ACUTE LYMPHOBLASTIC LEUKEMIA (ALL) is the most prevalent type of cancer, as well as the most common form of leukemia in children.1 This lymphoid malignancy, manifested by the proliferation of lymphopoietic blast cells, represents a heterogeneous group of diseases that vary with respect to morphological, cytogenetic, and immunologic features of the transformed cells. Technical improvements in immunofluorescence staining and flow cytometry together with the availability of numerous monoclonal antibodies (MoAbs) that recognize lineage-associated membrane molecules have illuminated the immunophenotypic heterogeneity in ALL. We now know that leukemia cells from patients with ALL may express various combinations of surface antigens that are found normally on lymphocyte precursors at discrete stages of maturation.2,3 Thus, the malignant clones in patients with ALL are thought to originate from normal lymphoid progenitor cells arrested at early stages of B- or T-lymphocyte ontogeny. Although cells from the majority (≈85%) of pediatric patients express B-lineage–associated antigens, those from approximately 15% of patients express the T-lineage–associated antigens CD1, CD2, CD3, CD4, CD5, CD7, or CD8.4-6 T-lineage ALL in children is associated with numerous unfavorable presenting features, thus it is not surprising that children with T-lineage ALL frequently have been reported to have a worse prognosis than children with B-lineage ALL.4,5,7-10 However, a number of encouraging reports from recent clinical studies using contemporary risk-adjusted multiagent chemotherapy programs have documented remarkably improved outcomes for patients with T-lineage ALL.6,10-14 Moreover, advanced preclinical studies have triggered much optimism that new agent discovery programs may lead to further improvements in outcome in the near future. In this review, we discuss current concepts regarding the etiology, biological characteristics, clinical features, and treatment of pediatric T-lineage ALL.

The role of numerous epidemiological factors, including maternal and paternal exposure to radiation, history of maternal fetal loss or fertility problems, higher birthweight at diagnosis, and use of exogenous growth hormone, remains controversial in the cause of pediatric ALL.15-17 A recent comprehensive review found no relationship between exposure to electromagnetic field (EMF) radiation and incidence of childhood ALL.18 The reported space-time clustering of ALL cases, which might suggest an etiologic agent such as a virus, is also controversial.19-23 Human T-cell leukemia virus-I and II may be associated with adult, but not pediatric T-lineage leukemia or lymphoma,24,25 and Epstein-Barr virus infection has been linked to a limited number of cases of T-cell lymphoma, but not T-lineage ALL, in children.26 

The autosomal recessive disorder ataxia telangiectasia (AT) appears to be a true etiologic factor because patients with AT have an increased risk of developing lymphoid malignancies, including T-lineage ALL.27 Translocations involving the T-cell receptor (TCR) loci are reported in approximately 10% of the T cells from patients with AT,28 but interestingly, the most frequent of these translocations appear to involve different regions within the TCR loci compared with those observed in patients with T-lineage ALL without AT.29-31 The molecular basis for these effects as well as other genetic abnormalities that may play a role in T-lineage leukemia will be discussed below. Taken together, these data suggest that multiple factors may be involved in the origin of T-lineage ALL.

Because leukemic cells are thought to originate from normal T-lymphocyte precursors arrested at early stages of ontogeny,2,32 every pathway that ensures homeostasis of a functional immune system is a potential target for disruption. Still, the fundamental issue of how many different mutations are required for malignant transformation to the leukemic state remains to be delineated. Nevertheless, clear associations have been identified between the occurrence of nonrandom translocations or other gene mutations and the development of T-lineage ALL. Below, we describe the specific molecular defects found in T-lineage leukemias and discuss altered signal transduction pathways that may contribute to the malignancy.

Chromosomal translocations.

An array of nonrandom translocations that are specific to T-lineage ALL have been identified; all appear to occur preferentially in the TCR loci on chromosomes 14 and 7.33 The breakpoints in many cases resemble TCR recombination signals, implying that the aberration arose during TCR rearrangement.34-39 Translocations involving chromosomes 1 and 14, such as t(1;14)(p33;q11) and t(1;14)(p32;q11), have been estimated to occur in approximately 3% of T-lineage ALL cases.40 In such rearrangements, the SCL/TCL5/TAL-1 gene from chromosome 1 and the TCRδ gene on chromosome 1436,41,42 are juxtaposed, resulting in deregulation of normal TAL-1 expression.41,43 TAL-1 was predicted to encode a protein containing a helix-loop-helix DNA binding motif,42,43 suggesting that the t(1;14) translocations could contribute to leukemogenesis by inducing aberrant expression of novel or TAL-1–regulated genes.

A distinct TAL-1 disruption occurs via an interstitial deletion between a locus called SIL (SCL interrupting locus) and the 5′UTR of SCL, resulting in a fusion transcript SIL/SCL, and is estimated to occur with a frequency of 16% to 26% in T-lineage ALL.44-46Presence of a TAL-1 disruption was correlated with high white blood cell (WBC) count, high hemoglobin level, and CD2+/CD10 immunophenotypes, and interestingly, 4-year event-free survival (EFS) was higher for patients with TAL-1 disruption compared to those without TAL-1 alterations (59% ± 11% v 44% ± 7%, respectively), although this difference did not reach conventional significance.46Although TAL-1 is required for development of all hematopoietic lineages in mice,47 the gene is not expressed in B- or T-lineage cells,41,48 and interestingly, SCL-transfected, v-ABL–transformed cells appear to be oncogenic in mice.49Taken together, these data suggest that disruption of normal TAL-1 expression may contribute to the transformation of T-cell precursors into leukemic blasts.

The t(10;14)(q24;q11) translocation, first identified in T-cell neoplasms including T-lineage ALL, involves the TCRα/TCRδ locus on chromosome 1434,35,50 and the TCL3 locus on chromosome 10.34,35 An open reading frame within TCL3 encodes a novel homeobox protein, HOX-11, whose expression is deregulated as a result of the translocation.51-53 Moreover, like TAL-1, HOX-11 is capable of DNA binding and transcriptional activation of reporter genes, suggesting a role for this gene in leukemic transformation.54 Additional studies showed that whereas HOX-11 was expressed in leukemic cell lines and leukemic blasts, it was not expressed in normal T lymphocytes,52,53,55 but was required for normal spleen development.56 Reverse transcriptase-polymerase chain reaction (RT-PCR) assays have suggested that HOX-11 alterations may occur with high frequency in patients with T-lineage ALL.57 Thus, deregulation of HOX-11 is likely to be a biologically significant factor in development of T-lineage ALL.

Translocations t(11;14)(p13;q11) and t(11;14)(p15;q11) also are observed frequently in T-lineage ALL58-60; both involve breakpoints within diversity or J segments the region the TCRα or TCRδ genes on chromosome 14.37,61,62 McGuire et al61 described multiple open reading frames near the chromosome 11 breakpoints and identified one at 11p15 as the open reading frame of the TTG-1 gene. Similarly, Boehm et al63 identified the involved region of 11p15 as the rhombotin gene. Both genes encode proteins characterized by duplicate cysteine-rich zinc-finger protein binding homology domains.61,63 A related gene, rhombotin-2/TTG-2, was shown to be deregulated in cases involving 11p13.63,64 Consistent with the predicted structure of the rhombotins, a recent report described the identification of an ets family transcription factor, ELF-2, that contains rhombotin-2 binding domains, suggesting a transcriptional regulatory role for rhombotin-2.65Clinically, several investigators have associated t(11;14) translocations with an immature stage of thymocyte development,37,59,60 but the overall prognostic significance of this translocation remains unclear.

Although translocations involving chromosome 7 occur in both B-precursor and T-lineage ALL, those involving the TCR-β locus at 7q32-36 are specific for T-lineage ALL.66 One such translocation, t(7;19), truncates the lyl-1 gene on chromosome 19,67 presumably resulting in altered DNA-binding ability for lyl-1.68 Another case, t(7;9), results in truncation of the TAN-1 gene on chromosome 9.69 The mouse homologue of TAN-1 is expressed ubiquitously, but is most abundant in lymphoid tissues, suggesting that normal expression of TAN-1 is disrupted in t(7;9)+ ALL.69 

The distinct translocation t(1;7)(p34;q34) was shown to juxtapose the TCR-β constant region enhancer upstream of the LCK gene, which encodes an SRC family protein tyrosine kinase that is involved in signal transduction through CD4.70,71 Notably, overexpression of LCK in transgenic mice causes thymomas or both thymomas and peripheral lymphoid malignancies,72,73suggesting a role for deregulated LCK expression in leukemogenesis. The c-myc locus on chromosome 8 defines yet another class of translocations associated with T-lineage ALL. In t(8;14)(q24;q11), c-myc is translocated with the TCRα loci on chromosome 14, resulting in deregulation of myc expression.74,75 In t(2;8), a fusion protein is produced that consists of c-myc and the product of an unidentified locus on chromosome 2.76 The frequency and significance of these translocations are unclear at present.

We have recently determined the frequency and clinical significance of chromosomal abnormalities in a large cohort of patients with T-lineage ALL enrolled on contemporary CCG studies (Heerema N., et al, submitted for publication). Translocations involving 14q11 and 7q32-q36 were among the most frequent abnormalities, but non-TCR loci, including 9p, 6q, 11q23, and 14q32, also were frequently altered. Notably, none of these abnormalities had prognostic significance in the context of the intensive therapies used in contemporary CCG studies. Nevertheless, the array of chromosomal rearrangements described above are a hallmark of the biological diversity of T-lineage ALL and are likely to result from alterations in underlying cellular control mechanisms. Indeed, recent advances in our understanding of cell signaling and cell cycle control suggest that defective cell surveillance mechanisms are likely to be the major factors leading both to unrestrained proliferation of leukemic cells and to the development of chromosomal abnormalities, including translocations, pseudodiploidy, and hyperdiploidy, that are associated with leukemic cells.77-80 Alterations in such control mechanisms are discussed below.

Mutation or loss of cell cycle control genes.

Mutations present in malignant cells allow them to circumnavigate regulators that control proliferation and differentiation. The retinoblastoma (Rb) gene was originally identified as a tumor suppressor gene because of its inactivation in cases of retinoblastoma; prostate, breast, and lung cancers; and leukemias.81Notably, the telomeric Rb1 gene is located on the long arm of chromosome 13 (13q14), which is inactivated or deleted in approximately 6% of T-lineage ALL cases.82,83 

In addition to Rb, other proteins that affect cell cycle progression include the cyclin-dependent kinase inhibitors p21, p27, and p57, as well as the inhibitors of Cdk4 (Ink4): p15Ink4b, p16Ink4a, p18Ink4c, and p19Ink4d.84-89 Among the Ink4 family of inhibitors, p15Ink4b and p16Ink4a have been implicated for a role in the biology of T-lineage ALL.90-95Both genes map to 9p21, a region on the short arm of chromosome 9 previously shown to be deleted frequently in T-lineage ALL.33,96-98 In addition, Batova et al95recently reported that the 5′ promoter region of the p15 gene is preferentially hypermethylated, presumably resulting in loss of transcriptional expression in 38% of newly diagnosed T-lineage ALL.

Another critical regulator of cell cycle progression, the p53 gene, is the most frequently mutated gene in human cancers.81 The major function of p53 is to ensure that cells arrest and attempt to repair genotoxic damage before replicating DNA and entering mitosis.99 In p53-deficient mice, the most common tumor that arises is a T-lineage lymphoid malignancy.100 Although p53 mutations are infrequently observed at diagnosis, they are associated with relapse in pediatric T-lineage ALL.101,102 

Another sensor for cell damage appears to be the ATM gene product, which is mutated in patients with AT.103 After insult with agents that induce sublethal DNA damage, cells from patients with AT fail to block DNA synthesis and thereby fail to repair the damaged DNA.104 These effects are apparently caused by a failure of the mutated ATM gene to regulate p53.105 ATM-deficient mice develop an aggressive form of T-lineage leukemia/lymphoma,106,107 and, as described above, children with AT frequently develop T-lineage ALL,27,108,109implicating ATM in leukemogenesis.

Other genes implicated in the malignant transformation of leukemic cells are Ets-1 and IKAROS. The Ets-1 T-lymphocyte transcription factor is thought to be important for normal thymic development and for prevention of cell death in normal mature T cells. A mutation in the DNA binding domain of the Ets-1 was reported in a case of T-lineage ALL,110 but the clinical significance of this finding remains to be proven. The IKAROS gene encodes a zinc finger DNA binding protein that is required for lymphoid cell differentiation.111 Heterozygous transgenic mice harboring a defective IKAROS gene develop a very aggressive form of T-cell leukemia, suggesting that IKAROS may serve as a suppressor of leukemic transformation.112 

Leukemic cells also appear to be altered in their responses to various stimuli that induce apoptosis. Debatin et al113,114reported that primary leukemic cells and cell lines from adult patients with T-cell leukemia were sensitive to FasL-induced cell killing in vitro, whereas leukemic cells from pediatric patients with T-lineage ALL were resistant. Resistance was unrelated to the quantity of Fas on the cell surface, but was reversed by treatment with the protein synthesis inhibitor cycloheximide, suggesting that short-lived proteins were required for maintenance of the resistant phenotype. In vivo treatment of a human T-lineage ALL-engrafted severe combined immunodeficiency (SCID) mouse with an anti-Fas antibody resulted in prolonged survival, but did not eradicate the disease, supporting the existence of Fas sensitive and insensitive leukemic cells.115 These data suggest that altered responses to apoptotic stimuli or regulatory factors may contribute to the ability of leukemic cells to escape killing by either immune surveillance or cytotoxic agents.

Bcl-2, which protects cells from non–Fas-mediated apoptosis,116,117 is expressed in both T-lineage and B-lineage leukemias, but it is not yet known how this affects their ability to survive cytotoxic treatments. A related protein, Bax,118 acts as an antagonist to Bcl-2 and may confer radiation sensitivity to cells.119 In a recent CCG study, we found a marked variation in Bcl-2 expression by primary leukemic cells from 238 children with newly diagnosed ALL, including 52 patients with T-lineage ALL.120 High-risk features, such as high WBC count, organomegaly, presence of MLL-AF4 or BCR-ABL fusion transcripts, or leukemic cell growth in SCID mice, were not associated with Bcl-2 expression in these patients. For patients with T-lineage ALL, high Bcl-2 expression was predictive of slow early response (ie, M3 day 14 marrow status). However, with limited follow-up and overall excellent outcome for patients, this correlation did not extend to EFS.

T-lineage ALL is distinct from B-lineage ALL not only biologically, but also clinically. Although the basis for these differences is not well understood, clinical characteristics have been useful prognostic factors for guiding the use of experimental treatments. Below, we describe common presenting features, prognostic variables, and treatment outcome of patients with T-lineage ALL based on data accumulated over the last decade. We then focus on causes for treatment failure and discuss new strategies for improving outcome among subgroups of patients who remain at risk for relapse despite intensive therapy.

Presenting features.

The relationship between T-lineage markers and unfavorable presenting characteristics was first noted by Borella, Sen, and others,121-124 and numerous studies have now confirmed that compared to patients with B-lineage ALL, those with T-lineage ALL more frequently show the highest WBC range (≥50,000/μL), are nonwhite, older, exhibit marked enlargement of the spleen, liver, and lymph nodes, and have a mediastinal mass.5,7,9,10,125 

Modal chromosome number is often abnormal among patients with ALL, with hyperdiploidy (>50 chromosomes) correlated with favorable outcome and pseudodiploidy associated with poor outcome.58,98,126-129 The hyperdiploid karyotype is more often associated with pre-B or early pre-B immunophenotypes,129 whereas the pseudodiploid karyotype is more often associated with the T-lineage immunophenotype.33Also, “near tetraploid” chromosome number (>65) is more often associated with T-lineage ALL and poor outcome.130 As described above, nonrandom translocations in T-lineage ALL preferentially occur in the TCR loci on chromosomes 7 and 14,33 and those involving the TCRβ locus at 7q32-36 and the TCRαδ 14q11 collectively occur in approximately 20% of all T-lineage ALL cases.33 

Risk classification of T-lineage ALL.

In general, treatment protocols for childhood leukemias have relied on the known prognostic factors of age and WBC count, as well as organomegaly rather than immunophenotype for risk assessment. As a result, even though many patients with T-lineage ALL were previously misclassified or not immunophenotyped, they were likely to receive treatment for high-risk ALL based on their other presenting features. In contemporary trials, various groups have used somewhat different criteria for classification, which has complicated comparisons of results between groups, but nevertheless has generally resulted in similar assignment of patients with T-lineage ALL to more intensive treatment protocols, such as Berlin-Frankfurt-Munster (BFM),131,132 modified BFM,12 and the New York (NY) regimen,13 as well as those of the St Jude Children's Research Hospital133 and Dana Farber Cancer Institute.11 

From 1983 through 1993, children daignosed with ALL who exhibited National Cancer Institute (NCI) standard risk features134were classified by the CCG as either low risk (ages 2 through 9 years and WBC <10,000/μL) or intermediate risk (ages 2 through 9 years and WBC <10,000 to 49,999/μL, or age 1 year and WBC <50,000/μL), whereas patients exhibiting NCI poor-risk characteristics were classified as follows: high risk, ages 1 through 9 years with WBC ≥50,000/μL or age >10 years; infants, age <1 year; lymphomatous, patients with specific high-risk features, as described.135 As shown in Table1, patients with T-lineage ALL more frequently were assigned to the higher risk than to the lower risk protocols, which is consistent with their clinical features described above.

Table 1.

CCG and NCI Risk Group Classification of Children With B-Lineage and T-Lineage Acute Lymphoblastic Leukemia

Risk GroupB-Lineage ALL N = 3,668T-Lineage ALL N = 730
N(%)*N(%)-151
CCG-Low  705  (19.2)  58 (8.0)  
CCG-Intermediate  1,575  (42.9)  71  (9.7) 
CCG-High  1,059  (28.9)  169  (23.2) 
CCG-Lymphomatous  216  (5.9)  425  (58.2) 
CCG-Infant  113  (3.1)  7  (1.0)  
Total  3,668 (100.0)  730  (100.0)  
NCI-Standard  2,213  (60.3) 211  (28.9)  
NCI-Poor  1,455  (39.7)  519  (71.1) 
Total  3,668  (100.0)  730  (100.0) 
Risk GroupB-Lineage ALL N = 3,668T-Lineage ALL N = 730
N(%)*N(%)-151
CCG-Low  705  (19.2)  58 (8.0)  
CCG-Intermediate  1,575  (42.9)  71  (9.7) 
CCG-High  1,059  (28.9)  169  (23.2) 
CCG-Lymphomatous  216  (5.9)  425  (58.2) 
CCG-Infant  113  (3.1)  7  (1.0)  
Total  3,668 (100.0)  730  (100.0)  
NCI-Standard  2,213  (60.3) 211  (28.9)  
NCI-Poor  1,455  (39.7)  519  (71.1) 
Total  3,668  (100.0)  730  (100.0) 

*Percentage of patients with B-lineage ALL classified into each risk group.

F0-151

Percentage of patients with T-lineage ALL classified into each risk group.

Treatment outcome in T-lineage ALL.

As noted above, previous studies showed poorer outcomes for patients with T-lineage ALL compared with patients with B-lineage ALL. For example, in the BFM group, Henze et al136 reported poor outcome for patients with T-lineage ALL who were treated on DAL (adapted from St Jude protocol VII), with 9-year probabilities of continuous complete remission (CCR) of 9% ± 9% and 41% ± 5%, for T-lineage and non–T-lineage, respectively. In contrast, patients treated on BFM achieved CCR of 52% ± 13% and 65% ± 5%, respectively, suggesting that BFM provided superior treatment for T-lineage ALL.

Investigators of the Pediatric Oncology Group7 treated 53 patients with T-lineage ALL with a modified LSA2L2 regimen that had been shown to be efficacious for treatment of T-cell non-Hodgkin's lymphoma.

Although complete remission was achieved for 88% of the patients, the projected overall 3-year EFS was only 40% (SE = 8.3%). Moreover, for patients with WBC count <50,000, the projected 3-year EFS was 67%, whereas for patients with WBC count >50,000, 3-year EFS was only 19%. In a follow-up study, 253 children with T-lineage ALL treated by a modified LSA2L2 regimen together with cranial radiation therapy and triple intrathecal therapy for presymptomatic treatment of central nervous system (CNS) disease achieved an overall 4-year EFS of 43% (SE = 4%).8 Thus, although outcomes improved, the LSA2L2 regimen remained ineffective for the majority of patients with T-lineage ALL. Similarly, in an analysis of data from St Jude studies X and XI, conducted from 1979 to 1983, 120 children with T-lineage ALL had a 5-year EFS of 46% (SE = 18%).137 In a French trial, Garand et al10 treated 88 pediatric patients with T-lineage ALL by protocols such as BFM or FRALLE,138 and an EFS of approximately 58% was reported for a median follow-up of 30 months, suggesting that such therapy could improve outcome for these patients.

Although the studies described above generally found unfavorable outcomes for patients with T-lineage ALL, other recent studies have reported improved outcomes through the use of highly intensive treatment protocols. For example, using an intensive four-drug induction and multidrug continuation, including doxorubicin and prednisone together with prophylaxis for CNS disease and high-dose L-asparaginase, Clavell et al11 reported improved outcome (4-year EFS of 71%) for high-risk patients, including those who had T-lineage ALL. More recently, in a study by Schorin et al1420 patients with T-lineage ALL treated with multiagent chemotherapy together with cranial irradiation and intrathecal methotrexate for 2 years also had favorable outcomes (7-year EFS of 70%, SE = 10%).14 The favorable outcome was attributed to the inclusion of L-asparaginase and doxorubicin in the treatment regimen.

Studies by the CCG also have shown improvements in EFS outcome for high-risk patients with ALL including those with the T-lineage immunophenotype. Steinherz et al13 used an intensive multidrug chemotherapy (NY regimen) to treat 100 patients with characteristics previously correlated with a high risk for relapse. This patient population included 13 patients with T-lineage ALL (defined as E-rosette–+). Four-year EFS for the entire cohort was 69% (SE = 5%), whereas 4-year EFS for patients with T-lineage ALL was 75%. Gaynon et al139 used a modified BFM therapy involving four-drug induction and aggressive continuation therapy to treat high-risk children, including 60 who were E-rosette–+. Overall 3-year EFS was 65% (SD = 3.5%); patients with WBC count >50,000 who were E-rosette–+ had a 3-year EFS of 75% (SD = 6.9%), whereas those who were E-rosette–− had an EFS of 51% (SD = 6.3%).

To investigate the outcome of patients with T-lineage ALL on these regimens more thoroughly, we recently analyzed data from the large cohort of patients enrolled on CCG studies conducted between 1983 and 1993.134 Notably, we observed a significant improvement in outcome of patients with T-lineage ALL compared with those on earlier studies because of marked decreases in the incidences of induction failures, early bone marrow relapses, and CNS relapses when more aggressive therapy was given (Fig 1). The probability of 3-year survival for patients with T-lineage ALL increased from 56% in studies conducted between 1978 and 1983, to 65% in studies conducted between 1983 and 1989, and to 78.8% in studies conducted between 1989 and 1993 (Table 2).Taken together, these various studies suggest that current risk for patients with T-lineage ALL treated by intensive therapeutic regimens is similar to that of patients with B-lineage ALL. Thus, a major improvement in treatment of T-lineage ALL has been achieved.

Fig. 1.

Improved EFS of patients with T-lineage ALL in the context of contemporary intensive chemotherapy programs. EFS for the entire cohort of patients with T-lineage and B-lineage ALL treated on the 1800 series and 100 series of CCG studies are shown. EFS values at designated points in follow-up are given in the text.

Fig. 1.

Improved EFS of patients with T-lineage ALL in the context of contemporary intensive chemotherapy programs. EFS for the entire cohort of patients with T-lineage and B-lineage ALL treated on the 1800 series and 100 series of CCG studies are shown. EFS values at designated points in follow-up are given in the text.

Close modal
Table 2.

Outcome for Patients With T-Lineage ALL Treated During Three Consecutive CCG Treatment Eras

CCG Study EraYearsEvent-Free Survival (%)
3-Year5-Year
CCG-160s  1978-1983  56.4  52.5 
CCG-100s  1983-1989  65.8  61.0  
CCG-1800s 1989-1993  78.2  75.2 
CCG Study EraYearsEvent-Free Survival (%)
3-Year5-Year
CCG-160s  1978-1983  56.4  52.5 
CCG-100s  1983-1989  65.8  61.0  
CCG-1800s 1989-1993  78.2  75.2 
Prognostic factors in T-lineage ALL.

A number of risk factors for T-lineage ALL were identified in the studies described above. For example, Dowell et al9 and Shuster et al8 reported that compared with patients with T-lineage ALL whose leukemic cells were CD10, those whose cells were CD10+ were more likely to achieve remission and have significantly improved EFS outcomes. In another study, Pui et al137 reported that CD3 positivity in association with an abnormal karyotype was a significant adverse risk factor; 5-year EFS for patients with both of these characteristics was 35%. In contrast, Shuster et al8 found no prognostic significance for CD3 expression; rather, the most important favorable prognostic factors for patients with low WBC count or high WBC count at diagnosis were CD5 positivity or expression of the THY antigen, respectively.

The findings that many patients with T-lineage ALL now can achieve a much improved outcome has motivated attempts to identify subgroups of patients within T-lineage ALL that may exhibit improved or reduced probabilities of survival. Two previous CCG studies described above noted a favorable association between outcome and E-rosette (CD2) positivity among high-risk patients.13,139To determine comprehensively the clinical significance of CD2 expression in T-lineage ALL, we prospectively immunophenotyped leukemic cells from the large cohort of children enrolled on CCG studies between 1983 and 1993.140 We noted a statistically significant correlation (P = .0006) between the CD2 antigen expression frequency (ie, the average percentage of blasts that were positive for CD2) and EFS. Compared with patients with the highest CD2 expression level, patients with intermediate and low CD2 expression frequencies had relative hazard rates (RHR) of 1.27 and 2.01, indicating an increased risk of treatment failure. After 6 years of follow-up, the EFS estimates for the three CD2 expression groups (low expression frequency to high expression frequency) were 49.3%, 63.5%, and 72.2%, respectively. CD2 expression remained a significant predictor of EFS after adjustment for the effects of other covariates by multivariate regression. Expression of other antigens (CD3, CD5, CD10, or CD34) by leukemic cells was not correlated with EFS. Thus, the expression frequency of CD2 antigen is a powerful predictor of EFS that may be useful for risk classification or assignment to novel therapies aimed at improving patient outcome.

Maturation stage of the predominant leukemic clones also has been suggested as a means for subgrouping patients with T-lineage ALL. Crist et al4 stratified 101 patients with T-lineage ALL into three maturation groups according to expression of T-lineage cell surface antigens, as follows: stage I, CD2+CD7+; stage II, CD2+CD7+CD1+CD4+CD8+; and stage III, CD2+CD7+CD1(CD4+ or CD8+)CD3+. Although the percentage of patients achieving remission following induction therapy was lower for patients with T-lineage ALL of the earliest maturation stage (79%, 100%, and 94% for stages I, II, and III, respectively), 4-year EFS was equally poor for all three groups (33%, 32%, and 38%, respectively).

Recently, we analyzed data from a large cohort of patients with T-lineage ALL treated on contemporary protocols of the CCG to further investigate the prognostic role of the apparent maturation stage of leukemic T-cell precursors.141 Patients were immunophenotypically classified as follows: pro-thymocyte leukemia (pro-TL), CD7+CD2CD5; immature TL, CD7+(CD2+ or CD5+)CD3; and mature TL, CD7+CD2+CD5+CD3+. No group had a preponderance of favorable or unfavorable presenting characteristics. Four-year EFS was lower for patients with pro-TL (57.1%; SD = 8.4%) compared with patients with immature and mature TL (68.5%, SD = 3.5%; and 77.1%, SD = 4.0%; respectively) with an overall significance of P = .05. Highly significant differences were found for overall survival (P = .005) as a result of the deaths of all patients with pro-TL who relapsed. Although CD2 also was a significant prognostic factor (P = .03), RHRs of 2.11, 1.51, and 1.17 for patients with pro-TL, CD2immature TL, and CD2+ immature TL, respectively, suggested that the pro-TL maturation stage had added prognostic significance (Fig2). Indeed, multivariate analysis indicated that the influence of ontogeny group was greater than that of CD2. Thus, leukemic cells of the pro-TL maturation stage identified a subgroup of patients with T-lineage ALL who have a significantly worse EFS outcome than patients whose leukemic cells correspond to a more mature stage of development.

Fig. 2.

EFS of patients with T-lineage ALL according to the apparent maturational stage of bone marrow leukemic blasts. EFS for (A) mature TL, (B) CD2+ immature TL, (C) CD2immature TL, and (D) pro-TL patients treated on the 1800 series of CCG protocols are shown. EFS values at designated points in follow-up are given in the text.

Fig. 2.

EFS of patients with T-lineage ALL according to the apparent maturational stage of bone marrow leukemic blasts. EFS for (A) mature TL, (B) CD2+ immature TL, (C) CD2immature TL, and (D) pro-TL patients treated on the 1800 series of CCG protocols are shown. EFS values at designated points in follow-up are given in the text.

Close modal

The variant immunophenotype in which leukemic cells coexpress T-lineage– and myeloid–associated antigens represents a controversial prognostic factor. Although numerous investigators have reported that coexpression of myeloid antigens predicted an adverse risk for patients with T-lineage ALL,142,143 others have found similar outcomes for myeloid antigen negative (My) and myeloid antigen positive (My+) T-lineage ALL.144,145 We recently evaluated the influence of myeloid antigen expression on treatment outcome in a large cohort of children with newly diagnosed ALL enrolled on risk-adjusted CCG studies.146 Patients were classified as Myor My+ T-lineage, according to expression of CD7, CD13, and CD33. Patients with My+ T-lineage ALL were more likely than patients with My T-lineage ALL to show favorable presenting features, but induction outcome and EFS outcome were similar for patients with My+ and My T-lineage ALL, with 4-year EFS of 72.7% (SD = 7.1%) and 70.1% (SD = 5.7%), respectively (P = .49; Fig 3).These results show that regardless of treatment intensity, mixed myeloid-lymphoid phenotype was not an adverse prognostic factor for childhood T-lineage ALL.

Fig. 3.

Myeloid antigen expression in T-lineage ALL is not associated with poor EFS. EFS for My+ TL and MY TL patients treated on the 1800 series of CCG protocols are shown. EFS values at designated points in follow-up are given in the text.

Fig. 3.

Myeloid antigen expression in T-lineage ALL is not associated with poor EFS. EFS for My+ TL and MY TL patients treated on the 1800 series of CCG protocols are shown. EFS values at designated points in follow-up are given in the text.

Close modal
Drug resistance.

Despite improvements in overall survival, relapse in the bone marrow, CNS, and other sites remains a significant problem for high-risk patients. Pieters et al147 showed that patients with T-lineage ALL were particularly resistant to prednisone (PRED), daunorubicin, cytarabine, mafosfamide, and L-asparaginase, but wide ranges of resistance levels were observed within each immunophenotypic group. For all patients, the probability of continuous complete remission decreased with increasing resistance to PRED. In a later study, these investigators reported that patients with T-lineage ALL were more resistant to a host of drugs including those mentioned above as well as teniposide, ifosfamide, vincristine, vindesine, and dexamethasone.148 Lauer et al149 found that a regimen of intensive rotating drug pairs was effective for prevention of drug resistance in high-risk patients with B-lineage, but not T-lineage ALL, again suggesting that immunophenotype plays a role in drug sensitivity. Others have attributed methotrexate (MTX) resistance in patients with T-lineage ALL to a decreased formation of MTX-polyglutamates, which is a determinant of toxicity.150,151 Resistance to glucocorticoids is thought to be caused by low glucocorticoid receptor (GR) levels. However, the relationship between GR and outcome within the T-lineage immunophenotype is unclear. Quddus et al152 reported that leukemic cell GR level did not predict outcome within the T-lineage group, whereas Costlow et al153 reported that lower GR levels were correlated with unfavorable presenting features including T-lineage. Finally, although multidrug resistance is thought to be mediated by overexpression of P-glycoprotein, the product of the multidrug resistance gene MDR-1,154 the specific significance of this phenomenon in T-lineage ALL has not been determined.

Current strategies for improving treatment of children with ALL have been aimed at maximizing efficacy of treatment according to risk. Reliable and accurate methods for predicting prognosis are required to achieve adequate treatment with the least intensive regimens. Identification of biological and clinical prognostic factors, as discussed above, has aided in stratifying patients according to risk. However, additional methods are required for identifying and more effectively treating subgroups of high-risk patients who are most likely to relapse despite intensive therapy.

Seventy-five percent of children with T-lineage ALL on CCG protocols fit within the NCI high-risk category based on presenting age and WBC count.134 Patients with T-lineage ALL with standard risk represent less than 4% of patients with ALL and less than 6% of all standard-risk patients. Treatment of patients who have relapsed generally has consisted of intensive chemotherapy to achieve a second remission and subsequent use of either nonablative chemotherapy or ablative radiochemotherapy followed by bone marrow transplantation (BMT), and recurrence of leukemia is the major obstacle to the success of either approach. Intensification of cytotoxic therapy using conventional drugs will likely cause overlapping toxicities and may result in delays which may erode the intensity of therapy. Overall, the outcome for patients with relapsed T-lineage ALL is dismal because only a very small fraction can be saved with high-dose radiochemotherapy followed by BMT. Consequently, the development of new potent antileukemia drugs and the design of combinative treatment protocols using these new agents have emerged as exceptional focal points for research in modern therapy of relapsed T-lineage ALL.

Immunotoxins and other targeted biotherapeutics.

Immunotoxins (MoAb-toxin conjugates) are a new class of immunopharmacologic agents that shows considerable promise for more effective treatment of T-lineage ALL. A vast number of MoAbs have been developed with the intent of specifically targeting cytotoxic agents to leukemia cells while limiting the deleterious effects on normal tissues. Immunoconjugates containing toxins such as pokeweed antiviral protein, ricin, Pseudomonas endotoxin, and diphtheria toxin directed against T-lineage–specific surface antigens have been developed for use as systemic therapy of T-lineage ALL.155-158 

Murphy et al159 as well as Kreitman et al160 have pioneered the use of genetic engineering to redirect the lethal action of diphtheria toxin towards effective targeting of growth factor receptors on leukemic cells. In one example, researchers have developed a recombinant fusion toxin, DAB486IL-2, in which the native receptor binding domain of diphtheria toxin has been replaced with interleukin-2.161 

Deoxyguanosine analogs.

Another new and promising treatment program for T-lineage ALL is based on the potent antileukemia activity of deoxyguanosine analogs. The accumulation and the resulting toxicity of dGTP in T lymphocytes was first described in patients with a genetic deficiency for the enzyme purine nucleoside phophorylase (PNP).162,163 This observation lead to the search for means by which cytotoxic levels of dGTP could be achieved in T-lineage leukemias. An analog of deoxyguanosine, Ara-G (9-β-D-arabinofuranosylguanine) accumulates in T cells and acts as a poor substrate for endogenous PNP, but is efficiently phosphorylated by deoxycytidine kinase164,165; in vitro studies have shown that Ara-G is selectively cytotoxic for T-cell lines and T-lineage leukemic cells.166,167 

Recently, a water soluble pro-drug derivative of Ara-G, known as compound 506U/C-506 (2-amino-6-methoxypurine arabinoside), was developed for in vivo therapeutic applications.168Preliminary results of a Phase I trial of C-506 in adult T-cell malignancies suggested that daily infusion of C-506 could achieve and maintain cytotoxic levels of Ara-GTP.169 These data indicate that C-506 warrants investigation as a new therapeutic drug for treatment of pediatric T-lineage ALL.

The adverse risk previously associated with T-lineage ALL in children has progressively been surmounted by intensive chemotherapeutic regimens. Still, approximately 20% to 25% of children with T-lineage ALL continue to fail therapy. Further augmentation of the currently used intensive chemotherapeutic regimens may not be warranted because of the likelihood of significant adverse effects. Thus, the current challenge is to apply our expanding knowledge of biological regulation in leukemic cells to the development of novel biologic therapeutics, particularly those that specifically target leukemic cells. Such agents could theoretically be used either to trigger cell killing directly or to alter the leukemic cell's response to radiation or chemotherapeutics. Finally, the identification of prognostically distinct patient subgroups may lead to tailored and risk-adjusted therapies for children with T-lineage ALL. Use of these various strategies, singly and in combination, should allow further improvements in outcome for patients with ALL who remain at risk for treatment failure.

Supported in part by research grants including CCG Chairman's Grants No. CA-13539, CA-51425, CA-42633, CA-42111, CA-60437, and CA-27137 from the National Cancer Institute, National Institutes of Health. F.M.U. is a Stohlman Scholar of the Leukemia Society of America, New York, NY.

Address reprint requests to Fatih M. Uckun, MD, PhD, Children's Cancer Group ALL Biology Reference Laboratory and Wayne Hughes Institute, 2665 Long Lake Rd, St Paul, MN 55113.

1
Poplack D: Acute lymphoblastic leukemia, in Pizzo P, Poplack D (eds): Principles and Practice of Pediatric Oncology (ed 2). Philadelphia, PA, Lippincott, 1993, p 431
2
Greaves
MF
Differentiation-linked leukemogenesis in lymphocytes.
Science
234
1986
697
3
Smith
LJ
Curtis
JE
Messner
HA
Senn
JS
Furthmayr
H
McCulloch
EA
Lineage infidelity in acute leukemia.
Blood
61
1983
1138
4
Crist
WM
Shuster
JJ
Falletta
J
Pullen
DJ
Berard
CW
Vietti
TJ
Alvarado
CS
Roper
MA
Prasthofer
E
Grossi
CE
Clinical features and outcome in childhood T-cell leukemia-lymphoma according to stage of thymocyte differentiation: A Pediatric Oncology Group study.
Blood
72
1988
1891
5
Borowitz
MJ
Dowell
BL
Boyett
JM
Pullen
DJ
Crist
WM
Quddus
FM
Falletta
JM
Metzgar
RS
Clinicopathologic aspects of E rosette negative T cell acute lymphocytic leukemia: A Pediatric Oncology Group study.
J Clin Oncol
4
1986
170
6
Uckun
F
Reaman
G
Steinherz
P
Arthur
D
Sather
H
Trigg
M
Tubergen
D
Gaynon
P
Improved outcome for children with T-lineage acute lymphoblastic leukemia after contemporary chemotherapy: A children's cancer group study.
Leuk Lymphoma
24
1996
57
7
Pullen
DJ
Sullivan
MP
Falletta
JM
Boyett
JM
Humphrey
GB
Starling
KA
Land
VJ
Dyment
PG
Vats
T
Duncan
MH
Modified LSA2-L2 treatment in 53 children with E-rosette-positive T-cell leukemia: Results and prognostic factors (a Pediatric Oncology Group study).
Blood
60
1982
1159
8
Shuster
JJ
Falletta
JM
Pullen
DJ
Crist
WM
Humphrey
GB
Dowell
BL
Wharam
MD
Borowitz
M
Prognostic factors in childhood T-cell acute lymphoblastic leukemia: A Pediatric Oncology Group study.
Blood
75
1990
166
9
Dowell
BL
Borowitz
MJ
Boyett
JM
Pullen
DJ
Crist
WM
Quddus
FF
Russell
EC
Falletta
JM
Metzgar
RS
Immunologic and clinicopathologic features of common acute lymphoblastic leukemia antigen-positive childhood T-cell leukemia. A Pediatric Oncology Group study.
Cancer
59
1987
2020
10
Garand
R
Vannier
JP
Bene
MC
Faure
G
Favre
M
Bernard
A
Comparison of outcome, clinical, laboratory, and immunological features in 164 children and adults with T-ALL. the Groupe D'Etude Immunologique Des Leucemies.
Leukemia
4
1990
739
11
Clavell
LA
Gelber
RD
Cohen
HJ
Hitchcock Bryan
S
Cassady
JR
Tarbell
NJ
Blattner
SR
Tantravahi
R
Leavitt
P
Sallan
SE
Four-agent induction and intensive asparaginase therapy for treatment of childhood acute lymphoblastic leukemia.
N Engl J Med
315
1986
657
12
Gaynon
P
Steinherz
P
Bleyer
WA
Ablin
A
Albo
V
Finkelstein
J
Grossman
N
Littman
P
Novak
L
Pyesmany
A
Reaman
G
Sather
H
Hammond
D
Intensive therapy for children with acute lymphoblastic leukemia and unfavorable presenting features.
Lancet
2
1988
921
13
Steinherz
PG
Gaynon
P
Miller
DR
Reaman
G
Bleyer
A
Finklestein
J
Evans
RG
Meyers
P
Steinherz
LJ
Sather
H
Hammond
D
Improved disease-free survival of children with acute lymphoblastic leukemia at high risk for early relapse with the New York regimen—A new intensive therapy protocol: A report from the Children's Cancer Study Group.
J Clin Oncol
4
1986
744
14
Schorin
MA
Blattner
S
Gelber
RD
Tarbell
NJ
Donnelly
M
Dalton
V
Cohen
HJ
Sallan
SE
Treatment of childhood acute lymphoblastic leukemia: Results of Dana-Farber Cancer Institute/Children's Hospital Acute Lymphoblastic Leukemia Consortium Protocol 85-01.
J Clin Oncol
12
1994
740
15
Blethen
SL
Allen
DB
Graves
D
August
G
Moshang
T
Rosenfeld
R
Safety of recombinant deoxyribonucleic acid-derived growth hormone: The National Cooperative Growth Study experience.
J Clin Endocrinol Metab
81
1996
1704
16
Lin
YW
Kubota
M
Wakazono
Y
Hirota
H
Okuda
A
Bessho
R
Usami
I
Kataoka
A
Yamanaka
C
Akiyama
Y
Furusho
K
Normal mutation frequencies of somatic cells in patients receiving growth hormone therapy.
Mutat Res
362
1996
97
17
Rapaport
R
Oberfield
SE
Robison
L
Salisbury
S
David
R
Rao
J
Redmond
GP
Relationship of growth hormone deficiency and leukemia.
J Pediatr
126
1995
759
18
Linet
MS
Hatch
EE
Kleinerman
RA
Robison
LL
Kaune
WT
Friedman
DR
Severson
RK
Haines
SM
Hartsock
CT
Niwa
S
Wacholder
S
Tarone
RE
Residential exposure to magnetic fields and acute lymphoblastic leukemia in children.
N Engl J Med
337
1997
1
19
Alexander
FE
Space-time clustering of childhood acute lymphoblastic leukaemia: Indirect evidence for a transmissible agent.
Br J Cancer
65
1992
589
20
Alexander FE: Viruses, clusters and clustering of childhood leukaemia: A new perspective? Eur J Cancer 29A:1424, 1993
21
Kinlen
L
Evidence for an infective cause of childhood leukaemia: Comparison of a Scottish New Town with nuclear reprocessing sites in Britain.
Lancet
2
1988
1323
22
Petridou
E
Revinthi
K
Alexander
FE
Haidas
S
Koliouskas
D
Kosmidis
H
Piperopoulou
F
Tzortzatou
F
Trichopoulos
D
Space-time clustering of childhood leukaemia in Greece: Evidence supporting a viral aetiology.
Br J Cancer
73
1996
1278
23
van Steensel Moll
HA
Valkenburg
HA
van Zanen
GE
Childhood leukemia and parental occupation: A register-based case-control study.
Am J Epidemiol
121
1985
216
24
Wachsman
W
Golde
D
Chen
I
HTLV and human leukemia: Perspectives.
Semin Hematol
23
1986
245
25
Williams
D
Ragab
A
McDougal
J
HTLV-I antibodies in childhood leukemia.
JAMA
253
1985
2496
26
Lin
KH
Su
IJ
Chen
RL
Lin
DT
Tien
HF
Chen
BW
Lin
KS
Peripheral T-cell lymphoma in childhood: A report of five cases in Taiwan.
Med Pediatr Oncol
23
1994
26
27
Toledano
SR
Lange
BJ
Ataxia-telangiectasia and acute lymphoblastic leukemia.
Cancer
45
1980
1675
28
Aurias
A
Dutrillaux
B
Buriot
D
Lejeune
J
High frequencies of inversions and translocations of chromosomes 7 and 14 in ataxia telangiectasia.
Mutat Res
69
1980
369
29
Heppell
A
Butterworth
SV
Hollis
RJ
Kennaugh
AA
Beatty
DW
Taylor
AM
Breakage of the T cell receptor alpha chain locus in non malignant clones from patients with ataxia telangiectasia.
Hum Genet
79
1988
360
30
Russo
G
Isobe
M
Pegoraro
L
Finan
J
Nowell
PC
Croce
CM
Molecular analysis of a t(7;14)(Q35;Q32) chromosome translocation in a T cell leukemia of a patient with ataxia telangiectasia.
Cell
53
1988
137
31
Hollis
RJ
Kennaugh
AA
Butterworth
SV
Taylor
AM
Growth of large chromosomally abnormal T cell clones in ataxiatelangiectasia patients is associated with translocation at 14q11: A model for other T cell neoplasia.
Hum Genet
76
1987
389
32
Champlin
R
Gale
RP
Acute lymphoblastic leukemia: Recent advances in biology and therapy [see comments].
Blood
73
1989
2051
33
Raimondi
SC
Behm
FG
Roberson
PK
Pui
CH
Rivera
GK
Murphy
SB
Williams
DL
Cytogenetics of childhood T-cell leukemia.
Blood
72
1988
1560
34
Kagan
J
Finger
LR
Letofsky
J
Finan
J
Nowell
PC
Croce
CM
Clustering of breakpoints on chromosome 10 in acute T-cell leukemias with the t(10;14) chromosome translocation.
Proc Natl Acad Sci USA
86
1989
4161
35
Zutter
M
Hockett
RD
Roberts
CW
McGuire
EA
Bloomstone
J
Morton
CC
Deaven
LL
Crist
WM
Carroll
AJ
Korsmeyer
SJ
The t(10;14)(q24;q11) of T-cell acute lymphoblastic leukemia juxtaposes the delta T-cell receptor with TCL3, a conserved and activated locus at 10q24.
Proc Natl Acad Sci USA
87
1990
3161
36
Begley
CG
Aplan
PD
Davey
MP
Nakahara
K
Tchorz
K
Kurtzberg
J
Hershfield
MS
Haynes
BF
Cohen
DI
Waldmann
TA
Kirsch
IR
Chromosomal translocation in a human leukemic stem-cell line disrupts the T-cell antigen receptor delta-chain diversity region and results in a previously unreported fusion transcript.
Proc Natl Acad Sci USA
86
1989
2031
37
Boehm
T
Baer
R
Lavenir
I
Forster
A
Waters
JJ
Nacheva
E
Rabbitts
TH
The mechanism of chromosomal translocation t(11;14) involving the T-cell receptor C delta locus on human chromosome 14q11 and a transcribed region of chromosome 11p15.
EMBO J
7
1988
385
38
Cheng
JT
Yang
CY
Hernandez
J
Embrey
J
Baer
R
The chromosome translocation (11;14)(p13;q11) associated with T cell acute leukemia. Asymmetric diversification of the translocational junctions.
J Exp Med
171
1990
489
39
Garcia
IS
Kaneko
Y
Gonzalez Sarmiento
R
Campbell
K
White
L
Boehm
T
Rabbitts
TH
A study of chromosome 11p13 translocations involving TCR beta and TCR delta in human T cell leukaemia.
Oncogene
6
1991
577
40
Carroll
AJ
Crist
WM
Link
MP
Amylon
MD
Pullen
DJ
Ragab
AH
Buchanan
GR
Wimmer
RS
Vietti
TJ
The t(1;14)(p34;q11) is nonrandom and restricted to T-cell acute lymphoblastic leukemia: A Pediatric Oncology Group study.
Blood
76
1990
1220
41
Finger
LR
Kagan
J
Christopher
G
Kurtzberg
J
Hershfield
MS
Nowell
PC
Croce
CM
Involvement of the TCL5 gene on human chromosome 1 in T-cell leukemia and melanoma.
Proc Natl Acad Sci USA
86
1989
5039
42
Chen
Q
Yang
CY
Tsan
JT
Xia
Y
Ragab
AH
Peiper
SC
Carroll
A
Baer
R
Coding sequences of the tal-1 gene are disrupted by chromosome translocation in human T cell leukemia.
J Exp Med
172
1990
1403
43
Begley
CG
Aplan
PD
Denning
SM
Haynes
BF
Waldmann
TA
Kirsch
IR
The gene SCL is expressed during early hematopoiesis and encodes a differentiation-related DNA-binding motif.
Proc Natl Acad Sci USA
86
1989
10128
44
Aplan
PD
Lombardi
DP
Ginsberg
AM
Cossman
J
Bertness
VL
Kirsch
IR
Disruption of the human SCL locus by “illegitimate” V-(D)-J recombinase activity.
Science
250
1990
1426
45
Aplan
PD
Lombardi
DP
Reaman
GH
Sather
HN
Hammond
GD
Kirsch
IR
Involvement of the putative hematopoietic transcription factor SCL in T-cell acute lymphoblastic leukemia.
Blood
79
1992
1327
46
Bash
RO
Crist
WM
Shuster
JJ
Link
MP
Amylon
M
Pullen
J
Carroll
AJ
Buchanan
GR
Smith
RG
Baer
R
Clinical features and outcome of T-cell acute lymphoblastic leukemia in childhood with respect to alterations at the TAL1 locus: A Pediatric Oncology Group study.
Blood
81
1993
2110
47
Porcher
C
Swat
W
Rockwell
K
Fujiwara
Y
Alt
FW
Orkin
SH
The T cell leukemia oncoprotein SCL/tal-1 is essential for development of all hematopoietic lineages.
Cell
86
1996
47
48
Pulford
K
Lecointe
N
Leroy Viard
K
Jones
M
Mathieu Mahul
D
Mason
DY
Expression of TAL-1 proteins in human tissues.
Blood
85
1995
675
49
Elwood
NJ
Cook
WD
Metcalf
D
Begley
CG
SCL, the gene implicated in human T-cell leukaemia, is oncogenic in a murine T-lymphocyte cell line.
Oncogene
8
1993
3093
50
Dube
ID
Raimondi
SC
Pi
D
Kalousek
DK
A new translocation, t(10;14)(q24;q11), in T cell neoplasia.
Blood
67
1986
1181
51
Kennedy
MA
Gonzalez Sarmiento
R
Kees
UR
Lampert
F
Dear
N
Boehm
T
Rabbitts
TH
HOX11, a homeobox-containing T-cell oncogene on human chromosome 10q24.
Proc Natl Acad Sci USA
88
1991
8900
52
Hatano
M
Roberts
CW
Minden
M
Crist
WM
Korsmeyer
SJ
Deregulation of a homeobox gene, HOX11, by the t(10;14) in T cell leukemia.
Science
253
1991
79
53
Dube
ID
Kamel Reid
S
Yuan
CC
Lu
M
Wu
X
Corpus
G
Raimondi
SC
Crist
WM
Carroll
AJ
Minowada
J
Baker
JB
A novel human homeobox gene lies at the chromosome 10 breakpoint in lymphoid neoplasias with chromosomal translocation t(10;14).
Blood
78
1991
2996
54
Dear
TN
Sanchez Garcia
I
Rabbitts
TH
The HOX11 gene encodes a DNA-binding nuclear transcription factor belonging to a distinct family of homeobox genes.
Proc Natl Acad Sci USA
90
1993
4431
55
Yamamoto
H
Hatano
M
Iitsuka
Y
Mahyar
NS
Yamamoto
M
Tokuhisa
T
Two forms of Hox11, a T cell leukemia oncogene, are expressed in fetal spleen but not in primary lymphocytes.
Mol Immunol
32
1995
1177
56
Roberts
CW
Shutter
JR
Korsmeyer
SJ
Hox11 controls the genesis of the spleen.
Nature
368
1994
747
57
Salvati
PD
Ranford
PR
Ford
J
Kees
UR
HOX11 expression in pediatric acute lymphoblastic leukemia is associated with T-cell phenotype.
Oncogene
11
1995
1333
58
Williams
DL
Look
AT
Melvin
SL
Roberson
PK
Dahl
G
Flake
T
Stass
S
New chromosomal translocations correlate with specific immunophenotypes of childhood acute lymphoblastic leukemia.
Cell
36
1984
101
59
Ribeiro
RC
Raimondi
SC
Behm
FG
Cherrie
J
Crist
WM
Pui
CH
Clinical and biologic features of childhood T-cell leukemia with the t(11;14).
Blood
78
1991
466
60
Zalcberg
IQ
Silva
ML
Abdelhay
E
Tabak
DG
Ornellas
MH
Simoes
FV
Pucheri
W
Ribeiro
R
Seuanez
HN
Translocation 11;14 in three children with acute lymphoblastic leukemia of T-cell origin.
Cancer Genet Cytogenet
84
1995
32
61
McGuire
EA
Hockett
RD
Pollock
KM
Bartholdi
MF
O'Brien
SJ
Korsmeyer
SJ
The t(11;14)(p15;q11) in a T-cell acute lymphoblastic leukemia cell line activates multiple transcripts, including Ttg-1, a gene encoding a potential zinc finger protein.
Mol Cell Biol
9
1989
2124
62
Royer Pokora
B
Fleischer
B
Ragg
S
Loos
U
Williams
D
Molecular cloning of the translocation breakpoint in T-ALL 11;14 (p13;q11): Genomic map of TCR alpha and delta region on chromosome 14q11 and long-range map of region 11p13.
Hum Genet
82
1989
264
63
Boehm
T
Foroni
L
Kaneko
Y
Perutz
MF
Rabbitts
TH
The rhombotin family of cysteine-rich LIM-domain oncogenes: Distinct members are involved in T-cell translocations to human chromosomes 11p15 and 11p13.
Proc Natl Acad Sci USA
88
1991
4367
64
Royer Pokora
B
Loos
U
Ludwig
WD
TTG-2, a new gene encoding a cysteine-rich protein with the lim motif, is overexpressed in acute T-cell leukaemia with the t(11;14)(P13;Q11).
Oncogene
6
1887
1887
65
Wilkinson
DA
Neale
GA
Mao
S
Naeve
CW
Goorha
RM
Elf-2, a rhombotin-2 binding ets transcription factor: Discovery and potential role in T cell leukemia.
Leukemia
11
1997
86
66
Raimondi
SC
Pui
CH
Behm
FG
Williams
DL
7q32-q36 translocations in childhood T cell leukemia: Cytogenetic evidence for involvement of the T cell receptor beta-chain gene.
Blood
69
1987
131
67
Cleary
ML
Mellentin
JD
Spies
J
Smith
SD
Chromosomal translocation involving the beta T cell receptor gene in acute leukemia.
J Exp Med
167
1988
682
68
Mellentin
JD
Smith
SD
Cleary
ML
lyl-1, a novel gene altered by chromosomal translocation in T cell leukemia, codes for a protein with a helix-loop-helix DNA binding motif.
Cell
58
1989
77
69
Ellisen
LW
Bird
J
West
DC
Soreng
AL
Reynolds
TC
Smith
SD
Sklar
J
TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms.
Cell
66
1991
649
70
Burnett
RC
David
JC
Harden
AM
Le Beau
MM
Rowley
JD
Diaz
MO
The Lck gene is involved in the t(1;7)(P34;Q34) in the T-cell acute lymphoblastic leukemia derived cell line, Hsb-2.
Gene Chromosom Cancer
3
1991
461
71
Tycko
B
Smith
SD
Sklar
J
Chromosomal translocations joining lck and tcrb loci in human T cell leukemia.
J Exp Med
174
1991
867
72
Abraham
KM
Levin
SD
Marth
JD
Forbush
KA
Perlmutter
RM
Thymic tumorigenesis induced by overexpression of P56lck.
Proc Natl Acad Sci USA
88
1991
3977
73
Wildin
RS
Garvin
AM
Pawar
S
Lewis
DB
Abraham
KM
Forbush
KA
Ziegler
SF
Allen
JM
Perlmutter
RM
Developmental regulation of lck gene expression in T lymphocytes.
J Exp Med
173
1991
383
74
Finver
SN
Nishikura
K
Finger
LR
Haluska
FG
Finan
J
Nowell
PC
Croce
CM
Sequence analysis of the MYC oncogene involved in the t(8;14)(q24;q11) chromosome translocation in a human leukemia T-cell line indicates that putative regulatory regions are not altered.
Proc Natl Acad Sci USA
85
1988
3052
75
Erikson
J
Williams
DL
Finan
J
Nowell
PC
Croce
CM
Locus of the alpha-chain of the T-cell receptor is split by chromosome translocation in T-cell leukemias.
Science
229
1985
784
76
Finger
LR
Huebner
K
Cannizzaro
LA
McLeod
K
Nowell
PC
Croce
CM
Chromosomal translocation in T-cell leukemia line HUT 78 results in a MYC fusion transcript.
Proc Natl Acad Sci USA
85
1988
9158
77
Elledge
SJ
Cell cycle checkpoints: Preventing an identity crisis.
Science
274
1996
1664
78
Boise
LH
Thompson
CB
Hierarchical control of lymphocyte survival.
Science
274
1996
67
79
Stillman
B
Cell cycle control of DNA replication.
Science
274
1996
1659
80
Lowsky
R
DeCoteau
JF
Reitmair
AH
Ichinohasama
R
Dong
WF
Xu
Y
Mak
TW
Kadin
ME
Minden
MD
Defects of the mismatch repair gene MSH2 are implicated in the development of murine and human lymphoblastic lymphomas and are associated with the aberrant expression of rhombotin-2 (Lmo-2) and Tal-1 (SCL).
Blood
89
1997
2276
81
Weinberg
RA
Tumor Suppressor Genes.
Science
254
1991
1138
82
(abstr 3095)
Hermanson
M
Liu
Y
Zabarovsky
E
Grander
D
Gahrton
G
Juliusson
G
Rasool
M
Wu
X
Buys
C
Yankovsky
N
et al
Chromosome 13q14 deletions in lymphoid malignancies (meeting abstract).
Proc Am Assoc Cancer Res
36
1995
104
83
(abstr 616)
Zhou
M
Zaki
SR
Ragab
AH
Findley
HW
Frequent alteration of Rb tumor-suppressor gene in childhood acute lymphoblastic leukemia (meeting abstract).
Proc Am Assoc Cancer Res
34
1993
104
84
Harper
JW
Adami
GR
Wei
N
Keyomarsi
K
Elledge
SJ
The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases.
Cell
75
1993
805
85
Hannon
GJ
Beach
D
p15ink4b is a potential effector of Tgf-beta-induced cell cycle arrest [see comments].
Nature
371
1994
257
86
Guan KL, Jenkins CW, Li Y, Nichols MA, Wu X, CL OK, Matera AG, Xiong Y: Growth suppression by p18, a p16INK4/MTS1- and p14INK4B/MTS2-related CDK6 inhibitor, correlates with wild-type pRb function. Genes Dev 8:2939, 1994
87
Chan
FK
Zhang
J
Cheng
L
Shapiro
DN
Winoto
A
Identification of human and mouse p19, a novel CDK4 and CDK6 inhibitor with homology to p16ink4.
Mol Cell Biol
15
1995
2682
88
Matsuoka
S
Edwards
MC
Bai
C
Parker
S
Zhang
P
Baldini
A
Harper
JW
Elledge
SJ
p57kip2, a structurally distinct member of the p21cip1 Cdk inhibitor family, is a candidate tumor suppressor gene.
Genes Dev
9
1995
650
89
Polyak
K
Kato
JY
Solomon
MJ
Sherr
CJ
Massague
J
Roberts
JM
Koff
A
p27kip1, a cyclin-Cdk inhibitor, links transforming growth factor-beta and contact inhibition to cell cycle arrest.
Genes Dev
8
1994
9
90
Rasool
O
Heyman
M
Brandter
LB
Liu
Y
Grander
D
Soderheall
S
Einhorn
S
p15ink4b and p16ink4 gene inactivation in acute lymphocytic leukemia.
Blood
85
1995
3431
91
Ohnishi
H
Kawamura
M
Ida
K
Sheng
XM
Hanada
R
Nobori
T
Yamamori
S
Hayashi
Y
Homozygous deletions of p16/MTS1 gene are frequent but mutations are infrequent in childhood T-cell acute lymphoblastic leukemia.
Blood
86
1995
1269
92
Okuda
T
Shurtleff
SA
Valentine
MB
Raimondi
SC
Head
DR
Behm
F
Curcio Brint
AM
Liu
Q
Pui
CH
Sherr
CJ
Beach
D
Look
AT
Downing
JR
Frequent deletion of p16INK4a/MTS1 and p15INK4b/MTS2 in pediatric acute lymphoblastic leukemia.
Blood
85
1995
2321
93
Guidal Giroux
C
Gerard
B
Cave
H
Duval
M
Rohrlich
P
Elion
J
Vilmer
E
Grandchamp
B
Deletion mapping indicates that MTS1 is the target of frequent deletions at chromosome 9p21 in paediatric acute lymphoblastic leukaemias.
Br J Haematol
92
1996
410
94
Fizzotti
M
Cimino
G
Pisegna
S
Alimena
G
Quartarone
C
Mandelli
F
Pelicci
PG
Lo Coco
F
Detection of homozygous deletions of the cyclin-dependent kinase 4 inhibitor (p16) gene in acute lymphoblastic leukemia and association with adverse prognostic features.
Blood
85
1995
2685
95
Batova
A
Diccianni
MB
Yu
JC
Nobori
T
Link
MP
Pullen
J
Yu
AL
Frequent and selective methylation of p15 and deletion of both p15 and p16 in T-cell acute lymphoblastic leukemia.
Cancer Res
57
1997
832
96
Chilcote
RR
Brown
E
Rowley
JD
Lymphoblastic leukemia with lymphomatous features associated with abnormalities of the short arm of chromosome 9.
N Engl J Med
313
1985
286
97
Uckun
FM
Gajl Peczalska
KJ
Provisor
AJ
Heerema
NA
Immunophenotype-karyotype associations in human acute lymphoblastic leukemia.
Blood
73
1989
271
98
Bloomfield
CD
Secker Walker
LM
Goldman
AI
Van Den Berghe
H
de la Chapelle
A
Ruutu
T
Alimena
G
Garson
OM
Golomb
HM
Rowley
JD
Kaneko
Y
Whang-Peng
J
Prigogina
E
Philip
P
Sandberg
AA
Lawler
SD
Mitleman
F
Six-year follow-up of the clinical significance of karyotype in acute lymphoblastic leukemia.
Cancer Genet Cytogenet
40
1989
171
99
Sherr
CJ
Cancer cell cycles.
Science
274
1996
1672
100
Harvey
M
Vogel
H
Morris
D
Bradley
A
Bernstein
A
Donehower
LA
A mutant p53 transgene accelerates tumour development in heterozygous but not nullizygous p53-deficient mice.
Nat Genet
9
1995
305
101
Yeargin J, Cheng J, Haas M: Role of the p53 tumor suppressor gene in the pathogenesis and in the suppression of acute lymphoblastic T-cell leukemia. Leukemia 6 Suppl 3:85s, 1992
102
Hsiao
M
Low
J
Dorn
E
Ku
D
Pattengale
P
Yeargin
J
Haas
M
Gain-of-function mutations of the p53 gene induce lymphohematopoietic metastatic potential and tissue invasiveness.
Am J Pathol
145
1994
702
103
Savitsky
K
Bar Shira
A
Gilad
S
Rotman
G
Ziv
Y
Vanagaite
L
Tagle
DA
Smith
S
Uziel
T
Sfez
S
et al
A single ataxia telangiectasia gene with a product similar to PI-3 kinase [see comments].
Science
268
1995
1749
104
Painter
RB
Young
BR
Radiosensitivity in ataxia-telangiectasia: A new explanation.
Proc Natl Acad Sci USA
77
1980
7315
105
Xu
Y
Baltimore
D
Dual roles of ATM in the cellular response to radiation and in cell growth control [see comments].
Genes Dev
10
1996
2401
106
Xu
Y
Ashley
T
Brainerd
EE
Bronson
RT
Meyn
MS
Baltimore
D
Targeted disruption of ATM leads to growth retardation, chromosomal fragmentation during meiosis, immune defects, and thymic lymphoma [see comments].
Genes Dev
10
1996
2411
107
Barlow
C
Hirotsune
S
Paylor
R
Liyanage
M
Eckhaus
M
Collins
F
Shiloh
Y
Crawley
JN
Ried
T
Tagle
D
Wynshaw Boris
A
Atm-deficient mice: A paradigm of ataxia telangiectasia.
Cell
86
1996
159
108
Taylor
A
Metalfe
J
Thick
J
Mak
Y
Leukemia and lymphoma in ataxia telangiectasia.
Blood
87
1996
423
109
Spector B, Filipovich A, Perry G, Kersey K: Epidemiology of cancer in ataxia telangiectasia, in Bridges B, Harnden D (eds): A Cellular and Molecular Link Between Cancer, Neuropathology, and Immune Deficiency. Chichester, UK, Wiley, 1982, p1
110
Collyn dHooghe M, Galiegue Zouitina S, Szymiczek D, Lantoine D, Quief S, Loucheux Lefebvre MH, Kerckaert JP: Quantitative and qualitative variation of ETS-1 transcripts in hematologic malignancies. Leukemia 7:1777, 1993
111
Georgopoulos
K
Moore
DD
Derfler
B
Ikaros, an early lymphoid-specific transcription factor and a putative mediator for T cell commitment.
Science
258
1992
808
112
Winandy
S
Wu
P
Georgopoulos
K
A dominant mutation in the Ikaros gene leads to rapid development of leukemia and lymphoma.
Cell
83
1995
289
113
Debatin
KM
Goldman
CK
Waldmann
TA
Krammer
PH
APO-1-induced apoptosis of leukemia cells from patients with adult T-cell leukemia.
Blood
81
1993
2972
114
Debatin
KM
Krammer
PH
Resistance to APO-1 (CD95) induced apoptosis in T-ALL is determined by a BCL-2 independent anti-apoptotic program.
Leukemia
9
1995
815
115
Lucking Famira
KM
Daniel
PT
Moller
P
Krammer
PH
Debatin
KM
APO-1 (CD95) mediated apoptosis in human T-ALL engrafted in SCID mice.
Leukemia
8
1994
1825
116
Cory
S
Regulation of lymphocyte survival by the bcl-2 gene family.
Annu Rev Immunol
13
1995
513
117
Conroy
LA
Alexander
DR
The role of intracellular signalling pathways regulating thymocyte and leukemic T cell apoptosis.
Leukemia
10
1996
1422
118
Oltvai
ZN
Milliman
CL
Korsmeyer
SJ
Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death.
Cell
74
1993
609
119
Kitada
S
Krajewski
S
Miyashita
T
Krajewska
M
Reed
JC
Gamma-radiation induces upregulation of Bax protein and apoptosis in radiosensitive cells in vivo.
Oncogene
12
1996
187
120
Uckun
FM
Yang
Z
Sather
HN
Steinherz
P
Nachman
J
Bostrom
B
Crotty
L
Sarquis
M
Ek
O
Zren
T
Tubergen
D
Reaman
G
Gaynon
P
Cellular expression of anti-apoptotic BCL-2 oncoprotein in newly diagnosed childhood acute lymphoblastic leukemia.
Blood
89
1997
3769
121
Sen
L
Borella
L
Clinical importance of lymphoblasts with T markers in childhood acute leukemia.
N Engl J Med
292
1975
828
122
Borella
L
Sen
L
T- and B-lymphocytes and lymphoblasts in untreated acute lymphocytic leukemia.
Cancer
34
1974
646
123
Williams
DL
Raimondi
S
Rivera
G
George
S
Berard
CW
Murphy
SB
Presence of clonal chromosome abnormalities in virtually all cases of acute lymphoblastic leukemia [letter].
N Engl J Med
313
1985
640
124
Pui
CH
Crist
WM
Look
AT
Biology and clinical significance of cytogenetic abnormalities in childhood acute lymphoblastic leukemia.
Blood
76
1990
1449
125
Crist
W
Boyett
J
Pullen
J
van Eys
J
Vietti
T
Clinical and biologic features predict poor prognosis in acute lymphoid leukemias in children and adolescents: A Pediatric Oncology Group review.
Med Pediatr Oncol
14
1986
135
126
Williams
DL
Harber
J
Murphy
SB
Look
AT
Kalwinsky
DK
Rivera
G
Melvin
SL
Stass
S
Dahl
GV
Chromosomal translocations play a unique role in influencing prognosis in childhood acute lymphoblastic leukemia.
Blood
68
1986
205
127
Bloomfield
CD
Goldman
AI
Alimena
G
Berger
R
Borgstrom
GH
Brandt
L
Catovsky
D
de la Chapelle
A
Dewald
GW
Garson
OM
et al
Chromosomal abnormalities identify high-risk and low-risk patients with acute lymphoblastic leukemia.
Blood
67
1986
415
128
Look
AT
Roberson
PK
Williams
DL
Rivera
G
Bowman
WP
Pui
CH
Ochs
J
Abromowitch
M
Kalwinsky
D
Dahl
GV
et al
Prognostic importance of blast cell DNA content in childhood acute lymphoblastic leukemia.
Blood
65
1985
1079
129
Jackson
JF
Boyett
J
Pullen
J
Brock
B
Patterson
R
Land
V
Borowitz
M
Head
D
Crist
W
Favorable prognosis associated with hyperdiploidy in children with acute lymphocytic leukemia correlates with extra chromosome 6: A Pediatric Oncology Group Study.
Cancer
66
1990
1183
130
Pui
CH
Carroll
AJ
Head
D
Raimondi
SC
Shuster
JJ
Crist
WM
Link
MP
Borowitz
MJ
Behm
FG
Land
VJ
et al
Near-triploid and near-tetraploid acute lymphoblastic leukemia of childhood.
Blood
76
1990
590
131
Zintl
F
Plenert
W
Malke
H
Results of acute lymphoblastic leukemia therapy in childhood with a modified BFM protocol in a multicenter study in the German Democratic Republic.
Hamatol Bluttransfus
30
1987
471
132
Riehm
H
Reiter
A
Schrappe
M
Berthold
F
Dopfer
R
Gerein
V
Ludwig
R
Ritter
J
Stollmann
B
Henze
G
Corticosteroid-dependent reduction of leukocyte count in blood as a prognostic factor in acute lymphoblastic leukemia in childhood (therapy study ALL-BFM 83).
Klin Padiatr
199
1987
151
133
Dahl
GV
Rivera
GK
Look
AT
Hustu
HO
Kalwinsky
DK
Abromowitch
M
Mirro
J
Ochs
J
Murphy
SB
Dodge
RK
et al
Teniposide plus cytarabine improves outcome in childhood acute lymphoblastic leukemia presenting with a leukocyte count greater than or equal to 100 × 109/L.
J Clin Oncol
5
1987
1015
134
Smith
M
Arthur
D
Camitta
B
Carroll
AJ
Crist
W
Gaynon
P
Gelber
R
Heerema
N
Korn
EL
Link
M
Murphy
S
Pui
CH
Pullen
J
Reamon
G
Sallan
SE
Sather
H
Shuster
J
Simon
R
Trigg
M
Tubergen
D
Uckun
F
Ungerleider
R
Uniform approach to risk classification and treatment assignment for children with acute lymphoblastic leukemia [see comments].
J Clin Oncol
14
1996
18
135
Steinherz PG, Siegel SE, Bleyer WA, Kersey J, Chard R, Jr., Coccia P, Leiken S, Lukens J, Neerhout R, Nesbit M, Miller DR, Reaman G, Sather H, Hammond D: Lymphomatous presentation of childhood acute lymphoblastic leukemia. Cancer 68:751, 1991
136
Henze
G
Langermann
HJ
Kaufmann
U
Ludwig
R
Schellong
G
Stollmann
B
Riehm
H
Thymic involvement and initial white blood count in childhood acute lymphoblastic leukemia.
Am J Pediatr Hematol Oncol
3
1981
369
137
Pui
CH
Behm
FG
Singh
B
Schell
MJ
Williams
DL
Rivera
GK
Kalwinsky
DK
Sandlund
JT
Crist
WM
Raimondi
SC
Heterogeneity of presenting features and their relation to treatment outcome in 120 children with T-cell acute lymphoblastic leukemia.
Blood
75
1990
174
138
Schaison
G
Leverger
G
Bancillon
A
Marty
M
Olive
D
Cornu
G
Griscelli
C
Lemerle
S
Harrousseau
J
Bonnet
M
Freycon
F
Dufillot
D
Demeocq
F
Bauters
F
Lamagnere
J
Taboureau
O
Intermediate risk childhood acute lymphoblastic leukemias: Anascrine + cytosine arabinoside versus intermediate dose methotrexate for consolidation, and 6 mercaptopurine + methotrexate + vincristine versus monthly pulses for maintenance.
Hamatol Bluttransfus
30
1987
461
139
Gaynon
PS
Bleyer
WA
Steinherz
PG
Finklestein
JZ
Littman
PS
Miller
DR
Reaman
GH
Sather
HN
Hammond
GD
Modified BFM therapy for children with previously untreated acute lymphoblastic leukemia and unfavorable prognostic features. Report of Children's Cancer Study Group Study CCG-193P.
Am J Pediatr Hematol Oncol
10
1988
42
140
Uckun
F
Steinherz
P
Sather
H
Trigg
M
Arthur
D
Tubergen
D
Gaynon
P
Reaman
G
CD2 antigen expression on leukemic cells as a predictor of event-free survival after chemotherapy for T-lineage acute lymphoblastic leukemia: A Children's Cancer Group Study.
Blood
88
1996
4288
141
Uckun
FM
Gaynon
P
Sensel
M
Nachman
J
Trigg
M
Steinherz
P
Bostrom
B
Sather
H
Reaman
G
Clinical features and treatment outcome of childhood T-lineage acute lymphoblastic leukemia according to the apparent maturational stage of T-lineage leukemic blasts: A Children's Cancer Group Study.
J Clin Oncol
15
1997
2214
142
Wiersma
SR
Ortega
J
Sobel
E
Weinberg
KI
Clinical importance of myeloid-antigen expression in acute lymphoblastic leukemia of childhood [see comments].
N Engl J Med
324
1991
800
143
Kurec
AS
Belair
P
Stefanu
C
Barrett
DM
Dubowy
RL
Davey
FR
Significance of aberrant immunophenotypes in childhood acute lymphoblastic leukemia.
Cancer
67
1991
3081
144
Pui
CH
Behm
FG
Singh
B
Rivera
GK
Schell
MJ
Roberts
WM
Crist
WM
Mirro
J
Jr
Myeloid-associated antigen expression lacks prognostic value in childhood acute lymphoblastic leukemia treated with intensive multiagent chemotherapy.
Blood
75
1990
198
145
Bradstock
KF
Kirk
J
Grimsley
PG
Kabral
A
Hughes
WG
Unusual immunophenotypes in acute leukaemias: Incidence and clinical correlations.
Br J Haematol
72
1989
512
146
Uckun
FM
Sather
HN
Gaynon
P
Arthur
D
Trigg
M
Tubergen
D
Nachman
J
Steinherz
P
Sensel
M
Reaman
G
Clinical features and treatment outcome of children with myeloid antigen positive acute lymphoblastic leukemia: A report from the Children's Cancer Group.
Blood
90
1997
28
147
Pieters
R
Kaspers
GJ
Klumper
E
Veerman
AJ
Clinical relevance of in vitro drug resistance testing in childhood acute lymphoblastic leukemia: The state of the art.
Med Pediatr Oncol
22
1994
299
148
Kaspers
GJ
Pieters
R
Van Zantwijk
CH
Van Wering
ER
Veerman
AJ
Clinical and cell biological features related to cellular drug resistance of childhood acute lymphoblastic leukemia cells.
Leuk Lymphoma
19
1995
407
149
Lauer
SJ
Camitta
BM
Leventhal
BG
Mahoney
DH
Jr
Shuster
JJ
Adair
S
Casper
JT
Civin
CI
Graham
M
Kiefer
G
Pullen
J
Steuber
CP
Kamen
B
Intensive alternating drug pairs for treatment of high-risk childhood acute lymphoblastic leukemia. A Pediatric Oncology Group pilot study.
Cancer
71
1993
2854
150
Goker
E
Lin
JT
Trippett
T
Elisseyeff
Y
Tong
WP
Niedzwiecki
D
Tan
C
Steinherz
P
Schweitzer
BI
Bertino
JR
Decreased polyglutamylation of methotrexate in acute lymphoblastic leukemia blasts in adults compared to children with this disease.
Leukemia
7
1993
1000
151
Barredo
JC
Synold
TW
Laver
J
Relling
MV
Pui
CH
Priest
DG
Evans
WE
Differences in constitutive and post-methotrexate folylpolyglutamate synthetase activity in B-lineage and T-lineage leukemia.
Blood
84
1994
564
152
Quddus
FF
Leventhal
BG
Boyett
JM
Pullen
DJ
Crist
WM
Borowitz
MJ
Glucocorticoid receptors in immunological subtypes of childhood acute lymphocytic leukemia cells: A Pediatric Oncology Group study.
Cancer Res
45
1985
6482
153
Costlow
ME
Pui
CH
Dahl
GV
Glucocorticoid receptors in childhood acute lymphocytic leukemia.
Cancer Res
42
1982
4801
154
Gros
P
Ben Neriah
YB
Croop
JM
Housman
DE
Isolation and expression of a complementary DNA that confers multidrug resistance.
Nature
323
1986
728
155
Laurent
G
Frankel
AE
Hertler
AA
Schlossman
DM
Casellas
P
Jansen
FK
Treatment of leukemia patients with T101 ricin a chain immunotoxins.
Cancer Treat Res
37
1988
483
156
Kreitman RJ, Chaudhary VK, Waldmann TA, Hanchard B, Cranston B, FitzGerald DJ, Pastan I: Cytotoxic activities of recombinant immunotoxins composed of pseudomonas toxin or diphtheria toxin toward lymphocytes from patients with adult T-cell leukemia. Leukemia 7:553, 1993
157
Waurzyniak
B
Schneider
E
Yanishevski
Y
Gunther
R
Chelstrom
LM
Wendorf
H
Myers
DE
Irvin
JD
Messinger
Y
Ek
O
Seren
T
Langlie
M
Evans
WE
Uckun
FM
In vivo toxicity, pharmacokinetics, and antileukemic activity of TXU (anti-CD7)-pokeweed antiviral protein (PAP) immunotoxin.
Clin Cancer Res
3
1997
881
158
Uckun
FM
Reaman
GH
Immunotoxins for treatment of leukemia and lymphoma.
Leuk Lymphoma
18
1995
195
159
Murphy
JR
Bishai
W
Borowski
M
Miyanohara
A
Boyd
J
Nagle
S
Genetic construction, expression, and melanoma-selective cytotoxicity of a diphtheria toxin-related alpha-melanocyte-stimulating hormone fusion protein.
Proc Natl Acad Sci USA
83
1986
8258
160
Kreitman
RJ
Chang
CN
Hudson
DV
Queen
C
Bailon
P
Pastan
I
Anti-Tac(Fab)-PE40, a recombinant double-chain immunotoxin which kills interleukin-2-receptor-bearing cells and induces complete remission in an in vivo tumor model.
Int J Cancer
57
1994
856
161
LeMaistre
CF
Craig
FE
Meneghetti
C
McMullin
B
Parker
K
Reuben
J
Boldt
DH
Rosenblum
M
Woodworth
T
Phase I trial of a 90-minute infusion of the fusion toxin DAB486IL-2 in hematological cancers.
Cancer Res
53
1993
3930
162
Giblett
ER
ADA and PNP deficiencies: How it all began.
Ann NY Acad Sci
451
1985
1
163
Kredich NM, Hershfield MS: Immunodeficiency diseases caused by adenosine deaminase deficiency and purine nucleoside phosphorylase deficiency, in Stanbury JB, Wyngaarden JB, Goldstein JL, Brown MS (eds): The Metabolic Basis of Inherited Disease. New York, NY, McGraw-Hill, 1983, p 1157
164
Ullman
B
Martin
DW
Jr
Specific cytotoxicity of arabinosylguanine toward cultured T lymphoblasts.
J Clin Invest
74
1984
951
165
Verhoef
V
Fridland
A
Metabolic basis of arabinonucleoside selectivity for human leukemic T- and B-lymphoblasts.
Cancer Res
45
1985
3646
166
Hebert
ME
Greenberg
ML
Chaffee
S
Gravatt
L
Hershfield
MS
Elion
GB
Kurtzberg
J
Pharmacologic purging of malignant T cells from human bone marrow using 9-beta-D-arabinofuranosylguanine.
Transplantation
52
1991
634
167
Gravatt
LC
Chaffee
S
Hebert
ME
Halperin
EC
Friedman
HS
Kurtzberg
J
Efficacy and toxicity of 9-beta-D-arabinofuranosylguanine (araG) as an agent to purge malignant T cells from murine bone marrow: Application to an in vivo T-leukemia model.
Leukemia
7
1993
1261
168
Lambe
CU
Averett
DR
Paff
MT
Reardon
JE
Wilson
JG
Krenitsky
TA
2-Amino-6-methoxypurine arabinoside: An agent for T-cell malignancies.
Cancer Res
55
1995
3352
169
Plunkett
W
Gandhi
V
Nowak
B
Du
M
Rodriguez
CO
Keating
MJ
Pharmacokinetics of compound 506, a soluble prodrug of arabinosylguanine, in adult leukemias (meeting abstract).
Proc Am Assoc Cancer Res
37
1996
125
Sign in via your Institution