Although numerous somatic mutations that contribute to the pathogenesis of childhood acute lymphoblastic leukemia (ALL) have been identified, no specific cytogenetic or molecular abnormalities are known to be consistently associated with relapse. Thep16INK4A (p16), which encodes for both p16INK4A and p19ARF proteins, andp15INK4B (p15) genes are inactivated by homozygous deletion and/or p15 promoter hypermethylation in a significant proportion of cases of childhood ALL at the time of initial diagnosis. To determine whether alterations in these genes play a role in disease progression, we analyzed a panel of 18 matched specimen pairs collected from children with ALL at the time of initial diagnosis and first bone marrow relapse for homozygous p16 and/orp15 deletions or p15 promoter hypermethylation. Four sample pairs contained homozygous p16 and p15 deletions at both diagnosis and relapse. Among the 14 pairs that werep16/p15 germline at diagnosis, three ALLs developed homozygous deletions of both p16 and p15, and two developed homozygous p16 deletions and retained p15germline status at relapse. In two patients, p15 promoter hypermethylation developed in the interval between initial diagnosis and relapse. In total, homozygous p16 deletions were present in nine of 18 cases, homozygous p15 deletions in seven of 18 cases, and p15 promoter hypermethylation in two of eight cases at relapse. These findings indicate that loss of function of proteins encoded by p16 and/or p15 plays an important role in the biology of relapsed childhood ALL, and is associated with disease progression in a subset of cases.

RELAPSED ACUTE lymphoblastic leukemia (ALL) is the fourth most common malignancy that occurs in children.1 While approximately 75% of children with newly diagnosed ALL are expected to be cured with contemporary treatment regimens, the outcome for children who experience a bone marrow relapse is poor, with a 6-year survival rate of 20% in a recent report from the Children’s Cancer Group.1 An important challenge is to define the biologic and genetic basis for these differences in treatment outcome.

Despite identification of a wide variety of oncogenes and tumor-suppressor genes (TSGs) that encode for proteins involved in leukemogenesis, relatively little is known about the genetic features of relapsed ALL. While it is likely that mutations in genes that encode proteins involved in transport or metabolism of chemotherapy agents (eg, MDR1) play a role in the progression of ALL, several observations indicate that mutations must also develop in specific oncogenes/TSGs. Relapsed ALL is often sensitive to the same drugs with which the patient was initially treated, arguing against selection for absolute drug resistance as a sole explanation for relapse. Permanent cell lines can be established more frequently from patients at relapse than at initial diagnosis.2 Similarly, relapse ALL cells engraft in irradiated immunodeficient mice more readily than cells from diagnosis.3 Confirming the multihit model of oncogenesis, oncogene and TSG mutations have been linked to progression in specific subtypes of leukemia.4-8 

Cytogenetic and molecular data suggest that TSG inactivation plays an important role in leukemogenesis; these same TSGs are also attractive candidates for the genes involved in progression of leukemia. To date,p16INK4A (p16, MTSI, CDKN2) and p15INK4B (p15, MTS2,CDKN2B) are the only TSGs identified that are inactivated in a significant percentage of leukemias at the time of initial diagnosis.9,10,p16 and p15, located within 25 kb of one another on the short arm of chromosome 9 (9p21), encode proteins (p16 and p15) that function as inhibitors of the cyclin-dependent kinases CDK4 and CDK6, which, complexed with cyclin D, phosphorylate the retinoblastoma protein (Rb) to allow progression through the cell cycle.11-13,p16 also encodes a second protein, p19ARF, via an alternative first exon and translation of common exons 2 and 3 in a different reading frame.14 Recent evidence suggests that p19ARF, which shares no amino acid homology with p16, enhances the functional activity of wild-type p53.15-17 Thus, homozygousp16 deletion is predicted to functionally inactivate growth-inhibitory proteins that act upstream of pathways involving both Rb and p53.18 Animal models support a critical role for proteins encoded by p16 in oncogenesis, as knockout mice that lack both p16 and p19ARF, or p19ARF alone, develop tumors, the most common of which are lymphomas, at greatly increased frequency.18 19 However, it remains uncertain whether inactivation of p16, p19ARF, or both is the critical event in human leukemogenesis.

Homozygous deletion of both p16 and p15 occurs in at least 60% of T- and 20% of B-lineage cases, and is the major means of functional inactivation of the proteins encoded by these genes in childhood ALL.20-22 In contrast to other tumors,p16 and p15 point mutations are rarely detected in childhood ALL.21,p15 is also frequently inactivated by promoter hypermethylation in T-ALL, but p16 promoter methylation is uncommon in childhood ALL.22 Limited information is available regarding potential changes inp16/p15 gene status in the interval between diagnosis and relapse in childhood ALL. However, p16/p15 lesions have been associated with progression of other hematolymphoid malignancies, including chronic myelogenous leukemia (CML) in lymphoid blast crisis, high-grade and transformed non-Hodgkin’s lymphomas, and myelodysplastic syndromes.7,8,23 24 

In this study, we analyzed a panel of 18 matched specimens obtained at diagnosis and first bone marrow relapse from children with ALL. We found that homozygous p16/p15 deletions and p15promoter hypermethylation are frequently acquired in the interval between initial diagnosis and relapse, suggesting that loss of function of the proteins encoded by these genes may play an important role in progression of childhood ALL.

Specimens.

Eighteen matched diagnosis and relapse bone marrow samples from children diagnosed with ALL were available from the cell bank of The Children’s Hospital in Denver. Diagnosis specimens were collected from 1982 to 1994, and relapse specimens were collected from 1982 to 1997. Mononuclear cells were collected by density centrifugation of fresh bone marrow aspirates, resuspended in 10% dimethyl sulfoxide (DMSO) and 20% fetal bovine serum, and maintained at either −70°C or in the vapor phase of liquid nitrogen. In the current study, we analyzed all matched diagnosis/first bone marrow relapse specimen pairs collected from patients with greater than 75% blasts in their bone marrow from whom sufficient material was available that we anticipated being able to isolate at least 10 μg of DNA. Immunophenotyping was performed at the time of presentation using panels of monoclonal antibodies that changed significantly between 1982 and 1997. In general, sufficient data were available to characterize leukemias as B or T lineage. All specimens were collected as part of protocols that had been approved by the Institutional Review Board (IRB) of the University of Colorado Health Sciences Center (UCHSC) and The Children’s Hospital, and the current retrospective analyses were also approved by the IRB.

Cytogenetics.

Giemsa-banded cytogenetic studies were performed from unstimulated cultures of bone marrow aspirate obtained at diagnosis or relapse. From 1982 to 1986, overnight cultures were synchronized using methodology described by Morse et al.25 After 1986, samples were prepared using a direct technique and overnight culture methods described previously with and without giant cell tumor supernatant supplementation.26,27 All cytogenetic results were reviewed by one of the authors (L.M.) and, whenever possible, described using the International System for Cytogenetic Nomenclature (ISCN, 1995).28 

Molecular analyses.

Genomic DNA was isolated by standard phenol/chloroform extraction. Southern blot analysis was performed as described previously.29 Briefly, membranes containingBamHI-digested DNAs were cohybridized with a 360-bp DNA fragment corresponding to p16 exon 2 and a previously describedMLL cDNA probe.30 This p16 probe cross-hybridizes with p15, allowing detection of deletions of either p16 or p15. Samples in which p16 and/orp15 bands were absent or were less than 10% to 20% of the intensities of control MLL bands were scored as containing homozygous deletions of the corresponding gene. For leukemias containing MLL translocations, blots were stripped and rehybridized with the p16 probe (along with a BCR cDNA probe) to insure that bands of altered migration did not representp16/p15 rearrangements.

To determine the methylation status of the p15 promoter region, membranes containing HindIII and EagI in combination with HindIII-digested DNAs were hybridized with a 270-bpp15 exon 1 probe as previously described.31Appropriate positive (DNA from the Kg1A cell line, which contains a hypermethylated p15 promoter) and negative (DNA from a healthy individual) controls were included on each blot.

Characteristics of study population.

We analyzed a panel of 18 paired specimens collected from children with ALL at initial diagnosis and first bone marrow relapse (Table1). Relapses occurred at a median of 36.7 months (range, 13 to 82) following initial diagnosis. Seven patients were classified as B lineage (CD10+, CD19+), three patients as T lineage (CD7+, CD2+), six patients were CD10+ but not further phenotyped, and there were two cases of CD10negative infant ALL. No major differences in immunophenotype developed in the interval between initial diagnosis and relapse.

Cytogenetic analyses.

Clonal karyotypic abnormalities were identified in 15 cases at initial diagnosis. Six ALLs were hyperdiploid with modal chromosome numbers of 52 to 60. Several cases contained recognized, nonrandom cytogenetic abnormalities, including del(6q) (DR-3, DR-9, DR-14), and t(4;11) (DR-16). At relapse, the clone observed at initial diagnosis was observed in eight of 15 cases. Additional numerical and structural changes were present within this clone in several cases. Of note, a del(9)(p22p24) developed in the interval between diagnosis and relapse in case DR-3. Two of 18 cases had incomplete analyses at relapse, and four of 18 had a 46,XX or 46,XY karyotype at relapse. Clonal abnormalities were observed at relapse in two of three cases that did not have clonal abnormalities detected at initial diagnosis. Cytogenetic analyses were unsuccessful due to culture failure for case DR-7 at initial diagnosis, but a t(2;11)(p1?12;q23) was identified at relapse. This translocation did not affect the 11q23 gene MLL, which was germline at both diagnosis and relapse (data not shown). Cytogenetic results were not available from DR-20 at initial diagnosis, when she was 2 months old. At relapse, a t(11;19)(q23;p13) was observed. Identical MLL rearrangements were present in both the diagnosis and relapse samples (data not shown), indicating that the t(11;19) was also present at initial diagnosis.

p16 and p15 gene deletions.

Southern blot analysis was used to determine p16 andp15 gene status (Fig 1 and Table1). Four sample pairs contained homozygous p16 and p15deletions at both diagnosis and relapse. Two of these patients had T-ALL, and the other two were classified as CD10+ ALLs. Among the 14 patients who were p16/p15 germline at diagnosis, three B-lineage ALLs exhibited homozygous deletions of bothp16 and p15 at relapse. In two additional patients (DR-10, a T-ALL, and DR-18, a CD10+ ALL), homozygous deletion of p16 was present in the relapse specimens, whilep15 retained germline status. In total, homozygous p16deletions were present in nine of 18 cases and homozygous p15deletions in seven of 18 cases at relapse.

Fig. 1.

p16/p15 deletions in matched diagnostic and relapse ALL specimen pairs. Autoradiogram of a blot containingBamH1-digested DNAs cohybridized with p16 and MLLcDNA probes. The locations of the germline p16,p15, and MLL bands are indicated by arrows at right. The migration of molecular size markers are shown in kilobases on the left. Samples include DNA from dilutions of the K562 cell line, which has homozygous p16 and p15 deletions, into normal DNA to simulate deletions in 50% and 10% of the cell population; a healthy control (NORM); K562; and four matched diagnostic (Dx) and relapse (R) patient samples (no. 4, 10, 17, 6). The gene status ofp16 and p15 is listed above each patient sample (G, germline; D, deleted). Faint residual p16 and p15 bands (<10%) in no. 6 relapse patient sample are due to contamination with small amounts of normal cells.

Fig. 1.

p16/p15 deletions in matched diagnostic and relapse ALL specimen pairs. Autoradiogram of a blot containingBamH1-digested DNAs cohybridized with p16 and MLLcDNA probes. The locations of the germline p16,p15, and MLL bands are indicated by arrows at right. The migration of molecular size markers are shown in kilobases on the left. Samples include DNA from dilutions of the K562 cell line, which has homozygous p16 and p15 deletions, into normal DNA to simulate deletions in 50% and 10% of the cell population; a healthy control (NORM); K562; and four matched diagnostic (Dx) and relapse (R) patient samples (no. 4, 10, 17, 6). The gene status ofp16 and p15 is listed above each patient sample (G, germline; D, deleted). Faint residual p16 and p15 bands (<10%) in no. 6 relapse patient sample are due to contamination with small amounts of normal cells.

Close modal
p15 promoter hypermethylation.

Using the strategy outlined in Fig 2, we determined whether or not the p15 promoter region was hypermethylated, which correlates with silencing of gene expression, in samples that did not contain p15 deletions. Sufficient DNA was available to perform these analyses for eight diagnostic samples.p15 exon 1 was homozygously hypermethylated in three of these eight patients, and unmethylated in the remaining five (Table 1). Sufficient DNA was available to perform these analyses for eight of 11p15-germline relapse specimens. Two cases reproducibly showed partial p15 promoter hypermethylation (Fig 2 and Table 1), suggesting either that one allele was hypermethylated in all cells, or that a subpopulation, accounting for about half the cells, was homozygously hypermethylated. We observed several different patterns ofp15 methylation status in the matched specimens. Particularly interesting is case DR-10, which had no evident p16/p15abnormalities at initial diagnosis, and contained homozygousp16 deletion and heterozygous p15 promoter hypermethylation at relapse (Figs 1 and 2).

Fig. 2.

p15 promoter hypermethylation. (A) Schematic representation of p15 exon 1 and surrounding HindIII (H3) and EagI (E) sites. The p15 probe used (indicated above) hybridizes to exon 1. A 2.8-kb band is seen when the samples are digested with HindIII alone. When the samples are digested withEagI (a methylation-sensitive enzyme) in combination withHindIII, a 0.6-kb band is recognized by the probe if the gene is unmethylated (cuts at EagI) and a 2.8-kb band is present if the sample is methylated (does not cut at EagI) (note that a 2.2-kb band is not seen, because the exon 1 probe does not hybridize to this region of DNA). (B) Autoradiogram of a blot containing HindIII and EagI-in combination with HindIII-digested DNAs hybridized with the p15 exon 1 probe. The locations of the expected 2.8-kb and 0.6-kb bands are indicated by arrows at the left. Samples include DNA from Kg1A cell line, a healthy control (NORM), DR#10 at diagnosis (Dx) and at relapse (R). Homozygous methylation is seen in Kg1A, and hemizygous methylation in DR#10 at relapse, but not at initial diagnosis.

Fig. 2.

p15 promoter hypermethylation. (A) Schematic representation of p15 exon 1 and surrounding HindIII (H3) and EagI (E) sites. The p15 probe used (indicated above) hybridizes to exon 1. A 2.8-kb band is seen when the samples are digested with HindIII alone. When the samples are digested withEagI (a methylation-sensitive enzyme) in combination withHindIII, a 0.6-kb band is recognized by the probe if the gene is unmethylated (cuts at EagI) and a 2.8-kb band is present if the sample is methylated (does not cut at EagI) (note that a 2.2-kb band is not seen, because the exon 1 probe does not hybridize to this region of DNA). (B) Autoradiogram of a blot containing HindIII and EagI-in combination with HindIII-digested DNAs hybridized with the p15 exon 1 probe. The locations of the expected 2.8-kb and 0.6-kb bands are indicated by arrows at the left. Samples include DNA from Kg1A cell line, a healthy control (NORM), DR#10 at diagnosis (Dx) and at relapse (R). Homozygous methylation is seen in Kg1A, and hemizygous methylation in DR#10 at relapse, but not at initial diagnosis.

Close modal

In the current study, we found that homozygous p16 deletions were common at the time of first bone marrow relapse of childhood ALL (9 of 18 cases). While four of these cases contained homozygous deletions at both diagnosis and relapse, 5 of the 14 cases (36%) that were p16/p15 germline at diagnosis acquired homozygousp16 deletions in the interval between diagnosis and relapse. Three of these five cases also acquired homozygous p15deletions, and one acquired a partial p15 exon 1 hypermethylation. One additional case acquired partial p15promoter hypermethylation at relapse in the absence of p16abnormalities. These results suggest that acquisition of p16and p15 abnormalities may be a critical event associated with disease progression in children with ALL. As all deletions involvedp16 exon 2, which is included in both p16 andp19ARF transcripts,14,19 it is not certain whether the critical protein target was p16, p19ARF, or both. Experimental data indicate that inactivation of these two proteins profoundly alters cell-cycle progression and growth arrest via loss of a negative regulator of the Rb pathway (p16) and a positive regulator of wild-type p53 activity (p19ARF),11-13 15-17 suggesting that these alterations may contribute to the clinical chemotherapy refractoriness observed in relapsed ALL.

It is important to emphasize that our results do not distinguish between two different potential explanations for the presence ofp16/p15 abnormalities in relapse, but not diagnosis, samples. First, the p16/p15 abnormalities may not have been present in any of the leukemic cells at diagnosis, but rather developed during (and perhaps instigated) the process of disease recurrence. Alternatively, a small subclone, that was below the limits of detection of Southern blot analysis, containing these genetic abnormalities could have been present at the time of initial diagnosis, and loss of p16/p15/p19ARF function could have conferred a proliferative and/or survival advantage that allowed this to become the dominant clone at relapse.

To our knowledge, this is the largest series of childhood ALLs in whichp16 and p15 gene status has been determined in matched diagnostic and relapse specimen pairs. Ohnishi et al reported thatp16 status was unchanged (one deleted, six germline) between diagnosis and relapse in seven paired samples.21 Takeuchi et al used microsatellite analysis to identify deletions within 9p in six matched pairs of childhood ALLs and found that no differences developed in the pattern of allelic loss between diagnosis and relapse.32 In contrast, Ogawa et al found that at least hemizygous loss of p16 developed between initial diagnosis and relapse in two of three paired samples.33 To determine whether there is any cytogenetic data to support our findings that acquisition of p16/p15 abnormalities are likely to play an important role in progression of childhood ALL, we reviewed the published literature on karyotypes from children with relapsed ALL, focusing on 9p abnormalities. In five series, complete karyotypes were available from 34 cases of childhood ALL that had an abnormal karyotype at diagnosis, and had additional changes within this clone at relapse.34-38 Deletions of 9p evolved between diagnosis and relapse in four of these 34 (12%) cases. As the majority of ALLs containing p16/p15 deletions do not have visible 9p abnormalities (and only one of the five cases that acquiredp16/p15 deletions in our study also acquired visible 9p abnormalities), these cytogenetic data likely underestimate the true incidence of new p16/p15 abnormalities.

Taken together with data from other studies,7,8,23,24 39these results strongly suggest that p16/p15 genetic abnormalities play an important role in the biology of relapsed hematologic and lymphoid malignancies, and may be directly related to disease progression. Our sample is too small, and the clinical characteristics and treatment of the patients too heterogeneous, to draw any conclusions regarding the potential prognostic import ofp16/p15 abnormalities at relapse. In the future, it will be important to analyze a larger, more homogeneous group of patients to accurately define the percentage of childhood ALLs that acquire p16/p15 abnormalities in the interval between initial diagnosis and relapse, and to determine whether this has prognostic significance. Increased understanding of the role ofp16/p15 inactivation in the initiation and progression of lymphoid malignancies should provide important insights into leukemia biology and lay the groundwork for rationally designed therapeutic interventions.

K.W.M. is the recipient of the Greg and Laura Norman Fellowship Award from the National Childhood Cancer Foundation and is also supported in part by a University of Colorado Cancer Center seed grant. S.P.H. was supported by a BLOOD/ASH Scholar Award and a Professional Development Award from The Children’s Hospital Research Institute, Denver, CO, and is a Leukemia Society Translational Research Awardee. Research supported by a grant from the Cancer League of Colorado to S.P.H. and a Cancer Center Core Grant (CA 46934).

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

1
Gaynon
PS
Qu
RP
Chappell
RJ
Willoughby
ML
Tubergen
DG
Steinherz
PG
Trigg
ME
Survival after relapse in childhood acute lymphoblastic leukemia: Impact of site and time to first relapse—The Children’s Cancer Group experience.
Cancer
82
1998
1387
2
Smith
SD
McFall
P
Morgan
R
Link
M
Hecht
F
Cleary
M
Sklar
J
Long-term growth of malignant thymocytes in vitro.
Blood
73
1989
2182
3
Kamel-Reid
S
Letarte
M
Doedens
M
Greaves
A
Murdoch
B
Grunberger
T
Lapidot
T
Thorner
P
Freedman
MH
Phillips
RA
Dick
JE
Bone marrow from children in relapse with pre-B acute lymphoblastic leukemia proliferates and disseminates rapidly in scid mice.
Blood
78
1991
2973
4
Ahuja
H
Bar-Eli
M
Advani
SH
Benchimol
S
Cline
MJ
Alterations in the p53 gene and the clonal evolution of the blast crisis of chronic myelocytic leukemia.
Proc Natl Acad Sci USA
86
1989
6783
5
Hsiao
MH
Yu
AL
Yeargin
J
Ku
D
Haas
M
Nonhereditary p53 mutations in T-cell acute lymphoblastic leukemia are associated with the relapse phase.
Blood
83
1994
2922
6
Wada
C
Shionoya
S
Fujino
Y
Tokuhiro
H
Akahoshi
T
Uchida
T
Ohtani
H
Genomic instability of microsatellite repeats and its association with the evolution of chronic myelogenous leukemia.
Blood
83
1994
3449
7
Serra
A
Gottardi
E
Della Ragione
F
Saglio
G
Iolascon
A
Involvement of the cyclin-dependent kinase-4 inhibitor (CDKN2) gene in the pathogenesis of lymphoid blast crisis of chronic myelogenous leukaemia.
Br J Haematol
91
1995
625
8
Sill
H
Goldman
JM
Cross
NC
Homozygous deletions of the p16 tumor-suppressor gene are associated with lymphoid transformation of chronic myeloid leukemia.
Blood
85
1995
2013
9
Haidar
MA
Cao
XB
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
10
Quesnel
B
Preudhomme
C
Philippe
N
Vanrumbeke
M
Dervite
I
Lai
JL
Bauters
F
Wattel
E
Fenaux
P
p16 gene homozygous deletions in acute lymphoblastic leukemia.
Blood
85
1995
657
11
Kamb
A
Gruis
NA
Weaver-Feldhaus
J
Liu
Q
Harshman
K
Tavtigian
SV
Stockert
E
Day
RSr
Johnson
BE
Skolnick
MH
A cell cycle regulator potentially involved in genesis of many tumor types.
Science
264
1994
436
12
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
13
Serrano
M
Hannon
GJ
Beach
D
A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4.
Nature
366
1993
704
14
Quelle
DE
Zindy
F
Ashmun
RA
Sherr
CJ
Alternative reading frames of the INK4a tumor suppressor gene encode two unrelated proteins capable of inducing cell cycle arrest.
Cell
83
1995
993
15
Pomerantz
J
Schreiber-Agus
N
Liegeois
NJ
Silverman
A
Alland
L
Chin
L
Potes
J
Chen
K
Orlow
I
Lee
HW
Cordon-Cardo
C
DePinho
RA
The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2’s inhibition of p53.
Cell
92
1998
713
16
Zhang
Y
Xiong
Y
Yarbrough
WG
ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways.
Cell
92
1998
725
17
Kamijo
T
Weber
JD
Zambetti
G
Zindy
F
Roussel
MF
Sherr
CJ
Functional and physical interactions of the ARF tumor suppressor with p53 and Mdm2.
Proc Natl Acad Sci USA
95
1998
8292
18
Kamijo
T
Zindy
F
Roussel
MF
Quelle
DE
Downing
JR
Ashmun
RA
Grosveld
G
Sherr
CJ
Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF.
Cell
91
1997
649
19
Serrano
M
Lee
H
Chin
L
Cordon-Cardo
C
Beach
D
DePinho
RA
Role of the INK4a locus in tumor suppression and cell mortality.
Cell
85
1996
27
20
Okuda
T
Shurtleff
SA
Valentine
MB
Raimondi
SC
Head
DR
Behm
F
Curcio-Brint
AM
Liu
Q
Pui
CH
Sherr
CJ
Beach
D
Look
AT
Downing
JR
Frequent deletion of p16INK4a/MTS1 and p15INK4b/MTS2 in pediatric acute lymphoblastic leukemia.
Blood
85
1995
2321
21
Ohnishi
H
Kawamura
M
Ida
K
Sheng
XM
Hanada
R
Nobori
T
Yamamori
S
Hayashi
Y
Homozygous deletions of p16/MTS1 gene are frequent but mutations are infrequent in childhood T-cell acute lymphoblastic leukemia.
Blood
86
1995
1269
22
Batova
A
Diccianni
MB
Yu
JC
Nobori
T
Link
MP
Pullen
J
Yu
AL
Frequent and selective methylation of p15 and deletion of both p15 and p16 in T-cell acute lymphoblastic leukemia.
Cancer Res
57
1997
832
23
Pinyol
M
Cobo
F
Bea
S
Jares
P
Nayach
P
Fernandez
PL
Montserrat
E
Cardesa
A
Campo
E
p16 INK4a gene inactivation by deletions, mutations, and hypermethylation is associated with transformed and aggressive variants of non-Hodgkin’s lymphomas.
Blood
91
1998
2977
24
Quesnel
B
Guillerm
G
Vereecque
R
Wattel
E
Preudhomme
C
Bauters
F
Vanrumbeke
M
Fenaux
P
Methylation of the p15(INK4b) gene in myelodysplastic syndromes is frequent and acquired during disease progression.
Blood
91
1998
2985
25
Morse
HG
Humbert
JR
Hutter
JJ
Robinson
A
Karyotyping of bone-marrow cells in hematologic diseases.
Hum Genet
37
1977
33
26
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
27
Hunger
SP
Sun
T
Boswell
AF
Carroll
AJ
McGavran
L
Hyperdiploidy and E2A-PBX1 fusion in an adult with t(1;19)+ acute lymphoblastic leukemia: Case report and review of the literature.
Genes Chromosomes Cancer
20
1997
392
28
Mitelman
F
ISCN (1995): An International System for Human Cytogenetic Nomenclature.
1995
Karger
Basel, Switzerland
29
Maloney
KW
Rubnitz
JE
Cleary
ML
Frankel
LS
Hakami
N
Link
MP
Pullen
DJ
Hunger
SP
Lack of ETV6 (TEL) gene rearrangements or p16INK4A/p15INK4B homozygous gene deletions in infant acute lymphoblastic leukemia.
Leukemia
11
1997
979
30
Hunger
SP
Tkachuk
DC
Amylon
MD
Link
MP
Carroll
AJ
Welborn
JL
Willman
CL
Cleary
ML
HRX involvement in de novo and secondary leukemias with diverse chromosome 11q23 abnormalities.
Blood
81
1993
3197
31
Maloney
KW
McGavran
L
Odom
LF
Hunger
SP
Different patterns of homozygous p16 INK4A and p15 INK4B deletions in childhood acute lymphoblastic leukemias containing distinct E2A translocations.
Leukemia
12
1998
1417
32
Takeuchi
S
Koike
M
Seriu
T
Bartram
CR
Slater
J
Park
S
Miyoshi
I
Koeffler
HP
Homozygous deletions at 9p21 in childhood acute lymphoblastic leukemia detected by microsatellite analysis.
Leukemia
11
1997
1636
33
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
34
Kaneko
Y
Rowley
JD
Variakojis
D
Chilcote
RR
Check
I
Sakurai
M
Correlation of karyotype with clinical features in acute lymphoblastic leukemia.
Cancer Res
42
1982
2918
35
Secker-Walker
LM
Alimena
G
Bloomfield
CD
Kaneko
Y
Whang-Peng
J
Arthur
DC
de la Chapelle
A
Reeves
BR
Rowley
JD
Lawler
SD
Mitelman
F
Cytogenetic studies of 21 patients with acute lymphoblastic leukemia in relapse.
Cancer Genet Cytogenet
40
1989
163
36
Shikano
T
Ishikawa
Y
Ohkawa
M
Hatayama
Y
Nakadate
H
Hatae
Y
Takeda
T
Karyotypic changes from initial diagnosis to relapse in childhood acute leukemia.
Leukemia
4
1990
419
37
Abshire
TC
Buchanan
GR
Jackson
JF
Shuster
JJ
Brock
B
Head
D
Behm
F
Crist
WM
Link
M
Borowitz
M
Pullen
DJ
Morphologic, immunologic and cytogenetic studies in children with acute lymphoblastic leukemia at diagnosis and relapse: A Pediatric Oncology Group study.
Leukemia
6
1992
357
38
Heerema
NA
Palmer
CG
Weetman
R
Bertolone
S
Cytogenetic analysis in relapsed childhood acute lymphoblastic leukemia.
Leukemia
6
1992
185
39
Hatta
Y
Hirama
T
Miller
CW
Yamada
Y
Tomonaga
M
Koeffler
HP
Homozygous deletions of the p15 (MTS2) and p16 (CDKN2/MTS1) genes in adult T-cell leukemia.
Blood
85
1995
2699

Author notes

Address reprint requests to Stephen P. Hunger, MD, UCHSC Campus Box C229, 4200 E Ninth Ave, Denver, CO 80262; e-mail:Stephen.Hunger@UCHSC.edu.

Sign in via your Institution