Leukemias diagnosed in the first 12 months of life are characterized by an equal distribution of lymphoid and myeloid subtypes and account for 2.5% to 5% of acute lymphoblastic leukemias (ALLs) and 6% to 14% of acute myeloid leukemias (AMLs) of childhood.1,2 In contrast to an excess of boys among older children with leukemia, there is a slight female predominance among infants with this disease.3-5 Infant leukemias display unique biological and clinical features that have provided important insights into the mechanisms governing normal and aberrant hemopoiesis in the fetus and young children, as well as reasons for the increased rates of treatment failure in infants as compared with older children. This review summarizes recent progress in understanding the biology of infant leukemias and the prospects for better treatment.

The risk of leukemia in children, as in cancer patients in general, reflects a complex interplay between inherited predisposition, exogenous exposures to agents with leukemogenic potential, and chance events. Infant leukemias afford unique investigative models for the study of leukemogenesis. Despite the fact that such leukemias arise very early in life, the leukemogenic contribution of abnormal alleles (transmission of parental mutations) is generally assumed to be small. Familial clustering is not seen in the infant leukemias, and constitutional predisposing alleles have not been identified. However, new germinal mutations in 1 parent could affect a single predisposed offspring if the alterations occur downstream in the spermatogenetic/oogenic pathway; reciprocal translocations that target cells of the developing hemopoietic system could also play a role.6 Infant leukemias have been associated with Down syndrome,7 with Turner syndrome, and with trisomy 9.8 In contrast to other cases, some infant leukemias associated with Down syndrome undergo spontaneous remission.7 These proliferations have distinguishing morphologic, immunophenotypic, and cytogenetic features.9Infants with acute lymphoblastic leukemia (ALL) or acute myeloid leukemia (AML) usually have acquired ALL1/MLL/HRX gene fusions as the major consistent genetic abnormality (see “ALL1/MLL/HRX cloning, structure, and function: clues to pathogenesis?”).

Epidemiological and molecular genetic studies demonstrate that most, if not all, cases of infant leukemia arise in utero. First, infant leukemias exhibit fetal-type DJH joining sequences in the immunoglobulin gene.10 Second, the very early onset of some cases with 11q23 chromosomal rearrangements strongly suggests a prenatal leukemogenic event.11 In fact, a report of fetal death due to AML with an ALL1/MLL/HRX rearrangement provided direct evidence of oncogenesis in utero.12 Third, molecular studies of identical twins with leukemias harboringALL1/MLL/HRX–rearranged genes have corroborated suggestions that ALL1/MLL/HRX rearrangement is acquired in utero. Indeed, the detection of an identical clonal, nonconstitutional rearrangement of the ALL1/MLL/HRX gene in the leukemic cells of both twins provided evidence for a single clonal event in utero in 1 twin, generating leukemic clonogenic progeny that metastasized to the other twin through placental anastomoses or by crossing the maternal circulation and reaching the second twin.13-17 Finally, the same clonotypic ALL1/MLL/HRX-AF4 genomic fusion sequences have recently been backtracked to neonatal blood spots from individuals who were diagnosed with ALL at ages 51 months to 2 years.18 The fetal origin of leukemia has also been established in older children (up to 14 years old) with T-cell ALL or B-precursor cell ALL withTEL-AML1 fusion by studies of monozygotic twin pairs19-21 or neonatal blood spots (Guthrie cards).22 One interpretation of this finding is that many childhood leukemias are initiated by a mutagenic event in utero. The presence of ALL1/MLL/HRX fusion in a susceptible cell type appears sufficient to induce leukemia, whereas with other genetic alterations, additional postnatal mutations are required. It is also likely that ALL1/MLL/HRX fusion occurs more frequently during fetal development, accounting for the high incidence of this genetic abnormality in infants with leukemia.

The types of exposures that give rise to leukemogenic somatic genetic changes in utero can be assessed with greater precision in infants than in older children. Thus, maternal alcohol consumption, but not smoking, during pregnancy has been correlated with an increased risk of infant leukemia, especially AML.23,24 It was postulated that ethanol induces microsomal enzymes, such as cytochrome P450, which in turn activate precarcinogens.25 Most studies have shown that an increased incidence of high birth weights and a low incidence of low birth weights correlate with higher rates of infant ALL and AML.26-31 It was suggested that high levels of insulinlike growth factor–1 might produce large babies and contribute to leukemogenesis, an interesting theory that remains to be proved.26 An increased maternal consumption of DNA topoisomerase-II-inhibitor–containing foods, such as specific fruits and vegetables that contain quercetin; soybeans (genistein); tea, cocoa, and wine (catechins); and caffeine have all been related to an increased risk of infant AML.32-34 

ALL1/MLL/HRX gene rearrangements are common in secondary acute leukemia (usually with monoblastic or myelomonoblastic morphology) arising after exposure to an epipodophyllotoxin or an anthyacycline, both of which inhibit topoisomerase-II.35,36This observation supports the theory that exposure of pregnant women to substances that inhibit topoisomerase-II might be a critical event in the development of leukemia in infancy. Anticancer drugs, quinolone antibiotics, flavonoids, catechins, podophylline resin, benzene metabolites, and estrogens can all inhibit topoisomerase-II in vivo or in vitro and may be considered potential mutagens in the induction of acute leukemias with rearranged ALL1/MLL/HRXgenes.37 Exposures of the pregnant mother and fetus to dietary, medical, and environmental chemicals that interact with topoisomerase-II may be order-of-magnitude lower in functional doses than in exposures to drugs used in cancer chemotherapy, even though the first class of agents are as biologically active as the latter class.38 It has been proposed that interindividual pharmacogenetic-based differences in metabolism between these types of chemicals might play an important role in dose-response relationships that modulate the risk of pediatric leukemia. Thus,ALL1/MLL/HRX rearrangement has been correlated with low NAD(P)H quinone oxidoreductase (NQO1) activity,39 the quinone moiety being shared by many topoisomerase-II-inhibiting drugs and other chemicals.40-42 Some of the different maternal exposures during pregnancy that have been implicated in the genesis of infant leukemia could therefore operate via the quinone metabolic pathway.

The possible role of parental genetic susceptibility factors in modulating the effects of parental carcinogen exposure was recently suggested by studies of glutathione-S-transferase (GST) genes polymorphisms in parents of diseased infants. Among the parents of infants with leukemias lacking ALL1/MLL/HRX gene rearrangements, the frequencies of single and double GST genes (class M-GSTM; class T-GSTT) deletions were significantly higher than expected. The deletion of both GSST1 and GSTM1 genes in either parent may therefore affect the risk of infant leukemia through a pathway independent of the ALL1/MLL/HRX gene.43Whether parental preconceptional or in utero exposure to radiation increases the risk of infant leukemia remains controversial. One report suggests that there might have been a transient increase in infant leukemia in northern Greece in association with radioactive fallout from the Chernobyl accident.44 However, the European Childhood Leukemia-Lymphoma Incidence Study failed to show any increase in the incidence of childhood leukemia as a consequence of this event.45 Likewise, in a subsequent study, German investigations were not able to correlate an increased incidence of infant leukemia with ionizing radiation from the accident.46 

The ALL1/MLL/HRX gene, located at cytogenetic band 11q23, is consistently altered in infant acute leukemia, being rearranged in more than 60% to 70% of cases.1,2 This gene was identified in 1991 and completely cloned and characterized in 1992. Somatic cell-hybrids or fluorescent in situ hybridization (FISH) was used to map the chromosomal 11q23 breakpoints into a region between theCD3γ and the porhobilinogen deaminase genes. Subsequently, a yeast artificial chromosome (YAC), containing the CD3δ gene was cloned and shown by FISH analysis to span the t(4;11), t(6;11), t(9;11), and t(11;19) chromosomal translocation breakpoints.47 When a similar YAC was used, a DNA insert was obtained in which Southern blot analysis detected rearranged bands in leukemic cells from patients with t(1;11), t(4;11), t(6;11), t(9;11), t(10;11), and del(11q23) abnormalities. Breakpoints were clustered in a small region of ≈8 kilobases (kb) within a gene namedALL1 by Cimino et al48 because rearrangements were first identified in a patient with ALL. Other investigators completed characterization of the gene and added the designation MLLbecause the gene could be altered in myeloid or lymphoid leukemias, and the designations HRX or Hrtx1 to indicate homology with the trithorax (trx) gene ofDrosophila.47,49-51 ALL1/MLL/HRX spans approximately 90 kb of DNA, encodes a major transcript of ≈15 kb, and consists of 36 exons, ranging in size from 65 to 4249 base pairs. The protein product consists of more than 3910 amino acids containing 3 regions homologous to sequences of the Drosophila trx gene, including cysteine-rich regions that can fold into 6 zinc-finger–like domains and a highly conserved 200–amino acid SET domain located at the carboxyl-terminal end.49-56 

In Drosophila, the trx gene acts to spatially maintain restricted expression pattern of the Antennapoedia andBithorax complexes during fruit fly development.trx activates transcription of multiple genes of the 2 complexes and, thus, counteracts the activity of Polycomb group (PcG) genes, which repress the transcription of the same genes. Gene-targeting studies demonstrated that ALL1/MLL/HRX is also a positive regulator of Hox genes in mice.57,Hox expression is shifted posteriorly in ALL1/MLL/HRXheterozygous (+/−) embryos and completely abolished inALL1/MLL/HRX homozygous null (−/−) embryos. Shifts in Hox expression are also observed in mice with targeted mutations in PcG.58,59 More recently, Yu et al60 showed that mice with deletions of theALL1/MLL/HRX gene had altered maintenance rather than activation of the Hox gene.

The ALL1/MLL/HRX gene product possesses 2 other regions that could be directly or indirectly involved in the control of gene transcription. These are (1) a region that is similar to the AT hook of high-mobility–group-I proteins and that binds to AT-rich regions of the minor groove of the DNA and (2) a cysteine-rich region homologous to the mammalian DNA methyltransferase double helix, which by favoring conformational DNA changes, facilitates the action of other regulatory genes.49-51 

To date, at least 16 different fusion partner genes involved in chromosomal translocations with ALL1/MLL/HRX have been characterized (Table 1).61-77Additionally, internal duplications within the amino-terminal part ofALL1/MLL/HRX and specific deletions of exon 8 have been detected in leukemic blast cells of some leukemia patients78-80 

Table 1.

Structural characteristics, location, and putative functions of ALL1/MLL/HRX partner genes

Partner gene Chromosomal locationTranscript size (kb) Protein size (aa)Putative function Shared homologyReferences
AF4 or FEL 4q21  12 and 15  1200  Transcription factor  None  61, 62  
AF9 9p22  5  578  Transcription factor  ENL 61 
ENL 19p13  4.7  559  Transcription factor AF9 61, 64  
AF6 6q27  8  1612 Cytoplasmic protein  None  63  
AF1p 1p32  4.4 896  Cytoplasmic protein  None  70  
AF10 10p12 5.5  1027  Cytoplasmic protein  AF17 72 
AF17 17q21  7.5 and 5    1093  Dimerization protein  AF10 66  
ELL 19p13.1  4.4  621 Transcription factor  None  65  
AF1q 1q21  1.7 90  Growth factor  None  69  
AFX1 Xq13 Not determined  Not determined  Transcription factor  AF6q21 68  
CBP 16p13  9  2491 Transcriptional coactivator  None  71  
AF6q21 6q21 6  403  Transcription factor  AFX 77  
ENN 19p13  2.6  368  Cytoplasmic protein  None  72 
ABI-1 10p11.2  2.9  387  Cytoplasmic protein  None 75  
HCDCrel 22q11.2  2.5 and 3.5  369 Cytoplasmic protein  MSF 74  
MSF 17q25 2.8  568  Cytoplasmic protein  HCDCrel 76 
Partner gene Chromosomal locationTranscript size (kb) Protein size (aa)Putative function Shared homologyReferences
AF4 or FEL 4q21  12 and 15  1200  Transcription factor  None  61, 62  
AF9 9p22  5  578  Transcription factor  ENL 61 
ENL 19p13  4.7  559  Transcription factor AF9 61, 64  
AF6 6q27  8  1612 Cytoplasmic protein  None  63  
AF1p 1p32  4.4 896  Cytoplasmic protein  None  70  
AF10 10p12 5.5  1027  Cytoplasmic protein  AF17 72 
AF17 17q21  7.5 and 5    1093  Dimerization protein  AF10 66  
ELL 19p13.1  4.4  621 Transcription factor  None  65  
AF1q 1q21  1.7 90  Growth factor  None  69  
AFX1 Xq13 Not determined  Not determined  Transcription factor  AF6q21 68  
CBP 16p13  9  2491 Transcriptional coactivator  None  71  
AF6q21 6q21 6  403  Transcription factor  AFX 77  
ENN 19p13  2.6  368  Cytoplasmic protein  None  72 
ABI-1 10p11.2  2.9  387  Cytoplasmic protein  None 75  
HCDCrel 22q11.2  2.5 and 3.5  369 Cytoplasmic protein  MSF 74  
MSF 17q25 2.8  568  Cytoplasmic protein  HCDCrel 76 

The impressive heterogeneity of ALL1/MLL/HRX recombination raises difficult questions as to how fusion proteins cause leukemia and the role of partner genes in activating the ALL1/MLL/HRX gene and determining the leukemic phenotype. Thus, several investigators have searched for structural similarities among the differentALL1/MLL/HRX partner genes. With the exception of the homology shown by AF9 with ENL, by AF10 withAF17, by AFX with AF6q21, and by MSFwith hCDCrel, sequence analysis did not reveal structural or functional similarities. Thus, it is unlikely that these fusion partners could play a role in the function of the hybrid gene by simply providing transcriptional modulation (activation or repression) domains. By contrast, the active functional contribution of partner genes in determining the oncogenic capacityof the resulting hybrid gene is strongly suggested by several observations. All partner genes are fused in frame to ALL1/MLL/HRX, generating a full-length fusion protein, whereas terminal deletions of ALL1/MLL/HRX have not been identified in leukemic cells. In addition, by testing a series ofALL1/MLL/HRX-ENL mutants to investigate the participation of several conserved sequence motifs in the oncogenic activity of the fusion product, Slany et al81 recently demonstrated that the DNA binding motifs of ALL1/MLL/HRX, as well as the transcriptional transactivation activity of ENL,are required for in vitro immortalization of murine myeloid cells. Finally, knock-in mice expressing anALL1/MLL/HRX-AF9 fusion gene under the control of the natural ALL1/MLL/HRX promoter developed AML, while mice expressing an ALL1/MLL/HRX/Myc tag fusion gene remained free of leukemia.82 

Despite extensive studies, it is still unclear as to how fusion products participate in leukemogenesis. One possibility is that they might supply a dimerization domain, which could activate theALL1/MLL/HRX chimeric genes. This hypothesis is supported, first, by the observation that several ALL1/MLL/HRX partner genes possess structures involved in the protein-protein interactions and, second, by the finding that the novel self-fusion genetic mechanism mentioned above leads to internal duplication of the amino-terminal part of the ALL1/MLL/HRX gene, which functionally could be equivalent to a dimer of the NH2 portion of theALL1/MLL/HRX. Since the RNA polymerase elongation factorELL and the transcriptional coactivator CBP (gene for CREB binding protein) are 2 ALL1/MLL/HRX fusion partners involved in transcriptional regulation, an interaction of theALL1/MLL/HRX fusion genes with the RNA polymerase-II transcription machinery has been proposed.

Of all the ALL1/MLL/HRX motifs present in all the fusion proteins, the AT hook region is the best characterized, and several lines of evidence suggest that this region has an important role in targeting and regulating transcriptional units for normal hematopoietic growth and differentiation. In this respect, it has been suggested that the Trx protein maintains target genes in a transcriptionally active state by an epigenetic mechanism that probably involves chromatin remodeling. Although ALL1/MLL/HRX has not been shown to remodel chromatin, the carboxy-terminal SET domain of ALL1/MLL/HRXinteracts with hSNT5/INI1, a component of the SNF/SWI complex, a chromatin-remodeling system.83,84 Importantly, the SET domain is lost when the aminoterminus of the ALL1/MLL/HRX and the carboxyl-terminal partner residues fuse to form ALL1/MLL/HRX fusion protein. The loss of this domain may explain the down-regulation of theARP1 gene in embryonic stem cells from ALL1/MLL/HRXdouble-knockout mice, as ARP1 was recently identified and characterized as a target of ALL1/MLL/HRX.85 

Finally, Adler et al86 have shown that the GADD34gene, which encodes a DNA damage–inducible factor, is another target of ALL1/MLL/HRX. These authors also showed that, while ALL1/MLL/HRX protein interacts directly with GADD34, resulting in a significant increase in apoptosis after treatment with ionizing radiation, the coexpression of 3 different ALL1/MLL/HRX fusion proteins (ie, ALL1/MLL/HRX-ENL, ALL1/MLL/HRX-AF9, and ALL1/MLL/HRX-ELL) had an antiapoptotic effect, abrogating GADD34-induced apoptosis. The authors also observed a difference between wild-type and leukemic ALL1/MLL/HRX fusion proteins, leading them to postulate a gain of function for ALL1/MLL/HRX compared with the wild-type protein, suggesting that the inhibition of apoptosis may be relevant to leukemogenesis.86 

With regard to the precise function of the wild-type ALL1/MLL/HRX protein, Hess et al,87 after examining the effects of the haploinsufficiency or absence of ALL1/MLL/HRX on the in vitro differentiation of yolk sac progenitor cells, concluded that ALL1/MLL/HRX is required for the generation of normal numbers of hematopoietic progenitors and their proper differentiation, especially along the granulocytic and monocytic lineages.

Chromosomal translocations leading to oncogene activation are common events in the pathogenesis of leukemia, but the molecular basis for this process is still incompletely understood. ALL1/MLL/HRX1offers a useful model for elucidating such mechanisms. First, the gene is altered by promiscuous chromosomal recombination with a variety of partner genes in various subsets of acute leukemias, including some childhood and adult acute lymphoid or myeloid leukemias, secondary leukemias associated with prior exposure to drugs that target topoisomerase-II (etoposide, tenoposide, and anthracyclines), and, especially, infant leukemias.88-92 Second, several DNA motifs implicated in DNA-recombination mechanisms have been recently identified and localized within the ALL1/MLL/HRX1 breakpoint cluster region (bcr). These include (1) recombinase signal sequences (heptamers and nonamers); (2) scaffold attachment regions (SARs); (3) high-affinity topoisomerase-II–binding sites, including a strong site in exon 9; and (4) Alu sequences.93-95 By comparingALL1/MLL/HRX rearrangements in de novo versus therapy-related acute leukemias, Broeker et al93 identified statistically significant differences in the breakpoint distribution between the 2 groups. In particular, they found that in therapy-related acute leukemias, the breakpoints clustered in the telomeric portion of the ALL1 bcr, which is characterized by the presence of SARs and high-affinity topoisomerase-II binding sites, in contrast to cases of de novo leukemias, whose breakpoints in most instances clustered in the centromeric or 5′ bcr. On the basis of these observations, the authors suggested that the mechanisms of translocation in de novo and treatment-related leukemias secondary to treatment with topoisomerase-II inhibitors might be different.93 This conclusion has important implications for attempts to understand the etiology and pathogenesis of infant leukemias. Molecular analyses ofALL1/MLL/HRX rearrangements in infant twins showed that these genetic aberrations arise during fetal hemopoiesis in utero.13 Epidemiologic evidence has also indicated that certain conditions during pregnancy, such as exposure to drugs, alcohol, and pesticides, are associated with an increased risk of infant leukemia.96 97 

Therefore, one mechanism leading to ALL1/MLL/HRX translocations might be chromosomal breakage induced by topoisomerase-II inhibitors within the ALL1/MLL/HRX gene, while another could be represented by mistakes in DNA-repair mechanisms. This hypothesis is supported by recent observations by Aplan et al98 showing that topoisomerase-II–inhibiting drugs cleave double-stranded DNA at a site in ALL1/MLL/HRX exon 9 both in vivo and in vitro. More recently, Gillert et al99 implicated “error-prone repair” as the DNA-repair process leading to ALL1/MLL/HRXtranslocations.

From these considerations and from analogy with the involvement of theALL1/MLL/HRX gene in treatment-related leukemia, it was suggested that the critical leukemogenic event(s) occurring in utero might similarly involve prenatal exposure to topoisomerase-II inhibitors as represented by several natural and medicinal substances. That infant leukemias and topoisomerase-II–related secondary leukemias show the same biased distribution of ALL1/MLL/HRXbreaks100 lends credence to this hypothesis. A substantial list of candidate leukemogenic agents are under investigation in international case-control epidemiological studies.

Clinical and biological features

ALL of infancy is associated with a high leukocyte count at presentation, hepatosplenomegaly, and central nervous system (CNS) involvement.1,101,102 The immunophenotype is usually that of immature B-lineage precursors and is characterized by a lack of CD10 expression and the coexpression of myeloid-associated antigens. A high frequency of myeloperoxidase-gene expression typifies infant ALL.103 These findings suggest that the classic form of infant ALL originates in a stem cell that has not fully committed to lymphoid differentiation. This hypothesis is supported by the observation that multipotential stem and progenitor cells prime the commitment and differentiation of several different hematopoietic lineages.104 The frequency of ALL1/MLL/HRX gene rearrangements is very high, possibly as high as 75% when studied with molecular techniques,1 2 as would be predicted from the frequency of the t(4;11), the translocation most often involved in the generation of this fusion gene.

These 3 characteristics—lack of CD10 expression, expression of myeloid-associated markers, and ALL1/MLL/HRX gene rearrangements—are correlated with one another, and their presence is inversely related to the age of the infant.2,102,105-111 For example,ALL1/MLL/HRX rearrangement is associated with 90% of the CD10 cases, contrasted with only 20% of the CD10+ cases.109,110,112 Among infants, a lack of CD10 expression, coexpression of myeloid-associated markers,ALL1/MLL/HRX rearrangement, and age of less than 6 months are associated with a poor prognosis.2,101,102,105-107,109-115The event-free survival of infants with CD10B–precursor-cell ALL is only about 25%, as compared with 50% to 55% for those with the CD10+ phenotype.102,106,113Similar estimates apply to cases with an ALL1/MLL/HRXrearrangement: 10% to 20% as compared with 50% in cases withALL1/MLL/HRX in a germ-line configuration.102,107,109-111,115 Two studies105,111 have analyzed the relation between myeloid-associated antigen expression and outcome in infant ALL: the event-free survival in cases expressing the antigens was 0% to 10%, compared with about 60% in the other cases. Finally, age itself can be used as a prognostic factor in infant ALL. Infants younger than 6 months of age have a worse outcome (10% to 20% event-free survival) than do infants between 6 and 12 months of age at diagnosis (40% to 45% event-free survival).102,106,107 111 

The above-mentioned high-risk factors are closely interrelated. In several analyses, the ALL1/MLL/HRX rearrangement emerged as an important adverse prognostic factor.109,114-117 In one large multivariate analysis, it was shown that ALL1/MLL/HRXrearrangement, age, CD10, and white blood cell count were all independent prognostic factors.115 Two other small studies showed that the ALL1/MLL/HRX rearrangement was of prognostic relevance independent of the white blood cell count.109,114 Recent studies by the Childrens Cancer Group (CCG) suggested that the t(4;11) is the onlyALL1/MLL/HRX-related translocation associated with a dismal outcome.107,112,118 In a combined Pediatric Oncology Group (POG)–St. Jude study, infant ALL cases with ALL1/MLL/HRX-ENLfusion due to the t(11;19) had an extremely poor prognosis.119 However, the adverse outcome cannot be attributed solely to the t(4;11) or the t(11;19), as children 1 to 9 years old with this abnormality have a reasonably favorable prognosis.119-121 Thus, other factors must contribute to the generally poor treatment results obtained in infants. In a recent study by the Berlin-Frankfurt-Münster (BFM) group, a poor early-treatment response to prednisone was found to be the strongest predictor of outcome in infant ALL, even in the subgroup with the t(4;11).122 Hence, a poor early-prednisone response is being used as the sole criterion for allogenic, hematopoietic stem cell transplantation in the Interfant '99 study of infant ALL, conducted by a consortium of European and US investigators.

In AML, age and the presence of 11q23 abnormalities have no clear adverse prognostic impact.1,111,123,124 Infant AML is characterized by myelomonoblastic or monoblastic morphology, a high percentage of CNS involvement, and a high leukocyte count.ALL1/MLL/HRX rearrangements are found in about 60% of infant AML cases.111,125 The prognostic factors that define infant AML are not clearly defined. In a St. Jude study, high presenting leukocyte counts and male sex were the only 2 independent adverse prognostic factors.111 The association of high leukocyte counts with a poor outcome is not unexpected; however, the basis for the predictive strength of male sex is uncertain. Lie et al126 have also found male gender to be a predictor of a poor outcome. The prognostic impact of M4 and M5 morphology is controversial, probably owing to the use of different treatment regimens.111,127 In a recent St. Jude study, the presence of the t(9;11)(p22;q22) conferred a favorable prognosis.128Clearly, additional studies are needed to ascertain the prognostic factors in infant AML.

Drug-resistance profile

Age, immunophenotype, and ALL1/MLL/HRX rearrangement reflect or cause differences in drug-resistance factors. These can be pharmacokinetic factors that determine the amount of drugs to which the leukemic cells are exposed or differences in cellular pharmacodynamics that determine the sensitivity of the cells to the drugs. There are no data suggesting that pharmacokinetic resistance might explain the poorer outcome of infant ALL; infants simply do not show increased clearance of antileukemic drugs.

Nonetheless, some studies suggest differences at the cellular level. Kumagai et al129 showed that leukemic cells from infants with 11q23 rearrangements grow better on stromal layers in vitro than do cells from other cases. Uckun et al130 similarly showed that cells from infants with ALL1/MLL/HRX-rearranged ALL are more readily recovered from severe combined immunodeficiency (SCID) mice than are cells from children with other types of ALL. In related studies, Kersey at al131 found that leukemic cells with the t(4;11) are more resistant to stress-induced death than are other B-lineage blast cells, while Pieters et al132 showed that cells from infants with ALL are significantly more resistant in vitro to prednisolone and L-asparaginase than are cells from older children. In vitro resistance to these drugs is a strong adverse prognostic factor.133-135 Of considerable therapeutic interest, the leukemic cells of infants with ALL are significantly more sensitive to cytarabine than are cells from older children.132 In addition, B-cell precursors that lacked CD10 expression were resistant to prednisone and L-asparaginase but showed significant sensitivity to cytarabine, in contrast to cases with CD10 expression. A study by the Dana-Farber Cancer Institute Consortium showed that high-dose cytarabine given immediately after remission induction is a feasible strategy and might benefit infants with ALL.136 Recent data from a German study of adult ALL patients showed that the long-term survival of adults with pro-B ALL with the t(4;11) has increased to about 40% with the introduction of high-dose cytarabine/mitoxantrone consolidation therapy.137 Finally, Reiter et al108 reported that infants with ALL more frequently show a poor in vivo response to prednisone than do older children. In their study, age lost its prognostic value in a multivariate analysis because of its association with a poor prednisone response in vivo. A recent update of the BFM 1986 and 1990 studies confirmed the prognostic significance of the steroid response: the 6-year event-free survival for infants with a good prednisone response was 58% versus only 16% for infants with a poor prednisone response.122 

Differences between infants and older children

Rapid changes in physiologic processes govern drug disposition in infants, especially neonates. First, the total body water content as a percentage of total body weight decreases from 75% in the newborn period to 60% at 1 year and 55% by adulthood; extracellular water as a percentage of total body water decreases from 45% in the neonate to 20% in the adult.138 Second, many drugs bind less avidly to serum proteins in the neonate than in the adult, leading to an increase in the unbound fraction (presumably the active drugs) and potentially enhanced pharmacologic responses in the former age group.139 Third, the activity of many P-450 enzymes is low during infancy.140,141 This decreased metabolic activity could result in reduced cytotoxic effects of antineoplastic drugs that require bioactivation (eg, cyclophosphamide) or in enhanced cytotoxicity of those that undergo inactivation (eg, vincristine and daunorubicin). Fourth, renal tubular function and the glomerular filtration rate reach adult levels by 7 months and 5 months of age, respectively142 143; hence, any drugs that depend on renal function for clearance will have increased systemic exposure and pharmacologic effects in infants.

There are, in addition, many important anatomical differences by age. For example, the volume of the CNS relative to body surface area or body weight is much larger in young children than in adults. While the CNS volume in infants approaches 80% to 90% of the adult value by age 4 to 6 years, body surface area does not reach adult values until approximately 16 to 18 years of age.144 Indeed, Bleyer144 demonstrated that the dosage of intrathecal chemotherapy should be based on age rather than body surface area to avoid undertreatment in young children. Another major age difference is the ratio of body weight (kg) to body surface area (m2); for example, the ratio for neonates is 18, which is lower than the ratio of 25 in 5-year-olds, which in turn is a lower value than the value of 40 in adults.145 Thus, if drug dosage were based solely on body weight for all age groups, infants would receive a substantially lower dosage by body surface area than would other children. Whether the dosage of any given drug in infants should be based on body surface area or body weight remains in question. This uncertainty is well illustrated by the empirical approach to drug dosing in different clinical trials of infant leukemias, some based entirely on body weight, others on body surface area, and still others on body surface area adjusted by age (ie, proportionally lower in young infants).

There are only limited pharmacokinetic and pharmacodynamic data on individual antileukemic drugs and on the tolerance to these agents in infants. An early report suggested that infants with leukemia are more susceptible to severe vincristine neurotoxicity than are children.146 In a subsequent study of remission-induction therapy with vincristine (1.5 mg/m2), prednisone, L-asparaginase, and intrathecal methotrexate, 7 of the 9 patients with a body surface area less than 0.5 m2 developed vincristine neurotoxicity, which was severe in 4.147 It was uncertain whether L-asparaginase or methotrexate contributed to increased toxicity by altering hepatic function or whether the infant nervous system is more sensitive to vincristine. Because infants have a large body surface area relative to body weight, the authors proposed that the drug doses in infants should be calculated on the basis of body weight (in kilograms), with dosages normalized from those of body surface area (dividing by 30). This conversion effectively lowers the final dosage and has proved adequate for vincristine treatment in infants. Limited data suggest that dosing of teniposide, etoposide, and cytarabine based on body surface area would yield similar systemic exposure in infants and adults.145 By contrast, normalized dosing of doxorubicin by body weight was more likely to achieve similar systemic exposure in these 2 age groups. The study also showed that methotrexate clearance tended to be lower in infants, but there was no need to reduce dosage, as methotrexate was better tolerated in these patients. Thus, uniform rules for dosage adjustment of all antileukemic agents used in infants is inappropriate; additional pharmacokinetic and pharmacodynamic studies are needed in infants younger than 2 months of age.

Acute lymphoblastic leukemia

Contemporary treatment for childhood ALL has cured approximately 80% of patients in some clinical trials,148 but results for infant ALL are still suboptimal. A variety of treatment regimens have been tested in infants, generally yielding event-free survival rates of 20% to 35% (Table2).101,102,106,122,132,136,149-154(See also A. Biondi, unpublished data, 1999.) In several recent clinical trials, high-dose methotrexate, high-dose cytarabine, and intensive consolidation/reinduction therapy appear to have improved clinical outcome,106,112,122,136,153 but these results should be viewed as preliminary because of the small numbers of patients enrolled, the lack of randomization, and the disproportionate numbers of cases with high-risk disease (ie, ALL1/MLL/HRX-AF4fusion). Moreover, the efficacy of any treatment component is affected by the overall therapeutic strategy. Hence, while clinical trials incorporating high-dose methotrexate with or without cytarabine have generally yielded improved results, 1 study with a similar therapeutic strategy resulted in an inferior outcome, partly because of an increase in remission deaths from infection or gastrointestinal complications due to combination treatment with etoposide and high-dose cytarabine.102 Likewise, excessive toxicities and treatment-related deaths, presumably due to high-dose daunorubicin in very young infants, were encountered in a POG study, despite encouraging overall results.153 Both studies underscore the need for pharmacokinetic and pharmacodynamic studies to ensure optimal dosing in infants.

Table 2.

Treatment regimens and results for infants with ALL

Study (year) No. of patientsEvent-free survivalTreatment
Cranial irradiationHigh-dose methotrexate High-dose cytarabineConsolidation/ reinduction therapyReference
AIEOP 88-91 (1988-1995)  61 5-y = 40%  12-18 Gy if >1 y (6 mo from diagnosis)  5 g/m2 × 4 doses  —  +  * 
ATRG (1984-1988)  24  2-y DFS = 20%  all patients  —  —  −  137  
BFM 83 (1983-1086)  13 6-y = 23% ± 12% SE  ± —  —  − 113  
BFM 86 (1986-1990)  34  6-y = 37% ± 8% SE ± 5 g/m2 × 4 doses  —  +  113 
BFM 90 (1990-1995)  59  6-y = 51% ± 7% SE  ± 5 g/m2 × 4 doses  —  +  113  
CCG 101, 143, 141, 141 A, 162, 163 (1972-1982)  115  4-y = 23% 12-15 Gy (n = 8); 18-20 Gy (n = 49); 24 Gy (n = 21)  —  —  −  93  
CCG 192P (1982-84)  27  4-y = 36% 18 Gy after 1 y old  —  —  −  138  
CCG 107 (1984-1987)  99  4-y = 33% ± 5% SE  —  7.2 g/m2 × 3 doses  —  +  103  
CCG 1883 (1987-1993)  135  4-y = 39% ± 4% SE  —  7.2 g/m2 × 4 doses  1.5 to 3 g/m2 q 12 h × 4 doses × 2 courses  +  103  
DFCI 73-01, 77-01 81-01 (1973-1985)  11  4-y = 9% ± 9% SE  22 Gy after 1 y of age  —  —  −  124  
DFCI 85-01, 87-01 91-01 (1985-1995)  23  4-y = 54% ± 11% SE  18-22 Gy 130 mg/kg × 2-3 doses  100 mg/kg q 12 h × 6 doses × 1 course  −  124  
EORTC 58831, 58832 (1982-1989) 28  4-y = 43%  12-20 Gy at the end of therapy (first 9 points)  2.5 g/m2 × 4 doses  —  +  97  
MRC UKALL VIII, X (1980-1987)  48  4-y = 30% 18 Gy after 2 y of age  —  —  −  94  
MRC UK Pilot (1987-90)  40  4-y = 20%  —  8 g/m2 × 3 doses  0.5 g/m2 q 12 h × 10 doses 1 g/m2 q 12 h × 6 doses +  94  
POG 8398 (1984-1990)  37 5-y = 17% ± 7.7% SE  —  —  —  − 140  
POG 8493 (1984-1990)  82  4-y = 28% ± 5% SE —  —  —  −  141  
POG 9407 Pilot (1994-1998)  21  2-y = 50%  —  4 g/m2 × 4 doses  3 g/m2 q 12 h × 4 doses  +  142  
SJCRH Study XI (1984-1988)  11 5-y = 36% ± 1% SE  18 Gy after 1 y of age  1 g/m2 × 2 doses  —  −  143 
Study (year) No. of patientsEvent-free survivalTreatment
Cranial irradiationHigh-dose methotrexate High-dose cytarabineConsolidation/ reinduction therapyReference
AIEOP 88-91 (1988-1995)  61 5-y = 40%  12-18 Gy if >1 y (6 mo from diagnosis)  5 g/m2 × 4 doses  —  +  * 
ATRG (1984-1988)  24  2-y DFS = 20%  all patients  —  —  −  137  
BFM 83 (1983-1086)  13 6-y = 23% ± 12% SE  ± —  —  − 113  
BFM 86 (1986-1990)  34  6-y = 37% ± 8% SE ± 5 g/m2 × 4 doses  —  +  113 
BFM 90 (1990-1995)  59  6-y = 51% ± 7% SE  ± 5 g/m2 × 4 doses  —  +  113  
CCG 101, 143, 141, 141 A, 162, 163 (1972-1982)  115  4-y = 23% 12-15 Gy (n = 8); 18-20 Gy (n = 49); 24 Gy (n = 21)  —  —  −  93  
CCG 192P (1982-84)  27  4-y = 36% 18 Gy after 1 y old  —  —  −  138  
CCG 107 (1984-1987)  99  4-y = 33% ± 5% SE  —  7.2 g/m2 × 3 doses  —  +  103  
CCG 1883 (1987-1993)  135  4-y = 39% ± 4% SE  —  7.2 g/m2 × 4 doses  1.5 to 3 g/m2 q 12 h × 4 doses × 2 courses  +  103  
DFCI 73-01, 77-01 81-01 (1973-1985)  11  4-y = 9% ± 9% SE  22 Gy after 1 y of age  —  —  −  124  
DFCI 85-01, 87-01 91-01 (1985-1995)  23  4-y = 54% ± 11% SE  18-22 Gy 130 mg/kg × 2-3 doses  100 mg/kg q 12 h × 6 doses × 1 course  −  124  
EORTC 58831, 58832 (1982-1989) 28  4-y = 43%  12-20 Gy at the end of therapy (first 9 points)  2.5 g/m2 × 4 doses  —  +  97  
MRC UKALL VIII, X (1980-1987)  48  4-y = 30% 18 Gy after 2 y of age  —  —  −  94  
MRC UK Pilot (1987-90)  40  4-y = 20%  —  8 g/m2 × 3 doses  0.5 g/m2 q 12 h × 10 doses 1 g/m2 q 12 h × 6 doses +  94  
POG 8398 (1984-1990)  37 5-y = 17% ± 7.7% SE  —  —  —  − 140  
POG 8493 (1984-1990)  82  4-y = 28% ± 5% SE —  —  —  −  141  
POG 9407 Pilot (1994-1998)  21  2-y = 50%  —  4 g/m2 × 4 doses  3 g/m2 q 12 h × 4 doses  +  142  
SJCRH Study XI (1984-1988)  11 5-y = 36% ± 1% SE  18 Gy after 1 y of age  1 g/m2 × 2 doses  —  −  143 

AIEOP indicates Associazione Italiana Ematologia Oncologia Pediatrica; ATRG, Aggressive Treatment Research Group; BFM (Berlin-Frankfurt-Münster group); CCG, Children's Cancer Group; DFCI, Dana-Farber Cancer Institute/Children's Hospital ALL Consortium; EORTC, European Organization for Research and Treatment of Cancer; MRC UK, Medical Research Council, United Kingdom; POG, Pediatric Oncology Group; SJCRH, St. Jude Children's Research Hospital; DFS, disease-free survival; EFS, event-free survival.

*

Biondi A, unpublished, 1999.

Infants <12 months of age at the intended time of cranial irradiation (24 to 26 weeks from diagnosis) were not given preventive (12 Gy) or therapeutic (20/18 Gy) irradiation.

Neuropsychological abnormalities are well-recognized complications of cranial irradiation, especially in very young children. Severe neurological deficits and learning disabilities were observed in 4 of 8, in 9 of 11, and in 2 of 4 long-term survivors of infant ALL who had received cranial irradiation in 3 separate studies.101,106,136 By contrast, in another study, infants who did not receive cranial irradiation showed normal neuropsychological development when tested at 5 years of age.155 In virtually all studies, attempts have been made to reduce neuropsychological complications by reducing the dosage of cranial irradiations, delaying the radiotherapy until the child was more than 1 year old, or avoiding irradiation altogether (Table 2). To date, most investigators favor eliminating irradiation in all infants with ALL, even in those with CNS leukemia at diagnosis, relying instead on intensive systemic and intrathecal treatments. Several observations support this approach. First, in the early studies of the CCG, cranial irradiation had no impact on treatment outcome.101 Second, in the recent CCG-1883 trial, which did not include cranial irradiation, the cumulative risk of isolated CNS relapse was only 3% ± 2%,112 despite a 14.2% prevalence of CNS leukemia at diagnosis. Third, in a POG study that employed only intrathecal treatment of the CNS for all patients, failure rates were similar in infant cases with and without CNS leukemia at diagnosis.152 In fact, none of the 21 patients with CNS leukemia at diagnosis had CNS involvement at the time of relapse.

There is a paucity of data on allogeneic hematopoietic stem-cell transplantation in infants with ALL. Two limited collaborative group studies have yielded dismal results: only 2 of 11 and none of 3 patients survived.102,112 The experience of the Fred Hutchinson Cancer Research Center is more encouraging, with 7 of 9 infants alive in first remission for 2 to 11 years after allogeneic transplantation.156 Additional studies are clearly needed to determine the role of hematopoietic stem-cell transplantation in high-risk infant cases.

Acute myeloid leukemia

While infants with ALL are treated on separate protocols in most clinical trials, those with AML receive essentially the same therapy as older children in virtually all studies.1,126,157-166 Infants with acute monoblastic leukemia are sometimes treated with epipodophyllotoxin-containing regimens,167 apparently because of the increased sensitivity of their leukemic cells to this class of agents.168,169 In most clinical trials of AML therapy, event-free survival rates are similar for infants and older children.126,157-160,164,165 In the BFM 1983 and 1987 trials, children younger than 2 years had an inferior treatment outcome, as compared with older children.166 However, in multivariate analyses, age lacked independent significance after adjustment for the FAB M5 or M7 subtypes, hyperleukocytosis, and an unfavorable karyotype. In the POG 8498 study, children younger than 2 years had a more favorable outcome than did older children.161 The inclusion of children 1 to 2 years of age made it difficult to determine the prognosis for infants younger than 12 months.

Since treatment outcome generally does not differ by age group in childhood AML, there is no compelling reason to develop separate trials for infant AML, with the following exception. Infants with megakaryoblastic leukemia and the t(1;22) (p13;q13) appear to have a particularly poor prognosis170,171 and may be candidates for innovative experimental therapy or perhaps allogeneic hematopoietic stem-cell transplantation. Although the latter procedure has yielded long-term survival in some infant cases,3,172 173 its relative efficacy compared with contemporary intensive chemotherapy is unknown owing to the lack of randomized studies.

Ongoing clinical trials

Currently, 2 large international prospective studies for the treatment of infant ALL are under way. One is a collaborative US study conducted by the POG and CCG. The other is a large international effort, Interfant '99, by European and US study groups. The POG/CCG trial tests the feasibility and efficacy of intensive therapy. Infants with an ALL1/MLL/HRX rearrangement are eligible for allogeneic hematopoietic stem-cell transplantation. The Interfant '99 protocol is based on a so-called hybrid form of therapy, consisting of elements from both ALL and AML treatments administered on an ALL-like schedule and combining both low-dose and high-dose cytarabine. Only patients with a poor initial response to prednisone are eligible for hematopoietic stem-cell transplantation. No CNS or total-body irradiation is used, and anthracyclines, epipodophyllotoxins, and alkylating agents are either avoided or used only sparingly. Both studies will prospectively analyze whether age, immunophenotype, leukocyte count, initial response to therapy, and ALL1/MLL/HRXrearrangement have independent prognostic value.

Conclusion

In most cases of infant ALL and AML, the discovery ofALL1/MLL/HRX gene involvement opened new opportunities for molecular diagnosis and monitoring, molecular epidemiology, and studies to unravel basic biologic mechanisms. Continued molecular investigations are needed to gain further insight into the basic differences between leukemias in infants and older children. Current therapy for infant ALL and AML is inadequate. Although intensification of chemotherapy and wider use of allogeneic hematopoietic stem-cell transplantation could improve this situation, there remains an urgent need to develop novel therapies by exploiting the unusual biologic properties of leukemic progenitor cells expressing the abnormalALL1/MLL/HRX gene product.

We thank E. Paccagnini for secretarial support and J. Gilbert for editorial review.

Supported by Fondazione M. Tettamanti, Associazione Italiana Ricerca sul Cancro (AIRC), and MURST (A.B.); by the Foundation Pediatric Oncology Center Rotterdam and grants 94-679, 95-921, and 97-1564 from the Dutch Cancer Society (R.P.); and by a Center of Excellence Grant from the State of Tennessee, by the American Lebanese Syrian Associated Charities (ALSAC), and by grants CA51001, CA21765, CA36401, CA78224, CA20180, and CA60419 from the National Institutes of Health (C-H.P).

Reprints:Andrea Biondi, Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università Milano-Bicocca, Ospedale S. Gerardo, Via Donizetti, 106, 20052-Monza (MI) Italy; e-mail: fondazione.tettamanti@galactica.it.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Pui
CH
Kane
JR
Crist
WM
Biology and treatment of infant leukemia.
Leukemia.
9
1995
762
769
2
Greaves
MF
Infant leukemia biology, aetiology and treatment.
Leukemia.
10
1996
372
377
3
Chessells
JM
Leukaemia in the young child.
Br J Cancer Suppl.
18
1992
S54
S57
4
Birch
JM
Blair
V
The epidemiology of infant cancers.
Br J Cancer Suppl.
18
1992
S2
S4
5
Pui
CH
Raimondi
SC
Murphy
SB
Ribeiro
RC
Kalwinsky
DK
Dahl
GV
An analysis of leukemic cell chromosomal features in infants.
Blood.
69
1987
1289
1293
6
Greaves
M
A natural history for pediatric acute leukemia.
Blood.
82
1993
1043
1051
7
Rosner
F
Lee
SL
Down's syndrome and acute leukemia: myloblastic or lymphoblastic? report of forty-three cases and review of the literature.
Am J Med.
53
1972
203
218
8
Van den Berghe
H
Fryns
JP
Verresen
H
Congenital leukaemia with 46, XX, t(Bq+, Cq) cells.
J Med Genet.
9
1972
468
470
9
Litz
CE
Davies
S
Brunning
RD
et al
Acute leukemia and the transient myeloproliferative disorder associated with Down syndrome: morphologic, immunophenotypic and cytogenetic manifestations.
Leukemia.
9
1995
1432
1439
10
Wasserman
R
Galili
N
Ito
Y
Reichard
BA
Shane
S
Rovera
G
Predominance of fetal type DJH joining in young children with B precursor lymphoblastic leukemia as evidence for an in utero transforming event.
J Exp Med.
176
1992
1577
1581
11
Sansone
R
Negri
D
Cytogenetic features of neonatal leukemias.
Cancer Genet Cytogenet.
63
1992
56
61
12
Hunger
SP
McGavran
L
Meltesen
L
Parker
NB
Kassenbrock
CK
Bitter
MA
Oncogenesis in utero: fetal death due to acute myelogenous leukaemia with an MLL translocation.
Br J Haematol.
103
1998
539
542
13
Ford
AM
Ridge
SA
Cabrera
ME
et al
In utero rearrangements in the trithorax-related oncogene in infant leukaemias.
Nature.
363
1993
358
360
14
Gill Super
HJ
Rothberg
PG
Kobayashi
H
Freeman
AI
Diaz
MO
Rowley
JD
Clonal, nonconstitutional rearrangements of the MLL gene in infant twins with the acute lymphoblastic leukemia: in utero chromosome rearrangement of 11q23.
Blood.
83
1994
641
644
15
Mahmoud
HH
Ridge
SA
Behm
FG
et al
Intrauterine monoclonal origin of neonatal concordant acute lymphoblastic leukemia in monozygotic twins.
Med Pediatr Oncol.
24
1995
77
81
16
Campbell
M
Cabrera
ME
Legues
ME
Ridge
S
Greaves
M
Discordant clinical presentation and outcome in infant twins sharing a common clonal leukaemia.
Br J Haematol.
93
1996
166
169
17
Richkind
KE
Loew
T
Meisner
L
Harris
C
Wason
D
Identical cytogenetic clones and clonal evolution in pediatric monozygotic twins with acute myeloid leukemia: presymptomatic disease detection by interphase fluorescence in situ hybridization and review of the literature.
J Pediatr Hematol Oncol.
20
1998
264
267
18
Gale
KB
Ford
AM
Repp
R
et al
Backtracking leukemia to birth: identification of clonotypic gene fusion sequences in neonatal blood spots.
Proc Natl Acad Sci U S A.
94
1997
13950
13954
19
Ford
AM
Pombo-de-Oliveira
MS
McCarthy
KP
et al
Monoclonal origin of concordant T-cell malignancy in identical twins.
Blood.
89
1997
281
285
20
Ford
AM
Bennett
CA
Price
CM
Bruin
MCA
Van Wering
ER
Greaves
M
Fetal origins of the TEL-AML1 fusion gene in identical twins with leukemia.
Proc Natl Acad Sci U S A.
95
1998
4584
4588
21
Wiemels
JL
Ford
AM
Van Wering
R
Postma
A
Greaves
M
Protracted and variable latency of acute lymphoblastic leukemia after TEL-AML1 gene fusion in utero.
Blood.
94
1999
1057
1062
22
Wiemels
JL
Cazzaniga
G
Daniotti
M
et al
Prenatal origin of acute lymphoblastic leukaemia in children.
Lancet.
354
1999
1499
1503
23
Severson
RK
Buckley
JD
Woods
WG
Benjamin
D
Robison
LL
Cigarette smoking and alcohol consumption by parents of children with acute myeloid leukemia: an analysis within morphological subgroups—a report from Childrens Cancer Group.
Cancer Epidemiol Biomarkers Prev.
5
1993
433
439
24
van Duijn
CM
van Steensel-Moll
HA
Coebergh
JW
van Zanen
GE
Risk factors for childhood acute non-lymphocytic leukemia: an association with maternal alcohol consumption during pregnancy?
Cancer Epidemiol Biomarkers Prev.
3
1994
457
460
25
Shu
XO
Ross
JA
Pendergrass
TW
Reaman
GH
Lampkin
B
Robison
LL
Parental alcohol consumption, cigarette smoking, and risk of infant leukemia: a Childrens Cancer Group study.
J Natl Cancer Inst.
88
1996
24
31
26
Ross
JA
Perentesis
JP
Robison
LL
Davies
SM
Big babies and infant leukemia: a role for insulin-like growth factor-1?
Cancer Causes Control.
7
1996
553
559
27
Ross
JA
Potter
JD
Shu
XO
Reaman
GH
Lamkin
B
Robison
LL
Evaluating the relationships among material reproductive history, birth characteristics, and infant leukemia: a report from the Children's Cancer Group.
Ann Epidemiol.
7
1997
172
179
28
Westergaard
T
Andersen
PK
Pedersen
JB
et al
Birth characteristics, sibling patterns and acute leukemia risk in childhood: a population-based cohort study.
J Natl Cancer Inst.
89
1997
939
947
29
Wertelecki
W
Mantel
N
Increased birthweight in leukemia.
Pediatr Res.
7
1973
132
138
30
Kaye
SA
Robison
LL
Smithson
WA
Gunderson
P
King
FL
Neglia
JP
Maternal reproductive history and birth characteristics in childhood acute lymphoblastic leukemia.
Cancer.
68
1991
1351
1355
31
Cnattingius
S
Zack
MM
Ekbom
A
et al
Prenatal and neonatal risk factors for childhood lymphatic leukemia.
J Natl Cancer Inst.
87
1995
908
914
32
Ross
JA
Potter
JD
Reaman
GH
Pendergrass
TW
Robison
LL
Maternal exposure to potential inhibitors of DNA topoisomerase-II and infant leukemia (United States): a report from the Children's Cancer Group.
Cancer Causes Control.
7
1996
581
590
33
Ross
JA
Potter
JD
Robison
LL
Infant leukemia, topoisomerase-II inhibitors, and the MLL gene.
J Natl Cancer Inst.
86
1994
1678
1680
34
Ross
JA
Maternal diet and infant leukemia: a role for DNA topoisomerase II inhibitors?
Int J Cancer Suppl.
11
1998
26
28
35
Pui
CH
Relling
MV
Rivera
GK
et al
Epipodophyllotoxin-related acute myeloid leukemia: a study of 35 cases.
Leukemia.
9
1995
1990
1996
36
Rowley
J.D
The critical role of chromosome translocations in human leukemias.
Annu Rev Genet.
32
1998
495
519
37
Greaves
M
Aetiology of acute leukemia.
Lancet.
349
1997
344
349
38
Yamashita
Y
Kawada
SZ
Nakano
H
Induction of mammalian topoisomerase II dependent DNA cleavage by nonintercalative flavonoids, genistein and orobol.
Biochem Pharmacol.
39
1990
737
744
39
Wiemels
JL
Pagnamenta
A
Taylor
GM
Eden
OB
Alexander
FE
Greaves
MF
the United Kingdom Childhood Cancer Study Investigators
A lack of function NAD(P)H:quinone oxudoreductase allele is selectively associated with pediatric leukemias that have MLL fusions. United Kingdom Childhood Cancer Study Investigators.
Cancer Res.
59
1999
4095
4099
40
Powis
G
Free radical formation by antitumor quinones.
Free Radic Biol Med.
6
1989
63
101
41
Chen
H
Eastmond
DA
Topoisomerase inhibition by phenolic metabolites: a potential mechanism for benzene's clastogenic effects.
Carcinogenesis.
16
1995
2301
2307
42
Frydman
B
Marton
LJ
Sun
JS
et al
Induction of topoisomerase-II mediated DNA cleavage by beta-lactone and related naphthoquinones.
Cancer Res.
57
1997
620
627
43
Biondi
A
Garte
S
Crosti
F
et al
Parental exposure and susceptibility factors in the etiology of infant leukemia.
Blood.
92
1998
391a
44
Petridou
E
Trichopoulos
D
Dessypris
N
et al
Infant leukaemia after in utero exposure to radiation from Chernobyl.
Nature.
382
1996
352
353
45
Parkin
DM
Clayton
D
Black
RJ
et al
Childhood leukaemia in Europe after Chernobyl: 5 year follow-up.
Br J Cancer.
73
1996
1006
1012
46
Michaelis
J
Kaletsch
U
Burkart
W
Grosche
B
Infant leukemia after Chernobyl accident.
Nature.
387
1997
246
47
Ziemin-van der Poel
S
McCabe
N
Gill
HJ
et al
Identification of a gene, MLL, that spans the breakpoint in 11q23 translocations associated with human leukemia.
Proc Natl Acad Sci U S A.
88
1991
10735
10739
48
Cimino
G
Moir
DT
Canaani
O
et al
Cloning of ALL-1, the locus involved in leukemias with the t(4;11)(q21;q23), t(9;11)(p22;q23), and t(11;19) (q23;p13) chromosome translocations.
Cancer Res.
51
1991
6712
6739
49
Gu
Y
Nakamura
T
Alder
H
et al
The t(4;11) chromosome translocation of human acute leukemias fuses the ALL-1 gene, related to Drosophila trithorax, to the AF-4 gene.
Cell.
71
1992
701
708
50
Tkachuk
DC
Kohler
S
Cleary
ML
Involvement of a homolog of Drosophila trithorax by 11q23 chromosomal translocations in acute leukemias.
Cell.
71
1992
691
700
51
Djabali
M
Selleri
L
Parry
P
Bower
M
Young
BD
Evans
GA
A trithorax-like gene is interrupted by chromosome 11q23 translocations in acute leukaemias.
Nat Genet.
4
1993
431
52
Cimino
G
Nakamura
T
Gu
Y
et al
An altered 11-kb transcript in leukemic cell lines with t(4;11) (q21;q23) chromosome translocation.
Cancer Res.
52
1992
3811
3813
53
Rasio
D
Schichman
SA
Negrini
M
Canaani
E
Croce
CM
Complete exon structure of the ALL1 gene.
Cancer Res.
56
1996
1766
1769
54
Nilson
I
Löchner
NK
Siegler
G
et al
Exon/intron structure of the human ALL1(MLL) gene involved in translocations to chromosomal region 11q23 and acute leukaemias.
Br J Haematol.
93
1996
966
972
55
Ingham
PW
Genetic control of the spatial pattern of selector gene expression in Drosophila.
Cold Spring Harb Symp Quant Biol.
50
1986
201
208
56
Wedeen
C
Harding
K
Levine
M
Spatial regulation of Antennapoedia and bithorax gene expression of the Polycomb locus in Drosophila.
Cell.
44
1986
739
748
57
Yu
BD
Hess
JL
Horning
SE
Brown
GA
Korsmeyer
SJ
Altered Hox expression and segmental identity in Mll-mutant mice.
Nature.
378
1995
505
508
58
Akasaka
T
Kanno
M
Balling
R
Mieza
MA
Taniguchi
M
Koseki
H
A role for mel-18, a Polycomb group-related vertebrate gene, during the anteroposterior specification of the axial skeleton.
Development.
134
1996
1513
1522
59
Core
N
Bel
S
Gaunt
SJ
et al
Altered cellular proliferation in polycomb-M33-deficient mice.
Development.
124
1997
721
729
60
Yu
BD
Hanson
RD
Hess
JL
Horning
SE
Korsmeyer
SJ
MLL, a mammalian trithorax-group gene, functions as a transcriptional maintenance factor in morphogenesis.
Proc Natl Acad Sci U S A.
95
1998
10632
10636
61
Nakamura
T
Alder
H
Gu
Y
et al
Genes on chromosomes 4, 9, and 19 involved in 11q23 abnormalities in acute leukemia share sequence homology and/or common motifs.
Proc Natl Acad Sci U S A.
90
1993
4631
4635
62
Morissey
J
Tkachuck
DC
Milatovich
A
Francke
U
Link
M
Cleary
ML
A serine-proline rich protein is fused to HRX in t(4;11) acute leukemias.
Blood.
81
1993
1124
1131
63
Prasad
R
Gu
Y
Alder
H
et al
Cloning of the ALL1 fusion partner, the AF6 gene, involved in acute leukemias with t(6;11) chromosome translocation.
Cancer Res.
53
1993
5624
5628
64
Rubnitz
JE
Morissey
J
Savage
PA
Cleary
ML
ENL, the gene fused with HRX in t(11;19) leukemias, encodes a nuclear protein with transcriptional activation potential in lymphoid and myeloid cells.
Blood.
84
1994
1747
1752
65
Thirman
MJ
Levitan
DA
Kobayashi
L
Simon
MC
Rowley
J
Cloning of ELL, a gene that fuses to MLL in a t(11;19)(q23;p13.1) in acute myeloid leukemia.
Proc Natl Acad Sci U S A.
91
1994
12110
12114
66
Prasad
R
Leshkowitz
D
Gu
Y
et al
Leucine zipper dimerization motif encoded by the AF17 gene fused to ALL-1 (MLL) in acute leukemia.
Proc Natl Acad Sci U S A.
91
1994
8107
8111
67
Corral
J
Forster
A
Thompson
S
et al
Acute leukemias of different lineages have similar MLL gene fusions encoding related chimeric proteins resulting from chromosomal translocation.
Proc Natl Acad Sci U S A.
90
1993
8538
8542
68
Parry
P
Wei
Y
Evans
G
Cloning and characterization of the t(X;11) breakpoint from a leukemic cell line identify a new member of the forkhead gene family.
Genes Chromosomes Cancer.
11
1994
79
84
69
Tse
W
Zhu
W
Chen
HS
Cohen
A
A novel gene, AF1q fused to MLL in t(1;11)(q21;q23), is specifically expressed in leukemic and immature hematopoietic cells.
Blood.
85
1995
650
656
70
Bernard
OA
Mauchauffe
M
Mecucci
C
Van den Berghe
H
Berger
R
A novel gene, AF-1p, fused to HRX in t(1;11)(p32;q23), is not related to AF-1, AF-9 nor ENL.
Oncogene.
9
1994
1039
1045
71
Taki
T
Sako
M
Tsuchida
M
Hayashi
Y
The t(11;16)(q23;p13) translocation in myelodysplastic syndrome fuses the MLL gene to the CBP gene.
Blood.
89
1997
3945
3950
72
Chaplin
T
Ayton
P
Bernard
OA
et al
A novel class of zinc finger-leucine zipper genes identified from the molecular cloning of the t(10;11) translocation in acute leukemia.
Blood.
85
1995
1435
1441
73
So
CW
Caldas
C
Liu
MM
et al
EEN encodes for a member of a new family of proteins containing an Src homology 3 domain and is the third gene located on chromosome 19p13 that fuses to MLL in human leukemia.
Proc Natl Acad Sci U S A.
94
1997
2563
2568
74
Megonigal
M
Rappaport
EF
Jones
DH
et al
t(11;22)(q23;q11.2) in acute myeloid leukemia of infant twins fuses MLL with hCDCrel, a cell division cycle gene in the genomic region of deletion in DiGeorge and velocardiofacial syndromes.
Proc Natl Acad Sci U S A.
95
1998
6413
6418
75
Taki
T
Shibuya
N
Taniwaki
M
et al
ABI-1, a human homolog to mouse Abl-interactor 1, fuses the MLL gene in acute myeloid leukemia with t(10;11)(p11.2;q23).
Blood.
92
1998
1125
1130
76
Osaka
M
Rowley
JD
Zeleznik-Le
NJ
MSF (MLL septin-like fusion), a fusion partner gene of MLL, in a therapy-related acute myeloid leukemia with a t(11;17)(q23;q25).
Proc Natl Acad Sci U S A.
96
1999
6428
6433
77
Hillion
J
Le Coniat
M
Jonveaux
P
Berger
R
Bernard
OA
AF6q21, a novel partner of the MLL gene in t(6;11)(q21;q23), defines a forkhead transcriptional factor subfamily.
Blood.
90
1997
3714
3719
78
Schichman
SA
Caligiuri
MA
Strout
MP
et al
ALL-1 tandem duplication in acute myeloid leukemia with normal karyotype involves homologous recombinations between Alu elements.
Cancer Res.
54
1994
4277
4280
79
Shichman
SA
Caligiuri
MA
Gu
Y
et al
ALL-1 partial duplication in acute myeloid leukemia.
Proc Natl Acad Sci U S A.
91
1994
6236
6239
80
Löchner
K
Siegler
G
Führer
M
et al
A specific deletion in the breakpoint cluster region of the ALL-1 gene is associated with acute lymphoblastic T-cell leukemias.
Cancer Res.
56
1996
2171
2177
81
Slany
RK
Lavau
C
Cleary
ML
The oncogenic capacity of HRX-ENL requires the transcriptional transactivation activity of ENL and the DNA binding motifs of HRX.
Mol Cell Biol.
18
1998
122
129
82
Corral
J
Lavenir
I
Impey
H
et al
An Mll-AF9 fusion gene made by homologous recombination causes acute leukemia in chimeric mice: a method to create fusion oncogenes.
Cell.
85
1996
853
861
83
Cui
X
De Vivo
I
Slany
R
Miyamoto
A
Firestein
R
Cleary
ML
Association of SET domain and myotubularin-related proteins modulates growth control.
Nat Genet.
18
1998
331
337
84
Rozenblatt-Rosen
O
Rozovskaia
T
Bukarov
D
et al
The C-terminal SET domains of ALL1 and TRITHORAX interact with the INI1 and SNR1 proteins, components of the SWI/SNF complex.
Proc Natl Acad Sci U S A.
95
1998
4152
4157
85
Arakawa
H
Nakamura
T
Zhadanov
AB
et al
Identification and characterization of the ARP1 gene, a target for the human acute leukemia ALL1 gene.
Proc Natl Acad Sci U S A.
95
1998
4573
4578
86
Adler
HT
Chinery
R
Wu
DY
et al
Leukemic HRX fusion proteins inhibit GADD34-induced apoptosis and associate with the GADD34 and hSNF5/INI1 proteins.
Mol Cell Biol.
19
1999
7050
7060
87
Hess
JL
Yu
BD
Li
B
Hanson
R
Korsmeyer
JS
Defects in yolk sac hematopoiesis in MLL-null embryos.
Blood.
90
1997
1799
1806
88
Cimino
G
Lo Coco
F
Biondi
A
et al
ALL-1 gene at chromosome 11q23 is consistently altered in acute leukemia of early infancy.
Blood.
82
1993
544
546
89
Bower
M
Parry
P
Carter
M
et al
Prevalence and clinical correlations of MLL gene rearrangements in AML-M4/5.
Blood.
84
1994
3776
3780
90
Cimino
G
Rapanotti
MC
Elia
L
et al
ALL-1 gene rearrangements in acute myeloid leukemia: association with M4-M5 French-American-British classification subtypes and young age.
Cancer Res.
55
1995
1625
1628
91
Poirel
H
Rack
K
Delabesse
E
et al
Incidence and characterization of MLL gene (11q23) rearrangements in acute myeloid leukemia M1 and M5.
Blood.
87
1996
2496
2505
92
Felix
CA
Lange
BJ
Hosler
MR
Fertala
J
Bjornsti
MA
Chromosome band 11q23 translocation breakpoints are DNA topoisomerase II cleavage sites.
Cancer Res.
55
1995
4287
4292
93
Broeker
PL
Super
HG
Thirman
MJ
et al
Distribution of 11q23 breakpoints within the MLL breakpoint cluster region in de novo acute leukemia and in treatment-related acute myeloid leukemia: correlation with scaffold attachment regions and topoisomerase II consensus binding sites.
Blood.
87
1996
1912
1922
94
Gu
Y
Cimino
G
Alder
H
et al
The t(4;11)(q21;q23) chromosome translocations in acute leukemias involve the VDJ recombinase.
Proc Natl Acad Sci U S A.
89
1992
10464
10468
95
Gu
Y
Alder
H
Nakamura
T
et al
Sequence analysis of the breakpoint cluster region in the ALL-1 gene involved in acute leukemia.
Cancer Res.
54
1994
2326
2330
96
Ross
JA
Davies
SM
Potter
JD
Robison
LL
Epidemiology of childhood leukemia with a focus on infants.
Epidemiol Rev.
16
1994
243
272
97
Buckley
JD
Robison
LL
Swotinsky
R
et al
Occupational exposures of parents of children with acute nonlymphocytic leukemia: a report from the Childrens Cancer Study Group.
Cancer Res.
49
1989
4030
4037
98
Aplan
PD
Chervinsky
DS
Stanulla
M
Burhans
WC
Site-specific DNA cleavage within the MLL breakpoint cluster region induced by topoisomerase II inhibitors.
Blood.
87
1996
2649
2658
99
Gillert
E
Leis
T
Repp
R
et al
A DNA damage repair mechanism is involved in the origin of chromosomal translocations t(4;11) in primary leukemic cells.
Oncogene.
18
1999
4663
4671
100
Cimino
G
Rapanotti
MC
Biondi
A
et al
Infant acute leukemias show the same biased distribution of ALL1 gene breaks as topoisomerase II related secondary acute leukemias.
Cancer Res.
57
1997
2879
2883
101
Reaman
G
Zeltzer
P
Bleyer
A
et al
Acute lymphoblastic leukemia in infants less than one year of age: a cumulative experience of the Children's Cancer Study Group.
J Clin Oncol.
3
1985
1513
1521
102
Chessells
JM
Eden
OB
Bailey
CC
Lilleyman
JS
Richards
SM
Acute lymphoblastic leukaemia in infancy: experience in MRC UKALL trials report from the Medical Research Council Working Party on Childhood Leukaemia.
Leukemia.
8
1994
1275
1279
103
Alvarado
CS
Austin
GE
Borowitz
MJ
et al
Myeloperoxidase gene expression in infant leukemia: a Pediatric Oncology Group Study.
Leuk Lymphoma.
29
1998
145
160
104
Hu
M
Krause
D
Greaves
M
et al
Multilineage gene expression precedes commitment in the hemopoietic system.
Genes Dev.
11
1997
774
785
105
Basso
G
Rondelli
R
Covezzoli
A
Putti
MC
The role of immunophenotype in acute lymphoblastic leukemia of infant age.
Leuk Lymphoma.
15
1994
51
60
106
Ferster
A
Bertrand
Y
Benoit
Y
et al
Improved survival for acute lymphoblastic leukaemia in infancy: the experience of EORTC-Childhood Leukaemia Cooperative Group.
Br J Haematol.
86
1994
284
290
107
Heerema
NA
Arthur
DC
Sather
H
et al
Cytogenetic features of infants less than 12 months of age at diagnosis of acute lymphoblastic leukemia: impact of the 11q23 breakpoint on outcome: a report of the Childrens Cancer Group.
Blood.
83
1994
2274
2284
108
Reiter
A
Schrappe
M
Ludwig
WD
et al
Chemotherapy in 998 unselected childhood acute lymphoblastic leukemia patients: results and conclusions of the multicenter trial ALL-BFM 86.
Blood.
84
1994
3122
3133
109
Cimino
G
Rapanotti
MC
Rivolta
A
et al
Prognostic relevance of ALL-1 gene rearrangement in infant leukemias.
Leukemia.
9
1995
391
395
110
Hilden
JM
Frestedt
JL
Moore
RO
et al
Molecular analysis of infant acute lymphoblastic leukemia: MLL rearrangement and reverse transcriptase-polymerase chain reaction for t(4;11)(q21;q23).
Blood.
86
1995
3876
3882
111
Pui
CH
Ribeiro
RC
Campana
D
et al
Prognostic factors in the acute lymphoid and myeloid leukemias of infants.
Leukemia.
10
1996
952
956
112
Reaman
GH
Sposto
R
Sensel
MG
et al
Treatment outcome and prognostic factors for infants with acute lymphoblastic leukemia treated on two consecutive trials of the Children's Cancer Group.
J Clin Oncol.
17
1999
445
455
113
Basso
G
Putti
MC
Cantu-Rajnoldi
A
et al
The immunophenotype in infant acute lymphoblastic leukaemia: correlation with clinical outcome. an Italian multicentre study (AIEOP).
Br J Haematol.
81
1992
184
191
114
Pui
CH
Behm
FG
Downing
JR
et al
11q23/MLL rearrangement confers a poor prognosis in infants with acute lymphoblastic leukemia.
J Clin Oncol.
12
1994
909
915
115
Rubnitz
JE
Link
MP
Shuster
JJ
et al
Frequency and prognostic significance of HRX rearrangements in infant acute lymphoblastic leukemia: a Pediatric Oncology Group study.
Blood.
2
1994
570
573
116
Chen
CS
Sorensen
PHB
Domer
PH
et al
Molecular rearrangements on chromosome 11q23 predominate in infant acute lymphoblastic leukemia and are associated with specific biologic variables and poor outcome.
Blood.
81
1993
2386
2393
117
Taki
T
Ida
K
Bessho
F
et al
Frequency and clinical significance of the MLL gene rearrangements in infant acute leukemia.
Leukemia.
10
1996
1303
1307
118
Heerema
NA
Sather
HN
Ge
J
et al
Cytogenetic studies of infant acute lymphoblastic leukemia: poor prognosis of infants with t(4;11): a report of the Children's Cancer Group.
Leukemia.
13
1999
679
686
119
Rubnitz
JE
Camitta
BM
Mahmoud
H
et al
Childhood acute lymphoblastic leukemia with the MLL-ENL fusion and t(11;19) (q23;p13.3) translocation.
J Clin Oncol.
17
1999
191
196
120
Pui
CH
Frankel
LS
Carroll
AJ
et al
Clinical characteristics and treatment outcome of childhood acute lymphoblastic leukemia with the t(4;11) (q21;q23): a collaborative study of 40 cases.
Blood.
77
1991
440
447
121
Johansson
B
Moorman
AV
Haas
OA
et al
Haematologic malignancies with t(4;11)(q21;q23): a cytogenetic, morphologic, immunophenotypic and clinical study of 183 cases.
Leukemia.
12
1998
779
787
122
Dördelmann
M
Reiter
A
Borkhardt
A
et al
Prednisone response is the strongest predictor of treatment outcome in infant acute lymphoblastic leukemia.
Blood.
94
1999
1209
1217
123
Vormoor J, Ritter J, Creutzig U, et al. Acute myelogenous leukaemia in children under 2 years: experiences of the West German AML studies BFM-78, -83 and -87. AML-BFM Study Group. Br J Cancer Suppl. 1992;S63-S67.
124
Satake
N
Maseki
N
Nishiyama
M
et al
Chromosome abnormalities and MLL rearrangement in acute myeloid leukemia of infants.
Leukemia.
13
1999
1013
1017
125
Sorensen
PH
Chen
CS
Smith
FO
et al
Molecular rearrangements of the MLL gene are present in most cases of infant acute myeloid leukemia and are strongly correlated with monocytic or myelomonocytic phenotypes.
J Clin Invest.
93
1994
429
437
126
Lie
SO
Jonmundsson
G
Mellander
L
Siimes
MA
Yssing
M
Gustafsson
G
A population-based study of 272 children with acute myeloid leukaemia treated on two consecutive protocols with different intensity: best outcome in girls, infants, and children with Down's syndrome. Nordic Society of Paediatric Haematology and Oncology (NOPHO).
Br J Haematol.
94
1996
82
88
127
Grier
HE
Gelber
RD
Camitta
BM
et al
Prognostic factors in childhood acute myelogenous leukemia.
J Clin Oncol.
5
1987
1026
1032
128
Pui
C-H
Raimondi
SC
Srivastava
DK
et al
Prognostic factors in infants with acute myeloid leukemia.
Leukemia.
14
2000
684
687
129
Kumagai
M
Manabe
A
Pui
CH
et al
Stroma-supported culture in childhood B-lineage acute lymphoblastic leukemia cells predicts treatment outcome.
J Clin Invest.
97
1996
755
760
130
Uckun
FM
Sather
H
Reaman
G
et al
Leukemic cell growth in SCID mice as a predictor of relapse in high-risk B-lineage acute lymphoblastic leukemia.
Blood.
85
1995
873
878
131
Kersey
JH
Wang
D
Oberto
M
Resistance of t(4;11) (MLL-AF4 fusion gene) leukemias to stress-induced cell death: possible mechanism for extensive extramedullary accumulation of cells and poor prognosis.
Leukemia.
12
1998
1561
1564
132
Pieters
R
den Boer
ML
Durian
M
et al
Relation between age, immunophenotype and in vitro drug resistance in 395 children with acute lymphoblastic leukemia: implications for treatment of infants.
Leukemia.
12
1998
1344
1348
133
Pieters
R
Huismans
DR
Loonen
AH
et al
Relation of cellular drug resistance to long-term clinical outcome in childhood acute lymphoblastic leukemia.
Lancet.
338
1991
399
403
134
Kaspers
GJ
Veerman
AJ
Pieters
R
et al
In vitro cellular drug resistance and prognosis in newly diagnosed childhood acute lymphoblastic leukemia.
Blood.
90
1997
2723
2729
135
Hongo
T
Yajima
S
Sakurai
M
Horiloshi
Y
Hanada
R
In vitro drug sensitivity testing can predict induction failure and early relapse of childhood acute lymphoblastic leukemia.
Blood.
89
1997
2959
2965
136
Silverman
LB
McLean
TW
Gelber
RD
et al
Intensified therapy for infants with acute lymphoblastic leukemia (ALL): results from the Dana-Farber Cancer Institute Consortium.
Cancer.
80
1997
2285
2295
137
Ludwig
WD
Rieder
H
Bartram
CR
et al
Immunophenotypic and genotypic features, clinical characteristics, and treatment outcome of adult pro-B acute lymphoblastic leukemia: results of the German Multicenter trials GMALL 03/87 and 04/89.
Blood.
92
1998
1898
1909
138
Friis-Hansen B. Body composition during growth: in vivo measurements and biochemical data correlated to differential anatomical growth. Pediatrics. 1971(suppl 2);47:264.
139
Stewart
CF
Hampton
EM
Effect of maturation on drug disposition in pediatric patients.
Clin Pharm.
6
1987
548
564
140
Pelkonen
O
Kaltiala
EH
Larmi
TKI
Karki
N
Comparison of activities of drug-metabolizing enzymes in human fetal and adult livers.
Clin Pharmacol Ther.
14
1973
840
846
141
Aranda
JV
Macleod
SM
Renton
KE
Eade
NR
Hepatic microsomeal drug oxidation and electron transport in newborn infants.
J Pediatr.
85
1974
534
542
142
Siegel
SE
Moran
RG
Problems in the chemotherapy of cancer in the neonate.
Am J Pediatr Hematol Oncol.
3
1981
287
296
143
Milsap
RL
Jusko
WJ
Pharmacokinetics in the infant.
Environ Health Perspect.
102
1994
107
110
144
Bleyer
WA
Clinical pharmacology of intrathecal methotrexate, II: an improved dosage regimen derived from age-related pharmacokinetics.
Cancer Treat Rep.
61
1977
1419
1425
145
McLeod
HL
Relling
MV
Crom
WR
et al
Disposition of antineoplastic agents in the very young child.
Br J Cancer Suppl.
66
1992
S23
S29
146
Allen
JC
The effects of cancer therapy on the nervous system.
J Pediatr.
93
1978
903
909
147
Woods
WG
O'Leary
M
Nesbit
ME
Life-threatening neuropathy and hepatotoxicity in infants during induction therapy for acute lymphoblastic leukemia.
J Pediatr.
98
1981
642
645
148
Pui
CH
Evans
WE
Acute lymphoblastic leukemia.
N Engl J Med.
339
1998
605
615
149
Ishii
E
Okamura
J
Tsuchida
M
et al
Infant leukemia in Japan: clinical and biological analysis of 48 cases.
Med Pediatr Oncol.
19
1991
28
32
150
Reaman
GH
Steinherz
PG
Gaynon
PS
et al
Improved survival of infants less than 1 year of age with acute lymphoblastic leukemia treated with intensive multiagent chemotherapy.
Cancer Treat Rep.
71
1987
1033
1038
151
Lauer
SJ
Camitta
BM
Leventhal
BG
et al
Intensive alternating drug pairs after remission induction for treatment of infants with acute lymphoblastic leukemia: a Pediatric Oncology Group pilot study.
J Pediatr Hematol Oncol.
20
1998
229
233
152
Frankel
LS
Ochs
J
Shuster
JJ
et al
Therapeutic trial for infant acute lymphoblastic leukemia: the Pediatric Oncology Group experience (POG 8493).
J Pediatr Hematol Oncol.
19
1997
35
42
153
Dreyer
ZE
Steuber
CP
Bowman
WP
et al
High risk infant ALL and improved survival with intensive chemotherapy.
Proc Am Soc Clin Oncol.
17
1998
529a
154
Rivera
GK
Raimondi
SC
Hancock
ML
et al
Improved outcome in childhood acute lymphoblastic leukaemia with reinforced early treatment and rotational combination chemotherapy.
Lancet.
337
1991
61
66
155
Kaleita
TA
Reaman
GH
MacLean
WE
Sather
HN
Whitt
JK
Neurodevelopmental outcome of infants with acute lymphoblastic leukemia: a Children's Cancer Group report.
Cancer.
85
1999
1859
1865
156
Pui
CH
Evans
WE
Acute lymphoblastic leukemia in infants.
J Clin Oncol.
17
1999
438
440
157
Grier
HE
Gelber
RD
Camitta
BM
et al
Prognostic factors in childhood acute myelogenous leukemia.
J Clin Oncol.
5
1987
1026
1032
158
Amadori
S
Ceci
A
Comelli
A
et al
Treatment of acute myelogenous leukemia in children: results of the Italian Cooperative Study AIEOP/LAM 8204.
J Clin Oncol.
5
1987
1356
1363
159
Pui
CH
Kalwinsky
DK
Schell
MJ
Mason
CA
Mirro
J
Jr
Dahl
GV
Acute nonlymphoblastic leukemia in infants: clinical presentation and outcome.
J Clin Oncol.
6
1988
1008
1013
160
Buckley
JD
Chard
RL
Baehner
RL
et al
Improvement in outcome for children with acute nonlymphocytic leukemia: a report from the Childrens Cancer Study Group.
Cancer.
63
1989
1457
1465
161
Ravindranath
Y
Steuber
CP
Krischer
J
et al
High-dose cytarabine for intensification of early therapy of childhood acute myeloid leukemia: a Pediatric Oncology Group study.
J Clin Oncol.
9
1991
572
580
162
Ravindranath
Y
Yeager
AM
Chang
MN
et al
Autologous bone marrow transplantation versus intensive consolidation chemotherapy for acute myeloid leukemia in childhood.
N Engl J Med.
334
1996
1428
1434
163
Béhar
C
Suciu
S
Benoit
Y
et al
Mitoxantrone-containing regimen for treatment of childhood acute leukemia (AML) and analysis of prognostic factors: results of the EORTC Children Leukemia Cooperative Study 58872.
Med Pediatr Oncol.
26
1996
173
179
164
Woods
WG
Kobrinsky
N
Buckley
JD
et al
Timed-sequential induction therapy improves postremission outcome in acute myeloid leukemia: a report from the Children's Cancer Group.
Blood.
87
1996
4979
4989
165
Stevens
RF
Hann
IM
Wheatley
K
Gray
RG
Marked improvements in outcome with chemotherapy alone in paediatric acute myeloid leukaemia: results of the United Kingdom Medical Research Council's 10th AML trial.
Br J Haematol.
101
1998
130
140
166
Creutzig
U
Zimmermann
M
Ritter
J
et al
Definition of a standard-risk group in children with AML.
Br J Haematol.
104
1999
630
639
167
Wells
RJ
Woods
WG
Buckley
JD
et al
Treatment of newly diagnosed children and adolescents with acute myeloid leukemia: a Childrens Cancer Group study.
J Clin Oncol.
12
1994
2367
2377
168
Odom
LF
Gordon
EM
Acute monoblastic leukemia in infancy and early childhood: successful treatment with an epipodophyllotoxin.
Blood.
64
1984
875
882
169
Nishikawa
A
Nakamura
Y
Nobori
U
et al
Acute monocytic leukemia in children: response to VP-16-213 as a single agent.
Cancer.
60
1987
2146
2149
170
Carroll
A
Civin
C
Schneider
N
et al
The t(1;22) (p13;q13) is nonrandom and restricted to infants with acute megakaryoblastic leukemia: a Pediatric Oncology Group study.
Blood.
78
1991
748
752
171
Lion
T
Haas
OA
Harbott
J
et al
The translocation t(1;22) (p13;q13) is a nonrandom marker specifically associated with acute megakaryocytic leukemia in young children.
Blood.
79
1992
3325
3330
172
Johnson
FL
Sanders
JE
Ruggiero
M
Chard
RL
Jr
Thomas
ED
Bone marrow transplantation for the treatment of acute nonlymphoblastic leukemia in children aged less than 2 years.
Blood.
71
1988
1277
1280
173
Emminger
W
Emminger-Schmidmeier
W
Haas
OA
et al
Treatment of infant leukemia with busulfan, cyclophosphamide ± etoposide and bone marrow transplantation.
Bone Marrow Transplant.
9
1992
313
318
Sign in via your Institution