody CD22-positive

    Alemtuzumab Anti-CD52 humanised monoclonal antibody CD52-positive

    Gemtu zu mab o zo gamicin Ant i- CD33 mo no clonal antibo dy co njugat ed

    with calicheamicin

    CD33-positive

    Imatinib mesilate ABL kinase inhibition BCR-ABL-positive

    Nilotinib ABL kinase inhibition BCR-ABL-positive

    Dasatinib BCR-ABL kinase inhibition BCR-ABL-positive

    MK-0457 Aurora kinase inhibition BCR-ABL-positive

    Lestaurtinib; midostaurin; tandutinib;

    sunitinib malate; IMC-EB10

    FMS-l ike ty ro sine k inase 3 inhibition MLL-rearranged;

    hyperdiploid

    Tipifarnib; lonafarnib Farnesyltransferase inhibition All

    Azacytidine; decitabine;

    temozolomide

    DNA methyltransferase inhibition All

    Romidepsin; vorinostat; valproic acid;

    MD-27-275; AN-9

    Histone deacetylase inhibition All

    Sirolimus; temsirolimus; everolimus;

    AP-23573

    Mammalian target-of-rapamycin inhibition All

    Bortzezomib Inhibition of ubiquitin proteasome pathway All

    Flavopiridol Serine-threonine cyclin-dependent kinase

    inhibition

    All

    Oblimersen Downregulation of BCL2 All

    17-AAG Heat shock protein-90 inhibitor BCR-ABL-positive;

    ZAP-70-positive

    Table:Selected antileukaemic drugs being tested in clinical trials

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    6/14

    Seminar

    www.thelancet.com Vol 371 March 22, 2008 1035

    outcome, several combined adult and paediatric

    consortia are using common regimens to treat patientsaged 150 years.

    Clinical factorsAge at diagnosis has a strong prognostic effect (figure 3).In work done at St Jude Childrens Research Hospital,847 children with acute lymphoblastic leukaemia wereenrolled in four consecutive treatment protocols from 1991to 2006. Children aged 19 years had a better outcome thaneither infants or adolescents.26,58 5-year event-free survivalestimates were 88% (SE 2) for children aged 19 years,73% (4) for adolescents aged 1015 years, 69% (7) for thoseolder than 15 years, and 44% (11) for babies younger than12 months. Babies younger than 6 months have an

    especially poor outcome.73,74The outcome of treatment inadults worsens with increasing age. Indeed, in the past,patients older than 60 years were not even included inclinical trials owing to their many coexisting healthproblems, their heightened susceptibility totreatment-related toxic effects, and their high frequency ofPhiladelphia chromosome-positive acute lymphoblasticleukaemia.1,4,75 This practice has begun to change, partlybecause of enhanced supportive care now available forolder adults and development of specific tyrosine kinaseinhibitors for Philadelphia chromosome-positive acutelymphoblastic leukaemia (table).75,76

    Leucocyte count is a continuous prognostic variable,with increasing counts conferring a poorer outcome,especially in patients with B-cell precursor disease.58,76In T-cell acute lymphoblastic leukaemia, a leucocytecount greater than 100x109/L is associated with anincreased risk of relapse in the CNS.77 Patients withextreme hyperleucocytosis (>400x109/L) are at high riskfor early complications such as CNS haemorrhage andpulmonary and neurological events due to leucostasis.78A uniform risk-classification system, based on both ageand leucocyte count, was devised to facilitatecomparisons of treatment results in childhood acutelymphoblastic leukaemia.26Two-thirds of patients aged19 years with a leucocyte count less than 50x109/L werejudged to have a standard (or low) risk of relapse,

    whereas the remaining third were classified as highrisk. This system by itself has limited value because upto a third of the so-called standard-risk patients couldrelapse, and individuals at very high riskwho needallogeneic haemopoietic stem-cell transplantationcannot be distinguished reliably from high-risk cases.26Moreover, risk criteria apply only to B-cell precursoracute lymphoblastic leukaemia and have little prognosticvalue in T-cell disease.

    In US cooperative group studies, black and Hispanicpatients fared worse than similarly treated whiteindividuals.79The poor prognosis for black people could berelated to their high frequency of T-cell acute lymphoblasticleukaemia and the t(1;19) chromosomal abnormality withE2A-PBX1 fusion.80However, in single-institution studies,

    black children had the same high cure rates as did whitechildren when given equal access to effective treatment,80underscoring the over-riding prognostic importance oftreatment. The adverse prognosis previously ascribed tomale sex has also been abolished with enhanced treatmentregimens.2,81

    Likewise, the effect of obesity on outcome of acutelymphoblastic leukaemia is also treatment dependent. Areport by the Childrens Oncology Group showed thatoverweight children with acute lymphoblastic leukaemia,aged 10 years or older, have a poor treatment outcome.82By contrast, we noted no association between thebody-mass index of patients with acute lymphoblasticleukaemia and clinical outcome, toxic effects, or thepharmacokinetics of several drugs tested.83

    Biological factorsT-cell and mature B-cell immunophenotypes, once

    associated with a poor outcome, have little prognosticimportance in childhood acute lymphoblastic leukaemiaand are actually favourable features in adult disease in thecontext of contemporary treatment.2,76,81,84Although geneticabnormalities do not account entirely for treatmentoutcome they still provide indispensable prognosticinformation (figure 4). 841 children with acutelymphoblastic leukaemia and successful cytogenetic andimmunophenotypic studies were enrolled in fourconsecutive treatment protocols at St Jude ChildrensResearch Hospital from 1991 to 2006. Patients withhyperdiploidy (>50 chromosomes), TEL-AML1fusion, andt(1;19)/E2A-PBX1fusion had the most favourable outcome,whereas those with the t(9;22)/BCR-ABLfusion or t(4;11)/MLL-AF4fusion had a dismal prognosis. 5-year event-free

    0

    0

    01

    02

    03

    04

    05

    06

    07

    08

    09

    1

    2 4 6 8Years from diagnosis

    10 12 14 16

    1815 years

    15 years (n=78)

    11 8 8 6 1 1 0 0

    598 528 378 295 213 149 85 31 4

    153 130 100 77 50 26 11 6 1

    78 60 40 29 20 7 1 0 0

    Probability

    Figure :Kaplan-Meier estimates of event-free survival according to age at diagnosis of acute

    lymphoblastic leukaemia

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    7/14

    Seminar

    1036 www.thelancet.com Vol 371 March 22, 2008

    survival estimates were 91% (SE 3) for hyperdiploidy, 89%

    (3) for TEL-AML1 fusion, 86% (7) for E2A-PBX1 fusion,82% (3) for other B-lineage disease, 73% (5) for T-cell acutelymphoblastic leukaemia, 37% (12) for BCR-ABL fusion,and 32% (12) for MLL-AF4fusion.

    In general, the Philadelphia chromosome, t(4;11) withMLL-AF4 fusion, and hypodiploidy (50 chromosomes), TEL-AML1fusion, andtrisomy 4, 10, and 17 are associated with favourableprognosis.6,8587About 2% of childhood cases were noted tohave intrachromosomal amplification of chromosome 21,which is associated with a B-cell precursor imm-unophenotype, older age, low white-cell counts, and, moreimportantly, a threefold increase in risk of relapse.88The

    high frequency of unfavourable genetic features and lowrate of favourable genetic abnormalities in adults withacute lymphoblastic leukaemia partly explain their inferioroutcome compared with childhood cases.76

    Age affects the prognostic importance of geneticabnormalities for unknown reasons. In children withPhiladelphia chromosome-positive acute lymphoblasticleukaemia, those aged 19 years fared better than didadolescents,89 who in turn had a better prognosis thanadults.90,91 In patients with MLL-AF4 fusion, infants andadults have a worse prognosis than children.9193 Thet(1;19) with E2A-PBX1 fusion has no prognosticimplications in childhood acute lymphoblastic leukaemiabut is still associated with a poor prognosis in some adultcases.76 Findings are scarce to suggest that activatingmutations of the NOTCH1 gene are associated with a

    favourable prognosis in childhood T-cell acute

    lymphoblastic leukaemia

    94

    but an unfavourable outcomein adults.95

    Data of microarray analyses of leukaemic cells identifiedgenes that affect the intracellular disposition ofantileukaemic drugs and have shown distinct sets ofgenes that are associated with resistance to differentclasses of antileukaemic agents.5961,9699 Aberrantexpression of some genes also seemed to haveprognostic relevance.59,60 It is also noteworthy thatnumerical chromosomal abnormalities, depending onwhether the affected chromosomes contain the wild-typeor variant allele of the genes, can greatly affect thepharmacogenomics of cancer treatment and, thus,clinical outcome.100

    Response to treatmentResponse to treatment is determined by the entire con-stellation of leukaemic-cell biological features (intrinsicdrug sensitivity) in concert with the pharmacodynamicsand pharmacogenomics of the host, the regimensadministered, and treatment adherence. Not surprisingly,the degree of reduction of the leukaemic cell clone earlyduring remission induction therapy has independentprognostic importance1,26,101104even in low-risk cases definedby clinical and biological features.105 However,morphology-based methods traditionally used to assesstreatment response are neither precise enough norsensitive enough to measure this cytoreduction reliably.26,106Molecular and flow-cytometric methods, which are at least100-fold more sensitive than morphological determinations,now allow minimal residual leukaemia to be detected atvery low levels (

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    8/14

    Seminar

    www.thelancet.com Vol 371 March 22, 2008 1037

    Treatment

    With the exception of patients with mature B-cell acutelymphoblastic leukaemia, who are treated with short-term intensive chemotherapy (including high-dosemethotrexate, cytarabine, and cyclophosphamide),109111treatment for acute lymphoblastic leukaemia typicallyconsists of a remission-induction phase, an intensification(or consolidation) phase, and continuation therapy toeliminate residual disease. Treatment is also directed tothe CNS early in the clinical course to prevent relapseattributable to leukaemic cells sequestered in this site.1The drugs currently in use for these phases weredeveloped and tested between the 1950s and 1970s, butefforts to identify new antileukaemic agents have begunto intensify (table).

    Remission-induction phaseThe goal of remission-induction treatment is to eradicatemore than 99% of the initial leukaemic cell burden andto restore normal haemopoiesis and healthy performancestatus. This approach typically includes administration ofa glucocorticoid (prednisone or dexamethasone),vincristine, and at least a third drug (asparaginase,anthracycline, or both). A three-drug induction regimenseems suffi cient for most standard-risk cases providedthey receive intensified post-remission treatment.Children with high-risk or very high-risk acute lymph-oblastic leukaemia, and virtually all adult cases of thedisease, are treated with four or more drugs for remissioninduction.1 We measure levels of minimal residualleukaemia after 2 weeks of remission induction and weintensify treatment in patients with high amounts ofresidual blasts (>1%). Clinical remission can now beinduced in 9699% of children and 7893% of adults.1Although no induction regimen is clearly superior to anyothers, addition of cyclophosphamide and intensivetreatment with asparaginase are widely consideredbeneficial to patients with T-cell acute lymphoblasticleukaemia,2,76and imatinib mesilate has greatly enhancedthe remission-induction rate, duration of disease-freesurvival, and quality of life of patients with Philadelphiachromosome-positive acute lymphoblastic leukaemia.112114

    Whether the cure rate of this subtype of leukaemia canbe raised with imatinib or the newly developed, morepotent, tyrosine kinase inhibitors nilotinib and dasatinibremains unknown.115,116

    Presumably because of its longer half-life andincreased penetration into the CNS, dexamethasonehas been deemed more effective than either prednisoneor prednisolone for treatment of acute lymphoblasticleukaemia.117,118However, findings of a small randomisedstudy showed that an augmented dose of prednisoloneproduced results comparable with those achieved withdexamethasone in the context of other intensivetreatment.119 Similarly, the pharmacodynamics ofasparaginase differ by formulation,120 and in terms ofleukaemia control, the dose intensity and duration of

    asparaginase treatment (ie, the amount of asparagine

    depletion) are far more important than the type ofasparaginase used.1,2 Compared with Escherichia coliasparaginase, Erwiniaasparaginase was associated withinferior antileukaemic response but fewer toxiceffects,121,122a finding now attributed to use of inadequatedoses of the Erwiniadrug. In some current protocols,polyethylene glycol-conjugated asparaginasea long-acting and less allergenic formhas replaced the nativeproduct in initial treatment.123126 Many complicationsrecorded during remission induction are attributable tothe synergistic effects of corticosteroid and asparaginase.In the context of multiagent treatment, a fairly smallincrease in dose of dexamethasone or asparaginase canresult in excessive toxic effects and death, especially in

    older children and adults.

    Consolidation (intensification) treatmentWith the restoration of normal haemopoiesis and bodyfunction, intensification treatment is generally used toeradicate drug-resistant residual leukaemic cells, thusreducing the risk of relapse. For example, patients withTEL-AML1-positive disease have an especially goodoutcome in clinical trials of intensive post-remissiontherapy with corticosteroids, vincristine, and aspara-ginase.81,127 Although the importance of this treatmentphase is rarely disputed, consensus is scarce on the bestregimens and duration of treatment. Frequently usedstrategies include high-dose methotrexate plusmercaptopurine, reinduction treatment with the sameagent that was given initially, frequent pulses ofvincristine and corticosteroid plus high-dose asparaginasefor 2030 weeks, and an augmented regimen consistingof reinduction treatment and additional doses ofvincristine, asparaginase, and intravenous methotrexateduring periods of myelosuppression.13,128 For patientswith high-risk or very high-risk acute lymphoblasticleukaemia, incorporation of high-dose methotrexate plusmercaptopurine into a regimen based on intensiveasparaginase treatment could be desirable. Findings ofongoing studies will establish if these approaches inchildren are effective and tolerable in adults.

    Reinduction treatment has become an integralcomponent of contemporary protocols. In one randomisedstudy of intermediate-risk acute lymphoblastic leukaemia,double reinduction further enhanced treatment outcome,whereas additional pulses of vincristine and prednisoneafter one reinduction course were not beneficial, suggestingthat the increased dose-intensity of other drugssuch asasparaginaseled to the noted improvement.129Althougha standard intensification regimen for adult acutelymphoblastic leukaemia is absent, post-remissiontreatment with cytarabine, cyclophosphamide, anthra-cyclines, and methotrexate has improved outcome in somenon-randomised studies.130132

    The best dose of methotrexate depends on theleukaemic-cell genotype and phenotype and host pharma-

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    9/14

    Seminar

    1038 www.thelancet.com Vol 371 March 22, 2008

    cogenetic and pharmacokinetic variables. Methotrexate

    at 12 g/m is adequate for most patients withstandard-risk acute lymphoblastic leukaemia, but ahigher dose (eg, 5 g/m) might benefit individuals withT-cell or high-risk B-cell precursor disease.133,134The fairlylow accumulation of methotrexate polyglutamates inblast cells with either TEL-AML1 or E2A-PBX1 fusionsuggests that patients with these genotypes could alsobenefit from an increased dose of methotrexate.98However, mega doses of methotrexate (eg, 336 g/m) donot seem necessary for patients with acute lymphoblasticleukaemia.135 Finally, leucovorin rescue, althoughnecessary after treatment with high-dose methotrexate,must not be given too early or at too high a dosagebecause it might counteract the antileukaemic effects of

    methotrexate.134136

    Allogeneic haemopoietic stem-celltransplantationAllogeneic haemopoietic stem-cell transplantation is themost intensive form of treatment for acute lymphoblasticleukaemia. Comparisons between this modality andintensive chemotherapy have yielded inconsistent resultsowing to the few patients studied and differences incase-selection criteria.137140Nonetheless, allogeneic trans-plantation clearly benefits several subgroups of patientswith high-risk acute lymphoblastic leukaemia, such asindividuals with Philadelphia chromosome-positivedisease (even when treated with a tyrosine kinaseinhibitor) and those with a poor initial response totreatment.89,113,137,140142 It also improves the outcome ofadults with the t(4;11) subtype of acute lymphoblasticleukaemia, but its benefits in infants with this genotypeare controversial.74,92,143,144Findings of studies suggest thatmatched unrelated-donor or cord-blood transplantationcould produce results comparable with those obtainedwith matched related-donor transplantation.145,146In viewof the substantial morbidity and mortality associatedwith this procedure and the growing prospects foreffective targeted therapy, the need for allogeneictransplantation should be reassessed continuously.Autologous transplantation, despite several practical

    advantages, has failed to enhance outcome in either adultor paediatric acute lymphoblastic leukaemia.138,147

    Continuation treatmentFor reasons that (currently) remain elusive, patients withacute lymphoblastic leukaemia need continuationtreatment to prevent or forestall relapse. Although abouttwo-thirds of childhood cases can be treated successfullywith only 12 months of therapy, they cannot be identifiedprospectively with any degree of certainty.148 Hence, allpatients receive chemotherapy for 2025 years. Dailymercaptopurine and methotrexate every week constitutethe backbone of continuation regimens. Many investigatorsadvocate that drug dosages be adjusted to maintainleucocyte counts below 310/L and neutrophil counts

    between 05 and 1510/L to ensure adequate dose

    intensity during the continuation phase.

    1

    Since thioguanine is more potent than mercaptopurinein model systems and leads to higher concentrations ofthioguanine nucleotides in cells and cytotoxic concentra-tions in cerebrospinal fluid,149 several randomised trialshave been done to compare the effectiveness of these twodrugs.150152Thioguanine, given at a daily dose of 40 mg/mor more, produced superior antileukaemic responses tomercaptopurine but was associated with profoundthrombocytopenia, an increased risk of death inremission, and an unacceptably high rate (1020%) ofhepatic veno-occlusive disease.150152 Although the loweractivity of thiopurine methyltransferase is associated withthioguanine-related liver damage, this measure cannot

    identify reliably patients at risk.153 Mercaptopurine,therefore, remains the drug of choice for acute lymph-oblastic leukaemia, although thioguanine could still begiven in short-term courses during the intensificationphase of treatment.

    In a multicentre randomised trial, addition of six pulsesof vincristine and dexamethasone during early continu-ation treatment failed to improve outcome of childrenwith intermediate-risk acute lymphoblastic leukaemia.154Whether more intensive pulse therapy would enhanceoutcome in the context of contemporary therapy remainsto be studied.

    CNS-directed treatmentCNS relapse is a major obstacle to cure, accounting for3040% of initial relapses in some studies.117,129,155Factorsassociated with an increased risk of CNS relapse includea T-cell immunophenotype, hyperleucocytosis, high-riskgenetic abnormalities, and presence of leukaemic cells incerebrospinal fluid (even from iatrogenic introductiondue to a traumatic lumbar puncture).3,77,156158 Poly-morphisms in genes that code for proteins implicated inthe pharmacodynamics of antileukaemic drugs have alsobeen associated with risk of CNS relapse.63

    Because of its many associated acute and latecomplications,159,160cranial irradiation is now administeredto only 520% of patients at high risk for CNS relapse. 77

    With effective systemic treatment, the radiation dose canbe lowered to 12 Gy for most patients and to 18 Gy forthose with CNS leukaemia at diagnosis.133In fact, 18 Gyirradiation was shown to be effective even in patientswith late isolated CNS relapse, in the context of intensivesystemic chemotherapy.161 We are testing the feasibilityof omitting radiation for all patients with acutelymphoblastic leukaemia, reserving its use exclusivelyfor remission retrieval therapy. Whether or not cranialradiation is used, the best regimen of intrathecal therapyshould be administered.162 To avoid traumatic lumbarpuncture from the repeated procedure and potentialCNS seeding, we give intrathecal therapy with the veryfirst diagnostic lumbar puncture, after the diagnosis ofleukaemia has been established. Some investigators

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    10/14

    Seminar

    www.thelancet.com Vol 371 March 22, 2008 1039

    recommend an Ommaya reservoir for this treatment in

    adults with acute lymphoblastic leukaemia.

    76

    In onerandomised trial, triple intrathecal therapy withmethotrexate, cytarabine, and hydrocortisone was moreeffective than intrathecal methotrexate in preventingCNS relapse, but it was associated with an increasedfrequency of bone marrow or testicular relapse.163 Oneexplanation for this seemingly paradoxical finding is thatan isolated CNS relapse is, in fact, an early manifestationof systemic relapse, and the better CNS control securedwith triple intrathecal therapy does not obviate overtleukaemic relapse in other sites. If so, more effectivesystemic chemotherapy is needed before the full benefitof triple intrathecal therapy can be realised. Indeed,systemic treatment has a substantial role in prevention

    of CNS relapse.117,118,122,152

    Remaining questions and the futureWhat are the major causative factors in the developmentof acute lymphoblastic leukaemia? Apart from isolatedcases that can be attributed to inherited genetic syn-dromes or exposures to known leukaemogenic agents,identification of causal factors with a predictable effecton substantial numbers of children or adults has notbeen possible, impeding efforts to develop effective prev-entive measures against acute lymphoblastic leukaemia.In view of the failure of large-scale epidemiologicalstudies to find such associations, future research in thisarea will probably restrict its focus to patients with acommon primary genetic lesion, such as those witheither BCR-ABL, MLL-AF4, or TEL-AML1 fusion, orhyperdiploidy.

    Assuming that molecular therapeutics will eventuallyreplace standard combination chemotherapy andhaemopoietic stem-cell transplantation in the manage-ment of patients with acute lymphoblastic leukaemia,which molecules implicated in disease pathogenesis aremost likely to yield substantial clinical benefits?Experience to date shows that transient responses can beobtained by inhibition of certain key enzymes, such astyrosine kinases, DNA methyltransferase, histonedeacetylase, secretase, serine-threonine kinases, and

    proteosomes (table).164 However, rapid development ofdrug resistance suggests that curative treatment willneed alternative strategies. For example, short-livedremissions induced in BCR-ABL-positive acute lymph-oblastic leukaemia by imatinib mesilate suggest a needto combine this drug with newly developed ABL-kinaseinhibitors, agents whose mechanism of action differsfrom that of imatinib, or with specific inhibitors ofpathways downstream of, or parallel, to the BCR-ABLpathway.39,115,116,165 A different situation arises whenNOTCH signalling is interrupted. That is, proliferativeintestinal crypt cells are destined to become post-mitoticgoblet cells in the absence of NOTCH signals,47,166raisingthe spectre of on-target toxic effects in human trials of-secretase inhibitors and other targeted therapeutics. In

    the case of -secretase inhibitors, alleviation of adverse

    effects on gastrointestinal stem cells seems to be possiblethrough an intermittent schedule that is still effectiveagainst leukaemic cells. This pitfall, and possibleavoidance strategies based on drug scheduling, will loomespecially large in leukaemia subtypes in which malignantcells have become addicted to signalling pathways thatare also essential for maintenance and renewal of healthytissues. As daunting as these challenges can seem, thepayoff in terms of understanding the pathobiology ofacute lymphoblastic leukaemia and devising noveleffective treatments with few or no toxic effects could beenormous, making it our charge to bring this promise tofruition.

    Are cancer stem cells likely to affect development of

    future targeted treatments for acute lymphoblasticleukaemia? Current evidence suggests that the stem-cellproperties of certain human cancers could cause aresurgence of tumour unless the malignant stem cells arespecifically targeted by treatment.167Findings show thattransformation of committed haemopoietic progenitorsby the MLL-AF9 oncoprotein can impart stem-cellproperties, especially a self-renewal-associated geneticprogramme.168 Whether important subpopulations ofleukaemic cells with stem-cell properties underlie somecases of relapsed acute lymphoblastic leukaemia remainsto be determined and, therefore, they must be consideredin the design of molecular therapeutics. Finally, increasingevidence suggests that the homing and engraftmentproperties of leukaemic stem cells differ from those ofnormal haemopoietic stem cells169,170and that bone-marrowmesenchymal cells can protect leukaemic cells from thecytotoxic effects of chemotherapy.171 Possibly, enhancedunderstanding of the molecular interactions betweenleukaemic cells and the bone-marrow microenvironmentwill lead to treatment strategies that enhance theantileukaemic effects of chemotherapy.

    Conflict of interest statement

    C-HP received honoraria from Enzon, Sanofi Aventis, and Mundipharmfor lectures. ATL received research support from Merck.

    Acknowledgments

    Supported in part by grants CA21765, CA51001, CA60419, CA36401,CA78224, GM61393, NR07610, CA90246, CA52259, CA68484, CA06516from the National Institutes of Health, and by the Amercian LebaneseSyrian Associated Charities. These sponsors had no role in writing of theSeminar.

    References1 Pui CH, Evans WE. Treatment of acute lymphoblastic leukemia.

    N Engl J Med2006; 354:16678.

    2 Silverman LB, Gelber RD, Dalton VK, et al. Improved outcome forchildren with acute lymphoblastic leukemia: results of Dana-FarberConsortium Protocol 91-01. Blood2001; 97:121118.

    3 Brger B, Zimmermann M, Mann G, et al. Diagnosticcerebrospinal fluid examination in children with acutelymphoblastic leukemia: significance of low leukocyte counts withblasts or traumatic lumbar puncture.J Clin Oncol2003; 21:17981.

    4 Vitale A, Guarini A, Chiaretti S, Fo R. The changing scene of adultacute lymphoblastic leukemia. Curr Opin Oncol 2006; 18:65259.

    5 Romn-Gmez J, Cordeu L, Agirre X, et al. Epigenetic regulation of

    Wnt-signaling pathway in acute lymphoblastic leukemia. Blood2007; 109:346269.

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    11/14

    Seminar

    1040 www.thelancet.com Vol 371 March 22, 2008

    6 Pui CH, Relling MV, Downing JR. Acute lymphoblastic leukemia.N Engl J Med2004; 350:153548.

    7 Evans WE, Relling MV. Moving towards individualized medicinewith pharmacogenomics. Nature2004; 429:46468.

    8 Hjalgrim LL, Westergaard T, Rostgaard K, et al. Birth weight as arisk factor for childhood leukemia: a meta-analysis of18 epidemiologic studies. Am J Epidemiol2003; 158:72435.

    9 Ahlbom A, Day N, Feychting M, et al. A pooled analysis of magneticfields and childhood leukaemia. Br J Cancer2000; 83:69298.

    10 Buffl er PA, Kwan ML, Reynolds P, Urayama KY. Environmental andgenetic risk factors for childhood leukemia: appraising theevidence. Cancer Invest2005; 23: 6075.

    11 Kinlen L. Infections and immune factors in cancer: the role ofepidemiology. Oncogene2004; 23:634148.

    12 Greaves M. Infection, immune responses and the aetiology ofchildhood leukaemia. Nat Rev Cancer2006; 6:193203.

    13 Wiemels JL, Cazzaniga G, Daniotti M, et al. Prenatal origin of acutelymphoblastic leukaemia in children. Lancet1999; 354:1499503.

    14 Maia AT, van der Velden VH, Harrison CJ, et al. Prenatal origin of

    hyperdiploid acute lymphoblastic leukemia in identical twins.Leukemia 2003; 17:200206.

    15 Hong D, Gupta R, Ancliff O, et al. Initiating and cancer-propagatingcells in TEL-AML1-associated childhood leukemia. Science2008;319: 33639.

    16 Mori H, Colman SM, Xiao Z, et al. Chromosome translocations andcovert leukemic clones are generated during normal fetaldevelopment. Proc Natl Acad Sci USA2002; 99:824247.

    17 Greaves MF, Wiemels J. Origins of chromosome translocations inchildhood leukaemia. Nat Rev Cancer2003; 3:63949.

    18 Lanciotti M, Dufour C, Corral L, et al. Genetic polymorphism ofNAD(P)H: quinone oxidoreductase is associated with an increasedrisk of infant acute lymphoblastic leukemia without MLLgenerearrangements. Leukemia2005; 19:21416.

    19 Krajinovic M, Labuda D, Sinnett D. Glutathione S-transferase P1genetic polymorphisms and susceptibility to childhood acutelymphoblastic leukaemia. Pharmacogenetics2002; 12:65558.

    20 Skibola CF, Smith MT, Kane E, et al. Polymorphisms in the methylenetetrahydrofolate reductase gene are associated with susceptibility toacute leukemia in adults. Proc Natl Acad Sci USA1999; 96:1281015.

    21 Skibola CF, Smith MT, Hubbard A, et al. Polymorphisms in thethymidylate synthase and serine hydroxymethyltransferase genes andrisk of adult acute lymphocytic leukemia. Blood 2002; 99:378691.

    22 Healy J, Blanger H, Beaulieu P, et al. Promoter SNPs in G1/Scheckpoint regulators and their impact on the susceptibility tochildhood leukemia. Blood2007; 109:68392.

    23 Gast A, Bermejo JL, Flohr T, et al. Folate metabolic genepolymorphisms and childhood acute lymphoblastic leukemia: acase-control study. Leukemia2007; 21: 32025.

    24 Spector LG, Xie Y, Robison LL, et al. Maternal diet and infantleukemia: the DNA topoisomerase II inhibitor hypothesis: a reportfrom the Childrens Oncology Group.Cancer Epidemiol Biomarkers Prev2005; 14:65155.

    25 Pui CH, Relling MV. Topoisomerase II inhibitor-related acutemyeloid leukaemia. Br J Haematol 2000; 109:1323.

    26 Pui CH, Campana D, Evans WE. Childhood acute lymphoblasticleukaemia: current status and future perspectives. Lancet Oncol2001; 2:597607.

    27 Biondi A, Cimino G, Pieters R, Pui CH. Biological and therapeuticaspects of infant leukemia. Blood2000; 96:2433.

    28 Armstrong SA, Look AT. Molecular genetics of acute lymphoblasticleukemia.J Clin Oncol2005; 23:630615.

    29 Wang JC, Dick JE. Cancer stem cells: lessons from leukemia.Trends Cell Biol2005; 15:494501.

    30 Ferrando AA, Neuberg DS, Staunton J, et al. Gene expressiondefine novel oncogenic pathways in T-cell acute lymphoblasticleukemia. Cancer Cell2002; 1:7587.

    31 Weinstein IB, Joe AK. Mechanisms of disease: oncogeneaddictiona rationale for molecular targeting in cancer therapy.Nat Clin Pract Oncol2006; 3:44857.

    32 Yeoh EJ, Ross ME, Shurtleff SA, et al. Classification, subtypediscovery, and prediction of outcome in pediatric acute

    lymphoblastic leukemia by gene expression profiling. Cancer Cell2002; 1:13343.

    33 Armstrong SA, Staunton JE, Silverman LB, et al. MLLtranslocations specify a distinct gene expression profile that

    distinguishes a unique leukemia. Nat Genet2002; 30:4147.34 Ferrando AA, Armstrong SA, Neuberg DS, et al. Gene expression

    signatures in MLL-rearranged T-lineage and B-precursor acuteleukemias: dominance of HOX dysregulation. Blood 2003;102:26268.

    35 Lu J, Getz G, Miska EA, et al. MicroRNA expression profilesclassify human cancers. Nature2005; 435:83438.

    36 Hock H, Meade E, Medeiros S, et al. Tel/Etv6 is an essential andselective regulator of adult hematopoietic stem cell survival.Genes Dev2004; 18:233641.

    37 Okuda T, van Deursen J, Hiebert SW, Grosveld G, Downing JR.AML1, the target of multiple chromosomal translocations inhuman leukemia, is essential for normal fetal liver hematopoiesis.Cell 1996; 84:32130.

    38 Wang Q, Stacy T, Binder M, et al. Disruption of the Cbfa2 genecauses necrosis and hemorrhaging in the central nervous system andblocks definitive hematopoiesis. Proc Natl Acad Sci USA1996;93:344449.

    39 Ren R. Mechanisms of BCR-ABL in the pathogenesis of chronicmyelogenous leukaemia. Nat Rev Cancer2005; 5:17283.

    40 Druker BJ. Imatinib as a paradigm of targeted therapies.Adv Cancer Res2004; 91:130.

    41 Weng AP, Ferrando AA, Lee W, et al. Activating mutations ofNOTCH1 in human T cell acute lymphoblastic leukemia. Science2004; 306:26971.

    42 Grabher C, von Boehmer H, Look AT. Notch 1 activation in themolecular pathogenesis of T-cell acute lymphoblastic leukaemia.Nat Rev Cancer2006; 6:34759.

    43 Weng AP, Millholland JM, Yashiro-Ohtani Y, et al. c-Myc is animportant direct target of Notch1 in T-cell acute lymphoblasticleukemia/lymphoma. Genes Dev2006; 20:2096109.

    44 Palomero T, Lim WK, Odom DT, et al. NOTCH1 directly regulatesc-MYC and activates a feed-forward-loop transcriptional networkpromoting leukemic cell growth. Proc Natl Acad Sci USA2006;103:18 26166.

    45 Sharma VM, Calvo JA, Draheim KM, et al. Notch1 contributes tomouse T-cell leukemia by directly inducing the expression ofc-myc. Mol Cell Biol 2006; 26:802231.

    46 Hoemann CD, Beaulieu N, Girard L, Rebai N, Jolicoeur P. Twodistinct Notch1 mutant alleles are involved in the induction ofT-cell leukemia in c-myc transgenic mice. Mol Cell Biol2000;20:383142.

    47 Wong GT, Manfra D, Poulet FM, et al. Chronic treatment with thegamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptideproduction and alters lymphopoiesis and intestinal celldifferentiation.J Biol Chem2004; 279:12 87682.

    48 Mullighan CG, Goorha S, Radtke I, et al. Genome-wide analysis ofgenetic alterations in acute lymphoblastic leukaemia.Nature2007;446:75864.

    49 Van Dyke T, Jacks T. Cancer modeling in the modern era: progressand challenges. Cell2002; 108:13544.

    50 ONeil J, Shank J, Cusson N, Murre C, Kelliher M. TAL1/SCLinduces leukemia by inhibiting the transcriptional activity ofE47/HEB. Cancer Cell2004; 5:58796.

    51 Eischen CM, Weber JD, Roussel MF, Sherr CJ, Cleveland JL.Disruption of the ARF-Mdm2-p53 tumor suppressor pathway inMyc-induced lymphomagenesis. Genes Dev1999; 13:265869.

    52 Dupuy AJ, Akagi K, Largaespada DA, Copeland NG, Jenkins NA.Mammalian mutagenesis using a highly mobile somatic SleepingBeauty transposon system. Nature2005: 436:22126.

    53 Amatruda JF, Shepard JL, Stern HM, Zon LI. Zebrafish as a cancermodel system. Cancer Cell2002; 1:22931.

    54 Langenau DM, Traver D, Ferrando AA, et al. Myc-induced T cellleukemia in transgenic zebrafish. Science2003; 299:88790.

    55 Langenau DM, Feng H, Berghmans S, et al. Cre/lox-regulatedtransgenic zebrafish model with conditional myc-induced T cellacute lymphoblastic leukemia. Proc Natl Acad Sci USA2005;102:606873.

    56 Sabaawy HE, Azuma M, Embree LJ, et al. TEL-AML1 transgeniczebrafish model of precursor B cell acute lymphoblastic leukemia.

    Proc Natl Acad Sci USA2006; 103:15 16671.57 Croce CM. Oncogenes and cancer. N Engl J Med 2008; 358:50211.

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    12/14

    Seminar

    www.thelancet.com Vol 371 March 22, 2008 1041

    58 Pui CH, Evans WE. Acute lymphoblastic leukemia. N Engl J Med1998; 339:60515.

    59 Mi S, Lu J, Sun M. MicroRNA expression signatures accuratelydiscriminate acute lymphoblastic leukemia from acute myeloidleukemia. Proc Natl Acad Sci USA; 2007; 104:1997176.

    60 Kawamata N, Ogawa S, Zimmermann M, et al. Molecularallelokaryotyping of pediatric acute lymphoblastic leukemias byhigh-resolution single nucleotide polymorphism oligonucleotidegenomic microarray. Blood2008; 111: 77684.

    61 Tissing WJ, den Boer ML, Meijerink JP, et al. Genomewideidentification of prednisolone-responsive gene in acutelymphoblastic leukemia cells. Blood2007; 109:392935.

    62 Kishi S, Cheng C, French D, et al. Ancestry and pharmacogeneticsof antileukemic drug toxicity. Blood 2007; 109:415157.

    63 Rocha JC, Cheng C, Liu W, et al. Pharmacogenetics of outcome inchildren with acute lymphoblastic leukemia. Blood2005; 105:475258.

    64 Relling MV, Pui CH, Cheng C, Evans WE. Thiopurinemethyltransferase in acute lymphoblastic leukemia. Blood2006;107:84344.

    65 Stanulla M, Schaeffeler E, Flohr T, et al. Thiopurinemethyltransferase (TPMT) genotype and early treatment responseto mercaptopurine in childhood acute lymphoblastic leukemia.JAMA2005; 293:148589.

    66 Relling MV, Rubnitz JE, Rivera GK, et al. High incidence ofsecondary brain tumours after radiotherapy and antimetabolites.Lancet1999; 354:3439.

    67 Evans WE, Hon YY, Bongaars L, et al. Preponderance ofthiopurine S-methyltransferase deficiency and heterozygosityamong patients intolerant to mercaptopurine or azathioprine.J Clin Oncol2001; 19:2293301.

    68 Boissel N, Auclerc MF, Lhritier V, et al. Should adolescents withacute lymphoblastic leukemia be treated as old children or youngadults? Comparison of the French FRALLE-93 and LALA-94 trials.J Clin Oncol2003; 21:77480.

    69 de Bont JM, Holt B, Dekker AW, et al. Significant difference inoutcome for adolescents with acute lymphoblastic leukemiatreated on pediatric vs adult protocols in the Netherlands.

    Leukemia2004; 18:203235.70 Ramanujachar R, Richards S, Hann I, et al. Adolescents with

    acute lymphoblastic leukaemia: outcome on UK nationalpaediatric (ALL97) and adult (UKALLXII/E2993) trials.Pediatr Blood Cancer2007; 48:25461.

    71 Hallbk H, Gustafsson G, Smedmyr B, et al. Treatment outcomein young adults and children >10 years of age with acutelymphoblastic leukemia in Sweden: a comparison between apediatric protocol and an adult protocol. Cancer2006; 107:155161.

    72 Barry E, DeAngelo DJ, Neuberg D, et al. Favorable outcome foradolescents with acute lymphoblastic leukemia treated onDana-Farber Cancer Institute Acute Lymphoblastic LeukemiaConsortium Protocols.J Clin Oncol2007; 25:81319.

    73 Hilden JM, Dinndorf PA, Meerbaum SO, et al. Analysis of prognosticfactors of acute lymphoblastic leukemia in infants: report on CCG1953 from the Childrens Oncology Group. Blood2006; 108:44151.

    74 Pieters R, Schrappe M, De Lorenzo P, et al. A treatment protocolfor infants younger than 1 year with acute lymphoblastic

    leukaemia (Interfant-99): an observational study and a multicentrerandomised trial.l. Lancet2007; 370:24050.

    75 Larson RA. Management of acute lymphoblastic leukemia in olderpatients. Semin Hematol2006; 43:12633.

    76 Landau H, Lamanna N. Clinical manifestations and treatment ofnewly diagnosed acute lymphoblastic leukemia in adults.Curr Hematol Malig Rep2006; 1:17179.

    77 Pui C-H. Central nervous system disease in acute lymphoblasticleukemia: prophylaxis and treatment. HematologyAm Soc HematolEduc Program2006:14246.

    78 Lowe EJ, Pui CH, Hancock ML, et al. Early complications inchildren with acute lymphoblastic leukemia presenting withhyperleukocytosis. Pediatr Blood Cancer2005; 45:1015.

    79 Kadan-Lott ick NS, Ness KK, Bhata S, Gurney JE. Survivalvariability by race and ethnicity in childhood acute lymphoblasticleukemia.JAMA2003; 290:200814.

    80 Pui CH, Sandlund JT, Pei D, et al. Results of therapy for acute

    lymphoblastic leukemia in black and white children.JAMA2003;290:200107.

    81 Pui CH, Sandlund JT, Pei D, et al. Improved outcome forchildren with acute lymphoblastic leukemia: results of Total

    Therapy Study XIIIB at St Jude Childrens Research Hospital.Blood2004; 104:269096.

    82 Butturini AM, Dorey FJ, Lange BJ, et al. Obesity and outcome inpediatric acute lymphoblastic leukemia.J Clin Oncol 2007;25:206369.

    83 Hijiya N, Panetta JC, Zhou Y, et al. Body mass index does notinfluence pharmacokinetics or outcome of treatment in childrenwith acute lymphoblastic leukemia. Blood2006; 108:39974002.

    84 Rowe JM, Buck G, Burnett AK, et al. Induction therapy for adultswith acute lymphoblastic leukemia: results of more than 1500patients from the international ALL trialMRC UKALL XII/ECOG E2993. Blood 2005; 106:376067.

    85 Moorman AV, Harrison CJ, Buck GA, et al. Karyotype is anindependent prognostic factor in adult acute lymphoblasticleukemia (ALL): analysis of cytogenetic data from patients treatedon the Medical Research Council (MRC) UKALLXII/EasternCooperative Oncology Group (ECOG) 2993 trial.Blood2007;109:318997.

    86 Nachman JB, Heerema NA, Sather H, et al. Outcome oftreatment in children with hypodiploid acute lymphoblasticleukemia. Blood2007; 110: 111215.

    87 Schultz KR, Pullen DJ, Sather HN, et al. Risk- and response-basedclassification of childhood B-precursor acute lymphoblasticleukemia: a combined analysis of prognostic markers from thePediatric Oncology Group (POG) and Childrens Cancer Group(CCG). Blood2007; 109:92635.

    88 Moorman AV, Richards SM, Robinson HM, et al. Prognosis ofchildren with acute lymphoblastic leukemia (ALL) andintrachromosomal amplification of chromosome 21 (iAMP21).Blood2007; 109:232730.

    89 Arico M, Valsecchi MG, Camitta B, et al. Outcome of treatmentin children with Philadelphia chromosome-positive acutelymphoblastic leukemia. N Engl J Med2000; 342:9981006.

    90 Gleissner B, Gokbuget N, Bartram CR, et al. Leading prognosticrelevance of the BCR-ABL translocation in adult acute B-lineage

    lymphoblastic leukemia: a prospective study of the GermanMulticenter Trial group and confirmed polymerase chain reactionanalysis. Blood2002; 99:153643.

    91 Mancini M, Scappaticci D, Cimino G, et al. A comprehensivegenetic classification of adult acute lymphoblastic leukemia(ALL): analysis of the GIMEMA 0496 protocol. Blood2005;105:343441.

    92 Pui CH, Gaynon PS, Boyett JM, et al. Outcome of treatment inchildhood acute lymphoblastic leukaemia with rearrangements ofthe 11q23 chromosomal region. Lancet2002; 359:190915.

    93 Gleissner B, Goekbuget N, Rieder H, et al. CD10- pre-B acutelymphoblastic leukemia (ALL) is a distinct high-risk subgroup ofadult ALL associated with a high frequency of MLL aberrations:results of the German Multicenter Trials for Adult ALL (GMALL).Blood2005; 106:405456.

    94 Breit S, Stanulla M, Flohr T, et al. Activating NOTCH1 mutationspredict favorable early treatment response and long-term outcomein childhood precursor T-cell lymphoblastic leukemia. Blood2006;

    108:115157.95 Zhu YM, Zhao WL, Fu JF, et al. NOTCH1 mutations in T-cell

    acute lymphoblastic leukemia: prognostic significance andimplication in multifactorial leukemogenesis. Clin Cancer Res2006; 12:304349.

    96 Cheok MH, Yang W, Pui CH, et al. Treatment-specific changes ingene expression discriminate in vivo drug response in humanleukemia cells. Nat Genet2003;34: 8590.

    97 Zaza G, Cheok M, Yang M, et al. Gene expression andthioguanine nucleotide disposition in acute lymphoblasticleukemia after in vivo mercaptopurine treatment. Blood2005;106:177885.

    98 Kager L, Cheok M, Yang W, et al. Folate pathway gene expressiondiffers in subtypes of acute lymphoblastic leukemia andinfluences methotrexate pharmacodynamics.J Clin Invest2005;115:11017.

    99 Holleman A, Cheok MH, den Boer ML, et al. Gene-expressionpatterns in drug-resistant acute lymphoblastic leukemia cells and

    response to treatment. N Engl J Med2004; 351:53342.

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    13/14

    Seminar

    1042 www.thelancet.com Vol 371 March 22, 2008

    100 Cheng Q, Yang W, Raimondi SC, et al. Karyotypic abnormal itiescreate discordance of germline genotype and cancer cell

    phenotypes. Nat Genet2005; 37:87882.101 Panzer-Grmayer ER, Schneider M, Panzer S, et al. Rapid

    molecular response during early induction chemotherapypredicts a good outcome in childhood acute lymphoblasticleukemia. Blood2000; 95:79094.

    102 Dworzak MN, Frschl G, Printz D, et al. Prognostic significance andmodalities of flow cytometric minimal residual disease detection inchildhood acute lymphoblastic leukemia. Blood2002; 99:195258.

    103 Zhou J, Goldwasser MA, Li A, et al. Quantitative analysis ofminimal residual disease predicts relapse in children withB-lineage acute lymphoblastic leukemia in DFCI ALL ConsortiumProtocol 95-01. Blood2007; 110: 160711.

    104 Bruggemann M, Raff T, Flohr T, et al. Clinical significance of minimalresidual disease quantification in adult patients with standard-riskacute lymphoblastic leukemia. Blood2006; 107:111623.

    105 Chauvenet AR, Martin PL, Devidas M, et al. Antimetabolitetherapy for lesser-risk B-lineage acute lymphoblastic leukemia ofchildhood: a report from Childrens Oncology Group Study P9201.Blood2007; 110: 110511.

    106 Coustan-Smith E, Sancho J, Behm FG, et al. Prognostic importanceof measuring early clearance of leukemic cells by flow cytometry inchildhood acute lymphoblastic leukemia. Blood2002; 100:5258.

    107 Raff T, Gkbget N, Luschen S, et al. Molecular relapse in adultstandard-risk ALL patients detected by prospective MRDmonitoring during and after maintenance treatment: data fromthe GMALL 06/99 and 07/03 trials. Blood2007; 109:91015.

    108 Coustan-Smith E, Ribeiro RC, Stow P, et al. A simplified flowcytometric assay identifies children with acute lymphoblastic leukemiawho have a superior clinical outcome. Blood2006; 108:97102.

    109 Patte C, Auperin A, Michon J, Behrendt H. The Socit FranaisedOncologie Pdiatrique LMB89 protocol: highly effectivemultiagent chemotherapy tailored to the tumor burden and initialresponse in 561 unselected children with B-cell lymphomas andL3 leukemia. Blood2001; 97:337079.

    110 Woessmann W, Seidemann K, Mann G, et al. The impact of the

    methotrexate administration schedule and dose in the treatmentof children and adolescents with B-cell neoplasms: a report of theBFM Group Study NHL-BFM95. Blood2005; 105:94858.

    111 Lee EJ, Petroni GR, Schiffer CA, et al. Brief-duration high-intensitychemotherapy for patients with small noncleaved-cell lymphomaor FAB L3 acute lymphocytic leukemia: results of cancer andleukemia group B study 9251.J Clin Oncol2001; 19:401422.

    112 Yanada M, Takeuchi J, Sugiura I, et al. High complete remissionrate and promising outcome by combination of imatinib andchemotherapy for newly diagnosed BCR-ABL-positive acutelymphoblastic leukemia: a phase II study by the Japan AdultLeukemia Study Group.J Clin Oncol2006; 24:46066.

    113 de Labarthe A, Rousselot P, Huguet-Rigal F, et al. Imatinibcombined with induction or consolidation chemotherapy inpatients with de novo Philadelphia chromosome-positive acutelymphoblastic leukemia: results of the GRAAPH-2003 study. Blood2007; 109:140813.

    114 Delannoy A, Delabesse E, Lheritier V, et al. Imatinib and

    methylprednisolone alternated with chemotherapy improve theoutcome of elderly patients with Philadelphia-positive acutelymphoblastic leukemia: results of the GRAALL AFR09 study.Leukemia2006; 20:152632.

    115 Kantarjian H, Giles F, Wunderle L, et al. Nilotinib inimatinib-resistant CML and Philadelphia chromosome-positiveALL. N Engl J Med2006; 354:254251.

    116 Talpaz M, Shah NP, Kantarjian H, et al. Dasatinib inimatinib-resistant Philadelphia chromosome-positive leukemias.N Engl J Med2006; 354:253141.

    117 Bostrom BC, Sensel MR, Sather HN, et al. Dexamethasone versusprednisone and daily oral versus weekly intravenousmercaptopurine for patients with standard-risk acute lymphoblasticleukemia: a report from the Childrens Cancer Group. Blood2003;101:380917.

    118 Mitchell CD, Richards SM, Kinsey SE, et al. Benefit ofdexamethasone compared with prednisolone for childhood acutelymphoblastic leukaemia: results of the UK Medical Research

    Council ALL97 randomized trial. Br J Haematol2005; 129:73445.

    119 Igarashi S, Manabe A, Ohara A, et al. No advantage ofdexamethasone over prednisolone for the outcome of standard- and

    intermediate-risk childhood acute lymphoblastic leukemia in theTokyo Childrens Cancer Study Group L95-14 protocol.J Clin Oncol2005; 23:648998.

    120 Pinheiro JP, Boos J. The best way to use asparaginase in childhoodacute lymphoblastic leukemia: still to be defined? Br J Haematol2004; 125:11727.

    121 Duval M, Suciu S, Ferster A, et al. Comparison of Escherichiacoli-asparaginase with Erwinia-asparaginase in the treatment ofchildhood lymphoid malignancies: results of a randomized EuropeanOrganisation for Research and Treatment of Cancer-ChildrensLeukemia Group phase 3 trial.Blood2002; 99:273439.

    122 Moghrabi A, Levy DE, Asselin B, et al. Results of the Dana-FarberCancer Institute ALL Consortium Protocol 95-01 for children withacute lymphoblastic leukemia. Blood2007; 109:896904.

    123 Avramis VI, Sencer S, Periclou AP, et al. A randomized comparisonof native Escherichia coliasparaginase and polyethylene glycolconjugated asparaginase for treatment of children with newlydiagnosed standard-risk acute lymphoblastic leukemia: a Childrens

    Cancer Group study. Blood2002; 99:198694.124 Rizzari C, Citterio M, Zucchetti M, et al. A pharmacological study

    on pegylated asparaginase used in front-line treatment of childrenwith acute lymphoblastic leukemia. Haematologica2006; 91:2431.

    125 Douer D, Yampolsky H, Cohen LJ, et al. Pharmacodynamics andsafety of intravenous pegaspargase during remission induction inadults aged 55 years or younger with newly diagnosed acutelymphoblastic leukemia. Blood2007; 109:274450.

    126 Wetzler M, Sanford BL, Kurtzberg J, et al. Effective asparaginedepletion with pegylated asparaginase results in improvedoutcomes in adult acute lymphoblastic leukemia: Cancer andLeukemia Group B Study 9511. Blood2007; 109:416467.

    127 Loh ML, Goldwasser MA, Silverman LB, et al. Prospective analysisof TEL/AML1-positive patients treated on Dana-Farber CancerInstitute Consortium Protocol 95-01. Blood2006; 107:450813.

    128 Nachman JB, Sather HN, Sensel MG, et al. Augmentedpost-induction therapy for children with high-risk acute

    lymphoblastic leukemia and a slow response to initial therapy.N Engl J Med1998; 338:166371.

    129 Lange BJ, Bostrom BC, Cherlow JM, et al. Double-delayedintensification improves event-free survival for children withintermediate-risk acute lymphoblastic leukemia: a report from theChildrens Cancer Group. Blood2002; 99:82533.

    130 Hoelzer D, Gokbuget N. New approaches to lymphoblasticleukemia in adults: where do we go? Semin Oncol2000; 27:54059.

    131 Kantarjian H, Thomas D, OBrien S, et al. Long-term follow-upresults of hyperfractionated cyclophosphamide, vincristine,doxorubicin, and dexamethasone (Hyper-CVAD), a dose-intensiveregimen, in adult acute lymphocytic leukemia. Cancer2004;101:2788801.

    132 Linker C, Damon L, Ries C, Navarro W. Intensified and shortenedcyclical chemotherapy for adult acute lymphoblastic leukemia.J Clin Oncol2002; 20:246471.

    133 Schrappe M, Reiter A, Ludwig WD, et al. Improved outcome inchildhood acute lymphoblastic leukemia despite reduced use of

    anthracyclines and cranial radiotherapy: results of trialALL-BFM 90. Blood2000; 95:331022.

    134 Pui CH, Relling MV, Evans WE. Is mega dose of methotrexatebeneficial to patients with acute lymphoblastic leukemia?Leuk Lymphoma2006; 47:243132.

    135 Nathan PC, Whitcomb T, Wolters PL, et al. Very high-dosemethotrexate (336 g/m2) as central nervous system preventivetherapy for childhood acute lymphoblastic leukemia: results ofNational Cancer Institute/Childrens Cancer Group trialsCCG-191P, CCG-134P, and CCG-144P. Leuk Lymphoma2006,47:2488504.

    136 Skrby TV, Anderson H, Heldrup J, et al. High leucovorin dosesduring high-dose methotrexate treatment may reduce the cure ratein childhood acute lymphoblastic leukemia. Leukemia2006;20:195562.

    137 Hunault M, Harousseau JL, Delain M, et al. Better outcome of adultacute lymphoblastic leukemia after early genoidentical allogeneicbone marrow transplantation (BMT) than after late high-dose therapy

    and autologous BMT: a GOELAMS trial. Blood2004; 104:302837.

  • 7/27/2019 Acute Lymphoblastic Leukaemia Lancet 2008

    14/14

    Seminar

    138 Ribera JM, Ortega JJ, Oriol A et al. Comparison of intensivechemotherapy, allogeneic or autologous stem cell transplantation as

    postremission treatment, for children with very high-risk acutelymphoblastic leukemia: PETHEMA ALL-93 Trial. J Clin Oncol2007; 25:1624.

    139 Balduzzi A, Valsecchi MG, Uderzo C, et al. Chemotherapy versusallogeneic transplantation for very-high-risk childhood acutelymphoblastic leukaemia in first complete remission: comparisonby genetic randomisation in an international prospective study.Lancet2005; 366:63542.

    140 Thomas X, Boiron JM, Huguet F, et al. Outcome of treatment inadults with acute lymphoblastic leukemia: analysis of the LALA-94trial.J Clin Oncol2004; 22:407586.

    141 Schrauder A, Reiter A, Gadner H, et al. Superiority of allogeneichematopoietic stem cell transplantation compared withchemotherapy alone in high-risk childhood T-cell acutelymphoblastic leukemia: results from ALL-BFM 90 and 95.J Clin Oncol2006; 24:574249.

    142 Vey N, Thomas X, Picard C, et al. Allogeneic stem celltransplantation improves the outcome of adults with

    t(1;19)/E2A-PBX1and t(4;11)/MLL-AF4positive B-cell acutelymphoblastic leukemia: results of the prospective multicenterLALA-94 study. Leukemia2006; 20:215561.

    143 Kosaka Y, Koh K, Kinukawa N, et al. Infant acute lymphoblasticleukemia with MLL gene rearrangements: outcome followingintensive chemotherapy and hematopoietic stem celltransplantation. Blood2004; 104:352734.

    144 Sanders JE, Im HJ, Hoffmeister PA, et al. Allogeneic hematopoieticcell transplantation for infants with acute lymphoblastic leukemia.Blood2005; 105:374956.

    145 Rocha V, Labopin M, Sanz G, et al. Transplants of umbilical-cordblood or bone marrow from unrelated donors in adults with acuteleukemia. N Engl J Med2004; 351:227685.

    146 Eapen M, Rubinstein P, Zhang MJ, et al. Outcomes oftransplantation of unrelated donor umbilical cord blood and bonemarrow in children with acute leukaemia: a comparison study.Lancet2007; 369:194754.

    147 Dhedin N, Dombret H, Thomas X, et al. Autologous stem celltransplantation in adults with acute lymphoblastic leukemia in firstcomplete remission: analysis of the LALA-85, -87 and -94 trials.Leukemia2006; 20:33644.

    148 Toyoda Y, Manabe A, Tsuchida M, et al. Six months of maintenancechemotherapy after intensified treatment for acute lymphoblasticleukemia of childhood.J Clin Oncol 2000; 18:150816.

    149 Jacobs SS, Stork LC, Bostrom BC, et al. Substitution of oral andintravenous thioguanine for mercaptopurine in a treatmentregimen for children with standard risk acute lymphoblasticleukemia: a collaborative Childrens Oncology Group/NationalCancer Institute pilot trial (CCG-1942). Pediatr Blood Cancer2007;49:25055.

    150 Harms DO, Gobel U, Spaar HJ, et al. Thioguanine offers noadvantage over mercaptopurine in maintenance treatment ofchildhood ALL: results of the randomized trial COALL-92. Blood2003; 102:273640.

    151 Stork LC, Sather H, Hutchinson RJ, et al. Comparison of

    mercaptopurine (MP) with thioguanine (TG) and IT methotrexate(ITM) with IT triples (ITT) in children with SR-ALL: results ofCCG-1952. Blood2002; 100:156a.

    152 Vora A, Mitchell CD, Lennard L, et al. Toxicity and effi cacy of6-thioguanine versus 6-mercaptopurine in childhood lymphoblasticleukaemia: a randomised trial. Lancet2006; 368:133948.

    153 Lennard L, Richards S, Cartwright CS, et al. The thiopurinemethyltransferase genetic polymorphism is associated withthioguanine-related veno-occlusive disease of the liver in childrenwith acute lymphoblastic leukemia. Clin Pharmacol Ther2006;80:37583.

    154 Conter V, Valsecchi MG, Silvestri D, et al. Pulses of vincristine anddexamethasone in addition to intensive chemotherapy for children

    with intermediate-risk acute lymphoblastic leukaemia: amulticentre randomised trial. Lancet2007; 369:12331.

    155 Hutchinson RJ, Gaynon PS, Sather H, et al. Intensification oftherapy for children with lower-risk acute lymphoblastic leukemia:long-term follow-up of patients treated on Childrens Cancer GroupTrial 1881.J Clin Oncol2003; 21:179097.

    156 Gajjar A, Harrison PL, Sandlund JT, et al. Traumatic lumbarpuncture at diagnosis adversely affects outcome in childhood acutelymphoblastic leukemia. Blood 2000; 96:338184.

    157 te Loo DM, Kamps WA, Van der Does-van den Berg AV, Van WeringER, de Graaf SS. Prognostic significance of blasts in thecerebrospinal fluid without pleiocytosis or a traumatic lumbarpuncture in children with acute lymphoblastic leukemia: theexperience of the Dutch Childhood Oncology Group.J Clin Oncol2006; 24:233236.

    158 Lazarus HM, Richards SM, Chopra R, et al. Central nervous systeminvolvement in adult acute lymphoblastic leukemia at diagnosis:results from the international ALL trial MRC UKALL XII/ECOG

    E2993. Blood2006; 108:46572.159 Pui CH, Cheng C, Leung W, et al. Extended follow-up of long-term

    survivors of childhood acute lymphoblastic leukemia. N Engl J Med2003; 349:64049.

    160 Hijiya N, Hudson MM, Lensing S, et al. Cumulative incidence ofsecondary neoplasms as a first event after childhood acutelymphoblastic leukemia.JAMA2007; 297:120715.

    161 Barredo JC, Devidas M, Lauer SJ, et al. Isolated CNS relapse ofacute lymphoblastic leukemia treated with intensive systemicchemotherapy and delayed CNS radiation: a pediatric oncologygroup study.J Clin Oncol2006; 24:314249.

    162 Pui CH. Toward optimal central nervous system-directed treatmentin childhood acute lymphoblastic leukemia.J Clin Oncol2003;21:17981.

    163 Matloub Y, Lindemulder S, Gaynon PS, et al. Intrathecal tripletherapy decreases central nervous system relapse but fails toimprove event-free survival when compared with intrathecal

    methotrexate: results of the Childrens Cancer Group (CCG) 1952study for standard-risk acute lymphoblastic leukemia, reported bythe Childrens Oncology Group. Blood2006; 108:116573.

    164 Pui CH, Jeha S. New therapeutic strategies for the treatmentof acute lymphoblastic leukaemia. Nat Rev Drug Discov2007;6:14965.

    165 Burgess MR, Skaggs BJ, Shah NP, Lee FY, Sawyers CL. Comparativeanalysis of two clinically active BCR-ABL kinase inhibitors revealsthe role of conformation-specific binding in resistance.Proc Natl Acad Sci U S A2005; 102:3395400.

    166 van Es JH, van Gijn ME, Riccio O, et al. Notch/gamma-secretaseinhibition turns proliferative cells in intestinal crypts and adenomasinto goblet cells. Nature2005; 435:95963.

    167 Jordan CT, Guzman ML, Noble M. Cancer stem cells. N Engl J Med2006; 355:125361.

    168 Krivtsov AV, Twomey D, Feng Z, et al. Transformation fromcommitted progenitor to leukaemia stem cell initiated by MLL-AF9.Nature2006; 442:81822.

    169 Krause DS, Lazarides K, von Andrian UH, van Etten RA.Requirement for CD44 in homing and engraftment ofBCR-ABL-expressing leukemic stem cells. Nat Med2006;12:117580.

    170 Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44eradicates human acute myeloid leukemic stem cells. Nat Med2006; 12:116774.

    171 Iwamoto S, Mihara K, Downing JR, Pui CH, Campana D.Mesenchymal cells regulate the response of acute lymphoblasticleukemia cells to asparaginase.J Clin Invest2007; 117:104957.


Recommended