ACUTE LYMPHOBLASTIC leukemias (ALL) are characterized by clonal proliferation, accumulation, and tissue infiltration of neoplastic cells. They are mainly regarded as childhood diseases, with an early incidence peak at 2 to 5 years of age, where they represent about 80% of the childhood leukemias in the United States, and occur with an incidence of up to 30 cases per 1 million population per year.1 The age-adjusted incidence of ALL in adults (usually defined as 15 years of age and older) amounts to about one third of that in children.1 However, ALL has a bimodal distribution, with a second peak around age 50 and a low but steady rise in incidence with increasing age.2 

Improvements in cytogenetic techniques have yielded significant insight as to the importance of cytogenetic abnormalities in the pathophysiology and prognosis of hematologic malignancies. Heim and Mitelman3 reported an overall increase in the number of reported cases of cancer with cytogenetic alterations from 3,844 in 1983 to more than 22,000 in 1994. Of all neoplasms, leukemias have been by far the most intensively investigated and account for more than 60% of all listed chromosomal aberrations, including more than 3,000 cases of ALL.

The majority of cases of ALL demonstrate an abnormal karyotype, either in chromosome number (ploidy) or as structural changes such as translocations, inversions, or deletions. These changes were detected in only half of ALL patients in the first banding studies.3The scantiness of information gained from chromosomal findings in ALL has been, in large part, due to technical difficulties. Chromosome studies in ALL exhibit poor morphology; chromosomes tend to spread poorly, and appear blurred and fuzzy with indistinct margins, making banding studies challenging or even impossible.4,5Improvements in spreading and banding techniques have resulted in higher rates of detection, and most studies now report chromosomal changes in 60% to 85% of ALL cases.6-10 The Third International Workshop on Chromosomes in Leukemia (TIWCL) found the majority of cytogenetic changes in cases of B-precursor ALL, with only 39% occurring in T-cell ALL.6,9 Williams et al4 used a direct technique of bone marrow (BM) chromosomal analysis developed particularly for studies in ALL, which paid attention to sampling and processing steps using specific flaming techniques and modified G-banding procedures. They identified clonal karyotypic abnormalities in 94% to 98% of cases of ALL. Such improved techniques also detected nonrandomly occurring cytogenetic abnormalities in cases with hyperdiploid chromosome numbers (>50) that had previously been classified as normal in karyotype.11 These results showed a high prevalence of clonal chromosomal abnormalities in ALL, as was shown for acute nonlymphoblastic leukemias by Yunis,12 who used high-resolution banding techniques. These and similar studies underscore the significant yield achieved with thorough cytogenetic studies in ALL.

That cytogenetic abnormalities confer important prognostic information in ALL was first reported by Secker-Walker et al13 in 1978 in a series of childhood ALL. The investigators reported better clinical outcomes in cases with hyperdiploid karyotypes than in those with hypodiploidy or pseudodiploidy, and these findings were confirmed in the follow-up study14 and by other researchers.8,15-17 The TIWCL examined 330 newly diagnosed ALL patients (172 adults and 157 children) and found that chromosomal abnormalities distinguished high-risk from low-risk patients. Complete remission (CR) rates, remission durations, as well as disease-free-survivals (DFS) were significantly affected by the karyotypic abnormalities.6 Among adult patients the highest likelihood of cure (21% to 30%) was projected in patients with chromosome numbers of >50, or 47 to 50, with 6q−, or with a normal karyotype.18 As with children, karyotypes in adults were significant independent predictors of remission duration or DFS, after considering covariates such as age, leukocyte count at presentation, or French-American-British (FAB) morphology.19 Secker-Walker et al10 reported the prognostic effect of certain structural rearrangements, such as t(9;22), to be independent of other single variables.

Most studies on karyotypic abnormalities and their clinical significance have been performed in childhood ALL.15,20-24Adult ALL may show nonrandom chromosomal abnormalities similar to those found in childhood ALL, but their distribution and, possibly, their biological significance are different. Few studies have addressed these issues in adult ALL.6-8,18,25,26 This review focuses on the most important chromosomal abnormalities found in adult ALL and their prognostic and therapeutic implications.

Numerical chromosome abnormalities, either alone or in association with structural changes, are found in about half of ALL cases. Several ploidy groups have been identified (Table1).15,27 These include low hyperdiploidy (modal number 47 to 50), high or massive hyperdiploidy (>50), hypodiploidy (46 and lower), pseudodiploidy (normal number of chromosomes, but with associated structural changes), as well as gain or loss of a single chromosome as the sole karyotypic change. Sole numerical aberrations are less frequent than the numerical aberrations in combination with structural changes, which occur in about 40% to 70% of cases.5,7 

Table 1.

Frequency of Ploidy Groups in Adult ALL Compared With Childhood ALL

Ploidy Group Frequency in:
Adult ALL (%) Childhood ALL (%)
Normal karyotype 26-34  8-56  
Hypodiploid <46  2-8  5-6 
Pseudodiploid  7-59  3-42  
Hyperdiploid 47-50  7-17 8-16  
Hyperdiploid >50  4-9  14-27 
Near triploidy  3  <1 
Near tetraploidy  2  
Ploidy Group Frequency in:
Adult ALL (%) Childhood ALL (%)
Normal karyotype 26-34  8-56  
Hypodiploid <46  2-8  5-6 
Pseudodiploid  7-59  3-42  
Hyperdiploid 47-50  7-17 8-16  
Hyperdiploid >50  4-9  14-27 
Near triploidy  3  <1 
Near tetraploidy  2  

Data from references 7, 8, 15, 18, 26, 27, 32, and 37.

Hyperdiploidy

Chromosome numbers of 47 or more are found in up to one fourth of adult ALL cases, making the hyperdiploid group one of the most frequent ploidy groups (Table 1). Hyperdiploid chromosome numbers, particularly those with >50 chromosomes, are more frequent in children than in adults. Chromosome numbers cluster around 47 and between 51 to 55 so that a bimodal distribution becomes evident, similar to the distribution in childhood ALL.15 A low hyperdiploidy group with 47 to 50 chromosomes can be distinguished from a massive hyperdiploidy group with >50 chromosomes. Disomies and, particularly, trisomies or tetrasomies, are frequent and are dominant in the group with >50 chromosomes.

Although various chromosomes are involved in hyperdiploidy, certain karyotypes seem to prevail. In a series of 31 patients, Ankathil et al28 found that hyperdiploid karyotypes were mainly characterized by trisomies of chromosomes 8, 18, 19, and 21. The TIWCL study also showed, in both hyperdiploidy groups, gains in chromosomes 4 and 8, as well as 21 and 18, to be more frequent. Additional chromosomes 6, 10, and 14 appeared almost exclusively in the massive hyperdiploidy group.6 The Groupe Français de Cytogénétique Hématologique (GFCH) found in the group of >50 chromosomes mainly chromosomes 4, 6, 8, 10, 14, 17, and 21 and in the 47 to 50 group mainly chromosomes 5, 8, 10, and 21.7About half of all adult ALL cases with hyperdiploidy of a modal number of >50 can be expected to show additional chromosomal rearrangements, with translocation t(9;22) being the most common.7,9 In contrast, the TIWCL found relatively few structural rearrangements in both hyperdiploidy groups. This difference reflects the impact of advances in technology and expertise over little more than a decade on the yield of cytogenetic studies. In addition, the technical quality of hyperdiploid metaphase spreads is frequently substandard, and structural aberrations are still not reliably identified in a significant number of cases.29 

In the TIWCL study, almost all cases with >50 chromosomes were of precursor B-cell type and associated with favorable prognostic features such as low white blood cell count (WBC) at presentation, low lactate dehydrogenase (LDH) levels, and FAB L1 or L2 morphology.6 

In children with ALL, chromosomal numbers of >50 have been reported to have better response duration as well as median survival time (MST), whereas numbers between 47 and 50 conferred an intermediate prognosis. In this context, the combination of trisomies of both chromosomes 4 and 10 in children with hyperdiploid ALL identified a subgroup of patients with an extremely favorable DFS, and likelihood of cure with antimetabolite-based chemotherapy.30 

No such favorable constellation could be identified in adult ALL, where the impact of hyperdiploidy on prognosis appeared less significant (Table 2). Although the TIWCL showed higher CR rates, as well as increased CR duration and MST in this ploidy group for both children and adults, children still survived significantly longer than adults for modal numbers >50 or 47 to 50. The GFCH study on 443 adult ALL patients found a favorable prognosis in patients with hyperdiploidy of >50 chromosomes without the Philadelphia chromosome (Ph), as well as in patients with tetraploidy.7 The UKALL XA trial observed better outcomes with hyperdiploidy which added prognostic significance to age, sex, and WBC, unless these variables were combined.10 Campbell et al31 showed a survival advantage for adult patients with only numerical abnormalities and particularly for those with chromosome numbers more than 50. However, other studies could not confirm the association of hyperdiploidy with good outcome in adult ALL. Fenaux et al8found no significant differences in CR rate or duration between cytogenetic groups in 73 adult patients, except for a slight increase in CR duration with normal karyotypes, and worse outcomes with the Ph abnormality. This contrasted with their findings in pediatric patients, where a significant difference in response to treatment was found between children with hyperdiploidy and those with hypodiploidy. Walters et al,32 in a series of 91 adult patients, could not demonstrate superior outcome with hyperdiploidy.

Table 2.

Characteristics and Clinical Outcome in Various Ploidy Groups

Karyotype References
Normal <46 46 abnl 47-50 >50
FAB category (%)  
 L1  36  11  14  30  TIWCL 
 47  100  50  40  20  Fenaux et al* 
 71  48  53  33  60  GFCH  
 NA NA  NA  NA  NA  UKALL XA  
 L2  62  89  86  70  100  TIWCL 
 53 0  50  60  80  Fenaux et al* 
 NA NA  NA  NA  NA  GFCH 
 NA  NA  NA NA  NA  UKALL XA  
LSM (%)  
 Pre-B 62  75  69  71  100  TIWCL 
 77  66  075  100  Fenaux et al* 
 65  92  78 67  100  GFCH  
 75  91  71  75  96 UKALL XA  
 T  38  25  31  29  0  TIWCL 
 23  100  33  25  0  Fenaux et al* 
 35 5  19  33  0  GFCH  
 25  9  29 25  4  UKALL XA  
CR rate (%)  86  50  67  67 78  TIWCL 
 63  0 100  75  40 Fenaux et al  
 79  65  71  84  74  GFCH 
 90  57  89  96  96  UKALL XA 
Median CR   duration (mo)  9  5  11 10  17  TIWCL 
 17  NA  6  15.5  Fenaux et al  
 NA  NA NA  NA  NA  GFCH 
 NA  NA  NA NA  NA  UKALL XA  
Median survival   time (mo)  24  7  10  8  21  TIWCL 
 NA  NA  NA  NA NA  Fenaux et al  
 NA  NA NA  NA  NA  GFCH 
 NA  NA  NA NA  NA  UKALL XA  
Median DFS (mo) 8  2  6  6  13  TIWCL 
 NA NA  NA  NA  NA  Fenaux et al  
 24  4  7  10  8  GFCH  
 18  13  32  36  UKALL XA 
Karyotype References
Normal <46 46 abnl 47-50 >50
FAB category (%)  
 L1  36  11  14  30  TIWCL 
 47  100  50  40  20  Fenaux et al* 
 71  48  53  33  60  GFCH  
 NA NA  NA  NA  NA  UKALL XA  
 L2  62  89  86  70  100  TIWCL 
 53 0  50  60  80  Fenaux et al* 
 NA NA  NA  NA  NA  GFCH 
 NA  NA  NA NA  NA  UKALL XA  
LSM (%)  
 Pre-B 62  75  69  71  100  TIWCL 
 77  66  075  100  Fenaux et al* 
 65  92  78 67  100  GFCH  
 75  91  71  75  96 UKALL XA  
 T  38  25  31  29  0  TIWCL 
 23  100  33  25  0  Fenaux et al* 
 35 5  19  33  0  GFCH  
 25  9  29 25  4  UKALL XA  
CR rate (%)  86  50  67  67 78  TIWCL 
 63  0 100  75  40 Fenaux et al  
 79  65  71  84  74  GFCH 
 90  57  89  96  96  UKALL XA 
Median CR   duration (mo)  9  5  11 10  17  TIWCL 
 17  NA  6  15.5  Fenaux et al  
 NA  NA NA  NA  NA  GFCH 
 NA  NA  NA NA  NA  UKALL XA  
Median survival   time (mo)  24  7  10  8  21  TIWCL 
 NA  NA  NA  NA NA  Fenaux et al  
 NA  NA NA  NA  NA  GFCH 
 NA  NA  NA NA  NA  UKALL XA  
Median DFS (mo) 8  2  6  6  13  TIWCL 
 NA NA  NA  NA  NA  Fenaux et al  
 24  4  7  10  8  GFCH  
 18  13  32  36  UKALL XA 

For each karyotype group the percentage of patients who had each characteristic is given, as is the response and survival.

Abbreviations: abnl, abnormal; NA, not available; LSM, lymphocyte surface marker; TIWCL, Third International Workshop on Chromosomes in Leukemia (refs 6, 18, 25, 36, 37); Fenaux et al (ref 8); GFCH, Groupe Français de Cytogénétique Hématologique (ref 7); UKALL XA, MRC Adult Leukaemia Working Party (ref 10).

*

Low patient numbers.

Based on one patient only.

Data included from Bloomfield et al (ref 18, 25).

The less favorable prognosis in adult ALL with hyperdiploid karyotype, compared with children, may be explained by the higher prevalence of associated unfavorable structural changes. The GFCH study, for example, reported the additional presence of Ph in 11 of 30 patients with hyperdiploid (>50) karyotypes.7 The UKALL XA trial showed that the prognostic impact of structural changes overrode the prognostic significance of ploidy groups. Poor-risk structural rearrangements (such as Ph) confer a bad outcome even if they occur in otherwise good-risk ploidy groups.10 

Hypodiploidy

Modal chromosome numbers of 45 and less are rare, particularly the nearly haploid numbers of 24 to 36. In most series of adult patients, hypodiploid chromosome numbers were found in 2% to 8% (Table 1). Sandberg33 compiled, from various references, 26 patients with near-haploid or severe hypodiploid leukemias, including seven adults. The most common losses involved chromosomes 1, 5, 6, 10, 11, 18, 19, 21, and 22. Interestingly, these were the same chromosomes involved in the hyperdiploid karyotypes.6,7,33 The reason for this preference is not known. Near-haploid cases almost always have only numerical changes, whereas cases with modal numbers of 30 to 44 show frequent structural aberrations, mainly translocations.6,34 In nearly all hypodiploid cases analyzed by Rieder et al,9 additional structural abnormalities were identified (half of them involving the Ph).

How massive chromosome losses contribute to leukemogenesis is unclear. Oshimura et al35 proposed multipolar mitosis with subsequent misdivision as a possible explanation. Conceivably, a near-haploid karyotype allows expression of recessive genes that would otherwise have been under the dominance of their allelic counterparts, with loss of regulatory control of growth and differentiation of lymphoid cells.33 Many ALL patients also have a second hyperdiploid population with 52 to 56 chromosomes, twice the near-haploid number, at some time in their disease course, possibly arising by secondary endoreduplication.34 In these instances hyperdiploidy may be an expression of clonal evolution from a near-haploid stem line.33 

ALL associated with a hypodiploid karyotype is usually of a precursor B-cell type, although in the TIWCL series, 20% of hypodiploid cases had T-cell ALL.6 Median initial WBC and percentage of blasts were higher than with diploid or hyperdiploid karyotype, and morphology was predominantly FAB L2.6 Hypodiploid adult patients had the worst 3-year survival in the UKALL XA trial, and hypodiploidy was shown to have prognostic significance independent of other important variables (including the combination of age, sex, and WBC).10 Like hyperdiploidy, hypodiploidy appeared to have more effect on outcome in children than in adults, where data are still limited.25 Nevertheless, hypodiploidy conferred poor prognosis in most studies of adult ALL, comparable to other poor-risk chromosomal translocations such as t(4;11) and t(1;19) (Table2).7,10,18 

Pseudodiploidy

A normal chromosome number with structural changes is the most frequently found abnormal karyotype in adult ALL, and provides for the most heterogeneous group of patients (Table 1). The GFCH showed a pseudodiploid karyotype in 59% of ALL patients, 60% of whom had recurrent translocations (Ph in 40%). This group was also characterized by the highest peripheral WBC counts.7 The TIWCL study found that, compared to other karyotype groups, the group with pseudodiploidy had increased ratios of adults to children, and of L2 to L1 morphology, as well as high initial leukocyte and blast counts.36 The pseudodiploid group was also notable for having the highest percentage of T-cell ALL.37 Most cases had structural changes, with only a few combining structural and numerical aberrations. Chromosomes frequently involved were 1, 6, 9, and 14 but included every autosome with the exception of 4, 16, 18, and 21.36 In the UKALL XA trial, pseudodiploidy was observed in association with translocation t(9;22), with abnormalities of 6q or 9p, or, less often, with translocations t(4;11) and t(1;19).10In some studies, initial leukemic cell burden was high in pseudodiploid cases, which was reflected by increased leukocyte counts and LDH levels.6,14 The poor prognosis associated with pseudodiploidy (Table 2) is likely a reflection of structural rearrangements and other poor-risk features. More accurate identification of underlying specific structural abnormalities may soon make the pseudodiploid group redundant in a prognostically useful classification.

Single Chromosomal Gains or Losses

Nonrandom single chromosome gains or losses occur frequently in ALL,38 although their incidence is lower than in myeloid leukemias.34,39-42 Rarely are they the sole karyotypic abnormality.38 The mechanisms by which such changes contribute to leukemogenesis are unknown. Heim and Mitelman43 offered two possible explanations: a dose effect, with abundance of certain gene products resulting in abnormal proliferation or differentiation, or a duplication of a small genetic defect with oncogenic potential. Most reports on single chromosome gains or losses have been published in childhood ALL, mainly with trisomy 8, monosomy 20, and trisomy 21.34,42,44-46 Only in 10% to 20% of these were the trisomy or monosomy the only karyotypic anomaly, and no particular distinguishing characteristics could be observed.

The TIWCL demonstrated trisomy 21 as the most frequent chromosomal gain in ALL, and further studies confirmed its relatively high incidence, but mainly as part of other cytogenetic changes such as hyperdiploidy.36,47 Trisomy 8 as the only karyotypic change is a frequent occurrence, mainly in myeloid leukemias.48Garipidou et al44 estimated its incidence in ALL to be 1% to 2%. It is indicative of a poor prognosis in acute myeloid leukemias (AML), but its prognostic significance in either adults or children with ALL is not established.49 

Trisomy 4 has been observed in a broad range of hematologic malignancies,38,50 but is rare in ALL51-53 and, as the sole abnormality, tends to be associated with myeloid leukemias, while no such association existed when there were additional cytogenetic changes.

The first report of trisomy 5 in ALL, by Sandberg et al,54was of a 26-year-old male patient with B-lineage ALL. Nagesh Rao et al55 described a 24-year-old woman with T-lymphoblastic lymphoma and trisomy 5 as the sole cytogenetic abnormality. Their review of the literature found only five cases in addition to the case reported by Sandberg et al: 3 AML, 1 non-Hodgkin's lymphoma, and 1 case of Hodgkin's disease. Chen et al56 delineated certain regions on chromosome 5, such as 5p13 and 5q11-31, that were supposedly more specific to ALL.

Monosomies of chromosomes 5 and 7 through deletion or losses of chromosomal material are frequently found in AML and myelodysplastic syndromes. However, their occurrence and significance is not well established in ALL. Dabaja et al57 analyzed 468 adult patients with ALL and found abnormalities of chromosome 5 in 3 and of chromosome 7 in 31 patients; Ph was an associated abnormality in one third. This association translated into lower CR and 3-year survival rates compared with patients who did not have abnormalities of chromosomes 5 or 7. However, when patients with −5 and −7, but without Ph were considered, clinical outcome was not different. The GFCH reported 45 cases with monosomy 7 among 443 adult patients. It was the sole abnormality in only one patient with T-cell ALL.7Rieder et al9 reported only 1 case out of 100 with loss of chromosome 7 as the sole cytogenetic abnormality, confirming the rarity of this karyotype.

Monosomy 20 rarely presents as the single anomaly in ALL. Most studies refer to pediatric patients but occasionally describe this finding in adults, where it correlated with FAB L1 morphology, although this association was not consistent.45,58 

Overall, the significance of monosomies or trisomies as isolated karyotypic changes is unclear, and no specific disease characteristics have been established. Furthermore, reports of single chromosomal gains or losses in adults were too sporadic to draw conclusions of clinical relevance.

More than 30 different nonrandomly occurring rearrangements are presently known in ALL (Table3).34 The GFCH found structural abnormalities in 78% of cases distributed across all ploidy groups.7 Translocations constituted the most common changes. They were found in 30% to 37% of adult cases, with the t(9;22) translocation being the most frequent.7,33,37 The TIWCL showed significant differences between chromosome groups regarding central nervous system (CNS) involvement, leukocyte and blast counts, FAB morphology, and immunophenotype.37 In adults, translocation t(4;11) and the Ph karyotypes were associated with higher leukocyte and blast counts than were other chromosomal rearrangements. Translocations t(8;14) and t(4;11), as well as 14q+, correlated with a higher risk of CNS involvement. Both children and adults with chromosomal translocations had worse survival than those with normal karyotypes. In children the presence of cytogenetic abnormalities other than translocations was associated with outcome similar to normal karyotypes, but in adults all structural abnormalities adversely influenced survival.

Table 3.

Nonrandom Structural Chromosomal Aberrations in Adult ALL

Chromosome Aberration Involved Genes Protein ProductFunction of Protein Product Frequency PhenotypeFAB-Morphology References
del(6q)  ?  ?  ?  <5%  B- or T-lineage  L1, L2  6, 7, 10, 18, 25, 28, 33, 240-247  
i(6p)  ?  ?  ?  <5%    34, 248-251  
i(7q)  ?  ?  ?  <5%    34, 248-252  
7q32-35 TCR-β    <5%  T-lineage  L1 > L2  6, 7, 157, 168-193  
t(1;7)(p32;q35)  TAL1/TCR-β  Basic HLH protein  Transcription factor  
t(1;7)(p34;q34) LCK/TCR-β  Protein kinase  Signal transduction 
t(1;7)(q11-21;  q35-36)  ?  ?  ?  
t(7;9)(q34;q32)  TAL2/TCR-β  Basic HLH protein Transcription factor  
t(7;9)(q34;p34)  TAN1/TCR-β Drosophila notch  Signal transduction  
  Homologue  
t(7;10)(q35;q24)  RHOM2/RCR-β  LIM domain protein  Transcription factor  
t(7;11)(q35;p13) RHOM2/TCR-β  LIM domain protein  Transcription factor 
 LYL1/TCR-β  Basic HLH protein  Transcription factor  
8q24  c-myc Basic HLH protein Transcription factor  <3-5%   B  L3  5-8, 18, 25, 33, 34, 36, 37, 151, 171, 194-208  
t(2;8)(p12;q24) lgκ/c-myc 
t(8;14)(q24;q32)  c-myc/lgH 
t(8;22)(q24;q11)  c-myclgλ  
i(9q)  ?  ?  ?  <5%  Pre-B  L2  34, 248-251 
t(9;22)(q34;q11)  BCR, ABL e1a2 b2a2/b3a2 e19a2  p190 p210 p230   Increased tyrosine kinase activity (? p23)   20-30%  Pre-B L1, L2  7, 8, 36, 54, 59-87  
del(9q)(p21-22) MTS1/MTS2  p16INK4A/p15INK4B Cyclin-dependent kinase inhibitors    7-15%  B- or T-lineage  L1, L2  7, 92-119 
t/dic(9;12)(p11-12; p11-13)  ?  ?  ?  <5% Early-pre-B or pre-B  L1, L2  22, 34, 120-123 
11q23 t(1;11)(p32;q23) t(4;11)(q21;q23) t(10;11)(p12-14; q14-23) t(11;19)(q23;  p13)  MLL (HRX, ALL-1, HTRX-1) AF1P/MLL AF4/MLL AF10/MLL MLL/ENL  MLL fusion proteins with conservation of 5′-N-terminal sequences including AT-hook and DNA-methyltransferase regions   Probably involved in transcriptional regulation    3-10%   Early-pre-B T-lineage biphenotypic    L1 L1, L2 L2 L1, ,L2  7, 9, 18, 33, 90, 124-143  
12q t(9;12)(q34;p13) t(12;21)(p11-12;  q22)  TEL (ETV6) ABL/TEL TEL/AML1  HLH protein tyrosine kinase runt/transactivating domain   Phosphorylation Transcription regulation  3-4%  pre-B  L1, L2  7, 10, 24, 209-239 
14q11  TCR-α/δ     T-lineage  L1 > L2 6, 7, 157, 168-193  
t(1;14)(p32-34;q11)  TAL1/TCR-δ Basic HLH protein  Transcription factor  ∼20% 
t(5;14)(p15;q11)  ?  ?  ?  
t(8;14)(q24;q11)  c-MYC/TCR-α  Basic HLH protein Transcription factor  <1%  
t(10;14)(q24;q11) HOX11/TCR-δ  Homeodomain protein  Transcription factor  3-7%  
t(11;14)(p13;q11)  RHOM2/TCR-δ  LIM domain protein  Transcription factor   5-7% 
t(11;14)(p15;q11)  RHOM1/TCR-δ  LIM domain protein Transcription factor  <1%  
inv(14)(q11;q32.1) TCL1/TCR-α  ?  ?  <1%  
inv(14)(q11;q32.3) IgH/TCR-α/δ  Ig  ?  <1%  
i(17q)  ?  ?  ?   7-9%  Pre-B   34, 248-251, 253  
19q13 t(1;19)(q23;p13) t(17;19)(q22;p13) E2A PBX1/E2A HLF/E2A Fusion proteins with preservation of E2A activation domains  Transcription factor  <5%   Pre-B Pre-B    L1 L1  7, 8, 15, 29, 33, 34, 90, 145-166  
i(21)q  ?  ?  ?  <5%  B-lineage   34, 248-251 
Chromosome Aberration Involved Genes Protein ProductFunction of Protein Product Frequency PhenotypeFAB-Morphology References
del(6q)  ?  ?  ?  <5%  B- or T-lineage  L1, L2  6, 7, 10, 18, 25, 28, 33, 240-247  
i(6p)  ?  ?  ?  <5%    34, 248-251  
i(7q)  ?  ?  ?  <5%    34, 248-252  
7q32-35 TCR-β    <5%  T-lineage  L1 > L2  6, 7, 157, 168-193  
t(1;7)(p32;q35)  TAL1/TCR-β  Basic HLH protein  Transcription factor  
t(1;7)(p34;q34) LCK/TCR-β  Protein kinase  Signal transduction 
t(1;7)(q11-21;  q35-36)  ?  ?  ?  
t(7;9)(q34;q32)  TAL2/TCR-β  Basic HLH protein Transcription factor  
t(7;9)(q34;p34)  TAN1/TCR-β Drosophila notch  Signal transduction  
  Homologue  
t(7;10)(q35;q24)  RHOM2/RCR-β  LIM domain protein  Transcription factor  
t(7;11)(q35;p13) RHOM2/TCR-β  LIM domain protein  Transcription factor 
 LYL1/TCR-β  Basic HLH protein  Transcription factor  
8q24  c-myc Basic HLH protein Transcription factor  <3-5%   B  L3  5-8, 18, 25, 33, 34, 36, 37, 151, 171, 194-208  
t(2;8)(p12;q24) lgκ/c-myc 
t(8;14)(q24;q32)  c-myc/lgH 
t(8;22)(q24;q11)  c-myclgλ  
i(9q)  ?  ?  ?  <5%  Pre-B  L2  34, 248-251 
t(9;22)(q34;q11)  BCR, ABL e1a2 b2a2/b3a2 e19a2  p190 p210 p230   Increased tyrosine kinase activity (? p23)   20-30%  Pre-B L1, L2  7, 8, 36, 54, 59-87  
del(9q)(p21-22) MTS1/MTS2  p16INK4A/p15INK4B Cyclin-dependent kinase inhibitors    7-15%  B- or T-lineage  L1, L2  7, 92-119 
t/dic(9;12)(p11-12; p11-13)  ?  ?  ?  <5% Early-pre-B or pre-B  L1, L2  22, 34, 120-123 
11q23 t(1;11)(p32;q23) t(4;11)(q21;q23) t(10;11)(p12-14; q14-23) t(11;19)(q23;  p13)  MLL (HRX, ALL-1, HTRX-1) AF1P/MLL AF4/MLL AF10/MLL MLL/ENL  MLL fusion proteins with conservation of 5′-N-terminal sequences including AT-hook and DNA-methyltransferase regions   Probably involved in transcriptional regulation    3-10%   Early-pre-B T-lineage biphenotypic    L1 L1, L2 L2 L1, ,L2  7, 9, 18, 33, 90, 124-143  
12q t(9;12)(q34;p13) t(12;21)(p11-12;  q22)  TEL (ETV6) ABL/TEL TEL/AML1  HLH protein tyrosine kinase runt/transactivating domain   Phosphorylation Transcription regulation  3-4%  pre-B  L1, L2  7, 10, 24, 209-239 
14q11  TCR-α/δ     T-lineage  L1 > L2 6, 7, 157, 168-193  
t(1;14)(p32-34;q11)  TAL1/TCR-δ Basic HLH protein  Transcription factor  ∼20% 
t(5;14)(p15;q11)  ?  ?  ?  
t(8;14)(q24;q11)  c-MYC/TCR-α  Basic HLH protein Transcription factor  <1%  
t(10;14)(q24;q11) HOX11/TCR-δ  Homeodomain protein  Transcription factor  3-7%  
t(11;14)(p13;q11)  RHOM2/TCR-δ  LIM domain protein  Transcription factor   5-7% 
t(11;14)(p15;q11)  RHOM1/TCR-δ  LIM domain protein Transcription factor  <1%  
inv(14)(q11;q32.1) TCL1/TCR-α  ?  ?  <1%  
inv(14)(q11;q32.3) IgH/TCR-α/δ  Ig  ?  <1%  
i(17q)  ?  ?  ?   7-9%  Pre-B   34, 248-251, 253  
19q13 t(1;19)(q23;p13) t(17;19)(q22;p13) E2A PBX1/E2A HLF/E2A Fusion proteins with preservation of E2A activation domains  Transcription factor  <5%   Pre-B Pre-B    L1 L1  7, 8, 15, 29, 33, 34, 90, 145-166  
i(21)q  ?  ?  ?  <5%  B-lineage   34, 248-251 

Translocation t(9;22)(q34;q11)

In 1960 Nowell and Hungerford59 discovered the Ph as a distinct chromosomal abnormality in chronic myeloid leukemia (CML). It was the first chromosomal abnormality to be associated with a specific malignant disease in humans, and became a karyotypic hallmark of CML. In 1970 Propp and Lizzi60 reported a 53-year-old patient with ALL who had the classic Ph in a high percentage of marrow cells. It is now well established that a t(9;22) translocation can be observed in up to 95% of patients with CML, in about 1% to 2% of patients with AML, as well as in up to 5% of children and 15% to 30% of adults with ALL, making it the most common ALL-associated chromosomal abnormality in the latter group.61-64 

The Ph is a shortened chromosome 22 that results from a reciprocal translocation between the long arms of chromosomes 9 and 22 (Fig 1).62 This transposes the large 3′ segment of the c-ABL gene from chromosome 9 to the 5′ part of the BCR gene on chromosome 22, creating a hybridBCR-ABL gene that is transcribed into a chimericBCR-ABL mRNA.65,66 The breakpoint in theABL region can occur anywhere in a 300-kb intron, but is usually 5′ of ABL exon a2. Deletions of ABL exon a2 and in-frame joining at the mRNA level of 5′ BCR sequences to theABL exon a3 have also been described.67 In almost all cases of CML, as well as in about half of adult Ph+ALL, the ABL gene from chromosome 9 transposes into the major breakpoint cluster region (M-bcr) on chromosome 22, spanning exons 12 to 16 (historically named b1 to b5), giving rise to BCR-ABLfusion transcript mRNAs with a b2a2 or b3a2 junction. The fusion mRNAs translate into a fusion protein of 210 kD called p210BCR-ABL.68 In very rare cases of CML or AML, but in about 50% of adult Ph+ ALL and in 80% of childhood Ph+ ALL, the breakpoint on chromosome 22 falls 5′ of the M-bcr, within a long intron segment separating the alternative exon e2′ from e2 called the minor breakpoint cluster region (m-bcr).69 Splicing out exons e1′ and e2′ creates an e1a2 junction in the BCR-ABL transcript, translating into a smaller BCR-ABL fusion protein called p190BCR-ABL. Both p210BCR-ABL and p190BCR-ABL have significantly increased tyrosine phosphokinase activity compared to the normal human c-ABL protein, p145. Saglio et al70 described a novel position for a breakpoint in the BCR gene that is located in a 3′ direction from the M-bcr region, between exons e19 and e20 (historically c3 and c4), creating a new fusion transcript with an e19a2 junction. This transcript contains a significantly larger part of the BCR gene and codes for the p230BCR-ABL fusion protein. In other cases, complex variant translocations involve three or more chromosomes, mask the Ph marker, or a fusion gene is created by the insertion of ABL sequences into a normal appearing chromosome 22.

Fig. 1.

Translocation t(9;22)(q34;q11) and fusion products. The Ph is a shortened chromosome 22 that results from the transposition of 3′-ABL segments on chromosome 9 to 5′-BCR segments on chromosome 22. Whereas breakpoint locations on chromosome 9 appear rather constant 5′ of ABL exon a2, several breakpoint cluster regions have been identified along the BCR gene on chromosome 22. Depending on which breakpoints are involved on chromosome 22, differently sized segments from the BCR gene are joined together with the 3′-sequences of the ABL gene. The translocation thus results in fusion mRNA molecules of different length (e1a2, b2a2, b3a2, e19a2) and, subsequently, chimeric protein products with variable molecular weights and presumably function (p190, p210, p230) (see text for details).

Fig. 1.

Translocation t(9;22)(q34;q11) and fusion products. The Ph is a shortened chromosome 22 that results from the transposition of 3′-ABL segments on chromosome 9 to 5′-BCR segments on chromosome 22. Whereas breakpoint locations on chromosome 9 appear rather constant 5′ of ABL exon a2, several breakpoint cluster regions have been identified along the BCR gene on chromosome 22. Depending on which breakpoints are involved on chromosome 22, differently sized segments from the BCR gene are joined together with the 3′-sequences of the ABL gene. The translocation thus results in fusion mRNA molecules of different length (e1a2, b2a2, b3a2, e19a2) and, subsequently, chimeric protein products with variable molecular weights and presumably function (p190, p210, p230) (see text for details).

Close modal

Traditional karyotypic studies underestimate the incidence of theBCR-ABL fusion gene, and Ph cytogenetics with positive molecular tests for the BCR-ABL fusion gene have been documented.62,71-73 Molecular tools for the detection of the BCR-ABL fusion gene include fluorescent in situ hybridization (FISH),74 polymerase chain reaction (PCR),75 and pulsed-field gel electrophoresis,76 which are complementary to cytogenetic analysis.73 Southern blot analysis, as it is applied in CML, is inadequate as a single diagnostic tool for Ph+ ALL, since it detects p210BCR-ABL but not p190BCR-ABL which is found in 50% to 80% of Ph+ ALL, because breakpoints on the BCR gene occur outside the M-bcr region in an area too large to be reliably recognized.71 

The variability of the breakpoint locations within the BCRgene, and the fact that p190BCR-ABL is predominantly associated with ALL, while p210BCR-ABL is most consistently associated with CML suggests a distinction between true de novo ALL (m-bcr, p190BCR-ABL) from CML in lymphoid blastic phase (M-bcr, p210BCR-ABL). There is also a difference in the frequency of expression of p190BCR-ABL between pediatric and adult ALL (80% v 50% of Ph+ ALL cases).77,78 In vitro studies showing that p190BCR-ABL is a more active tyrosine kinase than is p210BCR-ABL also suggest that these entities are different on the basis of their breakpoint location.77,79,80 The relation among these disease groups is still not well understood.81 Melo61 reported that the location of the breakpoint in the BCR gene (and/or possibly in the ABL gene) determined the disease phenotype. The hybrid gene product p190BCR-ABL may function preferentially during lymphoid and monocytic differentiation, while p210BCR-ABLcontains additional BCR-sequences that appear to affect pathways common to both ALL and CML precursors. However, p190BCR-ABLtranscripts are not confined to primary acute leukemias. Using quantitative reverse transcription-PCR (RT-PCR) for p210BCR-ABL and p190BCR-ABL mRNA, Van Rhee et al,82 in a study of adult leukemias, detected p190BCR-ABL mRNA in 88% (14 of 16) of patients with CML in chronic phase, 100% (10 of 10) of patients with CML in lymphoid blastic phase, and 100% (10 of 10) of cases with p210BCR-ABL-positive ALL. However, the p190BCR-ABL/ p210BCR-ABL ratio was 10 times greater in ALL than CML, whereas the ratio was similar in CML at diagnosis, in chronic phase, and in lymphoid blastic phase.

Several studies reported on the association between breakpoint location and clinical features and prognosis in Ph+ALL.79,80 Secker-Walker et al80 studied 113 adult patients with Ph+ ALL and found no significant difference between those with an M-bcr breakpoint and those with a different breakpoint location for age, immunophenotype, or outcome. Only the peripheral WBC count was significantly higher with an M-bcr breakpoint. Likewise, Kantarjian et al77 found no significant clinical, laboratory, or karyotypic differences in patients with p210BCR-ABL versus p190BCR-ABL ALL. Patients with either abnormality had similar incidences of older age, organomegaly, anemia, leukocytosis, thrombocytopenia, and blastosis. Although patients with p190BCR-ABL disease showed a trend for longer CR duration, overall survival was not influenced by the expression of either p190BCR-ABL or p210BCR-ABL. When maintenance therapy was not given after CR, some patients with p210BCR-ABL disease demonstrated a “second chronic phase” CML-like hematologic picture in the blood.77 

In patients treated with bone marrow transplantation (BMT) for Ph+ ALL, a different clinical behavior was reported for Ph+ ALL expressing p190BCR-ABL compared with p210BCR-ABL. In a study of 36 patients (29 adults) with Ph+ ALL, Radich et al83 showed that detection of BCR-ABL by PCR after BMT correlated with a high risk of relapse, and that the expression of p190BCR-ABL was associated with a higher risk of relapse than was the expression of p210BCR-ABL. All of the relapsed patients were adults.

Adult patients with Ph+ ALL tended to be older, had higher WBC and blast counts and, in some studies, more frequent lymphadenopathy and splenomegaly than Phpatients.84,85 Secker-Walker et al80 found that 44% of ALL patients older than 50 years were Ph+. Almost all cases of Ph+ ALL had a pre-B phenotype,7,8,36,84 and other immunophenotypes were rare.7,36,86,87 Increased expression of CD10 (“common ALL antigen” or CALLA) was found with Ph+ ALL, and in CALLA+ cases BCR-ABL identified a group of patients with short remission duration and poor DFS.73,84 Myeloid markers were present in 40% to 65% of Ph+ ALL cases.6,86 

In children, Ph+ ALL has a dismal prognosis even with intensive chemotherapy programs that have improved survival in other cytogenetic subgroups.15,18,88,89 A similarly poor prognosis was reported in adults where, despite high remission rates comparable to those of Ph ALL, remission duration and survival times were short (Table4).25,54,73,84,90 Therefore, patients with Ph+ALL are suitable candidates for innovative and intensified strategies. In younger patients, allogeneic BMT with a related or unrelated donor, or peripheral blood stem cell transplantation during first CR are indicated, although relapse rates tend to be high ranging from 40% to 80%, and disease recurrence after BMT has a particularly devastating prognosis in adults.83,91 In cases where no match can be found or marrow transplantation is not an option for other reasons, high-dose ara-C containing regimens can achieve considerable response rates.84 Other treatment alternatives include interferon, adoptive immunotherapy, and antibody-based therapies.83Unfortunately, to date, no satisfying track-record of durable responses has been established with any of these approaches.

Table 4.

Characteristics and Clinical Outcomes of Ph+ Adult ALL

No. Ph (%) Median Age (yr) Median WBC Count CR (%) CRD (mo) MST (mo) References
334  12  46% > 50  34% > 50 56  9  11  Preti et al84  
15  50 26.5  89.5  50  5-10  7.5  Bloomfield  et al85  
113  20  39.7  73.3  63.6 NA  7  Secker-Walker  et al80  
56 30  39  26.3  71  10  11.2  Westbrook  et al73  
56  26.8  39  123-150 73.33-150 NA  Specchia  et al86  
443  29 45  30  59   NA3-151 NA GFCH7  
58  21  51  57  75  8.5 NA  Fenaux et al8  
350  11 NA  NA  83  NA  NA3-152 UKALL XA10 
172  17 35  24  46  7  11 TIWCL18,373-153 
No. Ph (%) Median Age (yr) Median WBC Count CR (%) CRD (mo) MST (mo) References
334  12  46% > 50  34% > 50 56  9  11  Preti et al84  
15  50 26.5  89.5  50  5-10  7.5  Bloomfield  et al85  
113  20  39.7  73.3  63.6 NA  7  Secker-Walker  et al80  
56 30  39  26.3  71  10  11.2  Westbrook  et al73  
56  26.8  39  123-150 73.33-150 NA  Specchia  et al86  
443  29 45  30  59   NA3-151 NA GFCH7  
58  21  51  57  75  8.5 NA  Fenaux et al8  
350  11 NA  NA  83  NA  NA3-152 UKALL XA10 
172  17 35  24  46  7  11 TIWCL18,373-153 

Abbreviation: NA, not available.

F3-150

Not significantly different from Ph ALL.

F3-151

Five months median DFS.

F3-152

13% DFS at 3 years.

F3-153

Includes data from Bloomfield et al.18 

Abnormalities of the Short Arm of Chromosome 9

Loss or mutation of 9p21-22.

In 1983 Kowalczyk and Sandberg92 described 7 children with ALL, 5 with deletions of the short arm of chromosome 9 (9p−) and 2 with total loss of chromosome 9. In all cases segment 9p21-pter was missing. Compared with other children with ALL, the group had distinct clinical features such as older age, prominent lymphadenopathy and splenomegaly, and high WBC count and percentage of blasts. Four of the seven children had T-cell ALL. Median survival time for children with 9p− abnormalities was 1 year shorter than for children without these anomalies. In 1985 Chilcote et al93reported 65 patients with ALL (41 children and 24 adults). They singled out 8 patients (4 children and 4 adults) with “lymphomatous” features and showed that 6 of them (3 children and 3 adults) had loss of bands p21-p22 on the short arm of chromosome 9, whereas these bands were missing in only 1 out of the remaining 57 cases. The mechanisms involved included deletions, unbalanced translocations, or loss of the entire chromosome. The outcome for these patients was poor, as in the Kowalczyk and Sandberg study. The investigators hypothesized a possible suppressor gene in or near 9p21-22 that is involved in the control of proliferation of lymphoid precursors. Finally, in 1987 Pollack and Hagemeijer94described 32 patients (18 adults and 14 children) with a 9p− karyotype, including 20 with ALL (10 adults and 10 children). Among these, 3 adults showed 9p− as the sole karyotypic anomaly, whereas none of the children had 9p− without other chromosomal changes. On average, the karyotypes in children appeared more complex than in adults. However, no association of 9p− with T-cell ALL or “lymphomatous” features was seen. Other studies have confirmed that 9p anomalies are more likely in ALL than in other hematological malignancies, with a reported frequency of 7% to 13%, and no apparent difference in childhood versus adult ALL.92,93,95-97 An association with high-risk clinical features such as older age, higher leukocyte and blast counts, bulky disease, increased incidence of CNS disease, or T-cell immunophenotype was found in some, but not in all studies.

The smallest segment lost included 9p21. Specific deletions of 9p21 have also been described in a variety of human, rodent, and simian cancer cell lines, pointing at 9p21 as a possible location for a tumor suppressor gene.98,99 Detailed molecular analyses of this location, searching for a possible tumor suppressor gene, have been performed with a multitude of different probes.100 Trent et al101 have localized the interferon gene cluster to 9p21, and the assignment by Carrera et al102 of the methyladenosine phosphorylase (MTAPase) gene, coding for an essential enzyme in the purine salvage pathways, to the same region assumed new clinical relevance. Using molecular analysis, the interferon gene cluster was shown to be deleted in 43% of leukemia-derived cell lines and in 29% of primary leukemia samples,95,103 whereas MTAPase deficiency could be demonstrated in 10% of leukemias and, in particular, in 38% of cases of T-cell ALL.104 Kamb et al105 and Nobori et al106 analyzed 9p21 deletions in detail and localized two previously cloned genes to the 9p21 segment: p16INK4A and the structurally homologous gene, p15INK4B, located only 25 kb centromeric to p16INK4A. They named these genes Multiple Tumor Suppressor Gene 1 (MTS1) and Multiple Tumor Suppressor Gene 2 (MTS2), respectively, as they could demonstrate high frequencies of homozygous deletions in a variety of human tumor cell lines. Kamb et al105 also studied four leukemia cell lines and found deletions within 9p21 in one. Nobori et al106 analyzed 14 leukemia cell lines by PCR and foundMTS1 deletions in 9 of them (64%), with deletions of the MTAPase and interferon genes in 29% and 50% of cases, respectively. Both MTS1 and MTS2 encode for proteins that inhibit the cyclin-dependent kinases CDK4 and CDK6 and play a crucial role in cell cycle progression,100,107,108 which makes them ideal candidates for a putative tumor suppressor gene at 9p21.

The main mechanism for p16INK4A inactivation is biallelic deletions, with p15INK4B gene codeletions in most but not all cases (Table5).108 Current data suggest thatp16INK4A is the primary target of such deletions. This is underscored by the fact that in most studiesp15INK4B deletions occur only inp16INK4A-deleted cases, whereas the converse does not.109-112 This has also been shown forinterferon-α gene deletions.113 The frequency of homozygous deletions of theinterferon-α gene is lower than that forp16INK4A in one published report.103 

Table 5.

p16INK4A/p15INK4B Deletions (in %) in ALL

References p16 Ho p16 He p16 Mu p15 DelComments
Hebert et al109 and Cayuela et al110 45  NA  NA  36 Adults and children Strong correlation of p16 and p15 deletion with T-cell ALL  
Cayuela et al97 76  10  50 (2/4 He) NA  Adults and children  
     Only T-cell samples analyzed  
Quesnel  14  NA  1.1 (1/88)  NA  Mainly adults  
  et al113      No association with T-cell ALL  
     Associated with poor prognostic features  
     33% IFN-α gene deletion in p16-deleted cases  
Rasool et al114  27  NA  11 (1/9)  Children  
     9 hemizygous deletions for 9q21 markers  
     No association with T-cell ALL  
Haidar et al111  22  0  NA 11  Adults  
     No association with T-cell ALL  
Ogawa et al112  27  16  0  34 Mainly adults  
     p16 deletions more frequent in pre-B ALL  
     All patients with loss of p15 also had loss of p16  
     IFN-α gene loss only with loss of p16  
Dreyling et al115  45  45  0  45  Only adult data presented, children omitted  
Schröder et al118  17  10  NA  NA Adults and children  
     p16 deletions in 1/23 B-lineage and 6/7 T-cell ALL 
References p16 Ho p16 He p16 Mu p15 DelComments
Hebert et al109 and Cayuela et al110 45  NA  NA  36 Adults and children Strong correlation of p16 and p15 deletion with T-cell ALL  
Cayuela et al97 76  10  50 (2/4 He) NA  Adults and children  
     Only T-cell samples analyzed  
Quesnel  14  NA  1.1 (1/88)  NA  Mainly adults  
  et al113      No association with T-cell ALL  
     Associated with poor prognostic features  
     33% IFN-α gene deletion in p16-deleted cases  
Rasool et al114  27  NA  11 (1/9)  Children  
     9 hemizygous deletions for 9q21 markers  
     No association with T-cell ALL  
Haidar et al111  22  0  NA 11  Adults  
     No association with T-cell ALL  
Ogawa et al112  27  16  0  34 Mainly adults  
     p16 deletions more frequent in pre-B ALL  
     All patients with loss of p15 also had loss of p16  
     IFN-α gene loss only with loss of p16  
Dreyling et al115  45  45  0  45  Only adult data presented, children omitted  
Schröder et al118  17  10  NA  NA Adults and children  
     p16 deletions in 1/23 B-lineage and 6/7 T-cell ALL 

Abbreviations: IFN-α, interferon-α; Ho, homozygous deletion; He, hemizygous deletion; Mu, point mutation; del, deletion; NA, not available.

Data analysis similar to Quesnel et al.108 

Point mutations within p16INK4A are rare (Table 5). Quesnel et al113 analyzed p16INK4A gene deletions by Southern blot in 63 ALL patients (61 adults) and found 9 cases with homozygous deletions of p16INK4A (3 T-cell ALL, 6 precursor B-cell ALL). Single-stranded conformation polymorphism (SSCP) analysis of exons 1 and 2 ofp16INK4A was performed in 88 cases of ALL, including the 63 cases mentioned. Only one missense mutation at codon 49 was detected. Inactivation of p16INK4A occurred mainly through deletions of both copies of the gene, whereas deletion of one allele and point mutation of the other (as is frequently observed with the tumor suppressor gene p53) were rare.97,112,114,115 

More recently Herman et al116 and Batova et al117 presented evidence for an additional mechanism of gene inactivation. They showed that the p15INK4Bgene was preferentially hypermethylated at a 5′-CpG island. This was demonstrated in 17 of 45 children with T-cell ALL at diagnosis and in 7 of 32 at relapse. Methylation of the p16INK4A gene was rare; it occurred in only 2 of 49 patients at diagnosis and none at relapse. Methylation correlated with loss of transcription, and the preferred methylation site at p15INK4B in ALL lended strong support for a role of p15INK4Binactivation in the pathogenesis of ALL. Whether a similar mechanism is operative in adult ALL patients remains to be investigated.

Disagreement still exists about whether 9p21 abnormalities correlate with immunophenotypic or prognostic features. Strong correlations with T-cell ALL have been reported by some investigators96,109,110,118 but not by others.111,112 Quesnel et al113 found at least one poor prognostic factor, such as bulky disease or high WBC count, in patients with ALL and p16INK4A gene deletions, and most of these patients relapsed. However, no strong association with T-cell lineage was evident. Likewise, Fizzotti et al119reported a significant relationship betweenp16INK4A/p15INK4B gene deletions and leukemic cell mass and WBC count, but no difference by immunophenotype. The GFCH found 9p21 abnormalities in 15% of ALL patients.7The abnormalities had no effect on prognosis and no significant correlation with T or B lineage. Nevertheless, abnormalities of chromosome 9p21 with deletions of thep16INK4A/p15INK4B region are highly specific to lymphoid tumors and have been among the most consistent genetic defects found in ALL to date.

t/dic(9;12)(p11-12)(p11-13).

This rare group of chromosomal abnormalities represents, for the most part, unbalanced translocations with loss of part of the short arms of chromosomes 9 and 12. Considerable breakpoint variation exists on 9p11, where the translocation can fall anywhere in a 300-kb segment.34 This abnormality was first discovered in 1985 by Heerema et al,22 who described a child with the karyotype 45, XY, −9, −12, +der(12)t(9;12)(q1?;p13). In 1987 Carroll et al120 reported eight children with precursor-B-cell ALL and t/dic(9;12)(p11-13;p11-12); seven were hypodiploid, with loss of both chromosomes 9 and 12 and a remaining der(12)t/dic(9;12), whereas the eighth child retained two normal chromosomes 9, with one normal chromosome 12 and a der(12)t/dic(9;12). In four children this abnormality was the only cytogenetic change. Since then other dicentric translocations involving the short arms of chromosomes 9 and 12 have been described. Larger series were published in 1992 by the United Kingdom Cancer Cytogenetics Group121 and by Mahmoud et al.122 In the latter series 15 of 2,303 children carried a t/dic(9;12). There was a striking association with a pre-B/early-pre-B-cell ALL phenotype, male gender, and an excellent prognosis. In 1995 a dic(9;12) study group analyzed additional 14 cases of dic(9;12)(p11-13;p11-12),123 including 11 cases of ALL with a precursor-B phenotype (5 children and 6 adults). The ALL patients were characterized by young age (median age 15 years, all but one under 25 years), a median WBC count of 6.3 × 109/L, no CNS involvement, predominantly FAB L1 morphology, early pre-B-cell phenotype, positivity for CD10, and an excellent prognosis. Almost all achieved CR, with DFS and overall survivals (OS) of 93% and 97%, respectively, at a median follow-up of 61 months. In 41% of cases dic(9;12) was the sole cytogenetic abnormality, in which cases the dicentric chromosomes resulted in hypodiploidy. Additional numerical abnormalities were found in 34%, with trisomy 8 being the most common.

Behrendt et al123 described an interesting case of relapsed ALL with both dic(9;12) and t(21;21)(q21;q22). Translocations involving 21q22 occur in secondary leukemia patients after treatment with topoisomerase II inhibitors, and the patient had been previously treated with VP16.

Comparable data for adult patients have to be largely extrapolated from the available information in children or adolescents. This chromosomal abnormality is rare in adults, and little is known about its effect on prognosis.

Abnormalities Involving 11q23

Abnormalities of 11q23 are among the most frequent cytogenetic abnormalities in a variety of adult hematopoietic malignancies. They also occur in 60% to 70% of acute leukemias in infants.124-126 Their frequency in older children and adults with ALL is lower (up to 10%).124-126 When there has been previous therapy with topoisomerase II inhibitors, frequencies as high as 80% can be observed.127,128 

The common molecular denominator is the disruption of a gene located at band q23 of chromosome 11. In 1991 Ziemin-van der Poel et al129 and others identified this gene at 11q23 as the “mixed-lineage leukemia,” or “myeloid-lymphoid leukemia,” gene (MLL, also referred to asALL-1, HRX, or HTRX1).129-131 The gene contains at least 21 exons and spans about 100 kb. It encodes a protein of 3,968 amino acids with a molecular weight of 431 kD. It contains two central zinc-finger domains and a 210-amino acid C-terminal and shows significant homology with the Drosophila trithoraxprotein.130,131 At the N-terminal are a methyltransferase domain and three AT-hook motifs through which the protein can bind to AT-rich regions of the minor groove of the DNA double helix.132 

The function of the MLL protein is not entirely clear, but it appears to act as a transcription factor in the regulation of differentiation pathways by direct interaction with DNA or with other DNA-binding proteins.133,134 In 1993 Thirman et al135identified a single complementary DNA (cDNA) probe from the MLLgene that could detect rearrangements with a breakpoint at band 11q23 when DNA from leukemia patients was digested with a single enzyme. The breakpoints cluster within an 8.3 kb-region between exons 5 and 11. This is a comparatively restricted area that can be identified by Southern blot analysis of genomic DNA probed with a small cDNA fragment that overlaps the cluster region.

More than 20 reciprocal chromosomal loci are known to participate in 11q23 translocations, the most common are 4q21, 9p22, 19p13, and 1p32, and many partner genes have been identified (Table 6).133,136 With the exception of AF10 (10p12-14) and AF17 (17q21), as well as ENL (19p13) and AF9 (9p22), no close relation exists among the various partner genes on a structural or functional basis.134 The breakpoints on the reciprocal chromosomes are also scattered over larger segments (in the case of 4q21, over at least 38 kb) than is the case with MLL, giving rise to fusion transcripts that vary in length and exon composition.137Janssen et al125 used RT-PCR to show fusion transcripts containing MLL and AF4, the gene located at 4q21. They demonstrated that a large number of differently sized fusion transcripts was probably resulting from alternative splicing events. Overall they identified eight different MLL-AF4 versions. The fusion genes contain nearly identical 5′ portions of MLLtransposed to various partner genes. The resultant chimeric proteins contain the N-terminal portions of the MLL protein, including the AT-hook motifs and methyltransferase domain, but not the C-terminal segments including the central zinc-finger motifs.133,138 

Table 6.

11q23 Partner Chromosome Breakpoints and Fusion Transcripts in ALL

Chromosome Breakpoint Gene Involved Fusion Transcript Comments
1p32  AF1p  AF1p-MLL  Normally expressed as 4.4 kb transcript in various tissues 
   No known similarity with other genes known to fuse withMLL, but 88% homology with eps15 (refs 133, 134) 
4q21  AF4 (FEL,  MLLT2)  AF4-MLL  Expressed as two 12-kb and 10.5-kb transcripts in various tissues 
   Encodes protein of 140 kD with putative nuclear targeting sequence and consensus sequence for ATP/GTP binding 
   Proline/serine-rich protein  
   Most common 11q23 fusion gene (refs 133, 134)  
10p12-14  AF10 AF10-MLL  5.5-kb mRNA encoding for protein of 1,027 amino acids  
   Contains zinc-finger motif near N-terminus and leucine-zipper motif near C-terminus  
   High degree of similarity with AF17 
   IL-2 gene maps to breakpoint region (refs 133, 134)  
19p13  ENL (MLLT1,  LTG19)  MLL-ENL  4.7-kb mRNA encoding for protein of 559 amino acids  
   Contains putative nuclear targeting sequence and consensus sequence for ATP/GTP binding 
   Proline/serine-rich protein  
   56% identical with AF9 (refs 133, 134) 
Chromosome Breakpoint Gene Involved Fusion Transcript Comments
1p32  AF1p  AF1p-MLL  Normally expressed as 4.4 kb transcript in various tissues 
   No known similarity with other genes known to fuse withMLL, but 88% homology with eps15 (refs 133, 134) 
4q21  AF4 (FEL,  MLLT2)  AF4-MLL  Expressed as two 12-kb and 10.5-kb transcripts in various tissues 
   Encodes protein of 140 kD with putative nuclear targeting sequence and consensus sequence for ATP/GTP binding 
   Proline/serine-rich protein  
   Most common 11q23 fusion gene (refs 133, 134)  
10p12-14  AF10 AF10-MLL  5.5-kb mRNA encoding for protein of 1,027 amino acids  
   Contains zinc-finger motif near N-terminus and leucine-zipper motif near C-terminus  
   High degree of similarity with AF17 
   IL-2 gene maps to breakpoint region (refs 133, 134)  
19p13  ENL (MLLT1,  LTG19)  MLL-ENL  4.7-kb mRNA encoding for protein of 559 amino acids  
   Contains putative nuclear targeting sequence and consensus sequence for ATP/GTP binding 
   Proline/serine-rich protein  
   56% identical with AF9 (refs 133, 134) 

Several mechanisms for MLL gene rearrangements are possible including aberrant V-D-J recombination, homologous recombination between Alu-repeats, and topoisomerase-II–mediated nonhomologous recombinations.133 Truncation or loss of function of theMLL gene alone may be the crucial event in leukemogenesis, with partner genes assuming a minor role and being interchangeable. However, Rogaia et al139 recently showed that fusion of theMLL gene with eps15 (on chromosome 1, band p32) in AML altered the cellular compartmentalization of the fusion protein, providing a putative mechanism for activation of MLL in 11q23 recombinations and suggesting a more active role of the fusion partner genes in leukemogenesis.

t(4;11)(q21;q23).

The most common translocation involving 11q23 is translocation t(4;11)(q21;q23), first described by Oshimura et al140 in 1977. It is observed in more than 60% of infants with ALL, 2% of children with ALL, and 3% to 6% of adults with ALL.7,9,18,37 It is invariably associated with young age (generally under 2 years), female sex, and high WBC counts, and is frequently associated with organomegaly and involvement of sanctuary sites such as the CNS.33,90,125,141 The immunophenotype is of the early pre-B or pre-B-cell type, is positive for TdT, HLA-DR, and CD19 with rearrangements of the Ig heavy-chain (IgH) genes, and is variably CD10. Cells frequently coexpress myeloid antigens, such as CD13, CD15, or CD33. Janssen et al125found a significant association of MLL-AF4 recombinations in particular with CDw65, compared with early pre-B-cell ALL without this genetic translocation. This association emphasizes an important characteristic of 11q23 abnormalities suggesting that the transforming event originates at the stage of a pluripotential progenitor cell with the capacity to differentiate to both lymphoid and myeloid lineages.142 In fact, 11q23 abnormalities can frequently be detected in biphenotypic or acute myeloid leukemias.128,143 

The clinical outcome for both adults and children with the t(4;11)(q21;q23) translocation is poor (Table7).33

Table 7.

Clinical Outcomes for Patients With t(4;11)(q21;q23)

Proportion With t(4;11)(q21;q23)UKALL XA AdultsTIWCL Adults TIWCL Children
10/350 9/172 9/157
CR (%)  70  50  88 
MCRD (mo)  NA  NA  3  
MST (mo) NA  7  9  
MDFS (mo)  4  2  
DFS  24% at 3 yr  0% at 5 yr  0% at 5 yr 
Proportion With t(4;11)(q21;q23)UKALL XA AdultsTIWCL Adults TIWCL Children
10/350 9/172 9/157
CR (%)  70  50  88 
MCRD (mo)  NA  NA  3  
MST (mo) NA  7  9  
MDFS (mo)  4  2  
DFS  24% at 3 yr  0% at 5 yr  0% at 5 yr 

Abbreviations: MCRD, median complete remission duration; MDFS, median disease-free survival.

Data from the UKALL XA trial,10 the Third International Workshop on Chromosomes in Leukemia (TIWCL),37 and Bloomfield et al.18 

The GFCH demonstrated 11q23 abnormalities in 7% of adult patients with ALL.7 Half of these patients had a t(4;11), 50% of whom had coexpression of myeloid markers: their CR rate was 75%, with a median DFS of 7 months and no DFS at 3 years. This was slightly worse than for the whole 11q23 group, in whom the CR rate was 72% and the median DFS was 8 months, but the 3-year DFS was 26%. No differences in DFS were noted with translocations as opposed to deletions of 11q23.

Janssen et al125 analyzed MLL-AF4 rearrangements by PCR in a series of 46 patients with early pre-B-ALL (34 adults, 12 children). They demonstrated fusion transcripts in 39% of the patients (14 adults, 4 children). Using intensified treatment strategies which included BMT in 4 adult cases, they reported 9 of 19 ALL patients withMLL-AF4 to be in remission for up to 54 months (median CR duration [CRD], 26 months), with 7 of the 9 showing PCR negativity for minimal residual disease.

These results emphasize that survival can be improved substantially by applying intensive therapy to cytogenetic groups that had been previously defined as poor risk by their response to standard treatments. Although the data in adult ALL with translocation t(4;11) are scarce, risk-adapted therapy might benefit this group as it did in children.33 

t(11;19)(q23;p13).

Translocation t(11;19)(q23;p13) shares similar clinical and prognostic features with t(4;11). It results in a fusion of the ENL gene (also called LTG19 or MLLT1) from chromosome 19 to the 5′ end of MLL on 11q23.144 The translocation is associated with younger age and high WBC count, as well as IgHgene rearrangements in blast cells and coexpression of lymphoid and myelomonocytoid antigens.

Abnormalities Involving 19p13

t(1;19)(q23;p13).

The two known translocations involving band p13 of chromosome 19 are t(1;19)(q23;p13) and its rarer variant t(17;19)(q21-22;p13). Translocation t(1;19) was first reported in 1984 by Carroll et al,145 who found that some leukemias with a pre-B-cell ALL phenotype (cytoplasmic Ig [clg]-positive and surface Ig [slg]-negative) carried this abnormality. It was confirmed in subsequent studies as one of the most common recurring translocations in childhood ALL, with a frequency of 5% to 6% overall, and of 25% in pre-B-cell ALL.15,90,146 It is also seen in 1% of childhood cases of early pre-B-cell ALL (clg, slg) and occasionally in ALL patients with a transitional pre-B phenotype (clg+, μ heavy-chain Igs detectable on cell surface).147-150 It is present in less than 5% of adult ALL cases.7,8,151 

The translocation can be observed in two principal forms. In its unbalanced form, −19, +der(19)t(1;19), two normal chromosomes 1 are present. Shikano et al152 suggested that this anomaly may result from loss of the der(1)t(1;19)(q23;p13.3) by nondisjunction or asynchronous replication, with replacement of the normal chromosome 1 by a second copy, which ultimately resulted in trisomy for 1q23 → 1qter. Abnormalities of chromosome 1, such as complete or partial trisomies for the long arm, are known to arise during clonal evolution and can be observed in recurrent hematologic malignancies.153-155 The other form of the translocation is a balanced rearrangement, a simple reciprocal translocation without net loss or gain of genetic material. Unbalanced translocations appear to be more common than balanced rearrangements (about 75% are unbalanced). No differences in clinical presentation or prognosis exist.34,153 

At a molecular level, the breakpoint on chromosome 19 has been mapped to a tightly clustered region on band p13.2-p13.3. This segment contains a gene, E2A, which encodes the two transcription factors E12 and E47 via alternative splicing of precursor mRNA.156 E12 and E47 are ubiquitous helix-loop-helix (HLH)-containing proteins that bind to the E-box element in the κ light-chain DNA-enhancer region.157 They are considered essential for normal lymphopoiesis and regulation of B-cell development.158 Breakpoints in the E2A gene occur almost exclusively in a 3.5-kb intron segment between exons 13 and 14.159 The breakpoint region on chromosome 1q23 appears to be more dispersed and lies within an intron of at least 50 kb in size.159 It disrupts the homeobox-containing “pre-B-cell leukemia” gene (PRL, also referred to as PBX1), which is transcriptionally silent in lymphoid cells. The genomic organization of PBX1 has not yet been detailed in entirety.158 The chromosomal rearrangement fuses the 5′ sequences of the E2A gene with 3′ sequences of PBX1.The resulting fusion transcript is a chimeric transcription factor which preserves the activation domain of E2A but has its DNA-binding domain and HLH dimerization domain replaced byPBX1.159E2A-PBX1 appears to function as a potent transcriptional activator.160,161 Despite the size of the breakpoint location on 1q23, an unvarying portion ofPBX1 is fused with E2A as most mRNAs generated by the translocation have the same sequence, suggesting that the site-specific fusion between E2A and PBX1 is pathogenetically important.157 However, differential splicing seems to occur at the mRNA level, leading to the production of two E2A-PBX1 chimeric proteins that differ in their extreme carboxyl-terminal end.158 Several groups have detected E2A-PBX1fusion transcripts that differ in their mRNA sequences.162,163 Their exact derivation, as well as their clinical significance, are unknown. Other members of the PBXfamily with homeodomains that are nearly identical to that ofPBX1, namely PBX2 and PBX3, have been identified, but their role in lymphoid cell transformation is unknown.164 

A strong association exists between t(1;19) and pre-B-ALL, especially in children,145 where the translocation is present in 25% to 30% of cases.15,33,90 Among 73 adults and 101 children with ALL, Fenaux et al8 found that all patients with t(1;19) (1 adult and 7 children) had a pre-B-ALL phenotype. No other translocation demonstrated a comparably close relationship to a specific immunophenotype in this study. Raimondi et al147found that most cases with pre-B-ALL and t(1;19) were pseudodiploid, and hyperdiploidy with more than 50 chromosomes was virtually absent. The immunophenotype in the majority of cases was positive for CD10, CD19, and clg. Borowitz et al165 analyzed the surface marker expression of t(1;19) ALL in more detail. They found 22 cases with t(1;19) among 697 patients with pre-B-ALL. Twenty of them were characterized by an identical, complex phenotype with homogenous expression of CD10, CD19, and CD9, complete absence of CD34, and partial absence of CD20. All 12 cases analyzed with molecular studies showed E2A-PBX1 abnormalities. The same complex phenotype was seen in only 8% of children with pre-B-ALL without t(1;19). Pui et al153 later added positivity for CD22 and negativity for CD21 to the characteristic surface-marker profile.

In rare cases, t(1;19) is seen in clg, early-pre-B-ALL. Involvement of either E2A or PBX1 in these cases could not be consistently shown.150,162 In contrast to the adverse clinical prognosis of patients with pre-B-ALL and translocation t(1;19) (see below), patients with early pre-B-ALL and t(1;19) appear to fall into a standard-risk category.148,162 

Clinical characteristics of pre-B-ALL with t(1;19) include presentation with high WBC counts, high LDH levels, and a DNA index of less than 1.6, which underscores that this type of leukemia is usually associated with a pseudodiploid karyotype. It frequently occurs in black people. The GFCH7 observed t(1;19)-positive leukemias in 11 (3%) of its adult ALL patients. Nine had a pseudodiploid karyotype, whereas two were hyperdiploid, with 47 to 50 chromosomes. Contrary to most other studies with t(1;19), patients tended to be younger and presented with lower WBC counts. Although all patients achieved a CR, the median DFS was only 6 months, and the 3-year DFS was 20%.

Pediatric Oncology Group studies showed that children with ALL who express E2A-PBX1 and t(1;19) and do poorly with standard or less aggressive therapy, have good responses to more intensified treatment.148,162 Therefore, to plan appropriate therapy, adequate and thorough cytogenetic studies, including molecular analysis, assume a critical role in diagnosis.148Comparable data for adult patients with ALL and expression ofE2A-PBX1 who share equally dismal outcomes with standard therapy as do children are not available.

t(17;19)(q21-q22;p13).

This less well-recognized translocation fuses the E2A gene to the “hepatic leukemia factor” (HLF) gene, a basic leucine zipper transcription factor gene, on chromosome 17.166 Whereas HLF breakpoints on chromosome 17 appear to lie consistently in intron 3, E2A breakpoints differ with respect to the inclusion of exon 13 and the insertion of a cryptic exon that contains E2A intronic sequences at its 5′ end andHLF intronic sequences at its 3′ end. These translocations involving the E2A gene have been reviewed by Hunger.158 

Abnormalities Involving the T-Cell Receptor (TCR) Genes

Translocations involving the TCR genes are among the most common abnormalities in T-cell ALL,90 whereas TCR gene rearrangements are found at a lower frequency in patients with B-lymphoid markers.167 T-cell ALL is somewhat peculiar in that the majority of patients do not have a cytogenetically detectable chromosomal abnormality, more patients have pseudodiploid karyotypes, and only few have hyperdiploidy.6,168 Raimondi et al169 described an overall frequency of 40% to 45% of translocations in childhood T-cell ALL, half having breakpoints mapped to chromosomal regions encoding for TCR genes. These loci include chromosome 14 band q11 (TCR-α and -δ genes), 7q32-36 (TCR-β gene), as well as 7p15 (TCR-γ gene).170 The β-chain locus on 7q32-36 is less frequently rearranged than the α-δ chain loci proximally on 14q. Rearrangements of theTCR-γ gene on 7p15 are extremely rare. Although no specific karyotypic abnormality can be associated with a distinct clinical subtype of T-cell ALL (as in B-cell ALL [eg, Burkitt's lymphoma and t(8;14)]),171 a number of distinct chromosomal translocations have been identified.

A common theme is the juxtaposition of TCR promotor/enhancer elements to a variety of putative or proven transcription factors located at or near breakpoints on the partner chromosomes. Several families of involved transcription factors can be distinguished on the basis of structural motifs that characterize their DNA binding or dimerization domains (Table 8).157 For example, the TAL1 (also referred to as SCL, TCL5),TAL2, LYL1, and c-myc genes, encode for transcription factors that have a characteristic HLH motif. This structure consists of two amphipathic α helices that are separated by an intervening nonhelical loop and which stretch over 50 to 60 amino acids.157,172 The HLH motif allows for specific protein-protein interactions such as homodimerization or heterodimerization. A stretch of basic residues at the N terminus seems to mediate sequence-specific DNA-binding.172,TAL1, TAL2, and LYL1 share more than 85% homology in their basic HLH domains. A second class of transcription factors are encoded for by the rhombotin gene family located on chromosomes 11 and 12.173 The two genes RBTN1 (or Lmo1, Ttg1, Rhom1) and RBTN2 (or Lmo2, Ttg2, Rhom2) are both involved in translocations associated with T-cell ALL. Their protein products contain two cysteine-rich regions that are referred to as LIM domains and, through stabilization of their tertiary folds by zinc, are able to interact and bind with nucleic acids or with other proteins. Finally, HOX11 is a homeobox-containing gene that is translated into a helix-turn-helix structure that shows remarkable conservation across species of a region containing about 60 amino acids that is responsible for DNA binding.157 

Table 8.

Protein Domains and Transcription Factor Families Involved in Rearrangements With TCR Gene Loci

Protein Domain Involved Gene Location Translocation
Transcription factors  
 Basic HLH proteins  TAL1 (SCL, TCL5)  1p32  t(1;14)(p32-34;q11)  
   t(1;7)(p32;q35)  
 TAL2  9q32  t(7;9)(q34;q32) 
 LYL1  19p13  t(7;19)(q34;p13)  
 c-myc 8q24 t(8;14)(q24;q11) 
 LIM domain proteins RBTN1 (RHOM1, Ttg1, Lmo1) 11p15 t(11;14)(p15;q11) 
 RBTN2 (RHOM2, Ttg2, Lmo2) 11p13 t(11;14)(p13;q11) t(7;11)(q35;p13) 
 RHOM3 12p12-13 None known 
 Homeodomain HOX11 (TCL3) 10q24 t(10;14)(q24;q11) 
  proteins   t(7;10)(q35;q24) 
Protein kinases LCK 1p34 t(1;7)(p34;q34) 
Notch homologue TAN1 9p34 t(7;9)(q34;p34) 
Chimeric Ig-TCR gene IgH 14q32.3 inv(14)(q11;q32.3) 
Unknown TCL1 14q32.1 inv(14)(q11;q32.1) 
   t(14;14)(q11;q32.1) 
Protein Domain Involved Gene Location Translocation
Transcription factors  
 Basic HLH proteins  TAL1 (SCL, TCL5)  1p32  t(1;14)(p32-34;q11)  
   t(1;7)(p32;q35)  
 TAL2  9q32  t(7;9)(q34;q32) 
 LYL1  19p13  t(7;19)(q34;p13)  
 c-myc 8q24 t(8;14)(q24;q11) 
 LIM domain proteins RBTN1 (RHOM1, Ttg1, Lmo1) 11p15 t(11;14)(p15;q11) 
 RBTN2 (RHOM2, Ttg2, Lmo2) 11p13 t(11;14)(p13;q11) t(7;11)(q35;p13) 
 RHOM3 12p12-13 None known 
 Homeodomain HOX11 (TCL3) 10q24 t(10;14)(q24;q11) 
  proteins   t(7;10)(q35;q24) 
Protein kinases LCK 1p34 t(1;7)(p34;q34) 
Notch homologue TAN1 9p34 t(7;9)(q34;p34) 
Chimeric Ig-TCR gene IgH 14q32.3 inv(14)(q11;q32.3) 
Unknown TCL1 14q32.1 inv(14)(q11;q32.1) 
   t(14;14)(q11;q32.1) 

Abbreviation: HLH, helix-loop-helix.

Data from refs 164, 167, 172, 173, and 194.

TAL1 gene rearrangements.

TAL1 gene rearrangements are the most common abnormalities identified in children with T-cell ALL. They are associated with CD3 leukemias with commitment to the α/β lineage.174 Two mechanisms of rearrangement exist. About 3% of cases occur via a chromosomal translocation that transposes the TAL1 gene on chromosome 1 to either theTCR-α/δ locus on chromosome 14q11, as in translocation t(1;14)(p33;q11), or to theTCR-β location on 7q35, as in translocation t(1;7)(p33;q35). The latter translocation is less frequent. Another translocation, t(1;3)(p32;p21) that involves an uncharacterized segment on chromosome 3 and a segment 5′ to TAL1 has been described by Aplan et al.175 In 25% to 30% of pediatric patients withTAL1 alterations, the tumor-specific rearrangement ofTAL1 is not detectable by routine cytogenetic analysis.157,172,176,177 In these patients site-specific, submicroscopic deletion of a 90-kb to 100-kb segment fuses the coding exons of TAL1 to the first noncoding exon of the “SCL interrupting locus” (SIL) gene centromeric in a 5′ orientation on chromosome 1p33.176,178 Either translocation or interstitial deletion results in the juxtaposition of the TAL1gene to regulatory elements of either the SIL gene or the TCR genes. Although the breakpoint on the SIL gene remains remarkably constant, several breakpoints of the TAL1 gene have been identified (tald1-d3), of which the most frequent, tald1 and tald2, leave all coding exons ofTAL1 intact. The site specificity and precision of the gene rearrangement process, as well as the fact that the deletions are mediated by specific heptamer and nonamer flanking sequences that share homology to the heptamers in the recombination sequences of Ig and TCR genes, suggest that abnormal V-D-J recombination is effective in activating the TAL1 gene locus.157,179 AlthoughSIL and TCR genes are normally expressed in T cells,TAL1 is transcriptionally silent in normal T cells although it is expressed in megakaryocytes, basophilic granulocytes, and erythroblasts.172 Alternative splicing produces a variety of TAL1 transcripts.172 The two most relevant are the full-length, 42-kD TAL1α polypeptide (pp42TAL1, amino acids 1-331), which contains all three TAL1 coding exons (exons 4 to 6), and the 22-kD, N-terminally truncated polypeptide, TAL1β (pp22TAL1, amino acids 176-331), which lacks coding exon 4. Both polypeptides can be found in T-cell ALL, and both are able to form heterodimers with other basic HLH proteins such as E12 or E47, thus specifically recognizing E-box DNA transcriptional enhancer motifs.157,172 

A subset of T-cell ALL patients displays cytogenetic alterations of both the TAL1 gene and RBTN2 (see below), and theTAL1 and RBTN2 polypeptides seem to dimerize with each other easily in vivo.180 Both genes appear to play a crucial role in normal hematopoiesis as severe defects in erythropoiesis occur in mice that are deficient in either gene.181 Ono et al182 recently showed in T-cell ALL cell lines that aberrant expression of TAL1 was invariably accompanied by aberrant expression of either RBTN1 orRBTN2. The coexpression also strongly induced TALLA1, a highly specific T-cell marker. It is conceivable that the TAL1and RBTN gene products act synergistically in the genesis of T-cell ALL, and that their influence on the expression of downstream genes, such as TALLA1 or other unknown genes, is part of the mechanism of leukemogenesis.

Disruptions of the TAL1 gene locus are less frequent in adults than in children, where they occur with a frequency of up to 30% of T-cell ALL cases. Stock et al183 performed Southern Blot and PCR analysis in 33 newly diagnosed adult patients with T-cell ALL and detected only 1 case with a t(1;14)(q34;q11) molecular rearrangement, but no submicroscopic deletions. No comprehensive data exist on the clinical significance of TAL1 gene rearrangements in adults with ALL. In one of the largest studies to date, Bash et al184 found that 26% of 182 children with newly diagnosed T-cell ALL had local rearrangements of the TAL1 locus. No significant clinical correlations were obvious except that WBC counts and hemoglobin were higher at diagnosis in the group with theTAL1 rearrangements. The immunophenotype was characterized by positivity for CD2 and absence of CD10 expression. This marker profile was the same that Stock et al183 had found in their single adult patient with TAL1 gene rearrangement. Response to treatment was not significantly different with or without the rearrangement, although a trend for better DFS was observed with expression of TAL1. The latter finding concurs with results of a smaller study in children by Kikuchi et al,185 who suggested that TAL1 gene rearrangements may identify a subgroup of T-cell ALL with a favorable prognosis.

The closely related genes, LYL1 and TAL2, are rearranged in chromosomal translocations t(7;19)(q34;p13) and t(7;9)(q34q32), respectively. The result is a juxtaposition of both genes to promoter/enhancer elements of the TCR-β gene. These translocations occur infrequently.172 

t(10;14)(q24;q11).

Translocation t(10;14)(q24;q11) rearranges the homeobox-containing geneHOX11 (TCL3) with the TCR-δ gene; it is seen in 4% to 7% of T-cell acute leukemias.169,186 As in the TAL1 rearrangements, the coding regions of HOX11 are not disrupted by the translocation.187 The breakpoints on chromosome 10 cluster within a 15-kb region. Immediately telomeric to the breakpoint location lies the coding sequence of HOX11.188HOX11transcripts occur abundantly in leukemic blasts.187 The variant translocation t(7;10)(q35;q24) juxtaposes HOX11 to theTCR-β gene region. Both translocations result in overexpression of normal HOX11 mRNA by bringing HOX11under the influence of TCR promoter sequences.187,188 

The GFCH7 found t(10;14)(q24;q11) to be the most frequent chromosomal rearrangement in patients with T-cell ALL. This translocation was associated with a favorable outcome: the CR rate was 100%, with a median DFS of 46 months and a 3-year DFS rate of 75%.7 If other 14q11 abnormalities, such as t(11;14)(p13;q11), t(1;14)(p32;q11), inv(14)(q11;q32), and del(14)(q11) were included, the clinical outcomes were less favorable, with a median DFS of 24 months and a 3-year DFS rate of 42%.

t(11;14)(p15;p11) and t(11;14)(p13;q11).

Translocation t(11;14)(p15;p11) recombines the RBTN1 gene with the TCR-δ chain gene. RBTN1 encodes an 18-kD protein characterized by the presence of two cysteine-rich structural motifs that establish it in the LIM family of transcription factors.173,189 It is a rare but consistent translocation in T-cell ALL. The RBTN2 gene is rearranged in the closely related chromosomal translocation t(11;14)(p13;q11), as well as in t(7;11)(q35;p13). The breakpoint region (T-ALLbcr) on 11p13 lies within a 26-kb segment upstream of the RBTN2promoter.190 Neither RBTN1 nor RBTN2 is expressed in normal T cells, an attribute they share with TAL1.Their increased expression appears to contribute to the pathogenesis of T-cell lymphocytic leukemia by as yet undefined mechanisms.

t(8;14)(q24;q11).

Translocation t(8;14)(q24;q11) is not specific for T-cell ALL, and has been described in pre-B-ALL.191 The rearrangements are similar to those between Ig gene loci and c-myc in B-cell malignancies such as Burkitt's lymphoma (BL), but in the translocations mentioned here, c-myc recombines with theTCR-α gene in 14q11. Breakpoints in theTCR-α gene are located 5′ of one of the joining (J) segments, and breakpoints in c-myc are located distal to coding sequences in a 3′ orientation of the third exon.191-193 These chromosomal rearrangements are too infrequent both in adults and children to provide definite correlation with clinical characteristics and with prognosis.

Translocation t(8;14)(q24;q32) and Its Variants

t(8;14)(q24;q32) is a highly specific and consistent chromosomal rearrangement in mature B-cell neoplasms.194 Mature B-cell leukemias, which account for less than 5% of all cases of ALL in children or adults, represent the leukemic phase of BL.195The chromosome rearrangement does not appear to be different in BL and in its leukemic counterpart. It is present in both the sporadic, nonendemic form of BL and the endemic African tumor-type presentation.34 Patients in the leukemic phase present with bulky extramedullary disease, frequent and early CNS involvement, and a rapidly progressive clinical course due to an unusually high blast cell proliferative rate.33,151 The malignant cells are characterized by strong expression of surface Igs, and cell morphology is almost always L3.

B-cell ALL typically contains one of three specific chromosomal translocations. In about 85% of cases t(8;14)(q24;q32) can be detected. Less frequent are the variants t(2;8)(p12;q24), with 5% prevalence, and t(8;22)(q24;q11), with about 15% prevalence.34 However, association with these chromosomal changes is not absolute. There are cases in which the typical translocations are not found, and additional karyotypic abnormalities such as dup(1q), t(1;19)(q23;p13), del(6q), 13q34 abnormalities, trisomy 20, t(X;8)(p23;q24), the Ph chromosome, or complex three-way translocations have been detected.196-198 

The common denominator behind the classic translocations is a break in 8q24. This band contains the c-myc proto-ongene, which in t(8;14) is translocated to the IgH gene locus on chromosome 14q32.34,199 In the variant translocations t(2;8) and t(8;22), c-myc recombines with the Ig kappa light chain (Igκ) gene locus on chromosome 2p12 or the Ig lambda light chain (lgλ) gene locus on chromosome 22q11, respectively.34,199 These rearrangements bring c-myc under the influence of transcription-stimulating regulatory sequences in the proximity of constitutively active Ig loci.34 

Of the three exons in c-myc, the first most likely remains untranslated. The protein products encoded in exons 2 and 3 share structural motifs, such as a basic domain with an HLH-segment and leucine zipper regions. Other nuclear transcription factors also share these motifs. C-myc is able to form dimers with another nuclear protein called MAX, and together they show sequence-specific binding to DNA.200 

In t(8;14) the breakpoint almost always falls proximal on the centromeric side of exon 2 of c-myc, either in the 5′ flanking region or in the first exon or intron, so that the entire coding segment of c-myc becomes translocated.201 The translocation to 14q32 happens in a head-to-head (5′ to 5′) direction, either in the switch (S) or joining (J) regions on 14q32. The breakpoints at the IgH gene locus may be dispersed over a large area of approximately 170 to 190 kb.202 The 8q24 breakpoints in the variant translocations differ. They are located 3′ on the telomeric side of the c-myc locus, which means that c-myc remains on the rearranged chromosome 8, while the Ig light chain (IgL) gene loci on 2p12 and 22q11 are translocated to chromosome 8 and rearranged with c-myc. TheIgL breakpoints can fall within either the J, diversity (D), or S regions of the gene. Because of the different orientation of the variable regions of the IgL genes the rearrangements happen in a 5′ to 3′ (head-to-tail) fashion.34,203,204 The mechanisms by which c-myc becomes leukemogenic are not entirely clear yet. The dominant hypothesis is that c-myc is brought under the influence of highly active regulatory sequences of Ig genes, and thus itself becomes inappropriately expressed.34 However, other possible mechanisms are submicroscopic mutations, such as deletions of c-myc, or the utilization of different transcriptional promotors.205-207 The normal allele of c-mycremains silent.

The TIWCL found abnormalities of 8q24 (mostly 8q−, 14q+) in 6% of adult ALL patients. All patients had typical L3 morphology. Adults were affected more frequently than children, and males more than females. The majority (15 of 16 patients, including 6 children) had pseudodiploid karyotypes. The CR rate was 44%, the median CRD and survival time 5 months each, and 5-year DFS was 18%. Adult patients had a lower CR rate.5,6,18,25,36,37 The GFCH found t(8;14)(q24;q32) in 5% of patients. Two of these 21 patients showed variant translocations, all were of B-cell type, and 20 of 21 had L3 morphology. Sixty-two percent achieved CR, the median DFS was 2 months, and the 3-year DFS rate was 17%.7 Fenaux et al8 detected only 1 t(8;14) among 73 adult patients with ALL; it was slg+ but of L2 morphology. No survival data are available. Ankathil et al28 detected one t(8;14) in 46 adults and reported a survival time of 10 months.

Although mature B-cell ALL was long believed to be associated with a poor prognosis, the introduction of short-term dose-intensive regimens such as hyperfractionated cyclophosphamide, high-dose methotrexate, and cytarabine have significantly improved the clinical outcomes.151,171,208 This provided additional evidence that a change in therapy can have a beneficial impact on a disease category that has traditionally been considered “poor-risk.” This effect can also be shown in adult patients.145 

Abnormalities of the Short Arm of Chromosome 12

Prigogina et al209 first described abnormalities of the short arm of chromosome 12 in ALL. Raimondi et al210 showed that rearrangements of 12p, including deletions and translocations with various partner chromosomes such as 1, 3, 9, 10, and 17, were frequent events in childhood ALL, occurring in about 10% of patients. Most cases were characterized by a pre-B-phenotype and L1 morphology. As the common denominator of these rearrangements was a breakage in 12p, it could be suspected that in these instances a location on the short arm of chromosome 12 was critical to the process of leukemogenesis. The UKALL XA trial10 showed 12p abnormalities in 13 (4%) of 350 adult patients. In 11 cases, translocations involving various partner chromosomes were identified [14q11, 14q13, 10q11, 12q11, 17q12, 22q11 and 3 cases of dic(9;12)(p11;p11)]. The whole group with 12p abnormalities was characterized by a favorable outcome, with a CR rate of 92%, a median DFS of 36 months, and a 3-year DFS rate of 74% (for the total cohort of adult ALL patients: CR rate 88%, median DFS 19 months, 3 year-DFS rate 36%). The GFCH study7 identified 12p abnormalities in 23 (5%) of 443 adults. Twenty of them had monosomy 12 (8 deletions, 12 unbalanced translocations). The 12p abnormalities were most often observed in complex karyotypes and, overall, had no impact on prognosis.

Deletions of 12p13.

Several investigators have identified 12p13 as a hot spot frequently deleted in childhood ALL211,212 but rarely involved in adult ALL.213 Fine mapping of this segment showed that the deleted region lies between two genes, TEL (for “translocation, Ets, leukemia,” or ETV6), andCDKN1B (or KIP1). TEL is located at the telomeric end and encodes for a transcription-regulating protein of the Ets family of transcription factors. CDKN1B has a more centromeric location and encodes for p27kip1, an inhibitor of the cyclin E-cdk2 complex.214 The TEL gene was initially cloned by virtue of its involvement in translocation t(5;12)(q33;p13) in chronic myelomonocytic leukemia, where it is fused with the platelet-derived growth factor-β gene.215 Other chimeric TEL transcripts have been identified in both myeloid and lymphocytic leukemias, as well as in acute and chronic leukemias. Examples are TEL-MN1 in t(12;22)(p13;q11),216,TEL-ABL in t(9;12)(q34;p13),217,218 or TEL-10q24 in t(10;12)(q24;p13), a variant of t(5;12).219 More recentlyTEL was shown to be fused to the AML1 (orCBFA2) gene on chromosome 21q22 in childhood B-lineage ALL.220,221 It is noteworthy that the region of loss of heterozygosity on 12p13 not only comprises TEL but also extends centromeric into the KIP1 gene, raising the possibility that loss of function of KIP1, in association with deletions and/or rearrangements of TEL, may play a crucial role in ALL.222-225 

t(12;21)(p11-12;q22).

The TEL protein is characterized by two distinct regions. The Ets motif consists of a 70-amino acid, sequence-specific DNA-binding domain that is located at the C-terminal end of the protein. The N-terminus contains an HLH motif, which is able to mediate interactions with other proteins. The fusion partner in t(12;21) is AML1, a heterodimer consisting of an α- and β-subunit. The α-chain possesses a DNA-binding and transactivating motif called “runt” (in homology to the Drosophila runt gene). In translocation t(12;21) the HLH domain of TEL is fused to intron 2 of the AML1 gene in such a way that both the DNA-binding and runt domain remain intact and are expressed.225-227 Of interest, the nonrearrangedTEL allele seems to be deleted in almost all t(12;21) cases.228 

Translocation t(12;21) was long considered a rarity, as it was detected in less than 0.05% of patients analyzed with cytogenetic studies.24 Therefore, it was thought to be of no significant prognostic value. However, t(12;21) is difficult to detect, and routine cytogenetic studies grossly underestimated its frequency. In several studies, the application of molecular tools such as FISH, Southern blot analysis, and RT-PCR have shown loss of heterozygosity and TEL-AML1 fusion transcripts in up to 27% of children with B-lineage ALL, making it the most common karyotypic-molecular abnormality in pediatric ALL.222,229-236 TheTEL-AML1 rearrangement conferred an exceptionally good outcome in childhood B-precursor ALL and provided prognostic information independent of the consensus workshop risk groups, age, and WBC count at presentation.231,233,234,236 Lanza et al237described a more heterogeneous behavior of t(12;21) in childhood ALL with regard to lineage involvement and outcome. They studied 51 children with ALL by RT-PCR and found a TEL-AML1 rearrangement in 11 samples. Three of these children showed a hybrid phenotype, coexpressing both lymphoid and myeloid markers. Two of the 11 children (one with hybrid phenotype) died in relapse, in the absence of other poor-risk factors.

Available data are more limited in adult ALL. Raynaud et al238 analyzed 118 adult ALL patients (30 with T-cell ALL, 88 with B-lineage ALL) by RT-PCR for the TEL-AML1 fusion transcript and found only one positive case among the B-cell samples. Aguiar et al239 studied 101 patients by RT-PCR followed by Southern blotting and direct sequencing and detected only 3 cases of common ALL that were positive for TEL-AML1 rearrangements. Overall, the frequency of TEL-AML1+ ALL was low in adult B-lineage ALL (3% to 4%), and little is known about its prognostic significance.236,238,239 

Abnormalities of the Long Arm of Chromosome 6

In 1977 Oshimura et al35 described deletion of 6q as a recurrent karyotypic event in leukemias. Del(6q) occurs in about 4% to 6% of childhood ALL and probably somewhat less frequently in adults. Abnormalities of 6q are associated with other karyotypic abnormalities, although in one third of cases they occur as the sole change.33,34 The TIWCL found abnormalities of 6q in 1.7% of adults and 6.4% of children.6,18,25,37 Most deletions involved 6q21. The most frequently associated karyotypic abnormality was +21, followed by +4, +10, +11, and +14. For children, the prognosis was similar to that with no detectable cytogenetic abnormality; for adults, there were too few cases to make definite conclusions.6,18,25,37 Studies have either confirmed no difference in prognosis or reported improved outcomes for 6q−, with longer DFS or prolonged median survival times.7,10,28 

Although 6q deletions can be either interstitial or terminal, most seem to be interstitial. Menasce et al240 showed that even if conventional cytogenetic analysis showed a terminal deletion, FISH study revealed it to be interstitial. Although the breakpoint locations vary, they center around a proximal region flanked by 6q14-15 and 6q21 and a more telomeric region distal to 6q23.241-243 This raises the possibility of a putative tumor suppressor gene in that area. Indeed, the c-myb oncogene is localized to 6q22-24; however, it does not appear to be involved in the 6q abnormalities of lymphoid malignancies.244 

The estrogen receptor (ER) gene is located on 6q25.1.245 It has been shown to have suppressive activity on growth and metastasis in many different cell lines.246Issa et al247 demonstrated that the promoter region of theER gene is aberrantly methylated at CpG islands in leukemia cell lines, pediatric and adult acute lymphocytic as well as myeloid leukemias, and most hematopoietic tumors, including lymphomas, in which it was associated with a very low or even absent expression of theER gene. The methylation of specific gene loci has not been fully investigated but may possibly contribute or even be an important step in human leukemogenesis.

Isochromosomes

Isochromosomes are common tumor-associated cytogenetic abnormalities. They can be either monomeric or dicentric, in which case they have two homologous arms that are mirror images of one another. They result from aberrant transverse instead of longitudinal chromosome division or, alternatively, by chromatid exchange between the two homologous chromosome arms.248,249 The consequence is gain and/or loss of genetic material of the affected chromosome segments. Isochromosomes have been observed in 7% to 9% of ALL cases.250,251 The most frequent chromosomal changes are i(7q), i(9p), and i(17q). Recently i(21q) has been described in ALL.250 Isochromosomes have been detected more frequently in adults than in children (7.8% v 4.5%).250 

i(7q).

The isochromosome i(7q) is almost always one of several changes and is particularly associated with primary abnormalities, such as t(4;11)(q21;q23) or der(19)t(1;19)(q23;p13).251 At least in childhood ALL i(7q) seems to be correlated with hyperdiploid karyotypes.252 

i(9q).

Like i(7q), i(9q) is usually not the sole anomaly. It is associated with L2 morphology and a pre-B-cell phenotype.

i(17q).

The isochromosome i(17q) is one of the most frequent isochromosomes in ALL. It has a tendency to occur with early pre-B- and pre-B-cell leukemias and has a well-described association with primary aberrations t(9;22) and t(4;11).251 In t(9;22), i(17q) is rarer in Ph+ ALL than in Ph+ CML, where it serves as a paradigmatic marker for disease transformation. Frequently, i(17q) is observed with hyperdiploid karyotypes.34 Although hyperdiploidy is an indicator of good prognosis, Pui et al253 reported poor remission duration in children with hyperdiploidy and i(17q).

i(21q).

Martineau et al250 recently described i(21q) in ALL at a higher frequency than was found in previous studies. It identified patients with B-lineage disease and low WBC counts at presentation, was more frequent with male gender, and was associated with pseudodiploidy or low hyperdiploidy. The study included both children and adults. Follow-up was too short to permit conclusions regarding prognosis.

Overall, except for the association of isochromosomes with certain established primary abnormalities and ploidy groups, no significant clinical or hematologic characteristics distinguished this group of chromosomal anomalies.34,250,251 

Tumor suppressor genes define a group of genes whose expression can block the development of a tumorigenic phenotype and, upon inactivation, will facilitate the malignant process. The presence of these genes can be suspected in certain chromosomal locations by identifying loss of material from specific chromosomes. However, tumor suppressor genes are frequently altered by mechanisms that are operational at a submicroscopic level such as point mutations, deletions of coding or regulatory gene sequences, transcriptional silencing of genes by hypermethylation, altered protein expression, or expression of inhibiting proteins. Thus, most tumor suppressor gene abnormalities are detected not by conventional cytogenetics, but by molecular techniques such as Southern blot analysis, PCR, SSCP, nucleotide sequencing, or pulsed-field gel electrophoresis. Although tumor suppressor genes are frequently deleted in solid tumors,254-257 they appear to be present in only a fraction of cases of human leukemias and may not be major contributors to human leukemogenesis.

A gene frequently deleted in acute leukemias (mainly T-cell–derived leukemias) is p16INK4A, with or without codeletion of p15INK4B, and both are located on the short arm of chromosome 9. The incidence of homozygous deletions ofp16INK4A has been as high as 80% in some series.97,109,110,115 

Of the tumor suppressor genes, the retinoblastoma (RB1) and thep53 genes have been the most thoroughly investigated. TheRB1 gene, on chromosome 13q14, was isolated and cloned by virtue of its deletion in retinoblastoma.258 It binds to and inhibits several transcription factors (among them, E2F) and inhibits cell-cycle progression at the G1-S phase. It was shown to be deleted in 12% of T-ALL patients and about 20% of patients with CD10+ early pre-B-ALL in a study by Ahuja et al,259 which included both children and adults.

The p53 gene is located on the short arm of chromosome 17, band p13.1.260 Loss of wild-type function of p53 appears to facilitate growth and transformation of malignant cells and, in some cases, defer entry of cells into apoptotic pathways.261,262Several studies have analyzed p53 mutations in lymphoid malignancies, including ALL and non-Hodgkin's lymphomas, by SSCP, PCR, and nucleotide sequencing. The incidence of p53 alterations was low, ranging from 2% to 15%, and included both pediatric and adult cases.263-265 There is a correlation between p53mutation and progressive or relapsing disease, especially in T-cell ALL. In B-lineage disease, p53 mutations are frequently observed with translocation t(1;19) and with t(8;14) and FAB L3 morphology.266-270 In L3 ALL, mutations of p53 do not correlate with extensive tumor load or poor response to intensified treatment, at least as long as a normal p53 allele persists.271 Most data are based on results in children with ALL and very few have included adults.

How do tumor suppressor gene alterations influence each other and affect outcome?

Tsai et al272 analyzed RB1 and p53 gene expression in adult ALL (89 de novo, 26 relapsed or refractory). Lack of RB1 expression was not different between de novo and relapsing disease (64% v 58%), but overexpression ofp53 was significantly higher in cases of relapsed patients (42% v 21%). In about 10% of cases p53 andRB1 expression were both abnormal. Although the presence of the double abnormality did not distinguish between de novo and relapsing disease, it was associated with increased early mortality and suggested a poor response to therapy. There was no definite relationship between abnormalities of the tumor suppressor genes alone and disease outcome. However, loss of RB1 expression and abnormal p53 were more frequent in adult than in pediatric ALL populations.

Hangaishi et al273 examined inactivations of several tumor suppressor genes in lymphoid malignancies, includingp16INK4A, p15INK4B, p53, andRB1, by Southern blot analysis, immunoblotting, PCR, SSCP, and nucleotide sequencing of the p53 gene. Among 117 ALL cases,p16 deletions were found in 41% (homozygous deletions in 27%), deletions of p15 in 43% (homozygous deletions in 36%), deletions of the RB1 gene in 2% (but loss of the retinoblastoma protein in 33%), and aberrations of the p53gene in 10% (with loss of the second allele in 5%). In 45% of the samples, one or more aberrations of the four tumor suppressor genes were reported. Noteworthy is the high degree of loss of the retinoblastoma protein, which was also found in other lymphoid malignancies and was also observed by Tsai et al.272Inactivations of the tumor suppressor genes occurred independently of each other.

Most if not all cases of ALL harbor cytogenetic or submicroscopic molecular abnormalities. Abundant evidence now exists that these aberrations are not “innocent bystanders,” but that they represent decisive early steps in a process that leads to the disruption of hematopoiesis, the development of leukemia, and then continues in the leukemic clone as part of a “clonal evolution.” Associations exist between some chromosomal rearrangements and morphologic and immunophenotypic characteristics of the leukemia cell (Table 3). Examples are translocation t(8;14), which occurs in mature B-cell ALL with almost exclusively L3 morphology. Others include TCR rearrangements in T-cell ALL, and the Ph translocation or abnormalities of 19p13, both of which are closely linked to a pre-B immunophenotype. More importantly, karyotypic changes provide important prognostic information independent of other variables such as age or initial leukocyte count, allowing “low-risk” and “high-risk” patients to be distinguished at the time of diagnosis.10,25,274 

This has led to the concept of risk-adapted therapy in which treatment is tailored according to risk groups, and intensive therapy approaches, including BMT, can be reserved for patients who, by virtue of their cytogenetic profile, will otherwise do poorly with chemotherapy. Examples include the Ph translocation, 11q23 abnormalities, and translocation t(1;19).7,8,10,18,25,37,84,90 On the other hand, patients at low risk can be spared the morbidity or mortality of high-dose chemotherapy, when it is not needed, as for t(12;21) or trisomies 4 and 10.30,231,233,234,236 This strategy is beginning to be applied successfully in cases of childhood ALL, where a large body of data guides clinical decision making. Outcomes often improve significantly once poor-prognosis groups are identified and adequately treated, as was seen in cases of t(1;19) or mature B-cell ALL (Burkitt's leukemia).148,151,171,208 

These developments are in sharp contrast to the situation in adult ALL where an insufficient database and concomitant lack of experience have delayed the adoption of risk-oriented therapeutic concepts in most cases. Expansion of our knowledge of cytogenetic abnormalities in adult ALL, and investigation of how these changes pertain to clinical behavior and prognosis are of paramount importance. To achieve this goal, cytogenetic and molecular abnormalities at the time of diagnosis should be defined. Traditional cytogenetic studies are not sufficient, and thorough molecular analyses are needed. Examples for which evaluations by routine cytogenetics underestimate the frequency of rearrangements include Ph butBCR-ABL+ leukemias,71-76 TCR gene rearrangements involving the TAL1gene,157,172,176,177 cryptic t(12;21) with rearrangements of the TEL gene,229,234,235 and deletions or mutations of the p16 gene in abnormalities of the short arm of chromosome 9.109-113 

The analysis of cytogenetic abnormalities is more than a prognostic tool and should provide better understanding of the molecular biology of leukemias. Many genes have been implicated in leukemogenesis (Table3). These include oncogenes, putative or proven tumor suppressor genes, genes that act as transcription-regulating factors, and genes that are involved in the regulation of apoptosis and differentiation. The study of these genes, as well as their transcriptional and translational products, will further increase our understanding of crucial pathways of leukemic transformation.

We are grateful to Arlene Hoffman for her excellent assistance in editing this manuscript and many helpful suggestions. Due to the vast amount of literature and space limitations, we could not include many reports that also made important contributions to the topic discussed, and we apologize to their authors.

Address reprint requests to Hagop M. Kantarjian, MD, Department of Leukemia, Box 61, M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030.

1
Young JL Jr
Ries
LG
Silvergerg
E
Horm
JW
Miller
RW
Cancer incidence, survival and mortality for children younger than age 15 years.
Cancer
58
1986
589
2
Mauer
AM
Acute lymphocytic leukemia
Williams
WJ
Beutler
E
Erslev
AJ
Lichtman
MA
Hematology
4
1990
994
McGraw-Hill
New York, NY
3
Mitelman
F
Cytogenetic methods and terminology
Heim
S
Mitelman
F
Cancer Cytogenetics
2
1995
20
Wiley-Liss
New York, NY
4
Williams
DL
Harris
A
Williams
KJ
Brosius
MJ
Lemonds
W
A direct bone marrow chromosome technique for acute lymphoblastic leukemia.
Cancer Genet Cytogenet
13
1984
239
5
Mitelman
F
The Third International Workshop on Chromosomes in Leukemia.
Cancer Genet Cytogenet
4
1981
96
6
Third International Workshop on Chromosomes in Leukemia
Chromosomal abnormalities and their clinical significance in acute lymphoblastic leukemia.
Cancer Res
43
1983
868
7
Groupe Français de Cytogénétique Hématologique
Cytogenetic abnormalities in adult acute lymphoblastic leukemia: Correlations with hematologic findings and outcome. A collaborative study of the Groupe Français de Cytogénétique Hématologique.
Blood
87
1996
3135
8
Fenaux
P
Laı̈
JL
Morel
P
Nelken
B
Taboureau
O
Deminatti
M
Bauters
F
Cytogenetics and their prognostic value in childhood and adult acute lymphoblastic leukemia (ALL) excluding L3.
Hematol Oncol
7
1989
307
9
Rieder
H
Ludwig
W-D
Gassmann
W
Thiel
E
Löffler
H
Hoelzer
D
Fonatsch
C
Chromosomal abnormalities in adult acute lymphoblastic leukemia: Results of the German ALL/AUL study group.
Recent Results Cancer Res
131
1993
133
10
Secker-Walker
LM
Prentice
HG
Durrant
J
Richards
S
Hall
E
Harrison
G
Cytogenetics adds independent prognostic information in adults with acute lymphoblastic leukaemia on MRC trial UKALL XA.
Br J Haematol
96
1997
601
11
(letter)
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.
N Engl J Med
313
1985
640
12
Yunis
JJ
Recurrent chromosomal defects are found in most patients with acute nonlymphocytic leukemia.
Cancer Genet Cytogenet
11
1984
125
13
Secker-Walker
LM
Lawler
SD
Hardisty
RM
Prognostic implications of chromosomal findings in acute lymphoblastic leukemia at diagnosis.
Br Med J
2
1978
1529
14
Secker-Walker
LM
Chessells
JM
Stewart
EL
Swansbury
GJ
Richards
S
Lawler
SD
Chromosomes and other prognostic factors in acute lymphoblastic leukaemia: A long-term follow-up.
Br J Haematol
72
1989
336
15
Pui
C-H
Crist
WM
Look
T
Biology and clinical significance of cytogenetic abnormalities in childhood acute lymphoblastic leukemia.
Blood
76
1990
1449
16
Rubin
CM
Le Beau
MM
Cytogenetic abnormalities in childhood acute lymphoblastic leukemia.
Am J Pediatr Hematol Oncol
13
1991
202
17
Crist
WM
Pui
C-H
Clinical implications and molecular genetic analyses of pediatric acute lymphoblastic leukemia.
Stem Cells
11
1993
81
18
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
Mitelman
F
Six-year follow-up of the clinical significance of karyotype in acute lymphoblastic leukemia.
Cancer Genet Cytogenet
40
1989
171
19
Bennett
JM
Catovsky
D
Daniel
MT
Flandrin
G
Galton
DAG
Gralnick
HR
Sultan
C
Proposals for the classification of acute leukemia.
Br J Haematol
33
1976
451
20
Williams
DL
Raimondi
SC
Pui
C-H
Rivera
GK
Evolving chromosome patterns and new cytogenetic concepts in childhood acute lymphoblastic leukemia
Acute Lymphoblastic Leukemia: UCLA Symposia on Molecular and Cellular Biology, New Series, vol 108.
Gale
RP
Hoelzer
D
1990
Wiley-Liss
New York, NY
21
Williams
DL
Tsiatis
A
Brodeur
GM
Look
AT
Melvin
SL
Bowman
WP
Kalwinsky
DK
Rivera
G
Dahl
GV
Prognostic importance of chromosome number in 136 untreated children with acute lymphoblastic leukemia.
Blood
60
1982
864
22
Heerema
NA
Palmer
CG
Baehner
AL
Karyotypic and clinical findings in a consecutive series of children with acute lymphocytic leukemia.
Cancer Genet Cytogenet
17
1985
165
23
Groupe Français de Cytogénétique Hématologique
Collaborative study of karyotypes in childhood acute lymphoblastic leukemia.
Leukemia
7
1993
10
24
Raimondi
SC
Current status of cytogenetic research in childhood acute lymphoblastic leukemia.
Blood
81
1993
2237
25
Bloomfield
CD
Goldman
AI
Alimena
G
Berger
R
Borgström
GH
Brandt
L
Catovsky
D
de la Chapelle
A
Dewald
GW
Garson
OM
Garwicz
S
Golomb
HM
Hossfeld
DK
Lawler
SD
Mitelman
F
Nilsson
P
Pierre
RV
Philip
P
Prigogina
E
Rowley
JD
Sakurai
M
Sandberg
AA
Secker-Walker
LM
Tricot
G
Van Den Berghe
H
Van Orshoven
A
Vuopio
P
Whang-Peng
J
Chromosomal abnormalities identify high-risk and low-risk patients with acute lymphoblastic leukemia.
Blood
67
1986
415
26
Walters
R
Kantarjian
HM
Keating
MJ
Estey
EH
Trujillo
J
Cork
A
McCredie
KB
Freireich
EJ
The importance of cytogenetic studies in adult acute lymphocytic leukemia.
Am J Med
89
1990
579
27
Micallef-Eynaud
PD
Eden
OB
Grace
E
Ellis
PM
Cytogenetic abnormalities in childhood acute lymphoblastic leukemia.
Pediatr Hematol Oncol
10
1993
25
28
Ankathil
R
Geetha
N
Remani
P
Gangadharan
VP
Rajasekharan
PG
Krishnan
NM
Clinical implications of cytogenetic classification in adult acute lymphoblastic leukaemia patients.
J Cancer Res Clin Oncol
122
1996
370
29
Heerema NA: Cytogenetic abnormalities and molecular markers of acute lymphoblastic leukemia, in Pochebly C, Civin CI (eds): Childhood Acute Lymphoblastic Leukemia—Part I. Hematol Oncol Clin North Am 4:795, 1990
30
Harris
MB
Shuster
JJ
Carroll
A
Look
AT
Borowitz
MJ
Crist
WM
Nitschke
R
Pullen
J
Steuber
CP
Land
VJ
Trisomy of leukemic cell chromosomes 4 and 10 identifies children with B-progenitor cell acute lymphoblastic leukemia with a very low risk of treatment failure: A Pediatric Oncology Group study.
Blood
79
1992
3316
31
(abstr, suppl 1).
Campbell
LJ
Michael
PM
White
JS
Matthews
JP
Prognostic implications of karyotypes in 159 newly diagnosed adult patients with acute lymphoblastic leukaemia.
Blood
86
1995
678a
32
(abstr, suppl 1)
Walters
R
Kantarjian
HM
Keating
M
Estey
E
Beran
M
Andersson
B
Koller
C
McCredie
K
Freireich
E
Response and prognosis related to pretreatment bone marrow cytogenetics in adult acute lymphocytic leukemia.
Blood
70
1987
212a
33
Sandberg
AA
Acute lymphoblastic leukemia
Sandberg
AA
The Chromosomes in Human Cancer and Leukemia
2
1990
313
Elsevier
New York, NY
34
Mitelman
F
Acute lymphoblastic leukemia
Heim
S
Mitelman
F
Cancer Cytogenetics
2
1995
180
Wiley-Liss
New York, NY
35
Oshimura
M
Freeman
AI
Sandberg
AA
Chromosomes and causation of human cancers and leukemia. XXIII. Near-haploidy in acute leukemia.
Cancer
40
1977
1143
36
Third International Workshop on Chromosomes in Leukemia
Chromosomal abnormalities in acute lymphoblastic leukemia: Structural and numerical changes in 234 cases.
Cancer Genet Cytogenet
4
1981
101
37
Third International Workshop on Chromosomes in Leukemia
Clinical significance of chromosomal abnormalities in acute lymphoblastic leukemia.
Cancer Genet Cytogenet
4
1981
111
38
United Kingdom Cancer Cytogenetics Group
Primary, single, autosomal trisomies associated with haematological disorders.
Leuk Res
16
1992
841
39
Pettenati
MJ
Rao
N
Wofford
M
Shuster
JJ
Pullen
DJ
Ling
MP
Steuber
CP
Carroll
AJ
Presenting characteristics of trisomy 8 as the primary cytogenetic abnormality associated with childhood acute lymphoblastic leukemia.
Cancer Genet Cytogenet
75
1994
6
40
Secker-Walker
LM
Prognostic and biologic importance of chromosome findings in acute lymphoblastic leukemia.
Cancer Genet Cytogenet
49
1990
1
41
Raimondi
SC
Roberson
PK
Pui
C-H
Behm
FG
Rivera
GK
Hyperdiploid (47-50) acute lymphoblastic leukemia in children.
Blood
79
1992
3245
42
Betts
DR
Kingston
JE
Dorey
EL
Young
BD
Webb
D
Katz
FE
Gibbons
B
Monosomy 20: A nonrandom finding in acute lymphoblastic leukemia.
Genes Chromosomes Cancer
2
1990
182
43
Heim
S
Mitelman
F
Numerical chromosome aberrations in human neoplasia.
Cancer Genet Cytogenet
22
1986
99
44
Garipidou
V
Yamada
T
Prentice
HG
Secker-Walker
LM
Trisomy 8 in acute lymphoblastic leukemia (ALL): A case report and update of the literature.
Leukemia
4
1990
717
45
Silengo
M
Vassallo
E
Barisone
E
Miniero
R
Madon
E
Monosomy 20 in childhood acute lymphoblastic leukemia.
Cancer Genet Cytogenet
59
1992
177
46
Raimondi
SC
Pui
C-H
Head
D
Behm
F
Privitera
E
Roberson
PK
Rivera
GK
Williams
DL
Trisomy 21 as the sole acquired chromosomal abnormality in children with acute lymphoblastic leukemia.
Leukemia
6
1992
171
47
Berger
R
Acute lymphoblastic leukemia and chromosome 21.
Cancer Genet Cytogenet
94
1997
8
48
Mitelman
F
Heim
S
Quantitative acute leukemia cytogenetics.
Genes Chromosomes Cancer
5
1992
57
49
Schiffer
CA
Lee
ET
Tomiyasu
T
Wiernik
PH
Testa
JR
Prognostic impact of cytogenetic abnormalities in patients with de novo acute nonlymphoblastic leukemia.
Blood
73
1989
263
50
Sandberg
AA
Morgan
R
Jani Sait
S
Berger
R
Flandrin
G
Schrier
S
Hecht
F
Trisomy 4: An entity within acute nonlymphocytic leukemia.
Cancer Genet Cytogenet
26
1987
117
51
Kwong
YL
Ha
SY
Liu
HW
Chan
LC
Trisomy 4 may occur in a broad range of hematologic malignancies.
Cancer Genet Cytogenet
69
1993
139
52
Hodohara
K
Fujiyama
Y
Inoue
T
Niwakawa
M
Kitoh
K
Andoh
A
Bamba
T
Hosoda
S
Abe
T
Trisomy 4 in a case of acute lymphocytic leukemia (L1).
Cancer Genet Cytogenet
62
1992
88
53
Zollino
M
Leone
G
Sica
S
Bajer
J
Neri
G
Trisomy 4 in acute myeloblastic and acute lymphoblastic leukemia.
Cancer Genet Cytogenet
65
1993
115
54
Sandberg
AA
Morgan
SS
Morgan
R
Boros
L
Trisomy 5 as sole anomaly in acute lymphoblastic leukemia.
Cancer Genet Cytogenet
36
1988
31
55
Nagesh Rao
P
Buss
D
Brown
S
O'Connor
M
Hurd
D
Pettenati
J
Trisomy 5 as the sole abnormality in acute lymphoblastic leukemia.
Cancer Genet Cytogenet
75
1994
117
56
(letter)
Chen
Z
Morgan
R
Sandberg
AA
Non-random involvement of chromosome 5 in ALL.
Cancer Genet Cytogenet
61
1992
106
57
(abstr, suppl 1)
Dabaja
BS
Kantarjian
H
O'Brien
S
Estey
E
Keating
M
Deletion or losses in chromosomes 5 or 7 in adults with acute lymphocytic leukemia: Incidence, associations and implications.
Blood
90
1997
211b
58
(letter)
Bonet
C
Solé
F
Woessner
S
Florensa
L
Besses
C
Sans-Sabrafen
J
A case of monosomy 20 in an adult acute lymphoblastic leukemia.
Cancer Genet Cytogenet
69
1993
165
59
Nowell
PC
Hungerford
DA
A minute chromosome in human chronic granulocytic leukemia.
Science
132
1960
1497
60
Propp
S
Lizzi
FA
Philadelphia chromosome in acute lymphocytic leukemia.
Blood
36
1970
353
61
Melo
JV
The diversity of bcr-abl fusion proteins and their relationship to leukemia phenotype.
Blood
88
1996
2375
62
Kurzrock
R
Gutterman
JU
Talpaz
M
The molecular genetics of Philadelphia chromosome-positive leukemias.
N Engl J Med
319
1988
990
63
Rowley
JD
Chromosome abnormalities in human leukemia.
Annu Rev Genet
14
1980
17
64
Berger
R
Chen
SJ
Chen
Z
Philadelphia-positive leukemia. Cytogenetic and molecular aspects.
Cancer Genet Cytogenet
44
1990
143
65
Rowley
JD
A new consistent chromosomal abnormality in chronic myelogenous leukemia identified by quinacrine fluorescence and Giemsa staining.
Nature
243
1973
290
66
Bartram
CR
de Klein
A
Hagemeijer
A
van Agthoven
T
van Kessel
AG
Bootsma
D
Grosveld
G
Ferguson-Smith
MA
Davies
T
Stone
M
Heisterkamp
N
Stephenson
JR
Groffen
J
Translocation of c-abl oncogene correlates with the presence of a Philadelphia chromosome in chronic myelocytic leukemia.
Nature
306
1983
277
67
Soekarman
D
van Denderen
J
Hoefsloot
L
Moret
M
Meeuwsen
T
van Baal
Hagemeijer
A
Grosveld
G
A novel variant of the bcr-abl fusion product in Philadelphia chromosome-positive acute lymphoblastic leukemia.
Leukemia
4
1990
397
68
de Klein
A
Hagemeijer
A
Bartram
CR
Houwen
R
Hoefsloot
L
Carbonell
F
Chen
L
Barnett
M
Greaves
M
Kleihauer
E
Heisterkamp
N
Groffen
J
Grosvels
G
Bcr rearrangement and translocation of the c-abl oncogene in Philadelphia positive acute lymphoblastic leukemia.
Blood
68
1986
1369
69
Kurzrock
R
Shtalrid
M
Romero
P
Kloetzer
WS
Talpaz
M
Trujillo
JM
Blick
M
Beran
M
Gutterman
JU
A novel c-abl protein product in Philadelphia-positive acute lymphoblastic leukemia.
Nature
325
1987
631
70
Saglio
G
Guerrasio
A
Rosso
C
Zaccaria
A
Tassinari
A
Serra
A
Rege-Cambrin
G
Mazza
U
Gavosto
F
New type of Bcr/Abl junction in Philadelphia chromosome-positive chronic myelogenous leukemia.
Blood
76
1990
1819
71
Tuszynski
A
Dhut
S
Young
BD
Lister
TA
Rohatiner
AZ
Amess
JAL
Chaplin
T
Dorey
E
Gibbons
B
Detection and significance of bcr-abl mRNA transcripts and fusion proteins in Philadelphia-positive adult acute lymphoblastic leukemia.
Leukemia
7
1993
1504
72
Saglio
G
Guerrasio
A
Rosso
C
Lo Coco
F
Frontani
M
Annino
L
Mandelli
F
Detection of Ph′-positive acute lymphoblastic leukemia by PCR.
Lancet
338
1991
958
73
Westbrook
CA
Hooberman
L
Spino
C
Dodge
RK
Larson
RA
Davey
F
Wurster-Hill
DH
Sobol
RE
Schiffer
C
Bloomfield
C
Clinical significance of the bcr-abl fusion gene in adult acute lymphoblastic leukemia: A Cancer and Leukemia Group B Study (8762).
Blood
80
1992
2983
74
Tkachuk
DC
Westbrook
CA
Andreeff
M
Donlon
TA
Cleary
ML
Suryanarayan
K
Homge
M
Redner
A
Gray
J
Pinkel
D
Detection of bcr-abl fusion in chronic myelogenous leukemia by in situ hybridization.
Science
250
1990
539
75
Maurer
J
Janssen
JWG
Thiel
E
van Denderen
J
Ludwig
W-D
Aydemir
U
Heinze
B
Fonatsch
C
Harbott
J
Reiter
A
Riehm
H
Hoelzer
D
Bartram
CR
Detection of chimeric bcr-abl genes in acute lymphoblastic-leukemia by the polymerase chain reaction.
Lancet
337
1991
1055
76
Hooberman
AL
Rubin
CM
Barton
KP
Westbrook
CA
Detection of the Philadelphia chromosome in acute lymphoblastic leukemia by pulsed-field gel electrophoresis.
Blood
74
1989
1101
77
Kantarjian
HM
Talpaz
M
Dhingra
K
Estey
E
Keating
MJ
Ku
S
Trujillo
J
Huh
Y
Stass
S
Kurzrock
R
Significance of the p210 versus p190 molecular abnormalities in adults with Philadelphia chromosome-positive acute leukemia.
Blood
78
1991
2411
78
Kurzrock
R
Shtalrid
M
Gutterman
JU
Koller
CA
Walters
R
Trujillo
JM
Talpaz
M
Molecular analysis of chromosome 22 breakpoints in adult Philadelphia-positive acute lymphoblastic leukaemia.
Br J Haematol
67
1987
55
79
Secker-Walker
LM
Craig
JM
Prognostic implications of breakpoint and lineage heterogeneity in Philadelphia-positive acute lymphoblastic leukemia: A review.
Leukemia
7
1993
147
80
Secker-Walker
LM
Craig
JM
Hawkins
JM
Hoffbrand
AV
Philadelphia positive acute lymphoblastic leukemia in adults: Age distribution, bcr breakpoint and prognostic significance.
Leukemia
5
1991
196
81
Gale
RP
Butturini
A
Ph-chromosome positive acute leukemias and acute phase CML: One or two diseases? Two.
Leuk Res
14
1990
295
82
Van Rhee
F
Hochhaus
A
Lin
F
Melo
JV
Goldman
JM
Cross
N
p190 bcr-abl mRNA is expressed at low levels in p210-positive chronic myeloid and acute lymphoblastic leukemias.
Blood
87
1996
5213
83
Radich
J
Gehly
G
Lee
A
Avery
R
Bryant
E
Edmands
S
Gooley
T
Kessler
P
Kirk
J
Ladne
P
Thomas
ED
Appelbaum
FR
Detection of bcr-abl transcripts in Philadelphia chromosome-positive acute lymphoblastic leukemia after bone marrow transplantation.
Blood
89
1997
2602
84
Preti
HA
O'Brien
S
Giralt
S
Beran
M
Pierce
S
Kantarjian
HM
Philadelphia-chromosome-positive adult acute lymphocytic leukemia: Characteristics, treatment results, and prognosis in 41 patients.
Am J Med
97
1994
60
85
Bloomfield
CD
Peterson
LC
Yunis
JJ
Brunning
RD
The Philadelphia chromosome (Ph1) in adults presenting with acute leukemia: A comparison of Ph1+ and Ph1− patients.
Br J Haematol
36
1977
347
86
Specchia
G
Mininni
A
Guerrasio
A
Palumbo
G
Pastore
D
Liso
V
Ph positive acute lymphoblastic leukemia in adults: Molecular and clinical studies.
Leuk Lymphoma
18
1995
37
87
(letter)
Colleoni
GWB
Yamamoto
M
Kerbauy
J
Serafim
RC
Lindsey
CJ
Costa
FF
Grignolli
CRE
Saad
STO
Bcr-abl rearrangement in adult T-cell acute lymphoblastic leukemia.
Am J Hematol
53
1996
277
88
Crist
W
Carroll
A
Shuster
J
Jackson
J
Head
D
Borowitz
M
Behm
F
Link
M
Steuber
P
Ragab
A
Hirt
A
Brock
B
Land
V
Pullen
J
Philadelphia chromosome positive childhood acute lymphoblastic leukemia: Clinical and cytogenetic characteristics and treatment outcome. A Pediatric Oncology Group (POG) Study.
Blood
76
1990
489
89
Ribeiro
RC
Abromowitch
M
Raimondi
SC
Murphy
SB
Behm
F
Williams
DL
Clinical and biologic hallmarks of the Philadelphia chromosome in childhood acute lymphoblastic leukemia.
Blood
70
1987
948
90
Copelan
EA
McGuire
EA
The biology and treatment of acute lymphoblastic leukemia in adults.
Blood
5
1995
1151
91
Radich
J
Sanders
J
Buckner
CD
Martin
PJ
Petersen
FB
Hansen
JA
Second allogeneic bone marrow transplants for patients relapsing after initial transplant with TBI-containing regimens.
J Clin Oncol
11
1993
304
92
Kowalczyk
J
Sandberg
AA
A possible subgroup of ALL with 9p−.
Cancer Genet Cytogenet
9
1983
383
93
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
94
Pollak
C
Hagemeijer
A
Abnormalities of the short arm of chromosome 9 with partial loss of material in hematological disorders.
Leukemia
1
1987
541
95
Diaz
MO
Rubin
CM
Harden
A
Ziemin
S
Larson
RA
LeBeau
MM
Rowley
JD
Deletions of interferon genes in acute lymphoblastic leukemia.
N Engl J Med
322
1990
77
96
Laı̈
JL
Fenaux
P
Pollet
JP
Estienne
MH
Savary
JB
Huart
JJ
Deminatti
M
Acute lymphocytic leukemia with 9p anomalies.
Cancer Genet Cytogenet
33
1988
99
97
Cayuela
J-M
Madani
A
Sanhes
L
Stern
M-H
Sigaux
F
Multiple tumor-suppressor gene 1 inactivation is the most frequent genetic alteration in T-cell acute lymphoblastic leukemia.
Blood
87
1996
2180
98
Liu
Q
Neuhausen
S
McClure
M
Frye
C
Weaver-Feldhaus
J
Gruis
NA
Eddington
K
Allalunis-Turner
MJ
Skolnick
MH
Fujimura
FK
Kamb
A
CDKN 2 (MTS1) tumor suppressor gene mutations in human tumor cell lines.
Oncogene
10
1995
1061
99
Olopade
OI
Jenkins
RB
Ransom
DT
Malik
K
Pomykala
H
Nobori
T
Cowan
JM
Rowley
JD
Diaz
MO
Molecular analysis of deletions of the short arm of chromosome 9 in human gliomas.
Cancer Res
52
1992
2523
100
Della Ragione
F
Mercurio
C
Iolascon
A
Cell cycle regulation and human leukemias: The role of p16INK4 gene inactivation in the development of human acute lymphoblastic leukemia.
Haematologica
80
1995
557
101
Trent
JM
Olson
S
Lawn
RM
Chromosomal localization of human leukocyte, fibroblast, and immune interferon genes by means of in situ hybridization.
Proc Natl Acad Sci USA
79
1982
7809
102
Carrera
CJ
Eddy
RL
Shows
TB
Carson
DA
Assignment of the gene for methylthioadenosine phosphorylase to human chromosome 9 by mouse-human somatic cell hybridization.
Proc Natl Acad Sci USA
81
1984
2665
103
Diaz
MO
Ziemin
S
Le Beau
MM
Pitha
P
Smith
SD
Chilcote
RR
Rowley
JD
Homozygous deletion of the α- and β-interferon genes in human leukemia and derived cell lines.
Proc Natl Acad Sci USA
85
1988
5259
104
Traweek
ST
Riscoe
MK
Ferro
AJ
Braziel
RM
Magenis
RE
Fichten
JH
Methylthioadenosine phosphorylase deficiency in acute leukemia: Pathologic, cytogenetic, and clinical features.
Blood
71
1988
1568
105
Kamb
A
Gruis
NA
Weaver-Feldhaus
J
Liu
Q
Harshman
K
Tavtigian
SV
Stockert
E
Day RS
III
Johnson
BE
Skolnick
MH
A cell cycle regulator potentially involved in genesis of many tumor types.
Science
264
1994
436
106
Nobori
T
Miura
K
Wu
DJ
Lois
A
Takabayashi
K
Carson
DA
Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers.
Nature
368
1994
753
107
DelSal
G
Loda
M
Pagano
M
Cell cycle and cancer: Critical events at the G1 restriction point.
Crit Rev Oncog
7
1996
127
108
Quesnel
B
Preudhomme
C
Fenaux
P
p16ink4a gene and hematological malignancies.
Leuk Lymphoma
22
1996
11
109
Hebert
J
Cayuela
J-M
Berkeley
J
Sigaux
F
Candidate tumor-suppressor genes MTS1 (p16INK4A) and MTS2 (p15INK4B) display frequent homozygous deletions in primary cells from T- but not from B-cell lineage acute lymphoblastic leukemias.
Blood
84
1994
4038
110
(letter)
Cayuela
J-M
Hebert
J
Sigaux
F
Homozygous MTS1 (p16INK4A) deletion in primary tumor cells of 163 leukemic patients.
Blood
85
1995
854
111
Haidar
MA
Cao
X-B
Manshouri
T
Chan
LL
Glassman
A
Kantarjian
HM
Keating
MJ
Beran
MS
Albitar
M
p16INK4A and p15INK4B gene deletions in primary leukemias.
Blood
86
1995
311
112
Ogawa
S
Hangaishi
A
Miyawaki
S
Hirosawa
S
Miura
Y
Takeyama
K
Kamada
N
Ohtake
S
Uike
N
Shimazaki
C
Toyama
K
Hirano
M
Mizoguchi
H
Kobayashi
Y
Furusawa
S
Saito
M
Emi
N
Yazaki
Y
Ueda
R
Hirai
H
Loss of the cyclin-dependent kinase 4-inhibitor (p16; MTS1) gene is frequent in and highly specific to lymphoid tumors in primary human hematopoietic malignancies.
Blood
86
1995
1548
113
Quesnel
B
Preudhomme
C
Philippe
N
Vanrumbeke
M
Dervite
I
Laı̈
JL
Bauters
F
Wattel
E
Fenaux
P
p16 gene homozygous deletions in acute lymphoblastic leukemia.
Blood
85
1995
657
114
Rasool
O
Heyman
M
Borgonovo Brandter
L
Liu
Y
Grandér
D
Söderhäll
S
Einhorn
S
p15ink4B and p16ink4 gene inactivation in acute lymphocytic leukemia.
Blood
85
1995
3431
115
Dreyling
MH
Bohlander
SK
Le Beau
MM
Olopade
OI
Refined mapping of genomic rearrangements involving the short arm of chromosome 9 in acute lymphoblastic leukemias and other hematologic malignancies.
Blood
86
1995
1931
116
Herman
JG
Jen
J
Merlo
A
Baylin
SB
Hypermethylation-associated inactivation indicates a tumor suppressor role for p15INK4B1.
Cancer Res
56
1996
722
117
Batova
A
Diccianni
MB
Yu
JC
Nobori
T
Link
MP
Pullen
J
Yu
AL
Frequent and selective methylation of p15 and deletion of p16 in T-cell acute lymphoblastic leukemia.
Cancer Res
57
1997
832
118
Schröder
M
Mathieu
U
Dreyling
MH
Bohlander
SK
Hagemeijer
A
Beverloo
BH
Olopade
OI
Stilgenbauer
S
Fischer
K
Bentz
M
Lichter
P
Döhner
H
CDKN2 gene deletion is not found in chronic lymphoid leukaemias of B- and T-cell origin but is frequent in acute lymphoblastic leukaemia.
Br J Haematol
91
1995
865
119
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
120
Carroll
AJ
Raimondi
SC
Williams
DL
Behm
FG
Borowitz
M
Castleberry
RP
Harris
MB
Patterson
RB
Pullen
DJ
Crist
WM
tdic(9;12): A nonrandom chromosome abnormality in childhood B-cell precursor acute lymphoblastic leukemia: A Pediatric Oncology Group Study.
Blood
70
1987
1962
121
United Kingdom Cancer Cytogenetics Group
Translocations involving 9p and/or 12p in acute lymphoblastic leukemia.
Genes Chromosomes Cancer
5
1992
255
122
Mahmoud
H
Carroll
AJ
Behm
F
Raimondi
SC
Schuster
J
Borowitz
M
Land
V
Pullen
DJ
Vietti
TJ
Crist
W
The non-random dic(9;12) translocation in acute lymphoblastic leukemia is associated with B-progenitor phenotype and an excellent prognosis.
Leukemia
6
1992
703
123
Behrendt
H
Charrin
C
Gibbons
B
Harrison
CJ
Hawkins
JM
Heerema
NA
Horschler-Bötel
B
Huret
J-L
Laı̈
JL
Lampert
F
Nelken
B
Perot
C
Ritterbach
J
Schlegelberger
B
Secker-Walker
LM
Slater
R
Slovak
M
Tanzer
J
Van Den Akker
J
Dicentric (9;12) in acute lymphocytic leukemia and other hematological malignancies: Report from a dic(9;12) study group.
Leukemia
9
1995
102
124
Chen
C-S
Sorenson
PHB
Domer
PH
Reaman
GH
Korsmeyer
SJ
Heerema
NA
Hammond
GD
Kersey
JH
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
125
Janssen
JWG
Ludwig
W-D
Borkhardt
A
Spadinger
U
Rieder
H
Fonatsch
C
Hossfeld
DK
Harbott
J
Schulz
AS
Repp
R
Sykora
K-W
Hoelzer
D
Bartram
CR
Pre-pre-B acute lymphoblastic leukemia: High frequency of alternatively spliced ALL1-AF4 transcripts and absence of minimal residual disease during complete remission.
Blood
84
1994
3835
126
Kaneko
Y
Skikano
T
Maseki
N
Sakurai
M
Takeda
T
Hiyoshi
Y
Fujimoto
T
Clinical characteristics of infant acute leukemia with or without 11q23 translocations.
Leukemia
2
1988
672
127
Super
HJ
McCabe
NR
Thirman
MJ
Larson
RA
Le Beau
MM
Pedersen-Bjergaard
J
Philip
P
Diaz
MO
Rowley
JD
Rearrangements of the MLL gene in therapy-related acute myeloid leukemia in patients previously treated with agents targeting DNA-topoisomerase II.
Blood
82
1993
3705
128
Ohshima
A
Miura
I
Chubaki
A
Hashimoto
K
Nimura
T
Utsumi
S
Takahashi
N
Hayashi
Y
Seto
M
Ueda
R
Miura
AB
11q23 aberration is an additional chromosomal change in de novo acute leukemia after treatment with etoposide and mitoxantrone.
Am J Hematol
53
1996
264
129
Ziemin-van der Poel
S
McCabe
NR
Gill
HJ
Espinosa
R III
Patel
Y
Harden
A
Rubinelli
P
Smith
SD
LeBeau
MM
Rowley
JD
Diaz
MO
Identification of a gene, MLL, that spans the breakpoint in 11q23 translocations associated with human leukemias.
Proc Natl Acad Sci USA
88
1991
10735
130
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
2
1992
113
131
Tkachuk
DC
Kohler
S
Cleary
ML
Involvement of a homolog of Drosophila trithorax by 11q23 chromosomal translocations in acute leukemias.
Cell
71
1992
701
132
Reeves
R
Nissen
MS
The AT-DNA-binding domain of mammalian high-morbidity-group I chromosomal proteins.
J Biol Chem
265
1990
8573
133
Rubnitz
JE
Behm
FG
Downing
JR
11q23 rearrangements in acute leukemia.
Leukemia
10
1996
74
134
Bernard
OA
Berger
R
Molecular basis of 11q23 rearrangements in hematopoietic malignant proliferations.
Genes Chromosomes Cancer
13
1995
75
135
Thirman
MJ
Gill
HJ
Burnett
RC
Mbangkollo
D
McCabe
NR
Kobayashi
H
Ziemin-van der Poel
S
Kaneko
Y
Morgan
R
Sandberg
AA
Chaganti
RSK
Larson
RA
Le Beau
M
Diaz
MO
Rowley
JD
Rearrangement of the MLL gene in acute lymphoblastic and acute myeloid leukemias with 11q23 chromosomal translocations.
N Engl J Med
329
1993
909
136
Stock
W
Thirman
MJ
Dodge
RK
Rowley
JD
Diaz
MO
Wurster-Hill
D
Sobol
RE
Davey
FR
Larson
RA
Westbrook
CA
Bloomfield
CD
Detection of MLL gene rearrangements in adult acute lymphoblastic leukemia. A Cancer and Leukemia Group B Study.
Leukemia
8
1994
1918
137
Griesinger
F
Ludwig
W-D
Falk
M
Rieder
H
Harbott
J
Lampert
F
Heinze
B
Hoelzer
D
Thiel
E
Riehm
H
Wörmann
B
Fonatsch
C
Hiddemann
W
Detection of HRX-FEL fusion transcripts in pre-pre-B-ALL with and without cytogenetic demonstration of t(4;11).
Leukemia
8
1994
542
138
Zeleznik-Le
NJ
Harden
AM
Rowley
JD
11q23 translocations split the ‘AT-hook’ cruciform DNA-binding region and the transcriptional repression domain from the activation domain of the mixed-lineage leukemia (MLL) gene.
Proc Natl Acad Sci USA
91
1994
10610
139
Rogaia
D
Grignani
F
Carbone
R
Riganelli
D
Lo Coco
F
Nakamura
T
Croce
CM
Di Fiore
PP
Pelicci
PG
The localization of the HRX/ALL1 protein to specific subnuclear domains is altered by fusion with its eps15 translocation partner.
Cancer Res
57
1997
799
140
Oshimura
M
Freeman
AI
Sandberg
AA
Chromosomes and causation of human cancer and leukemia. XXVI. Banding studies in acute lymphoblastic leukemia (ALL).
Cancer
40
1977
1161
141
De Braekeleer
M
Lin
CC
4;11 translocation-associated acute leukemia: A comprehensive analysis.
Cancer Genet Cytogenet
21
1986
53
142
Arthur
DC
Bloomfield
CD
Linquist
LL
Nesbit
MEJ
Translocation 4;11 in acute lymphoblastic leukemia: Clinical characteristics and prognostic significance.
Blood
67
1982
1
143
Fidanza
V
Melotti
P
Yano
T
Nakamura
T
Bradley
A
Canaani
E
Calabretta
B
Croce
CM
Double knockout of the ALL-1 gene blocks hematopoietic differentiation in vitro.
Cancer Res
56
1996
1179
144
Rabbitts
TH
Chromosomal translocations in human cancer.
Nature
372
1994
143
145
Carroll
AJ
Crist
WM
Parmley
RT
Roper
M
Cooper
MD
Finley
WH
Pre-B cell leukemia associated with chromosome translocation 1;19.
Blood
63
1984
721
146
Pui
C-H
Crist
WM
Cytogenetic abnormalities in childhood acute lymphoblastic leukemia correlate with clinical features and treatment outcome.
Leuk Lymphoma
7
1992
259
147
Raimondi
SC
Behm
FG
Roberson
PK
Williams
DL
Pui
C-H
Crist
WM
Look
AT
Rivera
GK
Cytogenetics of pre-B acute lymphoblastic leukemia with emphasis on prognostic implications of the t(1;19).
J Clin Oncol
8
1990
1380
148
Crist
WM
Carroll
AJ
Shuster
JJ
Behm
FG
Whitehead
M
Vietti
TJ
Look
AT
Mahoney
D
Ragab
A
Pullen
DJ
Land
VJ
Poor prognosis of children with pre-B acute lymphoblastic leukemia is associated with the t(1;19)(q23;p13). A Pediatric Oncology Group Study.
Blood
76
1990
117
149
Pui
C-H
Childhood leukemias.
N Engl J Med
332
1995
1618
150
Devaraj
PE
Foroni
L
Hoffbrand
AV
Secker-Walker
LM
Expression of the E2A-PBX1 fusion transcripts in t(1;19)(q23;p13) and der(19)t(1;19) at diagnosis and in remission of acute lymphoblastic leukemia with different B lineage immunophenotypes.
Leukemia
9
1995
821
151
Cortes
JE
Kantarjian
HM
Acute lymphoblastic leukemia.
Cancer
76
1995
2393
152
Shikano
T
Kaneko
Y
Takazawa
M
Ueno
N
Ohkawa
M
Fujimoto
T
Balanced and unbalanced 1;19 translocation-associated acute lymphoblastic leukemias.
Cancer
58
1986
2239
153
Pui
C-H
Raimondi
SC
Hancock
ML
Rivera
GK
Ribeiro
RC
Mahmoud
HH
Sandlund
JT
Crist
WM
Behm
FG
Immunologic, cytogenetic, and clinical characterization of childhood acute lymphoblastic leukemia with the t(1;19)(q23;p13) or its derivative.
J Clin Oncol
12
1994
2601
154
Rowley
JD
Abnormalities of chromosome no.1: Significance in malignant transformation.
Virchows Arch B Cell Path
29
1978
139
155
Mamaeva
SE
Mamaev
NN
Jartseva
NM
Belyaeva
LV
Scherbakova
EG
Complete or partial trisomy for the long arm of chromosome 1 in patients with various hematologic malignancies.
Hum Genet
63
1983
107
156
Mellentin
JD
Murre
C
Donlon
TA
McCaw
PS
Smith
SD
Carroll
AJ
McDonald
ME
Baltimore
D
Cleary
ML
The gene for enhancer binding proteins E12/E47 lies at the t(1;19) breakpoint in acute leukemias.
Science
246
1989
379
157
Nichols
J
Nimer
D
Transcription factors, translocations, and leukemia.
Blood
80
1992
2953
158
Hunger
S
Chromosomal translocations involving the E2A gene in acute lymphoblastic leukemia: Clinical features and molecular pathogenesis.
Blood
87
1996
1211
159
Mellentin
JD
Nourse
J
Hunger
SP
Smith
SD
Cleary
ML
Molecular analysis of the t(1;19) breakpoint cluster region in pre-B-ALL.
Genes Chromosomes Cancer
2
1990
239
160
Lu
Q
Wright
DD
Kamps
MP
Fusion with E2A converts the Pbx1 homeodomain protein into a constitutive transcriptional activator in human leukemias carrying the t(1;19).
Mol Cell Biol
14
1994
3938
161
LeBrun
DP
Cleary
ML
Fusion with E2A alters the transcriptional properties of the homeodomain protein PBX1 in t(1;19) leukemias.
Oncogene
9
1994
1641
162
Privitera
E
Luciano
A
Ronchetti
D
Arico
M
Santostasi
T
Basso
G
Biondi
A
Molecular variants of the 1;19 chromosomal translocation in pediatric acute lymphoblastic leukemia.
Leukemia
8
1994
554
163
Izraeli
S
Kovar
H
Gadner
H
Lion
T
Unexpected heterogeneity in E2A/PBX1 fusion messenger RNA detected by the polymerase chain reaction in pediatric patients with acute lymphoblastic leukemia.
Blood
80
1992
1413
164
Monica
K
Galili
N
Nourse
J
Saltman
D
Cleary
ML
PBX2 and PBX3, new homeobox genes with extensive homology to the human proto-oncogene PBX1.
Mol Cell Biol
11
1991
6149
165
Borowitz
MJ
Hunger
SP
Carroll
AJ
Shuster
JJ
Pullen
DJ
Steuber
CP
Cleary
ML
Predictability of the t(1;19)(q23;p13) from surface antigen phenotype: Implications for screening cases of childhood acute lymphoblastic leukemia for molecular analysis: A Pediatric Oncology Group Study.
Blood
82
1993
1086
166
Inaba
T
Roberts
WM
Shapiro
LH
Jolly
KW
Raimondi
SC
Smith
SD
Look
AT
Fusion of the leucine zipper gene HLF to the E2A gene in human acute B-lineage leukemia.
Science
257
1992
531
167
Bernard
O
Barin
C
Charrin
C
Mathieu-Mahul
D
Berger
R
Characterization of translocation t(1;14)(p32;q11) in a T and in a B acute leukemia.
Leukemia
7
1993
1509
168
Uckun
FM
Gajl-Peczalska
KJ
Provisor
AJ
Heerema
NA
Immunophenotype-karyotype associations in human acute lymphoblastic leukemia.
Blood
73
1989
271
169
Raimondi
SC
Behm
FG
Roberson
PK
Pui
C-H
Rivera
GK
Murphy
SB
Williams
DL
Cytogenetics of childhood T-cell leukemia.
Blood
72
1988
1560
170
Human gene mapping 8
Helsinki conference: Eighth International Workshop on Human Gene Mapping.
Cytogenet Cell Genet
40
1985
1
171
Fenaux
P
Laı̈
JL
Miaux
O
Zandecki
M
Jouet
JP
Bauters
F
Burkitt cell acute leukemia (L3 ALL) in adults: A report of 18 cases.
Br J Haematol
71
1989
371
172
Baer
R
TAL1, TAL2 and LYL1: A family of basic helix-loop-helix proteins implicated in T cell acute leukemia.
Cancer Biol
4
1993
341
173
Sánchez-Garcı́a
I
Rabbitts
TH
LIM domain proteins in leukemia and development.
Cancer Biol
4
1993
349
174
Breit
TM
Mol
EJ
Wolvers-Tettero
ILM
Ludwig
W-D
van Wering
ER
van Dongen
JM
Site-specific deletions involving the tal-1 and sil genes are restricted to cells of the T-cell receptor α/β lineage: T cell receptor δ gene deletion mechanism affects multiple genes.
J Exp Med
177
1993
965
175
Aplan
PD
Raimondi
SC
Kirsch
IR
Disruption of the SCL gene by a t(1;3) translocation in a patient with T-cell acute lymphoblastic leukemia.
J Exp Med
176
1992
1303
176
Janssen
JWG
Ludwig
W-D
Sterry
W
Bartram
CR
SIL-TAL1 deletion in T-cell acute lymphoblastic leukemia.
Leukemia
8
1993
1204
177
Bash
RO
Hall
S
Timmons
CF
Crist
WM
Amylon
M
Graham Smith
R
Baer
R
Does activation of the TAL1 gene occur in a majority of patients with T-cell acute lymphoblastic leukemia? A Pediatric Oncology Group Study.
Blood
86
1995
666
178
Kwong
YL
Chan
D
Liang
R
SIL/TAL1 recombination in adult T-acute lymphoblastic leukemia and T-lymphoblastic lymphoma.
Cancer Genet Cytogenet
85
1995
159
179
van Dongen
JM
Breit
TM
Adriaansen
HJ
Beishuizen
A
Hooijkaas
H
Detection of minimal residual disease in ALL.
Recent Results Cancer Res
131
1993
157
180
Wadman
I
Li
J
Bash
RO
Forster
A
Osada
H
Rabbitts
TH
Baer
R
Specific in vivo association between the bHLH and LIM proteins implicated in human T cell leukemia.
EMBO J
13
1994
4831
181
Shivdasani
RA
Mayer
EL
Orkin
SH
Absence of blood formation in mice lacking the T-cell leukemia oncoprotein tal-1/SCL.
Nature
373
1995
432
182
Ono
Y
Fukuhara
N
Yoshie
O
Transcriptional activity of TAL1 in T cell acute lymphoblastic leukemia (T-ALL) requires RBTN1 or -2 and induces TALLA1, a highly specific tumor marker of T-ALL.
J Biol Chem
272
1997
4576
183
Stock
W
Westbrook
CA
Sher
D
Dodge
R
Sobol
RE
Wurster-Hill
D
Davey
FR
Larson
RA
LeBeau
MM
Aplan
PD
Frankel
SR
Stewart
CC
Bloomfield
CD
Low incidence of TAL1 gene rearrangement in adult acute lymphoblastic leukemia: A Cancer and Leukemia Group B Study (8762).
Clin Cancer Res
1
1995
459
184
Bash
RO
Crist
WM
Shuster
JJ
Link
MP
Amylon
M
Pullen
J
Carroll
AJ
Buchanan
GR
Graham Smith
R
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
8
1993
2110
185
Kikuchi
A
Hayashi
Y
Kobayashi
S
Hanada
R
Moriwaki
K
Yamamoto
K
Fujimoto
J-I
Kaneko
Y
Yamamori
S
Clinical significance of TAL1 gene alteration in childhood T-cell acute lymphoblastic leukemia and lymphoma.
Leukemia
7
1993
933
186
Dubé
ID
Raimondi
SC
Pi
D
Kalousek
DK
A new translocation t(10;14)(q24;q11) in T-cell neoplasia.
Blood
67
1986
1181
187
Hatano
M
Roberts
CWM
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
188
Dubé
ID
Kamel-Reid
S
Yuan
CC
Lu
M
Wu
X
Corpus
G
Raimondi
SC
Crist
WM
Carroll
AJ
Minowada
J
Baker
JB
A novel homeobox gene lies at the chromosome 10 breakpoint in lymphoid neoplasias with chromosomal translocation t(10;14).
Blood
11
1991
2996
189
Boehm
T
Foroni
L
Kennedy
M
Rabbitts
TH
The rhombotin gene belongs to a class of transcriptional regulators with a potential novel protein dimerization motif.
Oncogene
5
1990
1103
190
Boehm
T
Buluwela
L
Williams
D
White
L
Rabbitts
TH
A cluster of chromosome 11p13 translocations found via distinct D-D and D-D-J rearrangements of the human T cell receptor δ chain gene.
EMBO J
7
1988
2011
191
Erikson
J
Finger
L
Sun
L
Ar-Rushdi
A
Nishikura
K
Minowada
J
Finan
J
Emanuel
BS
Nowell
PC
Croce
CM
Deregulation of c-myc by translocation of the alpha-locus of the T-cell receptor in T-cell leukemias.
Science
232
1986
884
192
Mathieu-Mahul
D
Sigaux
F
Zhu
C
Bernheim
A
Mauchaffe
M
Daniel
M-T
Berger
R
Larsen
C-J
A t98;14)(q24;q11) translocation in a T-cell leukemia (L1-ALL) with c-myc and TCR-alpha chain locus rearrangements.
Int J Cancer
38
1986
835
193
Shima
EA
LeBeau
MM
McKeithan
TW
Minowada
J
Shove
LC
Mak
TW
Minden
MD
Rowley
JD
Diaz
MO
Gene encoding the α-chain of the T-cell receptor is moved immediately downstream of c-myc in a chromosomal 8;14 translocation in a cell line from a human T-cell leukemia.
Proc Natl Acad Sci USA
83
1986
3439
194
Berger
R
Bernheim
J
Broquet
JC
Daniel
MT
Flandrin
G
t(8;14) translocation in a Burkitt's type of lymphoblastic leukemia.
Br J Haematol
43
1979
87
195
Magrath
IT
Ziegler
JL
Bone marrow involvement in Burkitt's lymphoma and its relationship to acute B-cell leukemia.
Leuk Res
4
1979
33
196
Sullivan
MP
Pullen
DJ
Crist
WM
Brecher
M
Ramirez
I
Sabio
H
Borowitz
MJ
Head
DR
Cerezo
L
Shuster
JJ
Murphy
SB
Clinical and biologic heterogeneity of childhood B cell acute lymphocytic leukemia: Implications for clinical trials.
Leukemia
4
1990
6
197
Gladstone
B
Kadam
PR
Balsara
BR
Pai
SK
Gopal
R
Nair
CN
Parikh
PM
Saikia
T
Advani
SH
Cytogenetic studies on patients of acute lymphoblastic leukemia Burkitt's type with (8;14) & (14;18) translocations.
Indian J Med Res
99
1994
264
198
Laı̈
JL
Fenaux
P
Zandecki
M
Nelken
B
Huart
JJ
Deminatti
M
Cytogenetic studies in 30 patients with Burkitt's lymphoma of L3 acute lymphoblastic leukemia with special reference to additional chromosome abnormalities.
Ann Genet
32
1989
26
199
Croce
CM
Nowell
PC
Molecular basis of human B cell neoplasia.
Blood
65
1985
1
200
Korsmeyer
SJ
Chromosomal translocations in lymphoid malignancies reveal novel proto-oncogenes.
Annu Rev Immunol
10
1992
785
201
Wiman
KG
Clarkson
B
Hayday
AC
Saito
H
Tonegawa
S
Hayward
WS
Activation of a translocated c-myc gene: Role of structural alterations in the upstream region.
Proc Natl Acad Sci USA
81
1984
6798
202
Joos
S
Haluska
FG
Falk
MH
Henglein
B
Hameister
H
Croce
CM
Bornkamm
GW
Mapping chromosomal breakpoints of Burkitt's t(8;14) translocations far upstream of c-myc.
Cancer Res
52
1992
6547
203
Hollis
GF
Mitchell
KF
Battey
J
Potter
H
Taub
R
Lenoir
GM
Leder
P
A variant translocation places the λ immunoglobulin genes 3′ to the c-myc oncogene in Burkitt's lymphoma.
Nature
307
1984
752
204
Emanuel
BS
Selden
JR
Chaganti
RSK
Jhanwar
S
Nowell
PC
Croce
CM
The 2p breakpoint of a 2;8 translocation in Burkitt lymphoma interrupts the Vκ locus.
Proc Natl Acad Sci USA
81
1984
2444
205
Cesarman
E
Dalla-Favera
R
Bentley
D
Groudine
M
Mutations in the first exon are associated with altered transcription of c-myc in Burkitt lymphoma.
Science
238
1987
1272
206
Rabbitts
TH
Hamlyn
PH
Baer
R
Altered nucleotide sequences of a translocated c-myc gene in Burkitt lymphoma.
Nature
306
1983
760
207
Bhatia
K
Huppi
K
Spangler
G
Point mutations in the c-myc transactivation domain are common in Burkitt's lymphoma and mouse plasmacytomas.
Nature Genet
5
1993
55
208
Thandla
S
Aplan
P
Molecular biology of acute lymphocytic leukemia.
Semin Oncol
24
1997
45
209
Prigogina
EL
Fleischman
EW
Puchkova
GP
Kulagina
OE
Majakova
SA
Balakirev
SA
Frenkel
MA
Khvatova
NV
Peterson
IS
Chromosomes in acute leukemia.
Hum Genet
53
1979
5
210
Raimondi
SC
Williams
DL
Callihan
T
Peiper
S
Rivera
GK
Murphy
SB
Nonrandom involvement of the 12p12 breakpoint in chromosome abnormalities of childhood acute lymphoblastic leukemia.
Blood
68
1986
69
211
Cave
H
Gerard
B
Martin
E
Guidal
C
Devaux
I
Weissenbach
J
Elion
J
Vilmer
E
Grandchamp
B
Loss of heterozygosity in the chromosomal region 12p12-13 is very common in childhood acute lymphoblastic leukemia and permits the precise localization of a tumor-suppressor gene distinct from p27KIP1.
Blood
86
1995
3869
212
Stegmaier
K
Pendse
S
Barker
GF
Bray-Ward
P
Ward
DC
Montgomery
KT
Krauter
KS
Reynolds
C
Sklar
J
Donnelly
M
Bohlander
SK
Rowley
SD
Sallan
SE
Gilliland
DG
Golub
TR
Frequent loss of heterozygosity at the TEL gene locus in acute lymphoblastic leukemia of childhood.
Blood
86
1995
38
213
Aguiar
RC
Sohal
J
Van Rhee
F
Carapeti
M
Franklin
IM
Goldstone
AH
Goldman
JM
Cross
NC
TEL-AML1 fusion in acute lymphoblastic leukaemia of adults. M.R.C. Adult Leukaemia Working Party.
Br J Haematol
95
1996
673
214
Sato
Y
Suto
Y
Pietenpol
J
Golub
TR
Gilliland
DG
Roberts
JM
Trask
BJ
LeBeau
MM
Vogelstein
B
Rowley
JD
Bohlander
SK
TEL and KIP1 define the smallest region of deletions in 12p13 in hematopoietic malignancies.
Blood
86
1995
1525
215
Golub
TR
Barker
GF
Lovett
M
Gillilland
DG
Fusion of PDGF receptor β to a novel ets-like gene, tel, in chronic myelomonocytic leukemia with t(5;12) chromosomal translocation.
Cell
77
1994
307
216
Buijs
A
Sherr
S
van Baal
S
van Bezouw
S
van der Plas
D
van Kessel
A
Riegman
P
Deprez
R
Zwarhoff
E
Hagemeijer
A
Grosfeld
G
Translocation (12;22)(p13;q11) in myeloproliferative disorders results in fusion of the ETS-like TEL gene on 12p13 to the MN1 gene on 22q11.
Oncogene
10
1995
1511
217
Janssen
JWG
Ridge
SA
Papadopoulos
P
Cotter
F
Ludwig
W-D
Fonatsch
C
Rieder
H
Ostertag
W
Bartram
CR
Wiedemann
LM
The fusion of TEL and ABL in human acute lymphoblastic leukaemia is a rare event.
Br J Haematol
90
1995
222
218
Papadopoulos
P
Ridge
SA
Boucher
CA
Stocking
C
Wiedemann
LM
The novel activation of ABL by fusion to an ets-related gene, TEL.
Cancer Res
55
1995
34
219
Wlodarska
I
Mecucci
C
Marynen
P
Guo
C
Franckx
D
La Starza
R
Aventin
A
Bosley
MF
Martelli
JJ
Cassiman
JJ
Van den Berghe
H
TEL gene is involved in myelodysplastic syndromes with either the typical t(5;12)(q33;p13) or its variant t(10;12)(q24;p13).
Blood
85
1995
2848
220
Golub
TR
Barker
GF
Bohlander
SK
Hiebert
SW
Ward
DC
Bray-Ward
P
Morgan
E
Raimondi
SC
Rowley
JD
Gilliland
DG
Fusion of the TEL gene on 12p13 to the AML1 gene on 21q22 in acute lymphoblastic leukemia.
Proc Natl Acad Sci USA
92
1995
4917
221
Romana
SP
Mauchauffé
M
Le Coniat
M
Chumakov
I
Le Paslier
D
Berger
R
Bernard
OA
The t(12;21) of acute lymphoblastic leukemia results in a tel-AML1 gene fusion.
Blood
85
1995
3662
222
Golub
TR
Barker
GF
Stegmaier
K
Gilliland
DG
The TEL gene contributes to the pathogenesis of myeloid and lymphoid leukemias by diverse molecular genetic mechanisms.
Curr Top Microbiol Immunol
220
1997
67
223
Kim
D-H
Moldwin
RL
Vignon
C
Bohlander
SK
Suto
Y
Giordano
L
Gupta
R
Fears
S
Nucifora
G
Rowley
JD
Smith
SD
TEL-AML1 translocations with Tel and CDKN2 inactivation in acute lymphoblastic leukemia cell lines.
Blood
88
1996
785
224
Stegmaier
K
Takeuchi
S
Golub
TR
Bohlander
SK
Bartram
CR
Koeffler
HP
Gilliland
DG
Mutational analysis of the candidate tumor suppressor genes TEL and KIP1 in childhood acute lymphoblastic leukemia.
Cancer Res
56
1996
1413
225
Wlodarska
I
Baens
M
Peeters
P
Aerssens
J
Mecucci
C
Brock
P
Marynen
P
Van den Berghe
Biallelic alterations of both ETV6 and CDKN1B genes in a t(12;21) childhood acute lymphoblastic leukemia case.
Cancer Res
56
1996
2655
226
Golub TR, Barker GF, Stegmaier K, Gilliland DG: Involvement of the TEL gene in hematologic malignancy by diverse molecular genetic mechanisms, in Wolff L, Perkins AS (eds): Molecular Aspects of Myeloid Stem Cell Development. Curr Top Microbiol Immunol 211:279, 1996
227
Bernard
OA
Romana
SP
Poirel
H
Berger
R
Molecular cytogenetics of t(12;21)(p13;q22).
Leuk Lymphoma
23
1996
459
228
Raynaud
S
Cavé
H
Baens
M
Bastard
C
Cacheux
V
Grosgeorge
J
Guidal-Giroux
C
Guo
C
Vilmer
E
Marynen
P
Grandchamp
B
The 12;21 translocation involving TEL and deletion of the other TEL allele: Two frequently associated alterations found in childhood acute lymphoblastic leukemia.
Blood
87
1996
2891
229
Romana
SP
Poirel
H
Le Coniat
M
Flexor
M-A
Mauchauffé
M
Jonveaux
P
Macintyre
EA
Berger
R
Bernard
OA
High frequency of t(12;21) in childhood B-lineage acute lymphoblastic leukemia.
Blood
86
1995
4263
230
Fears
S
Vignon
C
Bohlander
SK
Smith
S
Rowley
JD
Nucifora
G
Correlation between the ETV6/CBFA2 (TEL/AML1) fusion gene and karyotypic abnormalities in children with B-cell precursor acute lymphoblastic leukemia.
Genes Chromosomes Cancer
17
1996
127
231
Borkhardt
A
Cazzaniga
G
Viehmann
S
Valsecchi
MG
Ludwig
W-D
Burci
L
Mangioni
S
Schrappe
M
Riehm
H
Lampert
F
Basso
G
Masera
G
Harbott
J
Biondi A for the “Associazione Italiana Ematologica Oncologia Pediatrica” and the “Berlin-Frankfurt-Münster” Study Group
Incidence and clinical relevance of TEL/AML1 fusion genes in children with acute lymphoblastic leukemia enrolled in the German and Italian multicenter therapy trials.
Blood
90
1997
571
232
Romana
SP
Le Coniat
M
Berger
R
t(12;21): A new recurrent translocation in acute lymphoblastic leukemia.
Genes Chromosomes Cancer
9
1994
186
233
Rubnitz
JE
Shuster
JJ
Land
VJ
Link
MP
Pullen
DJ
Camitta
BM
Pui
C-H
Downing
JR
Behm
FG
Case-control study suggests a favorable impact of TEL rearrangement in patients with B-lineage acute lymphoblastic leukemia treated with antimetabolite-based therapy: A Pediatric Oncology Group Study.
Blood
89
1997
1143
234
Rubnitz
JE
Downing
JR
Pui
C-H
Shurtleff
SA
Raimondi
SC
Evans
WE
Head
DR
Crist
WM
Rivera
GK
Hancock
ML
Boyett
JM
Bujis
A
Grosveld
G
Behm
FG
TEL gene rearrangement in acute lymphoblastic leukemia: A new genetic marker with prognostic significance.
J Clin Oncol
15
1997
1150
235
Shurtleff
SA
Bujis
A
Behm
FG
Rubnitz
JE
Raimondi
SC
Hancock
ML
Chan
G
Pui
C-H
Grosveld
G
Downing
JR
TEL/AML1 fusion resulting from a cryptic t(12;21) is the most common genetic lesion in pediatric ALL and defines a subgroup with an excellent prognosis.
Leukemia
9
1995
1985
236
McLean
T
Ringold
S
Neuberg
D
Stegmaier
K
Tantravahi
R
Ritz
J
Koeffler
HP
Takeuchi
S
Janssen
JWG
Seriu
T
Bartram
CR
Sallan
SE
Gilliland
DG
Golub
TR
TEL/AML-1 dimerizes and is associated with a favorable outcome in childhood acute lymphoblastic leukemia.
Blood
88
1996
4252
237
Lanza
C
Volpe
G
Basso
G
Gottardi
E
Barisone
E
Spinelli
M
Ricotti
E
Cilli
V
Perfetto
F
Madon
E
Saglio
G
Outcome and lineage involvement in t(12;21) childhood acute lymphoblastic leukaemia.
Br J Haematol
97
1997
460
238
Raynaud
S
Mauvieux
L
Cayuela
JM
Bastard
C
Bilhou-Nabera
C
Debuire
B
Bories
D
Boucheix
C
Charrin
C
Fière
D
Gabert
J
TEL/AML1 fusion gene is a rare event in adult acute lymphoblastic leukemia.
Leukemia
10
1996
1529
239
Aguiar
RC
Sohal
J
Van Rhee
F
Carapeti
M
Franklin
IM
Goldstone
AH
Goldman
JM
Cross
NCP
TEL-AML1 fusion in acute lymphoblastic leukaemia of adults.
on behalf of the MRC Adult Leukemia Working Party LRF Centre for Adult Leukaemia
Br J Haematol
95
1996
673
240
Menasce
LP
Orphanos
V
Santibanez-Koref
M
Boyle
JM
Harrison
CJ
Deletion of a common region on the long arm of chromosome 6 in acute lymphoblastic leukemia.
Genes Chromosomes Cancer
10
1994
26
241
Sherratt
T
Morelli
C
Boyle
JM
Harrison
CJ
Analysis of chromosome 6 deletions in lymphoid malignancies provides evidence for a region of minimal deletion within a 2-megabase segment of 6q21.
Chromosome Res
5
1997
118
242
Gerard
B
Cave
H
Guidal
C
Dastudue
N
Vilmer
E
Grandchamp
B
Delineation of a 6 cM commonly deleted region in childhood acute lymphoblastic leukemia on the 6q chromosomal arm.
Leukemia
11
1997
28
243
Menasce
LP
Orphanos
V
Santibanez-Koref
M
Boyle
JM
Harrison
CJ
Common region of deletion on the long arm of chromosome 6 in non-Hodgkin's lymphoma and acute lymphoblastic leukaemia.
Genes Chromosomes Cancer
10
1994
286
244
Park
JG
Reddy
EP
Large-scale molecular mapping of human c-myb locus: c-myb proto-oncogene is not involved in 6q- abnormalities of lymphoid tumors.
Oncogene
7
1992
1603
245
Menasce
LP
White
GR
Harrison
CJ
Boyle
JM
Localization of the estrogen receptor locus (ESR) to chromosome 6q25.1 by FISH and a simple post-FISH banding technique.
Genomics
17
1993
263
246
Garcia
M
Derocq
D
Freiss
G
Rochefort
H
Activation of estrogen receptor transfected into a receptor-negative breast cancer cell line decreases the metastatic and invasive potential of the cells.
Proc Natl Acad Sci USA
89
1992
11538
247
Issa
J-PJ
Zehnbauer
BA
Civin
CI
Collector
MI
Sharkis
SJ
Davidson
NE
Kaufmann
SH
Baylin
SB
The estrogen receptor CpG island is methylated in most hematopoietic neoplasms.
Cancer Res
56
1996
973
248
de la Chapelle
How do human isochromosomes arise?
Cancer Genet Cytogenet
5
1982
173
249
Grasso
M
Giovannucci Uziella
ML
Pierluigi
M
Tavellini
F
Perroni
L
Dagna Bricarelli
F
Isochromosome not translocation in trisomy 21q21.
Hum Genet
84
1989
63
250
Martineau
M
Clark
R
Farrell
DM
Hawkins
JM
Moorman
V
Secker-Walker
LM
Isochromosomes in acute lymphoblastic leukaemia: i(21q) is a significant finding.
Genes Chromosomes Cancer
17
1996
21
251
Mertens
F
Johansson
B
Mitelman
F
Isochromosomes in neoplasia.
Genes Chromosomes Cancer
10
1994
221
252
Pui
C-H
Carroll
AJ
Raimondi
SC
Schell
MJ
Head
DR
Shuster
JJ
Crist
WM
Borowitz
MJ
Link
MP
Behm
FG
Steuber
CP
Land
VJ
Isochromosomes in childhood acute lymphoblastic leukemia: A collaborative study of 83 cases.
Blood
79
1992
2384
253
Pui
C-H
Raimondi
SC
Williams
DL
Isochromosome 17q in childhood acute lymphoblastic leukemia: An adverse cytogenetic feature in association with hyperdiploidy?
Leukemia
2
1988
222
254
Baker
SJ
Markowits
S
Fearon
ER
Wilson
JKV
Vogelstein
B
Suppression of human colorectal carcinoma cell growth by wild-type p53.
Science
249
1990
912
255
Nevins
JR
E2F: A link between the Rb Tumor suppressor protein and viral oncoproteins.
Science
258
1992
424
256
Hollstein
M
Sidransky
D
Vogelstein
B
Harris
CC
p53 mutations in human cancers.
Science
253
1993
49
257
Levine
AJ
Momand
J
Tumor suppressor genes: The p53 and retinoblastoma sensitivity genes and gene products.
Biochim Biophys Acta
119
1990
1032
258
Friend
SH
Bernards
R
Rogeij
S
Weinberg
RA
Rapaport
JM
Albert
DM
Dryja
TP
A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma.
Nature
323
1986
643
259
Ahuja
HG
Jat
PS
Foti
A
Bar-Eli
M
Cline
MJ
Abnormalities of the retinoblastoma gene in the pathogenesis of acute leukemia.
Blood
78
1991
3259
260
Levine
AJ
Momand
J
Finlay
CA
The p53 tumor suppressor gene.
Nature
351
1991
453
261
Ryan
JJ
Danish
R
Gottlieb
CA
Clarke
MF
Cell cycle analysis of p53-induced death in murine erythroleukemia cells.
Mol Cell Biol
13
1993
711
262
Yonish-Rouach
E
Resnitzky
D
Lotem
J
Sachs
L
Kimchi
A
Oren
M
Wild-type p53 induces apoptosis of myeloid leukaemic cells that is inhibited by interleukin-6.
Nature
352
1991
345
263
Wada
M
Bartram
CR
Nakamura
H
Hachiya
M
Chen
D-L
Borenstein
J
Miller
CW
Ludwig
L
Hansen-Hagge
TE
Ludwid
W-D
Reiter
A
Mizoguchi
H
Koeffler
HP
Analysis of p53 mutations in a large series of lymphoid hematologic malignancies of childhood.
Blood
82
1993
3163
264
Sugimoto
K
Toyoshima
H
Sakai
R
Miyagawa
K
Hagiwara
K
Hirai
H
Ishikawa
F
Takaku
F
Mutations of the p53 gene in lymphoid leukemia.
Blood
77
1991
1153
265
Fenaux
P
Jonveaux
P
Quiquandron
I
Preudhomme
C
Laı̈
JL
Vanrumbeke
M
Loucheux-Lefebvre
MH
Bauters
F
Berger
R
Kerckaert
P
Mutations of the p53 gene in B-cell lymphoblastic acute leukemia: A report on 60 cases.
Leukemia
6
1992
42
266
(suppl)
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
1992
85S
267
Lanza
C
Gaidano
G
Cimino
G
Lo Coco
F
Basso
G
Sainati
L
Pastore
C
Nomdedeu
J
Volpe
G
Parvis
G
Barisone
E
Mazza
U
Madon
E
Saglio
G
p53 gene inactivation in acute lymphoblastic leukemia of B cell lineage associates with chromosomal breakpoints at 11q23 and 8q24.
Leukemia
9
1995
955
268
Kawamura
M
Kikuchi
A
Kobayashi
S
Hanada
R
Yamamoto
K
Horibe
K
Shikano
T
Ueda
K
Hayashi
K
Sekiya
T
Hayashi
Y
Mutations of the p53 and ras genes in childhood t(1;19)-acute lymphoblastic leukemia.
Blood
85
1995
2546
269
Diccianni
MB
Yu
J
Hsiao
M
Mukherjee
S
Shao
LE
Yu
AL
Clinical significance of p53 mutations in relapsed T-cell acute lymphoblastic leukemia.
Blood
84
1994
3105
270
Hsiao
MH
Yu
AL
Yeargin
J
Haas
M
Nonhereditary p53 mutations in T-cell acute lymphoblastic leukemia are associated with the relapse phase.
Blood
83
1994
2922
271
Preudhomme
C
Dervite
I
Wattel
E
Vanrumbeke
M
Flactif
M
Laı́
JL
Hecquet
B
Coppin
MC
Nelken
B
Gosselin
B
Fenaux
P
Clinical significance of p53 mutations in newly diagnosed Burkitt's lymphoma and acute lymphoblastic leukemia: A report of 48 cases.
J Clin Oncol
13
1995
812
272
Tsai
T
Davalath
S
Rankin
C
Radich
JP
Head
D
Appelbaum
FR
Boldt
DH
Tumor suppressor gene alteration in adult acute lymphoblastic leukemia (ALL). Analysis of retinoblastoma (Rb) and p53 gene expression in lymphoblasts of patients with de novo, relapsed, or refractory ALL treated in the Southwest Oncology Group studies.
Leukemia
10
1996
1901
273
Hangaishi
A
Ogawa
S
Imamura
N
Miyawaki
S
Miura
Y
Uike
N
Shimazaki
C
Emi
N
Takeyama
K
Hirosawa
S
Kamada
N
Kobayashi
Y
Takemoto
Y
Kitani
T
Toyama
K
Ohtake
S
Yazaki
Y
Ueda
R
Hirai
H
Inactivation of multiple tumor-suppressor genes involved in negative regulation of the cell cycle, MTS1/p16INK4A/CDKN2, MTS2/p15INK4B, p53, and Rb genes in primary lymphoid malignancies.
Blood
87
1996
4949
274
Rambaldi
A
Attuati
V
Bassan
R
Neonato
MG
Viero
P
Battista
R
Di Bona
E
Rossi
G
Pogliani
E
Ruggeri
M
Amaru
R
Rivolta
A
Giudici
G
Biondi
A
Barbui
T
Molecular diagnosis and clinical relevance of t(9;22), t(4;11) and t(1;19) chromosome abnormalities in a consecutive group of 141 adult patients with acute lymphoblastic leukemia.
Leuk Lymphoma
21
1996
457
Sign in via your Institution